scispace - formally typeset
Search or ask a question
Posted ContentDOI

PTEN mutant NSCLC require ATM to suppress pro-apoptotic signalling and evade radiotherapy

TL;DR: PTEN tumors are addicted to ATM to detect and repair radiation induced DNA damage, which creates an exploitable bottleneck and low concentration of ATM inhibitor is able to synergise with IR to treat PTEN-deficient tumors in genetically well-defined IR resistant lung cancer models.
Abstract: Background: Despite advances in treatment of patients with non-small cell lung cancer, carriers of certain genetic alterations are prone to failure. One such factor frequently mutated, is the tumor suppressor PTEN. These tumors are supposed to be more resistant to radiation, chemo- and immunotherapy. Methods: Using CRISPR genome editing, we deleted PTEN in a human tracheal stem cell-like cell line as well generated primary murine NSCLC, proficient or deficient for Pten, in vivo. These models were used to verify the impact of PTEN loss in vitro and in vivo by immunohistochemical staining, western blot and RNA-Sequencing. Radiation sensitivity was assessed by colony formation and growth assays. To elucidate putative treatment options, identified via the molecular characterisation, PTEN pro- and deficient cells were treated with PI3K/mTOR/DNA-PK-inhibitor PI-103 or the ATM-inhibitors KU-60019 und AZD 1390. Changes in radiation sensitivity were assessed by colony-formation assay, FACS, western-blot, phospho-proteomic mass spectrometry and ex vivo lung slice cultures. Results: We demonstrate that loss of PTEN led to altered expression of transcriptional programs which directly regulate therapy resistance, resulting in establishment of radiation resistance. While PTEN-deficient tumor cells were not dependent on DNA PK for IR resistance nor activated ATR during IR, they showed a significant dependence for the DNA damage kinase ATM. Pharmacologic inhibition of ATM, via KU-60019 and AZD1390 at non-toxic doses, restored and even synergized with IR in PTEN-deficient human and murine NSCLC cells as well in a multicellular organotypic ex vivo tumor model. Conclusion: PTEN tumors are addicted to ATM to detect and repair radiation induced DNA damage. This creates an exploitable bottleneck. At least in cellulo and ex vivo we show that low concentration of ATM inhibitor is able to synergise with IR to treat PTEN-deficient tumors in genetically well-defined IR resistant lung cancer models.

Summary (1 min read)

Jump to:  and [Introduction]

Introduction

  • For cell 97 detachment Trypsin (Sigma Aldrich) was used.
  • Result files were filtered 395 for the included genes to create pathway specific visualizations.
  • The authors study suggests that tumors harbouring a loss of function mutation in PTEN can 930 be therapeutically addressed by irradiation in combination with ATM inhibition.
  • BioRxiv preprint 26 h after radiation with 8 Gy (dashed lines) or without radiation (continuous 1108 lines) (dead cells stained with trypan blue excluded from analysis).

