ArticlePDF Available

Propranolol induced G0/G1/S phase arrest and apoptosis in melanoma cells via AKT/MAPK pathway

Authors:

Abstract and Figures

Both preclinical and epidemiology studies associate β-adrenoceptors-blockers (β-blockers) with activity against melanoma. However, the underlying mechanism is still unclear, especially in acral melanoma. In this study, we explored the effect of propranolol, a non-selective β-blocker, on the A375 melanoma cell line, two primary acral melanoma cell lines (P-3, P-6) and mice xenografts. Cell viability assay demonstrated that 50μM-400μM of propranolol inhibited viability in a concentration and time dependent manner with an IC50 ranging from 65.33μM to 148.60μM for 24h -72h treatment, but propranolol (less than 200μM) had no effect on HaCaT cell line. Western blots showed 100μM propranolol significantly reduced the expression of Bcl-2 while increasing the expressions of Bax, cytochrome c, cleaved capase-9 and cleaved caspase-3, and down-regulated the levels of p-AKT, p-BRAF, p-MEK1/2 and p-ERK1/2 in melanoma cells, after a 24h incubation. The in vivo data confirmed the isolation results. Mice received daily ip. administration of propranolol at the dose of 2 mg/kg for 3 weeks and the control group was treated with the same volume of saline. The mean tumor volume at day 21 in A375 xenografts was 82.33 ± 3.75mm3vs. 2044.67 ± 54.57mm3 for the propranolol-treated mice and the control group, respectively, and 31.66 ± 4.67 mm3vs. 1074.67 ± 32.17 mm3 for the P-3 xenografts. Propranolol also reduced Ki67, inhibited phosphorylation of AKT, BRAF, MEK1/2 and ERK1/2 in xenografts. These are the first data to demonstrate that propranolol might inhibit melanoma by activating the intrinsic apoptosis pathway and inactivating the MAPK and AKT pathways.
Content may be subject to copyright.
Oncotarget1
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Advance Publications 2016
Propranolol induced G0/G1/S phase arrest and apoptosis in
melanoma cells via AKT/MAPK pathway
Chengfang Zhou1, Xiang Chen3, Weiqi Zeng3, Cong Peng3, Gang Huang4, Xian’an
Li4, Zhengxiao Ouyang4, Yi Luo4, Xuezheng Xu4, Biaobo Xu1, Weili Wang1, Ruohui
He1, Xu Zhang3, Liyang Zhang5, Jie Liu1, Todd C. Knepper2, Yijing He1,2,3, Howard
L. McLeod1,2
1Department of Clinical Pharmacology, XiangYa Hospital, Central South University, Institute of Clinical Pharmacology, Central
South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
2Moftt Cancer Center, DeBartolo Family Personalized Medicine Institute, Tampa, FL, USA
3Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China
4Department of Orthopedics, Hunan Tumor Hospital, Changsha, China
5Department of Neurosurgery, Xiang-Ya Hospital, Central South University, Changsha, China
Correspondence to: Howard L. McLeod, email: Howard.McLeod@moftt.org
Yijing He, email: yijing.he@moftt.org
Keywords: melanoma, propranolol, apoptosis, AKT pathway, MAPK pathway
Received: February 15, 2016 Accepted: August 12, 2016 Published: August 25, 2016
ABSTRACT
Both preclinical and epidemiology studies associate β-adrenoceptors-blockers
(β-blockers) with activity against melanoma. However, the underlying mechanism
is still unclear, especially in acral melanoma. In this study, we explored the effect
of propranolol, a non-selective β-blocker, on the A375 melanoma cell line, two
primary acral melanoma cell lines (P-3, P-6) and mice xenografts. Cell viability assay
demonstrated that 50μM-400μM of propranolol inhibited viability in a concentration
and time dependent manner with an IC50 ranging from 65.33μM to 148.60μM for 24h
-72h treatment, but propranolol (less than 200μM) had no effect on HaCaT cell line.
Western blots showed 100μM propranolol signicantly reduced the expression of Bcl-2
while increasing the expressions of Bax, cytochrome c, cleaved capase-9 and cleaved
caspase-3, and down-regulated the levels of p-AKT, p-BRAF, p-MEK1/2 and p-ERK1/2
in melanoma cells, after a 24h incubation. The in vivo data conrmed the isolation
results. Mice received daily ip. administration of propranolol at the dose of 2 mg/kg
for 3 weeks and the control group was treated with the same volume of saline. The
mean tumor volume at day 21 in A375 xenografts was 82.33 ± 3.75mm3vs. 2044.67
± 54.57mm
3
for the propranolol-treated mice and the control group, respectively,
and 31.66 ± 4.67 mm3 vs. 1074.67 ± 32.17 mm3 for the P-3 xenografts. Propranolol
also reduced Ki67, inhibited phosphorylation of AKT, BRAF, MEK1/2 and ERK1/2 in
xenografts. These are the rst data to demonstrate that propranolol might inhibit
melanoma by activating the intrinsic apoptosis pathway and inactivating the MAPK
and AKT pathways.
INTRODUCTION
Melanoma, a cancer most commonly affecting
the skin, is derived from pigment-containing cells
known as melanocytes [1]. In China, the most common
subtype is acral lentiginous melanoma, which accounts
for approximately 40% of cutaneous melanoma
[2, 3]. Surgery followed by systemic chemotherapy
is recommended by National Comprehensive Cancer
Network (NCCN) for early stage melanoma patients.
For unresectable melanoma, BRAF/MEK inhibitors and
immune check points inhibitors are the rst line treatment
[4]. BRAF inhibitors, vemurafenib and dabrafenib, have
been approved for use in melanoma patients who are
Oncotarget2
www.impactjournals.com/oncotarget
BRAF V600E/V600K mutation carriers, these agents have
been shown to signicantly improve survival in BRAF-
mutated melanoma patients [5, 6]. The MEK inhibitors
trametinib can also improve survival in melanoma when
used in combination with BRAF inhibitors [7]. However,
the duration of response for single-agent vemurafenib
varied between 2 to 18 months [8]. Most patients
developed an acquired resistance to vemurafenib after 6
months of treatment [9]. Lack of durable response and the
development of acquired drug resistance are limitations to
the use of BRAF inhibitors, particularly as single-agents.
Mitogen-activated protein kinase (MAPK) and
AKT pathways play an important role in BRAF inhibitor
antitumor activity. Thep42/p44 MAPK and AKT are
highly activated during the growth stage of melanoma [10,
11]. Ras ultimately activates several signaling pathways
that are related to cell proliferation and anti-apoptotic
signalling cascades, including the Raf/MEK/extracellular
signal-regulated kinase (ERK) and phosphatidylinositol
3-kinase (PI3K)/ ATP-dependent tyrosine kinases (AKT)/
Phosphatase and PTEN pathways and NF-κB [12, 13].
β-adrenoceptors are also expressed in melanoma cells [14,
15]. Because of the important role of β-adrenergic receptor
in regulating AKT/MAPK pathway, we hypothesized
that blockage of β-adrenergic receptor may inhibit AKT/
MAPK pathway and lead to the death of melanoma cells.
Propranolol is a non-selective β-blocker widely
used for the treatment of hypertension, which has also
been demonstrated safety and efcacy for the treatment
of large hemangiomas in infants [16, 17]. Propranolol
has anti-proliferative, anti-migratory, anti-angiogenic and
cytotoxic properties in a wide variety of cancers [18–26].
In addition, a retrospective study revealed that melanoma
patients received β-blockers for hypertension achieved
longer survival than patients did not [27]. Recently,
Wrobelet al. reported that propranolol, not metoprolol,
inhibited proliferation of melanoma cell lines in vitro and
shrank the tumor size in vivo[15]. However, the underlying
mechanism of how β-blockers inhibit melanoma remains
unknown, especially with the acral melanoma subtype.
This study ascertained if propranolol could inhibit
proliferation and induce apoptosis in cutaneous/acral
melanoma and explored the potential mechanism of this
effect.
RESULTS
Propranolol cytotoxicity in cutaneous/acral
melanoma
The direct effect of propranolol on melanoma
proliferation was assessed using A375 cell, two acral
melanoma cell lines (P-3 and P-6 cell lines), and HaCaT,
a normal skin keratinocytes cell line. Cell viability was
measured by AlamarBlue® Cell Viability Assay. After
treatment for 24h, 48h and 72h with 25μM, 50μM,
100μM, 200μM and 400μM, propranolol respectively, cell
proliferation was signicantly reduced from 0.911 to 0.056
(Figure 1A, 1B, 1C, P<0.0001) in A375, 0.906 to 0.074
(Figure 1A, 1B, 1C, P<0.0001) in P-3, and 0.912 to 0.096
(Figure 1A, 1B, 1C, P<0.0001) in P-6, respectively. In
this assay, propranolol signicantly reduced cell viability
of the three melanoma cell lines (the range of IC50 in
A375: 65.33μM to 98.17, P-3: 116.86μM to 148.60μM,
P-6: 88.24μM to 118.23μM, Figure 1D, P < 0.05) in a
concentration and time dependent manner. Interestingly,
compared with melanoma cell lines, propranolol slightly
inhibited proliferation of HaCaT cell (the range of IC50
in HaCaT: 88.24μM to 466.11μM, Figure 1D) at the same
concentration (less than 200μM).
Propranolol induced apoptosis and inhibited cell
cycle in melanoma cell lines
After 100μM propranolol treatment for 24h, the
population of cells in G0/G1 greatly increased from
57.8%, 56.8% and 62.5% to 68.1%, 73.1% and 73.5%
in A375, P-3 and P-6 respectively (Figure 2A, Aa-
Ac, P< 0.0001), the distribution of S phase obviously
reduced from 27.9% to 9.0% in A375, 26.8% to 12.5%
in P-3 and 27.1% to 13.5% in P-6 (Figure 2A, Aa-Ac,
P< 0.0001), the number of G2M phase also signicantly
increased from 8.2% to 22.1% in A375, 7.6% to 13.1%
in P-6 (Figure 2A, Aa-Ac, P< 0.001), but no signicant
change in P-3 (Figure 2Ab). This redistribution of cells
within the cell cycle may be, at least in part, accounted for
propranolol-mediated apoptosis. Hoechst stain showed
the apoptotic cell number, dening with chromatin
condensation, nuclear shrinkage and formation of
apoptotic bodies, signicantly increased from 9.9% to
95.4% in A375 (Figure 2B, 2Ba, P< 0.0001), 6.6% to
88.3% in P-3 (Figure 2B, 2Bb, P< 0.0001), and 6.4% to
48.5% in P-6 (Figure 2B, 2Bc, P< 0.0001). These ndings
suggested that the propranolol might inhibit melanoma
cell lines by arresting cell progression at G0/G1 and S
phase and then inducing apoptosis.
