150 200 o 2 3 2 3it i 1 1* a t 150 a r 1Boehringer ...

1
In vivo PD Modulation and Efficacy for BI 1823911 Fabio Savarese 1 , Andreas Gollner 1 , Dorothea Rudolph 1 , Melanie Wurm 1 , Jesse Lipp 1 , Johannes Popow 1 , Marco H. Hofmann 1 , Heribert Arnhof 1 , Jörg Rinnenthal 1 , Francesca Trapani 1 , Michael Gmachl 1 , Daniel Gerlach 1 , Andreas Wernitznig 1 , Joachim Broeker 1 , Peter Ettmayer 1 , Andreas Mantoulidis 1 , Jason Phan 2 , Chris Smethurst 1 , Matthias Treu 1 , Alex G. Waterson 2 , Hengyu Lu 3 , Annette Machado 3 , Joseph Daniele 3 , Stephen W. Fesik 2 , Christopher P. Vellano 3 , Timothy P. Heffernan 3 , Joseph R. Marszalek 3 , Darryl B. McConnell 1 , Mark Petronczki 1 , Norbert Kraut 1 and Irene C. Waizenegger 1* In vitro and in vivo Characterization of BI 1823911 a Novel KRAS G12C Selective Small Molecule Inhibitor 1 Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria; 2 Vanderbilt University, Nashville, Tennessee, USA, 3 TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA Presented at the AACR Virtual Annual Meeting 2021 This study was funded by Boehringer Ingelheim. The authors were fully responsible for all content and editorial decisions, were involved at all stages of poster development and have approved the final version. *Corresponding author email address: [email protected] Key findings and conclusions Introduction Screening for Combination Partners with BI 1823911 In vivo Efficacy of BI 1823911 and SOS1 inhibitor KRAS G12C mutations are predominantly found in non-small cell lung cancer (NSCLC, 13%), in colorectal cancer (CRC, 3%), and with a lower prevalence of 1% in pancreatic ductal adenocarcinoma (PDAC). The amino acid exchange at position 12 from glycine to cysteine renders RAS insensitive to GAP-catalyzed hydrolysis but not to intrinsic hydrolysis and consequently, KRAS G12C is still dependent on GEF stimulation to achieve full activation. The active GTP–loaded form of KRAS G12C is favored and leads to activation of downstream signaling and proliferation. A number of recent publications has shown that targeting this mutant form of KRAS, using several covalent KRAS G12C inhibitors binding to the inactive GDP-KRAS G12C form, leads to anti-proliferative effects and induction of apoptosis in KRAS G12C mutant cancer cell lines, CDX and PDX models. Early clinical data for AMG- 510 and MRTX849 revealed a response rate of 35-45% in NSCLC and of 7-17% in CRC patients. Anti-proliferative Activity of BI 1823911 In vivo Efficacy of BI 1823911 on CDX and PDX Models In Vitro Pathway Modulation by BI 1823911 #1271 In Vitro Pathway Modulation by BI 1823911 and SOS1i Ten KRAS G12C and five non-KRAS G12C cell lines were treated with BI 1823911 in combination with listed combination partners, anti-proliferative activity was determined after five days of incubation using Cell-Titer-Glo®. Heat maps shows syngergy scores based on Loewe additivity. NCI-H2122 Dose [mg/kg] Schedule & Route TGI d19 [%] Tumor regressions [x/x] Natrosol -- qd po -- 0/8 BI 1823911 60 qd po 87 1/8 BI 1701963 (SOS1) 50 bid po 75 0/8 BI 1823911 BI 1701963 (SOS1) 20 50 qd bid po po 112 9/9 SW837 Dose [mg/kg] Schedule & Route TGI d27 [%] Tumor regressions [x/x] Natrosol -- qd po -- 0/8 BI 1823911 60 qd po 108 7/8 BI 1701963 (SOS1) 50 bid po 51 1/8 BI 1823911 BI 1701963 (SOS1) 20 50 qd bid po po 124 10/10 0 200 400 600 0 5 10 15 20 Median Tumor Volume [mm 3 ] Day Median Tumor Volume 25 26 27 28 29 30 31 0 5 10 15 20 Median Body Weight [gr] Day Median Body Weights 0 200 400 0 10 20 30 Tumor Volume [mm 3 ] Day Median Tumor Volumes 27 28 29 30 31 32 0 5 10 15 20 25 30 Median Body Weight [gr] Day Median Body Weights BI 1823911 has potent anti-proliferative activity in a panel of KRAS G12C mutant cancer cell lines with higher or similar potency compared to most advanced compounds in clinical development, AMG 510 and MRTX849. In a panel of KRAS G12C NSCLC cell lines, treatment with BI 1823911 results in downregulation of MAPK pathway-responsive genes, such as