discovery and preclinical development of mrtx849: co69 a ... · a mutation-selective kras g12c...

1
H1373 H2122 Compound 1 pERK IC 50 = 7.5 uM 1 Compound 2 pERK IC 50 = 70 nM 1 Compound 3 pERK IC 50 = 5 nM 1 MRTX1257 pERK IC 50 = 900 pM 1 pERK IC 50 = 6 nM 2 MRTX849 pERK IC 50 = 14 nM 1 pERK IC 50 = 5 nM 2 REFERENCES & ACKNOWLEDGEMENTS 1. Bos J, Cancer Res 49, 4682-4689 (1989). 2. Ostrem JM, et al. Nature 503, 548–551 (2013). 3. Lim SM, et al. Angew. Chem. 53, 199–204 (2014). 4. Lito, P. et al. Science 351, 604-608 (2016). 5. Patricelli, M.P. et al. Cancer Discovery 6, 317-329 (2016). 6. Ostrem JM, Shokat, KM Nat. Rev. Drug Discov. 15, 771–785 (2016). 7. Makas, A et al. Crit. Rev. Oncol. Hematol 110, 1-12 (2017). 8. Fell, JB et al. ACS Med. Chem Le. 9, 1230-1234 (2018). 9. Janes MR, et al. Cell 172, 578–589 (2018). 10. Sanchez-Vega, F et al. Cell 172, 321-337 (2018). Fig. 1: MRTX849 Key Small Molecule Interactions with KRAS G12C Protein Fig. 4: KRAS G12C Protein Modificaon 6hrs Post Single Dose of MRTX849 Fig. 6 MRTX849 at 100mg/kg leads to significant pERK and pS6 inhibion by IHC in mulple xenograſt models Fig. 5: Normalized pERK and pS6 Inhibion 6hrs Post Single Dose of MRTX849 Fig. 2: Cysteine Selectivity in H358 Cells with MRTX849 (1uM) Fig. 3: Antitumor Efficacy in MIA PaCa-2 Xenograft Model MRTX849 Treatment Achieves KRAS Modification, Biomarker Modulation, and Antitumor Efficacy MRTX849 Demonstrates Broad Spectrum Tumor Regression in KRAS G12C Nonclinical Models Clinical Target Concentration Projections Derived from Nonclinical PKPD Modeling Fig. 7: 17 of 26 KRAS G12C Xenografts Show >30% Regression with MRTX849 at 100mg/kg QD Discovery and Preclinical Development of MRTX849: A Mutation-Selective KRAS G12C Inhibitor CONCLUSIONS Opmizaon of the warhead reacvity and naphthyl substuent of tool compound MRTX1257 led to the opmized compound MRTX849 with favorable human PK and dose predicons. Dose response administraon of MRTX849 in preclinical models led to KRAS modificaon, pERK and pS6 modulaon, and antumor acvity which defined the PKPD relaonship and target AUC needed for human dose predicons. MRTX849 administered daily PO at well-tolerated dose levels induced 30% or greater tumor regression in ~65% of cell line and paent-derived xenograſts with durable complete regressions observed in a subset of models. Nonclinical PKPD modeling predict that target AUC is achievable at dose levels presently safely achieved in clinical trials. Projected human half-life exceeding 20 hours is predicted to be advantageous in liming KRAS-dependent signaling feedback reacvaon. CO69 Jill Hallin 1 , Matthew A. Marx 1 , Lars D. Engstrom 1 , Lauren Hargis 1 , Andrew Calinisan 1 , Ruth Aranda 1 , Aaron C. Burns 1 , David M. Briere 1 , Niranjan Sudhakar 1 , Vickie Bowcut 1 , Brian R. Baer 2 , James F. Blake 2 , Michael R. Burkard 2 , Jay B. Fell 2 , John P. Fischer 2 , Tony P. Tang 2 , Lance Wollenberg 2 , Guy P. Vigers 2 , Peter Olson 1 , James G. Christensen 1 1 Mira Therapeucs, Inc., San Diego, CA 92121, USA, 2 Array BioPharma Inc., Boulder, CO 80301, USA Relationship Between Dose, AUC 0-24 and Antitumor Efficacy in Nonclinical Human Xenograft Models BACKGROUND • Despite decades of research, efforts to directly target KRAS have been challenging. The MRTX chemical series is comprised of mutant-selecve, covalent inhibitors that target the GDP-bound form of KRAS G12C, locking it in the inacve state. The oral bioavailability and ADME properes of a previously described tool compound, MRTX1257, was limited by GST-mediated glutathione (GSH) scavenging, leading to unfavorable predicted human PK properes. Opmizaon of the 8-substuent of the naphthyl ring and modulaon of the acrylamide reacvity led to MRTX849, with improved GST-mediated GSH stability and favorable human PK projecons. The clinical candidate molecule MRTX849, a KRAS G12C mutant-selecve, covalent inhibitor of KRAS G12C was idenfied through structure-based drug design with low nanomolar cell potency and favorable oral properes. Previous Poster Disclosure: Hallin J. et al. Insight Towards Therapeuc Suscepbility of KRAS Mutant Cancers from MRTX1257: A prototype selecve inhibitor of KRAS G12C. Poster presented at: 2019 AACR Mar 29-Apr 3; Atlanta, GA. The addition of the C2 substituent significantly improved solubility and cellular potency compared with the lead compound 1 and demonstrated more rapid modification of the protein. The introduction of the cyanomethyl substituent on the piperazine further improved potency, allowing for the elimination of the 