ArticlePDF Available

Autophagy: A Novel Mechanism of Synergistic Cytotoxicity between Doxorubicin and Roscovitine in a Sarcoma Model

Authors:

Abstract and Figures

Doxorubicin is a genotoxic chemotherapy agent used in treatment of a wide variety of cancers. Significant clinical side effects, including cardiac toxicity and myelosuppression, severely limit the therapeutic index of this commonly used agent and methods which improve doxorubicin efficacy could benefit many patients. Because doxorubicin cytotoxicity is cell cycle specific, the cell cycle is a rational target to enhance its efficacy. We examined the direct, cyclin-dependent kinase inhibitor roscovitine as a means of enhancing doxorubicin cytotoxicity. This study showed synergistic cytotoxicity between doxorubicin and roscovitine in three sarcoma cell lines: SW-982 (synovial sarcoma), U2OS-LC3-GFP (osteosarcoma), and SK-LMS-1 (uterine leiomyosarcoma), but not the fibroblast cell line WI38. The combined treatment of doxorubicin and roscovitine was associated with a prolonged G(2)-M cell cycle arrest in the three sarcoma cell lines. Using three different methods for detecting apoptosis, our results revealed that apoptotic cell death did not account for the synergistic cytotoxicity between doxorubicin and roscovitine. However, morphologic changes observed by light microscopy and increased cytoplasmic LC3-GFP puncta in U20S-LC3-GFP cells after the combined treatment suggested the induction of autophagy. Induction of autophagy was also shown in SW-982 and SK-LMS-1 cells treated with both doxorubicin and roscovitine by acridine orange staining. These results suggest a novel role of autophagy in the enhanced cytotoxicity by cell cycle inhibition after genotoxic injury in tumor cells. Further investigation of this enhanced cytotoxicity as a treatment strategy for sarcomas is warranted.
Content may be subject to copyright.
Autophagy: A Novel Mechanism of Synergistic Cytotoxicity between
Doxorubicin and Roscovitine in a Sarcoma Model
Laura A. Lambert,1Na Qiao,2Kelly K. Hunt,1Donald H. Lambert,1Gordon B. Mills,3
Laurent Meijer,4and Khandan Keyomarsi1,2
Departments of 1Surgical Oncology, 2Experimental Radiation Oncology, and 3Systems Biology, University of Texas - M. D. Anderson Cancer
Center, Houston, Texas; and 4Protein Phosphorylation and Disease, Centre National de la Recherche Scientifique,
Station Biologique, Roscoff, France
Abstract
Doxorubicin is a genotoxic chemotherapy agent used in treat-
ment of a wide variety of cancers. Significant clinical side
effects, including cardiac toxicity and myelosuppression,
severely limit the therapeutic index of this commonly used
agent and methods which improve doxorubicin efficacy could
benefit many patients. Because doxorubicin cytotoxicity is
cell cycle specific, the cell cycle is a rational target to enhance
its efficacy. We examined the direct, cyclin-dependent kinase
inhibitor roscovitine as a means of enhancing doxorubi-
cin cytotoxicity. This study showed synergistic cytotoxicity
between doxorubicin and roscovitine in three sarcoma cell
lines: SW-982 (synovial sarcoma), U2OS-LC3-GFP (osteosarco-
ma), and SK-LMS-1 (uterine leiomyosarcoma), but not the
fibroblast cell line WI38. The combined treatment of doxoru-
bicin and roscovitine was associated with a prolonged G
2
-M
cell cycle arrest in the three sarcoma cell lines. Using three
different methods for detecting apoptosis, our results revealed
that apoptotic cell death did not account for the synergistic
cytotoxicity between doxorubicin and roscovitine. However,
morphologic changes observed by light microscopy and
increased cytoplasmic LC3-GFP puncta in U20S-LC3-GFP cells
after the combined treatment suggested the induction of
autophagy. Induction of autophagy was also shown in SW-982
and SK-LMS-1 cells treated with both doxorubicin and
roscovitine by acridine orange staining. These results sug-
gest a novel role of autophagy in the enhanced cytotoxicity by
cell cycle inhibition after genotoxic injury in tumor cells. Fur-
ther investigation of this enhanced cytotoxicity as a treat-
ment strategy for sarcomas is warranted. [Cancer Res 2008;
68(19):7966–74]
Introduction
Sarcomas are a broad group of mesenchymal tumors that are
notoriously chemoresistant. More than 50 histological types of
sarcoma are described (1) with an overall 5-year survival for all
stages of 50% to 60% (2–4). Only 20% of sarcomas respond to
doxorubicin, which is the current standard of systemic therapy care
for these tumors (5). Unfortunately, the clinical utility of
doxorubicin, which is also used to treat a wide range of solid
and nonsolid tumors, is limited by significant side effects,
particularly irreversible cardiac toxicity (6). For these reasons,
efforts to improve the efficacy of doxorubicin are essential and
could benefit many patients suffering from a variety of cancers.
The mechanism of doxorubicin cytotoxicity involves ‘‘poisoning’’
the topoisomerase II enzyme, thereby interfering with the
separation of daughter DNA strands and chromatin remodeling.
In addition, doxorubicin intercalates into double-stranded DNA
producing structural changes that interfere with DNA and RNA
synthesis (6). Because it primarily damages double-stranded DNA,
cells in the S phase of the cell cycle are more susceptible. Because
of this cell cycle specificity, the cell cycle is a rational target for
enhancing doxorubicin efficacy.
Cyclin-dependent kinases (Cdk) are essential cell cycle regula-
tory proteins, which ensure accurate and appropriate transition
between cell cycle phases and ultimately cell division (7). In
addition, Cdk function is an important determinant of the cellular
response to DNA damage, including that caused by genotoxic
chemotherapies, such as doxorubicin (8). To date, 13 different Cdks
have been identified (Cdk1–13; ref. 9). The activity of individual Cdk
types is restricted to specific phases of the cell cycle and dependent
on binding with activating cyclin proteins (10). Because Cdks are
involved in regulating many important aspects of the cell cycle,
they provide an appealing target for cell cycle-directed anticancer
therapy.
Currently, >100 direct and indirect Cdk inhibitors are available.
One of the most studied, and apparently selective, direct Cdk
inhibitors is the purine analogue, roscovitine (11). Roscovitine
directly inhibits Cdks by occupying the ATP binding site of the
catalytic subunit of the kinase protein and is relatively more
specific for Cdk1 and Cdk2 than other Cdk inhibitors (12). It is a
well-tolerated, oral agent, which has shown ability to arrest tumor
growth as a single agent in phase I clinical trials (13) and is
currently being tested in phase II clinical trials against non–small
cell lung cancer and nasopharyngeal cancer. Other preclinical
studies have shown enhanced cytotoxicity with a variety of chemo-
therapeutic agents when combined with roscovitine (14, 15).
Because roscovitine can inhibit both Cdk1 and Cdk2, it has the
potential to enhance the cytotoxicity of genotoxic chemotherapeu-
tic agents by the combined effect of delayed cell cycle progression
as well as inhibition of the DNA damage response pathway.
In the present study, we investigated the effect of the
combination of doxorubicin with roscovitine in three different
sarcoma cell lines and one immortalized fibroblast cell line. Our
results show a nonapoptotic synergistic cytotoxicity in all three
sarcoma cell lines, but not the fibroblasts, treated with both
doxorubicin and roscovitine. This synergistic cell death was
accompanied by both a prolonged G
2
-M arrest and significant
induction of autophagy.
Requests for reprints: Khandan Keyomarsi, Department of Experimental
Radiation Oncology, Unit 0066, University of Texas, M. D. Anderson Cancer Center,
1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-792-4845; Fax: 713-794-
5369; E-mail: kkeyomar@mdanderson.org.
I2008 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-08-1333
Cancer Res 2008; 68: (19). October 1, 2008 7966 www.aacrjournals.org
Research Article
Materials and Methods
Cell culture. Fetal bovine serum (FBS) and cell culture medium were
purchased from Hyclone Laboratories. All other chemicals were reagent
grade. SW-982, SK-LMS-1, and WI38 (immortalized fibroblasts) were
purchased from American Type Culture Collection and cultured in DMEM
(SW-982, SK-LMS-1) or a-MEM (WI38) with 10% FBS. U20S cells stably
transfected with LC3-GFP protein (U2OS-LC3-GFP) were kindly donated by
Dr. Gordon B. Mills (M. D. Anderson Cancer Center, Houston, TX) and
maintained in DMEM with 10% FBS. All cells were cultured and treated at
37jC in a humidified incubator containing 6.5% CO2. A 5-mmol/L stock
solution of doxorubicin (Sigma) was made in sterile water and maintained at
20jC. A 10-mmol/L stock solution of roscovitine (synthesized and kindly
provided by Drs. Herve Galons, Universite´ Rene´ Descartes, Paris, France and
Laurent Meijer, Protein Phosphorylation and Disease, Centre National de la
Recherche Scientifique, Station Biologique, Roscoff, France) was made in
DMSO and maintained at 20jC. Fresh drug was prepared for each
experiment.
Cell cycle analysis. For DNA content analysis, harvested cells were
centrifuged at 1,000 gfor 5 min, washed with PBS, and fixed in 70% ethanol.
