a5307 shp2 inhibition as the backbone of targeted therapy

1
-100 0 100 200 300 % Change in Tumor Volume 0 9 10 -50 -100 0 100 200 300 % Change in Tumor Volume 20 30 40 50 60 0 500 1000 1500 Days Post Implant Mean Tumor Volume (mm 3 ) Control RMC-4630 20 mg/kg po q2d Cobimetinib 2.5 mg/kg po qd Combination Dosing start 10 20 30 0 500 1000 1500 Days Post-implant Mean Tumor Volume (mm 3 ) Dosing start Control RMC-4630 10 mg/kg po qd AMG 510 10 mg/kg po qd Combination 15 25 35 45 0 250 500 750 1000 Days Post Implant Mean Tumor Volume (mm 3 ) Dosing start 0 20 40 60 0 200 400 600 Days on Study % Change in Tumor volume 5 10 15 20 25 30 35 0 500 1000 1500 2000 Days Post Implant Mean Tumor Volume (mm 3 ) Dosing start ✱✱ Control RMC-4550 20 mg/kg po q2d Cobimetinib 2.5 mg/kg po qd Combination 10 15 20 25 30 35 0 1000 2000 3000 Days Post Implant Mean Tumor Volume (mm 3 ) Dosing start * SHP2 inhibition as the backbone of targeted therapy combinations for the treatment of cancers driven by oncogenic mutations in the RAS pathway JAM Smith 1 , M Singh 1 , RJ Nichols 1 , ES Koltun 2 , YC Yang 1 , , D Wildes 1 , C Stahlhut 1 , D Lee 3 , CJ Schulze 1 , D Reyes 1 , A Marquez 1 , G Lee 1 , S Li 4 , C Marcireau 5 , L Debussche 5 , MA Goldsmith 1, 2, 3, 4 , ZP Wang 3 , AL Gill 2 , SM Kelsey 1, 2, 3, 4 Departments of Biology 1 , Chemistry 2 , Non-Clinical Development and Clinical Pharmacology 3 , and Clinical Development 4 , Revolution Medicines, Redwood City, California; Sanofi Oncology Research 5 , Vitry, France 1. RMC-4630 is a Potent, Selective and Orally Bioavailable Allosteric Inhibitor of SHP2 3. Single Agent and Combinatorial Benefits of RMC-4630: EGFR mut NSCLC Xenograft Models 2. Single Agent and Combinatorial Benefits of RMC-4630: KRAS G12C NSCLC 4. Single Agent and Combinatorial Benefits of RMC-4630: NF1 LOF and KRAS Amp Xenograft Models Conclusions RMC-4630 is a potent, selective and orally bioavailable SHP2 inhibitor which has shown preliminary signs of clinical activity in patients with NSCLC harboring KRAS mutations, particularly KRAS G12C In combination with mutant-selective KRAS G12C or EGFR inhibitors RMC-4630 can suppress oncoprotein-mediated signaling and adaptive resistance in preclinical models SHP2 inhibition alone, or in combination with targeted inhibition of another pathway node such as MEK, exhibited anti-tumor activity in mouse xenograft models with RAS pathway oncogenic drivers, such as NF1 LOF , KRAS Amp and KRAS G12D or KRAS G12V for which there are currently no mutant-selective inhibitors Translation of these preclinical findings into clinical benefit could position RMC-4630, an investigational therapeutic agent, as a backbone of targeted therapy combinations for patients bearing cancers with diverse oncogenic mutations in the RAS pathway SHP2 is a Frontier Target in Oncology Direct targeting of oncogenic mutations in the RAS pathway is a beneficial therapeutic strategy for patients with cancers with these mutations. Mutant-selective inhibitors offer a wide therapeutic window but are ultimately limited by emergence of drug resistance Escape from mutant-selective inhibitors frequently involves activation of wild-type signaling nodes, including hyperactivation of receptor tyrosine kinases (RTKs), that lead to robust re-activation of the RAS pathway SHP2 (PTPN11) is a phosphatase that functions as a convergent node downstream of multiple RTKs to regulate RAS activation. We have recently shown that single agent inhibition of SHP2 has anti-tumor activity in tumors harboring KRAS G12C both in the clinic and preclinical models 1, 2 In the context of adaptive resistance to mutant-selective inhibitors, SHP2 inhibition has the potential to suppress oncoprotein-mediated signaling and adaptive signaling driving escape from therapy For many RAS pathway oncogenic drivers, including KRAS G12D and KRAS G12V , NF1 LOF , KRAS Amp or BRAF Class3 , mutant-selective inhibitors are not currently available. Here, a combination strategy simultaneously targeting nodes both up- and down-stream of the oncoprotein (“oncoprotein clamping”) can drive tumor growth inhibition Here we show that SHP2 inhibitors have the potential to become the backbone of targeted therapy combinations across the spectrum of RAS-dependent tumors We have also shown that SHP2 inhibition, alone or in combination, can promote anti- tumor immunity in preclinical models via effects on the innate and adaptive immune systems 4, 5 . These effects may influence the overall profile of a SHP2 inhibitor Therapeutic Combinations for RAS Driven Tumors: Mutant-selective and RAS Pathway Node Inhibitors 5. Combination Benefit for SHP2 and MEK Inhibition in Other KRAS-Mutant Xenograft Models References 1. Nichols et al., Nat Cell Biol. 2018 20(9):1064-1073 2. Ou et al. AACR-IASCLC 2020 3. Clinical Trials.Gov: NCT03634982 4. Quintana et al., 2020 Cancer Research 10.1158/0008-5472 5. Shifrin et al., 2020 AACR A7744 P2837 6. Planchard et al., Annals Oncology 26: 2073–2078, 2015 Acknowledgements: Jingjing Jiang for expert input into design and execution of in vivo pharmacology models CRO support: WuXi AppTec (Suzhou, China); Champions Oncology (Maryland, USA); Charles River Laboratories/Oncotest Gmbh (Freiburg, Germany); Genendesign (Shanghai, China); Xentech (Evry, France); TD2 (Arizona, USA) Best Change in Tumor Burden from Baseline NSCLC with any KRAS Mutation for RMC-4630 Monotherapy 2, 3 Disease Control Rate: NSCLC KRAS mut 12/18 (67%) NSCLC KRAS G12C 6/8 (75%) Data presented for efficacy evaluable population (N = 18) defined as patients with baseline and at least one post-baseline scan or who died or had clinical progression prior to first post-baseline scan. Four patients are not represented in this figure: 2 patients had clinical progression prior to first scan, 1 patient did not have measurements for one of the target lesions but progressed due to new lesion, and 1 patient had missing tumor measurements in the database at the time of data extract. Confirmed PR # Unconfirmed PR Each animal represented as separate bar N = number of regressions >10% at end of study; 10 mice/group Clinical Preclinical NCI-H358 KRAS G12C NSCLC Xenograft All treatments were well-tolerated RMC-4630 (PO) PK/PD in vivo RMC-4630 anti-tumor activity in vivo NCI-H358 KRAS G12C NSCLC xenograft NCI-H358 KRAS G12C NSCLC xenograft Compound RMC-4630 RMC-4550 1 Tool Compound SHP2 biochemical potency (IC 50 , nM) 1.29 1.52 RAS pathway suppression (pERK IC 50 , nM) NCI-H358 KRAS