ArticlePDF Available

Rosemary (Rosmarinus officinalis L.) extract inhibits prostate cancer cell proliferation and survival by targeting Akt and mTOR

Authors:

Abstract and Figures

Prostate cancer is the most commonly diagnosed type of cancer in North American men and is typically classified as either androgen receptor positive or negative depending on the expression of the androgen receptor (AR). AR positive prostate cancer can be treated with hormone therapy while AR negative prostate cancer is aggressive and does not respond to hormone therapy. It has been previously reported that rosemary extract (RE) has antioxidant, anti-inflammatory and anti-cancer properties. In the present study, we found that treatment of the androgen-insensitive PC-3 prostate cancer cells with RE resulted in a significant inhibition of proliferation, survival, migration, Akt, and mTOR signaling. In addition, treatment of the androgen-sensitive 22RV1 prostate cancer cells with RE resulted in a significant inhibition of proliferation and survival while RE had no effect on normal prostate epithelial PNT1A cells. These findings suggest that RE has potent effects against prostate cancer and warrants further investigation.
Content may be subject to copyright.
Biomedicine & Pharmacotherapy 131 (2020) 110717
Available online 23 September 2020
0753-3322/© 2020 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Original article
Rosemary (Rosmarinus ofcinalis L.) extract inhibits prostate cancer cell
proliferation and survival by targeting Akt and mTOR
Alina Jaglanian
a
, Deborah Termini
a
, Evangelia Tsiani
a
,
b
,
*
a
Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
b
Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
ARTICLE INFO
Keywords:
Rosemary extract
Prostate cancer
Proliferation
Survival
Akt
mTOR
ABSTRACT
Prostate cancer is the most commonly diagnosed type of cancer in North American men and is typically classied
as either androgen receptor positive or negative depending on the expression of the androgen receptor (AR). AR
positive prostate cancer can be treated with hormone therapy while AR negative prostate cancer is aggressive
and does not respond to hormone therapy. It has been previously reported that rosemary extract (RE) has
antioxidant, anti-inammatory and anti-cancer properties. In the present study, we found that treatment of the
androgen-insensitive PC-3 prostate cancer cells with RE resulted in a signicant inhibition of proliferation,
survival, migration, Akt, and mTOR signaling. In addition, treatment of the androgen-sensitive 22RV1 prostate
cancer cells with RE resulted in a signicant inhibition of proliferation and survival while RE had no effect on
normal prostate epithelial PNT1A cells. These ndings suggest that RE has potent effects against prostate cancer
and warrants further investigation.
1. Introduction
Prostate cancer accounted for roughly 1.3 million cases and 359,000
deaths globally in 2018, and is the second leading cause of death in
North American men despite all available treatment strategies including
surgery, radiotherapy, and chemotherapy [1]. Finding novel approaches
to prevent and treat prostate cancer effectively is highly desirable. Major
disruptions of cellular homeostasis of the prostate gland leads to prostate
cancer [2]. The growth of prostate epithelial cells is inuenced by
growth factors, the expression and function of androgen receptors (AR)
and by the hypothalamic-pituitary axis [2,3]. Androgens provide
important growth stimuli for prostate cells, and ARs are typically
expressed in the stromal and epithelial compartments of the prostate
gland [2,4]. Production of luteinizing hormone (LH)-releasing hormone
(LHRH) by the hypothalamus induces the production of LH by the pi-
tuitary gland [5] leading to increased androgen production. Hypotha-
lamic LHRH and pituitary LH production are regulated by a negative
feedback mechanism [5]. Androgen binding to AR leads to transcrip-
tional activation of AR target genes which are involved in various bio-
logical processes such as proliferation and apoptosis [24]. Overall, AR
signaling is directly involved in maintaining normal prostate tissue ho-
meostasis [2].
Cancer cells are characterized by their ability to proliferate uncon-
trollably and evade apoptosis [6,7]. These characteristics are often ac-
quired as a result of mutations in key proteins involved in the signaling
pathways responsible for regulating cellular function and maintaining
homeostasis [813]. Molecular signaling pathways of growth factor
receptors; such as Epidermal Growth Factor (EGF) Receptor (EGFR)
initiate signal transduction pathways that lead to increased cell prolif-
eration and survival [8,1416]. The phosphatidylinositol 3-kinase
(PI3K)/Akt pathway is activated by growth factor [17] and androgen
receptor [18] signaling and plays a prominent role in prostate cancer.
Mutations that result in the overactivation of this cascade, activation of
other oncogenes, and/or the inactivation of proteins that serve as tumor
suppressors such as p53, p27, and phosphatase and tensin homologue
(PTEN) [19,20], contribute to carcinogenesis and the development of
prostatic tumors [2123].
Increased Akt activation is associated with carcinogenesis as well as
increased resistance to chemotherapeutic agents such as cisplatin,
methotrexate, and paclitaxel [24,25]. Akt expression/activation is often
elevated in human prostate cancers [26,27]. Aberrations in the
PI3K/Akt pathway have been reported in approximately 40 % of early
prostate cancer and 70100 % of advanced cases [28,29]. Specically,
the loss of PTEN leads to the constitutive activation of the PI3K/Akt
* Corresponding author at: Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
E-mail addresses: aj11fo@brocku.ca (A. Jaglanian), dt14@brocku.ca (D. Termini), etsiani@brocku.ca (E. Tsiani).
Contents lists available at ScienceDirect
Biomedicine & Pharmacotherapy
journal homepage: www.elsevier.com/locate/biopha
https://doi.org/10.1016/j.biopha.2020.110717
Received 28 July 2020; Received in revised form 28 August 2020; Accepted 30 August 2020
Biomedicine & Pharmacotherapy 131 (2020) 110717
2
pathway [4]. Due to the importance of Akt in prostate cancer, several
small molecules that target/inhibit Akt are currently in clinical devel-
opment [30,31].
Mechanistic target of rapamycin (mTOR), a 289 kDa serine/threo-
nine kinase, is a downstream effector of the PI3K/Akt pathway, and is
involved in the control of cell growth [32,33]. Twice the levels of total
and phosphorylated mTOR have been reported in prostate cancer tissue
when compared to normal prostate epithelium [34]. As a result mTOR is
an appealing therapeutic target and mTOR inhibitors such as sirolimus,
deforolimus, everolimus, and temsirolimus are used as a monotherapy
or combined therapy for various types of cancers [24,33,35].
The process of programmed cell death, known as apoptosis, is an
essential process in the maintenance of cell homeostasis. The most
common signaling cascades involved in regulating cellular apoptosis
promote the downstream activation of caspases and Poly (ADP-ribose)
polymerase-1 (PARP-1) cleavage to form 89 and 24 kDa fragments [36].
PARP-1 is an enzyme responsible for DNA repair and therefore, plays a
role in genomic maintenance [37,38]. Cleaved PARP is an established
indicator of apoptosis [36,38].
Over 50 % of modern chemotherapeutic agents that are used for
cancer treatment are derived from natural products [39,40]; for
example, the chemotherapeutic drug paclitaxel was originally isolated
from the bark of the Pacic yew tree (Taxus brevifolia) and the chemo-
therapeutic drug docetaxel was originally isolated from the needles of
the European yew tree (Taxus baccata) [41].
Scientic interest in nding chemicals in plants with anti-cancer
potential continues today. The rosemary (Rosmarinus ofcinalis L.)
plant, native to Mediterranean countries, contains the polyphenols
carnosic acid (CA), rosmarinic acid (RA), and carnosol (COH) in high
concentrations [42,43]. In recent years, rosemary extract (RE) and RE
polyphenols have been reported to have antioxidant, antimicrobial, and
anti-cancer properties [4448]. Limited data exists regarding the effects
of rosemary extract in prostate cancer [4952], and little is known about
the underlying signaling mechanisms involved in mediating its
pro-apoptotic and anti-proliferative effects.
In the present study, we investigated the effects of rosemary extract
in PC-3 and 22RV1 prostate cancer cells, as well as in PNT1A normal
prostate epithelial cells.
2. Materials and methods
2.1. Materials
The PC-3 human epithelial prostate cancer cells were obtained from
American Type Culture Collection (ATCC) (Manassas, VA, USA). The
Roswell Park Memorial Institute (RPMI) 1640 Medium, fetal bovine
serum (FBS), 0.25 % trypsin and the antibioticantimycotic solution
were purchased from GIBCO Life Technologies (Burlington, ON, Can-
ada). Akt (#9272) (1:1000 dilution), p-Akt (Ser473) (#9271) (1:1000
dilution), mTOR (#2972) (1:1000 dilution), p-mTOR (Ser2448)
(#2971) (1:1000 dilution), PARP (#9542) (1:1000 dilution), β-actin
(#8457) (1:1000 dilution), as well as secondary anti-rabbit IgG HRP-
linked antibodies (#7074) (1:2000 dilution) were from Cell Signaling
Technology via New England Biolabs (Mississauga, ON, Canada).
22RV1 human epithelial prostate cancer cells, PNT1A normal prostate
epithelial cells, bovine serum albumin (BSA), dimethyl sulfoxide
(DMSO), docetaxel and paclitaxel were from Sigma (Oakville, ON,
Canada). Clarity western enhanced chemiluminescence (ECL) reagent,
30 % acrylamide/bis solution 37 (5:1), ammonium persulfate (APS),
polyvinylidene diuoride (PVDF) membranes and reagents for electro-
phoresis were purchased from Bio-Rad (Hercules, CA, USA).
2.2. Rosemary extract preparation
Whole dried rosemary (Rosmarinus ofcinalis L.) leaves (purchased
from Compliments/Sobeys, Mississauga, ON, Canada) were used, and
the rosemary extract was prepared as previously reported [53]. Briey,
dried rosemary leaves were ground and steeped overnight (16 h) in
dichloromethane: methanol (1:1) followed by ltration the next day.
After ltering, the solvent was set aside while the leaves were boiled in
methanol for 30 min. The solvent obtained after boiling was combined
with the ltered solvent. The combined solvent was removed from the
nal extract by rotary evaporation and the green powder was collected
and stored at 20 C, protected from light. Aliquots were prepared in
dimethyl sulfoxide (DMSO) to yield a stock concentration of
100 mg/mL, stored at 20 C and protected from light.
2.3. Cell culture and treatment
The cells were cultured in RPMI 1640 media supplemented with 1%
(v/v) antibioticantimycotic solution (containing 100 units/mL of
penicillin, 100
μ
g/mL of streptomycin, and 0.25
μ
g/mL of Amphotericin
B) and 10 % (v/v) FBS in an incubator at 37 C.
Cells were treated with a working stock of RE (400
μ
g/mL in cell
culture media) and the nal concentration of DMSO in the RE-treated
cells was less than 0.1 %. Exposure of the cells to DMSO to match the
concentration of DMSO seen by cells exposed to RE (vehicle control) did
not have any effect on any assays/measurements used in the current
study.
2.4. Cell proliferation assay
Cells were seeded (1000 cells/well) in a 96-well plate supplemented
with DMEM and treated as indicated in the gures for 72 h. The cells
were xed with 10 % formalin and stained using 0.5 % crystal violet
stain. The next day solubilizer solution containing 0.05 M NaH
2
PO
4
was
added into each well and the absorbance was read at 570 nm using the
KC4 microplate reader.
2.5. Clonogenic survival assay
Cells were seeded (1000 cells/well) in six-well plates and allowed to
adhere for 24 h followed by treatment as indicated in the gures for
seven days. At the end of the treatment, the cells were washed twice with
sterile phosphate-buffered saline (PBS) and stained with 0.05 % w/v
methylene blue. The next day, colonies greater than 50 cells were
counted under the microscope.
2.6. Wound healing assay
The wound healing assay was used to assess cell migration [54,55].
