Blocking Genomic Instability Prevents Acquired Resistance To MAPK Inhibitor Therapy in Melanoma
Blocking Genomic Instability Prevents Acquired Resistance To MAPK Inhibitor Therapy in Melanoma
can be identified as intrachromosomal complex genomic rear- (NSG) mice at doses sufficient to elicit tumor regression
rangements (CGR) and extrachromosomal DNAs (ecDNA, and then generated acquired MAPKi-resistant tumors (n = 6
aka double minutes), which may be temporally related struc- vehicle-treated tumors; n = 12 acquired-resistant tumors;
tures that confer a range of genomic–signaling plasticity (13). n = 6 normal tissues; Supplementary Fig. S1A; refs. 11,
Earlier studies have established the prognostic importance of 22). Sequencing depths across MAPKi-sensitive/naive tumors
ecDNAs in clinical tumor tissues and the potential relevance of (mean, 38×; median, 32×; range, 18–94×), acquired-resistant
ecDNAs to therapeutic resistance in cancer cell lines (14, 15). tumors (mean, 33×; median, 27×; range, 11–85×), and patient-
The non-Mendelian inheritance of ecDNAs, their accessible matched normal tissues (mean, 38×; median, 33×; range,
chromatin, as well as enhancer hijacking and transcriptional 12–98×) were comparable. The median numbers of genetic
hub congregation by ecDNAs are all mechanisms that facili- alterations [somatic single-nucleotide variants (SNV), indels
tate rapid adaptations to extreme stress such as oncogene- (ID), and copy-number variations (CNV)] for sensitive and
targeted therapy (16–19). Finally, iterative cycles of CGR and resistant tumors were, respectively: (i) 81 SNVs/megabase
ecDNA biogenesis drive the amplification of a model resist- (Mb), 5 IDs/Mb, and 261 CNVs/genome and (ii) 97 SNVs/
ance gene (DHFR) to the chemotherapy methotrexate (20). Mb, 7 IDs/Mb, and 253 CNVs/genome.
To test the hypothesis that chromothripsis, CGRs, and
ecDNAs play key roles in the evolution of MAPKi resist- Recurrence, Quantity, Chromosomal Origin, and
ance in clinical melanoma, we analyzed by whole-genome Complexity of ecDNAs and CGRs in Acquired-
Resistant versus MAPKi-Naive Melanoma
Cohorts
Clinical
1q
Rapid autopsy
1p
2q PDX
2p
3q Genotype
3p
BRAF MUT
4q
5q NRAS MUT
Chromosome locations
5p
6q
Sensitive
6p
7q Acquired resistant
7p
8q
8p Genomic alterations
9q CGR
9p ecDNA
10q
M _P X R2
Pt P 1.D ne
PT Pt5.Dine
M l_Ple.DD.D P
M _P X V1
Pt P sel 3
Pt4.D ase P1
Pt .D .D line
Pt DD DP A
9. t7. ine
M l_P X2 .R2
M _P X V1
M l_PDX .V1
M l_PDX .R4
M _P X R5
X6 1
6. 5. P1
Ba D e
Pt t4.B t3.Dline
M PDX27.R1
M l_P DX .R3
el D 2. 2
M l_PDX .R2
AM M 0 Br n. .D ve
5. .D 2B
Pt P sel 2
M l_PDX 7.VP
Pt P sel 2
P1
A 4 0 . in D P
M l_PDX .R1
M _P X R4
D 6.V5
3
R RA M1 006 ren g.DD.D 9
R M1 .01 12. t9. D. P2
R M1 14 006Braal.D D.DP
R 1 .01 rai .S D.DP1
M l_PDX D.DP
el D 27 1
R RA 1301. rai DD siti 2
P sel C
14 06 Lymung .DD.DP
P1
P
7. t6. in
M l_PDX .R
P
_P X R
.R
A 4 d n D P
A 0 3 ra D P
00 .Ly ph .D .D
AM 2. .B n. en P
4. D. P1
M h D D
R M1 006 06 Liv .D .D
3
i
1 0 a D P
Ba D
el D 1.
el D 2.
Ba D
Ba .D
P D D
AM 2 .B 01 D D
el D 3.
Pt sel
el D 4.
el D 4.
Ba D
Ba P
4
3
P P e
3
P P e
4
1
s
s
2
Ba
t
4 D
1.
Pt
e
e
e
e
e
e
e
e
e
e
A M 4 .
AM R
A
R
Figure 1. Landscape of ecDNAs and CGRs in patient-matched MAPKi-sensitive and acquired resistant melanoma. Chromosomal distribution of ecDNA
and CGR amplicons in three cohorts: BRAFV600MUT clinical cohort (baseline tumors, n = 8; resistant tumors, n = 11; patients, n = 8), RAM tissues (sensitive
tumor, n = 1; resistant tumors, n = 12; deceased patients, n = 3), and PDXs (vehicle-treated or sensitive, n = 6; resistant tumors, n = 12; patients of origin,
n = 6). Os and Xs, ecDNAs and CGRs, respectively; sizes, numbers of CN variant segments (minimal CN of 4.5; reference sizes shown). Pretreatment and
post–acquired resistance tumors from Pt10 and Pt11 are not shown given no detection of ecDNAs and CGRs.
t test, P = 0.0002) association between acquired-resistant by RAC1 (23). Moreover, we validated a recurrent ecDNA by
tumors and CGRs/ecDNAs harboring bona fide MAPKi direct isolation and high-depth sequencing [NRAS amplifica-
resistance–driver genes. BRAF (CN range, 4.5–27), NRAS tion, CN 13, Pt9-double drug-disease progression (DD-DP)1;
(CN, 5–13), HRAS (CN, 13–16), MYC (CN, 12–15), and EGFR Fig. 2A] using a new approach referred to as CRISPR-CATCH
(CN, 4.6–5; Figs. 1 and 2A–D) when amplified are known to (24). This alternative technique confirmed the circularized
drive acquired MAPKi resistance and MAPK pathway reacti- junctions of an 890-kb driver ecDNA within this acquired-
vation (5, 7–9, 11). RAC1 (CN, 6–7) in an ecDNA amplicon resistant clinical tumor sample. In addition, we used DNA-
was specifically observed in acquired resistance in NRASMUT FISH to validate the resistance-specific presence and the CNs
melanoma PDXs, which suggests regulation of MLK3–CRAF of BRAF and NRAS amplicons (Fig. 2E). In total, among the 17
125 25
Mel_PDX27-V1
Mel_PDX1-V1
Mel_PDX2-V1
100 20
CN
75 15
50 10
25 5
0 0
TRIM33 NRAS
chr1
42518432
42539638
114441294
116371336
Mel_PDX27-R2
B
Mel_PDX1-R1
Mel_PDX2-R2
BRAF MUT—rapid autopsy (RAM12.01-Brain-DDP10)
1,200 80
1,000 60
Coverage
800
40
CN
600
400 20
200
0 0
CDK6 BRAF
Mel_PDX27 Mel_PDX1 Mel_PDX2
chr7 60 *** 15 *** 15 ***
92757501
93431000
140031001
141584000
NRAS CN
NRAS CN
10
BRAF CN
C 175
BRAF MUT
—PDX (Mel_PDX27-R2)
20 5 5
150 35
30
Coverage
125 25
100 20 0 0
CN
75 0
15 V1 R2 V1 R1 V1 R2
50 10
25 5
0 0
BRAF
chr7
139668001
141933200
1,500
CN
150 30 75
100 20 1,000 50
50 10 500 25
0 0 0 0
TRIM33 ATP1A1TENT5C NOTCH2 CREB2L2 TRIM24 BRAF EZH2 XRCC2
RBM15 NRAS
chr1 7 17 chr7 11
136414501
141637000
148317501
148882000
152566501
153202000
74487202
74497776
42051582
42072132
48185908
48196458
107638001
121714000
58086567
58097117
62266856
62277406
62374042
62384592
26734580
26755131
Figure 2. Genes amplified by ecDNAs and CGRs in acquired MAPKi resistance and enriched pathways. A–D, SV view of reconstructed amplicons in rep-
resentative acquired MAPKi-resistant melanoma tumors in the clinical (A), RAM (B), BRAFV600MUT PDX (C), and NRASMUT PDX (D) cohorts. Horizontal black
and red lines indicate, respectively, genomic segments with similar CNs and genes. Short vertical lines and arcs indicate discordant read pairs linking two
amplicons via an SV junction. Long vertical lines indicate break ends that map from amplicon into low-complexity regions that cannot be traced further.
Each line/arc representing discordant reads is colored based on differences from expected distance or orientation. Yellow arrows point out resistance-
driver genes within ecDNAs. DD-DP, double drug (BRAFi + MEKi)-disease progression. E, Representative images of DNA-FISH showing indicated patient-
matched tumors (n = 3 pairs) and CNs of BRAF or NRAS in either vehicle-treated/MAPKi-sensitive (top) or acquired MAPKi-resistant (middle) melanoma.
CEN, centromere; DAPI, nuclear stain. Scale bars, 25 μm. Quantification of DNA-FISH (bottom) from 40 nuclei per tumor; results shown as mean ± SD.