Propranolol induced apoptosis by activating an
intrinsic apoptosis pathway
An inuence of propranolol on apoptosis may
contribute to the mechanistic effects. Melanoma cell lines
were exposed to 100μM propranolol for 24h, Bax was
signicantly increased in A375 (Figure 3A, Aa, P <0.001),
P-3 (Figure 3A, Ab, P <0.01) and P-6 (Figure 3A, Ac,
P <0.01) cell lines, while Bcl-2 was decreased in the
three melanoma cell lines (Figure 3B, Ba-Bc, P <0.01).
Propranolol also caused releasing of cytochrome c in
three cell lines (Figure 3C, Ca-Cc, P <0.01). Capase-9
and caspase-3, initiator caspases of the intrinsic apoptotic
pathway and one of the downstream target proteins of
cytochrome c, were activated by propranolol. An obvious
Oncotarget3
www.impactjournals.com/oncotarget
increase of the cleavage of caspase-9 and caspase-3
was observed in A375 (Figure 3D, Da, Figure 3E, Ea, P
<0.001), P-3 (Figure 3D, Db, Figure 3E, Eb, P <0.01) and
P-6 (Figure 3D, Dc, Figure 3E, Ec, P <0.01) cell lines.
These data strongly suggested that propranolol induced
apoptosis of melanoma cells by activating intrinsic
apoptosis pathway.
Propranolol inhibited proliferation by inhibiting
MAPK pathway
Since AKT and MAPK pathways play a key role in
tumor proliferation and apoptosis, we hypothesized that
propranolol might produce its strong function through
down-regulating activities in the two pathways. As shown
in Figure 4, p-BRAF (Figure 4A, Aa-Ac, P < 0.001),
p-MEK1/2 (Figure 4B, Ba-Bc, P < 0.001), p-Erk1/2
(Figure 4C, Ca-Cc, P < 0.001) and p-AKT (Figure 4D,
Da-Dc, P < 0.001) were greatly reduced while total
expressions of them were slightly decreased in propranolol
100μM group after a 24-hour incubation, suggesting that
the anti-proliferation might be mediated by simultaneously
targeting both ERK and AKT phosphorylation.
Propranolol inhibited melanoma growth in vivo
To evaluate whether the strong action observed
against melanoma in vitro could be transferred to
xenografts, the three human melanoma cells were
engrafted in BALB/C nude mice, while P-6 xenografts
were failed to develop to a solid tumor, we compared
the effect of propranolol on tumor growth. Previous
studies showed propranolol was effective in suppressing
melanoma tumor growth in vivo at dosages of 2-10 mg/
kg/day [15, 24]. In this study, mice received a daily
ip. injection of propranolol at the dose of 2 or 10mg/
kg for 3 weeks, the control group was treated with the
same volume of saline, but 80% animals were weight
loss and hemafecal after the higher dosage propranolol
administration (Data not shown), interestingly, the lower
showed better effect. As shown in Figure 5, the mean
tumor sizes of propranolol-treated mice were smaller
than the PBS group on day 21 in A375 xenografts (82.33
±3.75mm3 versus 2044.67 ± 54.57mm3, unpaired t-test,
P<0.0001, Figure 5A, 5C) and P-3 xenografts (31.66
±4.67 mm3 versus 1074.67 ± 32.17 mm3, unpaired t-test
P<0.0001, Figure 5B, 5D). Notably, no mice in the
Figure 1: The effect of propranolol on cell survival in melanoma cell lines and normal skin cell line. A-C. Cell viability was
determined following treatment with increasing propranolol concentration (25, 50, 100, 200, 400μM) for 24h, 48h and 72h by AlamarBlue®
Cell Viability Assay, relative growth was calculated as the ratio of treated to untreated cells at each dose for each replicate. D. IC50 of
propranolol after a 24h, 48h and 72h incubation in the four cell lines, respectively. ****P<0.0001. All experiments were repeated for at least
three times independently.
Oncotarget4
www.impactjournals.com/oncotarget
Figure 2: Propranolol induced cell cycle arrest and apoptosis in melanoma cells. A. A375, P-3 and P-6 cells were exposed
to100μM propranolol for 24h and then stained by propidium iodide (PI) to determine cell cycle assay by ow cytometry. B. Hoechst staining
was performed on the three cell lines after 100μM propranolol treatment. Aa-Ac, Data from (A) in histogram. Ba-Bc, Quantication of
Hoechst staining. ***P<0.001, ****P<0.0001. All experiments were repeated for at least three times independently.
Oncotarget5
www.impactjournals.com/oncotarget
propranolol group lost body weight (more than 10%) both
in A375 and P-3 xenografts (Figure 5E, 5F), indicating the
absence of gross toxicity for the treatment.
Propranolol inhibited proliferation and induced
apoptosis in melanoma xenografts
To determine whether the inducing apoptosis by
propranolol observed in vitro also functioned in vivo, we
performed IHC analyses on tumor sections from all the
experimental groups. H&E staining assay was applied to
investigate the role of propranolol on cell morphology (Figure
6A). When compared with PBS treated group, the number of
nuclei with karyorhexis and karyolysis was greatly increased in
propranolol treated xenografts, implying that propranolol could
promote cell necrosis in tumors (Figure 6A, Aa, Ab, P <0.01).
We also assessed the proliferation level indicated by cell
marker Ki-67 in tumor sections. The Ki67 index signicantly
decreased in both A375 (47.67% ± 3.84 % versus 16.00% ±
2.65%, N=6, P= 0.0025, Figure 6B, Ba) and P-3 (41.67% ±
2.60% versus 21.33% ± 2.60%, N=6, P=0.0052, Figure 6B,
Bb) xenografts. To demonstrate the role of propranolol on cell
death, tunnel terminal deoxynucleotidyl transferase dUTP nick
end labeling (TUNEL) assay was performed. In A375 derived
tumors, TUNEL positive cell was signicantly (P < 0.0001)
higher of propranolol group (92.67% ± 1.86%) than PBS
group (3.67% ± 0.88%, Figure 6C, Ca). In P-3 derived tumors,
higher proportion of TUNEL positive cells was observed in the
propranolol treated group than in the control group (89.0% ±
2.65% vs 6.33% ± 1.76%, P<0.0001, N=6, Figure 6D, Da).
Figure 3: Propranolol activated mitochondria-mediated apoptosis pathway in melanoma. A-E. The expressions of Bax,
Bcl-2, cytochrome c, caspase-9 and caspase-3 following exposure to propranolol 100μM for 24h in A375, P-3 and P-6 cell lines. Aa-Ec.
Quantication of A-D. Results are presented as mean±SEM, *P<0.05, ***P<0.001, ****P<0.0001. All experiments were repeated for at least
three times independently.
Oncotarget6
www.impactjournals.com/oncotarget
Propranolol inhibited AKT pathway and MAPK
pathway in vivo
To determine whether the down-regulation of
MAPK and AKT signaling by propranolol observed in
vitro also functioned in vivo, immunohistochemistry
analysis was applied to determine the phosphorylation of
the AKT and MAPK pathways in tumors. As observed in
Figure 7, the data showed that the expression of p-AKT
was signicantly (P<0.0001) reduced in the propranolol
treated group compared with PBS group both in A375
(Figure 7A, Aa) and P-3 xenografts (Figure 7A, Ab). The
Figure 4: Propranolol inhibited MAPK pathway. A-D. BRAF, MEK1/2, ERK1/2 and AKT phosphorylation following treatment
with propranolol 100μM for 24h in A375, P-3 and P-6 cell lines. Aa-Df, Quantication of A-D. Results are presented as mean±SEM,
*P<0.05, **P<0.01,***P<0.001, ****P<0.0001. All experiments were repeated for at least three times independently.
Oncotarget7
www.impactjournals.com/oncotarget
expression of p-BRAF and p-MEK1/2 were moderately
lowered by propranolol in A375 (Figure 7B, 7C, Ba, Ca)
and P-3 xenografts (Figure 7B, 7C, Bb, Cb). Furthermore,
the phosphorylated ERK1/2 was reduced in A375 (Figure
7D, Da) and P-3 (Figure 7D, Db) derived tumors in the
propranolol treated group. These data revealed that
propranolol could inhibit the growth of melanoma by
inhibiting the phosphorylation of AKT and MAPK
pathway in vivo.
DISCUSSION
Accumulative studies have reported that
β-blockers have anti-tumor effects by inhibiting
proliferation, inducing apoptosis and inhibiting
metastasis of many types of tumor [24–26]. Calvani M
and colleagues found β3-AR activation in melanoma
accessory cells drives stromal reactivity by inducing
pro-inammatory cytokines secretion and de novo
angiogenesis, sustaining tumor growth and melanoma
aggressiveness, which validated selective β3-AR
antagonists as potential promising anti-metastatic agents
[28]. Another study demonstrated the combination
of propranolol with 5-FU or paclitaxel resulted in
more profound and sustained anti-tumor effects and
signicantly increased the survival benets in MDA-
MB-231 cell xenografts [20]. In 2015, Wrobelet al.
reported that propranolol could inhibit proliferation in
Figure 5: Propranolol inhibits tumor development in xenografts. A, B. Tumors excised from xenografts of A375 and P-3 model
mice, untreated mice (PBS group) and mice treated with propranolol (propranolol group). C, D. The growth curves of tumor measured by
average volume of 6 tumors in each group. E, F. Body weights of A375 and P-3 model mice were measured before and after the beginning
of the treatment by three days interval.
