Pharmacological inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models
Authors:
Frank T. Zenke , Astrid Zimmermann , Christian Sirrenberg , Heike Dahmen , Vladimir Kirkin , Ulrich Pehl , Thomas Grombacher , Claudia Wilm , Thomas
6
7
Fuchss , Christiane Amendt , Lyubomir T. Vassilev , Andree Blaukat Affiliations:
1
8
1
Merck KGaA, Biopharma R&D, Translational Innovation Platform Oncology,
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
Frankfurter Str. 250, 64293 Darmstadt, Germany; The Institute of Cancer Research,
Cancer Research UK Cancer Therapeutics Unit, 15 Cotswold Road, Sutton, London,
SM2 5NG, UK; Merck KGaA, Biopharma R&D, Discovery Development
Technologies, Frankfurter Str. 250, 64293 Darmstadt, Germany; Merck KGaA,
Biopharma R&D, Translational Medicine, Frankfurter Str. 250, 64293 Darmstadt,
Germany; EMD Serono Research and Development Institute Inc., Biopharma R&D,
Translational Innovation Platform Oncology, 45A Middlesex Turnpike, Billerica, MA 01821-3936, USA; a business of Merck KGaA, Darmstadt, Germany
Email addresses:
[email protected]
1
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Running title: Preclinical pharmacology of M3814 in cancer models
Suggested keywords (max 5): Non-homologous end joining; DNA-damage repair; DNA-dependent protein kinase; ionizing radiation; M3814
Additional information:
Financial support: This work was funded by Merck KGaA, Darmstadt, Germany
Corresponding author: Frank T. Zenke; Merck KGaA, Frankfurter Str. 250, 64293
Darmstadt, Germany; email: [email protected]; Tel: +49 (0)6151 72- 4730; Fax: +49 (0)6151 72-7398.
Conflict of interest disclosure statement:
All authors are employees of Merck KGaA or EMD Serono, except for Vladimir Kirkin, who is a former employee of Merck KGaA.
Word count: 4605/5000 (includes Introduction, Results, Discussion, Acknowledgements, Figure legends)
Figures/tables: 6/6
2
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Abstract (214/250 words)
Physical and chemical DNA-damaging agents are used widely in the treatment of
cancer. Double-strand break (DSB) lesions in DNA are the most deleterious form of
damage and, if left unrepaired, can effectively kill cancer cells. DNA-dependent
protein kinase (DNA-PK) is a critical component of non-homologous end joining
(NHEJ), one of the two major pathways for DSB repair. Whilst DNA-PK has been
considered an attractive target for cancer therapy, the development of
pharmacological DNA-PK inhibitors for clinical use has been lagging. Here, we report
the discovery and characterization of a potent, selective, and orally bioavailable DNA-
PK inhibitor, M3814, and provide in vivo proof of principle for DNA-PK inhibition as a
novel approach to combination radiotherapy. M3814 potently inhibits DNA-PK
catalytic activity and sensitizes multiple cancer cell lines to ionizing radiation (IR) and
DSB-inducing agents. Inhibition of DNA-PK autophosphorylation in cancer cells or
xenograft tumors led to an increased number of persistent DSBs. Oral administration
of M3814 to two xenograft models of human cancer, using a clinically established 6-
week fractionated radiation schedule, strongly potentiated the antitumor activity of IR
and led to complete tumor regression at non-toxic doses. Our results strongly support
DNA-PK inhibition as a novel approach for the combination radiotherapy of cancer.
M3814 is currently under investigation in combination with radiotherapy in clinical trials.
3
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Introduction
To ensure the accurate maintenance and transfer of genetic information to progeny,
mammalian cells have evolved sophisticated mechanisms to sense DNA damage,
coordinate its repair, and prevent potential tumorigenic effects; this is collectively
known as the DNA damage response (DDR). Defects in the DDR contribute to
genomic instability and represent one of the key hallmarks of cancer (1). DNA can be
damaged by multiple endogenous and exogenous factors. Many established
therapeutic modalities, such as radio- and chemotherapy that attack cancer cell DNA
are in clinical use but provide limited benefit to cancer patients. This is due, at least in part, to the competence of tumor cells to deal with DNA damage (2).
Diverse types of lesions can be generated in DNA, ranging from base modifications
to strand breaks, leading to large deletions or genomic rearrangements. Of those,
double-strand breaks (DSBs) are considered the most harmful and can have lethal
consequences for the cells and organism if left unrepaired (3). DSB repair is
accomplished through two major pathways, homologous recombination-guided repair
(HR) and non-homologous end joining (NHEJ) (3,4). HR requires an intact DNA
strand as a template for break repair and is restricted to the S and G2 phases of the
cell cycle. Therefore, HR is considered less error prone than NHEJ. Conversely,
NHEJ repairs DSBs in the absence of a template and leads to alterations in the
repaired DNA. However, NHEJ is functional in all phases of the cell cycle and is
believed to participate in the repair of over 80% of DSBs induced by ionizing radiation (IR) in cancer cells (5).
DNA-dependent protein kinase (DNA-PK) is a serine/threonine kinase and a key
driver of NHEJ repair, working in co-ordination with five additional factors, Ku70,
Ku80, XRCC4, ligase IV, and Artemis (6). A heterodimer consisting of Ku70 and 4
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
91
92
93
94
95
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Ku80 binds specifically to DSBs, recruits and activates the catalytic subunit DNA-
PKc, which in turn recruits the XRCC4/ligase IV heterodimer responsible for resealing
the break. Trimming of the DSB ends may require Artemis and other DNA
polymerases specialized in repair-mediated DNA polymerization. The activation of
DNA-PK through autophosphorylation is essential for proper execution of the repair process (7,8).
DNA-PK-knockout mice are viable, suggesting that pharmacological inhibition will not
affect essential functions in mammalian organisms and may be tolerated for the
duration of standard cancer therapy regimens (9,10). Several lines of experimental
evidence suggest that inhibition of DNA-PK activity can effectively sensitize cancer
cells to exogenous DSB DNA damage, such as IR and certain types of DSB-inducing
chemotherapies (11-17). These conclusions are derived from experiments using
molecular biology approaches (RNAi) to suppress DNA-PK expression or early
chemical inhibitors as laboratory tools in cultured cancer cells. However, most of
those tool compounds lacked the specificity and pharmacological properties needed
to establish a proof of principle for selective DNA-PK inhibition as a therapeutic approach in relevant in vivo models.
Here, we describe M3814, a novel potent and selective pharmacological DNA-PK
inhibitor. We show that M3814 effectively suppresses DSB repair in cancer cells in a
DNA-PK-dependent manner and strongly potentiates the antitumor effect of IR and
DSB-inducing chemotherapy in vitro and in vivo. Clinically relevant 6-week studies
using fractionated radiation and M3814 in two human xenograft models
demonstrated complete and durable tumor regression, providing a strong rationale for clinical testing.
