1
-10 -9 -8 -7 -6 -5 -4 0 25 50 75 100 125 Log 10 [Compound] (M) Percent of Control MK-6482 Compound 3 Compound 4 Series 1 inhibitors were optimized to improve potency and pharmacokinetic properties via structure-based design and iterative interpretation of structure activity relationships. A selection of optimized advanced prototypes are shown in Table 2 which potently inhibit HIF-2α function in numerous assays formats without appreciable off-target activity. 20 40 60 80 100 -1×10 5 -5×10 4 0 5×10 4 Temperature (°C) Derivative of Fluorescence Our advanced prototype HIF-2α inhibitor, Compound 3, exhibited a favorable in vitro pharmacokinetic profile with low intrinsic clearance in dog and human hepatocytes (Table 3). Furthermore, Compound 3 exhibited negligible inhibition against a panel of CYP isoforms (Table 4) and no time- dependent CYP inhibition (not shown). Compound 3 is further characterized by moderate-to-low clearance in rat and dog with high bioavailability in both species (Table 5). -9 -8 -7 -6 -5 -4 -3 0 25 50 75 100 125 Log 10 [Compound] (M) Percent of Activity Small molecules have been designed to inhibit HIF-2α/ARNT heterodimerization by binding a small, internal cavity in the HIF-2α PAS-B domain. This hydrophobic cavity (shown as blue slate surface below) is fully enclosed with a volume of 290Å 3 , and is occupied by 8 water molecules in the apo form. It has been demonstrated that small molecules can enter 21% O 2 DMSO 1% O 2 DMSO 10 1 0.1 10 1 0.1 0.0 0.2 0.4 0.6 0.8 1.0 Relative EPO Compound 3 ( M) MK-6482 ( M) 21% O 2 DMSO 1% O 2 DMSO 10 1 0.1 10 1 0.1 0 5 10 15 20 25 Relative PAI1 Compound 3 ( M) MK-6482 ( M) **** **** **** **** **** **** **** **** **** **** **** **** **** **** Kenneth V Lawson , Kelsey E Sivick Gauthier, Dana Piovesan, Jeremy T A Fournier, Brandon R Rosen, Artur Mailyan, Joel W Beatty, Lixia Jin, Manmohan Leleti, Elaine Ginn, Akshata R Udyavar, Ada Chen, Jennifer Au, Cesar A Meleza, Stephen W. Young, Matthew J Walters, Jay P Powers Arcus Biosciences, Inc.; 3928 Point Eden Way, Hayward, CA 94545 (USA) Discovery and Characterization of Novel, Potent, and Selective Hypoxia-Inducible Factor (HIF)-2α Inhibitors 2020 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Abstract 32 Three distinct compound series are undergoing iterative SAR optimization to develop novel HIF-2α antagonists. Representative compounds from each series show both HIF-2α binding and functional activity in cell-based assays (Table 1, Figure 3). Series 1 inhibitors have been optimized via structure-based design and interrogation of SAR trends to afford numerous potent advanced prototype HIF-2α inhibitors (Table 2). A prototypical example, Compound 3, strongly inhibited HIF-2α target gene expression in Hep3B cells. In contrast HIF-1α target genes expression was minimally altered indicating Arcus inhibitors are highly selective for HIF-2α (Figure 5). Optimized Arcus inhibitors, such as Compound 3, exhibit a favorable pharmacokinetic profile characterized by low intrinsic clearance in human hepatocytes and high oral bioavailability in preclinical species. The solid tumor microenvironment (TME) can be hypoxic and cancer cells require induction of genes associated with metabolism, proliferation, and angiogenesis to survive and metastasize 3 . The master transcriptional regulators of hypoxia-induced genes are the Hypoxia-Inducible Factor (HIF) proteins 4 . HIF consists of an oxygen-regulated alpha monomer, of which there are three isoforms (HIF-1α, HIF-2α, and HIF-3α) 4 . Alpha monomers heterodimerize with a constitutively-expressed beta monomer (HIF-1β/ARNT) using Per-ARNT-SIM (PAS) protein-protein interaction domains 4 . Disruption of HIF-α/HIF-1β heterodimer formation