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
PTEN mutant NSCLC require ATM to suppress pro-apoptotic signalling and evade
1
radiotherapy
2
3
Thomas Fischer
1,2,3
, Oliver Hartmann
2,3,*
, Michaela Reissland
2,3*
, Cristian Prieto-
4
Garcia
2,3
,
Kevin Klann
4
, Christina Schülein-Völk
5
, Bülent Polat
1,6
, Elena Gerhard-
5
Hartmann
6,7
, Mathias Rosenfeldt
6,7
,
Christian Münch
4
,
Michael Flentje
1
& Markus E.
6
Diefenbacher
2,3,
ŧ
7
8
9
1
Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
10
2
Protein Stability and Cancer Group, University of Würzburg, Department of Biochemistry and Molecular Biology,
11
Würzburg, Germany
12
3
Mildred Scheel Early Career Center, Würzburg, Germany
13
4
Protein Quality Control Group, Institute of Biochemistry II, Goethe University Frankfurt, Germany
14
5
Core Unit High-Content Microscopy, Biocenter, University of Würzburg, Germany
15
6
Comprehensive Cancer Centre Mainfranken, Würzburg, Germany
16
7
Institute for Pathology, University of Würzburg, Germany
17
18
*Equal contribution
19
ŧ
Corresponding author
20
21
Running Title: PTEN loss establishes radiotherapy resistance
22
23
Keywords: PTEN, ATM, IR, NSCLC, Radiotherapy, Cancer. DNA-PK, PI3K.
24
25
*Corresponding Author: Dr. Markus E. Diefenbacher. Lehrstuhl für Biochemie und
26
Molekularbiologie, Biozentrum, Am Hubland, Würzburg, 97074, Germany. Phone:
27
0931 31-88167; Fax: 0931 31-84113; E-mail: markus.diefenbacher@uni-
28
wuerzburg.de
29
30
Conflict of Interest: The authors declare no potential conflicts of interest.
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
.CC-BY 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 25, 2021. ; https://doi.org/10.1101/2021.07.24.453632doi: bioRxiv preprint

2
Abstract
47
48
Background
49
Despite advances in treatment of patients with non-small cell lung cancer, carriers of
50
certain genetic alterations are prone to failure. One such factor frequently mutated, is
51
the tumor suppressor PTEN. These tumors are supposed to be more resistant to
52
radiation, chemo- and immunotherapy.
53
54
Methods
55
Using CRISPR genome editing, we deleted PTEN in a human tracheal stem cell-like
56
cell line as well generated primary murine NSCLC, proficient or deficient for Pten, in
57
vivo. These models were used to verify the impact of PTEN loss in vitro and in vivo
58
by immunohistochemical staining, western blot and RNA-Sequencing. Radiation
59
sensitivity was assessed by colony formation and growth assays. To elucidate
60
putative treatment options, identified via the molecular characterisation, PTEN pro-
61
and deficient cells were treated with PI3K/mTOR/DNA-PK-inhibitor PI-103 or the
62
ATM-inhibitors KU-60019 und AZD 1390. Changes in radiation sensitivity were
63
assessed by colony-formation assay, FACS, western-blot, phospho-proteomic mass
64
spectrometry and ex vivo lung slice cultures.
65
66
Results
67
We demonstrate that loss of PTEN led to altered expression of transcriptional
68
programs which directly regulate therapy resistance, resulting in establishment of
69
radiation resistance. While PTEN-deficient tumor cells were not dependent on
70
DNA-PK for IR resistance nor activated ATR during IR, they showed a significant
71
dependence for the DNA damage kinase ATM. Pharmacologic inhibition of ATM, via
72
KU-60019 and AZD1390 at non-toxic doses, restored and even synergized with IR in
73
PTEN-deficient human and murine NSCLC cells as well in a multicellular organotypic
74
ex vivo tumor model.
75
76
Conclusion
77
PTEN tumors are addicted to ATM to detect and repair radiation induced DNA
78
damage. This creates an exploitable bottleneck. At least in cellulo and ex vivo we
79
show that low concentration of ATM inhibitor is able to synergise with IR to treat
80
PTEN-deficient tumors in genetically well-defined IR resistant lung cancer models.
81
82
83
84
85
86
87
88
89
90
.CC-BY 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 25, 2021. ; https://doi.org/10.1101/2021.07.24.453632doi: bioRxiv preprint