Cells were then treated with RNase (10 Ag/mL) for 30 min at 37jC, washed
with PBS, resuspended, and stained in 1 mL of 69 Amol/L propidium iodide
(PI) in 38 mmol/L sodium citrate for 30 min. The cell cycle phase
distribution was determined by analytic DNA flow cytometry as described
by Keyomarsi and colleagues (16). The percentage of cells in each phase of
the cell cycle was analyzed using Modfit software (Verity Software House).
High-throughput clonogenic assay and combination index. 3-(4,5-
Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were
modified for use as high-throughput clonogenic assays (HTCA) to
determine cell viability. Cells were plated in 96-well plates at a density of
1.5 to 3 10
3
cells per well. After 24 h, cells were treated with drug-free
medium, 0.001 to 0.02 Amol/L doxorubicin for 24 h, 5 to 25 Amol/L
roscovitine for 48 h, or doxorubicin for 24 h then roscovitine for 48 h. Drug-
free medium was replaced every 48 h after treatment. Six replicates were
plated for each treatment group. Ten days after plating, 50 AL of 2.5 mg/mL
MTT solution was added to each well and incubated for 4 h. The resultant
blue formazan crystals were solubilized in 100 AL of buffer (0.04 N HCl and
1% SDS in isopropyl alcohol) for 1 h. Absorbance was read at 590 nm using a
Wallac-1420 plate reader. Drug interactions were assessed using CalcuSyn
software version 2.1 (Biosoft, Inc.) to determine the combination index of
the combined treatment of doxorubicin and roscovitine.
Clonogenic assays. Cells (1 10
3
) were plated onto 100-mm
2
dishes.
After 24 h, cells were treated with fresh drug-free medium, 0.01 Amol/L
doxorubicin (SK-LMS-1), 0.005 Amol/L doxorubicin (U2OS-LC3-GFP and
SW-982) for 24 h, 20 Amol/L (SK-LMS-1) or 10 Amol/L (U2OS-LC3-GFP and
SW-982) roscovitine for 48 h, or doxorubicin for 24 h then roscovitine for
48 h. Drug-free medium was applied at the end of each treatment. After
14 d, the cells were fixed and stained with crystal violet in 100% ethanol
suspension. Plates were scored for the number of visible colonies of z2 mm.
Apoptosis assays. Both APC-conjugated Annexin V and terminal
deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays were
used to determine the presence of apoptosis. Cells (2 10
5
) were plated on
100-mm
2
plates. After 24 h, cells were treated with fresh drug-free medium,
doxorubicin for 24 h, roscovitine for 48 h, or doxorubicin for 24 h then
roscovitine for 48 h as described above. Cells were harvested and stained
with Annexin V-APC and PI (Trevigen, Inc.) or by TUNEL using the APO-
DIRECT kit (BD Biosciences PharMingen) according to the manufacturer’s
instructions (Trevigen, Inc.). Apoptosis was detected by flow cytometry.
Western blot analysis for apoptosis. For Western blot analysis, cells
were homogenized by sonication and high-speed centrifugation. Cell lysate
supernatant was assayed for total protein content and subjected to Western
blot analysis as described by Rao and colleagues (17). Briefly, 25 Ag of protein
from each condition was electropheresed on a 10% SDS-polyacrylamide gel
and transferred to Immunobilon P for 2 h. Membranes were washed at 4jCin
Blotto [5% nonfat dry milk in 20 mmol/L Tris, 137 mmol/L NaCl, 0.25% Tween
(pH 7.6)] overnight. After 6 washes in TBST [20 mmol/L Tris, 137 mmol/L
NaCl, and 0.05% Tween (pH 7.6)], membranes were incubated in primary
antibody (Parp; Cell Signaling) for 2 h (1 Ag/mL in Blotto). Membranes were
washed and incubated with goat anti-mouse horseradish peroxidase
conjugate at a dilution of 1:5,000 for 1 h, washed, and developed with
Renaissance chemiluminescence system as directed by the manufacturers
(NEN Life Sciences Products).
Autophagy assays. U2OS-LC3-GFP cells (1 10
4
) were plated on
coverslips in 6-well plates. After 24 h, cells were treated with fresh drug-free
medium, doxorubicin for 24 h, roscovitine for 48 h, or doxorubicin for
24 h then roscovitine for 48 h. The cells were fixed with 4% para-
formaldehyde, stained with 4’,6-diamidino-2-phenylindole, dilactate (Invi-
trogen), and observed with fluorescence microscopy (Leica DM 4000 B).
To detect autophagy in SK-LMS-1 and SW-982 cells, 1 10
4
cells were
plated on coverslips in 6-well plates. After 24 h, cells were treated with fresh
drug-free medium, doxorubicin for 24 h, roscovitine for 48 h, or doxorubicin
for 24 h then roscovitine for 48 h. The cells were fixed with 4%
paraformaldehyde for 15 min at room temperature then stained with
acridine orange (AO; Polysciences, Inc.), 1 Ag/mL in PBS at 37jC, in the
dark, and observed immediately with fluorescence microscopy. To quantify
the number of cell with acidic vesicles, cells were seeded into 6-well plates
at a density of 4 10
4
cells per well and cultured overnight. The cells were
stained with 1 Ag/mL AO in DMEM at 37jC for 15 min. After incubation,
the cells were washed with PBS and removed with trypsin-EDTA,
resuspended, and analyzed by flow cytometry.
Statistical analysis. Each experiment was repeated at least thrice. The
isobologram analysis and graphs were carried out using Calcusyn software
(version 2.1; Biosoft, Inc.), which performs multiple drug dose-effect
calculations using the median effects method described by T-C Chou and
P. Talalay (18). Combination index (CI) values of <0.9 indicates synergy, CI of
>0.9 and <1.2 indicate additivity, and CI of >1.2 indicate antagonism.
Comparisons among groups were analyzed by two-sided ttest. A difference
of Pvalue of V0.05 was considered to be statistically significant. All analyses
were done with SPSS software, Version 12.0. The data represent the means
of three or more samples with SE.
Results
Synergistic cytotoxicity between doxorubicin and roscovi-
tine. High-throughput clonogenic assay (HTCA) was used to
compare the cytotoxic effects of doxorubicin alone, roscovitine
alone, and doxorubicin followed by roscovitine in SW-982, U2OS-
LC3-GFP, SK-LMS-1 sarcoma, and WI38-immortalized fibroblast
cell lines. A sequential drug treatment strategy (see Materials and
Methods) was chosen based on previous reports in the literature
demonstrating sequence–specific synergistic effects with adminis-
tration of combination chemotherapy, and Cdk inhibitors (19, 20).
As shown in a three-dimensional graph in Fig. 1A, dose-dependent
increases in cell death were seen in all four cell lines when treated
with doxorubicin alone (Xaxis). For SW-982, the percentage of cell
death increased from 5% to 60% as cells were treated with
increasing doxorubicin doses from 0.001 to 0.015 Amol/L (IC
50
,
f0.0125 Amol/L). For U2OS-LC3-GFP, the percentage of cell death
increased from 5% to 85% as cell were treated with increasing
doxorubicin doses from 0.001 to 0.01 Amol/L (IC
50
, between 0.005
and 0.0075 Amol/L). For SK-LMS-1, the percentage of cell death
increased from 7% to 25% as cell were treated with increasing
doxorubicin doses from 0.001 to 0.02 Amol/L (IC
50
, not reached).
For WI38, the percentage of cell death increased from 13% to 50% as
cell were treated with increasing doxorubicin doses from 0.001 to
0.01 Amol/L (IC
50
, 0.01 Amol/L). The percentage of cell death for all
three sarcoma cell lines treated with roscovitine alone ranged
between 5% and 30% as cells were treated with increasing rosco-
vitine doses from 5 to 20 Amol/L (Yaxis). For WI38, the percentage
of cell death increased from 25% to 95% as cell were treated with
increasing roscovitine doses from 5 to 25 Amol/L (IC
50
,10Amol/L).
Synergistic Cytoxicity through Autophagy
www.aacrjournals.org 7967 Cancer Res 2008; 68: (19). October 1, 2008
When the two drugs were combined, there was a significant
increase in cell death (Zaxis) in all three sarcoma cell lines
compared with either doxorubicin or roscovitine alone in isobolo-
gram determinations. However, in the WI38 cells, the combination
of doxorubicin and roscovitine did not result in such a synergistic
cell death. CIs, as determined using the CalcuSyn software, showed
synergistic cytotoxicity between doxorubicin and roscovitine in the
three sarcoma cell lines but not the WI38 cells (Fig. 1B). For
Figure 1. Synergistic effect of doxorubicin
and roscovitine in sarcoma cells. A, HTCA
was used to compare the cytotoxic effects
of doxorubicin alone, roscovitine alone,
and doxorubicin followed by roscovitine in
SW-982, U2OS-LC3-GFP, SK-LMS-1,
and WI38 cell lines [X-axis: doxorubicin
(Dox)Amol/L; Y-axis: roscovitine (Rosc)
Amol/L; Z-axis: percentage cell death].
B, isobologram analysis showed
synergistic interactions in all three sarcoma
cell lines. Isobologram analysis and
graphs were obtained using CalcuSyn
software, which performs drug dose-effect
calculation using the median effect method
described by Chou and Talalay (18).
Cand D, clonogenic assays also showed
a synergistic effect between doxorubicin
and roscovitine in the three cell lines
(Cont, untreated; D, doxorubicin; R,
roscovitine; D+R, doxorubicin plus
roscovitine). Representative of three
experiments.