P values (unpaired two-tailed Student t test): ***, P < 0.001. F, SV view of reconstructed amplicons in clinically acquired resistance showing XRCC2 as a
putative resistance-driving gene. (continued on following page)
of 19 patients included in this study whose tumors harbored of MAPK-reactivation genes were highly positively correlated
ecDNAs/CGRs, MAPK-reactivation genes were amplified via with mRNA level changes (Spearman correlation rs = 0.64,
ecDNAs/CGRs specifically in acquired-resistant tumors (but P = 0.001; Supplementary Table S5 and Supplementary
not in any MAPKi-sensitive/naive tumor) at a high frequency Fig. S1F). In a minority of patients (Pt5, ecDNA+CGR+;
(60% or 23 of 38 resistant tumors; Supplementary Table S3). Pt11, ecDNA−CGR−; Supplementary Table S6), we observed
Expectedly, MAPKi resistance–driver gene amplification lower-level but linear amplifications of BRAF (CN, 3–4)
via ecDNAs/CGRs often co-occurred with other resistance- specifically in acquired-resistant tumors. The BRAFV600MUT
specific CN alterations and somatic mutations reported melanoma tumors from both patients were treated with
earlier (Supplementary Table S4; refs. 7–9). Using tumor- only BRAFi (not BRAFi + MEKi), suggesting that low-level
matched RNA-sequencing (RNA-seq) data available from a linear amplifications suffice in driving acquired resistance to
subset of tumors, CN alterations (resistant versus sensitive) BRAFi monotherapy.
G H
BRAF CGR genes in
ecDNA genes in EGFR
Sensitivity only
Sensitivity only 1,000 Resistance only
400 Resistance only NRAS Sensitivity & resistance
Sensitivity & resistance 800 Resistance & ecDNA
Resistance & CGR genes in sensitivity
Number of genes
Number of genes
300 genes in sensitivity 600
400
200
NRAS 300
BRAF
100 200
1
2
3
4
5
6
7
AM Pt9
_P 01
el 27
M PD 1
_ 2
_ 3
el X4
X6
1
2
3
4
5
6
7
M M1 9
_P 01
el 27
M PD 1
_ 2
_ 3
el X4
X6
Pt
Pt
Pt
Pt
Pt
Pt
Pt
el X
el X
el X
Pt
Pt
Pt
Pt
Pt
Pt
Pt
A Pt
el X
el X
el X
el 12
M DX
M PD
M PD
M PD
D
el 2
M DX
M PD
M PD
M PD
_P
_P
_
_
_
_
R
R
Mel_PDX27
Oncogenic MAPK signaling
Oncogenic MAPK signaling
Glycoside metabolic process
Acrosome assembly Pt3
Glycosyl compound metabolic process
Cellular component assembly involved in morphogenesis
Polyketide metabolic process
Secretory granular organization
0 2 4 6 8 0 2 4 6
–Log10 (P) –Log10 (P)
Mel_PDX1
Matrisome Potassium ion channel
Pt4
Mel_PDX4
Calcium activation phospholipid scrambling
Pt9
Mel_PDX6
Formation of the cornified envelope
Terminal pathway of complement Keratinization
Negative regulation of biosynthetic process GPCR signaling
Jak–STAT signaling pathway Cholesterol biosynthesis
0 2 4 6 0 5 10 15
–Log10 (P) –Log10 (P)
Figure 2. (Continued) G, Number of ecDNA-amplified genes (i) shared between pre/sensitive and post/acquired resistant tumors, (ii) specifically in
pre/sensitive tumors, (iii) specifically in post/acquired resistant tumors, and (iv) in post/acquired resistant tumors shared with CGR-amplified genes in
pre/sensitive tumors. Cohorts: clinical (n = 8 patients), RAM (n = 1 patient), and PDX models (n = 6 patients). Patients without sensitivity (n = 2) and with-
out ecDNAs and CGRs detected (n = 2) were excluded. H, As in G, except showing the number of CGR-amplified genes in the same (i) to (iii). (iv) Number of
CGR-amplified genes in post/acquired resistant tumors shared with ecDNA-amplified genes in pre/sensitive tumors. I, Pathway enrichment analysis of
genes amplified by ecDNAs and/or CGRs exclusively in acquired resistant tumors (n = 8). Only tumors with sufficient genes for enrichment analysis were
analyzed. GPCR, G protein-coupled receptor.
Nonhomologous end-joining (NHEJ) and homologous NRAS amplifications, respectively, harbored coamplification
recombination repair (HRR) of DSBs are thought to be of superenhancers (Supplementary Table S8), which suggests
critical for CGR and ecDNA generation after chromothripsis that genomic plasticity may facilitate epigenomic plasticity
(20). Intriguingly, we observed that XRCC2 (RAD51-like; CN, and alter gene expression on a large scale in MAPKi-resistant
49), a key HRR gene, and XRCC6 (KU70; CN, 14–15), a key tumors. In further support of ecDNAs playing a role in epige
NHEJ gene, were amplified specifically in acquired MAPKi- nomic plasticity, we observed that H3K27 acetylated genomic
resistant melanomas as ecDNAs (Fig. 2F; Supplementary regions (extracted from ENCODE data) were rearranged to the
Table S3). Moreover, acquired MAPKi resistance was associ- proximity of and coamplified with ecDNA genes (e.g., c-MYC
ated with ecDNA or CGR amplicons spanning other DSB and BRAF, respectively) in acquired resistance (Supplementary
repair genes in NHEJ (TRIM33 CN, 7–11; PAXIP1 CN, 3) and Fig. S1H). Enhancer docking sites within ecDNA amplicons
HRR (SSBP1 CN, 5; BRCA2 CN, 6; RFC3 CN, 6; TRIM33 CN, may also influence the differentiation state(s) of MAPKi-
7–11; SYCP1 CN, 7–9, TRIM24 CN, 5–11) pathways (Sup- resistant tumors by acting in trans on intrachromosomal genes
plementary Table S3). Thus, acquired MAPKi resistance is such as HOXA9, HOXA11, HOXA13, and LIFR (RAM12.01-
specifically associated with CGR and ecDNA amplicons in Brain-DD-DP10, RAM12.01-Brain-DD-DP3, and RAM12.01-
MAPK-reactivation and DSB repair genes, which suggests Brain-DD-DP9) and acting as mobile regulatory elements
functional interplay as codrivers of resistance. for genes such as SMO (Pt3-DP1; Supplementary Table S8)
Other resistance-specific genes amplified by ecDNAs and/ and ATP1A1 (Mel_PDX2-R2; Supplementary Fig. S1G). Thus,
If chromothripsis were a precursor step for ecDNA and acquired-resistant versus MAPKi-naive/sensitive tumors (Wil-
CGR generation during melanoma disease progression, we coxon rank sum test, P = 0.005; Supplementary Fig. S2C), sug-
expected a nonrandom overlap of affected genomic spans. gesting that MAPKi-selected chromothriptic regions in some
Consistent with expectation, we observed that the genomic individuals feature a relatively stronger mutator phenotype.
spans of ecDNAs and CGRs overlapped significantly or non- We have reported that MAPKi selection of clinical melano-
randomly with chromothriptic regions in ∼29% (5 of 17) mas alters the mutational spectra (9). To characterize how a
of MAPKi-sensitive/naive genomes and in ∼54% (22 of 41) MAPKi-associated mutator phenotype affects chromothrip-
of acquired MAPKi-resistant genomes (Fig. 3A). For every tic genomic spans selected by MAPKi, we culled chromothrip-
genome, we computed the sizes of ecDNAs + CGRs (a − x), sis-associated somatic mutations unique to MAPKi-sensitive/
chromothriptic regions (b − x), their overlap (x), and genomic naive (mean, 64 SNVs/Mb) versus MAPKi-resistant genomes
regions devoid of these genomic alterations (g − a − b). These (mean, 58 SNVs/Mb) and analyzed signatures of single-
four quantities were represented in 2 × 2 contingency tables base substitutions (SBS; Fig. 3B), double-base substitutions
to carry out the Fisher exact test. Using hypergeometric (DBS), and IDs (Supplementary Fig. S2D; 32, 33). In both
distribution, we then estimated the probability of observ- MAPKi-sensitive/naive and acquired-resistant tumors, we
ing random overlaps between ecDNA + CGR spans and frequently detected signatures of tobacco smoking (SBS4)
chromothriptic spans. In each tumor genome with over- and ultraviolet radiation (SBS7a and SBS38). Among chro-
laps between ecDNA + CGR and chromothriptic regions, we mothripsis-associated somatic mutations unique to either
A
Mel_PDX27-V1 Chromothripsis
Mel_PDX2-V1
Pt3-Baseline ecDNAs + CGRs
Pt7-Baseline overlapping
Pt10-Baseline chromothripsis
Mel_PDX1-V1 *** Nonoverlapping
Pt2-Baseline
ecDNAs + CGRs
Sensitive
Mel_PDX3-V1
Pt11-Baseline
Pt5-Baseline
Mel_PDX6-V1 ***
Pt6-Baseline
Pt4-Baseline ***
Pt1-Baseline
Pt9-Baseline ***
RAM12.01-Sensitive
Mel_PDX4-V1 ***
Mel_PDX27-R1
Mel_PDX6-R3
RAM14006-Lymph1-DD-DP
RAM14006-Liver-DD-DP
Pt5-DP2
Pt11-DP2
Pt4-DD-DP-2B ***
RAM13003-Brain-DD-DP ***
Pt9-DD-DP2 ***
RAM14006-Adrenal-DD-DP
Pt11-DP1
RAM13003-Lung-DD-DP ***
Pt9-DP2 ***
Pt7-DP1
Mel_PDX4-R5 ***
Mel_PDX2-R2 ***
RAM14006-Lymph2-DD-DP
Mel_PDX4-R4 ***
Mel_PDX3-R4 ***
Pt4-DD-DP-1A ***
RAM12.01-Brain-DD-DP9 ***
RAM14006-Muscle-DD-DP
Pt9-DD-DP1 ***
0 250 500 750
Size of region (Mb)
Figure 3. Associations of chromothripsis, ecDNAs, and CGRs with mutagenic and double-stranded DNA repair pathways. A, Pattern of overlap or
nonoverlap between ecDNA + CGR and chromothriptic genomic spans within individual sensitive (n = 17; top) and acquired resistant (n = 41; bottom) mel-
anoma tumors. Sizes (Mb) of chromothriptic regions are in cyan, ecDNAs + CGRs overlapping with chromothripsis in red, and ecDNAs + CGRs nonoverlap-
ping with chromothripsis in purple. P value (Fisher exact test) of nonrandom overlap between chromothriptic and ecDNA + CGR regions: ***, P < 0.00001.