Oncotarget8
www.impactjournals.com/oncotarget
Figure 6: The effect of propranolol on tumor histology in vivo. A. Hematoxylin- -eosin staining (HE) for cell morphology in
propranolol and PBS group. B. Ki67 was assessed by immunohistochemistry assay. C-D. Cell death was measured by TUNEL assay in
A375 and P-3 xenografts from untreated (left panels) and propranolol treated mice (right panels). Ba,-Bb, Quantication of Ki67 staining
in A375 (n=30 estimations on 6 tumors, both in propranolol and PBS groups) and P-3 (n=30 estimations on 6 tumors, both in propranolol
treated and PBS groups) xenografts mice. Ca, Da, Quantication of TUNEL staining in A375 (n=36 estimations on 6 tumors, both in
propranolol and PBS groups) and P-3 (n=36 estimations on 6 tumors, both in propranolol and PBS groups) xenografts mice. Results are
presented as mean±SEM, **P<0.01, ****P<0.0001. All experiments were repeated for at least three times independently.
Oncotarget9
www.impactjournals.com/oncotarget
melanoma cell lines without knowing its underlying
mechanism [15]. This study demonstrated that
propranolol inhibited melanoma growth by regulating
the AKT pathway and MAPK pathways and inducing
apoptosis. It is also the rst evidence showing that
activation of apoptosis by propranolol could down-
regulate the activity of AKT and MAPK pathways both
in cutaneous and acral melanoma.
Figure 7: The effect of propranolol on Akt and MAPK pathway in vivo. A-D. p-Akt, p-BARF, p-MEK, and p-ERK were
assessed by immunohistochemistry assay in A375 (n=30 estimations on 6 tumors, both in propranolol and PBS groups) and P-3 (n=40
estimations on 6 tumors, both in propranolol and PBS groups) derived xenografts. Aa-Db. Quantication of p-AKT, p-BRAF, p-MEK1/2
and p-ERK1/2 staining in A375 (n=30 estimations on 6 tumors, both in propranolol and PBS groups) and P-3 (n=30 estimations on 6
tumors, both in propranolol treated and PBS groups) xenografts mice, respectively. Results are presented as mean±SEM, ***P<0.001,
****P<0.0001. All experiments were repeated for at least three times independently.
Oncotarget10
www.impactjournals.com/oncotarget
Propranolol inhibited proliferation in the three
melanoma cell lines. Wrobel and his colleague also
addressed that propranolol signicantly reduced melanoma
cell viability with 100μM for 24h [15]. Similar result was
reported by Albiñana who found that propranolol reduced
viability and induced apoptosis in hemangioblastoma
cells from von Hippel-Lindau patients [29]. Consistent
with the in vitro studies, propranolol shrunk tumor size
and inhibited the expression of Ki67 in xenograft models.
In neuroblastoma and breast cancer, propranolol was
also observed to reduce tumor volume in xenografts
models [20, 24]. Propranolol inhibition of proliferation
of cutaneous and acral melanoma occurred in a time
and concentration dependent manner. This observation
is consistent with the hemangioblastoma study, which
found 50 and 100μM propranolol signicantly reduced
cell viability with longer incubation time (24h-96h)
[29]. Liao and his colleague also demonstrated that
propranolol reduced proliferation of gastric cancer cells
in a concentration-dependent manner (25μM-300μM)
[23]. Pharmacokinetic data suggested that the peak serum
concentrations of propranolol ranged from 200-400 ng/
ml, which is equivalent to 0.77-1.5μM in vivo[30]. But
the IC50 value of propranolol was more than 60 μM after
a 24-hour incubation in three melanoma lines. Large
discrepancy of drug concentration was observed between
in vivo and in vitro models. This may because propranolol
could exert the anti-tumor effect via multiple pathways in
vivo compared with in vitro models, such as propranolol,
could inuence angiogenesis [31, 32] and attenuate
immune-suppressionin tumor [33], thus may require less
drug concentrations to achieve similar effect. On the other
hand, propranolol showed no effect on normal skin cell
line at the same concentration. This is consistent with a
recent study, which demonstrated that propranolol did not
show any cytotoxicity in normal skin cells and normal
melanocytes [15].
The mechanisms of anti-tumor activity of β-blockers
have been explored by other investigators in other cancer.
Lin X et al. found that the non-selective β-AR agonist
isoproterenol signicantly increased the activation of
ERK/MAPK signal pathway in pancreatic cancer cell [25].
Huang’s study showed that norepinephrine also stimulated
pancreatic cancer cell proliferation, migration and invasion
via β-AR dependent activation of P38/MAPK pathway
[34]. Propranolol and other β-blockers reduced the activity
of MAPK in pancreatic cancer and Hemangioma [35, 36].
Meanwhile, inhibition of MAPK is a widely used strategy
for melanoma treatment [37]. Vemurafenib, a BRAFV600E
inhibitor, has achieved high response rate (more than 50%)
at the initial stage of vemurafenib treatment in advanced
melanoma patients [38–40]. However, more than 60%
patients developed acquired drug resistance (ADR) after
6 months [41–43]. One possible reason of vemurafenib
induced ADR was believed to be the reactivation of
MAPK pathway in melanoma [44]. This study further
advanced our knowledge on the therapeutic mechanism
of propranolol in melanoma. More signicantly, our data
also provide a scientic basis on a strategy of using the
propranolol inhibitory effects on the MAPK pathway
to overcoming the vemurafenib-induced resistance in
melanoma treatment.
In summary, this data implied that propranolol could
inhibit melanoma in vitro and in vivo. The study highlights
for the rst time that propranolol may exert the anti-tumor
effect in cutaneous and acral melanoma by suppressing
AKT and MAPK pathways. Clinical trials are needed to
verify the treatment effect of propranolol in melanoma
patients as an adjuvant regimen.
MATERIALS AND METHODS
Cell lines and reagents
The A375 cell line, derived from chronic sun-
induced damage (CSD) cutaneous melanoma [45], was
obtained from American type culture collection (ATCC).
All cell lines were cultured in DMEM medium (Gibco,
Life Technologies, China) supplemented with 10% FBS
(Gibco, Life Technologies Australia) at 37°C and 5%
CO2 in tissue culture incubator. The A375 cell line was
regularly veried to be mycoplasmae –free and no cross
cell contaminationusing ABI 3100 type genetic analyzer
(Applied Biosystems, USA). (Supplementary Figure S1).
Human melanoma primary cultures
Patient 3 cell line (P-3) and patient 6 cell line
(P-6) were derived from surgical resection samples
of acral melanoma from two patients (Supplementary
Figure S2). Written informed consent was obtained from
the patients. Melanoma specic antigens were detected
via western blotting for CD31, CD63, CD166 and CD146
conrmed that the cells were melanocytes (Supplementary
Figure S3, Supplementary Figure S6). Tumor tissues were
mechanically dissociated with a small tissue chopper prior
to sequential enzymatic digestion in 2 mg/ml Collagenase
(Sigma-Aldrich, Schnelldorf, Germany) and 1 mg/ml
Dispase I (Gibco/Invitrogen, Carlsbad, CA) in DMEM for
30 min at 37°C. Cells were ltered (100 μm cell strainer)
to obtain a single cell suspension and re-supplemented by
phosphate-buffered saline, thereafter centrifuged at 1000
r.p.m. for 5 minutes. Pellet was re-suspended in DMEM
with 10% FBS and then cultured in 75 cm
3
plastic asks
(Themo Fisher Scientic, China).
Adenylyl cyclase activity
Adenylyl cyclase activity was measured by a
protein binding assay [46]. Cell pellets were thawed and
homogenised in a glass douncehomogeniser containing
ice-cold homogenization buffer (0.3 M sucrose, 25
Oncotarget11
www.impactjournals.com/oncotarget
mMTris, pH 7.4). A 40 μL sample of homogenate
was then added to 30μL premix buffer (nal assay
concentration 50 mMTris (pH 7.5), 5 mM Mg2+, 1 mM
ATP (Sigma-Aldrich, Schnelldorf, Germany), 1μM GTP
(G9002-25MG,Sigma-Aldrich, Schnelldorf, Germany),
250μM 4- (3-Butoxy- 4-methoxybenzyl) imidazolidin-
2-one (RO20-1724, Abcam, USA), 20 mMcreatine
phosphate (000000010621714001, Roche) and 130 U/mL
creatine phosphokinase (C7886-500UN, Sigma-Aldrich,
Schnelldorf, Germany)). The tubes were incubated at 37
°C for 10 min and the reaction terminated by the addition
of 20 μL of 100% trichloroacetic acid and the tubes placed
on ice for 10 min. Precipitated protein was pelleted by
centrifugation at 2,900 × g for 20 min at 4 °C, the resulting
supernatant was used to determine cAmp level by cAMP
Direct Immunoassay Kit (Catalog#K371-100, BioVison,
USA) according to the instruction. The adenylyl cyclase
activity expressed as pmol cyclic AMP min
-1
mg
-1
protein.
Quantitative real-time PCR (qRT-PCR)
Total RNA was extracted from the three melanoma
and HaCaT cell lines by using Trizol reagent (Invitrogen,
Carlsbad, CA) according to the manufacturer’s
instructions. qRT-PCR was performed using the
SYBR® Green Realtime PCR Master Mix assay kit
(Toyobo, Osaka, Japan) according to the manufacturer’s
instructions. The primers of target genes were as follows:
β1AR: 5’-ATCGAGACCCTGTGTGTCATT-3’ (forward)
and 5’-GTAGAAGGAGACTACGGACGAG-3’ (reverse),
β2AR: 5’-TTGCTGGCACCCAATAGAAGC-3’ (forward)
and 5’-CAGACGCTCGAACTTGGCA-3’ (reverse),
β3AR: 5’-GACCAACGTGTTCGTGACTTC-3’ (forward)
and 5’-GCACAGGGTTTCGATGCTG-3’ (reverse),
and GAPDH: 5’-GAGTCAACGGATTTGGTCGT-3’
(forward) and 5’-TTGATTTTGGAGGGATCTCG-3’
(reverse). The results were analyzed using the 2-ΔΔCt
method as the following formula: ΔΔCt = ΔCtβ2/β3AR-
ΔCtβ1AR, ΔCt = Ct target gene -CtGAPDH.
Cell viability assays
The half-maximal inhibitory concentration (IC50)
value was determined by AlamarBlue
®
assay (Invitrogen,
Burlington, ON, Canada). A375 cells were plated in
96-well plates at a density of 2×103 and treated with
25μM-400μM propranolol (propranolol hydrochloride,
P0884, Sigma-Aldrich, USA) for 24h, 48h and 72h.
Cells were incubated with 10% of AlamarBlue overnight.
Fluorescence of each plate was measured using a
spectrophotometer at excitation 530 nm and emission 590
nm (Spectra MAX Gemini EM, Molecular Devices).