Cells were seeded at a density of 2.5 ×10
5
cells/mL into a 6-well plate
and the media was replaced every 48 h until the cells reached 90100 %
conuency. When conuency was reached the cells were incubated with
mitomycin-C (MMC) (1
μ
g/mL) for 1 h to prevent cell proliferation.
After the incubation period, a vertical line was drawn in the centre of
each well using a 100
μ
L pipette tip. The wells were drained of media
and washed twice with PBS to get rid of oating cells, followed by
treatment as indicated in the gures. Before taking photographs, hori-
zontal lines were drawn underneath the well plates to be used as a
reference for future time points. Photos were taken at 0 and 40 -h time
points. Wound closure percentage was calculated using the equation
0hr Area40 hr Area
0 Hr Area x 100. The area of each wound was measured using the
ImageJ software.
2.7. Immunoblotting
Cell lysate samples containing 20
μ
g of protein, determined using the
Bradford assay [56], were loaded onto 10 % polyacrylamide gel and
separated by sodium dodecyl sulfatepolyacrylamide gel electrophoresis
(SDS-PAGE). The separated proteins were then transferred onto a
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
3
polyvinylidene diuoride (PVDF), membrane which was exposed to
blocking buffer (5% (w/v) dry milk in Tris-buffered saline) for 1 h and
incubated with the primary antibody overnight at 4 C. The following
day the membrane was incubated with horseradish peroxidase
(HRP)-linked IgG anti-rabbit secondary antibody for 1 h at room tem-
perature. Enhanced chemiluminescence (ECL), the Bio-Rad clarity
western solution, was used to detect the bands corresponding to the
proteins of interest. Densitometric analysis was performed using ImageJ
software. The data (arbitrary densitometric units) were corrected to
β-actin levels and expressed as a percentage of untreated control cells.
2.8. Statistical analysis
The data are the mean ±standard error mean (SEM) of the indicated
number of independent experiments. Analysis of variance (ANOVA)
followed by Bonferronis post-hoc test was used to determine the sig-
nicance of the differences between groups. Signicance was assumed
at p <0.05. Statistical tests were performed using GraphPad Prism 8
software.
3. Results
3.1. Inhibition of PC-3 prostate cancer cell proliferation by rosemary
extract
The antiproliferative effects of RE were evaluated in the androgen
receptor negative PC-3 prostate cancer cells. PC-3 cells were exposed to
5, 10, 25, 50, 75, 100, or 150
μ
g/mL RE for 72 h and cell proliferation
was assessed using the crystal violet assay. The RE powder was dissolved
in dimethyl sulfoxide (DMSO) to create a stock solution (100 mg/mL).
This solution was diluted using cell culture media to create a working
stock (400
μ
g/mL) that was used to treat the cells. Treatment with RE
resulted in a dosedependent inhibition of cell proliferation (Fig. 1A). A
signicant inhibition (80.25 ±4.44 % of control, p <0.01) was seen
with 10
μ
g/mL RE while maximum inhibition (26.51 ±2.95 % of con-
trol, p <0.0001) was seen with 50
μ
g/mL RE (Fig. 1A-C). Higher RE
concentrations (75, 100 and 150
μ
g/mL) did not result in a statistically
greater inhibition of cell proliferation compared to 50
μ
g/mL (Fig. 1A).
The data from Fig. 1A were plotted on a log scale (Fig. 1B) and the
calculated RE concentration for the half maximal inhibition (IC
50
) of cell
proliferation was 19.72
μ
g/mL. Docetaxel (DTX), derived from Taxus
baccata and paclitaxel (PTX), derived from Taxus brevifolia, are estab-
lished medications used clinically in the treatment of prostate cancer
[57] and we used them in the present study to compare the effects of RE
to their effects. We used two different concentrations (5 and 10 nM) of
PTX and DTX based on other in vitro studies [58,59]. Treatment of the
cells with 5 nM PTX did not result in any signicant inhibition of cell
proliferation (91.27 ±2.79 % of control, p >0.05) (Fig. 1C), while
treatment with 10 nM of paclitaxel showed signicant inhibition of cell
proliferation (67.63 ±4.24 % of control, p <0.01) (Fig. 1C). Treatment
of the cells with 5 and 10 nM DTX resulted in signicant inhibition of
cell proliferation (38.11 ±1.16, 32.08 ±0.84 % of control respectively,
both p <0.0001) (Fig. 1C). The inhibition of cell proliferation seen with
50
μ
g/mL of rosemary extract (26.51 ±2.95 % of control, p <0.0001)
was greater than that seen with 10 nM PTX treatment and at the same
level achieved with DTX treatment.
Fig. 1. Inhibition of PC-3 prostate cancer cell proliferation by RE. PC-3 cells were treated with 5, 10, 25, 50, 75, 100, or 150
μ
g/mL of rosemary extract (RE) (A, B,
C), 5, 10 nM paclitaxel (PTX) (C), or 5, 10 nM docetaxel (DTX) (C) for 72 h, followed by xing and staining with 0.5 % crystal violet. The stain was solubilized, and
absorbance was read at 570 nm. Data are expressed as percent of control, untreated cells. Data are the mean ±SEM of 6 independent experiments.
**p <0.01, ****p <0.0001.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
4
3.2. Inhibition of PC-3 prostate cancer cell survival by rosemary extract
The ability of cancer cells to survive and form colonies was also
assessed through a clonogenic survival assay. Exposure of PC-3 cells to
0.5, 1, 2.5, 5, or 10
μ
g/mL of RE resulted in a concentration-dependent
inhibition of survival (Fig. 2A) with a signicant inhibition
(80.20 ±4.60 % of control, p <0.01) seen with 0.5
μ
g/mL RE. The
greatest inhibition (15.27 ±3.80 % of control, p <0.0001) of cell sur-
vival was seen at 10
μ
g/mL RE (Fig. 2A and 2C). The data from Fig. 2A
were plotted on a log scale and the calculated RE concentration for the
half maximal inhibition (IC
50
) of cell survival was 2.43
μ
g/mL (Fig. 2B).
Exposure of the cells to 0.5 nM (64.12 ±6.94 % of control, p <0.05) and
5 nM paclitaxel (23.81 ±11.92 % of control, p <0.0001) both resulted
in a signicant inhibition of cell survival (Fig. 2C). In addition, treat-
ment with both 0.5 nM (59.18 ±9.45 % of control p <0.01) and 5 nM
docetaxel (8.50 ±8.50 % of control p <0.0001) resulted in a signicant
inhibition of cell survival.
3.3. Inhibition of Akt signaling in PC-3 prostate cancer cells by rosemary
extract
We also examined the effects of RE treatment on Akt and measured
the levels of total Akt and Akt phosphorylation on the serine 473 res-
idue, an established indicator of Akt activity [60]. Treatment of PC-3
prostate cancer cells with 50
μ
g/mL RE for 24 and 48 h both
signicantly reduced Akt phosphorylation/activation (60.65 ±11.84 %
of control, p <0.001) and (36.46 ±4.79 % of control, p <0.0001),
respectively (Fig. 3A and B). The total Akt levels were signicantly
reduced by 48 h (69.93 ±4.66 % of control, p <0.01) but not 24 h
(87.52 ±9.50 % of control, p >0.05) RE treatment.
3.4. Inhibition of mTOR signaling in PC-3 prostate cancer cells by
rosemary extract
Next, we examined the effects of RE on mTOR activation by utilizing
an antibody that recognizes phosphorylation of the serine 2448 residue,
an established marker of mTOR activation [61]. Treatment of PC-3 cells
with RE for 24 (49.41 ±6.60 % of control, p <0.0001) or 48 h
(38.82 ±6.69 % of control, p <0.0001) resulted in a signicant
decrease in mTOR phosphorylation (Fig. 4A and B). Treatment with RE
for 24 and 48 h also showed a signicant decrease of total mTOR levels
(71.74 ±10.38 % of control, p <0.01) and (60.55 ±9.02 % of control,
p <0.001), respectively.
3.5. Increased apoptosis of PC-3 prostate cancer cells by rosemary extarct
The effect of RE on cell apoptosis was examined by measuring the
levels of cleaved PARP, an established indicator of apoptosis [37].
Exposing PC-3 prostate cancer cells to rosemary extract (50
μ
g/mL) for
24 h resulted in a signicant increase in cleaved PARP (177.9 ±14.50 %
Fig. 2. Inhibition of PC-3 prostate cancer cell survival by rosemary extract. PC-3 cells were seeded (1000 cells/well) in six-well plates and exposed to 0.5, 1, 2.5, 5, or
10
μ
g/mL of rosemary extract (RE) (A, B, C), 0.5, 5 nM paclitaxel (PTX) (C), or 0.5, 5 nM docetaxel (DTX) (C) for 7 days followed by xing and staining with 0.05 %
methylene blue. Colonies of more than 50 cells were counted. Data are expressed as percent of control, untreated cells. Data are the mean ±SEM of 6 independent
experiments. *p <0.05, **p <0.01, ****p <0.0001.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
5
of control, p <0.01) relative to the control, indicating enhanced
apoptosis (Fig. 5A and B).
We routinely examined microscopically the morphology of cells
before and after treatments. Fig. 6 shows a representative image of RE-
and DTX-treated PC-3 cells compared to the control untreated cells. No
changes in cell morphology were observed with any of the treatments. It
is important to note that the same number of cells were seeded in all
wells (six-well plates were used). As it can be seen from Fig. 6, treatment
with 50
μ
g/mL of RE for 24 or 48 h resulted in a substantially reduced
cell density relative to the control untreated group. Treatment with DTX
for 24 or 48 h showed a reduction in cell density that was not as great as
the reduction seen with RE.
3.6. Inhibition of PC-3 prostate cancer cell migration by rosemary extract
The wound healing assay was used to assess the ability of prostate
cancer cells to migrate. The cells were seeded at a density of 2.5 ×10
5
cells/mL into a 6-well plate and grown until the cells reached 90100 %
conuency. PC-3 cells were exposed to 1
μ
g/mL of mitomycin-C (MMC)
for 1 h to inhibit cell proliferation. After the MMC was removed a wound
was established by drawing a vertical line in the centre of each well
using a 100
μ
L pipette tip. The cells were then treated without (control)
or with either 50
μ
g/mL RE or 10 nM docetaxel for 40 h. Treatment with
rosemary extract was shown to signicantly inhibit wound closure
(56.14 ±3.48 % of control, p <0.0001) indicating properties against
Fig. 3. Inhibition of Akt signaling in PC-3 prostate cancer cells by rosemary extract. Cell lysates were prepared from PC-3 prostate cancer cells treated with 50
μ
g/mL
rosemary extract (RE) for either 24 or 48 h. Cell lysates (20
μ
g) were immunoblotted using specic antibodies against phosphorylated/activated Akt, total Akt, or
β-actin. A representative immunoblot is shown (A). The densitometry of the bands were corrected to β-actin levels and expressed in arbitrary densitometry units as
percent of control (B). The data are the mean ±SEM of 4 independent experiments. **p <0.01, ***p <0.001, ****p <0.0001.
Fig. 4. Inhibition of mTOR signaling in PC-3 prostate cancer cells by rosemary extract. Cell lysates were prepared from PC-3 cells treated with 50
μ
g/mL rosemary
extract (RE) for either 24 or 48 h. Cell lysates (20
μ
g) were immunoblotted using specic antibodies against phosphorylated/activated mTOR, total mTOR, or β-actin.
A representative immunoblot is shown (A). The densitometry of the bands were corrected to β-actin levels and expressed in arbitrary densitometry units as percent of
control (B). The data are the mean ±SEM of 5 independent experiments. **p <0.01, ***p <0.001 ****p <0.0001.
