Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Received: 11 November 2019 | Revised: 15 January 2020 | Accepted: 9 February 2020

DOI: 10.1111/jcmm.15156

ORIGINAL ARTICLE

CDK4/6 inhibition suppresses tumour growth and enhances


the effect of temozolomide in glioma cells

Yingxiao Cao1 | Xin Li2 | Shiqi Kong1 | Shuling Shang3 | Yanhui Qi4

1
Department of Neurosurgery, Xingtai
People’s Hospital, Xingtai, China Abstract
2
Department of Neurosurgery, The First In adults, glioma is the most commonly occurring and invasive brain tumour. For ma-
People's Hospital of Shenyang, Shenyang,
lignant gliomas, the current advanced chemotherapy includes TMZ (temozolomide).
China
3
Department of Operating Room, Xingtai
However, a sizeable number of gliomas are unyielding to TMZ, hence, giving rise to
People’s Hospital, Xingtai, China an urgent need for more efficient treatment choices. Here, we report that cyclin-
4
Department of Intensive Care Unit, Xingtai dependent kinases 4 (CDK4) is expressed at significantly high levels in glioma cell
People’s Hospital, Xingtai, China
lines and tissues. CDK4 overexpression enhances colony formation and proliferation
Correspondence of glioma cells and extends resistance to inhibition of TMZ-mediated cell proliferation
Yingxiao Cao, Department of Neurosurgery,
Xingtai People’s Hospital, Xingtai, Hebei and induction of apoptosis. However, CDK4 knockdown impedes colony formation
054031, China. and cell proliferation, and enhances sensitivity of glioma cells to TMZ. The selective
Email: caoyingxiao2019@163.com
inhibition of CDK4/6 impedes glioma cell proliferation and induces apoptotic induc-
tion. The selective inhibitors of CDK4/6 may enhance glioma cell sensitivity to TMZ.
We further showed the possible role of RB phosphorylation mediated by CDK4 for
its oncogenic function in glioma. The growth of glioma xenografts was inhibited in
vivo, through combination treatment, and corresponded to enhanced p-RB levels,
reduced staining of Ki-67 and enhanced activation of caspase 3. Therefore, CDK4
inhibition may be a favourable strategy for glioma treatment and overcomes TMZ
resistance.

KEYWORDS

CDK4/6, drug resistance, glioma, p-RB, temozolomide

1 | I NTRO D U C TI O N in treatment of glioma, the overall survival (OS) of glioma patients is


only 1-1.25 years.5
An important category of human brain tumours, gliomas, are the most Temozolomide (TMZ), an alkylating agent, is currently used as
invasive and fatal brain tumour of adulthood that are well-differenti- a first-line drug for treatment of glioma including GBM.6 TMZ leads
ated astrocytomas of low-grade, anaplastic astrocytomas and GBM to guanine methylation in the DNA at position O6, causing incorrect
1
(glioblastoma multiforme). The most aggressive subtype among the pairing of O6-methylguanine with thymine, activating the mismatch
gliomas is GBM with the poorest rates of survival.2 The commonly ap- repair system.7 This results in a break in the double-strand genome
plied treatments are chemotherapy, surgical resection and radiother- that causes cell cycle arrest and apoptotic induction.6,8 TMZ resistance
apy.3,4 Although the past few years have seen significant advancement is induced by MGMT (O6-methylguanine-DNA methyltransferase)

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

J Cell Mol Med. 2020;24:5135–5145.  wileyonlinelibrary.com/journal/jcmm | 5135


|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5136 CAO et al.