(continued on following page)
B Cohorts
Clinical
Rapid autopsy
1.00 PDX
Proportion of mutational signatures
Genotype
BRAF MUT
NRAS MUT
0.75
Sensitive
Acquired
resistant
0.50 Signatures
SBS3
SBS4
SBS5
0.25 SBS6
SBS7a
SBS9
SBS10d
0.00 SBS18
SBS20
Pt1-Baseline
Pt2-Baseline
Pt3-Baseline
Pt4-Baseline
Pt5-Baseline
Pt6-Baseline
Pt7-Baseline
Pt9-Baseline
Pt10-Baseline
Pt11-Baseline
RAM12.01-Sensitive
Mel_PDX1-V1
Mel_PDX2-V1
Mel_PDX3-V1
Mel_PDX4-V1
Mel_PDX6-V1
Pt1-DP1
Pt1-DP3
Pt2-DP1
Pt3-DP1
Pt4-DD-DP-1A
Pt4-DD-DP-2B
Pt4-DD-DP-3C
Pt5-DP1
Pt5-DP2
Pt6-DP2
Pt7-DP1
Pt9-DP2
Pt9-DD-DP1
Pt9-DD-DP2
Pt10-DP1
Pt11-DP1
Pt11-DP2
RAM12.01-DD-DP10
RAM12.01-DD-DP3
RAM12.01-DD-DP9
Mel_PDX27-R2
Mel_PDX27-R3
Mel_PDX1-R1
Mel_PDX1-R2
Mel_PDX2-R2
Mel_PDX3-R4
Mel_PDX3-R5
Mel_PDX4-R2
Mel_PDX4-R4
Mel_PDX4-R5
Mel_PDX6-R3
SBS25
SBS26
SBS30
SBS32
C
Signatures Cohorts
SBS2 Rapid autopsy
SBS3 Clinical
1.00 SBS4
SBS5 PDX
Proportion of mutational signatures
SBS6
SBS7a Genotype
0.75 SBS7b BRAF MUT
SBS9
SBS10b NRAS MUT
SBS10c
0.50 SBS25 Sensitive
SBS26 Acquired
SBS30 resistant
SBS38
0.25 SBS44
SBS86
SBS87
SBS92
0.00
SBS2 Signature score
SBS3 2.0
SBS4
SBS5 1.5
SBS6 1.0
SBS7a
Mutational signatures
SBS7b 0.5
SBS9
SBS10b
SBS10c
SBS25
SBS26
SBS30
SBS38
SBS44
SBS86
SBS87
SBS92
Pt2-Baseline
Pt3-Baseline
Pt4-Baseline
Pt5-Baseline
Pt6-Baseline
Pt7-Baseline
Pt9-Baseline
Mel_PDX27-V1
Mel_PDX1-V1
Mel_PDX2-V1
Mel_PDX4-V1
Mel_PDX6-V1
Pt1-DP1
Pt1-DP3
Pt2-DP1
Pt3-DP1
Pt4-DD-DP1A
Pt4-DD-DP2B
Pt4-DD-DP3C
Pt5-DP1
Pt5-DP2
Pt6-DP2
Pt7-DP1
Pt9-DD-DP1
Pt9-DD-DP2
Pt9-DP2
RAM1201-DD-DP3
RAM1201-DD-DP9
RAM1201-DD-DP10
Mel_PDX27-R1
Mel_PDX27-R2
Mel_PDX27-R3
Mel_PDX1-R1
Mel_PDX1-R2
Mel_PDX2-R2
Mel_PDX3-R4
Mel_PDX4-R2
Mel_PDX4-R4
Mel_PDX4-R5
Mel_PDX6-R3
Figure 3. (Continued) B, SBS signatures of chromothriptic genomes of MAPKi-sensitive/naive (left, n = 16) versus MAPKi-resistant (right, n = 31)
melanoma tumors. Excluded from analysis, acquired resistant tumors without patient-matched sensitive tumors and tumors without chromothripsis.
C, Top, SBS signatures of ecDNAs + CGRs of MAPKi-sensitive/naive (left, n = 12) versus MAPKi-resistant (right, n = 28) melanoma tumors. Bottom, SBS
signature enrichment scores are shown by both heat scale and dot size. Increasing dot sizes indicate increasing enrichment of indicated signatures
within ecDNAs + CGRs compared with regions devoid of these events (score > 1). Enrichment score = 1 is considered the cutoff; only scores >1 and <1
are shown. Excluded from analysis are acquired-resistant tumors without patient-matched sensitive tumors and tumors without ecDNAs and CGRs.
(continued on next page)
D E 100 100
80 80
HRAS
BRAF
100 60
% ecDNA + CGR breakpoints
Sensitive 40
Acquired 40
75 resistance 20 20
0 0
*** 100 100
50 80
80
NRAS
EGFR
60 60
25
40 40
20 20
0
0 0
NHEJ Alt-NHEJ HRR NHEJ Alt-NHEJ HRR NHEJ Alt-NHEJ HRR
Figure 3. (Continued) D, Breakpoint–junctional sequence analysis of ecDNAs + CGRs inferring indicated DNA DSB repair processes: NHEJ, alternative
NHEJ (alt-NHEJ), and HRR. Amplicons from all three cohorts of tumors combined for analysis (sensitive, n = 17; resistant, n = 41). Homologous sequences
ecDNA/CGR amplicons. CGR + ecDNA-associated TMB kataegis in ecDNAs (33). We also observed positive enrich-
trended higher in acquired-resistant (mean, 130 SNVs/Mb) ment scores of SBS3 (defective HRR) in 47% (9 of 19) of
compared with MAPKi-sensitive/naive (mean, 80 SNVs/Mb) ecDNA+ acquired-resistant tumors compared with 17% (4 of
tumors (Wilcoxon rank sum test, P = 0.06). Enrichment of 24) of CGR+ acquired-resistant tumors (Wilcoxon rank sum
SBS3 (defective HRR) was more recurrent in the ecDNAs/ test, P = 0.07; Supplementary Fig. S2F). In short, ecDNA
CGRs of resistant tumors (11 of 28) compared with patient- and CGR amplicons, whether in MAPKi-sensitive/naive or
matched sensitive tumors (1 of 12), although this difference acquired-resistant tumors, harbor SBS patterns sugges-
did not reach statistical significance (Wilcoxon rank sum tive of specific defects or deficiencies in HRR. In acquired
test, P = 0.08; Fig. 3C). Moreover, we derived mutational MAPKi resistance, APOBEC activity likely contributes to
signature enrichment scores by calculating the normalized ecDNA mutagenesis and hence shapes the mutanome of
ratios of signature proportions within ecDNAs/CGRs and ecDNA amplicons. Further analysis might lead to insights
within regions devoid of these events (scores >1 defined as into ecDNA biogenesis and suppressive strategies.