5
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
116
117
118
119
120
121
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Methods
Cell lines and reagents
M3814 (MSC2490484A) was synthesized in the department of Medicinal Chemistry
at Merck KGaA, Darmstadt (18). Cell lines were obtained commercially (ATCC,
Virginia, USA; ECACC, Salisbury, UK; JCRB, Ibaraki, Japan; RIKEN, Tsukuba,
Japan) and cultured in medium recommended or previously tested for these cells
122
(A549, BxPC-3, FaDu, HT-29: DMEM/10% FCS/10% CO
2
; Calu-6: DMEM/10%
123
FBS/NEAA/10% CO
2
; Capan-1: DMEM/15% FBS/10% CO
2
; DU-145: MEM
124
alpha/10% FBS/2 mM glutamine/5% CO
2;
EBC-1; MEM Eagle/10% FBS/2 mM
125
glutamine/5% CO
2
; HCT-116: MEM alpha/10% FBS/10% CO
2;
KP-4: DMEM/Hams
126
F12 1:1/10% FCS/2 mM glutamine/5% CO
2
; MiaPaCa-2: DMEM/10% FBS/2.5%
127
128
129
130
131
132
133
134
135
136
137
138
139
140
HS/10% CO2; MO59K, MO59K: DMEM/Nut Mix F12/10% FBS/2.5 mM glutamine/NEAA/5% CO2; A375, NCI-H460: RPMI 1640/2 mM glutamine/1 mM sodium pyruvate/5% CO2). Short tandem repeat analysis was performed to confirm
cell line identity. Mycoplasma infection was excluded using a PCR-based method.
The rabbit monoclonal antibody EM09912 was generated at Epitomics Inc.
(Burlingame, California). In brief, rabbits were immunized with KLH-coupled
phosphopeptide (YSYSpSQDPRPA). Supernatants from hybridoma cells were tested
for differential activity against the phospho- versus nonphosphopeptide using
enzyme-linked immunosorbent assays (ELISAs) and subsequently verified by
western blotting. The antibody was purified from hybridoma supernatant using a protein A affinity chromatography.
Protein kinase assays
DNA-PK enzymatic assays were performed at concentrations of ATP near the Km
(10 µM) or at 1 mM for time-resolved fluorescence energy transfer (TR-FRET). DNA- 6
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
PK purified from HeLa nuclear extracts was pre-incubated with M3814 at different
concentrations (4.0E-15–3.0E-5 M) or vehicle for 15 minutes at 22°C in assay buffer.
The reaction was started by addition of biotinylated STK-substrate (61ST1BLC,
Cisbio), Mg-ATP, calf thymus DNA, and staurosporine, followed by incubation for 60–
80 minutes at 22°C. The reaction was stopped with EDTA, and phospho-STK was
detected with an anti-phospho-STK antibody (61PSTKLB, Cisbio) labeled with
Europium as the donor, and streptavidin-labeled with XL665 (610SAXAC, Cisbio) as
the FRET acceptor. Following incubation for 60 minutes, plates were analyzed on a
Rubystar (BMG Labtech) microplate reader (excitation wavelength: 337 nm; emission wavelengths: 665 and 615 nm).
Ataxia telangiectasia-mutated (ATM) and ATM and rad3-related (ATR)/ATR-
interacting protein (ATRIP) assays were performed using TR-FRET. Human
recombinant ATM (14-933, Eurofins) or ATR/ATRIP (14-953, Eurofins) were pre-
incubated in assay buffer for 15 minutes at 22°C with different concentrations of
M3814 (4.0E-15–3.0E-5 M) or vehicle. The assay was started by addition of purified
c-myc-tagged p53 (23-034, Eurofins) and ATP, and then incubated for approximately
30 minutes at 22°C. Reactions were subsequently stopped and antibodies were
added (anti-phospho-p53(Ser15)-Eu [61P08KAY, Cisbio]; anti-cmyc [61MYCDAB,
Cisbio]). After incubation for 2 hours, plates were analyzed in an EnVision
(PerkinElmer) microplate reader (excitation at 340 nm, emission: 665 and 615 nm).
161
Data were normalized to a DMSO control, and IC
50
values were determined by non-
162
163
164
165
linear regression analysis.
Protein and lipid kinase profiling was performed at Merck Millipore, Dundee, UK.
Recombinantly produced protein and lipid kinases were used in enzyme activity
assays. Protein kinase reactions were initiated with Mg-ATP and stopped after 40 7
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
minutes by addition of phosphoric acid. The transfer of radiolabeled -phosphate to
peptide substrates was quantified by scintillation counting of peptide substrates
immobilized on filter membranes. A non-radioactive assay (homogeneous time
resolved fluorescence [HTRF]) was used for lipid kinases. The “percent of effect”
activity was determined compared with vehicle-treated controls corrected for
background activity. M3814 was tested at 1 µM (some cases 10 µM) or serially diluted for IC50 determination.
Western blot analysis
Exponentially growing HCT-116 and FaDu cancer cell lines were seeded in 12-well
plates. The next day, the medium was removed, and cells were incubated with fresh
medium containing a serial dilution of M3814 (concentration range: 1.5E-09–2.5E-05
M) and 10 µM bleomycin (for detection of phosphorylated DNA-PK) or 3 mM
hydroxyurea (for detection of phosphorylated CHK1) for 6 hours. Cells were washed
and lysed using HGNT buffer. After sodium dodecyl sulfate-polyacrylamide gel
electrophoresis (SDS-PAGE), proteins were transferred to nitrocellulose membranes
and incubated with primary antibodies (1:2000 anti-total DNA-PK, ab70250, Abcam;
1:1000 anti-pSer2056 DNA-PK, ab18192, Abcam; 1:1000 anti-pSer345 CHK1,2348,
Cell Signaling; 1:200 anti-total CHK1sc8408, Santa Cruz) overnight at 4°C.
Membranes were incubated with horseradish peroxidase (HRP)-conjugated
185
secondary antibodies and developed using Lumi-Light
PLUS
(Roche). Luminescence
186
187
188
189
was recorded using a VersaDoc luminescent imager (Bio-Rad, Munich, Germany).
pDNA-PK and pCHK1 values were normalized to total protein, and the
bleomycin/hydroxyurea and DMSO controls were set to 0 and −100, respectively.
Concentration-response curves were fitted using a nonlinear regression method to
190
determine IC50
8
values. The experiment was repeated at least twice.
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Immunofluorescence studies
A375 cells were seeded on eight-well glass chamber slides pre-coated with
fibronectin. After 48 hours, the medium was removed and fresh medium without
serum was added. The next day, the medium was removed and fresh medium with
and without 1 µM M3814 was added 30 minutes prior to IR (2.4 Gy); the cells were
further incubated for 0.5, 2, 4, 6, 8, or 24 h. Cells were fixed with methanol and
stained with primary antibodies against phospho-histone H2A.X (1:8000 dilution,
γH2A.X Ser139; clone 20E3; Cell Signaling 9718) overnight at 4°C, and
subsequently with Alexa Fluor-conjugated secondary antibodies (1:2000 dilution,
anti-rabbit IgG; Invitrogen A-11008). Immunofluorescence was determined at 40X
magnification, γH2A.X foci were counted in 50–80 nuclei, and the numbers of cells
with more than 10 γH2A.X foci per nucleus were counted and expressed as a percentage of total nuclei.
Immunohistochemistry
FaDu xenograft tumors were fixed in 4% buffered formaldehyde and embedded in
paraffin. Sections (3 µm) were deparaffinized and heated to 96–100°C for epitope
retrieval. Sections were incubated with primary antibodies (10 µg/mL anti-murine
CD31, clone SZ31, DIA-310, Dianova; 1:50 anti-human γH2A.X, clone 20E3, 9718,
NEB), and subsequently with Alexa Fluor-conjugated secondary antibodies (1:250
anti-rat IgG AF488: A-21208, Invitrogen; 1:250 anti-rabbit IgG AF594: A-11037,
Invitrogen). Immunofluorescence was imaged at 40X magnification and the γH2A.X
area inside nuclei (AF594 positive)/area nucleus (DAPI positive) was quantified. Data
from vehicle- and drug-treated animals were compared by one-way ANOVA
(Kruskal–Wallis test) followed by Dunn’s Multiple Comparison Test (two-sided,
9
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
GraphPad Prism). Data are presented as the mean ± standard error of the mean (SEM).