by eliminating methylation from guanine at O6 position.9 Therefore, of CDK4 deacetylase by specific CDK4 inhibitors impedes prolifer-
patients with methylated MGMT promoter and silenced MGMT ex- ation and enhances apoptosis in glioma cell lines. The glioma cells
pression showed a better median and 2-year survival through com- can be sensitized to TMZ by inhibitors of CDK4. Our outcome here
bined radiotherapy and TMZ chemotherapy.10 In patients with GBM indicates that CDK4 inhibition may be a possible treatment strategy
resistant to TMZ, resistance was anticipated to be inhibited by MGMT for glioma. In this study, we evaluated the effect of CDK4 overex-
pseudo-substrates through MGMT depletion, although no significant pression and its inhibition in glioma cells in vitro and in vivo, and
restoration of TMZ sensitivity was observed in clinical trials.11 Thus, determined the characteristics of glioma cell such as apoptosis and
new strategies are needed to sensitize patients to the efficiency of associated protein markers.
chemotherapy using TMZ.
The common features in several types of cancer, including mela-
noma, include constitutive CDK (cyclin-dependent kinases) activation 2 | M ATE R I A L S A N D M E TH O DS
and of cell cycle deregulation.12,13 In 38% of cancer, there is a deletion
of a tumour suppressor and a negative regulator of CDK4, P16INK4a.14 2.1 | Collection of samples from patients
Furthermore, there are CDK4 gene amplification and mutations in ger-
mline in melanoma, causing unhindered CDK4 activity and enhanced Between 2015 and 2018, tissues from normal brain (n = 6) and gli-
cell proliferation.15 CDK6 is regulated by cyclin, more specifically by cy- oma tissues of high grade (n = 12) were collected during surgery from
clin D protein and CDK inhibitor protein. CDK6 is overexpressed in can- Xingtai People's Hospital. The samples were frozen immediately in
cers including leukaemia, melanoma, medulloblastoma and lymphoma, liquid nitrogen till used further. The ethics committee of Xingtai
which associated with chromosomal rearrangements. Generally, the People's Hospital gave consent to the project.
cyclins control the cell cycle entry in proliferating adult mammalian cells
by binding and activating CDK4 and CDK6 and facilitation of retinoblas-
toma (RB) phosphorylation and G1 to S transition.16 As a result, CDK4 2.2 | Cell culture
activity deregulation correlated with tumorigenesis, although, there
are only rare reports of studies on the function of CDK4 in glioma.17 The ATCC (American Type Culture Collection) provided us with the
Palbociclib (PD0332991), a specific CDK4/6 inhibitor, was developed to cell lines for human glioma (U87, U251, H4 and A172). NHAs (Normal
18
arrest the cell cycle progression in proliferating tumour cells. It proved human astrocytes) were procured from Lonza (Switzerland) and cul-
to be beneficial especially in tumours lacking p16INK4a or overexpress- tured as per supplied instructions. These cell lines were maintained in
ing cyclin D, such as bladder and gastric cancers, while those without DMEM (HyClone) containing FBS (10%) and penicillin-streptomycin
functional RB1 have been refractory to palbociclib treatment.19 Upon (1%).
promising results in preclinical models of various cancers including
GBM, palbociclib was tested in several clinical trials in phase I/II and
has been approved by the FDA in combination with anti-oestrogen 2.3 | Assay to detect colony formation
therapies against hormone receptor-positive breast cancers.20,21 These
clinical studies indicate that as a single agent, palbociclib fails to provide Seeding of cells was done in 6-well plates at of the rate of 500 cells
durable responses, potentially due at least in part to tumour adaptation, in each well. Post-adhesion, treatment was done with TMZ with or
and suggesting a need of combination with other agents.22 Previously without CDK inhibitors. Every three days, renewal of the culture
study has shown that pharmacologic inhibition of cyclin-dependent ki- medium was done. After two weeks, the cells were crystal violet
nases 4 and 6 arrests the growth of glioblastoma multiforme intracranial stained and imaged.
xenografts.23 In addition, palbociclib significantly prolongs survival in a
genetically engineered mouse model of brainstem glioma.24 Previously
studies also have shown that combining CDK4/6 and mTOR inhibitors 2.4 | CCK-8 assay
offers increased benefits against glioma through a number of mech-
anisms, including blockade of compensatory signalling mechanisms, The cells cultured in 96-well plates were treated with the specified
improved brain penetration of palbociclib, enhanced metabolic effects compounds, or transfected with indicated siRNA or plasmids, and at
and cytostatic to cytotoxic conversion.25 Although modifications in indicated times, measurement of cell proliferation was done using
several cell cycle regulators, such as activation of CDK2, amplification CCK-8 kit (Sigma).
of cyclin D and loss of p21CIP1 or p27KIP1, may contribute to tumour
adaptation, the main resistance to palbociclib treatment is mediated by
RB1 inactivation.26 We therefore sought a combination regimen using 2.5 | Detection of apoptosis
CDK4/6 inhibitors that inhibits overall cell cycle progression.
In this study, we report the overexpression of CDK4 in glioma tis- The apoptosis assay was performed as previously study. 27,28
sues and cell lines, promoting proliferation of glioma and formation Briefly, cultured cells in six-well plates were treated with the speci-
of spheroid and bestow TMZ resistance. The inhibition of activity fied compounds, collected after trypsinization, given PBS wash
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAO et al. 5137

and suspended again in binding buffer. These were then stained nearly 100 mm3 tumour volume, the mice were arbitrarily grouped
for 15 minutes with Annexin V/PI as per instructions from Becton into four categories (6 mice/group). Treatment of mice per group
Dickinson. A Becton Dickinson flow cytometer was used to detect was done daily through oral gavage with vehicle, abemaciclib
fluorescence. (150 mg/kg/d), TMZ (50 mg/kg/d) or their combination for three
weeks. Tumours were measured Every two days, tumours were
measured through a Vernier caliper, and their volume was deter-
2.6 | Plasmids and transfection mined by the formula: V = (L × W2)/2 (W, width; L, Length). The
experiments were carried out as per the Regulations for the
The wild-type CDK4 (CDK4-WT) and CDK4-DN expressing plasmids Administration of Affairs Concerning Experimental Animals and
29
were procured from Addgene. Transfection of plasmids was done consented by the Experimental Animal Ethics Committee of Xingtai
into cells along with DNA transfection agent TurboFect as per in- People's Hospital.
structions (Thermo Scientific). At 20%-30% confluence of cells, 2 μg
plasmids were transfected per well of the six-well plate and 0.1 μg
plasmids in each well of a plate with 96-wells. The siRNAs target- 2.10 | Immunohistochemical (IHC) staining
ing human CDK4 and control siRNA were procured from RiboBio.
Transfection of siRNA was done along with Lipofectamine 3000 Immunohistochemical was performed as previously study.32,33
from Invitrogen into the cells as per the protocol of the manufac- Tissue sections (4 μm) were first fixed with 1:1 ratio of
turer. At 20%-30% confluence of cells, siRNA at 50 pmol was trans- Acetone:Methanol for 10 minutes. Peroxidase inhibitor (CM1) was
fected in each well of a six-well plate and at 2.5 pmol in each well of used to block the endogenous peroxidases for 8 minutes prior to
a 96-well plate. the incubation of the section with primary antibody at 1:100 di-
lution overnight at 4°C. The DISCOVERY OmniMap AntiRb HRP
detection system and DISCOVERY ChromoMap DAB Kit (Ventana
2.7 | Quantitative real-time PCR Co.) were used to detect antigen-antibody complex. Then, each
slide was counterstained with haematoxylin, dehydrated, cleared
For two days, the cells were treated and then extraction of RNA was and assessed after mounting.
done using TRIzol reagent (Sigma). Using PrimeScript™ RT reagent
Kit (Perfect Real Time), reverse transcription was carried out fol-
lowed by quantitative RT-PCR in a CFX96 Real-Time PCR Detection 2.11 | Analysis of statistical data
System from Bio-Rad using SYBR® Premix Ex Taq™ II master mix (Tli
RNaseH Plus) from Takara, China. Synthesis of the primers was done Data are presented as the mean ± SD (standard deviation).
at Sangon Biotech Co., Ltd. Per experiment was done independently Student's t test was applied to carry out all statistical assess-
in triplicates. The expression of β-actin was used for normalization ments and realized through GraphPad Prism VI statistical soft-
of relative gene expression which was determined through the 2-ΔΔCt ware. A difference representing P < .05 was deemed significant
method. statistically.