Cancer Research
Cancer Res 2008; 68: (19). October 1, 2008 7968 www.aacrjournals.org
example, in SW-982 cells, treatment with either 0.005 Amol/L
doxorubicin alone or 10 Amol/L roscovitine alone resulted in a
cell death rate of only 15%. However, when treated first with
0.005 Amol/L doxorubicin then 10 Amol/L roscovitine, the
percentage of cell death increased to 55% (CI, <0.9), suggesting
synergism. Similarly, in U2OS-LC3-GFP, treatment with either 0.005
Amol/L doxorubicin alone or 10 Amol/L roscovitine alone resulted
in a cell death of 30% and 7%, respectively. However, when treated
first with 0.005 Amol/L doxorubicin then 10 Amol/L roscovitine, the
percentage of cell death increased to 75% (CI<0.9). For SK-LMS-1,
treatment with either 0.02 Amol/L doxorubicin alone or 20 Amol/L
roscovitine alone resulted in a cell death of 25%. However, when
treated first with 0.02 Amol/L doxorubicin then 20 Amol/L
roscovitine, the percentage of cell death increased to 64% (CI,
<0.9). Hence, in all three sarcoma cell lines, the combination of
doxorubicin and roscovitine resulted in synergistic cell killings. On
the other hand, for the WI38-immortalized fibroblast cells, the CIs
were predominantly antagonistic (CI, >1.2) and not synergistic. This
finding suggests that the increased cytotoxicity of the combined
treatment is tumor cell specific.
The HTCA was complemented with conventional clonogenic
assays in the sarcoma cell lines, and also showed a synergistic
effect between doxorubicin and roscovitine in all three sarcoma
cell lines (Fig. 1Cand D). For SW-982 cells, there was a 10%
decrease in the number of colonies formed after treatment with
10 Amol/L roscovitine alone, a 30% decrease after treatment with
0.005 Amol/L doxorubicin alone, and a 64% decrease in the number
of colonies formed after treatment with doxorubicin (0.005 Amol/L)
then roscovitine (10 Amol/L; PV0.006). For U2OS-LC3-GFP cells,
there was a 6% decrease in the number of colonies formed after
treatment with 10 Amol/L roscovitine alone, a 64% decrease after
treatment with 0.005 Amol/L doxorubicin alone, and a 82%
decrease in the number of colonies formed after treatment with
doxorubicin (0.005 Amol/L) then roscovitine (10 Amol/L; PV0.001).
For SK-LMS-1 cells, there was no significant change in the number
of colonies formed after treatment with 20 Amol/L roscovitine
alone. There was a 47% decrease after treatment with 0.01 Amol/L
doxorubicin alone and a 85% decrease in the number of colonies
formed after treatment with doxorubicin (0.01 Amol/L) then
roscovitine (20 Amol/L; PV0.008). Fig. 1Cshows representative
clonogenic plates from each experiment, and Fig. 1Dshows
quantization of the clonogenic assays under each condition.
Treatment with doxorubicin followed by roscovitine induces
prolonged G
2
arrest. To determine the cell cycle effects of
doxorubicin and roscovitine, both alone and in combination, DNA
content was evaluated using PI staining followed by flow
cytometry. Cell cycle distributions for the three sarcoma cell lines,
both untreated and treated, are shown in Fig. 2A. After 24 hours of
Figure 2. Effect of doxorubicin and roscovitine on cell cycle. A, SW-982 cells, U2OS-LC3-GFP cells, and SK-LMS-1 cells were treated with either 0.005 Amol/L
(SW-982 and U20S-LC3-GFP) or 0.01 Amol/L (SK-LMS-1) doxorubicin for 24 h, 10 Amol/L (SW-982 and U20S-LC3-GFP) or 20 Amol/L (SK-LMS-1) roscovitine for 48 h,
the combination of doxorubicin and roscovitine, or were untreated. At the completion of drug treatment (T1) and 72 to 96 h after the completion of drug treatment (T2),
cells were harvested and the cell cycle distribution was determined using fluorescence-activated cell sorting (B). Cell cycle distribution histograms of SK-LMS-1.
Representative of three experiments.
Synergistic Cytoxicity through Autophagy
www.aacrjournals.org 7969 Cancer Res 2008; 68: (19). October 1, 2008
treatment with doxorubicin alone, all 3 sarcoma cell lines showed
an increase in the percentage of cells in G
2
-M phase: 5% in SW-982,
6% in U20S-LC3-GFP, and 9% in SK-LMS-1 (Fig. 2B) compared with
untreated cells. After 48 hours of treatment with roscovitine alone,
there was an increase in the percentage of cells in G
1
by 13% for
SW-982. U2OS-LC3-GFP and SK-LMS-1 showed a 7% and 25%
increase in the percentage of cells in G
2
-M phase compared with
untreated cells, respectively. Combined treatment with doxorubicin
for 24 hours followed by roscovitine for 48 hours resulted in an
increase in the percentage of cells in G
2
-M phase compared with
untreated cells by 5% for SW-982, 24% for U2OS-LC3-GFP, and 45%
for SK-LMS-1. Collectively, comparison of the changes in cell cycle
distribution in response to each drug alone versus in combination
suggests significant modulation of the cell cycle pathway by the
combination of the two drugs. To assess the involvement of the cell
cycle as the modulator of the observed synergistic response, we
next performed cell cycle analysis at various time points: at the
completion of drug treatment (T1) and 72 to 96 hours after the
completion of drug treatment (T2).
As shown in Fig. 2A, untreated cells in all three sarcoma cell lines
showed persistent cell division over the time course with increased
percentage of cells residing in G
1
phase at the time of final analysis
(T2), consistent with a contact-induced G
1
cell cycle arrest
(compare control cells in T1 with T2). In addition, the percentage
of cells in G
1
phase also increased in all three sarcoma cell lines
treated with either doxorubicin or roscovitine alone, suggesting the
persistence of cell division after drug treatment (compare
doxorubicin alone cells in T1 with T2). In comparison, after
combined treatment with doxorubicin and roscovitine, SW982 cells
showed a smaller percentage of cells in G
1
compared with the
initial time point (19%) and a persistence of cells in G
2
-M phase
compared with untreated cells at the final time point (T2, 13%
versus 3% of untreated cells). For U2OS-LC3-GFP cells, there was no
significant change in the percentage of cells in G
1
compared with
the initial time point (41% versus 46%, respectively) but a
persistence of cells in G
2
-M phase compared with untreated cells
at the final time point (14% versus 2%). SK-LMS-1 cells showed an
increase in the percentage of cells in G
1
compared with the
combined treatment at the initial time point (19%) as well as a
persistence of cells in the G
2
-M phase (15% versus 3%) compared
with untreated cells at the final time point (Fig. 2B). These findings
translated into a 4-, 7-, and 5-fold increase in the percentage of
SW982, U2OS-LC3-GFP, and SK-LMS-1 cells remaining in G
2
-M
phase, respectively, compared with untreated cells at the final time
point with the combination treatment. These results suggest that
treatment by roscovitine after doxorubicin not only inhibits the cell
cycle but also the ability of the cell to recover from cytotoxic injury.
All of these experiments were performed at least thrice in triplicate
with similar results. Figure 2Bis representative of a single
experiment in the SK-LMS-1 cell line.
Synergistic cytotoxicity by doxorubicin and roscovitine is
not due to increased apoptosis. We detected unique morpho-
logical changes by light microscopy of untreated and treated cells
particularly in U2OS-LC3-GFP and SK-LMS-1 cells (Fig. 3A–B ). As
compared with untreated cells, cells treated with both doxorubicin
and roscovitine became larger, displayed prominent fibrils, and
were often multinucleated (see arrows). Based on the synergistic
cytotoxicity observed with the combined drug treatment, it was
clear that cell death was present but not through apoptosis. Three
different methods were used to determine the degree of cell death
due to apoptosis after treatment with either the single agents or
the combination: Annexin V-APC staining, TUNEL staining, and
expression of Parp (Fig. 4).
As shown in Fig. 4A, there was a significant increase in Annexin
V-APC–positive SW-982 cells after treatment with either doxoru-
bicin or roscovitine alone or in combination compared with
untreated cells (PV0.004). Roscovitine treatment alone lead to
the greatest fold increase in Annexin V-APC–positive SW-982 cells
(4-fold), and this was significant when compared with either
doxorubicin treatment alone or in combination with roscovitine
Figure 3. Morphologic changes observed by light microscopy. A, U2OS-LC3-GFP cells and (B) SK-LMS-1 cells were treated with either 0.005 Amol/L
(U20S-LC3-GFP) or 0.01 Amol/L (SK-LMS-1) doxorubicin for 24 h, 10 Amol/L (U20S-LC3-GFP) or 20 Amol/L (SK-LMS-1) roscovitine for 48 h, the combination of
doxorubicin and roscovitine, or left untreated. At 6 d, the cells were observed by light microscopy. Arrows, enlarged cells with prominent micofibrils and multinucleated
cells seen in combination treatment.
Cancer Research
Cancer Res 2008; 68: (19). October 1, 2008 7970 www.aacrjournals.org
(PV0.021). However, as shown previously, the combined treatment
resulted in greater cell death than either doxorubicin or roscovitine
alone, and because apoptosis was not significantly increased after
the combination treatment, this finding supports the hypothesis of
a nonapoptotic mechanism of synergistic cell death.