Fig. 5. Effect of rosemary extract on
PARP signaling in PC-3 prostate cancer
cells. Cell lysates were prepared from
PC-3 cells treated with 50
μ
g/mL rose-
mary extract (RE) for 24 h. Cell lysates
(20
μ
g) were immunoblotted using spe-
cic antibodies against cleaved PARP or
β-actin. A representative immunoblot is
shown (A). The densitometry of the
bands were corrected to β-actin levels
and expressed in arbitrary densitometry
units as percent of control (B). The data
are the mean ±SEM of 3 independent
experiments. *p <0.05.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
6
cell migration (Fig. 7B). A signicant inhibition of cell migration was
also seen when treating the cells with 10 nM docetaxel (70.92 ±2.35 %
of control, p <0.001), (Fig. 7A and B).
3.7. Inhibition of 22RV1 prostate cancer cell proliferation and survival by
rosemary extract
We also examined the effects of RE on the androgen receptor positive
22RV1 prostate cancer cells. A signicant inhibition of cell proliferation
was seen with 25
μ
g/mL RE (86.20 ±4.53 % of control, p <0.01) while
the highest level of inhibition was seen with 150
μ
g/mL RE
(49.80 ±2.289 % of control, p <0.0001), (Fig. 8A). The IC
50
value of RE
for cell proliferation, calculated by graphing the data from Fig. 8A on a
log scale, was 43.41
μ
g/mL (Fig. 8B). The effects of RE on 22RV1
prostate cancer cell survival was investigated by treating the cells with
2.5, 5, 10, 15, and 20
μ
g/mL RE for 7 days (Fig. 8C). A dose-dependent
inhibition of cell survival was seen. The calculated RE concentration for
the half maximal inhibition (IC
50
) of cell survival calculated using the
data from Fig. 8C and graphing it on a log scale was 4.17
μ
g/mL
(Fig. 8D).
3.8. Effect of rosemary extract on PNT1A normal prostate epithelial cell
proliferation
PNT1A prostate epithelial cells represent normal healthy prostate
epithelium. Treatment of PNT1A cells with 5, 10, 25, 50, 75, 100, or
150
μ
g/mL RE for 72 h did not result in any signicant changes in cell
proliferation (p >0.05) (Fig. 9).
4. Discussion
The current treatment strategies for prostate cancer include surgery,
radiotherapy, and chemotherapy [62]. Patients with localized prostate
cancer are most often treated with radical prostatectomy or radical
radiotherapy, however advanced and metastatic prostate cancer is
treated with hormonal therapy [62]. Common hormonal therapies often
use androgen-receptor inhibitors or LHRH agonists (such as leuprolide,
goserelin, buserelin, or nafarelin) that initially increase testosterone
production, but with prolonged exposure downregulate the LHRH re-
ceptor and inhibit testosterone production [5]. LHRH antagonists (such
as cetrorelix, abarelix, or orgalutran) directly inhibit LHRH, which de-
creases testosterone production [5]. Surgical castration can also
decrease testosterone levels by removing the source of production. For
patients that do not respond to androgen therapies, cytotoxic chemo-
therapeutic agents, such as etoposide, doxorubicin, paclitaxel, and
docetaxel are used [5] but resistance often develops, indicating a need
for novel therapeutics to be used alone or in combination with existing
drugs to treat prostate cancer and improve patient outcome.
Plant extracts have been used traditionally for medicinal purposes
and more than half of all available chemotherapy agents used in cancer
treatment are derived from plants with paclitaxel and docetaxel repre-
senting two such chemotherapeutics [3941]. Finding chemicals in
plants with anti-cancer potential is the focus of many research labs,
including ours. In recent years a few studies provided evidence of
anticancer properties of rosemary extract [4248]. In the present study
we found a dose-dependent inhibition of PC-3 androgen-independent
and 22RV1 androgen-dependent prostate cancer cell proliferation with
rosemary extract treatment (Figs. 1,8, and 10). Similar to our ndings, in
other studies, treatment with RE dose-dependently inhibited the
viability of 22RV1 and LNCaP prostate cancer cells [51]. In addition, a
Fig. 6. Effect of rosemary extract on PC-3 prostate cancer cell morphology. Cells were seeded (200,000 cells/well) and after 24 h were treated without (control) or
with RE (50
μ
g/mL) or DTX (10 nM) for 24 or 48 h. Photographs were taken immediately after treatment using an EVOS XL Core Cell Imaging System by Life
Technologies (10×magnication).
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
7
dose-dependent decrease in proliferation and viability of PC-3, DU145,
and LNCaP prostate cancer cells was seen by RE treatment [49,52].
Based on our data, we calculated the RE concentration for half maximal
inhibition (IC
50
) of cell proliferation and comparing these IC
50
values it
appears that the androgen-independent PC-3 cells (19.72
μ
g/mL) are
more sensitive to RE treatment than the androgen-dependent 22RV1
cells (43.41
μ
g/mL). Importantly, treatment of the PNT1A normal
prostate epithelial cells with RE did not signicantly affect their rate of
proliferation (Fig. 9). Similarly to our ndings, Petiwala et al. [51], as
mentioned above, found a signicant inhibition of 22RV1 and LNCaP
prostate cancer cell viability but not an effect on normal prostate
epithelial cells derived from two different patients undergoing radical
prostatectomy [51]. These data indicate that RE is able to discriminate
and preferentially target prostate cancer cells while sparing normal
healthy prostate epithelial cells.
Apart from cell proliferation, treatment with RE resulted in a dose-
dependent inhibition of cell survival with IC
50
values of 2.43
μ
g/mL
and 4.17
μ
g/mL for PC-3 and 22RV1 cells, respectively (Figs. 2,8, and
10). These data indicate a higher sensitivity of the androgen-
independent PC-3 cells than the androgen-dependent 22RV1 cells to
RE treatment and are in agreement with our proliferation data.
The inhibition of prostate cancer cell (PC-3) proliferation and sur-
vival seen with RE treatment was robust and comparable to the inhibi-
tion seen with docetaxel (DTX) and paclitaxel (PTX) both routinely used
in the treatment of prostate cancer [57].
Cleaved PARP is an established indicator of apoptosis [36,38], and
our data showed an increase in cleaved PARP levels in PC-3 cells treated
with RE indicating an effect of RE to induce apoptosis (Fig. 5). Similar to
our ndings, treatment of 22RV1 and LNCaP prostate cancer cell with
RE resulted in a signicant increase in apoptosis [51]. In addition,
treatment of PC-3 cells with the RE polyphenol carnosic acid [63] and
PC-3 and DU145 cells with the RE polyphenol rosmarinic acid [64]
induced apoptosis as indicated by the increased levels of cleaved PARP.
Furthermore, RE treated PC-3 cells showed a signicant inhibition of
cell migration (56.14 ±3.48 % of control, p <0.0001) that was com-
parable to the response seen with docetaxel (70.92 ±2.35 % of control,
p <0.001) (Fig. 7). No other studies examining the anti-migratory or
anti-metastatic effects of RE in prostate cancer cells currently exist. We
previously found a signicant inhibition of MDA-MB-231 breast cancer
cell migration by RE treatment [65]. In a study by P´
erez-S´
anchez et al.,
Fig. 7. Inhibition of PC-3 prostate cancer cell migration by rosemary extract. PC-3 cells were exposed to 1
μ
g/mL of mitomycin-C for one hour, followed by a wound
induction and treatment without or with 50
μ
g/mL rosemary extract (RE) or 10 nM docetaxel (DTX) for 40 hs. Representative images are shown immediately after
wound induction (0 h) and after 40 h of treatment (A). Wound closure was calculated as indicated in the methods and expressed as a percent of control untreated cells
(B). The data are the mean ±SEM of 3 independent experiments. ***p <0.001, ****p <0.0001.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
8
treatment with RE inhibited the migration of HGUE-C-1, HT-29, and
SW480 human colon cells [66]. In prostate cancer metastasis, cells
migrate away from the primary tumor to other tissues. Once in other
tissues, their invasiveness depends on their clonogenic survival. Treat-
ment with RE signicantly reduced both, the cell migration, as assessed
by the wound-healing assay, and the clonogenic survival. Our data
indicate that RE has the potential to reduce prostate cancer cell prolif-
eration and tumor growth as well as reduce their migration and invasion
capabilities.
The expression and activation of the serine/threonine kinase Akt is
often elevated in human prostate cancer. Approximately 40 % of early
cases and 70100 % of advanced cases of prostate cancer have aberra-
tions in the PI3K/Akt signaling [2629]. This is often due to mutations
on PI3K [4], Akt [67] as well as the loss of the tumor suppressor gene
PTEN [68], all of which lead to overactivation of Akt resulting in
enhanced proliferation and survival. Our study showed a signicant
inhibition of Akt phosphorylation/activation with RE treatment (Fig. 3).
A search of the literature revealed that no other studied have examined
the effects of RE treatment on Akt in prostate cancer cells. It should be
noted that a signicant inhibition of Akt phosphorylation/activation
was seen in PC-3 prostate cancer cells treated with the RE polyphenol
carnosic acid [63] and carnosol [69]. In previous studies by our lab, we
found a signicant inhibition of Akt phosphorylation/ activation by RE
treatment of A549 lung cancer [53] and MDA-MB-231 breast cancer
cells [65].
It has been reported that the levels of total and phosphorylated
mTOR are twice as great in prostate cancer tissue when compared to
normal prostate epithelium [34]. mTOR is a downstream target of Akt
and its activation leads to increased protein synthesis and cell prolifer-
ation [70]. Our study is the rst to show a signicant inhibition of mTOR
in prostate cancer cells with RE treatment (Fig. 4). Similar to our study, a
signicant inhibition of mTOR phosphorylation/activation was seen in
PC-3 prostate cancer cells treated with the RE polyphenol carnosol [69].
Fig. 8. Inhibition of 22RV1 prostate cancer cell proliferation and survival by RE. 22RV1 cells were treated with 5, 10, 25, 50, 75, 100, or 150
μ
g/mL of rosemary
extract (RE) (A,B) for 72 h followed by xing and staining with 0.5 % crystal violet. The stain was solubilized, and absorbance was read at 570 nm. 22RV1 cells were
treated with 2.5, 5, 10, 15, or 20
μ
g/mL of rosemary extract (RE) (C,D) for 7 days followed by xing and staining with 0.05 % methylene blue. Colonies of more than
50 cells were counted. Data are expressed as percent of control, untreated cells. Data are the mean ±SEM of 5 independent experiments. *p <0.05,
**p <0.01, ****p <0.0001.
Fig. 9. Effect of RE on PNT1A normal prostate epithelial cell proliferation.
PNT1A cells were treated with 5, 10, 25, 50, 75, 100, or 150
μ
g/mL of rosemary
extract (RE) for 72 h, followed by xing and staining with 0.5 % crystal violet.
The stain was solubilized, and absorbance was read at 570 nm. Data are
expressed as percent of control, untreated cells. Data are the mean ±SEM of 3
independent experiments.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
9
The mechanisms involved in the RE-induced inhibition of Akt and mTOR
(Fig. 10) is not known. It is possible that components in RE act as allo-
steric inhibitors of Akt and/or mTOR or they act on a step upstream of
Akt. Another possibility is that components in RE increase the activity of
Akt and/or mTOR specic phosphatases [71,72] resulting in their
reduced phosphorylation/activation. It is also possible that the inhibi-
tion of mTOR is due to Akt inhibition. Future studies should examine
these possibilities and elucidate the mechanisms involved in these
inhibitory effects of RE.
PC-3 cells, contrary to 22RV1, contain PTEN mutations leading to
enhanced Akt activation [73]. It is possible that the increased activation
of the PI3K/Akt/mTOR cascade in PC-3 cells may explain their higher
sensitivity to RE treatment compared to 22RV1 cells. The notion that RE
targets prostate cancer cells characterized by increased Akt-mTOR
signaling should be explored in future studies to further dene REs
potential as a therapeutic agent.