Colony formation assay (CFA)
Cell survival was tested in response to IR at doses of 2.4 and 4.8 Gy using a Faxitron
RX-650 irradiation device (Tucson, Arizona). Exponentially growing cancer cells (for
cell lines see Table 2 and Fig. 1D) were seeded on six-well plates. Twenty-four hours
later, cells were treated with a serial dilution of M3814 (typical concentration range: 2
x 10 –5 x 10 M) for 1 hour prior to radiation. After an additional 24 hours in the
presence of M3814, the medium was substituted with fresh culture medium without
compound. Cells were incubated for several days to weeks until visible colonies
could be detected. Colonies were stained with neutral red or crystal violet and
quantified using a Gelcount Scanner (Oxford Optronix, UK, England). Inhibition under
each condition (single concentration and IR dose) was determined in singlicate in at
least three independent experiments. Raw data were normalized by setting the cell
number under DMSO treatment without IR to 100%. Concentration-response curves
230
were fitted using a nonlinear regression method to determine IC
50
values.
231
232
233
234
235
236
237
238
239
ELISA assays
Autophosphorylation of DNA-PK on serine 2056 was assessed by ELISA with a
pDNA-PK antibody (0.125 µg/mL, EM9912), a DNA-PK antibody (0.5 µg/mL,
WH0005591M2) for capture, and a biotinylated DNA-PK antibody (0.15 µg/mL,
Abcam, ab79444) for detection. The PathScan Phospho-Chk2 (Thr68) Kit (Cell
Signaling) was used according to the manufacturer’s instructions to determine CHK2
phosphorylation on threonine 68. Exponentially growing cancer cells were treated
with a serial dilution of M3814 (3E-10–3E-05 M) and 10 µM bleomycin for 6 hours.
Cell lysates prepared in HGNT buffer were incubated with a solid phase-bound 10
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
240
241
242
243
244
245
246
247
248
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
capture antibody and further processed with a detection antibody and reagents.
Phospho-DNA-PK and total DNA-PK chemiluminescence was quantified with a
Mithras LB940 reader (Berthold, Bad Wildbad, Germany). The absorbance signal for
phospho-CHK2 was measured at 450 nm using a Sunrise reader (Tecan, Männedorf,
Switzerland). Phospho-DNA-PK values were normalized to total DNA-PK values; the
bleomycin and DMSO controls were set to 0 and −100, respectively. For the
PathScan Phospho-Chk2 (Thr68) Kit (Cell Signaling), background-corrected values
were analyzed by setting the mean value of bleomycin-treated controls to 0% and
transforming the absorbance values measured for the compound-treated samples to
249
percentages. IC50
values were calculated by non-linear regression. Each experiment
250
251
252
253
254
255
256
was repeated at least three times.
Cancer cell line profiling in combination with IR or other drugs
Radiosensitization of 92 cancer cell lines and resting peripheral blood mononuclear
cells (PBMCs) by M3814 was performed at Oncolead (Karlsfeld, Germany). Cell
viability was determined with 3 Gy IR (Cobalt-60 source), M3814 (5 µM–5 nM), and a
combination of 3 Gy IR and M3814 (5 µM–5 nM). Treated cells were incubated for
120 hours, fixed, stained with sulforhodamine B, and quantified colorimetrically (18).
257
EC50
(half-maximal effective concentrations) values were calculated from the
258
concentration response data. Because 50% inhibition of viability was not observed in
259
a substantial proportion of cell lines, EC50
data were displayed and analyzed.
260
261
262
263
264
Drug combination profiling of M3814 with 72 antitumor agents was performed in 34
cancer cell lines at Oncolead (Karlsfeld, Germany). Cell viability was determined with
72 drugs in concentration response, M3814 at a fixed concentration of 0.3 µM, and a
combination of both. After 120-hour incubation, cells were fixed, stained with
sulforhodamine B, and quantified colorimetrically (19). Bliss independence was used 11
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
to calculate synergy (20). In brief, the Bliss independence method compares the observed effect size Emeasured of a drug combination with the calculated effect, with Ecalc assuming complete independence of the drug effects (Ecalc = EDrug1 + EDrug2 – EDrug1EDrug2). Calculated Bliss values represent the mean values from all
concentration combinations of a specific drug combination (synergy >0.1; 0.1≥ additivity ≥-0.1; antagonism <-0.1).
In vivo efficacy and pharmacokinetic (PK)/pharmacodynamic (PD) studies
In vivo efficacy data were generated in squamous cell head and neck FaDu (ATCC ,
HTB-43 ) and non-small cell lung cancer (ATCC , HTB-177 ) NCI-H460 human
xenograft models in mice. The study designs and animal usage were approved by
local animal welfare authorities (Regierungspräsidium Darmstadt, Hesse, Germany,
protocol registration numbers DA4/Anz.397 and DA4/Anz.398). Seven- to nine-week-
old female NMRI (nu/nu) mice were used (Charles River Laboratories, Sulzfeld,
Germany). Mice received subcutaneous injections in the right thigh with 2.5 million
FaDu or NCI-H460 cells. When tumor xenografts reached a mean volume of 50 –115
mm , mice (n=10 per treatment arm, randomized from 15 mice per arm to obtain a
similar mean and median within the treatment groups) received IR (2 Gy, X-RAD320
irradiation cabinet, Precision X-ray Inc.; settings: 10 mA, 250 kV, 58 s, 50 cm FSD
collimator, 2 mm A1 filter). Mice were irradiated in groups of 10 and were
anaesthetized during the irradiation process. IR was administered locally by
positioning the tumor-bearing part of the leg in the beam path while shielding other
body parts of the mice with lead. M3814 was formulated in vehicle (0.5% Methocel,
0.25% Tween20, 300 mM Sodium Citrate buffer pH 2.5) and administered orally at
different doses, or in combination with IR for 5 days/week for 1 or 6 weeks. Mice
were irradiated 10 minutes after oral administration of M3814. Tumor length (L) and 12
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
290
291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
width (W) were measured with calipers and tumor volumes were calculated using L×W^2/2.
Pharmacodynamic (PD) data were generated in FaDu human xenograft models in
mice. M3814 was administered orally once (5, 25, or 100 mg/kg) in combination with
10 Gy IR. Animals were sacrificed at different time points (0.16, 1.5, 3, 8, or 24 hours)
and tumors were removed, frozen in liquid nitrogen, lysed with HGNT buffer, and
homogenized using a Precellys-24 homogenizer. Cell lysates were transferred to
capture antibody-coated (mouse anti-DNA-PK 1B9, Abnova H00005591-M02, 2
µg/mL for coating) microtiter plates (Mesoscale, L15XB-6) and detected with either
total DNA-PK (mouse anti-DNA PK biotin 3H6; Cell Signaling, 12311BF, 0.1 µg/mL
followed by a secondary streptavidin Sulfo-TAG antibody, 0.2 µg/mL; Mesoscale,
R32AD-1) or phospho-DNA-PK antibodies (rabbit anti-phospho-DNA PK MKV-2;
Epitomics/Abcam, MKV-2-99-12 used at 0.01 µg/mL followed by secondary anti-
rabbit sulfo-TAG antibody; Mesoscale 0.1 µg/mL, R32AB-1). Plates were analyzed using an MSD Sector imager 6000 (Model 1200).