2.8 | Western blotting 3 | R E S U LT S

Western blotting was performed as previously research.30,31 The 3.1 | Glioma tissues and cell lines overexpress CDK4
lysis of cells was done in lysis buffer. The samples were resolved by
SDS-PAGE, transferred to PVDF (polyvinylidene fluoride, Roche) Up-regulation of CDK4 has been observed in several human cancers,
membrane and blocked with skim milk (5%) and kept along with the but its role in glioma tumorigenesis unknown to a large extent. To
specified primary antibody. Then, incubation with the appropriate ascertain CDK4 level in glioma, the comparison of CDK4 mRNA levels
secondary antibody conjugated with HRP (horseradish peroxidase) of normal brain tissues from humans and glioma tissues from pub-
was done. The bands that appeared were observed with Western lished gene profiling studies (TCGA data). CDK4 expression was ob-
Bright ECL HRP substrate and Kodak films were used to develop served higher compared to that in normal control tissues (Figure 1A).
them. Then, through IHC a Western blot assay, the expression of CDK4
in glioma tissues and normal human brain tissues was done. In ac-
cordance with mRNA data, high expression of CDK4 was confirmed
2.9 | Xenograft studies in glioma specimens when compared to nontumour brain tissues
(Figure 1B,C). A similar outcome was observed when the expression
The right back of BALB/c nude mice (5-week-old female) were in- of CDK4 in glioma cell lines and normal human brain tissue were com-
jected subcutaneously with U251 cells (2 × 10 6 cells). On reaching pared (Figure 1D).
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5138 CAO et al.

F I G U R E 1 CDK4 is up-regulated
in glioblastoma tissues and cell lines.
A, Analysis of TCGA datasets showing
high expression of CDK4 mRNA in
glioblastoma specimens compared with
nontumour brain tissues. B, IHC analysis
of CDK4 expression in glioblastoma
specimens and normal brain tissues. Scale
bar, 100 μm. C, Western blotting of CDK4
expression in glioblastoma specimens
and normal brain tissues. D, Comparison
of CDK4 expression between NHAs and
glioblastoma cell lines by western blotting

F I G U R E 2 CDK4 promotes the


proliferation and spheroid formation
of glioblastoma cells. A, U87 cells were
transfected with the indicated plasmids
and cell proliferation was monitored
by a CCK-8 assay. B, U251 cells were
transfected with the indicated plasmids,
and cell proliferation was monitored
by a CCK-8 assay. C, U87 cells were
transfected with the indicated siRNA,
and cell proliferation was monitored
by a CCK-8 assay. D, U251 cells were
transfected with the indicated siRNA,
and cell proliferation was monitored
by a CCK-8 assay. E, U87 cells were
cultured in a nanoculture plate and then
transfected with the indicated plasmids.
Spheroid formation was monitored
under a microscope. F, U87 cells were
cultured in a nanoculture plate and then
transfected with the indicated siRNA.
Spheroid formation was monitored under
a microscope
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAO et al. 5139