is an effective means to inhibition of HIF-2α-dependent gene transcription 4 . Figure 1. Overview of HIF-2α regulation. In normoxia (left), proline residues present in the oxygen-dependent degradation domain (ODDD) of HIF-2α are hydroxylated by prolyl hydroxylases (PHDs), allowing for recognition by the von Hippel-Lindau (pVHL) E3-ubiquitin ligase complex and subsequent ubiquitination and proteasomal degradation. Upon exposure to low oxygen conditions (hypoxia, right) or in the case of vhl mutation or silencing (pseudohypoxia), HIF-2α subunits accumulate and dimerize with HIF-1β/ARNT, resulting in transcription of various gene sets, some of which are pro-tumorigenic, downstream of hypoxia- response element (HRE) DNA binding sites. Adapted from Yu et al. 5 . 1) Jonasch et al. (2020) ASCO 2020, Abstract #5003. 2) Srinivasan et al. (2020) ESMO, Abstract #LBA26. 3) Hockel & Vaupel (2001) JNCI 93, 266-276. 4) Li et al. (2019) J Med Chem. OVERVIEW HIF-2α BIOLOGY & REGULATION INITIAL DESIGN, OPTIMIZATION, AND CHARACTERIZATION OF ARCUS HIF-2α INHIBITORS PHARMACOKINETIC PROFILING CITATIONS HYPOXIA HIF-2α HIF-1β HIF-2α Target Gene Transcription Hypoxia CBP/p300 HIF-2α HIF-1β HRE No Hydroxylation PHDs HIF-2α Nuclear Translocation & Heterodimer Formation NORMOXIA HIF-2α ELOC ELOB CUL2 RBX1 pVHL Hydroxylation HIF-2α Proteosomal Degradation PHDs Ub Ub Ub E3 Ligase Complex Association Ubiquitination SUMMARY Table 1. Representative initial lead examples for Arcus Series 1, Series 2, and Series 3 HIF-2α inhibitor compounds. (MST = microscale thermophoresis, ITC = isothermal calorimetry) Figure 3. A & B) HIF and Control Cellular Reporter Assay. 786-O renal adenocarcinoma cells (mutant for VHL and HIF-1α) stably expressing HIF or control CMV luciferase reporter constructs (Qiagen) were treated with Arcus compounds for 20 hours (h) at 37C 5% CO 2 . C) Scintillation Proximity Assay (SPA). 50 nM PAS-B was incubated at room temperature with Arcus compounds in 2% DMSO for 60 min and 3 μg copper chelate PVT SPA beads for an additional 45 min before to addition of 25 nM 3 H-tracer and luciferase measurement. D) Thermal Shift Assay (TSA). Arcus compounds were incubated with PAS-B prior to addition of dye and fluorescence measurement. ΔT m was calculated by normalizing compound T m to DMSO T m . Dotted lines, DMSO only. Fundamentals of Targeting the HIF-2α/ARNT Complex + O 2 HIF-2α OH OH HIF-2α OH OH HIF-2α OH OH Ub B A C D Control Reporter SPA TSA Figure 5. Prototype HIF-2α inhibitor (Compound 3) inhibits HIF-2α-, but not HIF-1α-, mediated transcription of pro- tumorigenic gene sets. Hep3B hepatocellular carcinoma cells (wild- type for VHL and HIF-1α) were treated with 0.1, 1.0 , or 10 μM of Compound 3 or MK- 6482 and exposed to hypoxia (1% O 2 ) for 16 h prior to RNA isolation. Gene expression levels of HIF-2α target genes (EPO and PAI1) and HIF-1α genes (PDK1 and PGK1) were determined by qPCR (2 -Ct method) relative to HPRT1. Optimization of Series 1 HIF-2α Inhibitors 5) Yu et al. (2019) Drug Disc Today 00, 1-9. 6) Rogers et. al. (2013) J Med Chem 56, 17391747 7) Data from Arcus test of molecule described in Wehn et al. (2018) J Med Chem 61, 9691-9721. 