3-hydroxyl group on the naphthyl ring, improving ADME properties. The 8-position of the naphthyl group was substituted to fill a hydrophobic pocket, increasing potency an additional 5-fold. Warhead modification and final optimization for reactivity and bioavailability provided MRTX849. KEY PROTEIN-LIGAND INTERACTIONS Cys12: Covalent bond formed to acrylamide olefin Lys16: Acrylamide carbonyl interacon with NH3+ Gly10: Nitrile H-bond to backbone N-H Glu62: C2 tail salt bridge to the acid His95: H-bond to pyrimidine nitrogen Tyr96: pi-pi interacon to the pyrimidine IHC shows near complete inhibion of KRAS-mediated pathway inhibion and provides a more accurate representaon of signal modulaon than bulk tumor lysate. Near complete durable inhibion of KRAS-dependent signaling for full dose interval correlates with maximum an- tumor acvity. Near complete durable inhibion of pERK is associated with deep and durable responses, while transient and/or incomplete pERK inhibion is associated with submaximal antumor acvity 100mg/kg MRTX849 PO daily for 16 days demonstrates complete response with no tumor recovery aſter dosing cessaon. The reactivity of the acrylamide species of MRTX1257 leads to short half-lives in whole blood, due in part to GST-mediated GSH uptake. Systematic adjustment of the acrylamide reactivity and further optimization of the naphthyl 8-substituent led to MRTX849, which shows greater stability in whole blood. The cross-species bioavailability of MRTX849 was improved relative to MRTX1257, leading to favorable human PK and dose predictions. PBPK modeling ulizing PK Sim and Gastro Plus projects 50% oral bioavailability and >20 hour half-life in humans. Present PK data in MRTX849-001 clinical trial supports these projecons. Schedule dependence and infusion studies were ulized to define plasma exposure parameters that best fit antumor acvity regardless of dose schedule. AUC and C avg were the closest fit for PK parameters driving antumor acvity. The AUC 0-24 values determined at the 30 mg/kg and 100 mg/kg dose levels which demonstrated maximum antumor acvity in the MIA PaCa-2 (sensive) and HCC44 (parally sensive) models, respecvely, corrected for mouse plasma protein binding were ulized for human efficacious exposure projecons. The free-fracon adjusted AUC 0-24 corrected for human plasma protein binding (98.3%) were calculated as 14.3 ug*h/mL and 37.1 ug*h/mL in these 2 models. PBPK Link modeling approaches ulizing PK-Sim TM or GastroPlus 9.5 TM soſtware were applied, and data inputs including the total human projected target AUC values, along with nonclinical ADME data inputs, and in vivo clearance determined in mouse, rat and dog studies were applied to project a human efficacious dose and map human exposure relave to target efficacious concentraons. The data inputs fit well and human PK data was predictable based on PBPK modeling. Mean human AUCs achievable at steady-state at 600 mg BID well-exceeded even the most aggressive target plasma concentraons. 1 ICW average value, pERK IC 50 . MIA PaCa-2 cells, 24 hour incubation 2 GST-mediated GSH conjugation, hours 1 LCMS-based KRAS G12C protein modification assay utilizing MRTX849 pre-loaded with GDP * human projected F% ** indicates p-value <0.01 by two-tailed Student’s t-test Brackets indicate p-value <0.05 compared to left-most sample, by two-tailed Student’s t-test” 1 pERK IC 50 , 3hrs in H358 cells, 2 pERK IC 50 , 24hrs in MIA PaCa-2 cells Drug Discovery Progression Toward MRTX849 MRTX849 Binds GDP-KRAS G12C and Locks the Protein in the Inactive State MRTX849 Is A Potent, Selective and Orally Bioavailable Clinical Compound www.mira.com WE WOULD LIKE TO THANK: • Crown Biosciences for their support with in vivo PDX models. • Flagship Biosciences for their contribuons to immunohistochemistry images and analysis. MRTX1257 MRTX849 Cellular Potency 1 6 nM 5 nM Half-life in whole blood 2 , m/r/d/h 8, 5, 4, 16 > 50 in all species Bioavailability, % m/r/d 31/8/4 (30mg/kg mouse and rat, 10mg/kg dog) 63/30/26 (30mg/kg) V d,ss L/Kg, m/r/d 2/6/6 2/21/17 Model Dose (mg/kg) AUC 0-24 (ug*h/mL) FF adj AUC 0-24 (ug*h/mL) % Regression (Day) Projected Efficacious Total/FF adj AUC (human, ug*h/mL) MIA PaCa-2 10 7 0.0697 -52% (13) - MIA PaCa-2 30 24 0.243 -96% (13) 14.3 HCC-44 100 63 0.63 -61% (13) 37.1 MRTX849 MW/cLogP/tPSA 604 / 5.8 / 87 K inact /K I 35 +/- 0.3mM -1 s -1 5 min / 3uM Protein Modificaon 1 66% Hepatocyte ER % (m/h) 61 / 50 Plasma Protein Binding % (m/h) 99.0 / 98.3 F % (m/h) 63 / 50* MIA PaCa-2 pERK 100mpk pERK Veh pS6 Veh pS6 100mpk