Apart from a reduction in phosphorylated/activated Akt and mTOR
levels, our study shows a reduction in the total levels of these proteins
with RE treatment (Figs. 3 and 4), which may be due to the inhibition of
gene transcription, inhibition of protein synthesis, upregulation of
Fig. 10. Rosemary extract inhibits proliferation and survival of PC-3 androgen-insensitive and 22RV1 androgen-sensitive prostate cancer cells. RE had no effect on
the proliferation of PNT1A healthy prostate epithelial cells (A). Rosemary extract inhibits Akt and mTOR signaling and enhances PARP cleavage in PC-3 prostate
cancer cells (B).
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
10
protein degradation or an effect on protein stability. Another study
showed that resveratrol, a polyphenol found in grapes, inhibited total
and phosphorylated levels of Akt in both androgen-dependent and in-
dependent prostate cancer cell lines [74]. Previous studies by our group
performed in A549 lung [53] and MDA-MB-231 breast [65] cancer cells
also observed a signicant reduction of both total Akt and mTOR levels
by RE treatment.
Activation of the PI3K/Akt/mTOR pathway in prostate cancer is
associated with disease progression, resistance to androgen deprivation
therapy, and poor prognosis [4,64] and in recent years many efforts
have been focused on the development of inhibitors that target this
pathway [25,75]. ATP-competitive and allosteric inhibitors of Akt have
been developed and used against prostate cancer [25,75,76]. In a phase
II clinical study the combination of the Akt inhibitor ipatasertib, with
the anti-androgen medication abiraterone acetate, in patients with
metastatic castration-resistant prostate cancer improved
progression-free survival [77]. In another study, it was found that the
dual PI3K/Akt and mTOR inhibitor, NVP-BEZ235 (40 mg/kg), when
used as a combined therapy with paclitaxel (4 mg/kg), resulted in a
greater inhibition of tumor growth in castrated mice xenografted with
C42AT6 prostate cancer cells when compared to monotherapy treat-
ments [78]. Treatment with NVP-BEZ235 was shown to overcome
docetaxel resistance in human castration resistant prostate cancer [78].
The inhibition of Akt and mTOR by RE treatment is strong and future
studies should explore if RE and its components have effects similar to
the above mentioned Akt and mTOR inhibitors.
A few in vivo animal studies exist examining the effects of RE in
prostate cancer. Treatment of athymic nude mice transplanted with
22RV1 human prostate cancer cells with RE (100 mg/kg/day) resulted
in a 46 % reduction in tumor size compared to the control untreated
mice [51]. Treatment resulted in a signicant reduction in androgen
receptor and prostate specic antigen (PSA) levels [51]. Similarly, mice
xenografted with 22RV1 human prostate cancer cells treated with the RE
polyphenol CA (100 mg/kg/day), showed a 53 % reduction in tumor
growth when compared to the control untreated mice [79]. Addition-
ally, CA decreased androgen receptor levels [79]. Although the studies
examining the effects of rosemary extract and rosemary extract poly-
phenols in vivo are limited, the data/evidence suggests they may be
effective in inhibiting prostate cancer tumor growth and warrants
further investigation.
We recognize that the concentration of bioactive, medicinal in-
gredients in plant extracts, and in this case rosemary extract, may be
inuenced by environmental factors (such as soil quality, water, and sun
exposure) and by the extraction method used. We used high-
performance liquid chromatography (HPLC) and measured the levels
of carnosic acid and rosmarinic acid. Our rosemary extract was found to
contain 2.12 ±0.22 % carnosic acid [80] and 13.39 ±0.23 % rosmarinic
acid [81]. Based on the molecular weight of carnosic acid
(332.42 g/mol) and rosmarinic acid (360.31 g/mol) the cells treated
with 50
μ
g/mL RE would have been exposed to 3
μ
M carnosic acid and
20
μ
M rosmarinic acid. Apart from the aforementioned two, RE contains
many other polyphenols and constituents. The exact bioactive constit-
uents responsible for the anti-cancer properties of RE are not known. We
plan to explore the effects of carnosic acid and rosmarinic acid in future
studies.
5. Conclusions
In the present study we found a signicant inhibition of proliferation
and survival of the PC-3 androgen-insensitive and the 22RV1 androgen-
sensitive prostate cancer cells by RE while the PNT1A normal prostate
epithelial cells were not affected (Fig. 10). Furthermore, treatment with
RE induced apoptosis and reduced migration of PC-3 prostate cancer
cells.
Importantly, RE signicantly reduced the phosphorylation/activa-
tion levels of Akt and mTOR (Fig. 10). Overactivation of the PI3K/Akt/
mTOR pathway in prostate cancer is associated with increased prolif-
eration and survival, resistance to treatment, and overall poor prognosis.
Our data provide strong evidence that treatment with RE targets this
pathway, but it is not known whether this is true in vivo. Future studies
using animal models xenografted with prostate cancer cells should be
performed to investigate this possibility as well as further examine the
anti-cancer properties of RE. Future studies should also examine the
exact polyphenolic constituent(s) of rosemary extract that contribute to
its anti-cancer effects.
Author contributions
E.T conceived and designed the experiments and contributed to data
interpretation and manuscript writing. A.J performed the majority of
the experiments, analyzed the data, prepared the gures and contrib-
uted to manuscript writing. D.T performed the experiments using 22RV1
and PNT1A cells. All authors have read and agreed to the published
version of the manuscript.
Funding
This work was funded by a research grant to E.T from the Prostate
Cancer Fight Foundation, Ontario, Canada.
Declaration of Competing Interest
The authors report no declarations of interest.
Appendix A. Supplementary data
Supplementary material related to this article can be found, in the
online version, at doi:https://doi.org/10.1016/j.biopha.2020.110717.
References
[1] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global cancer
statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36
cancers in 185 countries, CA Cancer J. Clin. 68 (2018) 394424, https://doi.org/
10.3322/caac.21492.
[2] M. Nieto, S. Finn, M. Loda, W.C. Hahn, Prostate cancer: Re-focusing on androgen
receptor signaling, Int. J. Boichem. Cell Biol. 39 (2007) 15621568 (accessed
January 17, 2019), www.uicc.org/tnm.
[3] S. Koochekpour, Androgen receptor signaling and mutations in prostate cancer,
Nat. Publ. Gr. 12 (2010) 639657, https://doi.org/10.1038/aja.2010.89.
[4] M. Crumbaker, L. Khoja, A.M. Joshua, AR Signaling and the PI3K Pathway in
Prostate Cancer, Cancers (Basel). 9 (2017), https://doi.org/10.3390/
cancers9040034.
[5] S.R. Denmeade, J.T. Isaacs, TimelineA history of prostate cancer treatment, Nat.
Rev. Cancer (2002), https://doi.org/10.1038/nrc801.
[6] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144
(2011) 646647, https://doi.org/10.1016/j.cell.2011.02.013.
[7] N.N. Pavlova, C.B. Thompson, The emerging hallmarks of cancer metabolism, Cell
Metab. 23 (2016) 2747, https://doi.org/10.1016/j.cmet.2015.12.006.
[8] M. Martini, M. Chiara, D. Santis, L. Braccini, F. Gulluni, E. Hirsch, PI3K/AKT
signaling pathway and cancer: an updated review, Ann. Med. 46 (2014) 372383,
https://doi.org/10.3109/07853890.2014.912836.
[9] Q. Wang, X. Chen, N. Hay, Akt as a target for cancer therapy: more is not always
better (lessons from studies in mice), Br. J. Cancer (2017), https://doi.org/
10.1038/bjc.2017.153.
[10] K. Nakatani, D.A. Thompson, A. Barthel, H. Sakaue, W. Liu, R.J. Weigel, R.A. Roth,
Up-regulation of Akt3 in estrogen receptor-decient breast cancers and androgen-
independent prostate cancer lines, J. Biol. Chem. (1999), https://doi.org/10.1074/
jbc.274.31.21528.
[11] D. Mossmann, S. Park, M.N. Hall, mTOR signalling and cellular metabolism are
mutual determinants in cancer, Nat. Rev. Cancer (2018), https://doi.org/10.1038/
s41568-018-0074-8.
[12] A.S. Dhillon, S. Hagan, O. Rath, W. Kolch, MAP kinase signalling pathways in
cancer, Oncogene (2007) 32793290, https://doi.org/10.1038/sj.onc.1210421.
[13] G.A. Hobbs, C.J. Der, K.L. Rossman, RAS isoforms and mutations in cancer at a
glance, J. Cell. Sci. 129 (2016) 12891292, https://doi.org/10.1242/jcs.182873.
[14] K. Haglund, T.E. Rusten, H. Stenmark, Aberrant receptor signaling and trafcking
as mechanisms in oncogenesis, Crit. Rev. Oncog. (2007), https://doi.org/10.1615/
CritRevOncog.v13.i1.20.
[15] L.C. Cantley, R.J. Shaw, Ras, PI(3)K and mTOR signalling controls tumour cell
growth, Nature 441 (2006) 424430, https://doi.org/10.1038/nature04869.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
11
[16] F. Janku, T.A. Yap, F. Meric-Bernstam, Targeting the PI3K pathway in cancer: Are
we making headway? Nat. Rev. Clin. Oncol. (2018) 273291, https://doi.org/
10.1038/nrclinonc.2018.28.
[17] P. Liu, H. Cheng, T.M. Roberts, J.J. Zhao, Targeting the phosphoinositide 3-kinase
pathway in cancer, Nat. Rev. Drug Discov. 8 (2009) 627644, https://doi.org/
10.1038/nrd2926.
[18] R.S. Liao, S. Ma, L. Miao, R. Li, Y. Yin, G.V. Raj, Androgen receptor-mediated non-
genomic regulation of prostate cancer cell proliferation, Transl. Androl. Urol. 2
(2013) 187196, https://doi.org/10.3978/j.issn.2223-4683.2013.09.07.
[19] X.E. Guo, B. Ngo, A.S. Modrek, W.-H. Lee, Targeting tumor suppressor networks for
cancer therapeutics, Curr. Drug Targets 15 (2014) 216 (accessed January 30,
2019), http://www.ncbi.nlm.nih.gov/pubmed/24387338.
[20] R. Sever, J.S. Brugge, Signal transduction in cancer, Cold Spring Harb. Perspect.
Med. 5 (2015), https://doi.org/10.1101/cshperspect.a006098.
[21] Y.E. Whang, X. Wu, H. Suzuki, R.E. Reiter, C. Tran, R.L. Vessella, J.W. Said, W.
B. Isaacs, C.L. Sawyers, Inactivation of the tumor suppressor PTEN/MMAC1 in
advanced human prostate cancer through loss of expression, Proc. Natl. Acad. Sci.
(1998), https://doi.org/10.1073/pnas.95.9.5246.
[22] C. Cordon-Cardo, A. Koff, M. Drobnjak, P. Capodieci, I. Osman, S.S. Millard, P.
B. Gaudin, M. Fazzari, Z.F. Zhang, J. Massague, H.I. Scher, Distinct altered patterns
of p27(KIP1) gene expression in benign prostatic hyperplasia and prostatic
carcinoma, J. Natl. Cancer Inst. (1998), https://doi.org/10.1093/jnci/90.17.1284.
[23] I. Osman, M. Drobnjak, M. Fazzari, J. Ferrara, H.I. Scher, C. Cordon-Cardo,
Inactivation of the p53 pathway in prostate cancer: impact on tumor progression,
Clin. Cancer Res. (1999), https://doi.org/10.1158/1078-0432.ccr-05-0397.
[24] K.H. Khan, T.A. Yap, L. Yan, D. Cunningham, Targeting the PI3K-AKT-mTOR
signaling network in cancer, Chin. J. Cancer 32 (2013) 253265, https://doi.org/
10.5732/cjc.013.10057.