Cell cycle analysis by ow cytometry
A375,P-3 and P-6 cell lines were cultured at a
density of 1 × 106 cells in 100 mm2 culture dishes (Corning,
China) and were treated with 100 spectrophotometer 24h.
Cells were then harvested, washed, xed overnight in 70
% ethanol at −20°C, after washed with PBS, digested
with RNase A, stained with propidium iodide (50 with
propidiumilyzed by ow cytometry using BD Accuri
TM
C6
(Becton, Dickinson and Company, USA).
Western blot analysis
Western blot analysis was performed on cell extracts
of A375,P-3 and P-6 cell lines treated with 100μM
propranolol for 24h. Immunoblots were performed from
whole cell lysate prepared using RIPA Buffer (Cell
Signaling Technology) supplemented with dithiothreitol
(DTT), phenylmethylsulphonyl uoride (Sigma), and fresh
protease and phosphatase inhibitors (Sigma). Cell lysates
were quantied for protein content using a bicinchoninic
acid (BCA) protein assay kit (Thermo). Protein samples
were resolved on NuPAGE 12% Bis-Tris gels with MOPS
buffer or 3–8% Tris acetate gels with Tris acetate buffer
(Life Technologies) and then transferred to 0.45-mm
nitrocellulose membrane (Bio-Rad). After saturation in
Tris-buffered saline supplemented with 5% BSA, the
membranes were incubated with antibodies (diluted at
1:2,000) overnight at 4°C. Antibodies specic for the
following proteins were purchased from Cell Signaling
Technology: AKT (rabbit, 9272), B-Raf (rabbit, 9433),
MEK1/2 (L38C12) (mouse, 4694), ERK1/2 (rabbit, 9102),
phospho-ser473-AKT (rabbit, 9271), phosphor-ser445-
BRAF (rabbit, 2696), phosphor-ser221-MEK (rabbit,
2338), phospho-ERK1/2-Thr202/Tyr204 (rabbit, 4370),
cytochrome c-D18C7 (rabbit, 11940), caspase-9 (mouse,
9508) and caspase-3 (rabbit, 9662). The antibodies specic
for Bax (rabbit, sc-6236) and Bcl-2 (rabbit, sc-492), were
purchased from Santa Cruz Biotechnology. The antibodies
for BAD (rabbit, ab32445), p-BAD (rabbit, ab28824),
CD31 (mouse, ab9498), CD147 (rabbit, ab108317),
CD146 (mouse, ab24577), CD63 (mouse, ab8219) and
CD166 were purchased from Abcam. The antibody
specic for GAPDH (mouse, clone 6C5, MAB374) was
purchased from Millipore. Quantication of the bands was
done with Image J.
Hoechst staining
Cells treated with propranolol were xed, washed
twice with PBS and stained with Hoechst 33258 staining
solution according to the manufacturer’s instructions
(Beyotime, Jiangsu, China). Stained nuclei were observed
under a uorescence microscope (Leica-microsystems-
DM5000B+DFC1300,Leitz Wild Group, Germany).
Xenografts of human melanoma
The six weeks old NOD/SCID male mice (Hunan
Silaike experimental Animals Inc, China) was injected
subcutaneously with 106living melanoma cells that were
Oncotarget12
www.impactjournals.com/oncotarget
maintained in DMEM with 2% FBS and 15% Matrigel
(BD Bioscience, Franklin Lakes, NJ) in the left ank. A
solid tumor develops in A375 and P-3 xenografts within
two weeks, while P-6 xenografts were failed to develop.
In vivo propranolol treatment
The NOD/SCID mice with xenografts were
randomly divided into three groups after the tumor volume
reached 0.3 cm3. The propranolol group (n=6) received
a daily ip. injectionof propranolol (Sigma-Aldrich) at the
dose of 2 or 10mg/kgfor 3 weeks and the control group
(n=6) was treated with the same amount of saline. Tumor
volume was measured by length (l), width (w), and height
(h) twice a week with an external caliper and tumor size
was calculated by the modied ellipsoidal formula (Tumor
volume =π/6× l × w × h). The mice were sacriced at the
end of 3 weekstreatment. The xenografts were removed,
weighted and then snap-frozen in liquid nitrogen. Parafn-
embedded tumor blocks were prepared for further analysis
at the same time.
Immunohistochemistry and TUNEL assay
Deparafnized tissue sections were treated with
Antigen Retrieval Solution (made from citrate buffer, pH
6.0, concentrated 103, T0050 Diapath). Tissue sections
were then incubated with Peroxidase Blocking Solution
(S2023, Dako) for 15 min and Protein Block (X0909,
Dako) for 20 min. Primary antibody specic for phosphor-
ser221-MEK (rabbit, 2338, CST), phospho-ERK1/2-
Thr202/Tyr204 (rabbit, 4370, CST) and phospho-ser473-
AKT (rabbit, 9271, CST) and p-RAF-B (goat, sc-28006,
Santa Cruz Biotechnology) were applied, and the slides
were incubated overnight at 4°C. Signals were visualized
using rabbit HRP-conjugated secondary antibody (K4003,
Dako) and a haematoxylin (MHS32, Sigma) counterstain.
TUNEL assay was done using Cell-LightTMEdUTP
TUNEL In situ Detection Kit (Ruibo, China).
Statistical procedures
Drug sensitivity of propranolol and cell cycle
population analysis were done using an analysis of
variance on repeated measures with the Graphpad Prism
software (GraphPad Software, Inc., version 6.0).
ACKNOWLEDGMENTS
This work was supported by the National Natural
Science Foundation of China (Grant No. 81070902) and
by the Young Scientists Fund of the National Natural
Science Foundation of China (Grant No. 30600774
and No.81403022). Project supported by the China
Postdoctoral Science Foundation (Grant No. 201104515
and No. 2012M510138). The authors thank Prof.
William Tse from Division of Blood and Bone Marrow
Transplantation James Graham Brown Cancer Center
Department of Medicine University of Louisville School
of Medicine for editorial assistance on the text and study
suggestions.
CONFLICTS OF INTEREST
The authors state no conict of interest.
HIGHLIGHTS
1. Propranolol could inhibit the cell proliferation and
induce apoptosis of melanoma via activating the intrinsic
apoptosis pathway and inactivating the MAPK and AKT
pathways in vitro and in vivo.
2. Acral melanoma cells deprived from patients
were showed to response to propranolol treatment in vivo
and in vitro.
REFERENCES
1.
Melanoma Treatment–for health professionals (PDQ®).
National Cancer Institute, June 26, 2015.
2. Chi, Z., S. Li, X. Sheng, L. Si, C. Cui, M. Han, and J. Guo,
Clinical presentation, histology, and prognoses of malignant
melanoma in ethnic Chinese: a study of 522 consecutive
cases. BMC Cancer, 2011. 11: p. 85.
3.
Chang, J.W., Acral melanoma: a unique disease in Asia.
JAMA Dermatol, 2013. 149: p. 1272-3.
4.
NCCN Guidelines Version 1.2016 Melanoma. National
Comprehensive Ccancer Network, 2015.
5.
Sosman, J.A., K.B. Kim, L. Schuchter, R. Gonzalez,
A.C. Pavlick, J.S. Weber, G.A. McArthur, T.E. Hutson,
S.J. Moschos, K.T. Flaherty, P. Hersey, R. Kefford, D.
Lawrence, et al., Survival in BRAF V600-mutant advanced
melanoma treated with vemurafenib. N Engl J Med, 2012.
366: p. 707-14.
6.
Hauschild, A., J.J. Grob, L.V. Demidov, T. Jouary, R.
Gutzmer, M. Millward, P. Rutkowski, C.U. Blank,
W.H. Miller, Jr., E. Kaempgen, S. Martin-Algarra, B.
Karaszewska, C. Mauch, et al., Dabrafenib in BRAF-
mutated metastatic melanoma: a multicentre, open-label,
phase 3 randomised controlled trial. Lancet, 2012. 380:
p. 358-65.
7.
Long, G.V., D. Stroyakovskiy, H. Gogas, E. Levchenko,
F. de Braud, J. Larkin, C. Garbe, T. Jouary, A. Hauschild,
J.J. Grob, V. Chiarion-Sileni, C. Lebbe, M. Mandala, et al.,
Dabrafenib and trametinib versus dabrafenib and placebo
for Val600 BRAF-mutant melanoma: a multicentre, double-
blind, phase 3 randomised controlled trial. Lancet, 2015.
386: p. 444-51.
8.
Poulikakos, P.I. and N. Rosen, Mutant BRAF melanomas--
dependence and resistance. Cancer Cell, 2011. 19: p. 11-5.
Oncotarget13
www.impactjournals.com/oncotarget
9.
Flaherty, K.T., I. Puzanov, K.B. Kim, A. Ribas, G.A.
McArthur, J.A. Sosman, P.J. O’Dwyer, R.J. Lee, J.F.
Grippo, K. Nolop, and P.B. Chapman, Inhibition of mutated,
activated BRAF in metastatic melanoma. N Engl J Med,
2010. 363: p. 809-19.
10.
Govindarajan, B., J.E. Sligh, B.J. Vincent, M. Li, J.A.
Canter, B.J. Nickoloff, R.J. Rodenburg, J.A. Smeitink,
L. Oberley, Y. Zhang, J. Slingerland, R.S. Arnold, J.D.
Lambeth, et al., Overexpression of Akt converts radial
growth melanoma to vertical growth melanoma. J Clin
Invest, 2007. 117: p. 719-29.
11.
Cohen, C., A. Zavala-Pompa, J.H. Sequeira, M. Shoji, D.G.
Sexton, G. Cotsonis, F. Cerimele, B. Govindarajan, N.
Macaron, and J.L. Arbiser, Mitogen-actived protein kinase
activation is an early event in melanoma progression. Clin
Cancer Res, 2002. 8: p. 3728-33.
12.
McCubrey, J.A., L.S. Steelman, W.H. Chappell, S.L.
Abrams, E.W. Wong, F. Chang, B. Lehmann, D.M. Terrian,
M. Milella, A. Tafuri, F. Stivala, M. Libra, J. Basecke,
et al., Roles of the Raf/MEK/ERK pathway in cell growth,
malignant transformation and drug resistance. Biochim
Biophys Acta, 2007. 1773: p. 1263-84.
13.