3
MATERIAL AND METHODS
91
92
Cell lines
93
Human BEAS-2B and HEK 293T cell lines was obtained from ATCC. Cells were
94
maintained in high-glucose DMEM (Sigma Aldrich) supplemented with 10% FBS
95
(Capricorn Scientific) and 1% Pen-Strep (Sigma Aldrich) 1% Glutamin (Sigma
96
Aldrich) at 37°C in 5% CO
2
on 10 cm dishes (Greiner Bioscience). For cell
97
detachment Trypsin (Sigma Aldrich) was used. All the cells were maintained in
98
culture for 15 passages as maximum to maintain cell identity. Cells were routinely
99
tested for mycoplasma via PCR. The reagents were dissolved in Dimethyl sulfoxide
100
(DMSO) in specified concentrations and added to the cells.
101
102
DNA transfection and infection
103
For DNA transfection, a mix of 2,5
μ
g plasmid DNA, 200
μ
l free medium and 5
μ
l PEI
104
was added into the 6-well dish medium (60% confluence), after 6 h incubation at
105
37°C the medium was changed to full supplemented medium. For DNA infection
106
retroviruses or lentiviruses (MOI=10) were added to the cell medium in the presence
107
of polybrene (5
μ
g/ml) and incubated at 37°C for 72 h. After incubation, infected cells
108
were selected with 2
μ
g/ml puromycin for 72 h or 20 µg/ml blasticidin for 10 days.
109
110
X-ray irradiation
111
Irradiation was performed at room temperature using a 6 MV Siemens linear
112
accelerator (Siemens, Concord, CA) at a dose rate of 9,5 Gy/min.
113
114
Colony forming
115
Dependent on the experiment cells were treated with two different protocols. With
116
the direct seeding protocol exponential growing cells were seeded to 10 cm dishes in
117
adequate amount to be 50-80% confluent next day. Cells were trypsinized, counted
118
and diluted. The dilution was dispensed into different vials and cells were irradiated
119
in suspension. Cells were directly seeded in adequate amounts into 10 cm plates to
120
obtain 100-400 colonies per dish. With the re-seeding protocol exponential growing
121
cells were seeded to 10 cm dishes in adequate amount to be 25-30% confluent next
122
day. The attached cells were treated with different substances or DMSO as a control.
123
3 h after treatment cells were irradiated with 0, 2, 3, 5, 7, 8 Gy and cultured for 24 h,
124
then cells were trypsinized, counted and re-seeded in adequate amounts into 10 cm
125
plates to obtain 100-400 colonies per dish. For both protocols KP and KPP cells
126
formed colonies after 6 days, BEAS-2B cells formed colonies after 10-11 days. Cells
127
were fixed with ice cold 25% acidic acid in methanol and stained with 0,5% crystal
128
violet. Colonies were count manually. Only colonies containing at least 50 cells were
129
scored. Surviving fractions were calculated by dividing the plating efficiency for the
130
specified dose divided by the plating efficiency of untreated cells. Radiation
131
treatment survival curves were fitted to the linear-quadratic model formula S= exp[-
132
α
D-
β
D
2
] (S=survival fraction; D=radiation dose;
α
and
β
fitted parameters). Curves
133
were fitted and blotted using a non-linear regression and analysed with
OriginPro
134
.CC-BY 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 25, 2021. ; https://doi.org/10.1101/2021.07.24.453632doi: bioRxiv preprint