Upload: others

Post on 06-Apr-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Discovery and Preclinical Development of MRTX849: CO69 A ... · A Mutation-Selective KRAS G12C Inhibitor CONCLUSIONS • Optimization of the warhead reactivity and naphthyl substituent

H13

73H

2122

Compound 1pERK IC50 = 7.5 uM1

Compound 2pERK IC50 = 70 nM1

Compound 3 pERK IC50 = 5 nM1

MRTX1257pERK IC50 = 900 pM1

pERK IC50 = 6 nM2

MRTX849pERK IC50 = 14 nM1

pERK IC50 = 5 nM2

REFERENCES & ACKNOWLEDGEMENTS1. Bos J, Cancer Res 49, 4682-4689 (1989).

2. Ostrem JM, et al. Nature 503, 548–551 (2013).

3. Lim SM, et al. Angew. Chem. 53, 199–204 (2014).

4. Lito, P. et al. Science 351, 604-608 (2016).

5. Patricelli, M.P. et al. Cancer Discovery 6, 317-329 (2016).

6. Ostrem JM, Shokat, KM Nat. Rev. Drug Discov. 15, 771–785 (2016).

7. Matikas, A et al. Crit. Rev. Oncol. Hematol 110, 1-12 (2017).

8. Fell, JB et al. ACS Med. Chem Lett. 9, 1230-1234 (2018).

9. Janes MR, et al. Cell 172, 578–589 (2018).

10. Sanchez-Vega, F et al. Cell 172, 321-337 (2018).

Fig. 1: MRTX849 Key Small Molecule Interactions with KRAS G12C Protein Fig. 4: KRAS G12C Protein Modification 6hrs Post Single Dose of MRTX849

Fig. 6 MRTX849 at 100mg/kg leads to significant pERK and pS6 inhibition by IHC in multiple xenograft models

Fig. 5: Normalized pERK and pS6 Inhibition 6hrs Post Single Dose of MRTX849

Fig. 2: Cysteine Selectivity in H358 Cells with MRTX849 (1uM)

Fig. 3: Antitumor Efficacy in MIA PaCa-2 Xenograft Model

MRTX849 Treatment Achieves KRAS Modification, Biomarker Modulation, and Antitumor Efficacy

MRTX849 Demonstrates Broad Spectrum Tumor Regression in KRAS G12C Nonclinical Models

Clinical Target Concentration Projections Derived from Nonclinical PKPD Modeling

Fig. 7: 17 of 26 KRAS G12C Xenografts Show >30% Regression with MRTX849 at 100mg/kg QD

Discovery and Preclinical Development of MRTX849: A Mutation-Selective KRAS G12C Inhibitor

CONCLUSIONS• Optimization of the warhead reactivity and naphthyl substituent of tool compound

MRTX1257 led to the optimized compound MRTX849 with favorable human PK and dose predictions.

