+ All Categories
Transcript
Page 1: Acetyl-CoA Carboxylase Inhibition by ND-630 Inhibits Fatty ... · Geraldine Harriman, Jeremy Greenwood*, Sathesh Bhat*, Liang Tong#, Ruiying Wang#, Debamita Paul#, Rosana Kapeller

Nimbus Discovery - 25 First Street, Suite 404 - Cambridge, MA 02141 USA - Phone: (617) 588-2655 - Fax: (617) 231-0652 - Email:[email protected] - Web:www.nimbusdiscovery.com

Acetyl-CoA Carboxylase Inhibition by ND-630 Inhibits Fatty Acid Synthesis and Stimulates Fatty Acid Oxidation in Cultured Cells and in Experimental Animals

Geraldine Harriman, Jeremy Greenwood*, Sathesh Bhat*, Liang Tong#, Ruiying Wang#, Debamita Paul#, Rosana Kapeller and H. James Harwood Jr.Nimbus Discovery, Inc., Cambridge, MA, USA; *Schrödinger, Inc., New York, NY, USA; #Columbia University, New York, NY, USA

NO

N

O

N

O

O O

O

CH3O

OCH3

OHOH

OCH3

0 12

5,000

Months

Ideas

Milestone Achieved

Confirm binding mode (x-ray)

175 Total

synthesized compound

8,000 nM <10 nM Potency

ACC1, ACC2

1.3MM

In vivo target engagement

16

10,000

225

<1 nM

Rat DIO Studiesin progress

mg

TGs

per

g Li

ver T

issu

e

0

2

4

3 10 30Vehicle ND-630 (mg/kg)

Lean DIO

0

100

150

200

250

300

0.39 1.56 6.25 25 100Inhibitor (μM)

mA

50 Probe + cpd + ACC2Probe + cpd + buffer

-787

-387

-187

13

213

1013

30 40 50 60 70 80Temperature (°C)

Fluo

resc

ence

(dR

)

413

613

-587

813

5 μM hBC5 μM hBC + 50 μM Cp 5 μM hBC + 20 μM Soraphen5 μM hBC + 100 μM Ligand5 μM hBC + 50 μM Ligand5 μM hBC + 20 μM Ligand

0.001

0.010

0.100

1.000

10.000

100.000

0 4 8 12 16 20 24Time (hrs)

Con

cent

ratio

n (μ

M)

IV PO_A

0.001 0.01 0.1 1 100

10000

20000

30000

40000

cpm

ND-630CP640186(3 μM)

Inhibitor (μM)

ED50= 0.14 mg/kg PO

0

10

20

30

40

50

60

70

Vehicl

e 0.3 1mg/kg

3 10 30

DP

Ms/

mg

Tiss

ue

MED = 3 mg/kg PO

0.7

0.75

0.8

0.85

0.9

0.95

1

1.05

1.1

1.15

-60 0 60 120 180 240

RQ

(±S

EM

)

Vehicle 3mpk 10mpk 30mpk

Time (min)

Mito

chon

drion

Acetyl -CoA

ACC Inhibitor

ACC1

Malonyl-CoA

Malonyl-CoA

Fatty acid synthesis Fatty acid oxidation

ACC2

Hepatic Lipid Lowering

Liver Targeted

Liver Disease

Inhibitor Mediated Effects Potential For Disease Modification

Improvement in Insulin Sensitivity with

Weight Neutrality Type 2 Diabetes

Plasma Lipid Lowering Dyslipidemia

ABSTRACT Inhibition of acetyl-CoA carboxylase (ACC) reduces fatty acid synthesis and stimulates fatty acid oxidation and has the potential to favorably affect the morbidity and mortality asso-ciated with obesity, diabetes, and fatty liver diseases. ACC exists as two tissue-specific isozymes; ACC1 present in li-pogenic tissues and ACC2 present in oxidative tissues. To achieve maximal effectiveness an ACC inhibitor should inhibit both isozymes. Most efforts to discover ACC inhibitors have focused on in-teractions within the carboxyltransferase (CT) domain of the enzyme active center. By contrast, our efforts have focused on the allosteric subunit dimerization site on the biotin car-boxylase (BC) domain where Soraphen interacts. This is also the same site where the phospho-containing motif of phos-pho-ACC binds to prevent dimerization and subsequent acti-vation of the enzyme. Using state-of-the-art structure-based drug design tech-niques together with crystal structures of the BC domain of human ACC2, we identified a unique series of small mole-cule allosteric inhibitors that bind to the Soraphen binding site and inhibit enzymatic activity. Development of this series has yielded deep structure-activity relationships, sub-nanomolar enzyme inhibition, functional activity in HepG2 and C2C12 cells and favorable drug-like properties leading to in vivo proof of concept. For example, the series representative ND-630 inhibits both human ACC1 and ACC2 (IC50 = 2.0 nM), inhibits HepG2 cell fatty acid synthesis (EC50 = 66 nM), stimulates C2C12 cell fat-ty acid oxidation (2-fold at 200 nM), inhibits rat hepatic fatty acid synthesis (ED50 = 0.14 mg/kg) and stimulates rat whole body fatty acid oxidation (minimum effective dose 3 mg/kg). Together these observations suggest that allosteric ACC inhibition has the potential to favorably impact diabetes, obe-sity, fatty liver and lipid disorders.