4
(OriginPro, 2020, OriginLab Corporation, Northampton, MA, USA)
.
Mean survival
135
fractions at 2 Gy (SF2) and 4 Gy (SF4) were also obtained for each cell model and
136
each substance and used to calculate the radiation enhancement ratio at 2 Gy
137
(RER
2Gy
) and 4 Gy (RER
4Gy
) RER greater than 1 indicates enhancement of
138
radiosensitivity, RER below the value of 1 indicates a radio resistance effect.
139
Similarly, the radiation dose with 25% (D
25
) and 50% (D
50
) survival under different
140
conditions was calculated to obtain the dose enhancement ratio (DER
25
and DER
50
)
141
that is calculated by dividing D
25
without substance treatment by D
25
with substance
142
treatment, respectively D
50
without substance treatment by D
50
with substance. DER
143
greater than 1 indicates a radio sensitising effect, a DER below the value of 1
144
indicates a radio protecting effect. Plating efficiency was calculated by dividing the
145
number of colonies by the number of seeded cells. All calculated parameters are
146
listed in supplementary table 1 (Table S1)
147
148
Immunological methods
149
Cells were lysed in RIPA lysis buffer (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM
150
Na2EDTA, 1 mM EGTA, 1% NP-40 and 1% sodium deoxycholate), containing
151
proteinase inhibitor an phosphatase inhibitor (1/100; Bimake) by sonication using
152
Branson Sonifier 150 with a duty cycle at 25%, output control set on level 2 and the
153
timer set to 15 s. Protein concentration was quantified using Bradford assay (Biorad).
154
After mixing of Bradford reagent with 2
μ
l of sample, the photometer was used to
155
normalize the protein amounts with a previously performed bovine serum albumin
156
(BSA) standard curve. The quantified protein (40-80
μ
g) was heated in 4x sample
157
buffer (Thermo Fisher) and 10% sample reducing agent (Thermo Fisher) for 10 min
158
at 70°C and separated on 4-12% Bis/Tris-gels or 3-8% Tris/Acetat-Gels (Thermo
159
Fisher). After separation, protein was transferred to nitrocellulose membrane
160
(Thermo Fisher) in transfer buffer (Thermo Fisher) and then, incubated with blocking
161
buffer (5% low fat milk powder in TBS and 0.1% Tween20) for 60 min at RT. After
162
blocking, membranes were incubated with indicated Primary antibodies (1/1000
163
dilution in a buffer composed 5% low fat milk powder or 5% BSA in TBS and 0.1%
164
Tween20) over night at 4°C. Secondary HRP coupled antibody (Dako 1/1000 dilution
165
in a buffer composed 5% low fat milk powder or 5% BSA in TBS and 0.1% Tween20)
166
were incubated for 2 h at 4°C. Membranes were incubated for 5 min in luminol-
167
solution (250 mg luminol in 100 mM Tris pH 8,6) with 10% v/v cumarinic acid solution
168
(1,1 g cumarinic acid in DMS0 and 0,1% v/v H
2
O
2
)at RT, then membranes were
169
recorded with my ECL Imaging System. Analysis and quantifications of protein
170
expression was performed using Image Studio software (Licor Sciences, Lincoln,
171
NE, USA). Antibodies used for this publication are listed in supplemantary table 2
172
(Table S2)
173
174
AnnexinV/DAPI staining:
175
Cells growing as sub-confluent monolayers were pretreated with substance for 3 h
176
before radiation with 0 Gy and 8 Gy. The cells were kept under standard conditions
177
for normal cell growth. 24 h, 48 h, 72 h and 96 h after radiation cells were harvested
178
.CC-BY 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 25, 2021. ; https://doi.org/10.1101/2021.07.24.453632doi: bioRxiv preprint