Agell, N., O. Bachs, N. Rocamora, and P. Villalonga,
Modulation of the Ras/Raf/MEK/ERK pathway by Ca(2+),
and calmodulin. Cell Signal, 2002. 14: p. 649-54.
14.
Yang, E.V., S.J. Kim, E.L. Donovan, M. Chen, A.C.
Gross, J.I. Webster Marketon, S.H. Barsky, and R.
Glaser, Norepinephrine upregulates VEGF, IL-8, and
IL-6 expression in human melanoma tumor cell lines:
implications for stress-related enhancement of tumor
progression. Brain Behav Immun, 2009. 23: p. 267-75.
15.
Wrobel, L.J. and F.A. Le Gal, Inhibition of human
melanoma growth by a non-cardioselective beta-blocker. J
Invest Dermatol, 2015. 135: p. 525-31.
16.
Leaute-Labreze, C., E. Dumas de la Roque, T. Hubiche,
F. Boralevi, J.B. Thambo, and A. Taieb, Propranolol for
severe hemangiomas of infancy. N Engl J Med, 2008. 358:
p. 2649-51.
17.
Leaute-Labreze, C., P. Hoeger, J. Mazereeuw-Hautier, L.
Guibaud, E. Baselga, G. Posiunas, R.J. Phillips, H. Caceres,
J.C. Lopez Gutierrez, R. Ballona, S.F. Friedlander, J.
Powell, D. Perek, et al., A randomized, controlled trial of
oral propranolol in infantile hemangioma. N Engl J Med,
2015. 372: p. 735-46.
18.
Yang, E.V., A.K. Sood, M. Chen, Y. Li, T.D. Eubank,
C.B. Marsh, S. Jewell, N.A. Flavahan, C. Morrison,
P.E. Yeh, S. Lemeshow, and R. Glaser, Norepinephrine
up-regulates the expression of vascular endothelial growth
factor, matrix metalloproteinase (MMP)-2, and MMP-9 in
nasopharyngeal carcinoma tumor cells. Cancer Res, 2006.
66: p. 10357-64.
19.
Wang, H., D.R. Gomez, and Z. Liao, beta-Blockers and
metastasis in non-small-cell lung cancer. Expert Rev
Anticancer Ther, 2013. 13: p. 641-3.
20.
Pasquier, E., J. Ciccolini, M. Carre, S. Giacometti, R.
Fanciullino, C. Pouchy, M.P. Montero, C. Serdjebi, M.
Kavallaris, and N. Andre, Propranolol potentiates the anti-
angiogenic effects and anti-tumor efcacy of chemotherapy
agents: implication in breast cancer treatment. Oncotarget,
2011. 2: p. 797-809.
21. Coelho, M., M. Moz, G. Correia, A. Teixeira, R. Medeiros,
and L. Ribeiro, Antiproliferative effects of beta-blockers
on human colorectal cancer cells. Oncol Rep, 2015. 33:
p. 2513-20.
22.
Landen, C.N., Jr., Y.G. Lin, G.N. Armaiz Pena, P.D. Das,
J.M. Arevalo, A.A. Kamat, L.Y. Han, N.B. Jennings, W.A.
Spannuth, P.H. Thaker, S.K. Lutgendorf, C.A. Savary, A.M.
Sanguino, et al., Neuroendocrine modulation of signal
transducer and activator of transcription-3 in ovarian
cancer. Cancer Res, 2007. 67: p. 10389-96.
23.
Liao, X., X. Che, W. Zhao, D. Zhang, T. Bi, and G. Wang,
The beta-adrenoceptor antagonist, propranolol, induces
human gastric cancer cell apoptosis and cell cycle arrest
via inhibiting nuclear factor kappaB signaling. Oncol Rep,
2010. 24: p. 1669-76.
24.
Wolter, J.K., N.E. Wolter, A. Blanch, T. Partridge, L. Cheng,
D.A. Morgenstern, M. Podkowa, D.R. Kaplan, and M.S.
Irwin, Anti-tumor activity of the beta-adrenergic receptor
antagonist propranolol in neuroblastoma. Oncotarget,
2014. 5: p. 161-72.
25.
Lin, X., K. Luo, Z. Lv, and J. Huang, Beta-adrenoceptor
action on pancreatic cancer cell proliferation and tumor
growth in mice. Hepatogastroenterology, 2012. 59: p. 584-8.
26.
Powe, D.G., M.J. Voss, K.S. Zanker, H.O. Habashy, A.R.
Green, I.O. Ellis, and F. Entschladen, Beta-blocker drug
therapy reduces secondary cancer formation in breast
cancer and improves cancer specic survival. Oncotarget,
2010. 1: p. 628-38.
27.
De Giorgi, V., M. Grazzini, S. Gandini, S. Benemei, T.
Lotti, N. Marchionni, and P. Geppetti, Treatment with beta-
blockers and reduced disease progression in patients with
thick melanoma. Arch Intern Med, 2011. 171: p. 779-81.
28.
Calvani, M., F. Pelon, G. Comito, M.L. Taddei, S. Moretti,
S. Innocenti, R. Nassini, G. Gerlini, L. Borgognoni,
F. Bambi, E. Giannoni, L. Filippi, and P. Chiarugi,
Norepinephrine promotes tumor microenvironment
reactivity through beta3-adrenoreceptors during melanoma
progression. Oncotarget, 2015. 6: p. 4615-32.
29.
Albinana, V., K. Villar Gomez de Las Heras, G. Serrano-
Heras, T. Segura, A.B. Perona-Moratalla, M. Mota-Perez,
J.M. de Campos, and L.M. Botella, Propranolol reduces
viability and induces apoptosis in hemangioblastoma cells
from von Hippel-Lindau patients. Orphanet J Rare Dis,
2015. 10: p. 118.
30. Dey, M., J. Brisson, G. Davis, R. Enever, K. Pray, B. Zaim,
and D. Dvornik, Relationship between plasma propranolol
concentration and dose in young, healthy volunteers.
Biopharm Drug Dispos, 1986. 7: p. 103-11.
Oncotarget14
www.impactjournals.com/oncotarget
31.
Xie, W., H. Xie, F. Liu, W. Li, J. Dan, Y. Mei, L. Dan, X.
Xiao, J. Li, and X. Chen, Propranolol induces apoptosis
of human umbilical vein endothelial cells through
downregulation of CD147. Br J Dermatol, 2013. 168:
p. 739-48.
32.
Pasquier, E., J. Street, C. Pouchy, M. Carre, A.J.
Gifford, J. Murray, M.D. Norris, T. Trahair, N. Andre,
and M. Kavallaris, beta-blockers increase response to
chemotherapy via direct antitumour and anti-angiogenic
mechanisms in neuroblastoma. Br J Cancer, 2013. 108: p.
2485-94.
33.
Shakhar, G. and S. Ben-Eliyahu, In vivo beta-adrenergic
stimulation suppresses natural killer activity and
compromises resistance to tumor metastasis in rats. J
Immunol, 1998. 160: p. 3251-8.
34.
Huang, X.Y., H.C. Wang, Z. Yuan, J. Huang, and Q.
Zheng, Norepinephrine stimulates pancreatic cancer cell
proliferation, migration and invasion via beta-adrenergic
receptor-dependent activation of P38/MAPK pathway.
Hepatogastroenterology, 2012. 59: p. 889-93.
35.
Munabi, N.C., R.W. England, A.K. Edwards, A.A. Kitajewski,
Q.K. Tan, A. Weinstein, J.E. Kung, M. Wilcox, J.K.
Kitajewski, C.J. Shawber, and J.K. Wu, Propranolol Targets
Hemangioma Stem Cells via cAMP and Mitogen-Activated
Protein Kinase Regulation. Stem Cells Transl Med, 2015.
36.
Zhang, D., Q. Ma, Z. Wang, M. Zhang, K. Guo, F. Wang,
and E. Wu, beta2-adrenoceptor blockage induces G1/S
phase arrest and apoptosis in pancreatic cancer cells via
Ras/Akt/NFkappaB pathway. Mol Cancer, 2011. 10: p. 146.
37.
Burotto, M., V.L. Chiou, J.M. Lee, and E.C. Kohn, The
MAPK pathway across different malignancies: a new
perspective. Cancer, 2014. 120: p. 3446-56.
38.
Lott, J.P., Vemurafenib in melanoma with BRAF V600E
mutation. N Engl J Med, 2011. 365: p. 1449-50; author
reply 1450.
39.
Dalle, S., N. Poulalhon, and L. Thomas, Vemurafenib in
melanoma with BRAF V600E mutation. N Engl J Med,
2011. 365: p. 1448-9; author reply 1450.
40.
Chapman, P.B., A. Hauschild, C. Robert, J.B. Haanen, P.
Ascierto, J. Larkin, R. Dummer, C. Garbe, A. Testori, M.
Maio, D. Hogg, P. Lorigan, C. Lebbe, et al., Improved
survival with vemurafenib in melanoma with BRAF V600E
mutation. N Engl J Med, 2011. 364: p. 2507-16.
41.
Toy, E.L., F. Vekeman, M.C. Lewis, A.K. Oglesby, and M.S.
Duh, Costs, resource utilization, and treatment patterns
for patients with metastatic melanoma in a commercially
insured setting. Curr Med Res Opin, 2015. 31: p. 1561-72.
42.
Zhang, C., W. Spevak, Y. Zhang, E.A. Burton, Y. Ma, G.
Habets, J. Zhang, J. Lin, T. Ewing, B. Matusow, G. Tsang,
A. Marimuthu, H. Cho, et al., RAF inhibitors that evade
paradoxical MAPK pathway activation. Nature, 2015. 526:
p. 583-6.
43. Brady, D.C., M.S. Crowe, M.L. Turski, G.A. Hobbs, X. Yao,
A. Chaikuad, S. Knapp, K. Xiao, S.L. Campbell, D.J. Thiele,
and C.M. Counter, Copper is required for oncogenic BRAF
signalling and tumorigenesis. Nature, 2014. 509: p. 492-6.
44.
Johannessen, C.M., J.S. Boehm, S.Y. Kim, S.R. Thomas,
L. Wardwell, L.A. Johnson, C.M. Emery, N. Stransky, A.P.
Cogdill, J. Barretina, G. Caponigro, H. Hieronymus, R.R.
Murray, et al., COT drives resistance to RAF inhibition
through MAP kinase pathway reactivation. Nature, 2010.
468: p. 968-72.
45.