Whole blood from healthy donors (N=6) was treated with M3814 (concentration
range: 1.0E-09–3.E-05 M) and bleomycin (100 µM) for 4 hours at 37°C. PBMCs were
isolated by density gradient separation with Leucosep filled tubes (VWR GREI
227288) and lysed using HGNT buffer. Cell lysates were transferred to capture
antibody-coated (2 and 4 μg/mL mouse anti- DNA PK; Sigma Aldrich
WH0005591M2) microtiter plates (Mesoscale; L15XB-6) and detected with either
total DNA-PK (1 μg/mL mouse anti-DNA PK; Abcam ab79444; followed by a 1 μg/mL
secondary streptavidin Sulfo-TAG antibody; Mesoscale; R32AD-1) or phospho-DNA-
PK (0.1 μg/mL rabbit anti-phospho-DNA PK MKV-2; Epitomics/Abcam; MKV-2-99-12
13
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
314
315
316
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
followed by a 1 μg/mL secondary anti-rabbit sulfo-TAG antibody; Mesoscale; R32AB- 1) antibodies. Plates were read using an MSD Sector imager 6000 (Model 1200).
14
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
317
318
319
320
321
322
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Results
M3814 is a potent and selective inhibitor of DNA-PK activity
M3814 (MSC2490484A) is the product of a drug discovery screening and
optimization program performed at Merck KGaA, Darmstadt, Germany. The structure
of M3814 is shown in Fig. 1A and its synthesis described (18). Key pharmacological
properties of M3814 are summarized in Table 1. M3814 inhibits DNA-PK activity with
323
0.6 nM IC50
) at an ATP concentration close to Km (10 μM). At a high ATP
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
concentration (1 mM), the potency was reduced more than 30-fold, suggesting that M3814 competes with ATP for binding to DNA-PK.
M3814 exhibited a high degree of selectivity when tested using a broad panel of
serine/threonine, tyrosine, and lipid kinases (Table 1). Only eight of 284
recombinantly expressed protein/lipid kinases, including mutant kinases, were
inhibited by at least 50% at 1 μM M3814. All represented wild-type isoforms or
mutant versions of lipid kinases of the PI3K family (Supplementary Table S1). PI3K
kinases, ATM, ATR, mammalian target of rapamycin (mTOR), and DNA-PK, are
members of the PI3K-related kinase family, which is characterized by high similarity
in the kinase domain (21). Despite their high similarity, PI3K lipid kinase isoforms
were affected with strongly reduced potency (>100-fold split). Other family members, ATR, ATM, and mTOR, were even less sensitive to M3814 (Table 1).
M3814 selectively inhibits DNA-PK activity and DSB repair in human cancer cell lines
M3814 inhibited DNA-PK autophosphorylation on Ser2056, a marker for DNA-PK
activity in bleomycin-treated cancer cells, determined by western blot (Fig. 1B) or
ELISA (Table 2). In bleomycin-treated HCT-116 cells, M3814 did not inhibit
phosphorylation of the ATM kinase substrate CHK2 (Table 1). In contrast, a 15
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
342
343
344
345
346
347
348
349
350
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
moderate and concentration-dependent increase in CHK2 phosphorylation was
observed, suggesting that DNA-PK blockade may trigger a compensatory
upregulation of DSB repair (22). Hydroxyurea-induced CHK1 phosphorylation is
induced by activation of ATR. M3814 did not block CHK1 phosphorylation,
suggesting that ATR is not inhibited in cells. Phosphorylation of AKT on serine 473 in
PC3 prostate cancer cells is driven by inactivation of phosphatase and tensin
homolog (PTEN) and generation of phosphatidylinositol-3,4,5-triphosphate (PIP3) by
PI3K activity (23). M3814 only moderately inhibited the phosphorylation of AKT, which is consistent with its weak inhibitory activity on the PI3K isoforms.
The reduced level of DNA-PK autophosphorylation by M3814 suggests that DNA-PK-
dependent DSB repair by NHEJ is inhibited in cancer cells. To monitor the DSB level
and repair kinetics in cancer cells, γH2AX-foci were counted at different time points
after IR (Fig. 1C). Thirty minutes after IR, the number of cells with more than 10 foci
increased and subsequently returned to baseline at 24 hours. Upon addition of
M3814 (1 uM), the reduced number of foci over the recovery period was significantly
inhibited compared with the DMSO-treated control, indicating that DNA-PK inhibition effectively suppressed DSB repair.
M3814 sensitizes cancer cells to IR
M3814 reduced colony formation in combination with IR in a concentration-
dependent manner but showed no or limited activity in the absence of IR (Fig. 1D,
Table 2). Overall, the antitumor potency correlated well with the inhibition of DNA-PK
autophosphorylation, indicating that DNA-PK inhibition sensitizes cells to IR. The
ability of M3814 to inhibit DNA-PK autophosphorylation and reduce colony formation
was observed in all cancer cell lines tested, regardless of tumor origin. However,
level of inhibition varied depending on the cancer cell line (Table 2). The antitumor 16
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
367
368
369
370
371
372
373
374
375
376
377
378
379
380
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
effect of M3814 was due to suppressing DNA-PK catalytic activity; in the
glioblastoma cell line, MO59J, which lacks DNA-PK activity (24), increasing
concentrations of M3814 did not further enhance sensitization to IR (Table 2, Fig.
1E). However, the DNA-PK-proficient cell line, MO59K, isolated from the same tumor
specimen, was less sensitive to IR alone but was sensitized by M3814 in a
concentration-dependent manner, suggesting that pharmacological inhibition of DNA-
PK is the key mechanism of radiosensitization. Enhancement factors at 10% colony
survival (EF10) for M3814 reached values between 2.5 and 6 in the 0.11–1 µM concentration range.
Suppression of colony formation is a widely used test to assess radiosensitization.
However, its application is limited because many cancer cell lines do not form
countable colonies (25). To assess M3814 activity in combination with IR in a large
panel of cancer cell lines, we used a sulforhodamine B-based cell growth/viability
assay. A random collection of 92 cancer cell lines, representing multiple tumor types,
381
was used in the assay. M3814 inhibited cell growth with a mean EC50
of 2.1 µM
382
383
384
385
(Fig. 2A, Supplementary Table S2). The effect on growth/viability in the absence of IR
was more pronounced than in the colony formation assay, probably due to extended
exposure to the compound. Nevertheless, the potency of inhibition by M3814 alone
was regarded as rather moderate. In combination with 3 Gy IR, synergistic growth
386
inhibition was observed in all cell lines, with a mean 12.4-fold EC50
shift in
387
sensitization (Mean EC50[M3814+IR]
= 0,17 µM; Supplementary Table S2,
388
389
390
391
Supplementary Fig. S1). Taken together, our data suggest that by inhibiting DNA-PK
activity and DSB repair, M3814 sensitizes cancer cells to IR through impaired colony outgrowth or proliferation/viability.
M3814 synergistically enhances the activity of DSB-inducing agents
17
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
392
393
394
395
396
397
398
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
To investigate the combination potential of M3814 with other anticancer agents, 35
randomly selected cancer cell lines were profiled with 72 drugs, representing diverse
antitumor mechanisms of action. The inhibitory effect of each agent was measured in
the presence or absence of 300 nM M3814 and Bliss synergy was calculated (20).