DUSP6. Likewise, strong and sustained inactivation of the MAPK pathway at the protein level using p-ERK as a pharmacodynamic (PD) biomarker was observed. NCI-H358 cell line-derived NSCLC and MIA PaCa-2 cell line-derived pancreatic cancer xenograft models were selected for extensive PK/PD/efficacy analyses in vivo. BI 1823911 was tested with a daily oral dose of 60 mg/kg in a panel of NSCLC or CRC CDX or PDX mouse models and showed comparable efficacy to 100 mg/kg AMG 510 and 100 mg/kg MRTX849, respectively. Preclinical and clinical data suggest that monotherapy with a KRAS G12C inhibitor will not be sufficient to achieve durable response. Combination therapy of a KRAS G12C inhibitor may therefore lead to enhanced anti-tumor efficacy and may address adaptive resistance mechanisms. A panel of KRAS G12C mutant cancer cell lines was tested with a large set of compounds in combination with BI 1823911 to identify synergistic anti-proliferative activity. Among other MAPK and PI3K pathway inhibitors, a SOS1:KRAS inhibitor was confirmed as promising combination partner. We report on results from in vitro and in vivo combination studies in NSCLC and CRC tumor models that show deep and durable responses upon combination of BI 1823911 with SOS1::KRAS inhibitor BI 1701963 providing a strong rationale for clinical investigation of this combination. BI 1823911 Oral Daily Dose [mg/kg] TGI d14 [%] Tumor regressions [x/8] 3 71 0 10 113 8 30 124 8 Pathway Output: DUSP6 mRNA Apoptosis: cleaved Caspase 3 Control 2h 60 mg/kg 2h Control 6h 60 mg/kg 6h Control 7h 60 mg/kg 7h Control 24h 60 mg/kg 24h 0 50 100 150 pERK1/2 day 1 relative pERK ratio Control 2h 60 mg/kg 2h Control 6h 60 mg/kg 6h Control 7h 60 mg/kg 7h Control 24h 60 mg/kg 24h 0 50 100 150 200 pERK1/2 day 5 relative pERK ratio Control 2h 60 mg/kg 2h Control 6h 60 mg/kg 6h Control 7h 60 mg/kg 7h Control 24h 60 mg/kg 24h 0 50 100 150 200 DUSP6 day 1 relative DUSP6/TBP ratio Control 2h 60 mg/kg 2h Control 6h 60 mg/kg 6h Control 7h 60 mg/kg 7h Control 24h 60 mg/kg 24h 0 50 100 150 DUSP6 day 5 relative DUSP6/TBP ratio Control 4h 3 mg/kg 4h 10 mg/kg 4h 30 mg/kg 4h 0 50 100 150 relative DUSP6/TBP ratio DUSP6 Control 4h 3 mg/kg 4h 10 mg/kg 4h 30 mg/kg 4h 0 1 2 3 4 cC3% marked area cC3 BI 1823911, AMG 510 and MRTX849 were dose titrated in a panel of either KRAS G12C mutant or non-KRAS G12C mutant colon cancer, NSCLC or PDAC cell lines. After five days of incubation cell viability was determined using Cell-Titer-Glo® as read-out (3 days - NCI-H441, NCI-H2122). Mean IC 50 of ≥2 biological replicates are shown. Figure 1: BI 1823911 is a potent and selective KRAS G12C inhibitor with higher anti- proliferative activity compared to AMG 510 or MRTX849 Figure 2: Dose- and time-dependent binding of BI 1823911 to KRAS shown by band shift, which leads to concomittant MAPK pathway modulation, G1 cell cycle arrest and induction of apoptosis NCI-H358 KRAS G12C heterozygous NSCLC cell line was dose-titrated with BI 1823911 over indicated time points, cells were lysed and indicated proteins were immunoblotted with respective antibodies. Markers within MAPK pathway (KRAS, p-ERK/ERK, DUSP6) are shown in orange, G1 cell cycle markers (Cyclin D1, p27) in blue, apoptosis marker (cleaved PARP) in green, activation of RTKs suggested by increase of p-EGFR and HER2. Figure 3: Time-dependent p-ERK modulation across 4 different G12C mutant cancer cell lines NCI-H1792, NCI-H2030, NCI-H358 and SW1463 cells were treated with BI 1823911, AMG 510, MRTX849 and Trametinib for 2, 24 and 48h and pERK IC 50 values were derived using the phospho/total ERK1/2 kit (MSD). Except for the known outlier NCI-H1792, G12C-inhibition leads to durable repression of pERK over time. Left: Heatmap showing gene expression changes of MAPK pathway activation score (MPAS) genes (Wagle et al., NPJ Precis Oncol. 