3.2 | CDK4 promotes the proliferation and spheroid anticancer drugs, CDK4 inhibitors led to increase in apoptosis in
formation of glioma cells glioma U87 cells (Figure 4D,F). Moreover, inhibition of apoptosis
by pan-caspase inhibitor Z-VAD-FMK in combination with inhibi-
Next, we examined the possible regulation of glioma cell prolif- tors of CDK4 improved the cell viability percentage remarkably in
eration by CDK4 as result of its overexpression in glioma cells. To comparison with CDK4 inhibitors alone (Figure 4G-I). To further
assess this, wild-type CDK4 (CDK4-WT) and CDK4-DN overex- verify the above results, we carried out Western blot to evalu-
pression plasmids were transfected in U87 and U251 cells, and ate the changes in apoptosis-related proteins (cleaved caspase-3,
CCK-8 assay was done to assess cell proliferation. A significant Bax and Bcl-2) in U87 and U251 cells after 24 hours of treatment
rise in the rate of cell proliferation was observed on up-regulation with abemaciclib. Our results indicated abemaciclib enhance Bax
of CDK4 (Figure 2A,B). The CDK4-DN–transfected group exhib- and decrease Bcl-2 levels, and increased caspase 3 activation
ited similar rate of proliferation as the control group (Figure 2C,D). (Figure 4J-K).
Furthermore, CDK4 overexpression mainly facilitated the sphe-
roid formation of glioma cells (Figure 2E), although CDK4-DN
overexpression exhibited no evident effect of glioma cells sphe- 3.5 | CDK4 inhibition resensitize glioma cells
roid formation. The ability of glioma to form a spheroid was nearly to TMZ
abolished as a result of CDK4 knockdown (Figure 2F). Thus, CDK4
promotes spheroid formation and cell proliferation ability of Overexpression CDK4 confers resistance to TMZ; thus, we speculated
glioma. that CDK4 inhibition might aid in an enhanced TMZ efficiency in glioma.
To assess any cooperation between TMZ and inhibitors of CDK4 in the
chemotherapy of glioma, we treated glioma cells with TMZ and abe-
3.3 | The TMZ-mediated killing of glioma cells is maciclib either singly or in combination. Accordingly, the cotreatment
resisted by CDK4 with TMZ and abemaciclib significantly lowered the cell viability when
compared to treatment with TMZ alone (Figure 5A,B). In addition, the
A standard therapy for glioma is surgical resection and radio- combination index (CI) values of abemaciclib and TMZ were evaluated,
therapy combined with TMZ. 5 However, the effectiveness of this and abemaciclib showed strong synergistic effects with TMZ in U87
modality is limited by acquired chemoresistance of glioma cells. and U251 cells (Figure 5C,D). After 2 weeks, we assessed long-term
Thus, it is imperative to study TMZ resistance and we conjecture survival by staining through crystal violet and observed that the com-
that CDK4 overexpression in glioma might bestow resistance to bination nearly fully eliminated U87 and U251 cell clonogenic survival
TMZ. Figure 3A,B and E shows that CDK4 overexpression signifi- (Figure 5E,F). Furthermore, an increase in apoptotic rate was also ob-
cantly enhanced the percentage of living cells post-TMZ treatment. served after treatment with the combination, as an increase in Annexin
Furthermore, after TMZ treatment, nearly the same percentage of V-positive cell number was found (Figure 5G,H). Thus, the combination
living cells was observed in the group transfected with CDK4-DN. treatment of TMZ and CDK4 inhibitor is therapeutically effective on
As anticipated, CDK4 knockdown made the glioma more sensitive to glioma.
TMZ (Figure 3C,D and F). We evaluated the role of CDK4 in regulat-
ing TMZ-mediated cell apoptosis in glioma cells because at higher
concentrations, TMZ causes apoptotic cell death. TMZ-induced ap- 3.6 | The sensitivity of glioma cells to TMZ is
optosis is remarkably reduced as a result of CDK4 overexpression enhanced by abemaciclib through p-RB repression
(Figure 3G,H), while CDK4 knockdown sensitized glioma cells to
TMZ-mediated apoptosis (Figure 3I-L). Our results thus indicate that We then analysed whether the inhibitory effect of TMZ and abe-
CDK4 overexpression in glioma may possibly be an innate process maciclib treatment is mediated by the p-RB suppression. For this,
that grants TMZ resistance. we evaluated the effect of the dosage of TMZ or abemaciclib on
p-RB, by treating U87 glioma cells for 48 hours and observed
up-regulation of p-RB (Figure 6A). However, when the cells were
3.4 | Selective inhibitors of CDK4/6 inhibit treated with abemaciclib alone, down-regulation of p-RB was ob-
proliferation and induce apoptosis of glioma cells served (Figure 6B). To assess the interaction of abemaciclib to TMZ,
studies were conducted in parallel where U87 cells were treated
From the data mentioned above, CDK4 was found to have a with TMZ and abemaciclib for one full day and the Western blot re-
crucial part in controlling the proliferation of glioma cell, to cor- sults showed that the drug combination resulted in more decline in
roborate this, tried to inhibit CDK4 activity using three selective RB phosphorylation (Figure 6C). Considering these results, RB acti-
inhibitors abemaciclib, palbociclib and ribociclib and assessed vation is induced by TMZ treatment, leading to induction of apopto-
growth inhibition in three human glioma cell lines: A172, U87 and sis in glioma cells. Nevertheless, p-RB induced by TMZ is repressed
U251. For all three chemicals tested, a decline in cell prolifera- by abemaciclib, and a possible cause of enhanced sensitivity of cells
tion dependent on dose was observed (Figure 4A-C). Like a few to TMZ.
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5140 CAO et al.

F I G U R E 3 Overexpression of CDK4 confers resistance to TMZ-mediated cell proliferation inhibition. A, U87 cells were transfected
with the indicated plasmids. Then, 24 h after transfection, the cells were treated with the indicated concentrations of TMZ. After another
48 h, cell viability was measured by a CCK-8 assay. B, U251 cells were transfected with the indicated siRNA. Then, 24 h after transfection,
the cells were treated with the indicated concentrations of TMZ. After another 48 h, cell viability was measured by a CCK-8 assay. C, U87
cells were transfected with the indicated plasmids. Then, 24 h after transfection, the cells were treated with the indicated concentrations
of TMZ. After another 48 h, cell viability was measured by a CCK-8 assay. D, U251 cells were transfected with the indicated siRNA. Then,
24 h after transfection, the cells were treated with the indicated concentrations of TMZ. After another 48 h, cell viability was measured by a
CCK-8 assay. E, Colony formation of U87 cells transfected with vector, CDK4 or CDK4-DN, was treated with TMZ for two weeks. F, Colony
formation of U87 cells transfected with siRNA against CDK4 was treated with TMZ for two weeks. G, U87 cells were transfected with
the indicated plasmids. Then, 24 h after transfection, the cells were treated with the indicated concentrations of TMZ. After another 48 h,
cell apoptosis was analysed. H, U251 cells were transfected with the indicated siRNA. Then, 24 h after transfection, the cells were treated
with the indicated concentrations of TMZ. After another 48 h, cell apoptosis was analysed. I, U87 cells were transfected with the indicated
plasmids. Then, 24 h after transfection, the cells were treated with the indicated concentrations of TMZ. After another 48 h, cell apoptosis
was analysed. J, U251 cells were transfected with the indicated siRNA. Then, 24 h after transfection, the cells were treated with the
indicated concentrations of TMZ. After another 48 h, cell apoptosis was analysed. K, U87 cells were transfected with the indicated plasmids.
Then, 24 h after transfection, the cells were treated with the indicated concentrations of TMZ. After another 48 h, cleaved caspase 3 was
analysed by Western blotting. L, U87 cells were transfected with the indicated siRNA. Then, 24 h after transfection, the cells were treated
with the indicated concentrations of TMZ. After another 48 h, cleaved caspase 3 was analysed by Western blotting