G12C 20 28 Anti-proliferative activity (3D CTG IC 50 , nM) NCI-H358 KRAS G12C NCI-H1975 EGFR L858R/T790M 32 25 43 63 Selectivity - Phosphatase panel - Kinase panel > 3,000 > 3,000 > 3,000 > 3,000 SHP2 inhibitors - in vitro profile KRAS G12V Pancreatic KRAS G12V NSCLC NCI-H441 Capan-2 KRAS G12D Pancreatic KP-4 HPAC LUN #150, NSCLC CO-04-0004, CRC STO#332 WT KRAS Amp (copy number = 4) 10 5 KRAS Amp NF1 LOF Compound Parental EGFR L858R/T790M (IC 50 , nM) Transfected EGFR L858R/T790M/C797S (IC 50 , nM) RMC-4630 265 218 to 557 Osimertinib 8 30 to 2616 EGFR L858R/T790M/C797S in vitro EGFR L858R/T790 /MET Amp NSCLC PDX 6 Osimertinib-Sensitive Osimertinib-Resistant NCI-H1975 EGFR L858R/T790M NSCLC CDX Anti-proliferative activity (2D CTG) in parental NCI-H1975 or cells transfected with human EGFR L858R/T790M/C797S under six different promoters N= 12-15 mice/group One-way Anova: * p< 0.05; *** p < 0.0001 N = 3 mice/group; graphs show tumor volume data for individual mice, expressed as % of initial tumor volume at time of study start. N = 12 mice/group; graphs in B show tumor volume data for individual animals shown in A, expressed as % change in tumor volume from time of study start A B Each animal represented as separate bar N = number of regressions >10% at end of study; 10 mice/group Ordinary one-way ANOVA, * p< 0.01, ** p< 0.05, ***p<0.001 LUN #352 All treatments were well-tolerated All treatments were well-tolerated 0 10 20 30 0 200 400 600 800 Days on Study Mean Tumor Volume (mm 3 ) Control RMC-4630 10 mg/kg po qd Cobimetinib 2.5 mg/kg po qd Combination PDX models of tumors bearing NF1 mutations predicted to result in loss of function (LOF): deletions, insertions, premature stops, truncations Tumor growth inhibition in 62% of NF1 LOF PDX models (n=55) Tumor regressions in 25% (23/93) of responders (93/166 mice) N= 10 mice/group 40 80 120 160 0 500 1000 1500 2000 2500 Days Post Implant Mean Tumor Volume (mm 3 ) Control Osimertinib 5 mg/kg po qd RMC-4630 30 mg/kg po qd Combination Initial tumor volume 256 mm 3 Dosing stopped 40 80 120 160 0 400 800 Days Post Implant -100 Dosing stopped 40 80 120 160 0 400 800 -100 Days Post-implant Dosing stopped 40 80 0 100 Days Post Implant -100 -50 40 80 0 100 -50 -100 Days Post-implant % Change in Tumor Volume Osimertinib Combination 1 2 4 8 10 16 24 1 10 100 1000 10000 0 20 40 60 80 100 Time post-dose (hours) Unbound Plasma Concn (nM) % Inhibition pErk/tErk Relative to Control 10 mg/kg 30 mg/kg -9 -8 -7 -6 -5 0 25 50 75 100 Log Molar [RMC-4630] plasma, unbound % Inhibition of pERK/tERK relative to control EC 50 = 27 nM Time post dose 2h 8h 16h 24h Time post second dose (bid) 2h 8h 0 10 20 30 -100 0 100 200 300 Days on Study % Change in Tumor Volume Control RMC-4630 30 mg/kg po qd Dose/ schedule www.revolutionmedicines.com A5307 P1943 Questions? [email protected] 0 10 20 30 0 500 1000 1500 2000 2500 Days on Study Mean Tumor Volume (mm 3 ) * *** * # *** *** *** * *** 0 5 10 15 20 25 30 0 400 800 1200 1600 Days Post Implant Mean Tumor Volume (mm 3 ) Control RMC-4630 10 mg/kg po qd RMC-4630 30 mg/kg po qd Dosing start *** * 0 5 10 15 20 25 0 1000 2000 3000 Days Post Implant Mean Tumor Volume (mm 3 ) Control Osimertinib 25 mg/kg po 5d on/2d off RMC-4630 30 mg/kg po qd