2018) at 24 hrs after BI 1823911 treatment (100 nM) versus untreated state for ten NSCLC KRAS G12C cancer cell lines. Upregulation is shown in red, downregulation in blue. Cell lines are ordered by extent of MAPK pathway modulation as estimated by MPAS (see bottom heatmap annotation). Right: Line graphs depicting MPAS signature values relative to untreated baseline state within the first 24 hours after BI 1823911 (100 nM) treatment. Ten NSCLC KRAS G12C cell lines are grouped into four classes (sensitive, medium sensitive, resistant, and inert) based on sensitivity to BI 1823911 factoring in IC 50 and maximum effect of the dose response curve. Average effects per group are shown in blue, effects of individual cell lines are shown as dashed lines labeled with the cell line name. Figure 4: Modulation of expression of MPAS genes under BI 1823911 treatment Two doses of BI 1823911 were used for the treatment of NCI-H358 tumor bearing mice. 30 mg/kg led mainly to tumor stasis whereas 60 mg/kg led to regressions. PD analysis of such tumors after 1 and 5 days of treatment demonstrate consistent and durable PD modulation (pERK/MSD or DUPS6 mRNA/quantigene). Dose dependent efficacy (3, 10 and 30 mg/kg), PD modulation (DUSP6 mRNA, quantigene, 4h post single dose) as well as disease marker modulation (cleaved Caspase 3, IHC, 4h post single dose) were observed in MIA PaCa-2 model. Figure 8: BI 1823911 shows good synergistic anti-proliferative activity in combination with PI3K/mTOR, EGFR inhibitors, and also SOS1 inhibitor KRAS G12C NSCLC and CRC cell lines, NCI-H2122 and SW837, respectively, were treated with indicated compounds and concentrations for 6 or 24h, cell lysates were analysed by immunoblotting with indicated antibodies. Markers within MAPK pathway (KRAS, p-ERK/ERK, DUSP6) are shown in orange, G1 cell cylce markers (Cyclin D1, p27) in blue, apoptosis marker (cleaved caspase 3, cleaved PARP) in green. Figure 10: Synergistic anti-tumor activity in NSCLC tumor model – NCI-H2122 Figure 11: Anti-tumor activity in CRC tumor model – SW837 NMRI nude nice (Taconic) were injected with NCI-H2122 or SW837 cells subcutaneously. Mice were randomized into groups 16 days (NCI-H2122) or 22 days (SW837) after cell injection. Tumor diameter were measured three times a week and bodyweight weight was measured daily. Mice were dosed orally with Natrosol as control (qd), BI 1823911 (60 mg/kg, qd), BI 1701963 (50 mg/kg, bid qd) or a combination of BI_1823911 and BI 1701963. BI 1823911 is a selective and potent KRAS G12C inhibitor In vivo efficacy at 60 mg/kg is comparable to 100 mg/kg AMG 510 or MRTX849 Combination of BI 1823911 with SOS1 inhibitor BI 1701963 shows increased, sustained PD modulation and deeper anti-tumor efficacy. Further supporting data on this combination are shown on poster #CT210 Figure 5: NSCLC KRAS G12C cell line derived xenograft – NCI-H358 Figure 6: Pancreatic cancer KRAS G12C cell line derived xenograft – MIA PaCa-2 Figure 7: Anti-tumor activity on KRAS G12C mutant NSCLC and CRC mouse xenograft models Left: BI 1823911 was dosed with 60 mg/kg daily (CRC PDX models were dosed with a schedule of 5 days on/2 days off). Change from baselinge calculated according to Hallin et al., Cancer Discovery, 2020, showing a spread of anti-tumor activity across models, available genetic markers did not reveal a correlation to response (data not shown). CDX - cell line derived xenograft; PDX – patient derived xenograft; MAF – mutant allele frequency Right: BI 1823911, AMG 510 and MRTX849 tested in three NSCLC PDX models showing comparable efficacy. Figure 9: Deepened PD modulation when BI 1823911 is combined with SOS1i BI 1701963 LU2529 LU2521 LU6405 KRAS G12C Vehicle BI 1823911, 60 mg/kg qd AMG 510, 100 mg/kg qd MRTX849, 100 mg/kg qd Vehicle BI 1823911, 60 mg/kg qd BI 1823911, 100 mg/kg qd AMG 510, 100 mg/kg qd MRTX849, 100 mg/kg qd Vehicle BI 1823911, 60 mg/kg qd AMG 510, 100 mg/kg qd MRTX849, 100 mg/kg qd BI 1823911 Oral Daily Dose [mg/kg] TGI d20 [%] Tumor regressions 30 89 2/8 60 166 5/5 0 100 200 300 400 500 600 700 0 5 10 15 Median Tumor Volume [mm 3 ] Day 0 50 100 150 200 250 300 350 400 450 0 10 20 Median Tumor Volume [mm 3 ] Day