3.7 | TMZ and abemaciclib combination treatment 3 revealed increase in apoptosis (Figure 7D,E). Therefore, a combi-
improved antitumour efficacy in a xenograft model nation of abemaciclib and TMZ exhibits a synergy in the therapeutic
effect in vivo.
On observing the in vitro results, we assessed the efficiency of com-
bination treatment in xenografts. The combination therapy led to
significantly reduced tumour volume compared to the controls or 4 | D I S CU S S I O N
after monotherapy at end of three weeks (Figure 7A,B). Therefore,
like in vitro outcome, combination therapy markedly repressed RB A type of primary tumour of the brain, glioma, is the most common
dephosphorylation (Figure 7C). Likewise, IHC staining for Ki-67 re- and the most aggressive subtype is GBM.1,34 Currently, the common
vealed a decline in proliferation, and IF staining for cleaved caspase treatment option, chemotherapy, is largely ineffective because of
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAO et al. 5141

F I G U R E 4 CDK4/6-selective inhibitors impair the proliferation of glioblastoma cells. A, U87 cells were plated in 96-well cell culture
plates, and one day later, they were treated with the increasing dose of CDK4/6 inhibitors. After 72 h of treatment, cell viability was
measured by a CCK-8 assay. B, U251 cells were plated in 96-well cell culture plates, and one day later, they were treated with the increasing
dose of CDK4/6 inhibitors. After 72 h of treatment, cell viability was measured by a CCK-8 assay. C, A172 cells were plated in 96-well cell
culture plates, and one day later, they were treated with the increasing dose of CDK4/6 inhibitors. After 72 h of treatment, cell viability was
measured by a CCK-8 assay. D, U87 cells were treated with abemaciclib for 48 h, cell apoptosis was measured by flow cytometry. E, U87
cells were treated with palbociclib for 48 h, cell apoptosis was measured by flow cytometry. F, U87 cells were treated with ribociclib for 48 h,
cell apoptosis was measured by flow cytometry. G, U87 cells were treated with abemaciclib with or without 10 μmol/L Z-VAD-FMK for 48 h,
cell apoptosis was measured by flow cytometry. H, U87 cells were treated with palbociclib with or without 10 μmol/L Z-VAD-FMK for 48 h,
cell apoptosis was measured by flow cytometry. I, U87 cells were treated with ribociclib with or without 10 μmol/L Z-VAD-FMK for 48 h,
cell apoptosis was measured by flow cytometry. J, U87 cells were treated with abemaciclib at indicated concentration, and indicated protein
level was analysed by Western blotting. K, U251 cells were treated with abemaciclib at indicated concentration, and indicated protein level
was analysed by Western blotting

chemoresistance, leading to a recurrence of cancer.35,36 Anti-TMZ abemaciclib against glioma, as well as the synergy between abemaci-
resistance, as a form of anti-chemoresistance, is a potentially prom- clib and TMZ. Indeed, abemaciclib significantly induce apoptosis in
ising option for glioma treatment.37 Abemaciclib exhibits favour- glioma cells in vitro, therefore, its repressed cell proliferation and
able therapeutic properties and potential anticancer efficacy.38 survival. Further, this pro-apoptotic effect was found to occur via
Therefore, we assessed the in vitro activity and in vivo activity of RB pathway, in addition to a decline in Bcl-2 level and activation of
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5142 CAO et al.

F I G U R E 5 CDK4/6 inhibitors sensitize


glioblastoma cells to TMZ. A, U87 cells
were treated with the given doses of
TMZ with abemaciclib. Cell viability was
measured by a CCK-8 assay. B, U251
cells were treated with the given doses
of TMZ with abemaciclib. Cell viability
was measured by a CCK-8 assay. C, Drug
combination results (CI) for abemaciclib
and TMZ examined in U87 cells. D, Drug
combination results (CI) for abemaciclib
and TMZ examined in U251 cells. E,
U87 cells were treated with 200 μmol/L
TMZ, 0.1 μmol/L abemaciclib or their
combination. Long-term survival was
visualized after 14 d by crystal violet
staining. F, U87 cells were treated with
50 μmol/L TMZ, 0.1 μmol/L abemaciclib
or their combination. Long-term survival
was visualized after 14 d by crystal
violet staining. G, U87 cells were treated
with 200 μmol/L TMZ, 0.1 μmol/L
abemaciclib or their combination for
48 h. Cell apoptosis was measured by
flow cytometry. H, U251 cells were
treated with 50 μmol/L TMZ, 0.1 μmol/L
abemaciclib or their combination for 48 h.
Cell apoptosis was measured by flow
cytometry