8) Xu et. al. (2019) J Med Chem 62, 68766893 PDB-4ZP4 (2.4 Å) PAS-A PAS-B PAS-B bHLH HIF-2a PAS-B binding site PAS-A bHLH Inhibitor design challenges: Small internal pocket limits ligand size Binding affinity may not correlate with functional activity High affinity ligands often possess undesirable physicochemical properties (high lipophilicity) Assay Series 1 Series 2 Series 3 HIF-2a 786-O Luc. (Cellular) IC 50 (M) 2.4 1.12 0.72 786-O Reporter (control, M) > 50 > 50 > 50 HIF-2a TSA T M (degrees) 5.1 5.3 6.3 HIF-2a MST K D (M) 1.5 0.44 0.13 HIF-2a ITC K D (M) 0.44 1.25 0.66 HIF-2a SPA IC 50 (M) 1.0 0.96 0.49 Extensive Characterization of Initial HIF-2α Initial Lead Series Basis for regulation of protein-protein interaction 6 : Small molecule binds to HIF-2α PAS-B cavity Conformational change HIF dimerization disrupted Gene transcription inactive Assay Arcus Compound 1 Arcus Compound 2 Arcus Compound 3 Arcus Compound 4 MK-6482 (PT2977, competitor) 7 HIF-2a 786-O Luc. IC 50 (nM) 61.5 9.4 15.5 4.2 22.8 786-O Reporter (control, nM) >10,000 >10,000 >10,000 >10,000 > 10,000 HIF-2a SPA IC 50 (nM) 64.0 22.7 14.9 14.6 30.4 VEGF Secretion IC 50 (nM) n.d. 75.9 51.1 21.1 93.1 Arcus Compound 3 Selectively Inhibits HIF-2α Gene Transcription Pharmacokinetic Characterization of Compound 3 21% O 2 DMSO 1% O 2 DMSO 10 1 0.1 10 1 0.1 0 2 4 6 8 Relative PDK1 Compound 3 ( M) MK-6482 ( M) 21% O 2 DMSO 1% O 2 DMSO 10 1 0.1 10 1 0.1 0 20 40 60 80 100 Relative PGK1 Compound 3 ( M) MK-6482 ( M) **** **** * *** * * HIF-2α Target Genes HIF-1α Target Genes ARNT PAS-B HIF-2a PAS-B ARNT PAS-B iHIF-2α iHIF-2α [Stable HIF-2α/ARNT Complex] [Inhibitor induced dissociation of HIF-2α/ARNT complex] + the cavity and induce a subtle conformation change of the HIF-2α PAS-B domain, which, in turn, results in destabilization of the HIF- 2α/ARNT complex. 6 -9 -8 -7 -6 -5 -4 0 50 100 Log 10 [Compound] (M) Percent of Control -9 -8 -7 -6 -5 -4 0 25 50 75 100 125 Log 10 [Compound] (M) Percent of Control Series 2 Series 1 Series 3 Panels A-D: PDB-3F1P HIF-2α / ARNT Preclinical Species Pharmacokinetics Species CL (L/h/kg) Vss (L/kg) T 1/2 (h) F (%) Rat 1.94 3.04 1.3 79 Dog 0.26 1.25 3.7 78 Table 5. Summary of experimental PK parameters in rat and dog. Rats were dosed 0.25 mg/kg IV in DMAC:Ethanol:Propylene Glycol:Saline (10:10:30:50) and 2 mg/kg PO in PEG400:Vitamin E TPGS (95:5). Dogs were dosed 0.33 mg/kg IV in DMA/PG/water (1:1:1) and 2 mg/kg PO in 1% HMPC. Hepatocyte Stability Mouse Rat Dog Human T 1/2 (min) 21 100 340 950 CL int (μL/min/10 6 cells) 33 6.9 2.1 0.7 Table 3. Summary of hepatocyte stability in various species. CYP Isoform 2C8 2C9 2C19 2D6 3A4 IC 50 (μM) >40 39.0 16.2 >40 >40 Table 4. Compound was evaluated in vitro for its potential to inhibit major human drug metabolizing enzymes of the cytochrome P450 family. VEGF Secretion B A Figure 4. A) Representative VEGF dose-response curve of optimized Arcus HIF-2α inhibitors. MK-6482 included for comparison. 7,8 B) X-ray co-crystal structure of prototypical series 1 analog, Arcus Compound 1 (orange spheres), bound to HIF-2α/ARNT complex confirms inhibitor binding to PAS-B domain hydrophobic cavity. Figure 2. X-ray structure of HIF-2α/ARNT complex. EPO PAI1 (Serpine1) PGK1 PDK1 Table 2. Potency of select series 1 inhibitors. HIF/control reporter and SPA assays performed as described in Table 1. VEGF Protein Secretion Assay - 786-O cells were treated with inhibitors for 48 hours at 37 C 5% CO 2 (Media replaced with fresh after 24 hr). VEGF in the cell supernatant was quantified by AlphaLISA (Perkin Elmer). HIF-2α 786-O Luc. HIF-2α PAS-B ARNT PAS-B Preclinical and clinical evidence suggests that HIF-2α inhibition is a valid approach to destroy tumor cells, particularly in clear cell renal carcinoma (ccRCC) 1,2 . Arcus Biosciences is developing novel HIF-2α-specific small- molecule inhibitors and investigating the biology of HIF-2α in various cancer and non-cancer cell subsets. Here we describe the application of a pharmacophore mapping and structure-based design approach to discover multiple novel series of HIF-2α inhibitors which are characterized via a collection of in vitro assays. Highly optimized inhibitors exhibit low-nanomolar potency against HIF-2α and a favorable pharmacokinetic profile.