Giard, D.J., S.A. Aaronson, G.J. Todaro, P. Arnstein, J.H.
Kersey, H. Dosik, and W.P. Parks, In vitro cultivation of human
tumors: establishment of cell lines derived from a series of
solid tumors. J Natl Cancer Inst, 1973. 51: p. 1417-23.
46.
Brown, B.L., J.D. Albano, R.P. Ekins, and A.M. Sgherzi,
A simple and sensitive saturation assay method for the
measurement of adenosine 3’:5’-cyclic monophosphate.
Biochem J, 1971. 121: p. 561-2.
... In our study, propranolol inhibited the proliferation and promoted apoptosis of ovarian cancer cells through the AKT signaling pathway. Bcl-2 and Bax genes are the regulatory factors associated with the inhibition and promotion of apoptosis in the Bcl-2 protein family, respectively [10][11]. The death receptor often activates the caspase-3 mediated apoptotic pathway [12]. ...
... In the present study, propranolol signi cantly decreased Bcl-2 mRNA and signi cantly increased Bax, inducing caspase-3 expression thereby inhibiting the proliferation and promoting the apoptosis of SKVO-3 cells. Zhou et al. [10] displayed that melanoma cells treated with 100 µM propranolol for 24 h had signi cantly reduced viability, while Liao et al. [11] showed that propranolol could signi cantly inhibit the proliferation of gastric cancer cells. Shujun Zhao et al. [12] also reported that propranolol signi cantly reduced the viability of human ovarian cancer cell lines SKOV-3 and A2780 in a dose and time-dependent manner. ...
Preprint
Full-text available
Objective: This study aims to explore whether the effect of propranolol on the viability and apoptosis of ovarian cancer cells is mediated by inhibiting AKT signaling pathway. Methods: SKOV-3 ovarian cancer cell was treated with 0, 25, 50, 100, 200 and 400μM propranolol hydrochloride for 0, 24, 48, and 72 h, and then the cell viability was detected by CCK-8. After treating SKOV-3 cells with 0, 50, 100, and 200μM propranolol for 24h, Q-PCR detected the mRNA levels of BCL-2, BAX, and AKT. Western blotwas employed to detect the protein expression of caspase-3 and AKT. The activator N-oleoylglycine and the inhibitor A2D5363 were combined with propranolol to treat SKOV-3 cells for 24h. Western blot detected the protein expression of AKT and CCK-8 method was used to detect cell proliferation. Results: Propranolol inhibited the proliferation of SKOV-3 cells in a concentration- and time-dependent manner. In addition, propranolol promoted the expressions of BAX and caspase-3 and inhibited BCL-2 and AKT. Propranolol combined with AKT inhibitor A2D5363 enhanced the propranolol anti-ovarian cancer effect and the combined with AKT activator N-Oleoylglycine reduced the anti-ovarian cancer effect of propranolol. Conclusions: Propranolol-induced apoptosis of ovarian cancer is mediated by inhibiting the AKT signaling pathway.
... An estimated ±100,350 new cases of melanoma and 6,850 deaths were reported in the US in 2020 (Hwang et al., 2020;Ahmed et al., 2021). MM originates from pigment-bearing cells in the basal layer of the epidermis known as melanocytes (Zhou et al., 2016;Saginala et al., 2021). Melanocytes originate from the neural crest and express many molecules and signaling factors that promote migration and metastasis (Saginala et al., 2021). ...
Article
Full-text available
Melanoma skin cancer is a malignant melanocyte tumor considered the most invasive and dangerous skin cancer, with an average five-year survival rate of less than 5% after metastasis. Thus, a new strategy for preventing and treating cancer from the natural product is required. Medicinal plants are the potential as an alternative against cancer. This review article aims to determine natural products from medicinal plants which have the potential as an anticancer in melanoma skin cancer in vitro and in vivo. 40 plants have been selected based on the selection criteria for anticancer compounds. In vitro studies showed that the plant can reduce cell viability through cell cycle inhibition and apoptosis induction and inhibit angiogenesis, invasion, and metastasis in human melanoma skin cancer. Therefore, further research is required to explore more plants, especially medicinal plants, their active compounds, and the mechanism of anticancer action to be used as standard herbal medicines.
... 15 A previous study showed that cancer cells treated with PRO can undergo cell cycle arrest at different cell cycle checkpoints, e.g., G0/G1, S/M, or G1/S. 16 These findings, along with ours, suggest that PRO can exert its anti-proliferative and anti-apoptotic effects by arresting cancer cells at several cell cycle checkpoints, thereby reducing the cancer cells' potential for avoiding ultimate cell death. Lastly, the mechanisms underlying the anti-tumorigenic actions of PRO have been extensively studied in the literature through various preclinical and clinical cancer models. ...
Article
Full-text available
Objective: Propranolol (PRO), a non-selective beta-adrenergic receptor inhibitor, has been recently discovered to possess anti-tumorigenic effects in cancer patients. There-fore, we aimed to investigate the in vitro effects of PRO in A549-derived lung cancer spheroids in terms of cell viability, spheroid formation, cell cycle regulation, cell differen-tiation, and apoptosis. Materials and Methods: The effect of 24-hour PRO treatment on A549 cell viability was assessed using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. A sub-cytotoxic PRO concentration (125 µM) was employed to evaluate its impact on the clonogenicity of A549-derived cancer spheroids after seven days of incubation. Messenger Ribonucleic Acid (mRNA) levels of cell cycle regulators including cyclin-dependent kinase inhibitor 1A (p21) and G2 checkpoint kinase (WEE1), apoptotic markers such as caspases 3, 8, 9 (CASP3, CASP8, CASP9), and stem cell differentiation markers, namely POU class 5 ho-meobox 1 (octamer-binding transcription factor 4 (OCT4)), prominin 1 (CD133), and adenos-ine triphosphate (ATP) binding cassette subfamily G member 2 (ABCG2) were measured us-ing reverse transcription quantitative polymerase chain reaction (RT-qPCR) after a 24-hour treatment of cancer spheroids with PRO. Results: PRO treatment reduced cell viability and inhibited the clonogenicity of cancer spheroids by activating intrinsic apoptotic markers CASP3 and CASP9, leading to cell cy-cle arrest via increased p21 expression. PRO did not significantly alter stem cell differen-tiation markers. Conclusion: The proliferation and clonogenic activity of lung cancer spheroids can be ef-fectively suppressed with PRO, primarily through inducing intrinsic apoptosis following p21-mediated cell cycle arrest. While short-term PRO exposure did not affect the gene ex-pression levels of stem cell differentiation markers, the notable decrease in both cell viability and spheroid formation efficiency suggests the potential of PRO as a therapeutic drug in lung cancer treatment.
... Hepatitis C is an RNA-containing virus (HCV) that often infects the liver cells, leading to liver cirrhosis and cancer [98]. HCV induces different cellular inflammatory molecules like IL-6 and TNF by activating JAK-STAT, MAPK, and PI3K-Akt signaling circuits in cells that promote tumor formation further [99][100][101][102]. Cervical cancer is another example of virus-mediated inflammation and malignancy. ...
Article
Full-text available
Among the most extensively researched processes in the development and treatment of cancer is inflammatory condition. Although acute inflammation is essential for the wound healing and reconstruction of tissues that have been damaged, chronic inflammation may contribute to the onset and growth of a number of diseases, including cancer. By disrupting the signaling processes of cells, which result in cancer induction, invasion, and development, a variety of inflammatory molecules are linked to the development of cancer. The microenvironment surrounding the tumor is greatly influenced by inflammatory cells and their subsequent secretions, which also contribute significantly to the tumor's growth, survivability, and potential migration. These inflammatory variables have been mentioned in several publications as prospective diagnostic tools for anticipating the onset of cancer. Targeting inflammation with various therapies can reduce the inflammatory response and potentially limit or block the proliferation of cancer cells. The scientific medical literature from the past three decades has been studied to determine how inflammatory chemicals and cell signaling pathways related to cancer invasion and metastasis are related. The current narrative review updates the relevant literature while highlighting the specifics of inflammatory signaling pathways in cancer and their possible therapeutic possibilities.
... Since stress-related catecholamines have been shown to maintain the progression of melanoma, the possibility of using b-blockers as a therapeutic intervention for melanoma drugs in cancer is gaining attention. Despite many studies that confirm the antitumor effect of other non-selective bblocker propranolol against melanoma cells (Zhou et al., 2016), reports of the effects on these cells are limited. The current study establishes the growth inhibitory effect of carvedilol at concentrations ! 5 lM on C32 amelanotic cells and A2058 melanotic melanoma cells after 72 h of treatment. ...
Article
Full-text available
Melanoma is an aggressive skin cancer. Increasing evidence has shown the role of β-adrenergic receptors in the pathogenesis of melanoma. Carvedilol is a widely used non-selective β-AR antagonist with potential anticancer activity. The purpose of the study was to estimate the influence of carvedilol and sorafenib alone and in combination on the growth and inflammatory response of C32 and A2058 melanoma cells. Furthermore, this study also aimed to predict the probable interaction of carvedilol and sorafenib when administered together. Predictive study of the interaction of carvedilol and sorafenib was performed using the ChemDIS-Mixture system. Carvedilol and sorafenib alone and in combination showed a growth inhibitory effect on cells. The greatest synergistic antiproliferative effect on both cell lines was observed at Car 5 μM combined with Sor 5 μM. Analysis in silico identified diseases, proteins, and metabolic pathways that can be affected by the interaction of carvedilol and sorafenib. The results obtained demonstrated that carvedilol and sorafenib modulated the secretion of IL-8 by IL-1β-stimulated by melanoma cell lines but the use of a combination of both drugs did not intensify the effect. In summary, the results presented indicate that the combination of carvedilol and sorafenib may have a promising anticancer effect on melanoma cells.
... Previous studies on propranolol-induced changes in gene expression in endothelial cells also included genes involved in lipid and sterol metabolism and ubiquitination [17]. Propranolol has also been shown to affect various other host factors and signaling pathways, including inhibition of the RAS/RAF/ERK and AKT pathways [18,19]. The inhibition of factors involved in these signaling pathways was also shown to affect SARS-CoV-2 replication [20]. ...