[25] G.M. Nitulescu, M. Van De Venter, G. Nitulescu, A. Ungurianu, P. Juzenas, Q. Peng,
O.T. Olaru, D. Gr˘
adinaru, A. Tsatsakis, D. Tsoukalas, D.A. Spandidos, D. Margina,
The Akt pathway in oncology therapy and beyond (Review), Int. J. Oncol. (2018),
https://doi.org/10.3892/ijo.2018.4597.
[26] M. Sun, G. Wang, J.E. Paciga, R.I. Feldman, Z.Q. Yuan, X.L. Ma, S.A. Shelley,
R. Jove, P.N. Tsichlis, S.V. Nicosia, J.Q. Cheng, AKT1/PKBalpha kinase is
frequently elevated in human cancers and its constitutive activation is required for
oncogenic transformation in NIH3T3 cells, Am. J. Pathol. 159 (2001) 431437,
https://doi.org/10.1016/S0002-9440(10)61714-2.
[27] J.M. Drake, N.A. Graham, J.K. Lee, T. Stoyanova, C.M. Faltermeier, S. Sud, B. Titz,
J. Huang, K.J. Pienta, T.G. Graeber, O.N. Witte, Metastatic castration-resistant
prostate cancer reveals intrapatient similarity and interpatient heterogeneity of
therapeutic kinase targets, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) E4762E4769,
https://doi.org/10.1073/pnas.1319948110.
[28] B.S. Taylor, N. Schultz, H. Hieronymus, A. Gopalan, Y. Xiao, B.S. Carver, V.
K. Arora, P. Kaushik, E. Cerami, B. Reva, Y. Antipin, N. Mitsiades, T. Landers,
I. Dolgalev, J.E. Major, M. Wilson, N.D. Socci, A.E. Lash, A. Heguy, J.A. Eastham,
H.I. Scher, V.E. Reuter, P.T. Scardino, C. Sander, C.L. Sawyers, W.L. Gerald,
Integrative genomic proling of human prostate cancer, Cancer Cell 18 (2010)
1122, https://doi.org/10.1016/j.ccr.2010.05.026.
[29] B.S. Carver, C. Chapinski, J. Wongvipat, H. Hieronymus, Y. Chen,
S. Chandarlapaty, V.K. Arora, C. Le, J. Koutcher, H. Scher, P.T. Scardino, N. Rosen,
C.L. Sawyers, Reciprocal feedback regulation of PI3K and androgen receptor
signaling in PTEN-Decient prostate cancer, Cancer Cell 19 (2011) 575586,
https://doi.org/10.1016/j.ccr.2011.04.008.
[30] F. Claudio, Targeting the PI3K/AKT/mTOR pathway in prostate Cancer
development and progression: insight to therapy, Clin. Cancer Drugs 3 (2016),
https://doi.org/10.2174/2212697x0301160328201324.
[31] B.T. Hennessy, D.L. Smith, P.T. Ram, Y. Lu, G.B. Mills, Exploiting the PI3K/AKT
pathway for cancer drug discovery, Nat. Rev. Drug Discov. 4 (2005) 9881004,
https://doi.org/10.1038/nrd1902.
[32] D.A. Guertin, D.M. Sabatini, Dening the role of mTOR in cancer, Cancer Cell 12
(2007) 922, https://doi.org/10.1016/j.ccr.2007.05.008.
[33] H. P´
opulo, J.M. Lopes, P. Soares, The mTOR signalling pathway in human cancer,
Int. J. Mol. Sci. 13 (2012) 18861918, https://doi.org/10.3390/ijms13021886.
[34] C.L. Kremer, R.R. Klein, J. Mendelson, W. Browne, L.K. Samadzedeh, K. Vanpatten,
L. Highstrom, G.A. Pestano, R.B. Nagle, Expression of mTOR signaling pathway
markers in prostate cancer progression, Prostate 66 (2006) 12031212, https://
doi.org/10.1002/pros.20410.
[35] H. Zhou, Y. Luo, S. Huang, Updates of mTOR inhibitors, Anticancer Agents Med.
Chem. 10 (2012) 571581, https://doi.org/10.2174/187152010793498663.
[36] G.V. Chaitanya, A.J. Steven, P. Prakash Babu, PARP-1 cleavage fragments:
signatures of cell-death proteases in neurodegeneration, Cell Commun. Signal 8
(2010) 31, https://doi.org/10.1186/1478-811X-8-31.
[37] A.H. Boulares, A.G. Yakovlev, V. Ivanova, B. a Stoica, G. Wang, S. Iyer,
M. Smulson, M.J.B. Chem, Role of poly (ADP-ribose) polymerase (PARP) cleavage
in apoptosis, J. Biol. Chem. 274 (1999) 2293222940, https://doi.org/10.1074/
jbc.274.33.22932.
[38] M.J. Schiewer, K.E. Knudsen, Transcriptional roles of PARP1 in cancer, Mol.
Cancer Res. 12 (2014) 10691080, https://doi.org/10.1158/1541-7786.MCR-13-
0672.
[39] D.J. Newman, G.M. Cragg, Natural products as sources of new drugs over the last
25 years, J. Nat. Prod. 70 (2007) 461477, https://doi.org/10.1021/np068054v.
[40] H.K. Kim, E.G. Wilson, Y.H. Choi, R. Verpoorte, Metabolomics: A tool for
anticancer lead-nding from natural products, Planta Med. 76 (2010) 10941102,
https://doi.org/10.1055/s-0030-1249898.
[41] A.L. Demain, P. Vaishnav, Natural products for cancer chemotherapy, Microb.
Biotechnol. (2011) 687699, https://doi.org/10.1111/j.1751-7915.2010.00221.x.
[42] A. Vallverdú-Queralt, J. Regueiro, M. Martínez-Hu´
elamo, J.F. Rinaldi Alvarenga, L.
N. Leal, R.M. Lamuela-Raventos, A comprehensive study on the phenolic prole of
widely used culinary herbs and spices: rosemary, thyme, oregano, cinnamon,
cumin and bay, Food Chem. 154 (2014) 299307, https://doi.org/10.1016/j.
foodchem.2013.12.106.
[43] J.M. Andrade, C. Faustino, C. Garcia, D. Ladeiras, C.P. Reis, P. Rijo, Rosmarinus
ofcinalis L.: an update review of its phytochemistry and biological activity, Futur.
Sci. OA. 4 (2018) FSO283, https://doi.org/10.4155/fsoa-2017-0124.
[44] P.F. Leal, M.E.M. Braga, D.N. Sato, J.E. Carvalho, M.O.M. Marques, M.A.
A. Meireles, Functional properties of spice extracts obtained via supercritical uid
extraction, J. Agric. Food Chem. 51 (2003) 25202525, https://doi.org/10.1021/
jf0260693.
[45] J.J. Johnson, Carnosol: A promising anti-cancer and anti-inammatory agent,
Cancer Lett. 305 (2011) 17, https://doi.org/10.1016/j.canlet.2011.02.005.
[46] M. Gonz´
alez-Vallinas, G. Reglero, A. Ramírez De Molina, Rosemary (Rosmarinus
ofcinalis L.) extract as a potential complementary agent in anticancer therapy,
Nutr. Cancer 67 (2015) 12211229, https://doi.org/10.1080/
01635581.2015.1082110.
[47] J. Moore, M. Yousef, E. Tsiani, Anticancer effects of Rosemary (Rosmarinus
ofcinalis L.) extract and rosemary extract polyphenols, Nutrients 8 (2016),
https://doi.org/10.3390/nu8110731.
[48] G. Nieto, G. Ros, J. Castillo, Medicines Antioxidant and Antimicrobial Properties of
Rosemary (Rosmarinus ofcinalis, L.): A Review, Medicines 5 (2018), https://doi.
org/10.3390/medicines5030098.
[49] O. Yesil-Celiktas, C. Sevimli, E. Bedir, F. Vardar-Sukan, Inhibitory effects of
rosemary extracts, carnosic acid and rosmarinic acid on the growth of various
human Cancer cell lines, Plant Foods Hum. Nutr. 65 (2010) 158163, https://doi.
org/10.1007/s11130-010-0166-4.
[50] S.M. Petiwala, A.G. Puthenveetil, J.J. Johnson, M. Heinrich, H. Yang, Polyphenols
from the Mediterranean herb rosemary (Rosmarinus ofcinalis) for prostate
cancer, Front. Pharmacol. 4 (2013), https://doi.org/10.3389/fphar.2013.00029.
[51] S.M. Petiwala, S. Berhe, G. Li, A.G. Puthenveetil, O. Rahman, L. Nonn, J.
J. Johnson, Rosemary (Rosmarinus ofcinalis) extract modulates CHOP/GADD153
to promote androgen receptor degradation and decreases xenograft tumor growth,
PLoS One 9 (2014), e89772, https://doi.org/10.1371/journal.pone.0089772.
[52] M. Bourhia, F.E. Laasri, H. Aourik, A. Boukhris, R. Ullah, A. Bari, S.S. Ali, M. El
Mzibri, L. Benbacer, S. Gmouh, Antioxidant and antiproliferative activities of
bioactive compounds contained in Rosmarinus ofcinalis used in the
mediterranean diet, evidence-based complement, Altern. Med. 2019 (2019),
https://doi.org/10.1155/2019/7623830.
[53] J. Moore, M. Megaly, A.J. Macneil, P. Klentrou, E. Tsiani, Rosemary extract reduces
Akt/mTOR/p70S6K activation and inhibits proliferation and survival of A549
human lung cancer cells, Biomed. Pharmacother. 83 (2016) 725732, https://doi.
org/10.1016/j.biopha.2016.07.043.
[54] C.-C. Liang, A.Y. Park, J.-L. Guan, In vitro scratch assay: a convenient and
inexpensive method for analysis of cell migration in vitro, Nat. Protoc. 2 (2007)
329333, https://doi.org/10.1038/nprot.2007.30.
[55] D. Iitaka, S. Moodley, H. Shimizu, X.-H. Bai, M. Liu, PKCδ–iPLA2PGE2PPARγ
signaling cascade mediates TNF-
α
induced Claudin 1 expression in human lung
carcinoma cells, Cell. Signal. 27 (2015) 568577, https://doi.org/10.1016/j.
cellsig.2014.12.015.
[56] M.M. Bradford, A rapid and sensitive method for the quantitation of microgram
quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem.
72 (1976) 248254, https://doi.org/10.1016/0003-2697(76)90527-3.
[57] C. Obasaju, G.R. Hudes, Paclitaxel and docetaxel in prostate cancer, Hematol.
Oncol. Clin. North Am. 15 (2001) 525545, https://doi.org/10.1016/S0889-8588
(05)70230-6.
[58] Y. Li, Y. Zeng, S.M. Mooney, B. Yin, A. Mizokami, M. Namiki, R.H. Getzenberg,
Resistance to paclitaxel increases the sensitivity to other microenvironmental
stresses in prostate cancer cells, J. Cell. Biochem. 112 (2011) 21252137, https://
doi.org/10.1002/jcb.23134.
[59] P. Xiao, T. Ma, C. Zhou, Y. Xu, Y. Liu, H. Zhang, Anticancer effect of docetaxel
induces apoptosis of prostate cancer via the colin-1 and paxillin signaling
pathway, Mol. Med. Rep. 13 (2016) 40794084, https://doi.org/10.3892/
mmr.2016.5000.
[60] J. Cicenas, The potential role of Akt phosphorylation in human cancers, Int. J. Biol.
Markers 23 (2008) 19, https://doi.org/10.5301/JBM.2008.618.