F I G U R E 6 CDK4 inhibition suppressed RB phosphorylation. A, U87 cells were treated with indicated concentration of TMZ. Indicated
protein levels were analysed by Western blotting. B, U87 cells were treated with indicated concentration of abemaciclib. Indicated protein
levels were analysed by Western blotting. C, U87 cells were treated with indicated concentration of TMZ and abemaciclib. Indicated protein
levels were analysed by Western blotting

caspase-3 and Bax in glioma cell lines. A preferred drug for GBM The acquired or inherent resistance to TMZ is considerable, and, the
treatment is TMZ, but it is not curative and, thus, more efficient resistance of glioma cells primarily involves the MGMT DNA-repair en-
treatment options are needed. zyme.39 MGMT, a 22 kD protein, repairs TMZ-induced lesions directly
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAO et al. 5143

F I G U R E 7 CDK inhibitor sensitized TMZ in vivo. A, Nude mice were injected s.c. with 5 × 106 U87 cells. After 1 wk, mice were treated
with TMZ, abemaciclib or their combination for 10 consecutive days. Tumour volume at indicated time-points after treatment was calculated
and plotted with P values, n = 6, in each group. B, Tumour weight was calculated at end of the experiments. C, The levels of indicated
proteins in randomly selected tumours were analysed by Western blotting. D, Ki-67 was analysed by IHC staining. E, Cleaved caspase 3 was
analysed by IF staining

by eliminating guanine site O6 methylation.39 Recently, GANT61, a TMZ combination is more effective in inhibition of tumour cell
specific GLI (glioma-associated oncogene) inhibitor, was shown to in- proliferation and apoptotic induction in comparison with TMZ or
crease DNA damage, repress MGMT expression and recover the TMZ abemaciclib singly. In addition, the combination led to significantly
sensitivity of glioma, implicating some association between MGMT increased expression of apoptosis-related proteins (such as Bax,
and the hedgehog signalling pathway.40 Likewise, in the primary glioma Bcl-2 and cleaved caspase-3). To better understand the underlying
tissues, the association of zinc finger protein Gli1 activity with MGMT, mechanism, we observed that p-RB levels up-regulated by TMZ
with Gli1 binding to promoter region of the MGMT gene, implicating could be reversed by abemaciclib. The results were further cor-
MGMT to be a downstream target of HH/Gli1 pathway.41 roborated by our in vitro study which showed that combination
Some CDKs have recently been conferred roles as immune re- treatment extended median survival significantly in tumour-bear-
sponse and oncogenesis modulators.42 Particularly, genetic or phar- ing mice. In preclinical mouse models, abemaciclib shows promise
macological inhibition of CDK4 and CDK6 could inhibit in vivo and in controlling solid tumours and enhances sensitivity to gefitinib
in vitro tumour growth and control tumour associated antigens ex- and radiotherapy.48,49 To our knowledge, effects of abemaciclib
43,44
pression. In the progression of cell cycle, CDK4 and CDK6, both on the cytotoxicity of TMZ have not been reported in glioma cells.
close homologs, interact with cyclin D and form heterodimers.45 While we show here the synergism between TMZ and abemaci-
One of the selective inhibitors of the CDK4/6-cyclin D complex is clib in inhibiting glioma cells proliferation and bringing about the
P16, encoded by CDKN2A.45 CDK4 contributes to tumorigenesis in much-enhanced rates of apoptosis, future studies need to be care-
several human cancers,46 and its inhibition can increase oncolytic fully conducted to assess the suitability of this drug combination
viral replication in glioma.47 Here, we showed that pharmacological with TMZ.
inhibition and genetic knockdown of CDK4 hinders growth of glioma To conclude, we report enhanced expression of CDK4 in glioma
and TMZ resistance, via RB pathway regulation. and impaired proliferation of glioma as a result of CDK4 inhibition.
We report here that CDK4 enables glioma cell lines resistant Further, the CDK4 inhibitors could sensitize glioma cells to TMZ-
to TMZ, although the association between CDK4 and TMZ resis- induced inhibition of cell proliferation and induction of apoptosis.
tance in terms of their levels in primary gliomas still remains to be Therefore, CDK4 inhibition may be a promising strategy for treating
unravelled. Therefore, larger sample sizes are required to assess glioma and surpassing TMZ resistance.
the relationship between TMZ resistance and CDK4 levels. For
this, larger number samples that are resistant to TMZ are being AC K N OW L E D G E M E N T S
collected from our hospital, and the results will be presented in None.
our next manuscript.
Here, we focused on the synergism between CDK4/6 inhibi- C O N FL I C T O F I N T E R E S T
tors and TMZ, and report for the first time that abemaciclib and The authors have no conflict of interest.
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5144 CAO et al.