Bliss scores were classified as synergistic (>0.1), additive ( –0.1≤X≤0.1), or
antagonistic (<-0.1; Fig. 2B, Supplementary Fig. S2). No significant combination
effect was observed with the majority of tested drugs in most cancer cell lines,
indicating that M3814 does not broadly synergize with anticancer agents. Notably,
M3814 synergy across different cell lines was observed with the radiomimetic drug,
bleomycin, and the topoisomerase inhibitors, doxorubicin and etoposide. These
agents induce DSBs via radical generation or topoisomerase 2 inhibition, indicating
that DNA-PK is important for repair of these lesions. Although sporadic synergism or
antagonism was observed in a few individual cell lines, implying that the genetic
make-up of the cancer cell lines may contribute to the combination effect, the overall
ability of M3814 to synergistically enhance the activity of other drugs was clearly
defined by their mechanism of action. Only three out of 72 drugs showed significant
synergism in 34 cancer cell lines; those drugs are known to induce DSBs. These
results clearly indicated that M3814 activity is derived from its molecular mechanism of action.
M3814 inhibits radiation-induced DNA-PK autophosphorylation and DSB repair in tumor xenograft models
Next, we asked whether M3814 could inhibit its target and the repair of IR-induced
DSBs in tumor tissues in vivo. We performed PK/PD analyses in tumor xenograft
studies. Initial results indicated that detection of DNA-PK autophosphorylation at low
IR doses of 1–5 Gy is technically difficult and would not allow accurate quantification, 18
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
417
418
419
420
421
422
423
424
425
426
427
428
429
430
431
432
433
434
435
436
437
438
439
440
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
while a higher IR dose (10 Gy) resulted in a detectable increase of pSer2056 in DNA-
PK, which returned to baseline levels over 24 hours (Fig. 3A). Following oral
administration of a single dose of 25 mg/kg M3814, the plasma concentration of the
parent compound increased to approximately 3 µM 1.5-hours post-dose, followed by
elimination over the next 24 hours. IR-induced Ser2056 phosphorylation of DNA-PK
was suppressed at 1.5 hours to below baseline levels and remained low 3 hours after
IR. The signal increased at 8 hours, when the plasma concentrations of M3814 were
lower, and returned to baseline levels at 24 hours. Autophosphorylation of DNA-PK was reduced in a concentration-dependent manner at 1.5 hours (Fig. 3B).
To assess the effect of DNA-PK inhibition on DSB repair, we developed an immunofluorescence-based assay for quantification of H2A.X foci in tumor tissue
(Fig. 3C, see methods), which was used to measure H2A.X levels in FaDu tumors.
A single dose of IR (10 Gy) was given in the presence or absence of M3814 (200 mg/kg) and H2A.X signals were quantified in xenograft tumors obtained at different
intervals post-IR. M3814 administration changed the kinetics of the H2A.X signal
compared with vehicle-treated tumors, indicating suppression of DSB repair (Fig. 3C,
D). Taken together, these results indicate that M3814 administration inhibited IR-
induced DNA-PK activation, leading to increased DSB levels, both consistent with the exposure dynamics of M3814 in mouse plasma.
Preincubation of human blood with M3814 attenuates DNA-PK phosphorylation
Pharmacodynamic (PD) data obtained from human tumor biopsies helps to confirm
the drug reaches its molecular target in the target tissue. However, the possibility to
sample tumor tissue in early clinical trials is rather low, making it difficult to build a
robust PK/PD model. Analyses of PD biomarkers from surrogate tissues, such as 19
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
441
442
443
444
445
446
447
448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
blood, offer the possibility for regular sampling in parallel to routine PK analyses. For
this purpose, we developed a PD biomarker assay to detect DNA-PK
autophosphorylation in peripheral blood cells. DNA-PK phosphorylation was induced
by incubation of PBMCs from healthy human donors with bleomycin for 4 hours (Fig.
3E). Preincubation of human blood with different concentrations of M3814 ranging
from 1 nM to 30 µM attenuated DNA-PK phosphorylation in a concentration-
dependent manner. The potency of inhibition was within the same range observed for
cancer cell lines. These data demonstrated that PD analyses from surrogate tissue are feasible and may be useful for clinical exploration of M3814.
M3814 strongly potentiates IR efficacy in xenograft models of human cancer
M3814 demonstrated good oral bioavailability and PK in mice (Supplementary Table
S3, Supplementary Fig. S3); thus, further studies were performed to assess
antitumor activity and evaluate safety in mouse xenograft models. Subcutaneously
established FaDu or NCI-H460 tumors were treated with a 5-day fractionated
radiation regimen (2 Gy IR fraction per tumor/mouse per day), with or without M3814
administered by oral gavage 10 minutes before IR. In both tumor models, tumor
growth was moderately but significantly retarded with IR alone (Fig. 4A, B). However,
co-administration of M3814 led to enhanced tumor growth inhibition in both tumor
models. Tumor growth inhibition was dependent on the dose of M3814 administered
(Fig. 4A). Additional human xenograft models of different origin were investigated
(A549, BxPC3, Capan-1, and HCT-116) using the same IR regimen. Again,
significant tumor growth inhibition was seen when M3814 was combined with IR for 1
week (Supplementary Fig. S4). NHEJ repair is independent of the cell cycle phase,
fast, and contributes to the repair of most DSB lesions during the first hours after
radiation (26). Indeed, the results of scheduling experiments of M3814 in relation to 20
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
466
467
468
469
470
471
472
473
474
475
476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
IR support this hypothesis (Supplementary Fig. S5). When M3814 was administered
10 minutes prior to IR, the combination benefit was clearly superior than when
administered 3 or 6 hours after IR. Since M3814 is rapidly cleared in mice,
administration of an additional, consecutive dose of M3814 (first dose 10 minutes
before IR, second dose 3-hours later) also improved antitumor activity. However,
administration of M3814 for additional days after IR did not further improve antitumor efficacy indicating that DSBs have already been repaired.
In clinical practice, fractionated radiation regimens are commonly used for the
treatment of multiple solid tumor types. The duration of radiation therapy with curative
intent usually extends several weeks of therapy and applies fractionated doses of
1.8–2.0 Gy, for a total dose of approximately 50–70 Gy, depending on the tumor type
and location (27). Therefore, we applied a 6-week radiotherapy regimen (5 days on, 2
days off) with 2 Gy fractions to subcutaneously implanted mouse xenograft tumors
and investigated the effect of different doses of M3814 together with IR (Fig. 4C, D,
Supplementary Fig. S6). In the FaDu head and neck cancer model, IR led to a partial
regression of tumors during the treatment phase (Days 1–42); however, tumor growth
progressed upon cessation of IR (Fig. 4C). With increasing doses of M3814
administered orally 10 minutes before each IR fraction, the tumor response was
significantly enhanced, with increased tumor growth retardation at 5 and 10 mg/kg
and complete regression of all tumors observed at 25 and 50 mg/kg doses. Of note,
the strong response to combination therapy extended beyond treatment throughout
the observation period (Days 43–106). Since the FaDu model is sensitive to IR alone
(28), we assessed the impact of combination therapy in the relatively radio-resistant
non-small cell lung cancer model, NCI-H460 (Fig. 4D). Indeed, this model was
significantly less responsive to IR alone compared with FaDu tumors. During the 21
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
491
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
treatment period, H460 xenografts continued to grow following treatment with IR
alone. However, co-administration of M3814 and IR significantly increased the
antitumor response during the treatment and observation period. In five of 10 animals
treated with 25 mg/kg M3814 plus IR, tumors progressed during the observation
period (Days 49–97). Conversely, durable tumor responses were observed in the other five mice, and in all 10 mice treated with 50 mg/kg M3814 plus IR.
In general, the combination of M3814 plus IR was well-tolerated (Supplementary Fig.
