Journal Pre-proof Combination Therapy with Reovirus and ATM Inhibitor Enhances Cell Death and Virus Replication in Canine Melanoma Masaya Igase, Shusaku Shibutani, Yosuke Kurogouchi, Noriyuki Fujiki, Chung Chew Hwang, Matt Coffey, Shunsuke Noguchi, Yuki Nemoto, Takuya Mizuno PII:
S2372-7705(19)30074-9
DOI:
https://doi.org/10.1016/j.omto.2019.08.003
Reference:
OMTO 132
To appear in:
Molecular Therapy: Oncolytics
Received Date: 13 November 2018 Accepted Date: 16 August 2019
Please cite this article as: Igase M, Shibutani S, Kurogouchi Y, Fujiki N, Hwang CC, Coffey M, Noguchi S, Nemoto Y, Mizuno T, Combination Therapy with Reovirus and ATM Inhibitor Enhances Cell Death and Virus Replication in Canine Melanoma, Molecular Therapy: Oncolytics (2019), doi: https:// doi.org/10.1016/j.omto.2019.08.003. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019
1
Original article
2 3
Combination Therapy with Reovirus and ATM Inhibitor Enhances Cell Death and Virus
4
Replication in Canine Melanoma
5 6
Masaya Igase1, Shusaku Shibutani2, Yosuke Kurogouchi3, Noriyuki Fujiki3, Chung
7
Chew Hwang1, Matt Coffey4, Shunsuke Noguchi5, Yuki Nemoto1,3 and Takuya
8
Mizuno1,3
9 10
1
11
of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
12
2
13
University, Yamaguchi, Japan
14
3
15
Medicine, Yamaguchi University, Yamaguchi, Japan
16
4
17
5
18
Sciences, Osaka Prefecture University, Osaka, Japan
Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School
Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi
Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary
Oncolytics Biotech Inc., Calgary, Alberta, Canada Laboratory of Veterinary Radiology, Graduate School of Life and Environmental
19 20
Correspondence should be addressed to T. M. (
[email protected]).
21
Takuya Mizuno, D.V.M., Ph.D., Professor
22
Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary
23
Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
24
Tel & Fax: +81-83-933-5894. E-mail:
[email protected]
1
25 26
Short title: Combined therapy with reovirus and ATM inhibitor
2
27
ABSTRACT
28
Oncolytic virotherapy using reovirus is a promising new anti-cancer treatment with
29
potential for use in humans and dogs. Because reovirus monotherapy shows limited
30
efficacy in human and canine cancer patients, the clinical development of a combination
31
therapy is necessary. To identify candidate components of such a combination, we
32
screened a 285-compound drug library for those that enhanced reovirus cytotoxicity in a
33
canine melanoma cell line. Here, we show that exposure to an inhibitor of the ataxia
34
telangiectasia mutated protein (ATM) enhances the oncolytic potential of reovirus in 5
35
of 6 tested canine melanoma cell lines. Specifically, the ATM inhibitor potentiated
36
reovirus replication in cancer cells along with promoting the lysosomal activity,
37
resulting in an increased proportion of caspase-dependent apoptosis and cell cycle arrest
38
at G2/M compared to those observed with reovirus alone. Overall, our study suggests
39
that the combination of reovirus and the ATM inhibitor may be an attractive option in
40
cancer therapy.
3
41
INTRODUCTION
42
Canine melanoma is among the most aggressive cancers, exhibiting the ability to
43
readily metastasize to other organs such as the lung, lymph nodes, and liver. Naturally
44
occurring canine melanoma is considered a preclinical model of human melanoma
45
Therefore, new therapeutic approaches with efficacy against canine cancers have
46
potential for application in the treatment of human cancers.
1,2
.
47
Oncolytic virotherapy has been studied extensively in human cancers. In 2015, a
48
gene-modified herpes simplex virus-1, talimogene laherparepvec (T-VEC), was
49
approved by the U.S. Food and Drug Administration (FDA) as a treatment for
50
melanoma in patients with unresectable lesions of the skin and lymph nodes 3.
51
Oncolytic viruses are considered attractive treatment options because such reagents are
52
known to activate anti-tumor innate and/or adaptive immune effector cells
53
multiple clinical trials have examined the efficacy against human cancers of oncolytic
54
viruses in combination with chemotherapeutic drugs, radiation, and small molecule
55
inhibitors
56
therapy with oncolytic viruses for treating human cancers.
4-7
. Indeed,
8-10
. There still needs to identify new candidate drugs for use in combination
57
In the veterinary field, there have been only a limited number of reports regarding
58
the use of oncolytic viruses against canine cancer, but several pre-clinical trials
59
suggesting possible application in human medicine have been reported recently
60
Reovirus is a double-stranded RNA virus with low pathogenicity in humans. Our
61
laboratory has previously reported that REOLYSIN®, a GMP-grade non-modified
62
reovirus serotype 3 Dearing strain reovirus, exhibits efficacy in the treatment of canine
63
cancers including mast cell tumors, lymphoma, mammary gland tumors, and melanoma
64
when tested in vitro and in vivo in mouse xenograft models
4
15-18
11-14
.
. We also have carried
65
out pilot clinical studies using REOLYSIN® to treat 19 dogs with spontaneously
66
occurring tumors, demonstrating that reovirus therapy was safe and well-tolerated in
67
tumor-bearing dogs 19. Although decreased tumor volume was observed in some of the
68
reovirus-treated dogs, complete tumor regression was not seen in any of the enrolled
69
dogs. REOLYSIN® has been used in multiple clinical trials in human cancer patients,
70
primarily in combination with chemotherapeutic agents, with the intent of enhancing the
71
efficacy of oncolytic therapy
72
enhance reovirus oncolysis for the treatment of dogs and humans with tumors.
10,20
. In general, other therapeutic options are needed to
73
Therefore, the objective of our current study was to develop a new combination
74
approach for oncolytic virotherapy using reovirus in canine cancers. By screening a
75
large number of small molecule inhibitors in combination with reovirus, we
76
successfully identified a novel inhibitor of the ataxia telangiectasia mutated protein
77
(ATM). Here, we report the first evidence to our knowledge that the cytotoxicity of
78
reovirus is potentiated by inhibition of ATM in canine melanoma cell lines. We also
79
show that ATM inhibition increases reovirus replication, the endosomal acidification
80
and the cathepsin B activity. Notably, reovirus was able to induce the phosphorylation
81
of ATM without inducing DNA damage. Thus, our study demonstrated that the
82
combination of reovirus and an ATM inhibitor may be an attractive option in cancer
83
therapy.
5
84
RESULTS
85
The combination of an ATM inhibitor and reovirus enhances anti-tumor effects in
86
cell lines
87
To identify drugs that enhance reovirus-induced anti-tumor effects, we screened a
88
285-compound signaling pathway inhibitor library for activity in the CMeC1 canine
89
melanoma cell line (Figure S1). This screen revealed that the ATM inhibitor KU55933
90
showed no effect on cell proliferation by itself, but potentiated the cytotoxicity of
91
reovirus when used in combination with reovirus. Moreover, the combination of
92
KU55933 and reovirus yielded dose-dependent suppression of CMeC1 cell growth
93
(Figure S2). For subsequent experiments, a higher specificity inhibitor of the ATM,
94
KU60019 21 was used in place of KU55933.
95
To confirm if KU60019 enhances reovirus-induced anti-tumor effects in other
96
types of canine melanoma cell lines, we also examined in vitro cell survival using
97
another 5 canine melanoma cell lines (Figure 1). KU60019 combined with reovirus
98
(multiplicity of infection (MOI) 100) significantly suppressed cell proliferation in
99
CMeC1, KMeC, CMM12, LMeC, and CMM10 cell lines, as shown with KU55933.
100
These results indicated that the combination of KU60019 and reovirus yielded
101
significant cell growth inhibition compared to compound or virus alone in 5 of 6 tested
102
canine melanoma cell lines excepting CMGD2. These data provided evidence that
103
combination treatment with reovirus and ATM inhibitor potentiated anti-tumor activity
104
in canine melanomas.
105
To confirm the specificity of the ATM inhibitor, the ATM-encoding gene was
106
knocked out in several canine melanoma cell lines (CMeC1, KMeC, CMM10, and
107
CMM12) using a CRISPR/Cas9-based system (Figure 2A). At 48 hr after infection with
6
108
reovirus, ATM knock-out cells showed greater inhibition of cell viability than did the
109
respective ATM+ cells (Figure 2B). Whereas the ATM knock-out did not influence on the
110
cell growth in CMeC1 and CMM10 compared to ATM+ (wild type) cells, but the growth
111
of KMeC and CMM12 were suppressed to some extent (Figure S3). These results
112
supported that inhibition of ATM sensitizes canine melanoma cell lines to
113
reovirus-induced cytotoxicity.
114 115
KU60019 enhances reovirus-induced apoptosis and cell cycle arrest
116
Next, we assessed the potential mechanism by which the combination of KU60019
117
and reovirus induces anti-tumor effects. In addition to cell proliferation assay, cell
118
viability was examined in 4 of these cell lines (CMeC1, KMeC, LMeC, and CMGD2)
119
by means of a trypan blue exclusion test (Figure 3A). Compared to reovirus alone, the
120
combination of KU60019 and reovirus strongly potentiated cell death in 3 of the 4
121
canine melanoma cell lines, excepting CMGD2. At the earlier time point (24 hr),
122
cleaved caspase-3 accumulated to higher levels in CMeC1 cells subjected to the
123
combination treatment than in the same cell line treated with reovirus alone (Figure 3B).