5
with trysinization.
Non irradiated cells, treated with camptothecin 5 µM (CPT) were
179
harvested 48 h after treatment. Supernatant of cell culture dishes was pooled with
180
trypsinized cells and pelleted by centrifugation. Further preparation for FACS
181
measurement was following the protocol of the BioLegend APC Annexin V Apoptosis
182
Detection Kit and DNA-staining with DAPI Reagent (25
μ
g/mL) (Biolegend, San
183
Diego, CA, USA). 20 000 cells were assayed using a flow cytometer FACSCantoII
184
(Becton Dickinson, San Jose, CA, USA). The output data presented as two-
185
dimensional dot plot. Samples were analyzed using the Flowing software gating
186
events to avoid debris, then dividing events in four quadrants. Flowing software was
187
obtained from P. Terho (Turku Centre for Biotechnology, Turku, Finland). Column
188
histograms and statistics were analyzed with Graphpad PRISM 8 (GraphPad
189
Software, San Diego, California USA) and OriginPro. (OriginPro, 2020, OriginLab
190
Corporation, Northampton, MA, USA)
.
191
192
sgRNA design
193
sgRNAs were designed using the CRISPRtool (https://zlab.bio/guide-design-
194
resources).
195
196
AAV and lentivirus production and purification
197
Virus was packaged and synthetized in HEK 293T cells seeded in 15 cm-dishes.
198
For AAV production, cells (70% confluence) were transfected with the plasmid of
199
interest (10
μ
g), pHelper (15
μ
g) and pAAV-DJ or pAAV-2/8 (10
μ
g) using PEI
200
(70
μ
g). After 96 h, the cells and medium of 3 dishes were transferred to a 50 ml
201
Falcon tube together with 5 ml chloroform. Then, the mixture was shaken at 37°C for
202
60 min and NaCl (1 M) was added to the mixture. After NaCl is dissolved, the tubes
203
were centrifuged at 20 000 x g at 4°C for 15 min and the chloroform layer was
204
transferred to another Falcon tube together with 10% PEG8000. As soon as the
205
PEG800 is dissolved, the mixture was incubated at 4°C overnight and pelleted at 20
206
000 x g at 4°C for 15 min. The pellet was resuspended in PBS with MgCl2 and
207
0.001% pluronic F68, then, the virus was purified using Chloroform and stored at -
208
80C. AAV viruses were titrated using Coomassie staining and RT-PCR using AAV-
209
ITR sequence specific primers.
210
For Lentivirus production, HEK 293T cells (70% confluence) were transfected with
211
the plasmid of interest (15
μ
g), pPAX (10
μ
g) and pPMD2 (10
μ
g) using PEI (70
μ
g).
212
After 96h, the medium containing lentivirus was filtered and stored at -80°C.
213
214
In vivo experiments and histology
215
All in vivo experiments were approved by the Regierung Unterfranken and the ethics
216
committee under the license numbers 2532-2-362, 2532-2-367, 2532-2-374 and
217
2532-2-1003. The mouse strains used for this publication are listed. All animals are
218
housed in standard cages in pathogen
free facilities on a 12 h light/dark cycle
219
with ad libitum access to food and water. FELASA2014 guidelines were followed for
220
animal maintenance.
221
222
.CC-BY 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 25, 2021. ; https://doi.org/10.1101/2021.07.24.453632doi: bioRxiv preprint

References
More filters
Journal ArticleDOI
TL;DR: EdgeR as mentioned in this paper is a Bioconductor software package for examining differential expression of replicated count data, which uses an overdispersed Poisson model to account for both biological and technical variability and empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference.
Abstract: Summary: It is expected that emerging digital gene expression (DGE) technologies will overtake microarray technologies in the near future for many functional genomics applications. One of the fundamental data analysis tasks, especially for gene expression studies, involves determining whether there is evidence that counts for a transcript or exon are significantly different across experimental conditions. edgeR is a Bioconductor software package for examining differential expression of replicated count data. An overdispersed Poisson model is used to account for both biological and technical variability. Empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference. The methodology can be used even with the most minimal levels of replication, provided at least one phenotype or experimental condition is replicated. The software may have other applications beyond sequencing data, such as proteome peptide count data. Availability: The package is freely available under the LGPL licence from the Bioconductor web site (http://bioconductor.org).