Upload: others

Post on 26-Dec-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A5307 SHP2 inhibition as the backbone of targeted therapy

-100

0

100

200

300

% C

hang

e in

Tum

or V

olum

e

0 9 10

-50

-100

0

100

200

300

% C

hang

e in

Tum

or V

olum

e

20 30 40 50 600

500

1000

1500

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

ControlRMC-4630 20 mg/kg po q2dCobimetinib 2.5 mg/kg po qdCombination

Dosingstart

10 20 300

500

1000

1500

Days Post-implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

Dosingstart

ControlRMC-4630 10 mg/kg po qdAMG 510 10 mg/kg po qdCombination

15 25 35 450

250

500

750

1000

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

Dosingstart

0 20 40 60

0

200

400

600

Days on Study

% C

hang

e in

Tum

or v

olum

e

5 10 15 20 25 30 350

500

1000

1500

2000

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

Dosingstart

✱✱

ControlRMC-4550 20 mg/kg po q2dCobimetinib 2.5 mg/kg po qdCombination

10 15 20 25 30 350

1000

2000

3000

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

Dosingstart

*

SHP2 inhibition as the backbone of targeted therapy combinations for the treatment of cancers driven by oncogenic mutations in the RAS pathway

JAM Smith1, M Singh1, RJ Nichols1, ES Koltun2, YC Yang1, , D Wildes1, C Stahlhut1, D Lee3, CJ Schulze1, D Reyes1, A Marquez1, G Lee1, S Li4, C Marcireau5, L Debussche5, MA Goldsmith1, 2, 3, 4, ZP Wang3, AL Gill2, SM Kelsey1, 2, 3, 4

Departments of Biology1, Chemistry2, Non-Clinical Development and Clinical Pharmacology3, and Clinical Development4, Revolution Medicines, Redwood City, California; Sanofi Oncology Research5, Vitry, France

1. RMC-4630 is a Potent, Selective and Orally Bioavailable Allosteric Inhibitor of SHP2

3. Single Agent and Combinatorial Benefits of RMC-4630: EGFRmut NSCLC Xenograft Models

2. Single Agent and Combinatorial Benefits of RMC-4630: KRASG12C NSCLC

4. Single Agent and Combinatorial Benefits of RMC-4630: NF1LOF and KRASAmp Xenograft Models

Conclusions• RMC-4630 is a potent, selective and orally bioavailable SHP2 inhibitor which has

shown preliminary signs of clinical activity in patients with NSCLC harboring KRAS mutations, particularly KRASG12C

• In combination with mutant-selective KRASG12C or EGFR inhibitors RMC-4630 can suppress oncoprotein-mediated signaling and adaptive resistance in preclinical models

• SHP2 inhibition alone, or in combination with targeted inhibition of another pathway node such as MEK, exhibited anti-tumor activity in mouse xenograft models with RAS pathway oncogenic drivers, such as NF1LOF, KRASAmp and KRASG12D or KRASG12V for which there are currently no mutant-selective inhibitors

• Translation of these preclinical findings into clinical benefit could position RMC-4630, an investigational therapeutic agent, as a backbone of targeted therapy combinations for patients bearing cancers with diverse oncogenic mutations in the RAS pathway

SHP2 is a Frontier Target in Oncology• Direct targeting of oncogenic mutations in the RAS pathway is a beneficial therapeutic

strategy for patients with cancers with these mutations. Mutant-selective inhibitors offer a wide therapeutic window but are ultimately limited by emergence of drug resistance

• Escape from mutant-selective inhibitors frequently involves activation of wild-type signaling nodes, including hyperactivation of receptor tyrosine kinases (RTKs), that lead to robust re-activation of the RAS pathway

• SHP2 (PTPN11) is a phosphatase that functions as a convergent node downstream of multiple RTKs to regulate RAS activation. We have recently shown that single agent inhibition of SHP2 has anti-tumor activity in tumors harboring KRASG12C both in the clinic and preclinical models1, 2