Article
Full-text available
The SARS-CoV-2 pandemic highlighted the need for broad-spectrum antivirals to increase our preparedness. Patients often require treatment by the time that blocking virus replication is less effective. Therefore, therapy should not only aim to inhibit the virus, but also to suppress pathogenic host responses, e.g., leading to microvascular changes and pulmonary damage. Clinical studies have previously linked SARS-CoV-2 infection to pathogenic intussusceptive angiogenesis in the lungs, involving the upregulation of angiogenic factors such as ANGPTL4. The β-blocker propranolol is used to suppress aberrant ANGPTL4 expression in the treatment of hemangiomas. Therefore, we investigated the effect of propranolol on SARS-CoV-2 infection and the expression of ANGPTL4. SARS-CoV-2 upregulated ANGPTL4 in endothelial and other cells, which could be suppressed with R-propranolol. The compound also inhibited the replication of SARS-CoV-2 in Vero-E6 cells and reduced the viral load by up to ~2 logs in various cell lines and primary human airway epithelial cultures. R-propranolol was as effective as S-propranolol but lacks the latter’s undesired β-blocker activity. R-propranolol also inhibited SARS-CoV and MERS-CoV. It inhibited a post-entry step of the replication cycle, likely via host factors. The broad-spectrum antiviral effect and suppression of factors involved in pathogenic angiogenesis make R-propranolol an interesting molecule to further explore for the treatment of coronavirus infections.
... No data are available for the IC50 value of propranolol in the Hep-2 cell line. The IC50 value of propranolol for other cell lines can vary in a wide range between 65.33 and 466.11 μM [19]. ...
Article
Full-text available
PurposeWe aimed to evaluate the effects of thymoquinone and propranolol on Hep-2 cells representing laryngeal Ca cell type in comparison with cisplatin. We also evaluated their combined effects.Methods Apoptotic effects were directly analyzed via mitochondrial membrane potential and caspase-3 assays. In addition, effects on apoptosis and cell cycle via Bcl-2, Bax, P53, and Cyclin D1 mRNA expressions and effects on angiogenesis via VEGFA mRNA expression were evaluated by RT-qPCR.ResultsAccording to our results, it was determined that the anticancer effects of thymoquinone on Hep-2 cells were higher than propranolol. Our JC-1 and caspase-3 results showed an effect close to cisplatin, especially for 50 µM thymoquinone. Significant differences were also obtained in Bcl-2, Bax, P53, and cyclin D1 results for similar concentrations compared to the control. No effect of thymoquinone was seen for VEGFA. Propranolol alone had no significant effect on JC-1 and Caspase-3. Propranolol had an effect on Bcl-2, Bax mRNA expressions compared to the control, only at 250 µM concentration. Propranolol and its combinations increased VEGFA mRNA expression-like cisplatin.Conclusion Thymoquinone induced apoptosis and blocked the cell cycle in Hep-2 cells. The effects of propranolol, which was reported to have an antiangiogenesis effect in some studies, on apoptosis and cell cycle were limited except at high concentrations. For this cell line, why propranolol causes an increase in VEGFA expression should be evaluated extensively. Thymoquinone shows promise for cancer therapy, but studies need to be designed in vivo to evaluate the effects more reliably.
Article
Full-text available
Heart failure (HF) patients have a significantly higher risk of new-onset cancer and cancer-associated mortality, compared to subjects free of HF. While both the prevention and treatment of new-onset HF in patients with cancer have been investigated extensively, less is known about the prevention and treatment of new-onset cancer in patients with HF, and whether and how guideline-directed medical therapy (GDMT) for HF should be modified when cancer is diagnosed in HF patients. The purpose of this review is to elaborate and discuss the effects of pillar HF pharmacotherapies, as well as digoxin and diuretics on cancer, and to identify areas for further research and novel therapeutic strategies. To this end, in this review, (i) proposed effects and mechanisms of action of guideline-directed HF drugs on cancer derived from pre-clinical data will be described, (ii) the evidence from both observational studies and randomized controlled trials on the effects of guideline-directed medical therapy on cancer incidence and cancer-related outcomes, as synthetized by meta-analyses will be reviewed, and (iii) considerations for future pre-clinical and clinical investigations will be provided.
Article
Full-text available
Compelling evidence supports the hypothesis that stress negatively impacts cancer development and prognosis. Irrespective of its physical, biological or psychological source, stress triggers a physiological response that is mediated by the hypothalamic-pituitary-adrenal axis and the sympathetic adrenal medullary axis. The resulting release of glucocorticoids and catecholamines into the systemic circulation leads to neuroendocrine and metabolic adaptations that can affect immune homeostasis and immunosurveillance, thus impairing the detection and eradication of malignant cells. Moreover, catecholamines directly act on β-adrenoreceptors present on tumor cells, thereby stimulating survival, proliferation, and migration of nascent neoplasms. Numerous preclinical studies have shown that blocking adrenergic receptors slows tumor growth, suggesting potential clinical benefits of using β-blockers in cancer therapy. Much of these positive effects of β-blockade are mediated by improved immunosurveillance. The present trial watch summarizes current knowledge from preclinical and clinical studies investigating the anticancer effects of β-blockers either as standalone agents or in combination with conventional antineoplastic treatments or immunotherapy.
Article
The recently uncovered key role of the peripheral and central nervous systems in controlling tumorigenesis and metastasis has opened a new area of research to identify innovative approaches against cancer. Although the 'neural addiction' of cancer is only partially understood, in this Perspective we discuss the current knowledge and perspectives on peripheral and central nerve circuitries and brain areas that can support tumorigenesis and metastasis and the possible reciprocal influence that the brain and peripheral tumours exert on one another. Tumours can build up local autonomic and sensory nerve networks and are able to develop a long-distance relationship with the brain through circulating adipokines, inflammatory cytokines, neurotrophic factors or afferent nerve inputs, to promote cancer initiation, growth and dissemination. In turn, the central nervous system can affect tumour development and metastasis through the activation or dysregulation of specific central neural areas or circuits, as well as neuroendocrine, neuroimmune or neurovascular systems. Studying neural circuitries in the brain and tumours, as well as understanding how the brain communicates with the tumour or how intratumour nerves interplay with the tumour microenvironment, can reveal unrecognized mechanisms that promote cancer development and progression and open up opportunities for the development of novel therapeutic strategies. Targeting the dysregulated peripheral and central nervous systems might represent a novel strategy for next-generation cancer treatment that could, in part, be achieved through the repurposing of neuropsychiatric drugs in oncology.
Article
Full-text available
Significance: The present study investigated the action of propranolol in infantile hemangiomas (IHs). IHs are the most common vascular tumor in children and have been proposed to arise from a hemangioma stem cell (HemSC). Propranolol, a nonselective β-adrenergic receptor (βAR) antagonist, has proven efficacy; however, understanding of its mechanism of action on HemSCs is limited. The presented data demonstrate that propranolol, via βAR perturbation, dose dependently suppresses cAMP levels and activated extracellular signal-regulated kinase 1/2. Furthermore, propranolol acts via perturbation of β2AR, and not β1AR, although both receptors are expressed in HemSCs. These results provide important insight into propranolol's action in IHs and can be used to guide the development of more targeted therapy.
Article
Full-text available
Background: Von Hippel-Lindau (VHL) disease is a rare oncological disease with an incidence of 1:36,000, and is characterized by the growth of different types of tumors: hemangioblastomas in the central nervous system (CNS) and retina, renal carcinoma, pheochromocytomas, pancreatic serous cystadenoma, and endolymphatic sac tumors. These tumors do not express VHL protein (pVHL). pVHL ubiquitinates hypoxia inducible factor (HIF) protein for degradation by the proteasome; in the absence of VHL, HIF translocates to the nucleus to activate the expression of its target genes. Targeting VHL-derived tumors with drugs that have reduced side effects is urgent to avoid repeat CNS surgeries. Recent reports have shown that propranolol, a β-blocker used for the treatment of hypertension and other cardiac and neurological diseases, is the best option for infantile hemangioma (IH). Propranolol could be an efficient treatment to control hemangioblastoma growth in VHL disease because of its antiangiogenic effects demonstrated in IH and the hypothetical impact on HIF levels. Methods: HeLa 9X (HRE) hypoxia responsive element cell line and primary hemangioblastoma-derived cells were subjected to propranolol treatment and cell viability and apoptosis were evaluated. HIF1-α and Hif-2α expression after propranolol treatment was analyzed by western blotting. Quantitative PCR was performed to study the mRNA expression of HIF target genes. Vascular endothelial growth factor (VEGF) was measured in culture supernatants by immunoassay. Results: Propranolol downregulated HIF-dependent transcription in HeLa 9XHRE cells. Under hypoxic conditions, propranolol decreased the expression of HIF target genes in hemangioblastoma cells, which stopped proliferating and died following long-term treatment. These results suggests that propranolol treatment promoted reduced HIF protein expression and corresponding downregulation of HIF target genes, and inhibited cell proliferation in parallel with induction of cell death by apoptosis. Conclusions: Our results suggest that propranolol could reduce the growth of HIF-dependent tumors and may thus be a promising treatment to delay surgery in VHL patients.
Article
Full-text available
Colon cancer is the fourth and third most common cancer, respectively in men and women worldwide and its incidence is on the increase. Stress response has been associated with the incidence and development of cancer. The catecholamines (CA), adrenaline (AD) and noradrenaline (NA), are crucial mediators of stress response, exerting their effects through interaction with α- and β-adrenergic receptors (AR). Colon cancer cells express β-AR, and their activation has been implicated in carcinogenesis and tumor progression. Interest concerning the efficacy of β-AR blockers as possible additions to cancer treatment has increased. The aim of this study was to investigate the effect of several AR agonists and β-blockers following cell proliferation of HT-29 cells, a human colon adenocarcinoma cell line. For this purpose, HT-29 cells were incubated in the absence (control) or in the presence of the AR-agonists, AD, NA and isoprenaline (ISO) (0.1-100 µM) for 12 or 24 h. The tested AR agonists revealed proliferative effects on HT-29 cells. In order to study the effect of several β-blockers following proliferation induced by AR activation, the cells were treated with propranolol (PRO; 50 µM), carvedilol (CAR; 5 µM), atenolol (ATE; 50 µM), or ICI 118,551 (ICI; 5 µM) for 45 min prior, and simultaneously, to incubation with each of the AR agonists, AD and ISO, both at 1 and 10 µM. The results suggested that adrenergic activation plays an important role in colon cancer cell proliferation, most probably through β-AR. The β-blockers under study were able to reverse the proliferation induced by AD and ISO, and some of these blockers significantly decreased the proliferation of HT-29 cells. The elucidation of the intracellular pathways involved in CA-induced proliferation of colon cancer cells, and in the reversion of this effect by β-blockers, may contribute to identifying promising strategies in cancer treatment.