In comparison, there was no significant difference in Annexin
V-APC–positive U2OS-LC3-GFP cells after treatment with either
doxorubicin or roscovitine alone or in combination compared with
untreated cells. For SK-LMS-1 cells, there was a significant increase
in the Annexin V-APC–positive cells treated with the combination
of doxorubicin and roscovitine compared with untreated cells and
cells treated with doxorubicin alone (PV0.05). This increase was
not significantly different when compared with roscovitine alone.
However, because the percentage of Annexin V-APC–positive cells
was only 12% in cells treated with the combination of doxorubicin
and roscovitine, these findings also support an alternative
mechanism of synergistic cell death due to the combination
treatment. Similar results were found using the TUNEL assay. SK-
LMS-1 cells showed a slight increase in the TUNEL-positive cells
treated with the combination of doxorubicin and roscovitine. This
Figure 4. A, fluorescence-activated cell
sorting–based apoptosis analyses. SW-982
(top left), U20S-LC3-GFP (bottom left ), and
SK-LMS-1 cells (right) were treated
with either 0.005 Amol/L (SW-982 and
U20S-LC3-GFP) or 0.01 Amol/L (SK-LMS-1)
doxorubicin for 24 h, 10 Amol/L (SW-982
and U20S-LC3-GFP) or 20 Amol/L
(SK-LMS-1) roscovitine for 48 h, the
combination of doxorubicin and roscovitine,
or were untreated. At 6 d, the cells were
harvested and stained with either Annexin
V-APC or TUNEL and subjected to
fluorescence-activated cell sorting. *,
significant difference compared with
untreated cells (P< 0.05); c, significant
difference compared with D + R (P< 0.05).
Representative of three individual experiments.
B, effect of combined treatment with
doxorubicin and roscovitine on apoptosis.
SW-982, U20S-LC3-GFP, and
SK-LMS-1 cells were treated with either
0.005 Amol/L (SW-982 and U20S-LC3-GFP)
or 0.01 Amol/L (SK-LMS-1) doxorubicin for
24 h, 10 Amol/L (SW-982 and U20S-LC3-GFP)
or 20 Amol/L (SK-LMS-1) roscovitine for 48 h,
the combination of doxorubicin and roscovitine,
or left untreated. Western blot analysis using
antibody specific for Parp (full-length and
cleaved) was performed at 0 and 72 h after
treatment. Positive cont, SW-982 cells
treated with 4 Amol/L camptothecin for 12 h.
Representative of three individual experiments.
Synergistic Cytoxicity through Autophagy
www.aacrjournals.org 7971 Cancer Res 2008; 68: (19). October 1, 2008
difference was statistically significant when compared with
doxorubicin alone (P= 0.05) but not roscovitine alone. However,
similar to the Annexin-APC analysis, the percentage of TUNEL-
positive cells was low (4%) in cells treated with the combination of
doxorubicin and roscovitine again supporting an alternative
mechanism of synergistic cell death due to the combination
treatment.
Western blot analysis was also performed to determine whether
the enhanced cell death was occurring through apoptosis. As
shown in Fig. 4B, in all three sarcoma cell lines, the expression of
the cleaved form of the apoptosis marker, Parp, decreased after
treatment with the combination of doxorubicin and roscovitine
compared with either doxorubicin or roscovitine alone. This
finding further suggests that apoptosis is not the only mechanism
of cell death due to the combined treatment of doxorubicin and
roscovitine. Because the morphologic changes we observed with
the drug combination were reminiscent of the homeostatic
condition of autophagy, we explored whether an autophagy-
mediated mechanism of cell death was associated with the
synergistic cytotoxicity.
Synergistic cytotoxicity by doxorubicin and roscovitine is
associated with autophagy. Microtubule-associated protein-1
light chain-3 (LC3), the homologue of the yeast Apg8/Aut7p gene,
localizes on the autophagosomal membrane during autophagy (21).
The LC3-GFP fused protein is used frequently to detect autophagy
through the increased presence of GFP puncta within the
cytoplasm (21). Figure 5Ashows the difference in the presence of
LC3-GFP punctuate structures in U2OS-LC3-GFP cells treated with
doxorubicin alone, roscovitine alone, or with doxorubicin followed
by roscovitine compared with untreated cells. The pictures clearly
Figure 5. Induction of autophagy by combined treatment with doxorubicin and roscovitine. A, U20S-LC3-GFP cells were treated with either 0.005 Amol/L doxorubicin
for 24 h, roscovitine for 48 h, the combination of doxorubicin and roscovitine, or left untreated. Cells were observed under fluorescence microscopy to detect the
presence of LC3-GFP puncta, which are indicative of autophagy. Representative of three separate experiments. B, SK-LMS-1 cells were treated with either 0.01 Amol/L
doxorubicin for 24 h, 20 Amol/L roscovitine for 48 h, the combination of doxorubicin and roscovitine, or left untreated. Seventy-two hours after treatment, cells were stained
with AO and observed with fluorescence microscopy to detect the presence of AO puncta. C, SW-982 and SK-LMS-1 cells were treated with either 0.005 Amol/L
(SW-982) or 0.01 Amol/L (SK-LMS-1) doxorubicin for 24 h, 10 Amol/L (SW-982) or 20 Amol/L (SK-LMS-1) for roscovitine 48 h, the combination of doxorubicin and
roscovitine, or left untreated. Seventy-two hours after treatment, cells were stained with AO and quantified by fluorescence-activated cell sorting. *, significant difference
compared with untreated cells (P< 0.05); c, significant difference compared with D + R (P< 0.05). Representative of three separate experiments.
Cancer Research
Cancer Res 2008; 68: (19). October 1, 2008 7972 www.aacrjournals.org
show that treatment with either drug alone leads to increased
autophagocytic LC3-GFP puncta, compared with untreated U2OS-
LC3-GFP cells. Furthermore, cells treated with doxorubicin alone
had more LC3-GFP puncta than cells treated with roscovitine
alone. However, U2OS-LC3-GFP cells treated with both doxorubicin
and roscovitine showed the most LC3-GFP puncta compared with
cells treated with either drug alone. These findings suggested that
autophagy may be associated with the mechanism of synergistic
cytotoxicity of doxorubicin and roscovitine.
To determine whether autophagy was also present in SW-982
and SK-LMS-1, the cells were stained with AO. AO is concentrated
in acidic vesicles such as the autpohagolysosome and has been
used as a measure of autophagy (22). SK-LMS-1 and SW-982 cells
treated with either doxorubicin or roscovitine developed signif-
icantly increased cytoplasmic AO puncta compared with untreat-
ed cells (Fig. 5B). Furthermore, SK-LMS-1 and SW-982 cells
treated with both doxorubicin and roscovitine showed more AO
puncta compared with cells treated with either doxorubicin or
roscovitine alone. Quantization of AO staining by flow cytometry
showed a significant increase in both the SW-982 and SK-LMS-1
cells treated with both doxorubicin and roscovitine compared
with untreated cells and cell treated with either doxorubicin or
roscovitine alone (PV0.003 for all conditions; Fig. 5C). Taken
together, these results suggested an autophagic mechanism of
synergistic cytotoxicity due to the combined treatment of
doxorubicin and roscovitine.
Discussion
Roscovitine synergistically increases doxorubicin cytotox-
icity in sarcoma cells but not fibroblasts. Doxorubicin is a
commonly used cytotoxic chemotherapy agent with a significant
and use-limiting side effect profile. The ability to increase
doxorubicin efficacy would positively effect many patients suffering
from a variety of cancers. In this report, we show synergistic
cytotoxicity between the Cdk inhibitor roscovitine and doxorubicin
in three different sarcoma cell lines. However, immortalized human
fibroblast cells were not responsive to such synergistic action,
suggesting that the synergism observed is tumor specific. Previous
studies have shown that Cdk inhibition mediates tumor cell–
specific cell cycle arrest and cell death (23, 24). These effects occur
through Cdk inhibition in both the early and late phases of the cell
cycle. For example, within the G
1
phase, Cdk-mediated phosphor-
ylation of Rb leads to E2F-1 transcription factor activation and up-
regulation of the genes required for the transition into S phase (25).
Cdk inhibition during G
1
can lead to G
1
arrest and in S phase can
lead to inappropriately persistent E2F-1, resulting in both S-phase
delay and apoptosis (26).
The G
2
-M transition is also susceptible to Cdk inhibition. Cdk1
activation is essential for progression from the G
2
to M cell cycle
phases, the progression of mitosis through metaphase and cell
survival during the mitotic checkpoint (27, 28). Conditional
knockdown of Cdk1 has been shown to result in extensive DNA
rereplication and apoptosis (29). Inhibition of Cdk1 has also been
shown to increase apoptosis after genotoxic stress, whereas
prolonged Cdk1 inhibition alone can cause significant tumor
cell–specific apoptosis (23, 24). Furthermore, Cdk1 inhibition
during the spindle assembly checkpoint has been shown to cause
tumor cell–specific cell cycle progression without cell division
resulting in mitotic catastrophe (30). Because most conventional
chemotherapy agents cause DNA damage and activate cell cycle
checkpoints, it is logical that disruption of these checkpoints
through a Cdk inhibitor, such as roscovitine, would increase their
cytotoxic effects.