Transcript of 150 200 o 2 3 2 3it i 1 1* a t 150 a r 1Boehringer ...

Page 1: 150 200 o 2 3 2 3it i 1 1* a t 150 a r 1Boehringer ...

In vivo PD Modulation and Efficacy for BI 1823911

Fabio Savarese1, Andreas Gollner1, Dorothea Rudolph1, Melanie Wurm1, Jesse Lipp1, Johannes Popow1, Marco H. Hofmann1, Heribert Arnhof1, Jörg Rinnenthal1, Francesca Trapani1, Michael Gmachl1, Daniel Gerlach1, Andreas Wernitznig1, Joachim Broeker1, Peter Ettmayer1, Andreas Mantoulidis1, Jason Phan2, Chris Smethurst1,

Matthias Treu1, Alex G. Waterson2, Hengyu Lu3, Annette Machado3, Joseph Daniele3, Stephen W. Fesik2, Christopher P. Vellano3, Timothy P. Heffernan3, Joseph R. Marszalek3, Darryl B. McConnell1, Mark Petronczki1, Norbert Kraut1 and Irene C. Waizenegger1*

In vitro and in vivo Characterization of BI 1823911 – a Novel KRASG12C Selective Small Molecule Inhibitor

1Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria; 2Vanderbilt University, Nashville, Tennessee, USA, 3TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Presented at the AACR Virtual Annual Meeting 2021

This study was funded by Boehringer Ingelheim. The authors were fully responsible for all content and editorial decisions, were involved at all stages of poster development and have approved the final version.

*Corresponding author email address: [email protected]

Key findings and conclusions

Introduction Screening for Combination Partners with BI 1823911 In vivo Efficacy of BI 1823911 and SOS1 inhibitor

• KRASG12C mutations are predominantly found in non-small

cell lung cancer (NSCLC, 13%), in colorectal cancer (CRC, 3%),

and with a lower prevalence of 1% in pancreatic ductal

adenocarcinoma (PDAC). The amino acid exchange at

position 12 from glycine to cysteine renders RAS insensitive

to GAP-catalyzed hydrolysis but not to intrinsic hydrolysis

and consequently, KRASG12C is still dependent on GEF

stimulation to achieve full activation. The active GTP–loaded

form of KRASG12C is favored and leads to activation of

downstream signaling and proliferation. A number of recent

publications has shown that targeting this mutant form of

KRAS, using several covalent KRASG12C inhibitors binding to

the inactive GDP-KRASG12C form, leads to anti-proliferative

effects and induction of apoptosis in KRASG12C mutant cancer

cell lines, CDX and PDX models. Early clinical data for AMG-

510 and MRTX849 revealed a response rate of 35-45% in

NSCLC and of 7-17% in CRC patients.