• A structure-based virtual screen and drug design approach utilizing WaterMap was used to identify allosteric inhibitors of ACC that bind to the BC domain.

• Inhibitors were successfully optimized for excellent potency and drug-like properties in 12 months.

• ND-630 demonstrated in vivo proof of concept in pharmaco-logically relevant models of target engagement (rat FASyn ED50 = 0.14 mpk, PO & rat RQ MED = 3 mpk, PO).

FIGURE 1: Acetyl CoA Carboxylase (ACC): Master Regulator of Fatty Acid Synthesis & Oxidation

SUMMARY

NEXT STEPS• Using this approach, a portfolio of nanomolar inhibitors with diverse functional-group driven bio-distribution patterns have been identified and are being utilized for a breadth of ACC-aligned indications.

• Nimbus will report on the detailed pharmacology of ND-630 and other molecules in metabolic disease models, diabe-tes models and oncology models (cancer metabolism) in the future.

• Beneficial effects on lipids, blood glucose, weight, potentially diabetes and CV risk.

• Nimbus: FIRST small molecule allosteric inhibitor successfully targeting BC domain.

FIGURE 2: Nimbus Allosteric Inhibitors Show Promise for Both Metabolic Disease & Fatty Liver Disease

FIGURE 3: Previous Approaches Identified Inhibitors of the ACC CT Domain (ex. CP-640186)

FIGURE 4: Nimbus’ Unique Approach Identified Potent ACC BC Domain Inhibitors Targeting Soraphen Binding Site

FIGURE 5: Nimbus Approach has Delivered ACC Inhibitors with Demonstrated in vivo PoC in 12 Months

• Most CT-domain inhibitor series identified exhibited poor drug-like properties.

• Nimbus took an orthogonal approach and designed inhibitors that bound to the Soraphen binding site in the BC domain of the ACC complex.

• A structure-based virtual screen of 1.3M lead-like molecules was performed utiliz-ing Schrodinger’s computational tools including WaterMap and Glide. This led to the identification of a high quality family of hit molecules with micromolar inhibitory effects against the enzyme. Co-crystal structures of members of this series in the human BC domain confirmed the model and our approach.

• In an iterative design fashion over the next 12 month period, the potency of this fam-ily of hits were improved 1000x utilizing the computational model focusing on the re-placement or stabilization of high energy hydration sites within the Soraphen binding site.

• Simultaneous to the potency improvements, drug-like properties were optimized to deliver Development Candidate quality molecules that are currently being profiled in chronic models of metabolic syndrome and diabetes.

FIGURE 6: In vitro Assays Confirm Binding of Early Hits to BC Domain Prior to Obtaining Co-crystal Structures

• Early in the program we utilized the Soraphen-TAMRA binding assay and BC domain thermal shift assays to provide us confidence that our inhibitors in fact were binding in the Soraphen binding site.

• These tools were utilized until the first Nimbus inhibitor was co-crystalized in the hu-man BC domain. This co-crystal structure taught us our computational model was highly predictive and also provided additional information for WaterMap calculations.

FIGURE 7: ND-630 Displays Excellent Drug-like Properties

FIGURE 8: ND-630 in vivo Proof of Concept: Target Engagement in the Liver and Muscle

TARGET POTENCY• ACC1 IC50 = 2 nM• ACC2 IC50 = 2 nM• HepG2 EC50 = 66 nM• C2C12 FAOxn stim. 2x @ 200 nM

ADMET• Low multispecies intrinsic clearance (human,

mouse, rat, dog, monkey)• High solubility (>300 mM)• P450 inhib >50 mM, hERG >30 mM• Protein binding (98%)• DrugMatrixPanel 0/120 hits; 1000x window

RAT PK (10 mg/kg, PO)• 37%F, Cmax 6 mM, Cl 33 mL/min/kg, AUC 3.4 mM.hr, T1/2 = 4.5h, Vss 2 L/kg

• Liver exposure: 81 mM• Muscle exposure: 0.45 mM

PHARMACOLOGY• Rat RQ MED = 3 mpk PO• Rat FASyn ED50 = 0.14 mpk, PO

Soraphen-TAMRA Assay(Hit displaces soraphen)

Thermal Shift Assay(Hit binds to the BC domain)

Plasma PK Profile SD Rats IV (3 mpk, n=3, solution dose)PO_A (10 mpk, n=3, suspension dose)

Hep-G2 cell FAsyn Inhibition [14C]acetate incorporation into FA)

Target Engagement in Liver(Fatty acid synthesis inhibition)

Target Engagement in Muscle(Respiratory quotient)

Confirmatory Co-crystal Structures

Soraphen A

CP-640186

Top Related