et al. Wehn MK-6482 DMSO · 2020. 11. 19. · pVHL Hydroxylation HIF-2αProteosomal Degradation PHDs Ub Ub Ub E3 Ligase Complex Association Ubiquitination SUMMARY Table 1. Representative

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: et al. Wehn MK-6482 DMSO · 2020. 11. 19. · pVHL Hydroxylation HIF-2αProteosomal Degradation PHDs Ub Ub Ub E3 Ligase Complex Association Ubiquitination SUMMARY Table 1. Representative

-10 -9 -8 -7 -6 -5 -4

0

25

50

75

100

125

Log 10 [Compound] (M)

Pe

rce

nt

of

Co

ntr

ol

MK-6482

Compound 3

Compound 4

Series 1 inhibitors were optimized to improve potency and pharmacokinetic properties via structure-based design

and iterative interpretation of structure activity relationships. A selection of optimized advanced prototypes are shown

in Table 2 which potently inhibit HIF-2α function in numerous assays formats without appreciable off-target activity.

20 40 60 80 100

-1×105

-5×104

0

5×104

Temperature (°C)

Deri

vati

ve o

f F

luo

rescen

ce

Our advanced prototype HIF-2α inhibitor, Compound 3, exhibited a

favorable in vitro pharmacokinetic profile with low intrinsic clearance in dog

and human hepatocytes (Table 3). Furthermore, Compound 3 exhibited

negligible inhibition against a panel of CYP isoforms (Table 4) and no time-

dependent CYP inhibition (not shown). Compound 3 is further

characterized by moderate-to-low clearance in rat and dog with high

bioavailability in both species (Table 5).

-9 -8 -7 -6 -5 -4 -3

0

25

50

75

100

125

Log 10 [Compound] (M)

Perc

en

t o

f A

cti

vit

y

Small molecules have been designed to inhibit HIF-2α/ARNT heterodimerization by binding a small, internal cavity in the

HIF-2α PAS-B domain. This hydrophobic cavity (shown as blue slate surface below) is fully enclosed with a volume of

290Å3, and is occupied by 8 water molecules in the apo form. It has been demonstrated that small molecules can enter

the cavity via localized melting and invoke subtle conformation change of the HIF-2a PAS-B domain which, in turn,

results in destabilization of the HIF-2a/ARNT complex.