positive enrichment; see Methods). We found that 25% (7 of
28) of acquired-resistant tumors displayed positive enrich- NHEJ Underlies the Formation of ecDNAs
ment scores for defective HRR signatures (SBS3) within and CGRs
ecDNAs and CGRs, whereas only 8% (1 of 12) of sensitive We analyzed the breakpoint–junctional sequences of CGRs
tumors displayed positive enrichment (Wilcoxon rank sum and ecDNAs to infer DSB repair processes underlying resistance-
test, P = 0.3; Fig. 3C). In either sensitive or resistant tumors, associated amplicons (Fig. 3D). Alternative end-joining refers
we observed positive enrichment scores for SBS signatures to mechanisms of DSB repair that may compensate for
reflective of APOBEC cytidine deaminase activity (SBS2; HRR- and NHEJ-based repairs and comprises single-strand
2 of 12 in sensitivity; 3 of 28 in resistance) and polymer- annealing (SSA), microhomology-mediated end-joining
ase epsilon exonuclease domain mutations (SBS10b; 3 of (MMEJ), and other end-joining pathways (34). SSA is indi-
12 in sensitivity; 3 of 28 in resistance). Positive enrich- cated in the breakpoint junctions by complementary repeat
ment scores were also noted with lower recurrence for sequences >25 nucleotides and MMEJ by shorter tracks of
SBS signatures of defective POLD1 proofreading (SBS10c), sequence homology (2–20 nucleotides). Moreover, replicative
defective MMR (SBS44), chemotherapy treatment (SBS86, processes, such as fork stalling and template switching as well
SBS87), and tobacco smoking (SBS82). Overall, 50% (6 of as MMEJ, can contribute to the generation of CGRs. Break-
12) of sensitive/naive and 32% (9 of 28) of acquired-resistant point junctions derived from replicative processes (e.g., repli-
tumors displayed enrichment of unique SBS signatures cative fork collapsing when it encounters a nick) are expected
within ecDNA + CGR sequences compared with uninvolved to have microhomologies, insertions, and relatively long tem-
genomic regions. Lastly, we addressed whether the enrich- plated insertions (35, 36). Indeed, signatures of replication
ment of unique SBS signatures differed within ecDNAs ver- processes and templated insertion, as well as that of NHEJ,
sus CGRs in acquired resistance (Supplementary Fig. S2E). were detected pan-cancer (12). Analysis of breakpoint–junc-
We observed positive enrichment scores of SBS2 (APOBEC tional sequences of all resistance- and sensitivity-associated
cytidine deaminase activity) in 26% (5 of 19) of ecDNA+ CGRs and ecDNAs inferred NHEJ as the main mechanism
acquired-resistant tumors compared with 4% (1 of 24) of of double-stranded DNA fragment joining or rearrange-
CGR+ acquired-resistant tumor (Wilcoxon rank sum test, ment (Fig. 3D; Supplementary Fig. S3A). A lower number of
P = 0.05; Supplementary Fig. S2F). This finding is consistent breakpoint–junctional sequences displayed short and long
with the recent discovery of the co-occurrence of APOBEC3 homologous sequences as well as insertions (>10 bp), which
suggested DSB repair by alternative NHEJ (alt-NHEJ) and we used next-generation DNA-PKi (VX984 and AZD7648)
HRR, respectively (Fig. 3D). Analysis of breakpoint–junctional and PARPi (olaparib) and corroborated their combinatorial
sequences of specifically ecDNAs and CGRs harboring MAPK- efficacy with MAPKi in parental cell lines (Supplementary
reactivation or MAPKi resistance–driver genes also revealed a Fig. S4C–S4E and Supplementary Fig. S5A–S5F). In M245,
similar pattern, with NHEJ dominating the landscape of DSB we further corroborated the pharmacologic findings with
repair mechanisms (Fig. 3E). Of unknown significance or genetic studies. Using independent short hairpin RNAs
etiology, a higher incidence of double-stranded DNA ligation (shRNA) against two critical NHEJ genes (PRKDC, which
by HRR was detected in NRASMUT melanoma, either sensitive encodes DNA-PKCS, and DNA ligase IV, or LIG4), we showed
or acquired-resistant to MAPKi, compared with BRAFV600MUT that their protein knockdown, while not having a significant
melanoma (Supplementary Fig. S3B). In acquired-resistant effect on growth without MAPKi, strongly suppressed the
genomes, we also detected a higher incidence of double- frequency of DTPP emergence (Fig. 4P and Q).
stranded DNA ligation by NHEJ for CGRs compared with The acquired-resistant sublines used in the prior experi-
ecDNAs (Supplementary Fig. S3C). ments have been adapted chronically (i.e., months to years)
to MAPKi. Hence, we also tested whether a shorter delay (i.e.,
Inhibitors of DNA-PKCS and/or PARP1/2 Prevent weeks) in the cotreatment of parental cell lines with DNA-PKi
Acquired MAPKi Resistance in Melanoma Cell (and/or PARPi) would hinder the efficacy in preventing DTPP
Lines formation. In both cell lines (M229 BRAFV600MUT and M245
1.5 1.5
Relative % area
Relative % area
**
1.0 1.0
**
***
1.5
Relative % area
Relative % area
1.0
1.0 **
** ***
*** *** 0.5
0.5 ***
***
*** *** *** *** *** *** *** *** *** *** *** *** *** *** *** *** ***
0.0 0.0
(μmol/L) 0 2 4 6 8 10 0 2 4 8 10 12 0 2 4 8 10 12 (μmol/L) 0 2 4 6 8 10 0 .5 1 2 3 4 0 .5 1 2 3 4
0 0 + + + +
2 + 4 + 8 +6 + 0 + 2 + 4 6 8 10
1
NU7026 ABT888 NU7026 + ABT888 NU7026 ABT888 NU7026 + ABT888
BRAFi + MEKi BRAFi + MEKi
Figure 4. DNA-PKi and/or PARPi cotreatment prevents acquired MAPKi resistance in human melanoma cell lines. A–O, Long-term clonogenic growth
of isogenic parental and acquired MAPKi-resistant BRAFV600MUT (A–H) or NRASQ61MUT (I–O) human melanoma cell lines showing acquired resistant colonies
to BRAFi + MEKi (PLX4032 at 0.5 μmol/L, AZD6244 at 0.5 μmol/L in A and G; PLX4032 at 0.25 μmol/L, AZD6244 at 0.25 μmol/L in D; PLX4032 at
1 μmol/L, AZD6244 at 1 μmol/L in B, C, E, F, and H) or MEKi (trametinib at 0.005 μmol/L in I and 0.01 μmol/L in J and N; trametinib at 0.1 μmol/L in K, L, M,
and O) and their suppression by indicated cotreatments with DNA-PKi (NU7026) and/or PARPi (ABT888) at indicated concentrations. Top, representative
cultures; bottom, quantifications over n = 4 fields (mean ± SD). Data representative of 2 to 4 independent repeats. Seeding densities (cells/well in 6-well
dishes) and culture durations (days): 5,000; 31 (A), 1,000; 17 (B), 5,000; 18 (C), and 40,000; 40 (D). (continued on following page)
BRAFi + MEKi or combinations with DNA-PKi, PARPi, or treatments with DNA-PKi and/or PARPi. During the earliest
DNA-PKi + PARPi (Fig. 5G). Consistent with BRAF HSRs as a window on MAPKi treatment, expansion of ecDNA- and
driver of acquired MAPKi resistance, we observed their induc- CGR-involved genomic spans may be critical for specific
tion by BRAFi + MEKi but suppression by combinatorial resistance-driving ecDNAs and CGRs to emerge later. We
Relative % area
1.0 * 1.0 *
*** ***
Relative % area
1.0
1.0 ** ***
** * ** *** *** *** *** *** ***
*** *** ***
0.5 0.5 ***
*** ***
*** *** *** *** *** *** ***
***
0.0 0.0
(μmol/L) 0 2 4 6 8 10 0 2 4 8 10 12 0 2 4 8 10 12 (μmol/L) 0 2 4 6 8 10 0 2 4 8 10 12 0 +2 4 8 10 12
2 + 4 + 8 +6 + 0 + 2 4 + 8 +6 + 0 +
1 1
NU7026 ABT888 NU7026 + ABT888 NU7026 ABT888 NU7026 + ABT888
BRAFi + MEKi BRAFi + MEKi
Figure 4. (Continued) Seeding densities (cells/well in 6-well dishes) and culture durations (days): 5,000; 14 (E), 5,000; 12 (F), 20,000; 30 (G), and
20,000; 21 (H). (continued on next page)
reasoned that DNA-PKi, which is generally more effective ecDNA and CGR spans detected in vehicle-treated genomes;
than PARPi in preventing acquired resistance (Fig. 4), should Supplementary Table S10). Consistent with our hypothesis,
suppress MAPKi-elicited ecDNA + CGR genomic spans. To DNA-PKi cotreatment with MAPKi blunted the expansion
test this hypothesis, we performed WGS on two parental of ecDNA + CGR genomic spans in both cell lines (Fig. 5H).
cell lines (M229 and M245) treated with vehicle, MAPKi, Compared with treatment with MEKi alone, M245 cells
or MAPKi + DNA-PKi for a short duration (10–11 days); treated with MEKi + DNA-PKi gained de novo ecDNAs (0.06
identified ecDNAs and CGRs; and calculated treatment- Mb in chr3) and lost CGRs (0.4 Mb in chr1 and chr2;
specific genomic spans of ecDNAs + CGRs (by filtering out Fig. 5H).