AU T H O R C O N T R I B U T I O N 17. Baker SJ, Reddy EP. CDK4: a key player in the cell cycle, develop-
ment, and cancer. Genes Cancer. 2012;3:658-669.
YC, XL and SK designed the study. YC, SS and YQ collated the
18. Chen F, Liu C, Zhang J, Xu W, Zhang Y. Progress of CDK4/6 inhibitor
data, carried out data analysis and generated the draft of the man- palbociclib in the treatment of cancer. Anticancer Agents Med Chem.
uscript. All authors have read and approved the final submitted 2018;18:1241-1251.
manuscript. 19. Gopalan PK, Villegas AG, Cao C, et al. CDK4/6 inhibition stabilizes
disease in patients with p16-null non-small cell lung cancer and is
synergistic with mTOR inhibition. Oncotarget. 2018;9:37352-37366.
DATA AVA I L A B I L I T Y S TAT E M E N T
20. Taylor JW, Parikh M, Phillips JJ, et al. Phase-2 trial of palboci-
The data that support the findings of this study are available from clib in adult patients with recurrent RB1-positive glioblastoma. J
the corresponding author upon reasonable request. Neurooncol. 2018;140:477-483.
21. Whittaker S, Madani D, Joshi S, et al. Combination of palbociclib
and radiotherapy for glioblastoma. Cell Death Discov. 2017;3:17033.
ORCID
22. Serra F, Lapidari P, Quaquarini E, Tagliaferri B, Sottotetti F, Palumbo
Yingxiao Cao https://orcid.org/0000-0002-2646-7608 R. Palbociclib in metastatic breast cancer: current evidence and re-
al-life data. Drugs Context. 2019;8:212579.
REFERENCES 23. Michaud K, Solomon DA, Oermann E, et al. Pharmacologic inhi-
bition of cyclin-dependent kinases 4 and 6 arrests the growth
1. Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee SHU. Glioblastoma
of glioblastoma multiforme intracranial xenografts. Cancer Res.
multiforme: a review of its epidemiology and pathogenesis through
2010;70:3228-3238.
clinical presentation and treatment. Asian Pac J Cancer Prev.
24. Barton KL, Misuraca K, Cordero F, et al. PD-0332991, a CDK4/6
2017;18:3-9.
inhibitor, significantly prolongs survival in a genetically engineered
2. Lukas RV, Wainwright DA, Ladomersky E, Sachdev S, Sonabend
mouse model of brainstem glioma. PLoS One. 2013;8:e77639.
AM, Stupp R. Newly diagnosed glioblastoma: a review on clinical
25. Olmez I, Brenneman B, Xiao A, et al. Combined CDK4/6 and mTOR
management. Oncology (Williston Park). 2019;33:91-100.
inhibition is synergistic against glioblastoma via multiple mecha-
3. Fu P, He YS, Huang Q, et al. Bevacizumab treatment for newly diag-
nisms. Clin Cancer Res. 2017;23:6958-6968.
nosed glioblastoma: systematic review and meta-analysis of clinical
26. Arnedos M, Bayar MA, Cheaib B, et al. Modulation of Rb phos-
trials. Mol Clin Oncol. 2016;4:833-838.
phorylation and antiproliferative response to palbociclib: the pre-
4. Amelio D, Lorentini S, Schwarz M, Amichetti M. Intensity-
operative-palbociclib (POP) randomized clinical trial. Ann Oncol.
modulated radiation therapy in newly diagnosed glioblastoma: a
2018;29:1755-1762.
systematic review on clinical and technical issues. Radiother Oncol.
27. Tong J, Zheng X, Tan X, et al. Mcl-1 phosphorylation without degra-
2010;97:361-369.
dation mediates sensitivity to HDAC inhibitors by liberating BH3-
5. Shergalis A, Bankhead 3rd A, Luesakul U, Muangsin N, Neamati
only proteins. Cancer Res. 2018;78:4704-4715.
N. Current challenges and opportunities in treating glioblastoma.
28. Tong J, Wang P, Tan S, et al. Mcl-1 degradation is required for tar-
Pharmacol Rev. 2018;70:412-445.
geted therapeutics to eradicate colon cancer cells. Cancer Res.
6. Choi S, Yu Y, Grimmer MR, Wahl M, Chang SM, Costello JF.
2017;77:2512-2521.
Temozolomide-associated hypermutation in gliomas. Neuro Oncol.
29. van den Heuvel S, Harlow E. Distinct roles for cyclin-dependent ki-
2018;20:1300-1309.
nases in cell cycle control. Science. 1993;262:2050-2054.
7. Daniel P, Sabri S, Chaddad A, et al. Temozolomide induced hyper-
30. Tong J, Tan S, Nikolovska-Coleska Z, Yu J, Zou F, Zhang L. FBW7-
mutation in glioma: evolutionary mechanisms and therapeutic op-
dependent Mcl-1 degradation mediates the anticancer effect of
portunities. Front Oncol. 2019;9:41.
Hsp90 inhibitors. Mol Cancer Ther. 2017;16:1979-1988.
8. Oshiro S, Tsugu H, Komatsu F, et al. Efficacy of temozolomide
31. Tong J, Tan S, Zou F, Yu J, Zhang L. FBW7 mutations mediate resis-
treatment in patients with high-grade glioma. Anticancer Res.
tance of colorectal cancer to targeted therapies by blocking Mcl-1
2009;29:911-917.
degradation. Oncogene. 2017;36:787-796.
9. Lee SY. Temozolomide resistance in glioblastoma multiforme. Genes
32. Chen D, Tong J, Yang L, et al. PUMA amplifies necroptosis signal-
Dis. 2016;3:198-210.
ing by activating cytosolic DNA sensors. Proc Natl Acad Sci USA.
10. Chen X, Zhang M, Gan H, et al. A novel enhancer regulates MGMT
2018;115:3930-3935.
expression and promotes temozolomide resistance in glioblastoma.
33. Knickelbein K, Tong J, Chen D, et al. Restoring PUMA induction
Nat Commun. 2018;9:2949.
overcomes KRAS-mediated resistance to anti-EGFR antibodies in
11. Jiapaer S, Furuta T, Tanaka S, Kitabayashi T, Nakada M. Potential
colorectal cancer. Oncogene. 2018;37:4599-4610.
strategies overcoming the temozolomide resistance for glioblas-
34. Lucas CG, Solomon DA, Perry A. A review of recently described
toma. Neurol Med Chir (Tokyo). 2018;58:405-421.
genetic alterations in central nervous system tumors. Hum Pathol.
12. Vijayaraghavan S, Moulder S, Keyomarsi K, Layman RM. Inhibiting
2019:30191-30201.
CDK in cancer therapy: current evidence and future directions.
35. Yu X, Wang M, Zuo J, et al. Nuclear factor I A promotes temozolo-
Target Oncol. 2018;13:21-38.
mide resistance in glioblastoma via activation of nuclear factor kap-
13. Mayer EL. Targeting breast cancer with CDK inhibitors. Curr Oncol
paB pathway. Life Sci. 2019;236:116917.
Rep. 2015;17:443.
36. Wang J, Zuo J, Wang MD, Xie WF, Bai XB, Ma XD. Receptor tyrosine
14. Spring L, Bardia A, Modi S. Targeting the cyclin D-cyclin-dependent
kinase AXL is correlated with poor prognosis and induces temozolo-
kinase (CDK) 4/6-retinoblastoma pathway with selective CDK 4/6
mide resistance in glioblastoma. CNS Neurosci Ther. 2019:13227.
inhibitors in hormone receptor-positive breast cancer: rationale,
37. Zhao M, Tan B, Dai X, et al. DHFR/TYMS are positive regulators
current status, and future directions. Discov Med. 2016;21:65-74.
of glioma cell growth and modulate chemo-sensitivity to temozolo-
15. Goel S, DeCristo MJ, McAllister SS, Zhao JJ. CDK4/6 inhibition in
mide. Eur J Pharmacol. 2019;863:172665.
cancer: beyond cell cycle arrest. Trends Cell Biol. 2018;28:911-925.
38. Kim ES, Scott LJ. Palbociclib: a review in HR-positive, HER2-
16. Preusser M, De Mattos-Arruda L, Thill M, et al. CDK4/6 inhibitors in
negative, advanced or metastatic breast cancer. Target Oncol.
the treatment of patients with breast cancer: summary of a multi-
2017;12:373-383.
disciplinary round-table discussion. ESMO Open. 2018;3:e000368.
|