21% O2DMSO

1% O2DMSO

10 1 0.1 10 1 0.10.0

0.2

0.4

0.6

0.8

1.0

Re

lati

ve

EP

O

Compound 3

(M)

MK-6482

(M)

21% O2DMSO

1% O2DMSO

10 1 0.1 10 1 0.10

5

10

15

20

25

Rela

tive P

AI1

Compound 3

(M)

MK-6482

(M)

******** **** **** **** **** **** ******** **** **** **** **** ****

Kenneth V Lawson, Kelsey E Sivick Gauthier, Dana Piovesan, Jeremy T A Fournier, Brandon R Rosen, Artur Mailyan, Joel W Beatty, Lixia Jin, Manmohan Leleti, Elaine Ginn, Akshata R Udyavar, Ada Chen,

Jennifer Au, Cesar A Meleza, Stephen W. Young, Matthew J Walters, Jay P Powers

Arcus Biosciences, Inc.; 3928 Point Eden Way, Hayward, CA 94545 (USA)

Discovery and Characterization of Novel, Potent, and Selective Hypoxia-Inducible Factor (HIF)-2α Inhibitors2020 AACR-NCI-EORTC

Molecular Targets and

Cancer Therapeutics

Abstract 32

❖ Three distinct compound series are undergoing iterative SAR

optimization to develop novel HIF-2α antagonists. Representative

compounds from each series show both HIF-2α binding and functional

activity in cell-based assays (Table 1, Figure 3).

❖ Series 1 inhibitors have been optimized via structure-based design and

interrogation of SAR trends to afford numerous potent advanced

prototype HIF-2α inhibitors (Table 2).

❖ A prototypical example, Compound 3, strongly inhibited HIF-2α target

gene expression in Hep3B cells. In contrast HIF-1α target genes

expression was minimally altered indicating Arcus inhibitors are highly

selective for HIF-2α (Figure 5).

❖ Optimized Arcus inhibitors, such as Compound 3, exhibit a favorable

pharmacokinetic profile characterized by low intrinsic clearance in

human hepatocytes and high oral bioavailability in preclinical species.

❖ The solid tumor microenvironment (TME) can be hypoxic and cancer

cells require induction of genes associated with metabolism,

proliferation, and angiogenesis to survive and metastasize3.

❖ The master transcriptional regulators of hypoxia-induced genes are

the Hypoxia-Inducible Factor (HIF) proteins4.

❖ HIF consists of an oxygen-regulated alpha monomer, of which there

are three isoforms (HIF-1α, HIF-2α, and HIF-3α)4.

❖ Alpha monomers heterodimerize with a constitutively-expressed beta

monomer (HIF-1β/ARNT) using Per-ARNT-SIM (PAS) protein-protein

interaction domains4.

❖ Disruption of HIF-α/HIF-1β heterodimer formation is an effective

means to inhibition of HIF-2α-dependent gene transcription4.

Figure 1. Overview of HIF-2α regulation. In normoxia (left), proline residues present in the

oxygen-dependent degradation domain (ODDD) of HIF-2α are hydroxylated by prolyl

hydroxylases (PHDs), allowing for recognition by the von Hippel-Lindau (pVHL) E3-ubiquitin

ligase complex and subsequent ubiquitination and proteasomal degradation. Upon exposure to

low oxygen conditions (hypoxia, right) or in the case of vhl mutation or silencing

(pseudohypoxia), HIF-2α subunits accumulate and dimerize with HIF-1β/ARNT, resulting in

transcription of various gene sets, some of which are pro-tumorigenic, downstream of hypoxia-

response element (HRE) DNA binding sites. Adapted from Yu et al.5.

1) Jonasch et al. (2020) ASCO 2020, Abstract #5003.