Relative % area
Relative % area
1.0 1.0 1.0
*** ** ** * ***
*** ***
0.5 *** ***
0.5 0.5
Relative % area
Relative % area
Figure 4. (Continued) Seeding densities (cells/well in 6-well dishes) and culture durations (days): 40,000; 30 (I), 50,000; 31 (J), 5,000; 17 (K), 5,000; 22
(L), 5,000; 16 (M), and 20,000; 16 (N). (continued on following page)
DNA-PKi Suppresses Acquired MAPKi Resistance the MAPKi regimen, we used MEKi (trametinib), KRASG12C
in KRASG12C Pancreatic Ductal Adenocarcinoma inhibitor (KRASG12Ci; AMG510 or MRTX849) + MEKi, or
and Non–Small Cell Lung Carcinoma Cell Lines type II RAF inhibitor (RAFi; BGB-283) + MEKi. As with mela-
As DNA-PKi plus MAPKi may constitute an effective noma cell lines, we first identified concentrations of DNA-
combination for BRAFV600MUT and NRASMUT melanoma, we PKi (NU7026) or PARPi (ABT888) that did not affect the
explored this combinatorial efficacy (± PARPi) in human pan- clonogenic growth of KRASG12C PDAC and NSCLC cell lines
creatic ductal adenocarcinoma (PDAC) and non–small cell without MAPKi (Supplementary Fig. S4F and S4G). We then
lung carcinoma (NSCLC) cell lines driven by KRASG12C. For assayed for acquired MAPKi-resistant growth in these cell
Cell viability
1.5 PRKDC sh1
PRKDC
1.0 PRKDC sh2
0.5 PRKDC sh3
TUBULIN PRKDC sh4
0.0
ABT888 (μmol/L) 0 5 10 15 0 1 2 3 4 5 6
Days
PRKDC
shVector sh1 sh2 sh3 sh4
14 days
DMSO
NU7026 (μmol/L) 0 5 10 15
ABT888 (μmol/L) 0 5 10 15
29 days
MEKi
Relative % area
Relative % area
Relative % area
*** 1.5
*** *** 1.0
r
1
2
3
4
r
1
2
3
4
5 +0 + 5 +
to
to
sh
sh
sh
sh
sh
sh
sh
sh
c
c
Ve
Ve
1 1 PRKDC PRKDC
sh
sh
NU7026 ABT888 NU7026 +
DMSO MEKi
ABT888
MEKi
Relative % area
Cell viability
N SO
T8 6
+ NU 88
T8 026
AB 02
LIG4
88
M
7
AB 7
U
D
shVector sh1 sh2
BRAFi + MEKi
14 days
DMSO
MEKi ***
NU7026 + 0.5
DMSO NU7026 ABT888 ABT888
Relative % area
Relative % area
0.0
1.0 *** 1.0
*** ***
N SO
T8 6
+ NU 88
T8 026
AB 02
0.5 0.5 88
M
7
AB 7
U
D
0.0 0.0
MEKi
r
1
2
r
1
2
to
to
sh
sh
sh
sh
c
c
Ve
Ve
LIG4 LIG4
sh
sh
DMSO DMSO
Figure 4. (Continued) Seeding density (cells/well in 6-well dishes) and culture duration (days): 5,000; 16 (O). Data representative of 2 to 4 independ-
ent repeats. P and Q, Western blot, MTT assay (top right), and clonogenic growth assay (bottom) of M245 cells transduced by lentivirus harboring shVec-
tor control or shRNAs of PRKDC (P) or LIG4 (Q). TUBULIN, loading control. Clonogenic growth with vehicle (14 days, 5,000 cells/well in 6-well dishes) or
MEKi (29 days, 50,000 cells/well in 6-well dishes) treatments. MEKi, trametinib at 0.01 μmol/L. Representative results of 2 independent experiments;
quantification of n = 4 fields (mean ± SD). R and S, As in A and J, respectively, except DNA-PKi and/or PARPi cotreatments were performed in M229 (R)
and M245 (S) at a later stage of MAPKi treatment (see time-point schema). Inhibitor concentrations for M229: BRAFi + MEKi (PLX4032 at 0.5 μmol/L,
AZD6244 at 0.5 μmol/L), NU7026 (8 μmol/L), ABT888 (4 μmol/L), and NU7026 + ABT888 (4 μmol/L + 4 μmol/L). Inhibitor concentrations for M245: MEKi
(trametinib at 0.01 μmol/L), NU7026 (5 μmol/L), ABT888 (1 μmol/L), and NU7026 + ABT888 (5 μmol/L + 1 μmol/L). Left, representative cultures; right,
quantifications over n = 4 fields (mean ± SD). Data representative of 2 independent repeats. (continued on next page)
DMSO DMSO
NU7026
NU7026
1.5 ABT888 1.5
ABT888
Relative % area
Relative % area
NU7026 + ABT888
NU7026 + ABT888
1.0 1.0
Figure 4. (Continued) T and U, As in A and J, respectively, except DNA-PKi and/or PARPi cotreatments were performed in M229 (T) and M245 (U)
cells at the same concentrations in R and S for the entire duration (day 1–28 in T; day 1–30 in U), the first half (day 1–14 in T; day 1–15 in U) or the second
half (day 14–28 in T; day 15–30 in U) of the total treatment course. Top, representative cultures; bottom, quantifications over n = 4 fields (mean ± SD).
Data representative of 2 independent repeats. P values (one-way ANOVA followed by the Tukey multiple comparisons test) comparing indicated cultures
versus MAPKi-only cultures (A–O and R–U) or shVector culture (P and Q): *, P < 0.05; **, P < 0.01; ***, P < 0.001.
lines, without or with DNA-PKi and/or PARPi (Fig. 6). Inter- resistance, without incurring overt signs of toxicities or
estingly, we observed that DNA-PKi effectively suppressed significant reductions in body weight. In NRASMUT mela-
acquired MAPKi resistance in two of two KRASG12C PDAC noma (Mel_PDX1; Fig. 7C), DNA-PKi treatment (10 mg/
cell lines (Fig. 6A–F) and one of two KRASG12C NSCLC cell kg/d) did not elicit tumor regression, and MEKi (trametinib,
lines (Fig. 6G–J). Moreover, PARPi was generally ineffective 3 mg/kg/d) elicited only transient tumor regression. In
as a combinatorial agent with MAPKi. Future studies should contrast, the combination of MEKi + DNA-PKi led to
explore the contributions of chromothripsis, ecDNAs, and minimally palpable tumors, without incurring toxicity. In
CGRs to acquired MAPKi resistance in human KRASG12C two additional NRASMUT melanoma PDXs (Mel_PDX2 and
PDAC and NSCLC. Mel_PDX4), we consistently observed superior efficacy of
MEKi + DNA-PKi over MEKi alone (Fig. 7D and E). In
DNA-PKi Forestalls Resistance In Vivo and Mel_PDX4, we initiated dosing with both DNA-PKi at 8
Reduces ecDNA and CGR Size mg/kg/day and MEKi at 3 mg/kg/day but stopped dosing
Given the broader resistance-preventive activity of DNA- DNA-PKi (in combination with MEKi) on day 67 (Fig. 7E).
PKi, we tested NU7026’s in vivo efficacy in five cutaneous With longer follow-up (Supplementary Fig. S6B), discon-
melanoma PDXs. In Mel_PDX16 (BRAFV600MUT) melanoma, tinuation of DNA-PKi dosing in this group of mice treated
using well-established tumors (∼500–700 mm3; Fig. 7A), initially with MEKi + DNA-PKi did not lead to tumor
BRAFi + MEKi (vemurafenib at 90 mg/kg/d, trametinib at relapse. Based on cell line findings (Fig. 4), we expected that
0.7 mg/kg/d) elicited only tumor growth inhibition, and delayed DNA-PKi cotreatment in vivo would also dimin-
DNA-PKi treatment (8 mg/kg/d) had no discernible effect ish its preventive mechanism of action. In Mel_PDX27
on tumor growth compared with vehicle-treated tumors. In (BRAFV600MUT) melanoma (Fig. 7B; Supplementary Fig. S6A),
contrast, the triplet of BRAFi + MEKi + DNA-PKi elicited we repeated BRAFi + MEKi (vemurafenib at 90 mg/kg/day,
tumor regression transiently for ∼14 days until the tumors trametinib at 0.7 mg/kg/day) treatment, and, at the time of
acquired resistance. There were no overt signs of toxici- disease progression (when the mean tumor volume returned
ties or significant reductions in body weight in any exper- to the mean pre-MAPKi volume of ∼500 mm3), we divided
imental group. In Mel_PDX27 (BRAFV600MUT) melanoma the tumors/mice into two groups. The first group continued
(Fig. 7B; Supplementary Fig. S6A), BRAFi + MEKi (vemu- on BRAFi + MEKi treatment at the same dosages, whereas
rafenib at 90 mg/kg/day, trametinib at 0.7 mg/kg/day) the second group received DNA-PKi treatment (8 mg/kg/d)
elicited transient tumor regression, whereas DNA-PKi treat- added on top of BRAFi + MEKi. Consistent with the cell
ment (8 mg/kg/day) did not elicit tumor regression com- line results (Fig. 4A–O), DNA-PKi cotreatment did not elicit
pared with vehicle-treated tumors. In contrast, the triplet of discernible tumor regression on tumors that had already
BRAFi + MEKi + DNA-PKi significantly forestalled acquired acquired MAPKi resistance in vivo (Fig. 7F).
20
**
cell
*
10
0
Withdrawal 0 8 26 0 8 26
(days) DDR4 DDR5
DDR5 – BRAFi – MEKi DDR5 – BRAFi – MEKi
Parental DDR5 + BRAFi + MEKi 8 days 26 days
HSR HSR HSR
BRAF/CEN7/DAPI
200
100
0
Withdrawal 0 7 25
(days) DDR
40
20
0
Withdrawal 0 14 26
(days) C3
100 ***
** *** 120 HSR
HSR HSR HSR
RAF1 HSR area/
80 ecDNA
RAF1/CEN3/DAPI
100
Frequency (%)
60 80
cell
40 60
40
20 20
0 0
Withdrawal 0 14 26 Withdrawal 0 14 26
(days) C3 (days) C3
Parental C5 + MEKi C5 – MEKi 9 days
HSR HSR 15 * HSR
120
NRAS HSR area/
NRAS/CEN1/DAPI
Non-HSR
100
Frequency (%)
10
80
cell
60
5 40
20
0 0
Withdrawal 0 9 Withdrawal 0 9
(days) C5 (days) C5
Figure 5. Resistance-driver ecDNAs and HSRs dynamically track with resistance, and DNA-PKi prevents the size expansion of ecDNAs and CGRs early
on MAPKi treatment. A–C, Metaphase DNA-FISH of paired parental and acquired resistant BRAFV600MUT (A and B) or NRASQ61MUT (C) cell lines without or
with drug withdrawal showing ecDNA or HSR amplicons harboring BRAF, RAF1, or NRAS. Left, representative images; right, quantifications per cell. Scale
bars, 15 μm. P values (unpaired two-tailed Student t test): *, P < 0.05; **, P < 0.01; ***, P < 0.001. (continued on next page)
***
***
***
***
***
0 0 0
2 0
5 2
5
1
2
5
2 0
5 2
5
1
2
5
2 0
5 2
5
1
2
5
0. 0.