Roscovitine-enhanced doxorubicin cell death is associated
with autophagy. We show here that apoptosis is not the only
mechanism of synergistic cell death caused by the combination of
doxorubicin and roscovitine. Instead, autophagy is significantly
increased in the cells treated with the combination of doxorubicin
and roscovitine. Autophagy is a membrane-trafficking process
which, under normal conditions, degrades cytosolic proteins and
organelles through engulfment into double-membraned vesicles
(autophagosomes). Autophagosomes fuse with lysosomes to form
autolysosomes and the contents are degraded (31). Autophagy is
induced above basal levels by a wide variety of stimuli including
nutrient deprivation and genotoxic stress and has a direct effect on
cell viability. In this study, we have shown that autophagy can also
be induced by the direct Cdk inhibitor, roscovitine, in the setting of
previous cytotoxic treatment.
Cell cycle and autophagy. One of the more intriguing findings
of this study is the induction of autophagy by the combination of
doxorubicin and roscovitine in all three sarcoma cell lines despite
different initial cell cycle effects of the individual drug treatments.
Interestingly, the combined treatment with doxorubicin and
roscovitine lead to a prolonged G
2
-M arrest in all three sarcoma
cell lines. Therefore, we postulate that this prolonged arrest, caused
by the combined activation of the DNA damage checkpoint by
doxorubicin followed by the inhibition of the Cdk1-cyclinB
complex by roscovitine, may be a trigger for the induction of
autophagy and ultimately cell death (Fig. 6).
Figure 6. Proposed mechanism of autophagy-mediated synergistic cell death
by genotoxic chemotherapy and cyclin-dependent kinase inhibition.
Synergistic Cytoxicity through Autophagy
www.aacrjournals.org 7973 Cancer Res 2008; 68: (19). October 1, 2008
The relationship between cell cycle regulation and autophagy is
not yet clearly defined. A variety of agents have shown induction
of autophagy and autophagic cell death in association with a G
2
cell cycle arrest (32, 33). Based on our findings of the combination
treatment of doxorubicin and roscovitine leading to synergistic
cell death, induction of autophagy, and prolonged G
2
cell cycle
arrest, we postulate the following model. DNA damage by
doxorubicin (and potentially other genotoxic chemotherapy agents
or treatments) activates the DNA-PK/ATM/ATR kinases, initiating
two potential Cdk1-cyclin B inactivating cascades: (a) phosphor-
ylation of Cdc25 by CHK1 and CHK2 and (b) (when present)
activation of p53 and up-regulation of direct Cdk1-cyclin B
inhibitors such as 14-3-3j, GADD45, and p21. Injured cells
undergo cell cycle arrest and autophagy to facilitate the repair
of the damaged DNA. Once repaired, passage into mitosis occurs.
However, pharmacological inhibition of the Cdk1-cyclin B complex
by roscovitine also induces cell cycle arrest and autophagy,
thereby preventing the cells from entering mitosis. Furthermore,
due to the pivotal role of Cdk1-cyclin B in the G
2
-M transition, the
G
2
-M DNA damage checkpoint may also be inhibited by
roscovitine. In this manner, autophagy induced by a G
2
cell cycle
arrest in the setting of genotoxic injury and Cdk inhibition, may be
exploitable as a mechanism of cell death. Further investigation
into the mechanism of synergy between cytotoxic agents and Cdk
inhibition as well the role of the cell cycle in the initiation and
outcome of autophagy (e.g., cell survival or cell death) will
potentially provide new approaches to tumor-specific anticancer
therapies.
Disclosure of Potential Conflicts of Interest
L. Meijer: coinventor on patent on roscovitine, licensed to Cyclacel. The other
authors disclosed no potential conflicts of interest.
Acknowledgments
Received 4/9/2008; revised 6/18/2008; accepted 8/4/2008.
Grant support: Grant number CA87458 from the NIH (K. Keyomarsi), National
Cancer Institute P50CA116199 (K. Keyomarsi), and NIH T32 CA009599 (L.A. Lambert).
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank Dr. Robert Bast for the helpful discussions.
Cancer Research
Cancer Res 2008; 68: (19). October 1, 2008 7974 www.aacrjournals.org
References
1. Zahm SH, Fraumeni JF, Jr. The epidemiology of soft
tissue sarcoma. Semin Oncol 1997;24:504–14.
2. Gaynor JJ, Tan CC, Casper ES, et al. Refinement of
clinicopathologic staging for localized soft tissue
sarcoma of the extremity: a study of 423 adults. J Clin
Oncol 1992;10:1317–29.
3. Jaques DP, Coit DG, Hajdu SI, Brennan MF. Manage-
ment of primary and recurrent soft-tissue sarcoma of
the retroperitoneum. Ann Surg 1990;212:51–9.
4. Singer S, Demetri GD, Baldini EH, Fletcher CD.
Management of soft-tissue sarcomas: an overview and
update. Lancet Oncol 2000;1:75–85.
5. Verweij J, Lee SM, Ruka W, et al. Randomized phase II
study of docetaxel versus doxorubicin in first- and
second-line chemotherapy for locally advance d or
metastatic soft tissue sarcomas in adults: a study of
the european organization for research and treatment of
cancer soft tissue and bone sarcoma group. J Clin Oncol
2000;18:2081–6.
6. Rubin E, Hait W. Drugs That Target DNA Top-
oisomerases. In: Kufe D, Bast R, Hait W, et al., editors.
Cancer Medicine 7. Hamilton (Ontario): BC Decker, Inc;
2006. p. 690–3.
7. Sherr CJ. G1 phase progression: cycling on cue. Cell
1994;79:551–5.
8. Melo J, Toczyski D. A unified view of the DNA-damage
checkpoint. Curr Opin Cell Biol 2002;14:237–45.
9. Shi J, Feng Y, Goulet AC, et al. The p34cdc2-related
cyclin-dependent kinase 11 interacts with the p47
subunit of eukaryotic initiation factor 3 during apopto-
sis. J Biol Chem 2003;278:5062–71.
10. Meyerson M, Enders GH, Wu CL, et al. A family of
human cdc2-related protein kinases. EMBO J 1992;11:
2909–17.
11. Meijer L, Borgne A, Mulner O, et al. Biochemical and
cellular effects of roscovitine, a potent and selective
inhibitor of the cyclin-dependent kinases cdc2, cdk2 and
cdk5. Eur J Biochem 1997;243:527–36.
12. Meijer L, Raymond E. Roscovitine and other purines
as kinase inhibitors. From starfish oocytes to clinical
trials. Acc Chem Res 2003;36:417–25.
13. Benson C, White J, De Bono J, et al. A phase I trial of
the selective oral cyclin-dependent kinase inhibitor
seliciclib (CYC202; R-Roscovitine), administered twice
daily for 7 days every 21 days. Br J Cancer 2007;96:29–37.
14. Abal M, Bras-Goncalves R, Judde JG, et al. Enhanced
sensitivity to irinotecan by Cdk1 inhibition in the p53-
deficient HT29 human colon cancer cell line. Oncogene
2004;23:1737–44.
15. Crescenzi E, Palumbo G, Brady HJ. Roscovitine
modulates DNA repair and senescence: implications
for combination chemotherapy. Clin Cancer Res 2005;
11:8158–71.
16. Keyomarsi K, Conte D, Jr., Toyofuku W, Fox MP.
Deregulation of cyclin E in breast cancer. Oncogene
1995;11:941–50.
17. Rao S, Lowe M, Herliczek TW, Keyomarsi K.
Lovastatin mediated G1 arrest in normal and tumor
breast cells is through inhibition of CDK2 activity and
redistribution of p21 and p27, independent of p53.
Oncogene 1998;17:2393–402.
18. Chou T-C, Talalay P. Analysis of combined drug
effects: a new look at a very old problem. Trends Pharm
Sci 1983;4:450–4.
19. Motwani M, Delohery TM, Schwartz GK. Sequential
dependent enhancement of caspase activation and
apoptosis by flavopiridol on paclitaxel-treated human
gastric and breast cancer cells. Clin Cancer Res 1999;5:
1876–83.
20. Shah MA, Kortmansky J, Motwani M, et al. A phase I
clinical trial of the sequential combination of irinotecan
followed by flavopiridol. Clin Cancer Res 2005;11:3836–45.
21. Klionsky DJ, Abeliovich H, Agostinis P, et al. Guide-
lines for the use and interpretation of assays for
monitoring autophagy in higher eukaryotes. Autophag y
2008;4:151–75.
22. Paglin S, Hollister T, Delohery T, et al. A novel
response of cancer cells to radiation involves autophagy
and formation of acidic vesicles. Cancer Res 2001;61:
439–44.
23. Ongkeko W, Ferguson DJ, Harris AL, Norbury C.
Inactivation of Cdc2 increases the level of apoptosis
induced by DNA damage. J Cell Sci 1995;108:2897–904.
24. Vassilev LT, Tovar C, Chen S, et al. Selective small-
molecule inhibitor reveals critical mitotic functions of
human CDK1. Proc Natl Acad Sci U S A 2006;103:
10660–5.
25. Lundberg AS, Weinberg RA. Functional inactivation
of the retinoblastoma protein requires sequential
modification by at least two distinct cyclin-cdk com-
plexes. Mol Cell Biol 1998;18:753–61.
26. Phillips AC, Vousden KH. E2F-1 induced apoptosis.
Apoptosis 2001;6:173–82.