Anti-proliferative Activity of BI 1823911

In vivo Efficacy of BI 1823911 on CDX and PDX Models

In Vitro Pathway Modulation by BI 1823911

#1271

In Vitro Pathway Modulation by BI 1823911 and SOS1i

Ten KRASG12C and five non-KRASG12C cell lines were treated with BI 1823911 in combination with listed combinationpartners, anti-proliferative activity was determined after five days of incubation using Cell-Titer-Glo®. Heat mapsshows syngergy scores based on Loewe additivity.

NCI-H2122Dose

[mg/kg]Schedule &

RouteTGI d19

[%]Tumor regressions

[x/x]

Natrosol -- qd po -- 0/8

BI 1823911 60 qd po 87 1/8

BI 1701963 (SOS1) 50 bid po 75 0/8

BI 1823911BI 1701963 (SOS1)

2050

qdbid

popo

112 9/9

SW837Dose

[mg/kg]Schedule &

RouteTGI d27

[%]Tumor regressions

[x/x]

Natrosol -- qd po -- 0/8

BI 1823911 60 qd po 108 7/8

BI 1701963 (SOS1) 50 bid po 51 1/8

BI 1823911BI 1701963 (SOS1)

2050

qdbid

popo

124 10/10

0

200

400

600

0 5 10 15 20

Me

dia

n T

um

or

Vo

lum

e [

mm

3 ]

Day

Median Tumor Volume

25

26

27

28

29

30

31

0 5 10 15 20

Me

dia

n B

od

y W

eig

ht

[gr]

Day

Median Body Weights

0

200

400

0 10 20 30

Tum

or

Vo

lum

e [

mm

3 ]

Day

Median Tumor Volumes

27

28

29

30

31

32

0 5 10 15 20 25 30

Me

dia

n B

od

y W

eig

ht

[gr]

Day

Median Body Weights

• BI 1823911 has potent anti-proliferative activity in a panel of KRASG12C mutant cancer cell lines with higher or

similar potency compared to most advanced compounds in clinical development, AMG 510 and MRTX849.

• In a panel of KRASG12C NSCLC cell lines, treatment with BI 1823911 results in downregulation of MAPK

pathway-responsive genes, such as DUSP6. Likewise, strong and sustained inactivation of the MAPK pathway

at the protein level using p-ERK as a pharmacodynamic (PD) biomarker was observed.

• NCI-H358 cell line-derived NSCLC and MIA PaCa-2 cell line-derived pancreatic cancer xenograft models were

selected for extensive PK/PD/efficacy analyses in vivo.

• BI 1823911 was tested with a daily oral dose of 60 mg/kg in a panel of NSCLC or CRC CDX or PDX mouse

models and showed comparable efficacy to 100 mg/kg AMG 510 and 100 mg/kg MRTX849, respectively.

• Preclinical and clinical data suggest that monotherapy with a KRASG12C inhibitor will not be sufficient to achieve

durable response. Combination therapy of a KRASG12C inhibitor may therefore lead to enhanced anti-tumor

efficacy and may address adaptive resistance mechanisms. A panel of KRASG12C mutant cancer cell lines was

tested with a large set of compounds in combination with BI 1823911 to identify synergistic anti-proliferative

activity. Among other MAPK and PI3K pathway inhibitors, a SOS1:KRAS inhibitor was confirmed as promising

combination partner. We report on results from in vitro and in vivo combination studies in NSCLC and CRC

tumor models that show deep and durable responses upon combination of BI 1823911 with SOS1::KRAS

inhibitor BI 1701963 providing a strong rationale for clinical investigation of this combination.