Article
Full-text available
Oral propranolol has been used to treat complicated infantile hemangiomas, although data from randomized, controlled trials to inform its use are limited. We performed a multicenter, randomized, double-blind, adaptive, phase 2-3 trial assessing the efficacy and safety of a pediatric-specific oral propranolol solution in infants 1 to 5 months of age with proliferating infantile hemangioma requiring systemic therapy. Infants were randomly assigned to receive placebo or one of four propranolol regimens (1 or 3 mg of propranolol base per kilogram of body weight per day for 3 or 6 months). A preplanned interim analysis was conducted to identify the regimen to study for the final efficacy analysis. The primary end point was success (complete or nearly complete resolution of the target hemangioma) or failure of trial treatment at week 24, as assessed by independent, centralized, blinded evaluations of standardized photographs. Of 460 infants who underwent randomization, 456 received treatment. On the basis of an interim analysis of the first 188 patients who completed 24 weeks of trial treatment, the regimen of 3 mg of propranolol per kilogram per day for 6 months was selected for the final efficacy analysis. The frequency of successful treatment was higher with this regimen than with placebo (60% vs. 4%, P<0.001). A total of 88% of patients who received the selected propranolol regimen showed improvement by week 5, versus 5% of patients who received placebo. A total of 10% of patients in whom treatment with propranolol was successful required systemic retreatment during follow-up. Known adverse events associated with propranolol (hypoglycemia, hypotension, bradycardia, and bronchospasm) occurred infrequently, with no significant difference in frequency between the placebo group and the groups receiving propranolol. This trial showed that propranolol was effective at a dose of 3 mg per kilogram per day for 6 months in the treatment of infantile hemangioma. (Funded by Pierre Fabre Dermatologie; ClinicalTrials.gov number, NCT01056341.).
Article
Oncogenic activation of BRAF fuels cancer growth by constitutively promoting RAS-independent mitogen-activated protein kinase (MAPK) pathway signalling. Accordingly, RAF inhibitors have brought substantially improved personalized treatment of metastatic melanoma. However, these targeted agents have also revealed an unexpected consequence: stimulated growth of certain cancers. Structurally diverse ATP-competitive RAF inhibitors can either inhibit or paradoxically activate the MAPK pathway, depending whether activation is by BRAF mutation or by an upstream event, such as RAS mutation or receptor tyrosine kinase activation. Here we have identified next-generation RAF inhibitors (dubbed 'paradox breakers') that suppress mutant BRAF cells without activating the MAPK pathway in cells bearing upstream activation. In cells that express the same HRAS mutation prevalent in squamous tumours from patients treated with RAF inhibitors, the first-generation RAF inhibitor vemurafenib stimulated in vitro and in vivo growth and induced expression of MAPK pathway response genes; by contrast the paradox breakers PLX7904 and PLX8394 had no effect. Paradox breakers also overcame several known mechanisms of resistance to first-generation RAF inhibitors. Dissociating MAPK pathway inhibition from paradoxical activation might yield both improved safety and more durable efficacy than first-generation RAF inhibitors, a concept currently undergoing human clinical evaluation with PLX8394.
Article
Background Oral propranolol has been used to treat complicated infantile hemangiomas, although data from randomized, controlled trials to inform its use are limited. Methods We performed a multicenter, randomized, double-blind, adaptive, phase 2–3 trial assessing the efficacy and safety of a pediatric-specific oral propranolol solution in infants 1 to 5 months of age with proliferating infantile hemangioma requiring systemic therapy. Infants were randomly assigned to receive placebo or one of four propranolol regimens (1 or 3 mg of propranolol base per kilogram of body weight per day for 3 or 6 months). A preplanned interim analysis was conducted to identify the regimen to study for the final efficacy analysis. The primary end point was success (complete or nearly complete resolution of the target hemangioma) or failure of trial treatment at week 24, as assessed by independent, centralized, blinded evaluations of standardized photographs. Results Of 460 infants who underwent randomization, 456 received tr...
Article
To estimate real-world healthcare costs, resource utilization, and treatment patterns among metastatic melanoma (MM) patients who received a therapy recommended in current treatment guidelines during 2011 and 2012, following approval in the US of novel therapies (ipilimumab and vemurafenib). Administrative claims data were used in a retrospective, longitudinal, open cohort study. Adult MM patients were identified using ICD-9 codes. Therapy-based patient cohorts and index dates were defined by the first receipt of a therapy of interest: ipilimumab, vemurafenib, paclitaxel (alone and in combination), interleukin-2, dacarbazine (alone and in combination), or temozolomide. The follow-up period extended until the end of eligibility or data availability. A multivariate regression model was used to compare outcomes of the ipilimumab and vemurafenib cohorts, controlling for baseline and demographic characteristics. Direct healthcare costs (2013 US dollars) and utilization (incidence rates) were measured on a per-patient-per-month (PPPM) basis for each treatment cohort. Treatment patterns were assessed, including the frequency of patients receiving a second therapy of interest. The study population included 834 patients (265 in the ipilimumab cohort, 234 vemurafenib, 174 paclitaxel, 104 interleukin-2, 46 dacarbazine, and 11 temozolomide). Costs ranged from $10,879 PPPM (temozolomide) to $35,472 PPPM (ipilimumab). Adjusted total costs were $18,337 PPPM higher for ipilimumab vs. vemurafenib cohorts (p<0.001), primarily due to higher outpatient costs. Multivariate analysis did not find significant differences in resource utilization between ipilimumab and vemurafenib, except ipilimumab patients had fewer outpatient visits (excluding treatment visits). Ipilimumab and vemurafenib patients received a second therapy of interest (12% and 11%, respectively) less frequently than interleukin-2 and dacarbazine patients. The cost and resource utilization burden of MM is high and varies substantially across treatment cohorts. The two novel therapies, ipilimumab and vemurafenib, have quickly been adopted and are the most frequently used therapies. The results observed during the approximately 6-month follow-up period may not be representative of the full clinical experience of patients with MM.
Article
Previously, a study of ours showed that the combination of dabrafenib and trametinib improves progression-free survival compared with dabrafenib and placebo in patients with BRAF Val600Lys/Glu mutation-positive metastatic melanoma. The study was continued to assess the secondary endpoint of overall survival, which we report in this Article. We did this double-blind phase 3 study at 113 sites in 14 countries. We enrolled previously untreated patients with BRAF Val600Glu or Val600Lys mutation-positive unresectable stage IIIC or stage IV melanoma. Participants were computer-randomised (1:1) to receive a combination of dabrafenib (150 mg orally twice daily) and trametinib (2 mg orally once daily), or dabrafenib and placebo. The primary endpoint was progression-free survival and overall survival was a secondary endpoint. This study is registered with ClinicalTrials.gov, number NCT01584648. Between May 4, 2012, and Nov 30, 2012, we screened 947 patients for eligibility, of whom 423 were randomly assigned to receive dabrafenib and trametinib (n=211) or dabrafenib only (n=212). The final data cutoff was Jan 12, 2015, at which time 222 patients had died. Median overall survival was 25·1 months (95% CI 19·2-not reached) in the dabrafenib and trametinib group versus 18·7 months (15·2-23·7) in the dabrafenib only group (hazard ratio [HR] 0·71, 95% CI 0·55-0·92; p=0·0107). Overall survival was 74% at 1 year and 51% at 2 years in the dabrafenib and trametinib group versus 68% and 42%, respectively, in the dabrafenib only group. Based on 301 events, median progression-free survival was 11·0 months (95% CI 8·0-13·9) in the dabrafenib and trametinib group and 8·8 months (5·9-9·3) in the dabrafenib only group (HR 0·67, 95% CI 0·53-0·84; p=0·0004; unadjusted for multiple testing). Treatment-related adverse events occurred in 181 (87%) of 209 patients in the dabrafenib and trametinib group and 189 (90%) of 211 patients in the dabrafenib only group; the most common was pyrexia (108 patients, 52%) in the dabrafenib and trametinib group, and hyperkeratosis (70 patients, 33%) in the dabrafenib only group. Grade 3 or 4 adverse events occurred in 67 (32%) patients in the dabrafenib and trametinib group and 66 (31%) patients in the dabrafenib only group. The improvement in overall survival establishes the combination of dabrafenib and trametinib as the standard targeted treatment for BRAF Val600 mutation-positive melanoma. Studies assessing dabrafenib and trametinib in combination with immunotherapies are ongoing. GlaxoSmithKline. Copyright © 2015 Elsevier Ltd. All rights reserved.
Article
The BRAF serine/threonine kinase is mutated, typically at V600, to remain in the active oncogenic state in a large fraction of melanoma, thyroid cancers, and hairy cell leukemia, and to a lesser extent in a wide spectrum of other cancers, thereby activating the kinases MEK1 and MEK2 to stimulate the MAPK pathway and promote cancer. Excitingly, ATP inhibitors of oncogenic BRAF and MEK provide a survival advantage in metastatic melanoma and early clinical studies suggest that coupling BRAF and MEK kinase inhibitors may be even more effective. Thus, the combination of multiple approaches to inhibit MAPK signaling holds great promise for the treatment of BRAF mutation-positive cancers, especially in terms of overcoming resistance. In this regard, we previously found that copper (Cu) influx enhanced MEK1 phosphorylation of its substrates ERK1/2 through a Cu-MEK1 interaction. We show here that genetic loss of the high affinity Cu transporter Ctr1 or mutations in MEK1 that disrupt Cu binding reduced MAPK signaling and oncogenic BRAFV600E-mediated tumorigenesis, which could be rescued by expressing activated ERK2. Importantly, a Cu chelator used in the treatment of Wilson's disease reduced tumor growth of not only BRAFV600E-transformed cells, but also cells resistant to a BRAF inhibitor. Taken together, these results suggest that Cu-chelation therapy could be repurposed for the treatment of BRAFV600E mutation-positive cancers. Citation Format: Donita C. Brady, Matthew S. Crowe, Michelle L. Turski, Dennis J. Thiele, Christopher M. Counter. Copper is required for oncogenic BRAF signaling and tumorigenesis. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4294. doi:10.1158/1538-7445.AM2013-4294