27. Nigg EA. Mitotic kinases as regulators of cell division
and its checkpoints. Nat Rev Mol Cell Biol 2001;2:21–32.
28. O’Connor DS, Wall NR, Porter AC, Altieri DC. A
p34(cdc2) survival checkpoint in cancer. Cancer Cell
2002;2:43–54.
29. Itzhaki JE, Gilbert CS, Porter AC. Construction by
gene targeting in human cells of a "conditional’ CDC2
mutant that rereplicates its DNA. Nat Genet 1997;15:
258–65.
30. Weaver BA, Cleveland DW. Decoding the links
between mitosis, cancer, and chemotherapy: the mitotic
checkpoint, adaptation, and cell death. Cancer Cell 2005;
8:7–12.
31. Baehrecke EH. Autophagy: dual roles in life and
death? Nat Rev Mol Cell Biol 2005;6:505–10.
32. Aoki H, Takada Y, Kondo S, Sawaya R, Aggarwal BB,
Kondo Y. Evidence that curcumin suppresses the growth
of malignant gliomas in vitro and in vivo through
induction of autophagy: role of Akt and extracellular
signal-regulated kinase signaling pathways. Mol Phar-
macol 2007;72:29–39.
33. Kuo PL, Hsu YL, Cho CY. Plumbagin induces G2-M
arrest and autophagy by inhibiting the AKT/mammalian
target of rapamycin pathway in breast cancer cells. Mol
Cancer Ther 2006;5:3209–21.
... Biomedicines 2023, 11, x 9 of 20 CDK-4/6 inhibitor responsiveness in cancer cells [143,144]. The co-administration of palbociclib (a CDK-4/6 inhibitor) and MLN0128 (an mTOR dual-inhibitor) reduced TNBC metastasis in a mouse model [145]. ...
... Cyclin-dependent kinase (CDK)-4/6 inhibitor is an FDA-approved drug for metastatic breast cancer that suppresses the cell cycle. Inhibiting autophagy increases tumor cell death by increasing CDK-4/6 inhibitor responsiveness in cancer cells [143,144]. The co-administration of palbociclib (a CDK-4/6 inhibitor) and MLN0128 (an mTOR dual-inhibitor) reduced TNBC metastasis in a mouse model [145]. ...
Article
Full-text available
Patient morbidity and mortality is significantly increased in metastatic breast cancer. The metastasis process of breast cancer is very complicated and is delicately controlled by various factors. Autophagy is one of the important regulatory factors affecting metastasis in breast cancer by engaging in cell mobility, metabolic adaptation, tumor dormancy, and cancer stem cells. Here, we discuss the effects of autophagy on metastasis in breast cancer and assess the potential use of autophagy modulators for metastasis treatment.
... One such combination is that of camptothecin and doxorubicin [61], which has been shown in lowdose studies to exhibit synergism (with the potential for a 5-fold reduction in dosage) in several breast cancer cell line models in vitro. Other Dox combinations are also used to reduce dosages, increase efficacy, and reduce the toxicity of formulations: (i) gemcitabine and doxorubicin [62]; (ii) BH3-interacting domain death agonist (BID) protein + Dox [63]; (iii) Dox + Roscovitine [64]; and (iv) Dox + Resveratrol [65]. However, our findings are superior to those reported in the literature, as we have improved each component (Dox and ASP) and demonstrated the synergistic effects in the combination in micellar formulations. ...
Article
Full-text available
L-asparaginases (ASP) and Doxorubicin (Dox) are both used in the treatment of leukemia, including in combination. We have attempted to investigate if their combination within the same targeted delivery vehicle can make such therapy more efficacious. We assembled a micellar system, where the inner hydrophobic core was loaded with Dox, while ASP would absorb at the surface due to electrostatic interactions. To make such absorption stronger, we conjugated the ASP with oligoamines, such as spermine, and the lipid components of the micelle—lipoic and oleic acids—with heparin. When loaded with Dox alone, the system yielded about a 10-fold improvement in cytotoxicity, as compared to free Dox. ASP alone showed about a 2.5-fold increase in cytotoxicity, so, assuming additivity of the effect, one could expect a 25-fold improvement when the two agents are applied in combination. But in reality, a combination of ASP + Dox loaded into the delivery system produced a synergy, with a whopping 50× improvement vs. free individual component. Pharmacokinetic studies have shown prolonged circulation of micellar formulations in the bloodstream as well as an increase in the effective concentration of Dox in micellar form and a reduction in Dox accumulation to the liver and heart (which reduces hepatotoxicity and cardiotoxicity). For the same reason, Dox’s liposomal formulation has been in use in the treatment of multiple types of cancer, almost replacing the free drug. We believe that an opportunity to deliver a combination of two types of drugs to the same target cell may represent a further step towards improvement in the risk–benefit ratio in cancer treatment.
... With MK-0752 and doxorubicin, there was a large increase in the percentage of cells in the G2/M phase of the cell cycle suggestive of a G2/M arrest. Prior research using doxorubicin in combination with roscovitine in SK-LMS-1 cells has also demonstrated a G2/M arrest, as have studies looking at doxorubicin in lymphoid and kidney tissue [20,[33][34][35]. Our RNA sequencing data support the findings we saw in cell cycle flow cytometry. ...
Article
Full-text available
Simple Summary Uterine leiomyosarcoma (uLMS) is a rare and aggressive malignancy with treatments that have limited efficacy and often have significant toxicities. We previously demonstrated the presence of Notch signaling in uLMS as well as decreased cell viability and Notch expression after exposure to the gamma secretase inhibitor MK-0752. The aim of this study was to assess the combination of MK-0752 and chemotherapeutics commonly used for the treatment of uLMS. Using two human uLMS cell lines, SK-UT-1B and SK-LMS-1, we demonstrated synergism between MK-0752 and docetaxel, doxorubicin, and gemcitabine using MTT assays. These synergistic combinations were used in transwell invasion assays, cell cycle flow cytometry, proliferation assays, and RNA sequencing. We found decreased invasion, an increase in the apoptotic sub-G1 cell cycle population, and differentially expressed genes and altered KEGG pathways after combination treatment. The combinations of MK-0752 and doxorubicin, MK-0752 and docetaxel, and MK-0752 with gemcitabine and docetaxel are potential novel therapeutic approaches for uLMS. Abstract Due to limited effective therapeutics for uterine leiomyosarcoma (uLMS), the impact of the gamma secretase inhibitor (GSI) MK-0752 with common chemotherapeutics was explored in uLMS. MTT assays were performed on two human uLMS cell lines, SK-UT-1B and SK-LMS-1, using MK-0752, docetaxel, doxorubicin, and gemcitabine, individually and in combination, to determine cell viability after treatment. Synergistic combinations were used in transwell invasion assays, cell cycle flow cytometry, proliferation assays, and RNA sequencing. In SK-UT-1B, MK-0752 was synergistic with doxorubicin and gemcitabine plus docetaxel. In SK-LMS-1, MK-0752 was synergistic with all individual agents and with the combination of gemcitabine plus docetaxel. MK-0752, gemcitabine, and docetaxel decreased invasion in SK-UT-1B 2.1-fold* and in SK-LMS-1 1.7-fold*. In SK-LMS-1, invasion decreased 1.2-fold* after treatment with MK-0752 and docetaxel and 2.2-fold* after treatment with MK-0752 and doxorubicin. Cell cycle analysis demonstrated increases in the apoptotic sub-G1 population with MK-0752 alone in SK-UT-1B (1.4-fold*) and SK-LMS-1 (2.7-fold**), along with increases with all combinations in both cell lines. The combination treatments had limited effects on proliferation, while MK-0752 alone decreased proliferation in SK-LMS-1 (0.63-fold**). Both MK-0752 alone and in combination altered gene expression and KEGG pathways. In conclusion, the combinations of MK-0752 with either doxorubicin, docetaxel, or gemcitabine plus docetaxel are potential novel therapeutic approaches for uLMS. (* p < 0.05, ** p < 0.01).
... Our results are also supported by the findings of other authors which show that doxorubicin induces an autophagic response in cancer cells. 60 Figure 8 summarises the main findings of this study and the proposed mechanism of action of doxorubicin and sanguinarine in breast cancer cells. ...
Article
Full-text available
Breast cancer is the most commonly diagnosed cancer and the second most common cause of cancer death in women. The anthracycline, doxorubicin, is a well-known and highly effective treatment for breast cancer patients; however, many patients present with resistance to chemotherapeutic drugs, which ultimately results in treatment failure and contributes to high mortality rates. It is well established that the mitogen-activated protein kinase phosphatase 1 (MKP-1) mediates the response to chemotherapy, where upregulated MKP-1 is associated with chemoresistance. We investigated whether MKP-1 inhibition or silencing can sensitise triple-negative MDA-MB-231 breast cancer cells to doxorubicin therapy. We found that MKP-1 inhibition and silencing sensitises breast cancer cells to doxorubicin-induced apoptosis. Additionally, the inhibition of MKP-1 in combination with doxorubicin treatment promotes autophagy induction, while doxorubicin and not MKP-1 modulation increased lysosomal acidic compartments. As such, this study demonstrated that MKP-1 inhibition has a potential therapeutic benefit for breast cancer patients by increasing the efficacy of conventional chemotherapy. Therefore, MKP-1 inhibition should be developed as a clinically relevant adjuvant therapy, which could provide a novel avenue for therapeutic intervention in combination with chemotherapy in breast cancer patients.