124
As expected, the percentage of sub-G1 cells increased from 24 hr to 48 hr in CMeC1
125
cells subjected to the combination treatment compared to the percentage in cells treated
126
with reovirus alone (Figure 3C). Notably, pre-treatment of CMeC1 and KMeC with 50
127
µM Z-VAD-FMK (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone,
128
an irreversible caspase inhibitor), before exposure to the combination of reovirus and
129
KU60019, resulted in the complete inhibition of reovirus-induced cell death, as assessed
130
by the trypan blue exclusion test (Figure 3D). However, in a cell proliferation assay, the
131
combination treatment was not sufficient to completely abrogate cell growth in either of
7
132
these cell lines (Figure 3E). Therefore, we inferred that the combination of reovirus and
133
KU60019 also affects cell cycle progression in canine melanoma cells.
134
Consistent with previous studies showing that reovirus infection results in cell 22-26
135
cycle arrest at G2/ M in several cancer cell lines
136
reovirus alone induced cell cycle arrest at G2/ M in CMeC1. Moreover, treatment of
137
CMeC1 with the combination of KU60019 and reovirus resulted in strong cell cycle
138
arrest at G2/ M and a reduction of the G1 interval (Figure 4A). In a previous report, the
139
viral nonstructural protein sigma 1s regulated the activity of CDK1 (the G2/ M
140
cyclin-dependent kinase; cdc2), which was required for cell cycle arrest at G2/ M 23. We
141
sought to extend that analysis by using immunoblotting to test treated cells for their
142
levels of CDK1 and p21 (the cyclin-dependent kinase inhibitor) (Figure 4B). After 24 hr,
143
cells treated with the combination of KU60019 and reovirus accumulated significantly
144
lower levels of CDK1 than cells treated with either reagent alone. In contrast, the 24-hr
145
levels of p21 did not differ significantly among cells treated with the combination or
146
with KU60019 or reovirus alone. Together, these results indicated that the treatment of
147
canine melanoma cells with the combination of KU60019 and reovirus induces both
148
caspase-dependent apoptosis and cell cycle arrest at G2/M.
, we observed that treatment with
149 150
KU60019 increases reovirus replication and progeny virus number
151
Next, we assessed the correlation between reovirus replication and anti-tumor
152
effect in canine melanoma cell lines. Western blotting detected reovirus proteins in
153
reovirus-infected CMeC1 and KMeC cells, and further revealed that combination
154
treatment (i.e., reovirus infection in the presence of KU60019) yielded increased levels
155
of these proteins at each time point (Figure S4). This analysis was extended by
8
156
quantifying the progeny virus in the supernatant using the TCID50 assay. In 3 of 4 tested
157
canine melanoma cell lines (CMeC1, KMeC, and CMGD2, but not in LMeC), the
158
combination treatment yielded apparent increases in the number of progeny viruses
159
compared to the input titer (Figure 5A). There was no statistical significance between
160
reovirus alone and combination of KU60019 because the fold increase of virus titer was
161
modest. To prove that viral replication is involved in the cell death effect of the
162
combination of reovirus and KU60019, we used western blotting to assess the
163
accumulation of reovirus structural proteins and cleavage of caspase-3 in CMeC1
164
subjected to the combination treatment in the presence of ribavirin, an anti-viral reagent
165
27
166
caspase-3 cleavage (Figure 5B).
. Notably, ribavirin treatment completely suppressed both reovirus replication and
167 168
The combination of KU60019 and reovirus dose not enhance reovirus entry
169
To investigate the biological roles and functions of ATM in the reovirus replication
170
cycle, each step of the replication cycle was analyzed during the treatment with reovirus
171
and ATM inhibitor. The upregulation of JAM-A, the reovirus receptor, might be
172
associated with sensitivity to virus-induced cell death 28,29. The surface expression level
173
of JAM-A was stable on cells treated with KU60019 for 1 hr and 24 hr by flow
174
cytometric analysis (Figure S5). This data indicated that KU60019 did not influence the
175
step of virus entry.
176
In addition to test whether the effect of ATM inhibition was mediated via the
177
proteolytic step of reovirus replication, we examined the enhancement of viral protein
178
expressions using protease-stripped reovirus virions (infectious subviral particles,
179
ISVPs), which are capable of infecting cells lacking virus receptors and cells that have
9
30-32
180
lost their ability to un-coat incoming virions
181
treatment of reovirus and KU60019 enhanced the expression of reoviral proteins at 12
182
and 24 hr compared to cells exposed to reovirus alone. In contrast, subsequent western
183
blotting (Figure 5D) demonstrated that KU60019 showed no effect on reoviral protein
184
expressions after addition of ISVPs at early time points (at 6 hr and 12 hr), indicating
185
that KU60019 influences an event before viral disassembly. However, at 24 hr, reovirus
186
protein levels were elevated in CMeC1 cells treated with the combination of KU60019
187
and ISVPs; at 48 hr, the combination treatment yielded significant potentiation of cell
188
growth inhibition compared to either component alone (Figure S6), in parallel to the
189
results of Figure 1. We infer that this delayed effect (at 24 hr and after) reflects the
190
production of and infection by intact virus after one replication cycle (approximately 18
191
hr
192
treatment. These results suggested that outer capsid proteins and the ATM inhibition
193
during the reovirus life-cycle might trigger the enhancement of virus replication.
33
. As shown in Figure 5C, combination
), given that the production of intact virus was potentiated by the combination
194 195
Reovirus infection induces the phosphorylation of ATM without phosphorylation of
196
DNA damage-related proteins
197
Infection by some viruses has been shown to induce phosphorylation of ATM, 34-36
198
resulting in the induction of DNA damage
199
activity to replication of mammalian reovirus remains unclear. At an early stage of
200
infection (12 hr), reovirus induced phosphorylation of ATM in cell line CMeC1, but this
201
modification was abrogated by KU60019 (Figure 6). In addition, we examined whether
202
reovirus induced DNA damage in cancer cells by determining the phosphorylation state
203
of p53, that is known to be regulated by ATM and are known to detect DNA damage 37.
10
; however, the mechanism linking ATM
204
Notably, reovirus did not induce phosphorylation of p53 in a canine melanoma cell line
205
(Figures S7). This observation suggested that reovirus infection leads to increases in the
206
phosphorylation of ATM in cancer cells without associated DNA damage, leading to the
207
enhancement of reovirus replication.
208 209
Endosomal acidification and protease activity by the combination of KU60019 and
210
reovirus
211
For early phase of infection, disassembly of the reovirus particle in the endosome
212
is required for the replication cycle following internalization of the virus and depends
213
on acidification of the endosome 38,39. To analyze the effect of the ATM inhibitor on the
214
pH of the endosome, we used LysoTracker-Red, a dye which exhibits increased
215
fluorescence at the low pH typical of endosomes, lysosomes or authophagosomes
216
(Figure 7A and 7B). Although the fluorescence intensity of LysoTracker-Red was not
217
significantly altered compared to mock-treated cells following treatment with either
218
reovirus or KU60019 alone, fluorescence was significantly increased with the
219
combination treatment (Figure 7B). Confocal microscopy confirmed this result,
220
showing that the number of LysoTracker-positive dots was elevated in cells treated with
221
combination of reovirus and KU60019 (Figure 7B). These data suggested that the
222
combination treatment potentiated the acidification of endosomes/lysosomes.
223
The endosomal acidification modulates the activity of some proteases such as
224
cathepsins 39-41. Therefore, we evaluated the cathepsin B activity in CMeC1 cells treated
225
as in the above experiments. As unexpected, the cathepsin B activity in cells treated
226
with KU60019 for 1 hr was little difference as compared to combination treatment of
227
reovirus (data not shown). Cells exposed to reovirus alone for 24 hr incubation
11
228
exhibited nominal decreases in cathepsin B activity, although the effect fell short of
229
significance (p = 0.06 compared to mock-treated cells; Figure 7C). However,
230
combination treatment restored the level of cathepsin B activity to a value similar to that
231
seen with mock treatment. Even though the effect of the simultaneous treatment with
232
reovirus and KU60019 on cathepsin B activity remains unclear, these results implied
233
that the effect of KU60019 treatment is mediated by enhanced the lysosomal activity in
234
the late phase of infection. Thus, combination treatment potentiated the decreased pH in
235
endosomes and lysosomes, followed by activating proteases, but these effects did not
236
commit to enhance viral proteolysis during virus entry step of first viral infection.
12
237
DISCUSSION
238
In this study, we screened a drug library to identify new drugs that enhance
239
reovirus cytotoxicity. As a result, we identified an ATM inhibitor. Previous reports
240
demonstrated that many kinds of anti-cancer drugs have synergistic anti-tumor effects
241
on cancer cells when combined with reovirus
242
represents the first report to our knowledge that an ATM inhibitor can be used as a
243
combination drug with reovirus treatment. Based on our observations, the combinatorial
244
effects of reovirus and the ATM inhibitor resulted in reovirus-related apoptosis and cell
245
cycle arrest at G2/M in most canine melanoma cell lines, with the exception of CMGD2
246
(a canine melanoma line). The specificity of the ATM inhibitor in our study was
247
supported by a cell survival assay that treated ATM knock-out canine melanoma cell
248
lines with reovirus. Canine melanoma knock-out cell lines exhibited higher
249
susceptibility to reovirus compared to the isogenic wild-type (ATM+) cell lines.
250
However, the cell growth inhibition of reovirus combined with KU60019 were higher
251
than that of reovirus with ATM knock-out. We hypothesize that KU60019 inhibits not
252
only ATM but also other kinases (members of the phosphatidylinositol 3-kinase
253
(PI3K)-related kinase (PIKK) family))
254
high-efficiency virus replication and cytotoxicity.