29,413 citations

Journal ArticleDOI
TL;DR: The combined cancer death rate dropped continuously from 1991 to 2015 by a total of 26%, translating to approximately 2,378,600 fewer cancer deaths than would have been expected if death rates had remained at their peak.
Abstract: Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data, available through 2014, were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data, available through 2015, were collected by the National Center for Health Statistics. In 2018, 1,735,350 new cancer cases and 609,640 cancer deaths are projected to occur in the United States. Over the past decade of data, the cancer incidence rate (2005-2014) was stable in women and declined by approximately 2% annually in men, while the cancer death rate (2006-2015) declined by about 1.5% annually in both men and women. The combined cancer death rate dropped continuously from 1991 to 2015 by a total of 26%, translating to approximately 2,378,600 fewer cancer deaths than would have been expected if death rates had remained at their peak. Of the 10 leading causes of death, only cancer declined from 2014 to 2015. In 2015, the cancer death rate was 14% higher in non-Hispanic blacks (NHBs) than non-Hispanic whites (NHWs) overall (death rate ratio [DRR], 1.14; 95% confidence interval [95% CI], 1.13-1.15), but the racial disparity was much larger for individuals aged <65 years (DRR, 1.31; 95% CI, 1.29-1.32) compared with those aged ≥65 years (DRR, 1.07; 95% CI, 1.06-1.09) and varied substantially by state. For example, the cancer death rate was lower in NHBs than NHWs in Massachusetts for all ages and in New York for individuals aged ≥65 years, whereas for those aged <65 years, it was 3 times higher in NHBs in the District of Columbia (DRR, 2.89; 95% CI, 2.16-3.91) and about 50% higher in Wisconsin (DRR, 1.78; 95% CI, 1.56-2.02), Kansas (DRR, 1.51; 95% CI, 1.25-1.81), Louisiana (DRR, 1.49; 95% CI, 1.38-1.60), Illinois (DRR, 1.48; 95% CI, 1.39-1.57), and California (DRR, 1.45; 95% CI, 1.38-1.54). Larger racial inequalities in young and middle-aged adults probably partly reflect less access to high-quality health care. CA Cancer J Clin 2018;68:7-30. © 2018 American Cancer Society.

14,011 citations

Journal ArticleDOI
TL;DR: The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.
Abstract: The cBio Cancer Genomics Portal (http://cbioportal.org) is an open-access resource for interactive exploration of multidimensional cancer genomics data sets, currently providing access to data from more than 5,000 tumor samples from 20 cancer studies. The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.

11,912 citations

Journal ArticleDOI
TL;DR: A practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics, which makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries.
Abstract: The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.

10,947 citations

Journal ArticleDOI
TL;DR: The Pan-Cancer initiative compares the first 12 tumor types profiled by TCGA with a major opportunity to develop an integrated picture of commonalities, differences and emergent themes across tumor lineages.
Abstract: The Cancer Genome Atlas (TCGA) Research Network has profiled and analyzed large numbers of human tumors to discover molecular aberrations at the DNA, RNA, protein and epigenetic levels. The resulting rich data provide a major opportunity to develop an integrated picture of commonalities, differences and emergent themes across tumor lineages. The Pan-Cancer initiative compares the first 12 tumor types profiled by TCGA. Analysis of the molecular aberrations and their functional roles across tumor types will teach us how to extend therapies effective in one cancer type to others with a similar genomic profile.

5,294 citations

Related Papers (5)
Frequently Asked Questions (4)
Q1. What is the role of a phosphorylated PTEN in cancer cells?

ATM-mediated PTEN phosphorylation promotes PTEN nuclear 1315 translocation and autophagy in response to DNA-damaging agents in cancer cells. 

%Genetic Alteration Inframe Mutation (putative driver) Missense Mutation (putative driver) Missense Mutation (unknown significance) Truncating Mutation (putative driver) Truncating Mutation (unknown significance) Fusion Amplification (putative driver) Amplification (unknown significance) Deep Deletion (putative driver) Deep Deletion (unknown significance)mRNA High mRNA Low No alterationsProfiled in mRNA expression z-scores relative to diploidsamples (RNA Seq V2 RSEM)Yes NoPTENKRASHRASNRASEGFRBRAFPIK3CA38%26%8%16%13%23%75%Genetic 

Implementation of CRISPR/Cas9 Genome Editing to Generate 1318 Murine Lung Cancer Models That Depict the Mutational Landscape of Human 1319Disease. 

1252.CC-BY 4.0 International licenseavailable under a was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.