... Unlike the findings from a previous report [31], however, doxorubicin acted as an inhibitor. Although other studies have also reported doxorubicin-induction of autophagy, this misinterpretation appears to be due to insufficient analyses of autophagic flux, as only the amounts of LC3-II (or the number of LC3 puncta) and formation of autophagosomes, not p62 expression or formation of autolysosome, were determined [31][32][33][34]. Indeed, recent reports have shown that doxorubicin blocks autophagy at the late stage of autophagic flux by impairing lysosomal acidification, so that it increases both LC3-II and p62, which is similar to action of CQ [35][36][37]. ...
Article
Numerous studies have examined the role of autophagy in thyroid cancer treatment; however there are discrepancies among the reported data, with some showing the pro-survival and others the anti-survival effects of autophagy. These discrepant results appear to be at least in part due to insufficient analyses or data misinterpretation as well as improper assessments of autophagic activity. Therefore, the present study re-evaluated the regulation of autophagic activity by various anticancer modalities and examined the role of autophagy in thyroid cancer treatment in three thyroid cancer cell lines (TPC1, ACT1 and KTC1). The immunofluorescence and DalGreen findings demonstrated that cisplatin, irradiation and sorafenib were all autophagy inducers as previously reported, but, unlike previous studies using thyroid cancer cells, doxorubicin acted as an inhibitor. KTC1 cells are unique because they only responded to cisplatin. The efficacy of anticancer therapeutics was significantly higher in chloroquine or 3-methyladenine-treated autophagy-defective cells than in autophagy-competent cells, thereby indicating the pro-survival effect of autophagy induced by anticancer therapeutics, which is partly due to inhibition of apoptosis. Thus, the present findings relating to several anticancer therapeutics and three thyroid cancer cell lines demonstrate the pro-survival effect of autophagy in thyroid cancer treatment. Although the present study only involved cell lines, it provides evidence for the beneficial combination of the anticancer therapeutic modalities with autophagy inhibitors, and proposes that autophagy inhibitors may serve as a possible adjunctive therapy for thyroid cancer.
... Recently, we and other researchers have reported that the combinations of epigenetic inhibitors with radiotherapy is capable of increasing apoptosis in several solid tumors and is regarded as a potential mechanism for radiosensitization (Qiu et al., 2009;Kim et al., 2012Kim et al., , 2014Kwon et al., 2017). Autophagy has been suggested as a novel mechanism of eliciting synergistic cytotoxicity via several chemotherapeutic agents in sarcomas in vitro or in vivo (Lambert et al., 2008;Li et al., 2013). Strikingly, in the present study, we report on the innovative finding that the combination treatment of DNA hypomethylating drugs and radiation is also capable of increasing autophagic cell death in sarcoma cell lines. ...
Article
Full-text available
The targeting of DNA methylation in cancer using DNA hypomethylating drugs has been well known to sensitize cancer cells to chemotherapy and immunotherapy by affecting multiple pathways. Herein, we investigated the combinational effects of DNA hypomethylating drugs and ionizing radiation (IR) in human sarcoma cell lines both in vitro and in vivo. Clonogenic assays were performed to determine the radiosensitizing properties of two DNA hypomethylating drugs on sarcoma cell lines we tested in this study with multiple doses of IR. We analyzed the effects of 5-aza-dC or SGI-110, as DNA hypomethylating drugs, in combination with IR in vitro on the proliferation, apoptosis, caspase-3/7 activity, migration/invasion, and Western blotting using apoptosis- or autophagy-related factors. To confirm the combined effect of DNA hypomethylating drugs and IR in our in vitro experiment, we generated the sarcoma cells in nude mouse xenograft models. Here, we found that the combination of DNA hypomethylating drugs and IR improved anticancer effects by inhibiting cell proliferation and by promoting synergistic cell death that is associated with both apoptosis and autophagy in vitro and in vivo. Our data demonstrated that the combination effects of DNA hypomethylating drugs with radiation exhibited greater cellular effects than the use of a single agent treatment, thus suggesting that the combination of DNA hypomethylating drugs and radiation may become a new radiotherapy to improve therapeutic efficacy for cancer treatment.
Article
Treatment strategies with a strong scientific rationale based on specific biomarkers are needed to improve outcomes in patients with advanced sarcomas. Suppression of cell-cycle progression through reactivation of the tumor suppressor retinoblastoma (Rb) using CDK4/6 inhibitors is a potential avenue for novel targeted therapies in sarcomas that harbor intact Rb signaling. Here, we evaluated combination treatment strategies (sequential and concomitant) with the CDK4/6 inhibitor abemacicib to identify optimal combination strategies. Expression of Rb was examined in 1,043 sarcoma tumor specimens, and 50% were found to be Rb-positive. Using in vitro and in vivo models, an effective two-step sequential combination strategy was developed. Abemaciclib was used first to prime Rb-positive sarcoma cells to reversibly arrest in G1 phase. Upon drug removal, cells synchronously traversed to S phase, where a second treatment with S-phase targeted agents (gemcitabine or Wee1 kinase inhibitor) mediated a synergistic response by inducing DNA damage. The response to treatment could be noninvasively monitored using real-time positron emission tomography imaging and serum thymidine kinase activity. Collectively, these results show that a novel, sequential treatment strategy with a CDK4/6 inhibitor followed by a DNA-damaging agent was effective, resulting in synergistic tumor cell killing. This approach can be readily translated into a clinical trial with noninvasive functional imaging and serum biomarkers as indicators of response and cell cycling. Significance An innovative sequential therapeutic strategy targeting Rb, followed by treatment with agents that perturb DNA synthesis pathways, results in synergistic killing of Rb-positive sarcomas that can be noninvasively monitored.
Article
A coordination compound of CoII with alizarin was prepared to see if it resembles various activities that are reported for anthracyclines as anticancer agents. Alizarin, which has a comparatively simpler structure than anthracyclines, contain the same hydroxy-9,10-anthraquinone as found in anthracyclines. Hence, if tried in cancer chemotherapy, it is expected to show some advantages related to cost and for all those reasons that become applicable in using a relatively simpler compound. However, to be acceptable, it must show comparable efficacy as known for anthracyclines. Like anthracyclines, alizarin forms the semiquinone radical anion that generates superoxide radical anion, responsible for cardiotoxicity. Complex formation decreases generation of superoxide, that was verified by an enzyme assay that might help to decrease toxic side effects. Although hydroxy-9,10-anthraquinones (here alizarin), resemble anthracyclines, biophysical and/or biochemical interactions of the former at the cellular level are less efficient than the latter. Besides, under physiological conditions alizarin is anionic. Hence, increase in pH affects DNA binding adversely. The prepared complex, which is anionic, when it has CoII and neutral when CoIII, binds DNA better than alizarin under varying conditions of ionic strength and pH. Since reactive oxygen species are present in cells, there is a high probability that the CoII complex could either completely or partially convert to CoIII. The CoIII species would then manifest in several biological responses affecting the growth of cells. Experiments pertaining to ROS were performed on three human cell lines (SIHA, HepG2, WI 38) that suggest a shift in mechanism for the complex/complexes from that known for anthracyclines or hydroxy-9,10-anthraquinones. An expected loss in efficacy due to decreased semiquinone formation is compensated by the CoII/CoIII couple owing to the metal ion being present in two oxidation states in cellular medium thus promoting cytotoxicity. The CoII/CoIII complex tried on cells reveal several attributes that enable it/them to overcome some of the shortcomings of alizarin to become comparable to anthracyclines.
Article
Full-text available
Cyclin-dependent kinases (cdk) play an essential role in the intracellular control of the cell division cycle (cdc). These kinases and their regulators are frequently deregulated in human tumours. Enzymatic screening has recently led to the discovery of specific inhibitors of cyclin-dependent kinases, such as butyrolactone I, flavopiridol and the purine olomoucine. Among a series of C2, N6, N9-substituted adenines tested on purified cdc2/cyclin B, 2-(1-ethyl-2-hydroxyethylamino)-6-benzylamino-9-isopropylpurine (roscovitine) displays high efficiency and high selectivity towards some cyclin-dependent kinases. The kinase specificity of roscovitine was investigated with 25 highly purified kinases (including protein kinase A, G and C isoforms, myosin light-chain kinase, casein kinase 2, insulin receptor tyrosine kinase, c-src, v-abl). Most kinases are not significantly inhibited by roscovitine. cdc2/cyclin B, cdk2/cyclin A, cdk2/cyclin E and cdk5/p35 only are substantially inhibited (IC50 values of 0.65, 0.7, 0.7 and 0.2 microM, respectively). cdk4/cyclin D1 and cdk6/cyclin D2 are very poorly inhibited by roscovitine (IC50 > 100 microM). Extracellular regulated kinases erk1 and erk2 are inhibited with an IC50 of 34 microM and 14 microM, respectively. Roscovitine reversibly arrests starfish oocytes and sea urchin embryos in late prophase. Roscovitine inhibits in vitro M-phase-promoting factor activity and in vitro DNA synthesis in Xenopus egg extracts. It blocks progesterone-induced oocyte maturation of Xenopus oocytes and in vivo phosphorylation of the elongation factor eEF-1. Roscovitine inhibits the proliferation of mammalian cell lines with an average IC50 of 16 microM. In the presence of roscovitine L1210 cells arrest in G1 and accumulate in G2. In vivo phosphorylation of vimentin on Ser55 by cdc2/cyclin B is inhibited by roscovitine. Through its unique selectivity for some cyclin-dependent kinases, roscovitine provides a useful antimitotic reagent for cell cycle studies and may prove interesting to control cells with deregulated cdc2, cdk2 or cdk5 kinase activities.