• In the context of adaptive resistance to mutant-selective inhibitors, SHP2 inhibition has the potential to suppress oncoprotein-mediated signaling and adaptive signaling driving escape from therapy

• For many RAS pathway oncogenic drivers, including KRASG12D and KRASG12V, NF1LOF, KRASAmp or BRAFClass3, mutant-selective inhibitors are not currently available. Here, a combination strategy simultaneously targeting nodes both up- and down-stream of the oncoprotein (“oncoprotein clamping”) can drive tumor growth inhibition

• Here we show that SHP2 inhibitors have the potential to become the backbone of targeted therapy combinations across the spectrum of RAS-dependent tumors

• We have also shown that SHP2 inhibition, alone or in combination, can promote anti-tumor immunity in preclinical models via effects on the innate and adaptive immune systems4, 5. These effects may influence the overall profile of a SHP2 inhibitor

Therapeutic Combinations for RAS Driven Tumors: Mutant-selective and RAS Pathway Node Inhibitors

5. Combination Benefit for SHP2 and MEK Inhibition in Other KRAS-Mutant Xenograft Models

References1. Nichols et al., Nat Cell Biol. 2018 20(9):1064-10732. Ou et al. AACR-IASCLC 20203. Clinical Trials.Gov: NCT036349824. Quintana et al., 2020 Cancer Research 10.1158/0008-54725. Shifrin et al., 2020 AACR A7744 P28376. Planchard et al., Annals Oncology 26: 2073–2078, 2015

Acknowledgements:Jingjing Jiang for expert input into design and execution of in vivo pharmacology modelsCRO support: WuXi AppTec (Suzhou, China); Champions Oncology (Maryland, USA); Charles River Laboratories/OncotestGmbh (Freiburg, Germany); Genendesign (Shanghai, China); Xentech (Evry, France); TD2 (Arizona, USA)

Best Change in Tumor Burden from Baseline NSCLC with any KRAS Mutation for RMC-4630 Monotherapy2, 3

Disease Control Rate: NSCLC KRASmut 12/18 (67%) NSCLC KRASG12C 6/8 (75%)

Data presented for efficacy evaluable population (N = 18) defined as patients with baseline and at least one post-baseline scan or who died or had clinical progression prior to first post-baseline scan.Four patients are not represented in this figure: 2 patients had clinical progression prior to first scan, 1 patient did not have measurements for one of the target lesions but progressed due to new lesion, and 1 patient had missing tumor measurements in the database at the time of data extract.• Confirmed PR # Unconfirmed PR

✘ ✘

Each animal represented as separate barN = number of regressions >10% at end of study; 10 mice/group

Clinical Preclinical NCI-H358 KRASG12C NSCLC Xenograft

• All treatments were well-tolerated

RMC-4630 (PO) PK/PD in vivo

RMC-4630 anti-tumor activity in vivoNCI-H358 KRASG12C NSCLC xenograft

NCI-H358 KRASG12C NSCLC xenograftCompound RMC-4630 RMC-45501

Tool Compound

SHP2 biochemical potency (IC50, nM) 1.29 1.52

RAS pathway suppression (pERK IC50, nM)NCI-H358 KRASG12C 20 28

Anti-proliferative activity (3D CTG IC50, nM)NCI-H358 KRASG12C

NCI-H1975 EGFRL858R/T790M3225

4363

Selectivity- Phosphatase panel- Kinase panel

> 3,000> 3,000

> 3,000> 3,000

SHP2 inhibitors - in vitro profile

KRASG12V Pancreatic

KRASG12V NSCLCNCI-H441

Capan-2

KRASG12D Pancreatic

KP-4 HPAC

LUN #150, NSCLC

CO-04-0004, CRC

STO#332 WT KRASAmp (copy number = 4)

105

KRASAmpNF1LOF

CompoundParental

EGFRL858R/T790M

(IC50, nM)Transfected

EGFRL858R/T790M/C797S (IC50, nM)