0.
0. 0.
0.
0. 0.
0.
+
+
+
+
+
+
+
+
+
+
+
+
+
1
2
5
1
2
5
+
+
1
2
5
0.
0.
0.
BRAFi + MEKi (μmol/L) BRAFi + MEKi (μmol/L) BRAFi + MEKi (μmol/L)
Relative units (6 days)
Relative units (6 days)
200
50
***
***
***
***
***
0 0 0
2 0
5 2
5
1
2
5
2 0
5 2
5
1
2
5
2 0
5 2
5
1
2
5
0. 0.
0.
0. 0.
0.
0. 0.
0.
+
+
+
+
+
+
+
+
+
+
+
1
2
5
+
+
1
2
5
+
+
1
2
5
0.
0.
0.
BRAFi + MEKi (μmol/L) BRAFi + MEKi (μmol/L) BRAFi + MEKi (μmol/L)
1.0
Average total span of ecDNAs + CGRs (Mb)
100 100
80 80
60 60 0.5
*** ***
*** ***
40 40
20 20 0.0
0 0
MAPKi MAPKi + DNA-PKi
0
0.02
0.05
0.1
0.2
0.5
1
2
5
0
0.02
0.05
0.1
0.2
0.5
1
2
5
M245-d10
MEKi (μmol/L) MEKi (μmol/L) 0.4 ecDNA
CGR
Relative units (6 days)
120 150
0.3
100
80 100
0.2
60
40 50
0.1
***
20
***
***
0
0
0
0.02
0.05
0.1
0.2
0.5
1
2
5
0
0.02
0.05
0.1
0.2
0.5
1
2
5
0.0
MEKi (μmol/L) MEKi (μmol/L) MEKi MEKi + DNA-PKi
cell
0
D O
N SO
AB 26
+ NU 88
T8 026
S
70
T8
88
M
M
AB 7
U
D
BRAFi + MEKi
We then explored early on-treatment pharmacodynamic insights into the underlying genomic instability processes
markers that are associated with the superiority of that generate preexisting and therapy-elicited clonal diver-
MAPKi + DNA-PKi over MAPKi alone in forestalling or sification in advanced cutaneous melanoma. We identified
preventing acquired resistance. We posited that the com- chromothripsis as well as ecDNAs and CGRs as highly
bination of DNA-PKi with MAPKi (versus MAPKi alone) recurrent and pervasive genomic SVs in MAPKi-naive/sen-
would suppress the total size of ecDNAs and CGRs spe- sitive and acquired MAPKi-resistant melanoma in both the
cifically generated due to MAPKi treatment, agnostic of clinical setting (BRAFV600MUT melanoma, in which MAPKi
content genes, regulatory elements, and known or putative therapy is a standard-of-care therapy) and the experi-
roles in driving resistance. Using all five PDX models, we mental setting (NRASMUT melanoma, in which there is a
collected early on-treatment tumors and vehicle-treated lack of targeted therapy options). CGRs can derive from
tumors (Supplementary Fig. S6C and S6D). Expectedly, reintegration of ecDNAs or breakage–fusion–bridge (BFB)
p-ERK was suppressed strongly in both groups of tumors cycles (20, 43). We did not detect any BFB event in any of
treated with MAPKi or MAPKi + DNA-PKi (Supplementary our tumors, MAPKi-sensitive/naive or acquired-resistant,
Fig. S6E). Binding of the DNA-PK holoenzyme (DNA- favoring ecDNA reintegration as the main route of CGR
PKCS + Ku70 + Ku80) to DSBs elicits autophosphorylation generation in the setting of metastatic cutaneous mela-
of DNA-PKCS on serine 2056. We found that DNA-PKi noma. Consistently, the acute stress of MAPKi therapy
treatment, with or without MAPKi, suppressed nuclear favors ecDNAs, whereas stable or chronic stress favors
Figure 5. (Continued) D–F, Three-day (top) and six-day (bottom) MTT assay of indicated cell lines treated with graded concentrations of BRAFi + MEKi
(D and E) or MEKi (F). Cell viability was normalized to respective DMSO/vehicle groups. Inner brackets, comparisons between acquired resistant sublines
without and with drug withdrawal. Outer brackets, comparisons between acquired resistant sublines and their isogenic parental cell lines. P values (two-
way ANOVA test): *, P < 0.05; ***, P < 0.001. G, Representative images (left) and quantification (right) of metaphase DNA-FISH showing HSR harboring
BRAF in M249 cells treated with vehicle or BRAFi + MEKi with or without NU7026 and/or ABT888 for 31 days. Inhibitor concentrations: BRAFi + MEKi
(PLX4032 at 0.25 μmol/L, AZD6244 at 0.25 μmol/L), NU7026 (4 μmol/L), ABT888 (2 μmol/L), and NU7026 + ABT888 (4 μmol/L + 2 μmol/L). CEN,
centromere; DAPI, nuclear stain. Scale bars, 15 μm. P values (unpaired two-tailed Student t test): *, P < 0.05. H, Average total genomic spans of treatment-
specific ecDNAs + CGRs in M229 and M245 cell lines (background ecDNA + CGR spans detected in vehicle-treated cells were filtered). Inhibitor concen-
trations: BRAFi + MEKi (PLX4032 at 1 μmol/L, AZD6244 at 1 μmol/L for M229) or MEKi (trametinib at 0.02 μmol/L for M245), DNA-PKi (NU7026 at
8 μmol/L for both cell lines).
Relative % area
Relative % area
1.0 1.0 ** ** 1.0
*** *** *** ***
*** ***
0.5 *** *** 0.5 *** *** *** 0.5 *** ***
Relative % area
Figure 6. DNA-PKi cotreatment prevents acquired MAPKi resistance in KRASG12 human PDAC and NSCLC cell lines. A–J, Long-term clonogenic growth
of PDAC (MIAPaCa-2, XWR200; A–F) or NSCLC (H358, H2122; G–J) cell lines showing acquired resistant colonies to MEKi alone, type II RAFi + MEKi, or
KRASG12Ci + MEKi and their suppression by indicated cotreatments with DNA-PKi (NU7026) and/or PARPi (ABT888) at indicated concentrations. Top, rep-
resentative cultures; bottom, quantifications over n = 4 fields (mean ± SD). Data representative of 2 independent repeats. KRASi, KRAS inhibitor. Seeding
densities (cells/well in 6-well dishes) and culture durations: MIAPaCa-2 (10,000; 30 days for A–C) and XWR200 (150,000; 23 days for D, 30 days for E, 26
days for F). (continued on following page)
a recent study proposed that a specific BER defect may predis- in promoting the total segment sizes of ecDNAs and CGRs
pose micronuclei-associated or cytoplasmic chromosomes to generated early on MAPKi therapy. This finding is consist-
breakage, a key step toward chromothripsis (49). ent with prior literature supporting NHEJ as key to ecDNA
Translationally, we produced in vivo evidence supportive formation (28, 50). Selection for numeric expansion of spe-
of the well-studied role of DNA-PKCS in NHEJ as critical cific ecDNAs and CGRs is considered a direct mechanism
Relative % area
Relative % area
ABT888 0 5 10 15 ABT888 0 1 2 5
(μmol/L) (μmol/L)
NU7026 NU7026
(μmol/L) 0 1 2 5 (μmol/L) 0 2 5 10
ABT888 0 5 10 15 ABT888 0 1 2 5
(μmol/L) (μmol/L)
Relative % area
Relative % area
1.0 1.0
*** ***
*** *** *** ***
*** ***
0.5 0.5
***
***
0.0 *** *** *** *** ***
0.0
(μmol/L) 0 1 2 5 0 5 10 15 0 5 10 15 (μmol/L) 0 2 5 10 0 1 2 15 0 1 2 15
1 +2 + 5 + +
2 5 +0 +
1
NU7026 ABT888
NU7026 + NU7026 ABT888 NU7026 +
ABT888 ABT888
KRASi + MEKi MEKi
promoting tumor fitness in response to a given stressor. chemosensitization (37). The rationale is based on cata-
Therefore, reduction of the total ecDNA/CGR genomic strophic DSBs that would result in excessive DNA damage
spans early on DNA-PKi cotreatment should serve to reduce repair stress and hence the synergistic induction of death
the reservoir of ecDNAs/CGRs available for natural selec- in cancer cells, especially those overexpressing DNA-PKCS.