[61] B.T. Nav´
e, D.M. Ouwens, D.J. Withers, D.R. Alessi, P.R. Shepherd, Mammalian
target of rapamycin is a direct target for protein kinase B: identication of a
convergence point for opposing effects of insulin and amino-acid deciency on
protein translation, Biochem. J. 344 (1999) 427431, https://doi.org/10.1042/
0264-6021:3440427.
[62] S. Sriprasad, M.R. Feneley, P.M. Thompson, History of prostate cancer treatment,
Surg. Oncol. 18 (2009) 185191, https://doi.org/10.1016/j.suronc.2009.07.001.
[63] S. Kar, S. Palit, W.B. Ball, P.K. Das, Carnosic acid modulates Akt/IKK/NF-jB
signaling by PP2A and induces intrinsic and extrinsic pathway mediated apoptosis
in human prostate carcinoma PC-3 cells, Apoptosis 17 (2012) 735747, https://
doi.org/10.1007/s10495-012-0715-4.
[64] Y.G. Jang, K.A. Hwang, K.C. Choi, Rosmarinic acid, a component of rosemary tea,
induced the cell cycle arrest and apoptosis through modulation of HDAC2
expression in prostate cancer cell lines, Nutrients 10 (2018), https://doi.org/
10.3390/nu10111784.
[65] A. Jaglanian, E. Tsiani, Rosemary extract inhibits proliferation, survival, akt, and
mTOR signaling in triple-negative breast cancer cells, Int. J. Mol. Sci. 21 (2020)
810, https://doi.org/10.3390/ijms21030810.
A. Jaglanian et al.
Biomedicine & Pharmacotherapy 131 (2020) 110717
12
[66] A. P´
erez-S´
anchez, E. Barraj´
on-Catal´
an, V. Ruiz-Torres, L. Agull´
o-Chazarra,
M. Herranz-L´
opez, A. Vald´
es, A. Cifuentes, V. Micol, Rosemary (Rosmarinus
ofcinalis) extract causes ROS-induced necrotic cell death and inhibits tumor
growth in vivo, Sci. Rep. (2019), https://doi.org/10.1038/s41598-018-37173-7.
[67] P.K. Majumder, W.R. Sellers, Akt-regulated pathways in prostate cancer, Oncogene
24 (2005) 74657474, https://doi.org/10.1038/sj.onc.1209096.
[68] M. Keniry, R. Parsons, The role of PTEN signaling perturbations in cancer and in
targeted therapy, Oncogene 27 (2008) 54775485, https://doi.org/10.1038/
onc.2008.248.
[69] J.J. Johnson, D.N. Syed, C.R. Heren, Y. Suh, V.M. Adhami, H. Mukhtar, Carnosol, a
dietary diterpene, displays growth inhibitory effects in human prostate cancer PC3
cells leading to G2-phase cell cycle arrest and targets the 5-AMP-activated protein
kinase (AMPK) pathway, Pharm. Res. 25 (2008) 21252134, https://doi.org/
10.1007/s11095-008-9552-0.
[70] J. Kim, K.L. Guan, mTOR as a central hub of nutrient signalling and cell growth,
Nat. Cell Biol. 21 (2019) 6367, https://doi.org/10.1038/s41556-018-0205-1.
[71] S. Yu, G. Shen, O. Khor, J.-H. Kim, A.-N. Kong, Curcumin inhibits Akt/mTOR
signaling through protein phosphatase-dependent mechanism, Mol. Cancer Ther. 7
(2008) 26092620, https://doi.org/10.1158/1535-7163.MCT-07-2400.
[72] B.L. Allen-Petersen, T. Risom, Z. Feng, Z. Wang, Z.P. Jenny, M.C. Thoma, K.R. Pelz,
J.P. Morton, O.J. Sansom, C.D. Lopez, B. Sheppard, D.J. Christensen, M. Ohlmeyer,
G. Narla, R.C. Sears, Activation of PP2A and inhibition of mTOR synergistically
reduce MYC signaling and decrease tumor growth in pancreatic ductal
adenocarcinoma, Cancer Res. 79 (2019) 209219, https://doi.org/10.1158/0008-
5472.CAN-18-0717.
[73] L. Li, A.H. Ross, Why is PTEN an important tumor suppressor? J. Cell. Biochem.
102 (2007) 13681374, https://doi.org/10.1002/jcb.21593.
[74] Y. Wang, T. Romigh, X. He, M.S. Orloff, R.H. Silverman, W.D. Heston, C. Eng,
Resveratrol regulates the PTEN/AKT pathway through androgen receptor-
dependent and -Independent mechanisms in prostate cancer cell lines, Hum. Mol.
Genet. 19 (2010) 43194329, https://doi.org/10.1093/HMG/DDQ354.
[75] J.S. Brown, U. Banerji, Maximising the potential of AKT inhibitors as anti-cancer
treatments, Pharmacol. Ther. 172 (2017) 101115, https://doi.org/10.1016/j.
pharmthera.2016.12.001.
[76] L. Craig, B. Stanley, Y. Melissa, B. Mark, L. Mark, Recent progress in the
development of ATP-Competitive and allosteric akt kinase inhibitors, Curr. Top.
Med. Chem. 7 (2007) 13491363, https://doi.org/10.2174/
156802607781696864.
[77] J.S. De Bono, U. De Giorgi, D.N. Rodrigues, C. Massard, S. Bracarda, A. Font, J.A.
A. Arija, K.C. Shih, G.D. Radavoi, N. Xu, W.Y. Chan, H. Ma, S. Gendreau,
R. Riisnaes, P.H. Patel, D.J. Maslyar, V. Jinga, Randomized phase II study
evaluating AKT blockade with ipatasertib, in combination with abiraterone, in
patients with metastatic prostate cancer with and without PTEN loss, Clin. Cancer
Res. 25 (2019), https://doi.org/10.1158/1078-0432.CCR-18-0981.
[78] X.X. Wei, A.C. Hsieh, W. Kim, T. Friedlander, A.M. Lin, M. Louttit, C.J. Ryan,
A phase I study of abiraterone acetate combined with BEZ235, a dual PI3K/mTOR
inhibitor, in metastatic castration resistant prostate Cancer, Oncologist 22 (2017),
https://doi.org/10.1634/theoncologist.2016-0432.
[79] S.M. Petiwala, G. Li, M.C. Bosland, D.D. Lantvit, P.A. Petukhov, J.J. Johnson,
Carnosic acid promotes degradation of the androgen receptor and is regulated by
the unfolded protein response pathway in vitro and in vivo, Carcinogenesis 37
(2016) 827838, https://doi.org/10.1093/carcin/bgw052.
[80] M. Naimi, F. Vlavcheski, B. Murphy, T. Hudlicky, E. Tsiani, Carnosic acid as a
component of rosemary extract stimulates skeletal muscle cell glucose uptake via
AMPK activation, Clin. Exp. Pharmacol. Physiol. 44 (2017) 94102, https://doi.
org/10.1111/1440-1681.12674.
[81] F. Vlavcheski, M. Naimi, B. Murphy, T. Hudlicky, E. Tsiani, Rosmarinic acid, a
rosemary extract polyphenol, increases skeletal muscle cell glucose uptake and
activates AMPK, Molecules 22 (2017), https://doi.org/10.3390/
molecules22101669.
A. Jaglanian et al.
... Primary active metabolites (COH, CA, and RA) found in rosemary leaf extracts u water and ethanol solvent systems were obtained from the existing literature [29,35 The over-expressed and under-expressed prostate adenocarcinoma (PRAD) genes differentially expressed genes (GSE179324) versus normal prostate cells for the DU cell line were identified. Figure 4 shows that the total number of shared genes betw significant rosemary metabolites in leaf extracts and PCa target genes is 37. ...
... Primary active metabolites (COH, CA, and RA) found in rosemary leaf extracts using water and ethanol solvent systems were obtained from the existing literature [29,[35][36][37][38][39][40]. The over-expressed and under-expressed prostate adenocarcinoma (PRAD) genes and differentially expressed genes (GSE179324) versus normal prostate cells for the DU-145 cell line were identified. ...
... water and ethanol solvent systems were obtained from the existing literature [29,[35][36][37][38][39][40]. The over-expressed and under-expressed prostate adenocarcinoma (PRAD) genes and differentially expressed genes (GSE179324) versus normal prostate cells for the DU-145 cell line were identified. ...
Article
Full-text available
Prostate cancer is one of the most prevalent cancer types diagnosed in older men. Investigations into traditional medicines like Rosmarinus officinalis L., popularly known as rosemary, are a current research interest due to its anti-cancer properties. This study investigates the cytotoxicity of aqueous and ethanolic rosemary leaf extracts in DU-145 cells and the interaction of its active metabolites with key prostate cancer targets using an in silico approach. The water extract of rosemary leaves showed greater cytotoxicity than the ethanol extract, with IC50 values of 1.4140 ± 0.1138 mg/mL and 1.8666 ± 0.0367 mg/mL, respectively; the highest cytotoxic effects for both extracts were observed at 5 mg/mL. These findings indicate significant cytotoxic differences based on concentration and solvent. Network pharmacology identified 37 genes linked to prostate adenocarcinoma, highlighting key genes like EGFR, TP53, ERBB2, IGFBP3, MMP-2, MMP-9, HDAC6, PDGFRB, and FGFR1. Molecular dynamics simulations and binding energy calculations revealed strong interactions between carnosol and rosmarinic acid with these targets, with TP53–carnosol showing the most stable conformation. Rosmarinic acid was identified as a promising candidate due to its low toxicity. This study demonstrates the potential anti-prostate cancer properties of rosemary leaf extracts for further investigations on the development of drugs against prostate cancer.
... Our findings align with previous studies, such as research involving Ls174-T colorectal cancer cells, which demonstrated that RA preserved over 70% cell viability and reduced metastasis without notable cytotoxicity 34,35 . Jaglanian et al. also found a dose-responsive decline in proliferation for both androgenindependent PC-3 and androgen-dependent 22RV1 prostate cancer cells when treated with RA 36 . Similarly, studies show that RA decreases cell viability in 22RV1 and LNCaP prostate cancer lines by modulating CHOP/ GADD153, promoting androgen receptor degradation, and reducing xenograft tumor growth 37 . ...
... Similarly, studies show that RA decreases cell viability in 22RV1 and LNCaP prostate cancer lines by modulating CHOP/ GADD153, promoting androgen receptor degradation, and reducing xenograft tumor growth 37 . Moreover, rosmarinic acid has been shown to induce a dose-dependent decrease in the proliferation and viability of PC-3, DU145, and LNCaP prostate cancer cells 36 . ...
Article
Full-text available
Prostate cancer remains a significant health concern due to its high mortality rate, emphasizing the need for innovative therapeutic approaches. This study aims to explore the potential anticancer effects of a drug nanocomplex containing rosmarinic acid in the treatment of prostate cancer, aiming to contribute to the development of safer and more effective treatment options for cancer patients. Nanocomposite Graphene Oxide was synthesized following the Hummers’ method. The resulted product dissolved in deionized water with rosmarinic acid to prepare the final product. To investigate the effects of rosmarinic acid@Se-TiO2-GO, PC3, LNCaP, and normal (HFF-1) cell lines were treated with varying concentrations (7.8, 15.6, 31.2, 62.5, 125, 250, and 500 µg/ml) of the nanocomplex. Cell viability was assessed using the Resazurin test, while levels of reactive oxygen species (ROS), gene expression (Bcl-2 and Bax), and total antioxidant capacity were measured in both cancerous and normal cells. The Se-TiO2-GO nanoparticles demonstrated high entrapment efficiency and loading capacity for rosmarinic acid. The IC50 values after 24 and 48 h of RA treatment were significantly greater than those recorded for treatments involving rosmarinic acid@Se-TiO2-GO. Treatment with rosmarinic acid@Se-TiO2-GO resulted in decreased cell viability and increased apoptosis in PC3 and LNCaP cells, while showing no inhibitory effects on the normal cell line (HFF-1) at concentrations toxic to cancer cells. Additionally, a dose-dependent increase in ROS levels, a decrease in total antioxidant capacity, elevated Bax gene expression, and reduced Bcl-2 expression were observed in the cancer cells following treatment with the nanocomplex. The cytotoxic effects of rosmarinic acid@Se-TiO2-GO nanoparticles on prostate cancer cells appear to be mediated through the generation of oxidative stress and induction of apoptosis. The unique formulation of these nanoparticles holds promise for future prostate cancer treatment strategies.