Article
Full-text available
A simple, general and novel method for the quantitative analysis of combined drug effects is presented. The procedure determines if the drug combinations are additive, synergistic or antagonistic, regardless of whether the dose-effect curves are hyperbolic (first order) or sigmoidal (higher order), or whether the effects of the drugs are totally independent or not. We believe that the proposed method (median-effect plot) suffers from fewer restrictions and is more widely applicable than the isobologram or the fractional product method. It provides insight into the nature of the dose-effect relationship with relatively few measurements. We illustrate the procedure by analysing the combined lethal effects of two insecticides on house-flies.
Article
Full-text available
Research in autophagy continues to accelerate, and as a result many new scientists are entering the field. Accordingly, it is important to establish a standard set of criteria for monitoring macroautophagy in different organisms. Recent reviews have described the range of assays that have been used for this purpose. There are many useful and convenient methods that can be used to monitor macroautophagy in yeast, but relatively few in other model systems, and there is much confusion regarding acceptable methods to measure macroautophagy in higher eukaryotes. A key point that needs to be emphasized is that there is a difference between measurements that monitor the numbers of autophagosomes versus those that measure flux through the autophagy pathway; thus, a block in macroautophagy that results in autophagosome accumulation needs to be differentiated from fully functional autophagy that includes delivery to, and degradation within, lysosomes (in most higher eukaryotes) or the vacuole (in plants and fungi). Here, we present a set of guidelines for the selection and interpretation of the methods that can be used by investigators who are attempting to examine macroautophagy and related processes, as well as by reviewers who need to provide realistic and reasonable critiques of papers that investigate these processes. This set of guidelines is not meant to be a formulaic set of rules, because the appropriate assays depend in part on the question being asked and the system being used. In addition, we emphasize that no individual assay is guaranteed to be the most appropriate one in every situation, and we strongly recommend the use of multiple assays to verify an autophagic response.
Article
Full-text available
The p34cdc2 protein kinase is known to regulate important transitions in the eukaryotic cell cycle. We have identified 10 human protein kinases based on their structural relation to p34cdc2. Seven of these kinases are novel and the products of five share greater than 50% amino acid sequence identity with p34cdc2. The seven novel genes are broadly expressed in human cell lines and tissues with each displaying some cell type or tissue specificity. The cdk3 gene, like cdc2 and cdk2, can complement cdc28 mutants of Saccharomyces cerevisiae, suggesting that all three of these protein kinases can play roles in the regulation of the mammalian cell cycle. The identification of a large family of cdc2-related kinases opens the possibility of combinatorial regulation of the cell cycle together with the emerging large family of cyclins.
Article
Full-text available
A number of lines of evidence have suggested a possible involvement of the mitosis-promoting protein kinase Cdc2 in the process of apoptotic cell death, and one recent study concluded that premature activation of Cdc2 is required for apoptosis. Here we have used a temperature-sensitive murine Cdc2 mutant cell line and Cdc2 inhibitor compounds to study the effect of inhibition of this protein kinase on apoptosis induced by DNA-damaging drugs. Inhibition of Cdc2 activity before or during exposure to DNA strand break-inducing drugs had the effect of increasing the level of subsequent apoptosis, as assessed by electron microscopy and flow cytometry. We conclude that, far from being required for cell death, a form of mammalian Cdc2 suppresses apoptosis induced by DNA damage. This form of Cdc2 appears to be active in G2-arrested cells and is therefore presumably distinct from the mitosis-promoting Cdc2-cyclin B heterodimer.
Article
PURPOSEThe prognostic value of factors used in clinicopathologic staging of localized soft tissue sarcoma (STS) of the extremity were analyzed comprehensively.PATIENTS AND METHODS Four hundred twenty-three patients with STS that was confined to the extremity were admitted to Memorial Sloan-Kettering Cancer Center from 1968 to 1978. Cox models for the hazards rates of tumor mortality, development of a distant metastasis, strictly local recurrence, and postmetastasis survival were developed. Tests of changes in the prognostic value of the important variables over time were performed, as well as an analysis of the effect of a local recurrence on the hazard rate of distant metastasis.RESULTSThree unfavorable characteristics contained independent prognostic value for the rates of distant metastasis and tumor mortality: high grade (P less than .00001), deep location (P less than .0002), and size greater than or equal to 5 cm (P less than .007). Their Cox model coefficients did not differ significantly (P greater...
Article
Autophagy is a response of cancer cells to various anticancer therapies. It is designated as programmed cell death type II and characterized by the formation of autophagic vacuoles in the cytoplasm. The Akt/mammalian target of rapamycin (mTOR)/p70 ribosomal protein S6 kinase (p70S6K) and the extracellular signal-regulated kinases 1/2 (ERK1/2) pathways are two major pathways that regulate autophagy induced by nutrient starvation. These pathways are also frequently associated with oncogenesis in a variety of cancer cell types, including malignant gliomas. However, few studies have examined both of these signal pathways in the context of anticancer therapy-induced autophagy in cancer cells, and the effect of autophagy on cell death remains unclear. Here, we examined the anticancer efficacy and mechanisms of curcumin, a natural compound with low toxicity in normal cells, in U87-MG and U373-MG malignant glioma cells. Curcumin induced G(2)/M arrest and nonapoptotic autophagic cell death in both cell types. It inhibited the Akt/mTOR/p70S6K pathway and activated the ERK1/2 pathway, resulting in induction of autophagy. It is interesting that activation of the Akt pathway inhibited curcumin-induced autophagy and cytotoxicity, whereas inhibition of the ERK1/2 pathway inhibited curcumin-induced autophagy and induced apoptosis, thus resulting in enhanced cytotoxicity. These results imply that the effect of autophagy on cell death may be pathway-specific. In the subcutaneous xenograft model of U87-MG cells, curcumin inhibited tumor growth significantly (P < 0.05) and induced autophagy. These results suggest that curcumin has high anticancer efficacy in vitro and in vivo by inducing autophagy and warrant further investigation toward possible clinical application in patients with malignant glioma.
Article
The prognostic value of factors used in clinicopathologic staging of localized soft tissue sarcoma (STS) of the extremity were analyzed comprehensively. Four hundred twenty-three patients with STS that was confined to the extremity were admitted to Memorial Sloan-Kettering Cancer Center from 1968 to 1978. Cox models for the hazards rates of tumor mortality, development of a distant metastasis, strictly local recurrence, and postmetastasis survival were developed. Tests of changes in the prognostic value of the important variables over time were performed, as well as an analysis of the effect of a local recurrence on the hazard rate of distant metastasis. Three unfavorable characteristics contained independent prognostic value for the rates of distant metastasis and tumor mortality: high grade (P less than .00001), deep location (P less than .0002), and size greater than or equal to 5 cm (P less than .007). Their Cox model coefficients did not differ significantly (P greater than or equal to .65); thus, a staging scheme based on the risk of ever developing a distant metastasis would assign equal prognostic weights to grade, depth, and size. The tumor grade effect during the initial 18 months was much larger in magnitude than those for depth and size, and its effect disappeared beyond that time (P = .0003). Thus, a staging scheme based on the risk of early metastatic spread would assign a distinctly larger prognostic weight to grade and lesser but equal weights to depth and size. There was no local recurrence effect on the rate of distant metastasis in the high-risk group (high grade, deep, and greater than or equal to 5 cm; P = .75), but there was a significant association among the remaining groups combined (P = .0039). The magnitude of this association actually increased according to the number of favorable characteristics presented (P = .0024). The refinement of clinicopathologic staging may depend on the choice of outcome variable: ultimate prognosis versus early metastatic spread. Additionally, the observed local recurrence effect may be explained by a tendency for some patients to acquire one or more unfavorable risk factors at the time of local recurrence.
Article
From 1982 to 1987, 114 patients underwent operation at Memorial Sloan-Kettering Cancer Center for soft-tissue sarcoma of the retroperitoneum. A retrospective analysis of these patients defines the biologic behavior, surgical management of primary and recurrent disease, predictive factors for outcome, and impact of multimodality therapy. Complete resection was possible in 65% of primary retroperitoneal sarcomas and strongly predicts outcome (p less than 0.001). The rate of complete resection was not altered by histologic type, size, or grade of tumor. These patients had a median survival of 60 months compared to 24 months for those undergoing partial resection and 12 months for those with unresectable tumors. Forty-nine per cent of completely resected patients have had local recurrence. This is the site of first recurrence in 75% of patients. These patients undergo reoperation when feasible. Complete resection of recurrent disease was performed in 39 of 88 (44%) operations, with a 41-month median survival time after reoperation. Tumor grade was a significant predictor of outcome (p less than 0.001). High-grade tumors (n = 65) were associated with a 20-month median survival time compared to 80 months for low-grade tumors (n = 49). Gender, histologic type, size, previous biopsy, and partial resection versus unresectable tumors did not predict outcome by univariate analysis. Adjuvant radiation therapy and chemotherapy could not be shown to have significant impact on survival. Concerted attempt at complete resection of both primary and recurrent retroperitoneal soft-tissue sarcoma is indicated.