BI 1823911

Oral Daily Dose

[mg/kg]

TGI d14

[%]

Tumor regressions

[x/8]

3 71 0

10 113 8

30 124 8

Pathway Output: DUSP6 mRNA Apoptosis: cleaved Caspase 3

Contr

ol 2h

60 m

g/kg 2

h

Contr

ol 6h

60 m

g/kg 6

h

Contr

ol 7h

60 m

g/kg 7

h

Contr

ol 24h

60 m

g/kg 2

4h

0

50

100

150

pERK1/2 day 1

rela

tive

pE

RK

rati

o

Contr

ol 2h

60 m

g/kg 2

h

Contr

ol 6h

60 m

g/kg 6

h

Contr

ol 7h

60 m

g/kg 7

h

Contr

ol 24h

60 m

g/kg 2

4h

0

50

100

150

200

pERK1/2 day 5

rela

tive

pE

RK

rati

o

Contr

ol 2h

60 m

g/kg 2

h

Contr

ol 6h

60 m

g/kg 6

h

Contr

ol 7h

60 m

g/kg 7

h

Contr

ol 24h

60 m

g/kg 2

4h

0

50

100

150

200

DUSP6 day 1

rela

tive

DU

SP

6/T

BP

rati

o

Contr

ol 2h

60 m

g/kg 2

h

Contr

ol 6h

60 m

g/kg 6

h

Contr

ol 7h

60 m

g/kg 7

h

Contr

ol 24h

60 m

g/kg 2

4h

0

50

100

150

DUSP6 day 5

rela

tive

DU

SP

6/T

BP

rati

o

Contr

ol 4h

3 m

g/kg 4

h

10 m

g/kg 4

h

30 m

g/kg 4

h

0

50

100

150

rela

tive

DU

SP

6/T

BP

rati

o

DUSP6

Contr

ol 4h

3 m

g/kg 4

h

10 m

g/kg 4

h

30 m

g/kg 4

h

0

1

2

3

4

cC

3%

mark

ed

are

a

cC3

BI 1823911, AMG 510 and MRTX849 were dose titrated in a panel of either KRASG12C mutant or non-KRASG12C

mutant colon cancer, NSCLC or PDAC cell lines. After five days of incubation cell viability was determined usingCell-Titer-Glo® as read-out (3 days - NCI-H441, NCI-H2122). Mean IC50 of ≥2 biological replicates are shown.

Figure 1: BI 1823911 is a potent and selective KRASG12C inhibitor with higher anti-proliferative activity compared to AMG 510 or MRTX849

Figure 2: Dose- and time-dependent binding of BI 1823911 to KRAS shown by band shift,which leads to concomittant MAPK pathway modulation, G1 cell cycle arrest and inductionof apoptosis

NCI-H358 KRASG12C heterozygous NSCLC cell line was dose-titrated with BI 1823911 over indicated time points,cells were lysed and indicated proteins were immunoblotted with respective antibodies. Markers within MAPKpathway (KRAS, p-ERK/ERK, DUSP6) are shown in orange, G1 cell cycle markers (Cyclin D1, p27) in blue,apoptosis marker (cleaved PARP) in green, activation of RTKs suggested by increase of p-EGFR and HER2.

Figure 3: Time-dependent p-ERK modulation across 4 different G12C mutant cancer cell lines

NCI-H1792, NCI-H2030, NCI-H358 and SW1463 cells were treated with BI 1823911, AMG 510, MRTX849 andTrametinib for 2, 24 and 48h and pERK IC50 values were derived using the phospho/total ERK1/2 kit (MSD).Except for the known outlier NCI-H1792, G12C-inhibition leads to durable repression of pERK over time.

Left: Heatmap showing gene expression changes of MAPK pathway activation score (MPAS) genes (Wagle etal., NPJ Precis Oncol. 2018) at 24 hrs after BI 1823911 treatment (100 nM) versus untreated state for tenNSCLC KRASG12C cancer cell lines. Upregulation is shown in red, downregulation in blue. Cell lines are orderedby extent of MAPK pathway modulation as estimated by MPAS (see bottom heatmap annotation).Right: Line graphs depicting MPAS signature values relative to untreated baseline state within the first 24hours after BI 1823911 (100 nM) treatment. Ten NSCLC KRAS G12C cell lines are grouped into four classes(sensitive, medium sensitive, resistant, and inert) based on sensitivity to BI 1823911 factoring in IC50 andmaximum effect of the dose response curve. Average effects per group are shown in blue, effects of individualcell lines are shown as dashed lines labeled with the cell line name.