6,26,42-45
. However, the present work
21
, in a dose-dependent manner, leading to
255
We further examined the effect of the combination treatment on the ATM
256
phosphorylation that is associated with reovirus replication in cancer cells. These
257
experiments were inspired by observations of the effects of human herpes simplex
258
virus-1 (HSV-1), hepatitis C virus and human immunodeficiency virus-1 infection in
259
other cell lines
260
supports viral replication, such that the inhibition of ATM negatively regulates
46-49
. For those viruses, ATM activation along with DNA damage
13
261
replication. In contrast, it was unknown whether ATM activation is associated with
262
mammalian reovirus replication. We are aware of only one relevant report, which
263
demonstrated that two ATM-deficient cell lines (L3 lymphoblastoid cells and Granta
264
mantle cell lymphoma cells) were more susceptible to reovirus than were other cell lines
265
50
266
deficiency was not proven in that study. Therefore, in the present work, we identified a
267
putative mechanism for the potentiation of reoviral oncolysis by the normal status and
268
knock-out of ATM. Interestingly, p53 phosphorylation, a marker of DNA damage, was
269
not observed in canine melanoma cells even though reovirus infection might trigger
270
unknown nuclear damage followed by activation of ATM. To confirm the biological
271
roles and functions of ATM in the reovirus replication cycle, each step of the replication
272
cycle was analyzed. We found that inhibition of ATM enhanced the endosomal
273
acidification and re-activation of protease only in late phase of reovirus infection.
274
Previous work has shown that endosomal acidification and protease activity are
275
essential for reovirus replication in early time points
276
another mechanism. Reoviridae family members including mammalian reovirus
277
facilitate the induction of autophagy and its autophagy elevates the virus replication 52-55.
278
To assess the effect of combination treatment of KU60019 and reovirus on autophagy,
279
we have examined the expression levels of the autophagy marker LC3B-II (the lipidated
280
form of LC3B) in CMeC1 and KMeC by the western blotting (data not shown). As a
281
result, the accumulation of LC3B-II was observed in canine melanoma cells treated with
282
reovirus alone at 24-hr and 48-hr, and then treatment of reovirus with KU60019
283
increased the LC3B-II expression. Although the overall mechanism of virus replication
284
induced by ATM inhibition remains unclear, these data suggested that, at late phase of
. However, the relationship between reovirus replication, oncolysis, and ATM
14
31,39,51
, therefore there might be
285
infection, combination of reovirus and KU60019 facilitates the induction of autophagy
286
(ie: the upregulation of cathepsin B activity, the acidification of ensdosomes/lysosomes
287
and the LC3B-II accumulation), which might be associated with enhancement of virus
288
replication.
289
Compared to KU55933 (the library-identified compound), KU60019 has greater
290
specificity and is considered a much improved ATM kinase inhibitor. However, neither
291
KU55933 or KU60019 are suitable for in vivo studies, given the low aqueous solubility
292
and low bioavailability of these compounds
293
recently reported
294
for oral dosing; this compound is the subject of an ongoing phase I clinical trial being
295
performed by AstraZeneca. We hope to evaluate the combination of reovirus and
296
AZD0156 in xenograft mice harboring canine melanoma cell lines.
57
56-58
. The ATM inhibitor AZD0156 was
and shown to exhibit superior pharmacokinetics with the potential
297
Taken together, our results suggested that the ATM inhibitor KU60019 enhances
298
reovirus-induced anti-tumor effects in canine cancers (Figure 8). This synergy may
299
provide a route to resolving the limited efficacy of reovirus treatment of cancers.
300
Although the mechanism linking ATM activity, endosomal acidification and reovirus
301
replication remains unknown, we confirmed that treatment with the ATM inhibitor
302
up-regulated viral replication, resulting in increased the cell death. Given that the
303
incidence of adverse events may be increased in combination treatment, in vivo
304
experiments (e.g., mouse xenograft studies) will be needed to test this combination
305
therapy. Nonetheless, these findings support further investigation of this combination
306
therapy for the treatment of canine cancer patients.
15
307
MATERIALS AND METHODS
308
Cell, reovirus, and inhibitors Seven canine malignant melanoma cell lines (CMeC1, KMeC, LMeC 59, CMGD2
309 310
60
311
(CMeC1, KMeC, LMeC, CMM10 and CMM12) were kindly provided by Dr. T.
312
Nakagawa (The University of Tokyo, Tokyo, Japan). CMGD2 was kindly provided by
313
Dr. J. Modiano (University of Minnesota, Minneapolis, MN). A mouse L929
314
fibroblastic cell line (used for the titration of viral progeny) was obtained from the Cell
315
Resource Center for Biomedical Research (Institute of Development, Aging and Cancer,
316
Tohoku University, Sendai, Japan). All cell lines were grown in R10 complete medium
317
(RPMI1640 supplemented with 10% Fetal Bovine Serum (FBS), 100 U/ml penicillin,
318
100 µg/ml streptomycin, and 55 µM 2-mercaptoethanol) and maintained at 37 °C in a
319
humidified 5 % CO2 incubator.
, CMM10, and CMM12
61
) were used in this study. Five of the canine cell lines
320
The Dearing strain of reovirus serotype 3 (Reolysin®; GMP-grade reovirus) was
321
obtained from Oncolytics Biotech, Inc. (Calgary, Canada). Infectious subviral particles
322
(ISVPs) were produced just before use, and were generated according to the protocol of
323
Alain et al. (2006)
324
digested with chymotrypsin-HCl (2 µg; Roche Diagnostics K.K., Tokyo, Japan) for 30
325
min at 37 °C. The digestion was terminated by adding phenymethylsulphonyl fluoride
326
(5 mM in ethanol) and shifting the reaction to 4 °C.
327 328
30
. Briefly, reovirus (2.835 x108 Plaque-forming unit (PFU)) was
The ATM inhibitors KU55933 and KU60019 were obtained from AdooQ Bioscience (Irvine, CA).
329 330
Drug screening
16
331
To identify the new combination drug, we screened the SCADS inhibitor kit (The
332
Ministry of Education, Culture, Sports, Science and Technology, Tokyo, Japan), which
333
includes 285 signaling pathway inhibitors. The canine melanoma cell line CMeC1
334
(5,000 cells/ well) was plated in 96-well plates containing each inhibitor, with or
335
without reovirus at a MOI of 100 PFU per cell, and the plates were incubated for 48 hr.
336
After treatment, cell growth inhibition was evaluated by use of the Cell Counting Kit-8
337
(CCK-8; Dojindo, Kumamoto, Japan) according to the manufacturer’s instructions.
338 339
Cell survival assay
340
Each cell line was seeded at the density of 5,000 cells (CMeC1, KMeC, LMeC,
341
CMGD2, and CMM12) or 10,000 cells (CMM10) in triplicates in 96-well plates and
342
mock-infected or infected with reovirus at a MOI of 10 or 100 PFU per cell in the
343
presence of the ATM inhibitor KU60019 at 1.25, 2.5, 5.0 or 10 µM; assays were
344
performed in triplicate wells. Cells were cultured for 48 hr before adding the CCK-8
345
reagent. The cell proliferation rate was calculated by measuring absorbance. In the
346
cytotoxicity assay, each cell line was treated with reovirus at a MOI of 100 PFU per cell
347
and KU60019 at 2.5 or 10 µM; assays were performed in triplicate wells. Cells were
348
cultured for 48 hr before performing the trypan blue exclusion test.
349
To assess the inhibition of apoptosis induced by treatment with reovirus and
350
inhibitor, canine melanoma cell lines (CMeC1 and KMeC) were seeded at 5,000 cells
351
per well in 96-well plates. The irreversible pan-caspase inhibitor, Z-VAD-FMK
352
(Calbiochem, Billerica, MA) was added to triplicate wells at a concentration of 50 µM
353
for 1 hr before the cells were treated with reovirus (MOI 100) and KU60019 (indicated
354
concentration). After 48 hr, cell proliferation and cell viability were evaluated by
17
355 356
CCK-8 and trypan blue exclusion test, respectively. All experiments were repeated at least three times.
357 358 359
Reovirus replication and progeny virus Reovirus replication was examined by western blotting analysis using anti-reovirus 15-18
). Cell lines were seeded at 3 x 105 cells per
360
antibody (produced by our laboratory
361
well in 6-well plates and treated with reovirus (MOI 100) and KU60019 (2.5 µM) for 24
362
and 48 hr. In some experiments examining the reovirus proteins at early stages, wells
363
were rinsed to remove unbound reovirus following adsorption. Briefly, CMeC1 was
364
plated at 5 x 105 cells per well in 6-well plates and cultured before use. On the
365
following day, cells were exposed to reovirus (MOI 100) or ISVPs (MOI 10) for 1 hr at
366
4 °C. Then, cells were washed and cultured in the presence of KU60019 (2.5 µM) for 6,
367
12, or 24 hr at 37 °C. Cells were harvested at the indicated time points and stored at
368
-80 °C pending use for western blotting.
369
Supernatants of the samples from the cytotoxicity assay were collected and stored
370
at -80°C pending use. Viral progeny in the supernatants were measured using the 50%
371
tissue culture dose (TCID50) assay on L929 cells, as previously described 15-18. Titration
372
of the viral progeny was performed on all the supernatants obtained from three
373
independent cytotoxicity assays.