RMC-4630 265 218 to 557

Osimertinib 8 30 to 2616

EGFRL858R/T790M/C797S in vitro

EGFRL858R/T790/METAmp NSCLC PDX6

Osimertinib-Sensitive Osimertinib-ResistantNCI-H1975 EGFRL858R/T790M NSCLC CDX

Anti-proliferative activity (2D CTG) in parental NCI-H1975 or cells transfected with human EGFRL858R/T790M/C797S under six different promotersN= 12-15 mice/group

One-way Anova: * p< 0.05; *** p < 0.0001

N = 3 mice/group; graphs show tumor volume data for individual mice, expressed as % of initial tumor volume at time of study start.

N = 12 mice/group; graphs in B show tumor volume data for individual animals shown in A, expressed as % change in tumor volume from time of study start

A

B

Each animal represented as separate barN = number of regressions >10% at end of study; 10 mice/group

Ordinary one-way ANOVA, * p< 0.01, ** p< 0.05, ***p<0.001

LUN #352

• All treatments were well-tolerated

• All treatments were well-tolerated

0 10 20 300

200

400

600

800

Days on Study

Mea

n Tu

mor

Vol

ume

(mm

3 ) ControlRMC-4630 10 mg/kg po qdCobimetinib 2.5 mg/kg po qdCombination

• PDX models of tumors bearing NF1 mutations predicted to result in loss of function (LOF): deletions, insertions, premature stops, truncations

• Tumor growth inhibition in 62% of NF1LOF PDX models (n=55)• Tumor regressions in 25% (23/93) of responders (93/166 mice)

N= 10 mice/group40 80 120 160

0

500

1000

1500

2000

2500

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

ControlOsimertinib 5 mg/kg po qdRMC-4630 30 mg/kg po qdCombination

Initial tumor volume256 mm3

Dosing stopped

40 80 120 160

0

400

800

Days Post Implant

-100

% C

hang

e in

Tum

or V

olum

e

Dosingstopped

Osimertinib 5 mg/kg po qd

40 80 120 160

0

400

800

-100

% C

hang

e in

Tum

or V

olum

e

Days Post-implant

RMC-4630 30 QD + Osimertinib 5 QD

Dosingstopped

40 80

0

100

Days Post Implant

-100

-50

40 80

0

100

-50

-100

Days Post-implant

% C

hang

e in

Tum

or V

olum

e Osimertinib

Combination

1 2 4 8 10 16 241

10

100

1000

10000

0

20

40

60

80

100

Time post-dose (hours)

Unbo

und

Plas

ma

Conc

n(n

M)

% Inhibition pErk/tErkRelative to Control

10 mg/kg30 mg/kg

-9 -8 -7 -6 -50

25

50

75

100

Log Molar [RMC-4630]plasma, unbound

% In

hibi

tion

of p

ERK/

tERK

rela

tive

to c

ontro

l

EC50 = 27 nM

Time post dose2h8h

16h24h

Time post second dose (bid)2h8h

0 10 20 30-100

0

100

200

300

Days on Study

% C

hang

e in

Tum

or V

olum

e

ControlRMC-4630 30 mg/kg po qd

Dose/ schedule

www.revolutionmedicines.com

A5307P1943

[email protected]

0 10 20 300

500

1000

1500

2000

2500

Days on Study

Mea

n Tu

mor

Vol

ume

(mm

3 )

*

***

*#

*** ***

***

*

***0 5 10 15 20 25 30

0

400

800

1200

1600

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 ) ControlRMC-4630 10 mg/kg po qdRMC-4630 30 mg/kg po qd

Dosingstart

***

*

0

5 10 15 20 250

1000

2000

3000

Days Post Implant

Mea

n Tu

mor

Vol

ume

(mm

3 )

ControlOsimertinib 25 mg/kg po 5d on/2d offRMC-4630 30 mg/kg po qd