15824934, 2020, 9, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/jcmm.15156 by Turkey Cochrane Evidence Aid, Wiley Online Library on [10/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAO et al. 5145

39. Chai RC, Chang YZ, Wang QW, et al. A novel DNA methyla- 46. Li P, Zhang X, Gu L, Zhou J, Deng D. P16 methylation increases the
tion-based signature can predict the responses of MGMT pro- sensitivity of cancer cells to the CDK4/6 inhibitor palbociclib. PLoS
moter unmethylated glioblastomas to temozolomide. Front Genet. One. 2019;14:e0223084.
2019;10:910. 47. Becher OJ. CDK4/6 and diffuse intrinsic pontine glioma - Evaluate
40. Li J, Cai J, Zhao S, et al. GANT61, a GLI inhibitor, sensitizes gli- at diagnosis? EBioMedicine. 2019;44:16-17.
oma cells to the temozolomide treatment. J Exp Clin Cancer Res. 48. Liu M, Xu S, Wang Y, et al. PD 0332991, a selective cyclin D kinase
2016;35:184. 4/6 inhibitor, sensitizes lung cancer cells to treatment with epider-
41. Wang K, Chen D, Qian Z, Cui D, Gao L, Lou M. Hedgehog/Gli1 mal growth factor receptor tyrosine kinase inhibitors. Oncotarget.
signaling pathway regulates MGMT expression and chemoresis- 2016;7:84951-84964.
tance to temozolomide in human glioblastoma. Cancer Cell Int. 49. Chowdhary M, Sen N, Chowdhary A, et al. Safety and efficacy of
2017;17:117. palbociclib and radiation therapy in patients with metastatic breast
42. Yu J, Yan J, Guo Q, et al. Genetic aberrations in the CDK4 path- cancer: initial results of a novel combination. Adv Radiat Oncol.
way are associated with innate resistance to PD-1 blockade in 2019;4:453-457.
chinese patients with non-cutaneous melanoma. Clin Cancer Res.
2019;25:6511-6523.
43. Deng J, Wang ES, Jenkins RW, et al. CDK4/6 inhibition augments
How to cite this article: Cao Y, Li X, Kong S, Shang S, Qi Y.
antitumor immunity by enhancing T-cell activation. Cancer Discov.
2018;8:216-233.
CDK4/6 inhibition suppresses tumour growth and enhances
44. Zhang J, Bu X, Wang H, et al. Cyclin D-CDK4 kinase destabilizes the effect of temozolomide in glioma cells. J Cell Mol Med.
PD-L1 via cullin 3-SPOP to control cancer immune surveillance. 2020;24:5135–5145. https://doi.org/10.1111/jcmm.15156
Nature. 2018;553:91-95.
45. Pandey K, An HJ, Kim SK, et al. Molecular mechanisms of resis-
tance to CDK4/6 inhibitors in breast cancer: a review. Int J Cancer.
2019;145:1179-1188.

You might also like