374 375 376
Genome editing A
small
guide
RNA
(sgRNA)
targeting
canine
ATM,
5'-
377
TAGTTTCAGGATCCCGAATC -3', was designed through the online CRISPR design
378
tool (http://crispr.mit.edu/); the resulting sgRNA was synthesized and subcloned into the
18
62
379
lentiCRISPRv2 plasmid which was gift from Dr. Feng Zhang
(obtained from
380
Addgene (Cambridge, MA) as accession #52961). The plasmid and packaging vectors
381
were transfected to HEK293T to produce the lentivirus expressing an sgRNA targeting
382
ATM. After transduction with the lentivirus, canine melanoma cell lines were cultured in
383
the presence of puromycin (Sigma Aldrich Japan K.K., Tokyo, Japan; 0.6 – 5.0 µg/ml)
384
to select for stably transduced cells. Individual clones were isolated by the limiting
385
dilution method and further analyzed by western blotting to confirm that the cells no
386
longer produced ATM protein. The resulting clones were used for further experiments.
387 388
Flow cytometric analysis
389
To assess the cell cycle distribution, canine melanoma cells (3 x105 cells/ well in
390
6-well plates) were treated with reovirus (MOI 100) and KU60019 (2.5 µM) for 24 and
391
48 hr. Cells were trypsinized and washed in Phosphate-buffered saline (PBS) and fixed
392
in 70 % ice cold ethanol at -30 °C for at least 24 hr. The fixed cells were re-suspended
393
in PBS supplemented with 0.1 mg/ml of RNase (Nacalai Tesque, Kyoto, Japan) and
394
incubated for 30 min at room temperature. The digested cells then were incubated with
395
0.1 mg/ml of propidium iodide (PI; Nacalai Tesque) for 5 min and analyzed.
396
To evaluate surface expression of reovirus receptor Junction adhesion molecule
397
(JAM)-A, a canine melanoma cell line (CMeC1) were seeded at 2 x105 cells per well in
398
12-well plates and treated with mock and KU60019 (2.5 µM) for 1 hr or 24 hr.
399
Incubated cells were stained with primary antibodies including mouse IgG1 isotype
400
control (eBioscience) and mouse anti-human JAM-A monoclonal antibody (Santa cruz),
401
followed by incubation with Dylight488-conjugated goat anti-mouse IgG antibody
402
(BioLegend). Those cells were stained with PI to gate out dead cells.
19
403
All stained samples were analyzed using a BD Accuri C6 Flow Cytometer (BD
404
Biosciences). All given data were analyzed using FlowJo software ver. 10.1 (FlowJo
405
LLC, Ashland, OR).
406 407
Western blotting
408
Cells were plated at 3 x105 cells per well in 6-well plates and treated with reovirus
409
(MOI 100) and KU60019 (2.5 µM) for 24 and 48 hr. For short-term incubation, CMeC1
410
cells were plated at 5 x105 cells per well in 6-well plates, and on the following day, cells
411
were treated as shown above (see Reovirus replication and progeny viruses). Harvested
412
cells were lysed at 4 °C with NP40 lysis buffer (1 % NP40, 10 mM Tris-HCl (pH 7.5),
413
150 mM NaCl, 1 mM EDTA) supplemented with protease inhibitor cocktails (Nacalai
414
Tesque), 1 mM Na3VO4, and 50 mM NaF. The resulting supernatants were collected and
415
used as cell lysates. These lysates were subjected to SDS-PAGE, and proteins then were
416
transferred to Hybond PVDF membrane (GE Healthcare Bio-Sciences, Pittsburgh, PA).
417
The membrane was blocked with Tris buffered saline (TBS) containing with 0.05 %
418
Tween20 (Polyoxyethylene sorbitane monolaureate) and 5 % skim milk, followed by
419
addition of the primary antibodies. The primary antibodies are listed in Table S1.
420
Horseradish peroxidase (HRP) -conjugated antibodies were used as the secondary
421
antibodies. The labeled proteins were detected using the ECL reagent (PerkinElmer).
422
The intensity of the bands was determined by ImageJ software ver. 1.48.
423 424
LysoTracker-Red staining
425
CMeC1 cells (1 x105 cells) were cultured on microcover glasses (Matunami
426
Garasu Kogyo, Osaka, Japan) in the wells of a 12-well plate. On the following day, cells
20
427
were exposed to reovirus (MOI 100) for 1 hr at 4 °C. Then, cells were washed and
428
cultured in the presence of KU60019 (2.5 µM) for 24 hr at 37 °C. After treatment, cells
429
were incubated with LysoTracker-Red DND-99 (Invitrogen, Carlsbad, CA) for 30 min
430
at 37 °C, followed by fixation with 4 % paraformaldehyde. The coverslips were
431
mounted onto glass slides with Molecular Probes Prolong Gold with DAPI (Invitrogen)
432
for staining of nuclei. Samples were visualized at a 100x magnification using LSM710
433
confocal microscope (Carl Zeiss, Oberkochen, Germany). Images were analyzed by
434
using ZEN software 2012 (Carl Zeiss). The percentage of LysoTracker-Red-positive
435
CMeC1 cells was quantified by flow cytometric analysis as described above.
436 437
Measurement of cathepsin B activity
438
Cathepsin activity was determined with a Cathepsin B Activity Assay Kit
439
(PromoCell, Heidelberg, Germany), according to the manufacturer's instructions.
440
CMeC1 cells were cultured at the density of 5 x105 cells per well in 6-well plates. Then,
441
cells were treated with reovirus (MOI 100) and KU60019 (2.5 µM) for 24 hr. After
442
treatments, cells were lysed with CB Cell Lysis Buffer, followed by mixing with the CB
443
Reaction Buffer. The cell lysate was incubated at 37 °C for 1 hr with 200 µM CB
444
Substrate Ac-RR-AFC. Cathepsin activity was measured in an ARVO X4 fluorometer
445
(PerkinElmer). The fluorescence values were normalized to values obtained for a
446
mock-treated sample after subtraction of background autofluorescence.
447 448
Statistical analysis
449
The mean values and standard deviation (SD) in each assay were calculated from the
450
mean of at least three biological independent experiments. The significance of
21
451
differences between samples were determined by the one-way factorial ANOVA test
452
followed by multiple comparison using Tukey-Kramer test. Differences were
453
considered statistically significant if the p values were less than 0.05. Multiple
454
comparison tests were performed using JMP software ver. 9.0 (SAS Institute Japan,
455
Tokyo, Japan).
22
456 457
CONFLICTS OF INTEREST
458
M.C. is president/chief executive officer and has financial interests with Oncolytics
459
Biotech, Inc. The other authors declare no conflict of interest.
460 461
ACKNOWLEDGMENTS
462
This study was supported by JSPS KAKENHI Grant Number 15H04598 and 16J06655.
463
The SCADS Inhibitor Kit was provided by The Screening Committee of Anticancer
464
Drugs supported by a Grant-in-Aid for Scientific Research on Innovative Areas,
465
Scientific Support Programs for Cancer Research, from The Ministry of Education,
466
Culture, Sports, Science and Technology, Japan. Dr. Takayuki Nakagawa (The
467
University of Tokyo) and Dr. Jaime Modiano (University of Michigan) kindly provided
468
canine melanoma cell lines to the authors. The authors would like to acknowledge the
469
technical expertise of the DNA Core facility of the Center for Gene Research,
470
Yamaguchi University, supported by a grant-in-aid from the Ministry of Education,
471
Science, Sports and Culture of Japan.
472 473
AUTHOR CONTRIBUTIONS
474
M.I. and T.M. conceived the study design and performed all experiments, in addition to
475
writing the paper. S.S and M.C. provided essential resources and developed methods.
476
Y.K. and N.F. obtained additional data for this study. C.C.H., S.N., and Y.N. analyzed
477
the data and provided critical interpretation. All authors read and approved the final
478
manuscript.
23
479
REFERENCES
480
1.
Gillard, M., Cadieu, E., De, Brito, C., Abadie, J., Vergier, B., Devauchelle, P., et
481
al. (2013). Naturally occurring melanomas in dogs as models for non-UV
482
pathways of human melanomas. Pigment Cell Melanoma Res. 27, 90–102.
483
2.
Hernandez, B., Adissu, H.A., Wei, B.R., Michael, H.T., Merlino, G., and
484
Simpson, R.M. (2018). Naturally Occurring Canine Melanoma as a Predictive
485
Comparative Oncology Model for Human Mucosal and Other Triple Wild-Type
486
Melanomas. Int. J. Mol. Sci. 19, 394.
487
3.
melanoma immunotherapy. oncoimmunology 5, e1115641.
488 489
Pol, J., Kroemer, G., and Galluzzi, L. (2016). First oncolytic virus approved for
4.
Marelli, G., Howells, A., Lemoine, N.R., and Wang, Y. (2018). Oncolytic Viral
490
Therapy and the Immune System: A Double-Edged Sword Against Cancer. Front.
491
Immunol. 9, 1565–1568.
492
5.
Virotherapy? Front. Oncol. 7, 106.
493 494
Filley, A.C., and Dey, M. (2017). Immune System, Friend or Foe of Oncolytic
6.
Rajani, K., Parrish, C., Kottke, T., Thompson, J., Zaidi, S., Ilett, L., et al. (2016).
495
Combination Therapy With Reovirus and Anti-PD-1 Blockade Controls Tumor
496
Growth Through Innate and Adaptive Immune Responses. Mol. Ther. 24, 166–
497
174.
498
7.
Mostafa, A.A., Meyers, D.E., Thirukkumaran, C.M., Liu, P.J., Gratton, K.,
499
Spurrell, J., et al. (2018). Oncolytic Reovirus and Immune Checkpoint Inhibition
500
as a Novel Immunotherapeutic Strategy for Breast Cancer. Cancers (Basel) 10,
501
205.
502
8.
Pol, J., Buqué, A., Aranda, F., Bloy, N., Cremer, I., Eggermont, A., et al. (2016).
503
Trial Watch-Oncolytic viruses and cancer therapy. oncoimmunology 5,
504
e1117740.
505
9.