S6). During the treatment period, animals in all treatment groups showed a moderate
loss of body weight (less than 10%), likely due to the daily (5 days on, 2 days off)
treatment procedure, including anesthesia, oral gavage, and IR over 6 weeks.
However, the body weight loss was fully reversible, and mice recovered during the
observation period. In the FaDu efficacy study, grade 1 acute dermatitis was
observed in only three out of 10 mice in the combination group receiving 50 mg/kg
M3814 after 42 days of treatment; however, this was fully reversible after 14 days. No signs of dermatitis were observed in the other groups (29).
Applying clinically relevant IR treatment regimens to human tumor models in mice
confirmed the remarkable antitumor activity of M3814 in combination with IR, which warrants clinical exploration.
22
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
509
510
511
512
513
514
515
516
517
518
519
520
521
522
523
524
525
526
527
528
529
530
531
532
533
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Discussion
M3814 is a potent inhibitor of DNA-PK catalytic activity with remarkable selectivity
against most of the 284 protein kinases we tested, including the closest members of
its own kinase family. Cell-based experiments indicated that the in vitro selectivity is
retained in cancer cells, albeit with reduced potency due to high ATP concentrations
and possibly other factors, such as the nuclear localization of the target. Neither the
ATR-CHK1 nor ATM-CHK2 pathways were significantly inhibited by M3814 in cancer
cells at concentrations that effectively suppressed the DNA-PK pathway. Residual inhibitory activity against three closely related lipid kinases, PI3K hasbeen
noted. However, phosphorylation of AKT in PTEN-mutated prostate carcinoma cells,
which can be potently blocked by PI3K inhibitors, was only marginally inhibited.
M3814 inhibited DSB repair and sensitized cancer cells to IR. Treatment of cancer
cells with M3814 attenuated foci resolution, indicating inhibition of DSB repair. Radio-
sensitization was observed in most tested cancer cell lines, regardless of tissue
origin, and confirmed using two different assay formats. The level of synergy varied
moderately between cancer cell lines, suggesting that other factors have a subtle
influence on sensitivity to the combination treatment. However, as expected, there
was a good correlation between sensitivity to IR alone and to combination treatment
with M3814. This suggests that the DNA-PK inhibitor enhances existing sensitivity to
radiation rather than engaging a different antitumor mechanism. Strikingly, the
glioblastoma line MO59J, which is devoid of DNA-PK activity (24), could not be
radiosensitized by M3814, whereas the DNA-PK-proficient cell line, MO59K, which
originates from the same tumor specimen, was sensitive to M3814. These data
indicate that inhibition of DNA-PK kinase activity is the key mechanism underlying
radiosensitization by M3814. Taken together, our results suggest that inhibition of the 23
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
534
535
536
537
538
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
canonical NHEJ pathway is responsible for the strong antitumor effects of M3814 when combined with IR.
Profiling M3814 in combination with 72 diverse established and developmental
antitumor agents revealed potential for combination with the chemotherapeutics
bleomycin, etoposide, and doxorubicin. These synergistic combinations were
anticipated based on their mechanisms of action, which involve the generation of
DSBs through inhibition of the topoisomerase enzymatic cycle leading to the
generation of Top2:DNA complexes. DNA-modifying agents (alkylators,
antimetabolites, and topoisomerase 1 inhibitors) did not synergize with M3814, since
their primary mechanism of repair does not require the canonical NHEJ. Similarly,
multiple other mechanisms represented by the large panel of tested drugs do not
provide combination benefit, further indicating that M3814 acts exclusively by inhibiting NHEJ.
Exposure-dependent inhibition of DNA-PK autophosphorylation, and the subsequent
delay in DSB repair kinetics observed in xenograft tissues in vivo, indicated that
M3814 possesses all the necessary properties to evaluate the therapeutic potential
of DNA-PK inhibition in xenograft models. To this aim, we selected two xenograft
tumor models with different sensitivity to IR and the most relevant clinically
established regimen for radiation therapy; 6-week fractionated radiation dosing. Pilot
1-week fractionated radiation studies showed that the combination of M3814 and IR
is beneficial over IR alone. However, the 6-week animal trials, which modeled a
clinically relevant fractionated radiation regimen with curative intent, demonstrated
strong and durable antitumor activity of the M3814/IR combination. Local tumor
radiation with M3814 exposures predicted to be achievable in man caused durable
tumor regression in multiple animals in both models. In general, M3814 was well- 24
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
559
560
561
562
563
564
565
566
567
568
569
570
571
572
573
574
575
576
577
578
579
580
581
582
583
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
tolerated with moderate but completely reversible body weight loss. In one xenograft
study grade 1 radiation-induced dermatitis was observed in three out of ten animals
receiving the highest dose of M3814 (50 mg/kg) in combination; however, the
symptoms were fully reversible. Lower M3814 doses, which also produced complete
regressions, did not lead to a skin reaction. However, the limited toxicological data
presented here do not sufficiently address the impact of M3814 on other normal
tissues in the radiation field, especially toxic effects that manifest themselves
significantly later post-radiation treatment. Further combination studies with M3814
and dedicated toxicological assessments will help to better evaluate radiation-related toxicities.
Data on the discovery and characterization of a novel DNA-PK inhibitor, M3814
presented in this manuscript offer a basis to explore its activity in combination with
radiotherapy and other DSB-inducing therapies in the clinic. Furthermore, the data
provide preclinical proof of concept for selective pharmacological inhibitors of DNA-
PK as combination partners of clinically established regimens for radiation therapy.
Currently ongoing clinical investigations with M3814 in both monotherapy and
radiotherapy combination settings should answer many outstanding questions
regarding their therapeutic potential (NCT02316197, NCT02516813). Acknowledgments:
We would like to thank Isabella Schmele, Nina Heiss, Jesse Alderson, and Florian
Szardenings for their help in the preparation of this manuscript. Editorial assistance
was provided by David Lester and Lisa Jolly, PhD, of Bioscript Science, Macclesfield, UK, and funded by Merck KGaA, Darmstadt, Germany.
Authors contributions:
Conception and design:
25
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, T. Grombacher, L. Vassilev, C. Wilm, T.
Development of methodology:
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, T. Grombacher, C. Wilm, T.
Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.):
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, C. Wilm Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
computational analysis):
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, T. Grombacher, C. Wilm, T., L. Vassilev
Writing, review, and/or revision of the manuscript:
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, T. Grombacher, C. Wilm, T. Fuchss, C. Amendt, L. Vassilev, A. Blaukat
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases):
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, U. Pehl, T. Grombacher, C. Wilm, C. Amendt, L. Vassilev
Study supervision:
F.T. Zenke, A. Zimmermann, C. Sirrenberg, H. Dahmen, V. Kirkin, U. Pehl, C. Wilm, Other (compound design and synthesis):
T. Fuchss
26
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
657
658
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
References
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646-74
doi 10.1016/j.cell.2011.02.013.
2. Desai A, Yan Y, Gerson SL. Advances in therapeutic targeting of the DNA damage response in
cancer. DNA Repair (Amst) 2018;66-67:24-9 doi 10.1016/j.dnarep.2018.04.004.
3. Chapman JR, Taylor MR, Boulton SJ. Playing the end game: DNA double-strand break repair pathway choice. Mol Cell 2012;47(4):497-510 doi 10.1016/j.molcel.2012.07.029.
4. Kasparek TR, Humphrey TC. DNA double-strand break repair pathways, chromosomal rearrangements and cancer. Semin Cell Dev Biol 2011;22(8):886-97 doi 10.1016/j.semcdb.2011.10.007.