... The wound-healing (scratch) assay demonstrated the ability of the tested extract to inhibit cancer cell migration, consistent with previous studies reporting that different extracts from Salvia spp. and their bioactive compounds suppress cancer cell migration and metastasis in in vitro and in vivo cancer models [33][34][35]. In addition to its inhibitory effect on cancer cell migration, the S. aethiopis water extract was also found to suppress cancer cell proliferation by inducing apoptosis, necrosis and cell cycle arrest. ...
... Taken together with previous reports, the present findings indicate that Salvia spp. extracts trigger cancer cell death through different mechanisms, including both necrosis and apoptosis [34][35][36][37][38]. ...
Article
Full-text available
Salvia aethiopis L. (Mediterranean sage) is a medicinal plant known for its rich phenolic content and different therapeutic properties. This study evaluated the phytochemical composition, antioxidant capacity and anticancer potential of water extracts from in vitro cultivated S. aethiopis. The extract exhibited a high total polyphenol (110.03 ± 0.7 mg GAE/g) and flavonoid (7.88 ± 0.25 mg QE/g) content, along with a strong oxygen radical absorbance capacity (an ORAC value of 3677.9 ± 24.8 µmol TE/g). LC-HRMS analysis identified 21 bioactive compounds, including salvianic acid C, rosmarinic acid, salvianolic acid K and various organic acids. A cytotoxicity evaluation using the Neutral Red Uptake assay showed that the extract had a low toxicity to non-cancerous BALB/3T3 cells. An antiproliferative activity assessment via the MTT assay revealed selective cytotoxicity against Hep G2 hepatocellular carcinoma cells (IC50 = 353.8 ± 21.8 µg/mL) and lung (A549) and prostate (PC-3) carcinoma cell lines. Migration assays and cytopathological evaluations confirmed the significant inhibition of cancer cell proliferation, the suppression of migration and G2/M cell cycle arrest. Flow cytometry revealed considerable increases in apoptotic and necrotic cell populations following treatment with S. aethiopis extract. These findings showed the potential of S. aethiopis as a promising source of bioactive compounds with antioxidant and anticancer properties, supporting its further exploration for therapeutic applications.
... Also, the US FDA granted it the status of Generally Recognized as Safe (GRAS) [61]. Anticancer activities reported include colon cancer (extract showed strong inhibition of proliferation, migration, and colony formation of colon cancer cells regardless of their phenotype) [62], hepatocellular carcinoma (inhibited proliferation of HepG2 Cells) [63], lung cancer (extract decreased the activation of AKT/mTOR/p70S6 kinase (p70S6K) and showed inhibited proliferation and survival of A549 cells) [64], skin cancer (anti-proliferative effect on Human Melanoma A375 Cells) [65], oral cancer (rosemary exerts anti-inflammatory effects, proapoptotic, antiproliferative, and anti-angiogenic potential in buccal pouch carcinogenesis in a hamster model) [66], prostate cancer (extract inhibits prostate cancer cell proliferation and survival by targeting AKT and mTOR) [67], and breast cancer (extract exerts antiproliferative effects, inhibits survival, AKT, and mTOR signaling in triplenegative breast cancer cells) [68]. Evodia rutaecarpa (Juss.) ...
Article
Full-text available
Natural products obtained from medicinal and aromatic plants are increasingly recognized as promising anticancer agents due to their structural richness, including terpene and flavonoid molecules, which induce apoptosis and modulate gene expression. These compounds offer an alternative to conventional treatments, often costly, which face challenges such as multidrug resistance. This review aims to provide a promising alternative approach to effectively control cancer by consolidating significant findings in identifying natural products and anticancer agent development from medicinal and aromatic plants. It synthesizes the findings of a comprehensive search of academic databases, such as PubMed and Springer, prioritizing articles published in recognized peer-reviewed journals that address the bioprospecting of medicinal and aromatic plants as anticancer agents. The review addresses the anticancer activities of plant extracts and essential oils, which were selected for their relevance to chemoprevention and chemotherapy. Compounds successfully used in cancer therapy include Docetaxel (an antimitotic agent), Etoposide VP-16 (an antimitotic agent and topoisomerase II inhibitor), Topotecan (a topoisomerase I inhibitor), Thymoquinone (a Reactive Oxygen Species-ROS inducer), and Phenethyl isothiocyanate (with multiple mechanisms). The review highlights natural products such as Hinokitiol, Mahanine, Hesperetin, Borneol, Carvacrol, Eugenol, Epigallocatechin gallate, and Capsaicin for their demonstrated efficacy against multiple cancer types, including breast, cervical, gastric, colorectal, pancreatic, lung, prostate, and skin cancer. Finally, it highlights the need for continued bioprospecting studies to identify novel natural products that can be successfully used in modern chemoprevention and chemotherapy. Graphical Abstract
... 41 It hinders the initial activation of the allergic signalling pathway, including NF-kappaB, 42 and significantly impedes the messenger RNA for various pro-inflammatory chemokines and cytokines, such as IL-6, TNF, and IL-13. 43 Additionally, rosemary extract extends the latency period of cancer, 44 It also inhibits fungal growth, 45 promotes hair growth, 24 improves cell protection against UV radiation, and reduces skin damage caused by the sun. 46 Moreover, rosemary extract inhibits the production of nitric oxide (NO) within activated macrophages, 47 and exhibits emulsion stabilizing properties. ...
Article
Full-text available
Rosmarinus officinalis, commonly called rosemary is an aromatic plant native to the Mediterranean region. It has been employed in traditional medicine for its diverse therapeutic benefits. Contemporary research has validated its pharmacological potentials, attributing its efficacy to its rich phytochemical profile. This review explores the pharmacological and nutritional properties of rosemary, with particular focus on its bioactive compounds, including carnosic acid, carnosol, and rosmarinic acid. A comprehensive literature search across multiple databases yielded a robust dataset on the composition and biological activities of rosemary. The nutritional profile, encompassing essential oils, flavonoids, triterpenic acids, vitamins, minerals, and macronutrients, was meticulously examined. In addition, the review elucidated advanced extraction techniques for optimal bioactive compound recovery. By unraveling the mechanisms underlying the health-promoting effects of rosemary, this study provided adequate information that could guide the clinical applications and nutritional use of rosemary especially within the context of functional foods.
... Moreover, edible flowers hold appreciable health benefits against obesity, cardiovascular diseases, hepatic dysfunctions, and cancer (Rivas García et al., 2021). For instance, flowering rosemary (Rosmarinus officinalis L.) inhibited the proliferation and survival of prostate cancer cell by targeting Protein Kinase B (Akt) and Mammalian Target of Rapamycin (mTOR) signaling pathways (Jaglanian et al., 2020). The flowers of dandelion (Taraxacum officinale L.) exhibited anti-cancer activity by triggering G2/M phase arrest and inhibiting cell apoptosis in MDA-MB-231 cells (Li et al., 2017). ...
Article
Full-text available
Edible flowers have been used in dietary practices since ancient times. In recent years, they have garnered increasingly more attentions for their potentials in the prevention and amelioration of pathological conditions. The present study employed in vitro BSA models to evaluate the antiglycative effect of some edible flowers. Results showed that butterfly pea flower (BFPF) exhibited the highest potential in preventing advanced glycation end products (AGEs) formation, which had an inhibition rate of 92.11% at 1 g/mL, 56.99% at 0.1 g/mL, and 9.94% at 0.01 g/mL, respectively. Moreover, the antiglycative components in BFPF were identified as four flavonol glycosides through chromatographic and spectral analyses, which were manghaslin (quercetin 3-2″-rhamnosylrutinoside, QCT-Rh), clitorin (kaempferol 3–2″-rhamnosylrutinoside, KFR-Rh), rutin (quercetin 3-rutinoside), and kaempferol 3-neohesperidoside (KFR-Ne). Notably, KFR-Rh and KFR-Ne were presented in higher concentrations in BFPF (764.31 mg/kg and 1135.10 mg/kg dry matter) and significantly contributed to the antiglycative activity (IC50 = 182.17 μM and IC50 = 131.03 μM). Molecular docking (MD) and nuclear magnetic resonance (NMR) analyses revealed that KFR-Rh and KFR-Ne formed hydrogen bonds and hydrophobic interactions with BSA, while KFR-Ne demonstrating a stronger interaction than KFR-Rh. Collectively, our findings highlight the beneficial effects of BFPF with clearly identified active components, which might further promote its application in functional food and medical industry.
... Aside from cell proliferation, RE treatment resulted in a dosedependent inhibition of cell survival with IC50 values of 4.17 μg/ml and 2.43 μg/ml for 22RV1 and PC-3 cell lines, respectively. 44 The most prevalent malignancy in women to be diagnosed is breast cancer. Chemotherapy agents have been established in part because of compounds originating from plants. ...
Article
Full-text available
Objective: Apoptosis resistance and increased proliferation rates are characteristics of cancer cells. The anticancer properties of rosemary (Rosmarinus officinalis L.) extract (RE) have been demonstrated in a small number of in vivo and in vitro animal studies; however, no research has investigated the role of RE in human non-small cell lung cancer (NSCLC) A549 and H1299 cells, and its underlying mechanism of action remains unknown. In the current study, we examined the effects of RE on human non-small cell lung cancer cell proliferation, survival, and apoptosis. Methods: Human non-small cell lung cancer (NSCLC) A549 and H1299 were incubated with (2.5 µg/ml, 5 µg/ml, 7.5 µg/ml, 10 µg/ml, and 12.5 µg/ml) doses of RE for 12, 24, and 48 hours. MTT, Annexin V-PI, and caspase 3/7 assay kit were performed to detect the cell viability and apoptosis. Results: According to MTT analysis, the viability of A549 and H1299 human lung cancer cells was reduced by approximately 49.74% and 47.76%, respectively, for 24 hours by treatment with a dose of 5 µg/ml RE. The results of Annexin V-PI staining and Caspase 3/7 activation showed that RE had a greater effect on inducing cell death. Conclusion: In conclusion, we can say that rosemary extract has both apoptotic and antiproliferative properties on human lung cancer cells. We might propose that additional investigation is necessary to ascertain the therapeutic impacts of rosemary extract.
Article
Plant-derived products have been proven as beneficial medicinal treatments for various cancers. Rosmarinus officinalis L. (Rosemary - RM) is gaining increasing attention as an agent in cancer chemoprevention and therapy, due to its antioxidant, immunomodulatory, and anti-inflammatory properties. Herein, the anti-proliferative effects of RM leaves methanolic extract on human breast (triple-negative MDA-MB-231 and estrogen receptor-positive MCF-7), and liver cancer cells (HepG2 and HUH-7) relative to non-cancerous human cartilage chondrocytes (C20A4) are evaluated. Methyl thiazolyl tetrazolium (MTT) cell viability assay is used to determine drug effectiveness after 24- and 48-hour incubation periods. The residual extract from RM leaves is prepared and directly used in a fine powder after all solvents are evaporated. Five dosages of RM extract (0.05, 0.0625, 0.1, 0.125, and 0.25 mg/mL), identified by trial and error, and a negative control group are investigated. The RM extract is characterized by GC-MS and consists of three main compounds: eucalyptol (1,8-cineole), camphor (2-bornanone), and borneol ((2S)-1,7,7-trimethylbicyclo[2,2,1]heptan-2-ol). Our study demonstrates that dose-dependent treatment of RM extract leads to selective antioxidant, anti-proliferative and cytotoxic effects on breast carcinoma cell lines MDA-MB-231 and MCF-7, in comparison to C20A4 cells. The effect of RM extract on both liver carcinoma cell lines HepG2 and HUH-7 is insignificant compared to C20A4 cells. The optimum dosage of RM extract identified to counteract cancer cells is 0.25 mg/mL. However, further research is needed to determine mechanistic data. Overall, RM extract may have potential therapeutic value in the prevention and/or treatment of various types of cancer.