Trial Watch:: Oncolytic viruses for cancer therapy. oncoimmunology 3, e28694.
506 507
Pol, J., Bloy, N., Obrist, F., Eggermont, A., Galon, J., Cremer, I., et al. (2014).
10.
Vacchelli, E., Eggermont, A., Sautès-Fridman, C., Galon, J., Zitvogel, L.,
508
Kroemer, G., et al. (2013). Trial watch: Oncolytic viruses for cancer therapy.
509
oncoimmunology 2, e24612.
24
510
11.
an Oncolytic Myxoma Virus in Dogs with Soft Tissue Sarcoma. Viruses 10, 398.
511 512
MacNeill, A.L., Weishaar, K.M., Séguin, B., and Powers, B.E. (2018). Safety of
12.
Naik, S., Galyon, G.D., Jenks, N.J., Steele, M.B., Miller, A.C., Allstadt, S.D., et
513
al. (2017). Comparative oncology evaluation of intravenous recombinant
514
oncolytic Vesicular Stomatitis Virus therapy in spontaneous canine cancer. Mol.
515
Cancer Ther. 17, 316-326.
516
13.
(2014). Oncolytic virotherapy of canine and feline cancer. Viruses 6, 2122–2137.
517 518
Gentschev, I., Patil, S.S., Petrov, I., Cappello, J., Adelfinger, M., and Szalay, A.A.
14.
Cejalvo, T., Perisé-Barrios, A.J., Del Portillo, I., Laborda, E., Rodriguez-Milla,
519
M.A., Cubillo, I., et al. (2018). Remission of Spontaneous Canine Tumors after
520
Systemic Cellular Viroimmunotherapy. Cancer Res. 78, 4891–4901.
521
15.
Hwang, C.C., Umeki, S., Igase, M., Coffey, M., Noguchi, S., Okuda, M., et al.
522
(2016). The effects of oncolytic reovirus in canine lymphoma cell lines. Vet.
523
Comp. Oncol. 14 Suppl. 1, 61–73.
524
16.
Igase, M., Hwang, C.C., Coffey, M., Okuda, M., Noguchi, S., and Mizuno, T.
525
(2015). The oncolytic effects of reovirus in canine solid tumor cell lines. J. Vet.
526
Med. Sci. 77, 541–548.
527
17.
Igase, M., Hwang, C.C., Kambayashi, S., Kubo, M., Coffey, M., Miyama, T.S.,
528
et al. (2016). Oncolytic reovirus synergizes with chemotherapeutic agents to
529
promote cell death in canine mammary gland tumor. Can. J. Vet. Res. 80, 21–31.
530
18.
al. (2013). Oncolytic reovirus in canine mast cell tumor. PLoS One 8, e73555.
531 532
Hwang, C.C., Umeki, S., Kubo, M., Hayashi, T., Shimoda, H., Mochizuki, M., et
19.
Hwang, C.C., Igase, M., Sakurai, M., Haraguchi, T., Tani, K., Itamoto, K., et al.
533
(2018). Oncolytic reovirus therapy: Pilot study in dogs with spontaneously
534
occurring tumours. Vet. Comp. Oncol. 16, 229–238.
535
20.
therapy: an evidence-based review. Oncolytic Virother. 3, 69–82.
536 537
Clements, D., Helson, E., Gujar, S.A., and Lee, P.W. (2014). Reovirus in cancer
21.
Golding, S.E., Rosenberg, E., Valerie, N., Hussaini, I., Frigerio, M., Cockcroft,
538
X.F., et al. (2009). Improved ATM kinase inhibitor KU-60019 radiosensitizes
539
glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and
540
inhibits migration and invasion. Mol. Cancer Ther. 8, 2894–2902.
541
22.
Clarke, P., Richardson-Burns, S.M., Debiasi, R.L., and Tyler, K.L. (2005). 25
542
Mechanisms of apoptosis during reovirus infection. Curr. Top. Microbiol.
543
Immunol. 289, 1–24.
544
23.
Poggioli, G.J., Dermody, T.S., and Tyler, K.L. (2001). Reovirus-induced
545
sigma1s-dependent G(2)/M phase cell cycle arrest is associated with inhibition of
546
p34(cdc2). J. Virol. 75, 7429–7434.
547
24.
Maitra, R., Seetharam, R., Tesfa, L., Augustine, T.A., Klampfer, L., Coffey,
548
M.C., et al. (2014). Oncolytic reovirus preferentially induces apoptosis in KRAS
549
mutant colorectal cancer cells, and synergizes with irinotecan. Oncotarget 5,
550
2807–2819.
551
25.
Twigger, K., Vidal, L., White, C.L., De, Bono, J.S., Bhide, S., Coffey, M., et al.
552
(2008). Enhanced in vitro and in vivo cytotoxicity of combined reovirus and
553
radiotherapy. Clin. Cancer Res. 14, 912–923.
554
26.
Sei, S., Mussio, J.K., Yang, Q.E., Nagashima, K., Parchment, R.E., Coffey, M.C.,
555
et al. (2009). Synergistic antitumor activity of oncolytic reovirus and
556
chemotherapeutic agents in non-small cell lung cancer cells. Mol. Cancer 8, 47.
557
27.
Rankin, J.T., Eppes, S.B., Antczak, J.B., and Joklik, W.K. (1989). Studies on the
558
mechanism of the antiviral activity of ribavirin against reovirus. Virology 168,
559
147–158.
560
28.
Stiff, A., Caserta, E., Sborov, D.W., Nuovo, G.J., Mo, X., Schlotter, S.Y., et al.
561
(2016). Histone Deacetylase Inhibitors Enhance the Therapeutic Potential of
562
Reovirus in Multiple Myeloma. Mol. Cancer Ther. 15, 830-841.
563
29.
Kelly, K.R., Espitia, C.M., Zhao, W., Wendlandt, E., Tricot, G., Zhan, F., et al.
564
(2015). Junctional adhesion molecule-A is overexpressed in advanced multiple
565
myeloma and determines response to oncolytic reovirus. Oncotarget 6,
566
41275-41289.
567
30.
Alain, T., Kim, M., Johnston, R.N., Urbanski, S., Kossakowska, A.E., Forsyth,
568
P.A., et al. (2006). The oncolytic effect in vivo of reovirus on tumour cells that
569
have survived reovirus cell killing in vitro. Br. J. Cancer 95, 1020–1027.
570
31.
Alain, T., Kim, T.S., Lun, X., Liacini, A., Schiff, L.A., Senger, D.L., et al. (2007).
571
Proteolytic disassembly is a critical determinant for reovirus oncolysis. Mol. Ther.
572
15, 1512–1521.
573
32.
Marcato, P., Shmulevitz, M., Pan, D., Stoltz, D., and Lee, P.W. (2007). Ras 26
574
transformation mediates reovirus oncolysis by enhancing virus uncoating,
575
particle infectivity, and apoptosis-dependent release. Mol. Ther. 15, 1522–1530.
576
33.
potentiates reovirus-induced necrosis. Virology 484, 386–394.
577 578
Hiller, B.E., Berger, A.K., and Danthi, P. (2015). Viral gene expression
34
Passaro, C., Abagnale, A., Libertini, S., Volpe, M., Botta, G., Cella, L., et al.
579
(2013). Ionizing radiation enhances dl922-947-mediated cell death of anaplastic
580
thyroid carcinoma cells. Endocr. Relat. Cancer 20, 633–647.
581
35.
pathways triggered by viral replication. Expert Rev. Mol. Med. 8, 1–11.
582 583
Sinclair, A., Yarranton, S., and Schelcher, C. (2006). DNA-damage response
36.
Hau, P.M., Deng, W., Jia, L., Yang, J., Tsurumi, T., Chiang, A.K.S., et al. (2015).
584
Role of ATM in the formation of the replication compartment during lytic
585
replication of Epstein-Barr virus in nasopharyngeal epithelial cells. J. Virol. 89,
586
652–668.
587
37.
Kozlov, S.V., Graham, M.E., Jakob, B., Tobias, F., Kijas, A.W., Tanuji, M., et al.
588
(2011). Autophosphorylation and ATM activation: additional sites add to the
589
complexity. J. Biol. Chem. 286, 9107–9119.
590
38.
required for efficient reovirus infection. J. Virol. 86, 8346–8358.
591 592
Mainou, B.A., and Dermody, T.S. (2012). Transport to late endosomes is
39.
Sturzenbecker, L.J., Nibert, M., Furlong, D., and Fields, B.N. (1987).
593
Intracellular digestion of reovirus particles requires a low pH and is an essential
594
step in the viral infectious cycle. J. Virol. 61, 2351–2361.
595
40.
Blum, J.S., Fiani, M.L., and Stahl, P.D. (1991). Proteolytic cleavage of ricin A
596
chain in endosomal vesicles. Evidence for the action of endosomal proteases at
597
both neutral and acidic pH. J. Biol. Chem. 266, 22091–22095.
598
41.
internalized proteins. FEBS Lett. 389, 55–60.
599 600
Authier, F., Posner, B.I., and Bergeron, J.J. (1996). Endosomal proteolysis of
42.
Roulstone, V., Twigger, K., Zaidi, S., Pencavel, T., Kyula, J.N., White, C., et al.
601
(2013). Synergistic cytotoxicity of oncolytic reovirus in combination with
602
cisplatin-paclitaxel doublet chemotherapy. Gene Ther. 20, 521–528.
603
43.
Heinemann, L., Simpson, G.R., Boxall, A., Kottke, T., Relph, K.L., Vile, R., et al.
604
(2011). Synergistic effects of oncolytic reovirus and docetaxel chemotherapy in
605
prostate cancer. BMC Cancer 11, 221. 27
606
44.