5. Kakarougkas A, Jeggo PA. DNA DSB repair pathway choice: an orchestrated handover mechanism. Br J Radiol 2014;87(1035):20130685 doi 10.1259/bjr.20130685.
6. Davis AJ, Chen BP, Chen DJ. DNA-PK: a dynamic enzyme in a versatile DSB repair pathway. DNA Repair (Amst) 2014;17:21-9 doi 10.1016/j.dnarep.2014.02.020.
7. Salles B, Calsou P, Frit P, Muller C. The DNA repair complex DNA-PK, a pharmacological target
in cancer chemotherapy and radiotherapy. Pathol Biol (Paris) 2006;54(4):185-93 doi 10.1016/j.patbio.2006.01.012.
8. Dobbs TA, Tainer JA, Lees-Miller SP. A structural model for regulation of NHEJ by DNA-PKcs autophosphorylation. DNA Repair (Amst) 2010;9(12):1307-14 doi 10.1016/j.dnarep.2010.09.019.
9. Taccioli GE, Amatucci AG, Beamish HJ, Gell D, Xiang XH, Torres Arzayus MI, et al. Targeted
disruption of the catalytic subunit of the DNA-PK gene in mice confers severe combined immunodeficiency and radiosensitivity. Immunity 1998;9(3):355-66.
10. Kurimasa A, Ouyang H, Dong LJ, Wang S, Li X, Cordon-Cardo C, et al. Catalytic subunit of DNA- dependent protein kinase: impact on lymphocyte development and tumorigenesis. Proc Natl Acad Sci U S A 1999;96(4):1403-8.
11. Azad A, Jackson S, Cullinane C, Natoli A, Neilsen PM, Callen DF , et al. Inhibition of DNA- dependent protein kinase induces accelerated senescence in irradiated human cancer cells. Mol Cancer Res 2011;9(12):1696-707 doi 10.1158/1541-7786.MCR-11-0312.
12. Block WD, Merkle D, Meek K, Lees-Miller SP. Selective inhibition of the DNA-dependent protein kinase (DNA-PK) by the radiosensitizing agent caffeine. Nucleic Acids Res 2004;32(6):1967-72 doi 10.1093/nar/gkh508.
13. Daido S, Yamamoto A, Fujiwara K, Sawaya R, Kondo S, Kondo Y. Inhibition of the DNA- dependent protein kinase catalytic subunit radiosensitizes malignant glioma cells by inducing autophagy. Cancer Res 2005 ;65(10):4368-75 doi 10.1158/0008-5472.CAN-04-4202.
14. Hashimoto M, Rao S, Tokuno O, Yamamoto K, Takata M, Takeda S , et al. DNA-PK: the major target for wortmannin-mediated radiosensitization by the inhibition of DSB repair via NHEJ pathway. J Radiat Res 2003;44(2):151-9.
15. Munck JM, Batey MA, Zhao Y, Jenkins H, Richardson CJ, Cano C , et al. Chemosensitization of cancer cells by KU-0060648, a dual inhibitor of DNA-PK and PI-3K. Mol Cancer Ther 2012;11(8):1789-98 doi 10.1158/1535-7163.MCT-11-0535.
16. Dong J, Ren Y, Zhang T, Wang Z, Ling CC, Li GC , et al. Inactivation of DNA-PK by knockdown DNA-PKcs or NU7441 impairs non-homologous end-joining of radiation-induced double strand break repair. Oncol Rep 2018;39(3):912-20 doi 10.3892/or.2018.6217.
17. Zhuang W, Li B, Long L, Chen L, Huang Q, Liang ZQ. Knockdown of the DNA-dependent protein kinase catalytic subunit radiosensitizes glioma-initiating cells by inducing autophagy. Brain Res 2011;1371:7-15 doi 10.1016/j.brainres.2010.11.044.
18. Fuchss T, Emde U, Buchstaller H-P, Mederski WKR; Arylquinazolines 2014;Patent WO2014183850A1, example 136, (S)-[2-Chloro-4-fluoro-5-(7-morpholin-4-yl-quinazolin-4-yl)-
phenyl]-(6-methoxy-pyridazin-3-yl)-methanol.
27
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
19. Vichai V, Kirtikara K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc 2006;1(3):1112-6 doi 10.1038/nprot.2006.179.
20. Berenbaum MC. What is synergy? Pharmacol Rev 1989;41(2):93-141.
21. Lempiainen H, Halazonetis TD. Emerging common themes in regulation of PIKKs and PI3Ks. EMBO J 2009;28(20):3067-73 doi 10.1038/emboj.2009.281.
22. Sun Q, Guo Y, Liu X, Czauderna F, Carr MI, Zenke FT , et al. Therapeutic implications of p53 status on cancer cell fate following exposure to ionizing radiation and the DNA-PK inhibitor M3814. Mol Cancer Res 2019 doi 10.1158/1541-7786.MCR-19-0362.
23. Sharrard RM, Maitland NJ. Regulation of protein kinase B activity by PTEN and SHIP2 in human prostate-derived cell lines. Cell Signal 2007;19(1):129-38 doi 10.1016/j.cellsig.2006.05.029.
24. Hoppe BS, Jensen RB, Kirchgessner CU. Complementation of the radiosensitive M059J cell
line. Radiat Res 2000;153(2):125-30.
25. Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in
vitro. Nat Protoc 2006;1(5):2315-9 doi 10.1038/nprot.2006.339.
26. Lobrich M, Jeggo P. A Process of Resection-Dependent Nonhomologous End Joining Involving the Goddess Artemis. Trends Biochem Sci 2017;42(9):690-701 doi 10.1016/j.tibs.2017.06.011.
27. Arnold KM, Flynn NJ, Raben A, Romak L, Yu Y, Dicker AP, et al. The Impact of Radiation on the Tumor Microenvironment: Effect of Dose and Fractionation Schedules. Cancer Growth Metastasis 2018 ;11:1179064418761639 doi 10.1177/1179064418761639.
28. Kasten-Pisula U, Menegakis A, Brammer I, Borgmann K, Mansour WY, Degenhardt S , et al. The extreme radiosensitivity of the squamous cell carcinoma SKX is due to a defect in double- strand break repair. Radiother Oncol 2009;90(2):257-64 doi 10.1016/j.radonc.2008.10.019.
29. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys 1995;31(5):1341-6 doi 10.1016/0360-3016(95)00060-C.
28
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
688
689
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Tables
Table 1: M3814 potency and selectivity
Assay IC50 [nM]
DNA-PK, IC50 [10 µM ATP] 0.6
DNA-PK, IC50 [1,000 µM ATP] 20
Protein Kinases Panel profiling 276/284 not inhibited ≥ 50% at 1µM
PIKK Family Members ATM 10,000
PIKK Family Members ATR 2,800
PIKK Family Members hPI3Kalpha 330
PIKK Family Members hPI3Kbeta 250
PIKK Family Members hPI3Kgamma >1,000
PIKK Family Members hPI3Kdelta 95
PIKK Family Members mTOR >10,000
HU-induced pS345-CHK1 >25,000
Bleomycin-induced pT68-CHK2 >5,000
Constitutive pS473-AKT in PC3 3,800
690
ATM, ataxia telangiectasia-mutated; ATP, adenosine triphosphate; ATR, ATM and
691
692
693
694
rad3-related; CHK, checkpoint kinase; DNA-PK, DNA-dependent protein kinase; IC50 half maximal inhibitory concentration; mTOR, mammalian target of rapamycin.