Figure 4: Modulation of expression of MPAS genes under BI 1823911 treatment

Two doses of BI 1823911 were used for the treatment of NCI-H358 tumor bearing mice. 30 mg/kg led mainly totumor stasis whereas 60 mg/kg led to regressions. PD analysis of such tumors after 1 and 5 days of treatmentdemonstrate consistent and durable PD modulation (pERK/MSD or DUPS6 mRNA/quantigene).

Dose dependent efficacy (3, 10 and 30 mg/kg), PD modulation (DUSP6 mRNA, quantigene, 4h post single dose)as well as disease marker modulation (cleaved Caspase 3, IHC, 4h post single dose) were observed in MIA PaCa-2model.

Figure 8: BI 1823911 shows good synergistic anti-proliferative activity in combination withPI3K/mTOR, EGFR inhibitors, and also SOS1 inhibitor

KRASG12C NSCLC and CRC cell lines, NCI-H2122 and SW837, respectively, were treated with indicated compoundsand concentrations for 6 or 24h, cell lysates were analysed by immunoblotting with indicated antibodies. Markers within MAPK pathway (KRAS, p-ERK/ERK, DUSP6) are shown in orange, G1 cell cylce markers (Cyclin D1, p27) in blue, apoptosis marker (cleaved caspase 3, cleaved PARP) in green.

Figure 10: Synergistic anti-tumor activity in NSCLC tumor model – NCI-H2122

Figure 11: Anti-tumor activity in CRC tumor model – SW837

NMRI nude nice (Taconic) were injected with NCI-H2122 or SW837 cells subcutaneously. Mice were randomizedinto groups 16 days (NCI-H2122) or 22 days (SW837) after cell injection. Tumor diameter were measured threetimes a week and bodyweight weight was measured daily. Mice were dosed orally with Natrosol as control (qd),BI 1823911 (60 mg/kg, qd), BI 1701963 (50 mg/kg, bid qd) or a combination of BI_1823911 and BI 1701963.

• BI 1823911 is a selective and potent KRASG12C inhibitor

• In vivo efficacy at 60 mg/kg is comparable to 100 mg/kg AMG 510 or

MRTX849

• Combination of BI 1823911 with SOS1 inhibitor BI 1701963 shows increased,

sustained PD modulation and deeper anti-tumor efficacy. Further supporting

data on this combination are shown on poster #CT210

Figure 5: NSCLC KRASG12C cell line derived xenograft – NCI-H358

Figure 6: Pancreatic cancer KRASG12C cell line derived xenograft – MIA PaCa-2

Figure 7: Anti-tumor activity on KRASG12C mutant NSCLC and CRC mouse xenograft models

Left: BI 1823911 was dosed with 60 mg/kg daily (CRC PDX models were dosed with a schedule of 5 days on/2days off). Change from baselinge calculated according to Hallin et al., Cancer Discovery, 2020, showing a spreadof anti-tumor activity across models, available genetic markers did not reveal a correlation to response (data notshown). CDX - cell line derived xenograft; PDX – patient derived xenograft; MAF – mutant allele frequencyRight: BI 1823911, AMG 510 and MRTX849 tested in three NSCLC PDX models showing comparable efficacy.

Figure 9: Deepened PD modulation when BI 1823911 is combined with SOS1i BI 1701963

LU

25

29

LU

25

21

LU

64

05

KRASG12C Vehicle

BI 1823911, 60 mg/kg qd

AMG 510, 100 mg/kg qd

MRTX849, 100 mg/kg qd

VehicleBI 1823911, 60 mg/kg qdBI 1823911, 100 mg/kg qdAMG 510, 100 mg/kg qdMRTX849, 100 mg/kg qd

Vehicle

BI 1823911, 60 mg/kg qd

AMG 510, 100 mg/kg qd

MRTX849, 100 mg/kg qd

BI 1823911

Oral Daily Dose

[mg/kg]

TGI d20 [%] Tumor regressions

30 89 2/8

60 166 5/5

0

100

200

300

400

500

600

700

0 5 10 15

Med

ian

Tu

mo

r V

olu

me

[mm

3]

Day

0

50

100

150

200

250

300

350

400

450

0 10 20

Med

ian

Tu

mo

r V

olu

me [

mm

3]

Day