Hamano, S., Mori, Y., Aoyama, M., Kataoka, H., Tanaka, M., Ebi, M., et al.
607
(2014). Oncolytic reovirus combined with trastuzumab enhances antitumor
608
efficacy through TRAIL signaling in human HER2-positive gastric cancer cells.
609
Cancer Lett. 356, 846–854.
610
45.
Kelly, K.R., Espitia, C.M., Mahalingam, D., Oyajobi, B.O., Coffey, M., Giles,
611
F.J., et al. (2012). Reovirus therapy stimulates endoplasmic reticular stress,
612
NOXA induction, and augments bortezomib-mediated apoptosis in multiple
613
myeloma. Oncogene 31, 3023–3038.
614
46.
Li, H., Baskaran, R., Krisky, D.M., Bein, K., Grandi, P., Cohen, J.B., et al.
615
(2008). Chk2 is required for HSV-1 ICP0-mediated G2/M arrest and
616
enhancement of virus growth. Virology 375, 13–23.
617
47.
Alekseev, O., Donovan, K., and Azizkhan-Clifford, J. (2014). Inhibition of ataxia
618
telangiectasia mutated (ATM) kinase suppresses herpes simplex virus type 1
619
(HSV-1) keratitis. Invest. Ophthalmol. Vis. Sci. 55, 706–715.
620
48.
Machida, K., Cheng, K.T.H., Lai, C.K., Jeng, K.S., Sung, V.M.H., and Lai,
621
M.M.C. (2006). Hepatitis C virus triggers mitochondrial permeability transition
622
with production of reactive oxygen species, leading to DNA damage and STAT3
623
activation. J. Virol. 80, 7199–7207.
624
49.
Damage Response by RNA Viruses. Biomolecules 6, 2.
625 626
Ryan, E.L., Hollingworth, R., and Grand, R.J. (2016). Activation of the DNA
50.
Kim, M., Williamson, C.T., Prudhomme, J., Bebb, D.G., Riabowol, K., Lee,
627
P.W., et al. (2010). The viral tropism of two distinct oncolytic viruses, reovirus
628
and myxoma virus, is modulated by cellular tumor suppressor gene status.
629
Oncogene 29, 3990–3996.
630
51.
Ebert, D.H., Deussing, J., Peters, C., and Dermody, T.S. (2002). Cathepsin L and
631
cathepsin B mediate reovirus disassembly in murine fibroblast cells. J. Biol.
632
Chem. 277, 24609–24617.
633
52.
Thirukkumaran, C.M., Shi, Z.Q., Luider, J., Kopciuk, K., Gao, H., Bahlis, N., et
634
al. (2012). Reovirus as a viable therapeutic option for the treatment of multiple
635
myeloma. Clin. Cancer Res. 18, 4962–4972.
636 637
53.
Huang, W.-R., Chi, P.-I., Chiu, H.-C., Hsu, J.-L., Nielsen, B.L., Liao, T.-L., et al. (2017). Avian reovirus p17 and σA act cooperatively to downregulate Akt by 28
638
suppressing mTORC2 and CDK2/cyclin A2 and upregulating proteasome
639
PSMB6. Sci. Rep. 7, 5226.
640
54.
Kemp, V., Dautzenberg, I.J.C., Limpens, R.W., van den Wollenberg, D.J.M. and
641
Hoeben, R.C. (2017). Oncolytic Reovirus Infection Is Facilitated by the
642
Autophagic Machinery. Viruses 9, 266.
643
55.
starvation accelerates replication of Ibaraki virus. Virus Res. 260, 94–101.
644 645
Onishi, K., Shibutani, S., Goto, N., Maeda, Y. and Iwata, H. (2019). Amino acid
56.
Biddlestone-Thorpe, L., Sajjad, M., Rosenberg, E., Beckta, J.M., Valerie, N.C.K.,
646
Tokarz, M., et al. (2013). ATM kinase inhibition preferentially sensitizes
647
p53-mutant glioma to ionizing radiation. Clin. Cancer Res. 19, 3189–3200.
648
57.
Pike, K.G., Barlaam, B., Cadogan, E., Campbell, A., Chen, Y., Colclough, N., et
649
al. (2018). The Identification of Potent, Selective, and Orally Available Inhibitors
650
of Ataxia Telangiectasia Mutated (ATM) Kinase: The Discovery of AZD0156
651
(8-{6-[3-(Dimethylamino)propoxy]pyridin-3-yl}-3-methyl-1-(tetrahydro-2
652
H-pyran-4-yl)-1,3-dihydro-2 H-imidazo[4,5- c]quinolin-2-one). J. Med. Chem.
653
61, 3823–3841.
654
58.
Vecchio, D., Daga, A., Carra, E., Marubbi, D., Raso, A., Mascelli, S., et al.
655
(2015). Pharmacokinetics, pharmacodynamics and efficacy on pediatric tumors
656
of the glioma radiosensitizer KU60019. Int. J. Cancer 136, 1445–1457.
657
59.
Inoue, K., Ohashi, E., Kadosawa, T., Hong, S.H., Matsunaga, S., Mochizuki, M.,
658
et al. (2004). Establishment and characterization of four canine melanoma cell
659
lines. J. Vet. Med. Sci. 66, 1437–1440.
660
60.
Bianco, S.R., Sun, J., Fosmire, S.P., Hance, K., Padilla, M.L., Ritt, M.G., et al.
661
(2003). Enhancing antimelanoma immune responses through apoptosis. Cancer
662
Gene Ther. 10, 726–736.
663
61.
Yoshitake, R., Saeki, K., Watanabe, M., Nakaoka, N., Ong, S.M., Hanafusa, M.,
664
et al. (2017). Molecular investigation of the direct anti-tumour effects of
665
nonsteroidal anti-inflammatory drugs in a panel of canine cancer cell lines. Vet. J.
666
221, 38–47.
667 668
62.
Sanjana, N.E., Shalem, O., and Zhang, F. (2014). Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784.
669
29
670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723
Figure legends Figure 1. ATM inhibitor KU60019 enhances reovirus-induced cell growth inhibition in canine melanoma cell lines. To evaluate cell proliferation, canine melanoma cell lines (CMeC1, KMeC, LMeC, CMM10, CMM12, and CMGD2) were treated with reovirus (MOI 100 for all cell lines except CMGD2 at MOI 10) and KU60019 (indicated concentration) for 48 hr before adding CCK-8 reagent. Data are expressed as the mean ± SD from at least three independent experiments. p values were calculated for the comparison between reovirus alone and reovirus combined with KU60019. To focus on the additional effects provided by KU60019, significance was tested only where no significant difference was observed between mock control and KU60019 alone. Tukey-Kramer test, *p < 0.05, **p < 0.01. Figure 2. Reovirus susceptibility is enhanced by ATM knock-out. (A) The accumulation of ATM in the indicated melanoma cell lines was examined by western blotting. Vinculin was used as a protein loading control. (B) To evaluate cell viability, ATM knock-out canine melanoma cell lines (CMeC1, KMeC, CMM10, and CMM12) were treated with reovirus (MOI 100) for 48 hr before the trypan blue exclusion test. Viable cells and dead cells were counted, and the percentage of viable cells were calculated. Data are expressed as the mean + SD derived from three independent experiments. Tukey-Kramer test, *p < 0.05, **p < 0.01, ns: not significant. Figure 3. The combination of KU60019 and reovirus induces apoptotic cell death. (A) CMeC1, KMeC, and LMeC were treated with reovirus (MOI 100) and KU60019 (2.5 and 10 µM), and CMGD2 was treated with reovirus (MOI 10) and KU60019 (2.5 and 10 µM). After a 48-hr incubation, cell viability was quantified by the trypan blue exclusion test. Data are expressed as the mean ± SD from at least three independent experiments. p values were calculated for the comparison to reovirus alone. Tukey-Kramer test, *p < 0.05, **p < 0.01. (B) Cleaved caspase-3 levels in CMeC1 treated with reovirus (MOI 100) and KU60019 (2.5 µM) for the indicated time are shown in the upper panel. The relative level of cleaved caspase-3 normalized to actin was quantified from three independent experiments. Data are expressed as the mean + SD. Tukey-Kramer test, *p < 0.05, **p < 0.01. (C) The sub-G1 population was analyzed in CMeC1 treated with reovirus (MOI 100) and KU60019 (2.5 µM) for 24 and 48 hr. The cell numbers and stages were quantified using flow cytometry. The percentage of sub-G1 was calculated in total cells. Data are expressed as the mean + SD derived from three independent experiments. Tukey-Kramer test , *p < 0.05, **p < 0.01. Histogram of PI staining of each sample at 48-hr is shown for representative data from one of three independent experiments. (D, E) The effects of co-treatment for 48 hr with Z-VAD-FMK (50 µM) along with the combination of reovirus (MOI 100) and KU60019 (2.5 µM or indicated various concentrations) were quantified using the trypan blue exclusion test (D) or the CCK-8 cell viability assay (E). Data are expressed as the mean ± SD from at least three independent experiments. p values were calculated for the comparison to the vehicle sample. Tukey-Kramer test , **p < 0.01. Figure 4. The combination of KU60019 and reovirus induces cell cycle arrest at G2/M and increases sub-G1 populations in canine melanoma cells.