,
29
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
695
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Table 2: Cellular profiling of M3814 in cancer cell lines
Cellular profiling of M3814
Cancer
cell line
Origin Bleomycin-
induced
pDNA-PK, IC
50
[M] Clonogenicity ± irradiation, IC50 [M]
Cancer
cell line
Origin Bleomycin-
induced
pDNA-PK, IC
50
[M]
0 Gy
2.4 Gy
4.8 Gy
MO59J Brain n.d. 4 x10-5 No concentration-
dependence
MO59K Brain 2 x 10 1 x 10 4 x 10 3 x 10
HCT-116 Colon 1 x 10 >1 x 10 2 x 10 8 x 10
HT29 Colon 1 x 10 2 x 10 4 x 10 2 x 10
FaDu Head and neck 6 x 10 1 x 10 1 x 10 5 x 10
A549 Lung 2 x 10 1 x 10 1 x 10 5 x 10
Calu-6 Lung 2 x 10 1 x 10 7 x 10 3 x 10
EBC-1 Lung 2 x 10 1 x 10 1 x 10 5 x 10
NCI-H460 Lung 1 x 10 3 x 10 2 x 10 1 x 10
BxPC-3 Pancreas 2 x 10 1 x 10 1 x 10 7 x 10
KP-4 Pancreas 4 x 10 3 x 10 2 x 10 7 x 10
MiaPaCa Pancreas 5 x 10 > 5 x 10 2 x 10 9 x 10
DU145 Prostate 3 x 10 2 x 10 3 x 10 1 x 10
696
697
698
DNA-PK, DNA-dependent protein kinase; Gy, Gray unit; IC50, half maximal inhibitory concentration.
30
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Figure legends
Figure 1: M3814 in combination with IR inhibits DNA-PK activity and reduces cancer cell survival in a DNA-PK-dependent manner.
A) Chemical structure. B) Concentration-dependent inhibition of DNA-PK
autophosphorylation by M3814 in bleomycin-treated cancer cell lines, HCT-116 and
FaDu, as shown by western blotting. Total DNA-PK was used as a loading control.
Representative images are shown. C) M3814 (1E-6 M) in combination with IR (2.4
Gy) suppressed DSB repair, as assessed by the number of γH2A.X foci in cells. A
high basal level of γH2A.X foci was noted in A375 cells; therefore, the percentage of
cells with >10 γH2A.X foci was calculated and the time course of foci dynamics is
plotted (bottom). Representative immunofluorescent images of vehicle-treated A375
cells without IR (top image) and 30-minutes after IR (2.4 Gy) (bottom image) are
shown. D) M3814 (4E-7 M, 2E-8 M) in combination with IR (2.4 Gy, 4.8 Gy) reduced
colony formation in FaDu and HCT-116 cancer cells in a clonogenic cell survival
assay. Visible colonies were stained with neutral red. Representative images of at
least three assays are shown. E) M3814 in combination with IR reduced colony
formation of glioblastoma cell lines in a DNA-PK-dependent manner. M3814
combined with IR had no effect on colony survival versus vehicle in the DNA-PK-
deficient glioblastoma cell line, MO59J (top panel). With increasing concentrations of
M3814 combined with IR, the surviving fraction of colonies was reduced versus
vehicle in the DNA-PK-proficient glioblastoma cell line, MO59K (bottom panel).
Figure 2: In combination with IR, M3814 broadly reduces the viability of cancer
cell lines and synergistically enhances the activity of DNA DSB-inducing drugs.
31
Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.
723
724
725
726
Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
A) Sulforhodamine B-based cell growth/viability of 92 cancer cell lines in response to M3814 alone and in combination with IR (3 Gy). Calculated EC50 from M3814 (5E-9–
5E-5 M) titrations alone (grey bars) or in combination with 3 Gy IR (red bars) are
presented for all tested cell lines. Cell lines were sorted from low to high sensitivity to
727
M3814 alone. Mean EC50[M3814]
= 2.1 µM and mean EC50
[combination]
= 0,17 µM are
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746
747
displayed as lines. B) Heat map of Bliss synergy scores for combination profiling of
72 drugs, including DNA-damaging agents, tested in 34 cancer cell lines; synergistic (≥0.1; red), additive (–0.1 Figure 3: M3814 inhibits radiation-induced DNA-PK activation leading to increased DSB levels in tumor tissue from mice xenograft models; assessment of DNA-PK phosphorylation and inhibition by M3814 in human blood. A) Time course of DNA-PK phosphorylation in the FaDu tumor xenograft model in response to IR (10 Gy) alone (black line) or the combination of M3814 (25 mg/kg) with IR (10 Gy, red line). Data from a representative PK/PD study are shown. The ratio of phosphorylated to total DNA-PK was determined in five tumors per treatment group and time points, and the mean +/- SEM are presented. The mean plasma concentration of M3814 over time following oral administration is shown in green. B) The ratio of phosphorylated to total DNA-PK was determined in FaDu xenograft tumors at baseline (no IR), after 10 Gy IR alone, and following treatment with IR (10 Gy) combined with M3814 (5, 25, 100 mg/kg). Tumors were processed 1.5 hours after treatment (five tumors per group). C) Impaired DSB repair over time in FaDu tumors following treatment with vehicle (left image) or 200 mg/kg M3814 (right image) and IR (10 Gy). Representative immunofluorescence images detecting H2A.X in tumor sections are shown (Scale bar = 2 µm). D) Quantification of time-dependent 32 Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research. 748 749 750 751 752 753 754 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. resolution of H2A.X staining from the experiment described under C. E) DNA-PK phosphorylation was induced by incubating whole blood from healthy donors with bleomycin for 4 hours (left). Preincubation of whole blood with M3814 attenuated bleomycin-induced DNA-PK phosphorylation in a concentration-dependent manner, cccccccccccccccccccccccccccc M3814 in human blood samples. Representative dddddddddddddddddddddddddddd (blood from 6 different donors was tested). Figure 4: M3814 in combination with IR has antitumor activity in mouse xenograft models. A) M3814 (oral gavage 10-minutes before IR; dose groups: 25, 100, 150 mg/kg) in combination with a 5-day fractionated radiation regime (2 Gy IR fraction/mouse per day) dose-dependently reduced tumor growth in a human FaDu xenograft model to a greater extent than IR alone. Data represent the mean +/- SEM (group size N=10). B) M3814 (oral gavage 10-minutes before IR; dose: 150 mg/kg) in combination with a 5-day fractionated radiation regime (2 Gy IR fraction/mouse per day) reduced tumor growth in a human NCI-H460 xenograft model. Data represent the mean +/- SEM (group size N=10). C) M3814 (5, 10, 25, 50 mg/kg) in combination with a 6-week radiotherapy regime (2 Gy fractions, 5 days on, 2 days off) demonstrated dose-dependent antitumor activity in a human FaDu xenograft model. Data represent the mean +/- SEM (group size N=10). D) M3814 (25 and 50 mg/kg) in combination with a 6-week radiotherapy regime (2 Gy fractions, 5 days on, 5 days off) demonstrated antitumor activity in a human radio-resistant NCI-H460 xenograft model (group size N=10). 33 Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research. Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research. Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Associat Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research. Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 Ame Author Manuscript Published OnlineFirst on March 27, 2020; DOI: 10.1158/1535-7163.MCT-19-0734 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Pharmacological inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models Frank T. Zenke, Astrid Zimmermann, Christian Sirrenberg, et al. Downloaded from mct.aacrjournals.org on April 6, 2020. © 2020 American Association for Cancer Research.Nedisertib
755
Mol Cancer Ther Published OnlineFirst March 27, 2020.