• Dose response administration of MRTX849 in preclinical models led to KRAS modification, pERK and pS6 modulation, and antitumor activity which defined the PKPD relationship and target AUC needed for human dose predictions.

• MRTX849 administered daily PO at well-tolerated dose levels induced 30% or greater tumor regression in ~65% of cell line and patient-derived xenografts with durable complete regressions observed in a subset of models.

• Nonclinical PKPD modeling predict that target AUC is achievable at dose levels presently safely achieved in clinical trials. Projected human half-life exceeding 20 hours is predicted to be advantageous in limiting KRAS-dependent signaling feedback reactivation.

CO69

Jill Hallin1, Matthew A. Marx1, Lars D. Engstrom1, Lauren Hargis1, Andrew Calinisan1, Ruth Aranda1, Aaron C. Burns1, David M. Briere1, Niranjan Sudhakar1, Vickie Bowcut1, Brian R. Baer2, James F. Blake2, Michael R. Burkard2, Jay B. Fell2, John P. Fischer2, Tony P. Tang2, Lance Wollenberg2, Guy P. Vigers2, Peter Olson1, James G. Christensen1 1Mirati Therapeutics, Inc., San Diego, CA 92121, USA, 2Array BioPharma Inc., Boulder, CO 80301, USA

Relationship Between Dose, AUC0-24 and Antitumor Efficacy in Nonclinical Human Xenograft Models

BACKGROUND• Despite decades of research, efforts to directly target KRAS have been challenging.• The MRTX chemical series is comprised of mutant-selective, covalent inhibitors that

target the GDP-bound form of KRAS G12C, locking it in the inactive state.• The oral bioavailability and ADME properties of a previously described tool

compound, MRTX1257, was limited by GST-mediated glutathione (GSH) scavenging, leading to unfavorable predicted human PK properties.

• Optimization of the 8-substituent of the naphthyl ring and modulation of the acrylamide reactivity led to MRTX849, with improved GST-mediated GSH stability and favorable human PK projections.

• The clinical candidate molecule MRTX849, a KRAS G12C mutant-selective, covalent inhibitor of KRAS G12C was identified through structure-based drug design with low nanomolar cell potency and favorable oral properties.

Previous Poster Disclosure: Hallin J. et al. Insight Towards Therapeutic Susceptibility of KRAS Mutant Cancers from MRTX1257: A prototype selective inhibitor of KRAS G12C. Poster presented at: 2019 AACR Mar 29-Apr 3; Atlanta, GA.

• The addition of the C2 substituent significantly improved solubility and cellular potency compared with the lead compound 1 and demonstrated more rapid modification of the protein.

• The introduction of the cyanomethyl substituent on the piperazine further improved potency, allowing for the elimination of the 3-hydroxyl group on the naphthyl ring, improving ADME properties.

• The 8-position of the naphthyl group was substituted to fill a hydrophobic pocket, increasing potency an additional 5-fold.

• Warhead modification and final optimization for reactivity and bioavailability provided MRTX849.

KEY PROTEIN-LIGAND INTERACTIONS

Cys12: Covalent bond formed to acrylamide olefin

Lys16: Acrylamide carbonyl interaction with NH3+

Gly10: Nitrile H-bond to backbone N-H

Glu62: C2 tail salt bridge to the acid

His95: H-bond to pyrimidine nitrogen

Tyr96: pi-pi interaction to the pyrimidine

• IHC shows near complete inhibition of KRAS-mediated pathway inhibition and provides a more accurate representation of signal modulation than bulk tumor lysate.

• Near complete durable inhibition of KRAS-dependent signaling for full dose interval correlates with maximum anti-tumor activity.