30
724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777
(A) CMeC1 was treated with reovirus (MOI 100) and KU60019 (2.5 µM). After 24-hr or 48-hr incubations, the cell cycle of CMeC1 cells was assessed with propidium iodide (PI) staining, as shown in Fig. 3C. Bar graphs indicate the percentages of each cell cycles (G1, S, and G2/M) at 24-hr and 48-hr in the total cells except sub-G1 populations. p values shown were calculated for the comparison between "Reo" and "KU+Reo".Tukey-Kramer test, *p < 0.05, **p < 0.01. In addition, p values shown were calculated for the comparison between "Mock" and "Reo".Tukey-Kramer test, †p < 0.01. (B) cdc2 and p21 protein expression levels in CMeC1 treated as described in (A) are shown in the upper panel. The relative level of each protein normalized to actin was quantified from three independent experiments. Mean + SD are shown. Tukey-Kramer test, *p < 0.05, **p < 0.01. The membrane in Figure 3B was reprobed and used for this panel. Figure 5. Reovirus replication is enhanced by KU60019 treatment. (A) Supernatants of reovirus (MOI 100 for CMeC1, KMeC, and LMeC, or MOI 10 for CMGD2) -infected cell lines treated with KU60019 (2.5 µM) were harvested after a 48-hr incubation, and virus titers were determined by the TCID50 assay. The fold increase of reovirus titer represents values calculated from the titer of progeny virus divided by the titer of input virus. The mean + SD was calculated from three independent experiments. p values were calculated by Tukey-Kramer test, *p < 0.05, **p < 0.01, ns: not significant. (B) Effects of ribavirin (200 µM) in CMeC1 treated with combination of reovirus (MOI 100) and KU60019 (2.5 µM) for 24 hr were assessed by western blotting using anti-reovirus antibody and anti-cleaved caspase-3 antibody. Actin was used as a protein loading control. (C, D) CMeC1 cells were pre-treated with reovirus virions (MOI 100) or ISVPs (MOI 10), and washed-out unattached virus followed by treatment with KU60019 (2.5 µM) for the indicated time points. Reovirus structural proteins were detected by western blotting using anti-reovirus polyclonal antibody. Actin was used as a protein loading control. The relative level of reovirus protein normalized to actin was quantified from at least three independent experiments. Mean + SD are shown. Tukey-Kramer test, *p < 0.05, **p < 0.01. Figure 6. Reovirus infection induces the phosphorylation of ATM in cancer cells. Phospho-ATM and total ATM expression levels were assessed by western blotting of lysates from CMeC1 cells treated with reovirus (MOI 100) and KU60019 (2.5 µM) for the indicated times. Vinculin was used as a protein loading control. The graph shows the relative level of each protein (normalized to vinculin) quantified from four independent experiments. The fold change of phospho-ATM divided by total ATM was normalized to the value of Mock at 0 hr. Mean + SD are shown. Tukey-Kramer test, *p < 0.05. Figure 7. The combination of reovirus and KU60019 promotes the endosomal acidification and activation of cathepsin B. (A, B) The acidification of endosomes/lysosomes was determined by LysoTracker-Red staining. CMeC1 cells were treated with reovirus (MOI 100) and KU60019 (2.5 µM) for 24 hr before staining. (A) Histogram overlay of LysoTracker-Red staining of each sample is shown for representative data from one of four independent experiments. Relative median fluorescence intensity (MFI) values relative to mock treatment are indicated. Mean + SD are shown from four independent experiments. Tukey-Kramer test, *p < 0.05, **p < 0.01. (B) Representative images of samples from each treatment
31
778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799
groups are visualized using confocal microscopy at a 100x magnification. Nuclei were stained with DAPI. Scale bar: 10 µm. (C) The enzyme activity of cathepsin B in CMeC1 cells treated as described above was measured and normalized to the value obtained with mock treatment. Mean + SD are shown from five independent experiments. Tukey-Kramer test, *p < 0.05.
Figure 8. Model of anti-tumor effects of reovirus combined with KU60019. In general, the phosphorylation of ATM protein is induced when DNA double-strand breaks are caused by UV or radiation. We newly found that reovirus infection activated the ATM protein unaccompanied with DNA double-strand breaks, at the late time points of reovirus infection (after 12 hr)(A) (Figure 6). This indicates that reovirus may cause an atypical ATM signaling pathways, which is abrogated by KU60019 (B). The combination treatment of reovirus and KU60019 leads to enhancement the virus replication (C), followed by cell cycle arrest at G2/ M and caspase-dependent apoptosis, potentiating anti-tumor effects. However, the mechanism between upregulation of lysosomal activities and reovirus cytotoxicity is still unknown.
32
Figure 7 A
2.5 Mock KU Reo KU + Reo Sample Name
250
**
Count
Tube - 9_Data Source - 1.fcs DsRed-Monomer-A subset 15656 Tube - 8_Data Source - 1.fcs Lymphocytes
17177
Tube - 7_Data Source - 1.fcs Lymphocytes
17129
Tube - 6_Data Source - 1.fcs Lymphocytes
17439
Count
150
100
50
0 10
3
10
4
10
5
LysoTracker-Red FL3-A :: DsRed-Monomer-A
Relative MFI ( /Mock )
200
Count
Subset Name
*
2
1.5 1 0.5 0
Mock
KU60019
KU
Reo
KU + Reo
C 160
Reovirus
KU + Reo
Cathepsin B activity (% of Mock)
B
Mock
*
140 120 100 80 60 40 20 0
Mock
KU
Reo
KU + Reo
Figure 8
DNA damage
Reovirus
X
X X
(A)
ATM (B)
p-ATM
(C)
Virus replication Cell cycle arrest at G2/M Caspase-dependent apoptosis
KU60019
?
Up-regulation of lysosomal activity
Figure 1 KMeC
100
100
100
60 **
40
** **
20
0
1.25 2.5 5 KU60019 (μM)
**
60 **
40 20 0 1.25 2.5 5 KU60019 (μM)
10
**
20
0
% of cell proliferation
80
0
**
40
1.25 2.5 5 KU60019 (μM)
80 **
60 40 20 0
10
0
CMM10
120
100
**
60
0
10
CMGD2
120
80
1.25 2.5 5 KU60019 (μM)
10
CMM12 120
100 80 60
**
40 20
% of cell proliferation
80
% of cell proliferation
120
0
% of cell proliferation
LMeC
120 % of cell proliferation
% of cell proliferation
CMeC1 120
100 80 60
KU60019 *
40
**
20
KU60019 + Reov irus **
**
0
0 0
1.25 2.5 5 KU60019 (μM)
10
0
1.25 2.5 5 KU60019 (μM)
10
Figure 2 A KMeC
CMeC1
CMM10
WT 1C10 1D11 WT 2C10 3G9 WT 1D9 3D7
CMM12 WT 3C12 3E9
ATM Vinculin
B
KMeC
CMeC1 120
** **
80 60 40 20
ns
80 60 40 20
0
0 WT
1C10
1D11
WT
CMM10 120
2C10
**
** ns
% of cell viability
100
80 60 40
3G9
CMM12
120
**
100 % of cell viability
**
100 % of cell viability
100 % of cell viability
120
80 Mock Reovirus
60 40 20
20
0
0 WT
1D9
3D7
WT
3C12
3E9
15M
15M
15M
800
200
200
0
0 5.0M
10M
15M
0
*
0
0
5.0M
*
A02.fcs
10M 3 10
2
10
15M 4
10
60
10
6
10
400
100
200
0 15M
0
0 10
5.0M
2 10M
FSC-A :: FSC-A FSC-A :: FSC-A
C 70A02.fcs
A01.fcs Live 8598
Ungated 10000