tion by MAPKi therapy. Here, we rationalize the combination of DNA-PKi and
DNA-PKCS, the target of DNA-PKi, subserves other less- MAPKi based on dual concepts. First, the rapid induc-
characterized cancer survival pathways (37). It is possible tion of genomic instability mechanisms, in particular
that another beneficial mechanism of action of DNA-PKi the generation of ecDNA and CGR amplicons, is criti-
coexists. DNA-PKi has been proposed in combination with cal for genomic diversification and perhaps epigenomic
agents that directly induce DNA damage, such as radi- reprogramming necessary for melanomas to adapt quickly
otherapy or chemotherapy, with the intent of radio- or to MAPKi therapy. In this rationale, DNA-PKi suppresses
Weight (g)
15
DNA-PKi
1,000 BRAFi + MEKi
10
BRAFi + MEKi + DNA-PKi
500 5
0 0
0 16 33 36 39 42 44 47 49 51 54 56 58 61 63 65 68 70 0 16 33 39 47 54 61 68
Days Days
Weight (g)
2,000 20 Vehicle
1,500 15 DNA-PKi
1,000 10 BRAFi + MEKi
Vehicle
Weight (g)
3,000 15
DNA-PKi
MEKi
2,000 10 MEKi + DNA-PKi
1,000 5
3.9E-6
0 0
0 17 34 37 41 43 45 48 50 52 55 57 59 62 64 66 0 17 24 41 48 55 62
Days Days
2,000 25
20
1,500
Weight (g)
15 Vehicle
1,000 DNA-PKi
10 MEKi
500 MEKi + DNA-PKi
3.2E-7 5
3.2E-7
0 0
0 21 43 48 50 52 55 57 59 62 64 66 69 71 73 76 78 80 0 21 43 50 57 64 71 78
Days Days
20
Vehicle
1,500
15 DNA-PKi
1,000 MEKi
10
P = 8.91E-15 MEKi + DNA-PKi
500 5
0 0
0 21 42 46 48 50 53 55 57 60 62 64 67 69 71 74 76 78 0 21 42 49 56 63 70 77
Days Days
Figure 7. DNA-PKi cotreatment with MAPKi reduces the size of ecDNAs and CGRs and prevents acquired resistance in vivo. A–E, Measurements of
tumor volumes (left) and body weights of mice (right) in two BRAFV600MUT (A and B) and three NRASMUT (C–E) cutaneous melanoma PDX models. Vehicle or
indicated treatments initiated on well-established tumors on days 33 (A), 48 (B), 34 (C), 43 (D), and 42 (E) after tumor fragment implantation, as marked
by upside-down red triangles. Dosage of inhibitors DNA-PKi (NU7026), BRAFi (vemurafenib), and MEKi (trametinib; mg/kg/day): DNA-PKi, 8; BRAFi, 90;
MEKi, 0.7 (A and B); DNA-PKi, 10; MEKi, 3 (C); DNA-PKi, 6; MEKi, 5 (D); and DNA-PKi, 8 (stopped on day 67 when in combination with MEKi, as marked by
an upside-down orange triangle); MEKi, 3 (E). One tumor per mouse; number of mice per experimental group (from top to bottom): 5, 5, 7, 7 (A); 4, 5, 8,
8 (B); 5, 5, 8, 5 (C); 3, 4, 8, 8 (D); and 5, 5, 6, 9 (E). Results are shown as mean ± SEM. P values, Student t test. Body weights are shown as average values.
(continued on following page)
1,200 30
Tumor volume (mm3)
1,000 25
Weight (g)
800 20
BRAFi + MEKi
600 15 BRAFi + MEKi + DNA-PKi
400 10
200 5
0 0
0 33 66 70 73 75 77 80 82 84 87 89 91 94 96 98 101 0 33 66 73 80 87 94 101
Days Days
0.2
0.2 0.15
0.10
0.1
0.1
0.05
Mel_PDX2 Mel_PDX4
ecDNA ecDNA
CGR CGR
0.09
ecDNAs + CGRs (Mb)
Average total span of
0.06
2
0.03
0.00 0
MEKi MEKi + DNA-PKi MEKi MEKi + DNA-PKi
Figure 7. (Continued) F, As in Fig. 5B (BRAFi + MEKi treatment group), except at the earliest time of acquired resistance (day 84, marked by an
upside-down orange triangle), tumors or mice were divided into two groups (n = 5 each). Group 1 continued on BRAFi + MEKi treatments. Group 2
received DNA-PKi (8 mg/kg/day) in combination with BRAFi + MEKi treatments. G, Average total spans of ecDNAs + CGRs specific to MEKi-treated
versus MEKi + DNA-PKi–treated PDXs (background ecDNAs + CGRs detected in vehicle-treated tumors were filtered) in Mel_PDX16 (BRAFi + MEKi–
treated, n = 3; BRAFi + MEKi + DNA-PKi–treated, n = 3), Mel_PDX27 (BRAFi + MEKi–treated, n = 3; BRAFi + MEKi + DNA-PKi (8 mg/kg/day)–treated, n = 3;
BRAFi + MEKi + DNA-PKi (10 mg/kg/day)–treated, n = 2), Mel_PDX1 (MEKi-treated, n = 2; MEKi + DNA-PKi–treated, n = 2), Mel_PDX2 (MEKi-treated,
n = 2; MEKi + DNA-PKi–treated, n = 2), and Mel_PDX4 (MEKi-treated, n = 2; MEKi + DNA-PKi–treated, n = 3). Tumors were collected for analysis on days 6
(Mel_PDX16), 7 (Mel_PDX27), 11 (Mel_PDX1), 9 (Mel_PDX2), and 8 (Mel_PDX4) after initiating treatments.
NHEJ, which is necessary for the efficient formation of (52). Excessive DNA SSBs can be converted into DNA DSBs,
ecDNAs and CGRs. Second, MAPKi is potentially an inducer engendering DNA damage repair stress and/or chromosome
of DNA damage and/or DNA damage repair deficiency. In breakage. The latter may be a pathway to chromothripsis
this context, MAPKi has been shown to induce DNA dam- and ecDNA/CGR generation (49).
age in early drug-tolerant persister subpopulations (42). A Our demonstration of ecDNAs and CGRs driving acquired
therapy-induced oxidative metabolic adaptation (51) has MAPKi resistance advances the concept that multiple resist-
been proposed to cause ROS-induced mutagenesis, which ance mechanisms, genetic and epigenetic, as well as direct
can be repaired by DNA single-stranded break (SSB) repair (drug–target or MAPK pathway reactivation) and indirect
processes such as BER and MMR. DNA-PKCS, in addition to (nondrug–target pathway activation), are simultaneously
DSB repair, can also bind to and is activated by DNA SSBs causal of clinically acquired resistance. Future work needs to
dissect this hybrid genomic–epigenomic model with ecDNAs enzymatic fragmentation of gDNA, the libraries were constructed
and CGRs at the center of therapeutic targeting efforts. Find- by end repairing and A-tailing the fragmented DNAs, ligation
ings here also advance the concepts that preventing, instead of adapters, and PCR amplification. After library construction,
of reversing, acquired resistant phenotypes may be more indexed libraries were quantified for equal molar pooling and
paired-end sequenced with a read length of 2 × 150 bp on the Illu-
impactful clinically and that targeting DNA-PKCS and NHEJ,
mina NovaSeq 6000 S4 platform.
and potentially MMEJ and HRR, lies at the center of this Whole-genome libraries were prepared for patient-matched nor-
approach in stabilizing cancer genomes during oncogene- mal tissues and tumors as follows: (i) 10 BRAFV600MUT clinical
targeted therapies. Finally, prevention of chromothripsis, patients (baseline tumors, n = 10; resistant tumors, n = 17), (ii) three
such as by reducing chromosome fusion and missegregation BRAFMUT RAM patients (sensitive tumor, n = 1; resistant tumors,
events and minimizing primary and micronuclei membrane n = 12), and (iii) six PDX models. PDX models included one BRAFMUT
ruptures, may expand our molecular armamentarium against PDX (vehicle-treated tumor, n = 1; resistant tumors n = 3) and five
targeted therapy resistance. NRASMUT PDXs (vehicle-treated tumors, n = 5; resistant tumors,
n = 9). For early on-treatment samples, we prepared whole-genome
libraries from (i) one human BRAFV600MUT (M229; DMSO, n = 1;
METHODS BRAFi + MEKi, n = 1; BRAFi + MEKi + DNA-PKi, n = 1) and one
Human Subjects NRASMUT (M245; DMSO, n = 1; MEKi, n = 1; MEKi + DNA-PKi,
n = 1) cutaneous melanoma cell line and (ii) two BRAFV600MUT (vehi-
Patient characteristics related to clinical tissues are presented cle, n = 6; BRAFi + MEKi, n = 6; BRAFi + MEKi + DNA-PKi, n = 9)
We carried out pathway enrichment analysis of genes within centromeric region of chr1; BRAF/CEN7 amplification probe (BRAF-
ecDNAs and/or CGRs using the Molecular Signatures Database CHR07-20-ORGR) targeting BRAF or centromeric region of chr7;
with pathways listed in Gene Ontology, Kyoto Encyclopedia of and RAF1/CEN3 amplification probe (RAF1-CHR03-20-ORGR) tar-
Genes and Genomes, Reactome, and Pathway Interaction Data- geting RAF1 or centromeric region of chr3, all from Empire Genom-
bases. For each tumor sample, we identified somatic SNVs using ics. The probes were mixed with the provided hybridization buffer
Strelka2 (61) with default parameters. Next, we estimated SBS sig- in a 1:4 ratio and applied onto the tissues or cells. FFPE samples
natures in regions within and without ecDNAs and CGRs using the were then denatured at 75°C in a slide moat for 7 minutes, whereas
nonnegative matrix factorization–based tool MutationalPatterns metaphase samples were denatured at 73°C for 2 minutes. We then
(62) with COSMIC SBS signatures V3.3 as reference. We carried performed hybridization overnight at 37°C in a humidified chamber.