• Near complete durable inhibition of pERK is associated with deep and durable responses, while transient and/or incomplete pERK inhibition is associated with submaximal antitumor activity

• 100mg/kg MRTX849 PO daily for 16 days demonstrates complete response with no tumor recovery after dosing cessation.

• The reactivity of the acrylamide species of MRTX1257 leads to short half-lives in whole blood, due in part to GST-mediated GSH uptake.

• Systematic adjustment of the acrylamide reactivity and further optimization of the naphthyl 8-substituent led to MRTX849, which shows greater stability in whole blood.

• The cross-species bioavailability of MRTX849 was improved relative to MRTX1257, leading to favorable human PK and dose predictions.

• PBPK modeling utilizing PK Sim and Gastro Plus projects 50% oral bioavailability and >20 hour half-life in humans. Present PK data in MRTX849-001 clinical trial supports these projections.

• Schedule dependence and infusion studies were utilized to define plasma exposure parameters that best fit antitumor activity regardless of dose schedule. AUC and Cavg were the closest fit for PK parameters driving antitumor activity.

• The AUC0-24 values determined at the 30 mg/kg and 100 mg/kg dose levels which demonstrated maximum antitumor activity in the MIA PaCa-2 (sensitive) and HCC44 (partially sensitive) models, respectively, corrected for mouse plasma protein binding were utilized for human efficacious exposure projections. The free-fraction adjusted AUC0-24 corrected for human plasma protein binding (98.3%) were calculated as 14.3 ug*h/mL and 37.1 ug*h/mL in these 2 models.

• PBPK Link modeling approaches utilizing PK-SimTM or GastroPlus 9.5TM software were applied, and data inputs including the total human projected target AUC values, along with nonclinical ADME data inputs, and in vivo clearance determined in mouse, rat and dog studies were applied to project a human efficacious dose and map human exposure relative to target efficacious concentrations.

• The data inputs fit well and human PK data was predictable based on PBPK modeling. Mean human AUCs achievable at steady-state at 600 mg BID well-exceeded even the most aggressive target plasma concentrations.

1 ICW average value, pERK IC50. MIA PaCa-2 cells, 24 hour incubation 2 GST-mediated GSH conjugation, hours

1 LCMS-based KRAS G12C protein modification assay utilizing MRTX849 pre-loaded with GDP * human projected F%

** indicates p-value <0.01 by two-tailed Student’s t-test

Brackets indicate p-value <0.05 compared to left-most sample, by two-tailed Student’s t-test”

1 pERK IC50, 3hrs in H358 cells, 2 pERK IC50, 24hrs in MIA PaCa-2 cells

Drug Discovery Progression Toward MRTX849

MRTX849 Binds GDP-KRAS G12C and Locks the Protein in the Inactive State

MRTX849 Is A Potent, Selective and Orally Bioavailable Clinical Compound

www.mirati.com

WE WOULD LIKE TO THANK:

• Crown Biosciences for their support with in vivo PDX models.

• Flagship Biosciences for their contributions to immunohistochemistry images and analysis.

MRTX1257 MRTX849

Cellular Potency1 6 nM 5 nM

Half-life in whole blood2, m/r/d/h 8, 5, 4, 16 > 50 in all species

Bioavailability, % m/r/d 31/8/4 (30mg/kg mouse and rat, 10mg/kg dog)

63/30/26 (30mg/kg)

Vd,ss L/Kg, m/r/d 2/6/6 2/21/17

Model Dose (mg/kg)

AUC0-24

(ug*h/mL)FF adj AUC0-24

(ug*h/mL)% Regression

(Day)Projected Efficacious

Total/FF adj AUC (human, ug*h/mL)

MIA PaCa-2 10 7 0.0697 -52% (13) -

MIA PaCa-2 30 24 0.243 -96% (13) 14.3

HCC-44 100 63 0.63 -61% (13) 37.1

MRTX849

MW/cLogP/tPSA 604 / 5.8 / 87

Kinact/KI 35 +/- 0.3mM-1s-1

5 min / 3uM Protein Modification1 66%

Hepatocyte ER % (m/h) 61 / 50

Plasma Protein Binding % (m/h) 99.0 / 98.3

F % (m/h) 63 / 50*

MIA

PaC

a-2

pERK 100mpkpERK Veh pS6 Veh pS6 100mpk