A01.fcs Live 8598
A03.fcs Ungated 15096
10
0 10
5
6 10 5.0M
2 10M 10
10
3
Mock
15M10 4
0 0 10
0 5.0M 5.0M 6 10
5
KU60 019 Reo viru s KU + Reo B01.fcs A04.fcs Ungated Ungated 7410 31343
A02.fcs Live 8504
A02.fcs Live 8504
300
10
0
Count
Count
SSC-A :: SSC-A
B02.fcs Ungated 7023
10M 3 10
2
10
15M 4
10
5
10
6
10
100
0
0
Count
SSC-A :: SSC-A
100
15M
200
400
Live 72.3
600
10M
0
5
B02.fcs Live 5077
0
40
10
5
2 10 10M
5.0M 6 10
6
10
2 10M
10
FSC-A :: FSC-A #1 #2
FL2-A subset 3.13
Live 30.0
3 15M
10
0 4
0
10
5
FL2-A :: FL2-A
DMSO 300
B02.fcs Ungated 7023
Live 72.3
100
10M
FL2-A subset 34.7
80
3
4 10 15M
5
10
6
Actin
FL2-A subset 20.3
0
5 100
10
0 6 5.0M 10
FL2-A :: FL2-A
#3
10M
2
10
#5
B03.fcs Ungated 20172
10
15M 4
3
10
FSC-A :: FSC-A
#4
10
5
10
#6 FL2-A ::#7 FL2-A
2
10
3
6
#8
10
4
10
5
10
6
FL2-A :: FL2-A
#9 B03.fcs Live 6056
B02.fcs Live 5077
Reovirus - 48 hr
KU + Reo - 48 hr 400
15M
Live 9.37
200
FL2-A subset 43.9
Count
Live 30.0
300
FL2-A subset 20.3
10M
Count
SSC-A :: SSC-A
FL2-A subset 3.13
400
200
5.0M
200
100
10M 2 10
10
2
10
FSC-A :: FSC-A
3
2 3 10 10M 10
5 6 6 10 10 5.0M 10
FL2-A FL2-A :: FL2-A :: FL2-A
B01.fcsA04.fcs Live Live 5096 7238
0
0
0 4 3 15M 4 5 0 10 10 10 10
10M2 3 10 10
4 10 15M
100
Live 30.0
FL2-A subset 10M 3.13
200
10
5
10
10
6
2
10M
10
FSC-A :: FSC-A
FL2-A :: FL2-A
B03.fcs Ungated 20172
A04.fcs Live 7238
0
10M
2
10
15M 4 10
3
10
5
10
6
10
2
10
400
3
** FL2-A subset 20.3
200
Live 9.37
3
15M 4 10
0 05 10
6 10 5.0M
FL2-A :: FL2-A
B02.fcs Live 5077
DMSO
400 15M
300
2 10M
10
3
4 10 15M
FL2-A :: FL2-A
FSC-A :: FSC-A
B03.fcs Live 6056
B04.fcs Ungated 88008
KU60019
10
Reovirus 400
KU + Reo
10
5
4
10
CMeC1
400
120
100
100
300
** FL2-A subset 43.9 **
80
**
200
**
**
60
100
40
10
6
0 10
2
20 0
10
3
10
4
10
5
10
6
FL2-A :: FL2-A
B04.fcs Live 8245
0
5
10
6
B04.fcs Live 8245
120
300
10
FL2-A :: FL2-A
B03.fcs Live 6056
E
Ungated 88008
100
5.0M
0 5.0M
0
10
FSC-A :: FSC-A FL2-A :: FL2-A
0
40
0
5.0M
6
15M
400
0
15M 4 10
10
DNA content B04.fcs
B02.fcs Live 5077
KMeC
50
FL2-A :: FL2-A
FSC-A :: FSC-A
B03.fcs Ungated 20172
0
10
60
KU60019 Reovirus KU + Reo 15M
10
100
KU60019 - 48 hr
20
A03.fcs Live 5746
A04.fcs Ungated 31343
6 0 10 5.0M
4
200
200
B01.fcs Live 5096
**
5.0M
FSC-A :: FSC-A
20 0
10
10
**
0
10 5.0M
5.0M
600
120
100
4 0
3
**
400
5.0M
200
Count
Live 72.3
FL2-A FL2-A subsetsubset 2.39 34.710M
Z-VAD
**
5.0M
0
10
300
FL2-A subset 2.39
15M
FSC-A :: FSC-A
B02.fcs Ungated 7023
60 10
5.0M 6 10
5
15M 300
Cleaved Caspase-3
2
FL2-A :: FL2-A
B01.fcs Live 5096
600
B02.fcs Ungated 7023
SSC-A :: SSC-A
Count
Count
200
FL2-A :: FL2-A
A03.fcs Live 5746
FL2-A subset 14.5 10M Live 23.1
80
10
Vehicle
300
200
0
15M 15M 4 10
15M
FL2-A :: FL2-A
A03.fcs Ungated 15096
0
10M 2 10M 3 10 10
FSC-A :: FSC-A FSC-A :: FSC-A
FL2-A :: FL2-A
FSC-A :: FSC-A
Count
Count
SSC-A :: SSC-A
FSC-A :: FSC-A
3
200
400
5.0M
0 0
0
0
100
15M
10
Live Live 68.8 23.1
Count
200
FL2-A subset10M 14.5
100
FL2-A subset 10M 1.95
10M 2
10
200
A03.fcs Ungated 15096
% of cell viability
0
24
600
5.0M 5.0M
20
5.0M
5.0M
48
KU60019 (2.5 μM) Reovirus (MOI 100) 48 Time (hr)
FSC-A :: FSC-A FL2-A :: FL2-A
600
SSC-A :: SSC-A
Live 400 38.1
**
0
24
+ +
200
300
30
D
200
0 4 10
A03.fcs Live 5746
15M
10M FL2-A subset 10M 1.95
15M
400
10M
3 15M
+ +
0
5.0M
FSC-A :: FSC-A
Mock - 48 hr
15M 15M
600
120
Live 38.1
10
FSC-A :: FSC-A
B01.fcs Ungated 7410
A02.fcs Live 8504
CMeC1
600
10M2 10
300
40
0 FL2-A :: FL2-A
800
10
0
15M
Count
A02.fcs Ungated 10000
10
6
10M
− +
0
% of cell proliferation
FSC-A :: FSC-A
5.0M
5
300
15M 4 10
3
10
5.0M 6 10
5
Count
10
10
4
48
− +
400
SSC-A :: SSC-A
2 10M
15M
3
Count
10
10
0
0
CMGD2
10M
2
SSC-A :: SSC-A
5.0M
6 0 10 5.0M
FSC-A :: FSC-A
24 hr
10
0
0
KMeC
5
10
FL2-A :: FL2-A
5.0M
200
0 10
FL2-A :: FL2-A
800
Count
SSC-A :: SSC-A
Count 400
4
% of cell viability
% of sub-G1 cells
50
FL2-A subset 1.40 10M
Live 85.0
10 15M
0
+ −
400
15M
600
LMeC
3
48 hr
60 800
10
SSC-A :: SSC-A SSC-A :: SSC-A
A01.fcs Ungated 10000
0
0
CMeC1
0
Count
10M
4
5.0M
200
5.0M
10
1.4 1.2 FL2-A subset 1 14.5 0.8 0.6 0.4 0.2 0 FL2-A :: FL2-A
Live 68.8
200
5.0M
5.0M
5.0M
0
3
200
24
15M
10M
FL2-A subset 1.95
Live 38.1
Count
Live 85.0
400
20
SSC-A :: SSC-A
Count
10M
600
48
B01.fcs Ungated 7410
300
Count
FL2-A subset 10M 1.40
600
10
*
15M
**
40
2
A02.fcs Live 8504
**
800
800
24
+ −
5.0M
0
FL2-A :: FL2-A
** 15M
15M
5
A01.fcs Live 8598
**Ungated 10000
SSC-A :: SSC-A
% of cell viability
A01.fcs Ungated 10000
SSC-A :: SSC-A
Live 86.0
10
FSC-A :: FSC-A FL2-A :: FL2-A
FSC-A :: FSC-A
80
0
− −
0
SSC-A :: SSC-A
0
− −
FL2-A subset 3.13
400
% of cell proliferation
100
− −
B 5.0M
Reovirus + KU 10 μM
FL2-A subset 2.39
Live 72.3
400
Count
Reovirus + KU 2.5 μM
400
Live 68.8
10M
SSC-A :: SSC-A
Reovirus
400 5.0M
5.0M
Count
Live 85.0
Relative expression levels of cleaved casp-3 (/Actin)
Mock
10M
SSC-A :: SSC-A
Live 86.0
FL2-A subset 10M 1.95
Count
A
10M
600 600
FL2-A subset 1.40
600
Count
SSC-A :: SSC-A
Figure 3
SSC-A :: SSC-A
800
Count
15M
10
**
** **
80
** **
60
**
**
**
Vehicle+KU Vehicle+KU+Reo Z-VAD+KU
40
Z-VAD+KU+Reo
20 0
1.25 2.5 5 KU60019 (μM)
KMeC
0
1.25 2.5 5 10 KU60019 (μM)
Figure 4 A
24 hr
120% 120
*
†
100 100%
Percentages of cells
100% 100 80% 80
% of cells
% of cells
S
40% 40
20 20%
**
20% 20
0%0
0%0 M
k oc KU
0 60
19 R
s ru vi o e
KU
+
R
eo
M
− −
− −
− −
+ −
+ −
− +
− +
+ +
0
24
48
24
48
24
48
24
k oc KU
+ +
01 60
9
s eo ru R vi + o e R KU
KU60019 (2.5 μM)
Reovirus (MOI 100) 48 Time (hr)
cdc2/CDK1 p21
Relative expression levels of p21 (/Actin)
Relative expression levels of cdc2/CDK1 (/Actin)
Actin 2.5 *
2 1.5 1 0.5
1.5
#1
#2
#3
#4
#5
#6
#7
#8
#9
#1
#2
#3
#4
#5
#6
#7
#8
#9
1
0.5
0
G2/M
†
60 60%
40 40%
0
**
80 80%
60 60%
B
48 hr
120 120%
G1
Figure 5
**
35
A
*
**
ns
B
ns
ns
Fold increase of virus titer (/Input)
30
–
–
–
–
+
+
+
– −
– +
+ −
+ +
– −
– +
+ −
25 20
Reovirus Protein
Input Reovirus + DMSO
15
Reovirus + KU60019 2.5μM 10
Cleaved Caspase-3
5
Actin
0
CMeC1
LMeC
KMeC
CMGD2
C
D − −
− +
− +
− +
+ +
+ +
+ +
0
6
12
24
6
12
24
− KU60019 (2.5 μM) − Reovirus (MOI 100) 24 Time (hr)
− −
− +
− +
− +
+ +
+ +
+ +
0
6
12
24
6
12
24
Actin
Actin
**
p > 0.05
2 Relative expression levels of reovirus protein (/Actin)
2.5
* 2
1.5
1
0.5
p > 0.05
1.5
1
0.5
0
0 o,
r 6h
,1
r 2h
,2
r 4h
, +r
r 6h
r,
1
r 2h
r,
2
r 4h
− KU60019 (2.5 μM) − ISVPs (MOI 10) 24 Time (hr)
Reovirus Protein
Reovirus Protein
Relative expression levels of reovirus protein (/Actin)
+ Ribavirin (200 μM) + Reovirus (MOI 100) + KU60019 (2.5 μM)
o,
6h
r o,
12
hr o,
24
hr
, +r
6h
r r,
12
hr r,
24
hr
Figure 6 – – 0
– – 12
– – 6
– – 24
– + 6
– + 12
– + 24
+ + 6
+ + 12
+ KU60019 (2.5 μM) + Reovirus (MOI 100) 24 Time (hr)
p-ATM (Ser1981)
ATM
Vinculin 12 Fold change of p-ATM/ATM (Normalized to Mock at 0 hr)
*
10 8
*
6 4 2 0
#1
#2
#3
#4
#5
#6
#7
#8
#9
#10