out mutational signature analysis using SNVs in ecDNAs and/or The samples were then washed at 73°C with 0.3% Igepal/0.4× SSC for
CGR regions of (i) acquired MAPKi-resistant (mutations in patient- 2 minutes, followed by another 2-minute wash with 0.1% Igepal/2×
matched sensitive tumors subtracted from mutations in resistant SSC at room temperature. Finally, the tissue samples were stained
tumors) and (ii) MAPKi-sensitive genomes. To characterize whether with ProLong Diamond Antifade Mountant with DAPI (Invitrogen,
SBS mutational signatures are preferentially detected within (ver- P36966) and covered by coverslips. Images were acquired on a Leica
sus outside of) ecDNAs and/or CGRs amplicons, we calculated Confocal SP8-STED/FLIM/FCS microscope. For ecDNA quantifica-
the ratios of the proportions of signatures within ecDNA and/or tion, we counted the number of specific gene foci in each nucleus by
CGR sequences and the proportion of signatures outside ecDNAs ImageJ (version, 1.52a). For HSR quantification, we counted the area
and/or CGR sequences and defined these ratios as SBS mutational of gene foci larger than 2 μm2 to exclude ecDNA and non-HSR gene
signature enrichment score. We considered an enrichment score foci in each nucleus by ImageJ (version, 1.52a).
within these regions for each tumor sample. Ratios between TMBs Human PRKDC sh4: GGTCATGGATTCAAGAAAT
within and outside of chromothriptic regions were calculated. Human LIG4 sh1: TTCGACGCCACACCGTTTATT
Mutational signature analysis was carried out using SNVs in chro- Human LIG4 sh2: TATGTCAGTGGACTAATGGAT
mothriptic regions of (i) acquired MAPKi-resistant (mutations in
patient-matched sensitive tumors subtracted from mutations in
Cell Growth Assays
resistant tumors) and (ii) MAPKI-sensitive genomes. Mutational
(SBS, DBS, and ID) signatures for each sample were predicted using For clonogenic assay, cells were plated at indicated cell densities
nonnegative matrix factorization–based tool MutationalPatterns in 6-well plates and treated with inhibitor(s) the next day. Inhibi-
(62), and the extracted signatures were compared with the COSMIC tors and media were replenished every 2 days for the number
SBS, DBS, and ID signature database. of days indicated. Colonies were fixed in 4% paraformaldehyde,
followed by staining with 0.1% crystal violet. For the MTT assay,
Junctional Sequence Analysis of Amplicon Breakpoints cells were plated at 2,000 cells per well in 96-well plates (Fig. 4P
and Q); parental and acquired resistant sublines were seeded at
Sequences of CGRs and ecDNAs identified in all the genomes were 4,000 cells per well and treated with graded concentrations of
analyzed. Breakpoints of amplicons harboring key MAPKi-resistance MAPKi the next day (Fig. 5D–F); and media were replenished every
genes, BRAF, NRAS, HRAS, and EGFR, were extracted, and sequences 2 to 3 days. Methylthiazolyldiphenyl-tetrazolium bromide (MTT;
spanning these junctions were further analyzed. Homologous 100 μL) solution (0.5 mg/mL, Sigma, M5655) was added to each well
sequences and insertions for the breakpoint junctions were obtained and incubated at 37°C for 2 hours for MTT formazan formation.
from SvABA, and a possible mechanism for DNA DSB repair was Formazan was then dissolved in 100 μL DMSO, and the absorbance
inferred. First, HRR was identified with large homologous sequences
Statistical Methods
Lentivirus Production, Transduction, and shRNA Sequences
Statistical analysis for genomic data was conducted in R.4.02,
shRNAs for PRKDC and vector control (pGIPZ) were obtained
Python 3.8.0, and Python 2.7.17. Statistical analyses, described for
from Robert Damoiseaux, PhD (Molecular Screening Shared
specific experiments, for cell line– or PDX tumor–based assays were
Resource, UCLA); shRNAs for LIG4 were from Sigma. All shRNAs
performed using GraphPad Prism.
were packaged into lentiviral particles for infection. The lentiviral
viruses were generated by transfection of the constructs together
with pMD2.G, pRSV-Rev, and pMDLg/pRRE into HEK-293T cells Data Availability
using calcium phosphate. Fourteen hours after transfection, media The BAM files of WGS data are deposited in the European
were replaced with preheated fresh media. Virus particles were Genome-phenome Archive (https://www.ebi.ac.uk/ega/) with the
harvested 24 and 48 hours later and filtered by a 0.45 μm filter unit accession number EGAS00001006874. This study did not generate
(Millipore). Cells were transduced with recombinant lentivirus with custom codes. Any additional information regarding data reported in
10 μg/mL polybrene (Millipore, TR-1003-G) for 48 hours and then this article is available from the corresponding author upon request.
selected by puromycin (Sigma, P8833) for 1 week. shRNA targeting There are no restrictions on data availability.
sequences were as follows:
and Vedanta, personal fees (scientific advisory board member, hono- fact, this article is hereby marked “advertisement” in accordance with
raria, and stock) from 4C Biomed, Appia, Apricity, Arcus, High- 18 USC section 1734.
light, Compugen, ImaginAb, Kalthera/ImmPACT Bio, MapKure,
Merus, Rgenix, Lutris, PACT Pharma, Synthekine, Tango, Advaxis, Note
CytomX, Five Prime, RAPT, Isoplexis, and Kite/Gilead, and grants
Supplementary data for this article are available at Cancer Discovery
from Agilent and Bristol Myers Squibb outside the submitted work.
Online (http://cancerdiscovery.aacrjournals.org/).
S.J. Moschos reports grants from Merck during the conduct of the
study, as well as grants from Merck, Amgen, and Syndax Pharma and Received July 12, 2022; revised November 27, 2022; accepted
other support from Castle Biosciences outside the submitted work. January 23, 2023; published first January 26, 2023.
R.S. Lo reports grants from Merck, OncoSec, Pfizer, and Bristol
Myers Squibb outside the submitted work, as well as a patent for
U.S. Patent Application Serial No.: 63/268,028 issued. No disclo-
REFERENCES
sures were reported by the other authors.
1. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA,
et al. Inhibition of mutated, activated BRAF in metastatic melanoma.
Authors’ Contributions N Engl J Med 2010;363:809–19.
P. Dharanipragada: Data curation, formal analysis, investigation, 2. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J,
visualization, methodology, writing–original draft, writing–review et al. Combined BRAF and MEK inhibition in melanoma with BRAF
and editing. X. Zhang: Formal analysis, investigation, visualiza- V600 mutations. N Engl J Med 2012;367:1694–703.
tion, methodology, writing–review and editing. S. Liu: Data cura- 3. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, et al. Melano-
circular MYCN amplicon architecture in neuroblastoma. Nat Com- 40. Song C, Piva M, Sun L, Hong A, Moriceau G, Kong X, et al. Recurrent
mun 2020;11:5823. tumor cell-intrinsic and -extrinsic alterations during MAPKi-induced mel-
17. Hung KL, Yost KE, Xie L, Shi Q, Helmsauer K, Luebeck J, et al. ecDNA anoma regression and early adaptation. Cancer Discov 2017;7:1248–65.
hubs drive cooperative intermolecular oncogene expression. Nature 41. Harnor SJ, Brennan A, Cano C. Targeting DNA-dependent protein
2021;600:731–6. kinase for cancer therapy. ChemMedChem 2017;12:895–900.
18. Wu S, Bafna V, Mischel PS. Extrachromosomal DNA (ecDNA) in 42. Yang C, Tian C, Hoffman TE, Jacobsen NK, Spencer SL. Melanoma
cancer pathogenesis. Curr Opin Genet Dev 2021;66:78–82. subpopulations that rapidly escape MAPK pathway inhibition incur
19. Wu S, Turner KM, Nguyen N, Raviram R, Erb M, Santini J, et al. DNA damage and rely on stress signalling. Nat Commun 2021;12:1747.
Circular ecDNA promotes accessible chromatin and high oncogene 43. Cowell JK, Miller OJ. Occurrence and evolution of homogeneously
expression. Nature 2019;575:699–703. staining regions may be due to breakage-fusion-bridge cycles follow-
20. Shoshani O, Brunner SF, Yaeger R, Ly P, Nechemia-Arbely Y, Kim DH, ing telomere loss. Chromosoma 1983;88:216–21.
et al. Chromothripsis drives the evolution of gene amplification in 44. Koche RP, Rodriguez-Fos E, Helmsauer K, Burkert M, MacArthur IC,
cancer. Nature 2021;591:137–41. Maag J, et al. Extrachromosomal circular DNA drives oncogenic
21. Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, genome remodeling in neuroblastoma. Nat Genet 2020;52:29–34.
Goetz EM, et al. The genetic landscape of clinical resistance to RAF 45. Rosswog C, Bartenhagen C, Welte A, Kahlert Y, Hemstedt N, Lorenz W,
inhibition in metastatic melanoma. Cancer Discov 2014;4:94–109. et al. Chromothripsis followed by circular recombination drives onco-
22. Hong A, Moriceau G, Sun L, Lomeli S, Piva M, Damoiseaux R, et al. gene amplification in human cancer. Nat Genet 2021;53:1673–85.
Exploiting drug addiction mechanisms to select against MAPKi- 46. Ly P, Cleveland DW. Rebuilding chromosomes after catastrophe:
resistant melanoma. Cancer Discov 2018;8:74–93. emerging mechanisms of chromothripsis. Trends Cell Biol 2017;27:
23. Chadee DN, Xu D, Hung G, Andalibi A, Lim DJ, Luo Z, et al. Mixed- 917–30.
64. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. 66. Robinson MD, Oshlack A. A scaling normalization method for differ-
STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. ential expression analysis of RNA-seq data. Genome Biol 2010;11:R25.
65. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose 67. Shi H, Hong A, Kong X, Koya RC, Song C, Moriceau G, et al. A Novel
program for assigning sequence reads to genomic features. Bioinfor- AKT1 mutant amplifies an adaptive melanoma response to BRAF
matics 2014;30:923–30. inhibition. Cancer Discov 2014;4:69–79.