Article
Full-text available
Breast cancer is the most commonly diagnosed cancer in women. Triple-negative (TN) breast cancer lacks expression of estrogen receptor (ER), progesterone receptor (PR) as well as the expression and/or gene amplification of human epidermal growth factor receptor 2 (HER2). TN breast cancer is aggressive and does not respond to hormone therapy, therefore new treatments are urgently needed. Plant-derived chemicals have contributed to the establishment of chemotherapy agents. In previous studies, rosemary extract (RE) has been found to reduce cell proliferation and increase apoptosis in some cancer cell lines. However, there are very few studies examining the effects of RE in TN breast cancer. In the present study, we examined the effects of RE on TN MDA-MB-231 breast cancer cell proliferation, survival/apoptosis, Akt, and mTOR signaling. RE inhibited MDA-MB-231 cell proliferation and survival in a dose-dependent manner. Furthermore, RE inhibited the phosphorylation/activation of Akt and mTOR and enhanced the cleavage of PARP, a marker of apoptosis. Our findings indicate that RE has potent anticancer properties against TN breast cancer and modulates key signaling molecules involved in cell proliferation and survival.
Article
Full-text available
Background: Rosmarinus officinalis (R. officinalis) is a medicinal plant called rosemary, largely used in the Mediterranean diet for many decades ago. Objective: The aim of the present study was to investigate the polyphenolic content, the antioxidant activity, and the antiproliferative effect against human prostate cancer cell lines (LNCaP) of carnosol and carnosic acid as bioactive compounds contained in R. officinalis growing in Morocco. Materials and methods: Polyphenolic content of R. officinalis ethanolic extract was studied using colorimetric assay. Carnosol and carnosic acid contained in R. officinalis extract were quantified using high-performance liquid chromatography (HPLC). The antiproliferative effect of the studied extracts on LNCaP was evaluated by WST-1 bioassay, and the antioxidant activity was assessed using DPPH assay. Results: The extracts of R. officinalis showed an important polyphenolic content ranging from 74.15 μg·GAE/mg to 146.63 μg·GAE/mg. The percentage of carnosol and carnosic acid in rosemary crops ranges from 11.7 to 17.3% and 1.09% to 3%, respectively. The extracts of R. officinalis exhibited a promoting antioxidant activity with IC50 ranging from 0.236 mg/mL to 0.176 mg/mL. Regarding the antiproliferative effect, the WST-1 assay revealed that all the tested extracts reduced notably the cell viability with IC50 values ranging from 14.15 to 15. 04 μg/mL. Conclusion: In the current work, carnosol and carnosic acid exhibit antioxidant and antiproliferative activities in a concentration-dependent manner.
Article
Full-text available
Colorectal cancer is the third most common diagnosed cancer globally. Although substantial advances have been obtained both in treatment and survival rates, there is still a need for new therapeutical approaches. Natural compounds are a realistic source of new bioactive compounds with anticancer activity. Among them, rosemary polyphenols have shown a vast antiproliferative capacity against colon cancer cells in vitro and in animal models. We have investigated the antitumor activity of a rosemary extract (RE) obtained by using supercritical fluid extraction through its capacity to inhibit various signatures of cancer progression and metastasis such as proliferation, migration, invasion and clonogenic survival. RE strongly inhibited proliferation, migration and colony formation of colon cancer cells regardless their phenotype. Treatment with RE led to a sharp increase of intracellular ROS that resulted in necrosis cell death. Nrf2 gene silencing increased RE cytotoxic effects, thus suggesting that this pathway was involved in cell survival. These in vitro results were in line with a reduction of tumor growth by oral administration of RE in a xenograft model of colon cancer cells using athymic nude mice. These findings indicate that targeting colon cancer cells by increasing intracellular ROS and decreasing cell survival mechanisms may suppose a therapeutic option in colon cancer through the combination of rosemary compounds and chemotherapeutic drugs.
Article
Full-text available
Rosmarinic acid (RA), a main phenolic compound contained in rosemary which is used as tea, oil, medicine and so on, has been known to present anti-inflammatory, anti-oxidant and anti-cancer effects. Histone deacetylases (HDACs) are enzymes that play important roles in gene expression by removing the acetyl group from histone. The aberrant expression of HDAC in human tumors is related with the onset of human cancer. Especially, HDAC2, which belongs to HDAC class I composed of HDAC 1, 2, 3 and 8, has been reported to be highly expressed in prostate cancer (PCa) where it downregulates the expression of p53, resulting in an inhibition of apoptosis. The purpose of this study is to investigate the effect of RA in comparison with suberoylanilide hydroxamic acid (SAHA), an HDAC inhibitor used as an anti-cancer agent, on survival and apoptosis of PCa cell lines, PC-3 and DU145, and the expression of HDAC. RA decreased the cell proliferation in cell viability assay, and inhibited the colony formation and tumor spheroid formation. Additionally, RA induced early- and late-stage apoptosis of PC-3 and DU145 cells in Annexin V assay and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, respectively. In western blot analysis, RA inhibited the expression of HDAC2, as SAHA did. Proliferating cell nuclear antigen (PCNA), cyclin D1 and cyclin E1 were downregulated by RA, whereas p21 was upregulated. In addition, RA modulated the protein expression of intrinsic mitochondrial apoptotic pathway-related genes, such as Bax, Bcl-2, caspase-3 and poly (ADP-ribose) polymerase 1 (PARP-1) (cleaved) via the upregulation of p53 derived from HDAC2 downregulation, leading to the increased apoptosis of PC-3 and DU145 cells. Taken together, treatment of RA to PCa cell lines inhibits the cell survival and induces cell apoptosis, and it can be used as a novel therapeutic agent toward PCa.
Article
Full-text available
In cancer, kinases are often activated and phosphatases suppressed, leading to aberrant activation of signaling pathways driving cellular proliferation, survival, and therapeutic resistance. Although pancreatic ductal adenocarcinoma (PDA) has historically been refractory to kinase inhibition, therapeutic activation of phosphatases is emerging as a promising strategy to restore balance to these hyperactive signaling cascades. In this study, we hypothesized that phosphatase activation combined with kinase inhibition could deplete oncogenic survival signals to reduce tumor growth. We screened PDA cell lines for kinase inhibitors that could synergize with activation of protein phosphatase 2A (PP2A), a tumor suppressor phosphatase, and determined that activation of PP2A and inhibition of mTOR synergistically increase apoptosis and reduce oncogenic phenotypes in vitro and in vivo. This combination treatment resulted in suppression of AKT/mTOR signaling coupled with reduced expression of c-MYC, an oncoprotein implicated in tumor progression and therapeutic resistance. Forced expression of c-MYC or loss of PP2A B56α, the specific PP2A subunit shown to negatively regulate c-MYC, increased resistance to mTOR inhibition. Conversely, decreased c-MYC expression increased the sensitivity of PDA cells to mTOR inhibition. Together, these studies demonstrate that combined targeting of PP2A and mTOR suppresses proliferative signaling and induces cell death and implicates this combination as a promising therapeutic strategy for patients with PDA. Significance These findings present a combinatorial strategy targeting serine/threonine protein phosphatase PP2A and mTOR in PDA, a cancer for which there are currently no targeted therapeutic options.
Article
Full-text available
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt‑regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side‑effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Article
Full-text available
Purpose PI3K–Akt–mTOR and androgen receptor (AR) signaling are commonly aberrantly activated in metastatic castration-resistant prostate cancer (mCRPC), with PTEN loss associating with poor prognosis. We therefore conducted a phase Ib/II study of the combination of ipatasertib, an Akt inhibitor, with the CYP17 inhibitor abiraterone in patients with mCRPC. Patients and Methods: Patients were randomized 1:1:1 to ipatasertib 400 mg, ipatasertib 200 mg, or placebo, with abiraterone 1,000 mg orally. Coprimary efficacy endpoints were radiographic progression-free survival (rPFS) in the intent-to-treat population and in patients with PTEN-loss tumors. Results rPFS was prolonged in the ipatasertib cohort versus placebo, with similar trends in overall survival and time-to-PSA progression. A larger rPFS prolongation for the combination was demonstrated in PTEN-loss tumors versus those without. The combination was well tolerated, with no treatment-related deaths. Conclusions In mCRPC, combined blockade with abiraterone and ipatasertib showed superior antitumor activity to abiraterone alone, especially in patients with PTEN-loss tumors. See related commentary by Zhang et al., p. 901
Article
The highly conserved protein kinase mechanistic target of rapamycin (mTOR; originally known as mammalian target of rapamycin) is a central cell growth regulator connecting cellular metabolism and growth with a wide range of environmental inputs as part of mTOR complex 1 (mTORC1) and mTORC2. In this Review, we introduce the landmark discoveries in the mTOR field, starting from the isolation of rapamycin to the molecular characterizations of key components of the mTORC signalling network with an emphasis on amino acid sensing, and discuss the perspectives of mTORC inhibitors in therapeutic applications.
Article
Oncogenic signalling and metabolic alterations are interrelated in cancer cells. mTOR, which is frequently activated in cancer, controls cell growth and metabolism. mTOR signalling regulates amino acid, glucose, nucleotide, fatty acid and lipid metabolism. Conversely, metabolic inputs, such as amino acids, activate mTOR. In this Review, we discuss how mTOR signalling rewires cancer cell metabolism and delineate how changes in metabolism, in turn, sustain mTOR signalling and tumorigenicity. Several drugs are being developed to perturb cancer cell metabolism. However, their efficacy as stand-alone therapies, similar to mTOR inhibitors, is limited. Here, we discuss how the interdependence of mTOR signalling and metabolism can be exploited for cancer therapy.
Article
This article provides a status report on the global burden of cancer worldwide using the GLOBOCAN 2018 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer, with a focus on geographic variability across 20 world regions. There will be an estimated 18.1 million new cancer cases (17.0 million excluding nonmelanoma skin cancer) and 9.6 million cancer deaths (9.5 million excluding nonmelanoma skin cancer) in 2018. In both sexes combined, lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer death (18.4% of the total cancer deaths), closely followed by female breast cancer (11.6%), prostate cancer (7.1%), and colorectal cancer (6.1%) for incidence and colorectal cancer (9.2%), stomach cancer (8.2%), and liver cancer (8.2%) for mortality. Lung cancer is the most frequent cancer and the leading cause of cancer death among males, followed by prostate and colorectal cancer (for incidence) and liver and stomach cancer (for mortality). Among females, breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death, followed by colorectal and lung cancer (for incidence), and vice versa (for mortality); cervical cancer ranks fourth for both incidence and mortality. The most frequently diagnosed cancer and the leading cause of cancer death, however, substantially vary across countries and within each country depending on the degree of economic development and associated social and life style factors. It is noteworthy that high‐quality cancer registry data, the basis for planning and implementing evidence‐based cancer control programs, are not available in most low‐ and middle‐income countries. The Global Initiative for Cancer Registry Development is an international partnership that supports better estimation, as well as the collection and use of local data, to prioritize and evaluate national cancer control efforts. CA: A Cancer Journal for Clinicians 2018;0:1‐31. © 2018 American Cancer Society