2) Srinivasan et al. (2020) ESMO, Abstract #LBA26.

3) Hockel & Vaupel (2001) JNCI 93, 266-276.

4) Li et al. (2019) J Med Chem.

OVERVIEW

HIF-2α BIOLOGY & REGULATION

INITIAL DESIGN, OPTIMIZATION, AND CHARACTERIZATION OF ARCUS HIF-2α INHIBITORS PHARMACOKINETIC PROFILING

CITATIONS

HYPOXIA

HIF-2α

HIF-1β

HIF-2α Target Gene Transcription

Hypoxia

CBP/p300

HIF-2α HIF-1β

HRE

No

Hydroxylation

PHDs

HIF-2α

Nuclear

Translocation

& Heterodimer

Formation

NORMOXIA

HIF-2α

ELOC

ELOB

CUL2

RBX1

pVHL

Hydroxylation

HIF-2α Proteosomal Degradation

PHDs

Ub

UbUb

E3 Ligase

Complex

Association

Ubiquitination

SUMMARY

Table 1. Representative initial lead examples for Arcus Series 1, Series 2, and Series 3 HIF-2α inhibitor compounds. (MST =

microscale thermophoresis, ITC = isothermal calorimetry) Figure 3. A & B) HIF and Control Cellular Reporter Assay. 786-O renal

adenocarcinoma cells (mutant for VHL and HIF-1α) stably expressing HIF or control CMV luciferase reporter constructs (Qiagen) were

treated with Arcus compounds for 20 hours (h) at 37C 5% CO2. C) Scintillation Proximity Assay (SPA). 50 nM PAS-B was incubated at

room temperature with Arcus compounds in 2% DMSO for 60 min and 3 µg copper chelate PVT SPA beads for an additional 45 min

before to addition of 25 nM 3H-tracer and luciferase measurement. D) Thermal Shift Assay (TSA). Arcus compounds were incubated

with PAS-B prior to addition of dye and fluorescence measurement. ΔTm was calculated by normalizing compound Tm to DMSO Tm.

Dotted lines, DMSO only.

Fundamentals of Targeting the HIF-2α/ARNT Complex

+ O2

HIF-2α

OHOH

HIF-2α

OHOH

HIF-2α

OHOH

Ub

BA

C D

Control Reporter

SPA TSA

Figure 5. Prototype HIF-2α inhibitor

(Compound 3) inhibits HIF-2α-, but not

HIF-1α-, mediated transcription of pro-

tumorigenic gene sets.

Hep3B hepatocellular carcinoma cells (wild-

type for VHL and HIF-1α) were treated with

0.1, 1.0 , or 10 µM of Compound 3 or MK-

6482 and exposed to hypoxia (1% O2) for 16

h prior to RNA isolation. Gene expression

levels of HIF-2α target genes (EPO and PAI1)

and HIF-1α genes (PDK1 and PGK1) were

determined by qPCR (2-Ct method) relative

to HPRT1.

Optimization of Series 1 HIF-2α Inhibitors

5) Yu et al. (2019) Drug Disc Today 00, 1-9.

6) Rogers et. al. (2013) J Med Chem 56, 1739–1747

7) Data from Arcus test of molecule described in

Wehn et al. (2018) J Med Chem 61, 9691-9721.

8) Xu et. al. (2019) J Med Chem 62, 6876–6893

PDB-4ZP4 (2.4 Å)

PAS-A

PAS-B

PAS-B

bHLH

HIF-2a PAS-B binding site

PAS-A

bHLH

• Inhibitor design challenges:

‒ Small internal pocket limits ligand size

‒ Binding affinity may not correlate with

functional activity

‒ High affinity ligands often possess undesirable

physicochemical properties (high lipophilicity)

Assay Series 1 Series 2 Series 3

HIF-2a 786-O Luc.

(Cellular) IC50 (M)2.4 1.12 0.72

786-O Reporter

(control, M)> 50 > 50 > 50

HIF-2a TSA TM

(degrees)5.1 5.3 6.3

HIF-2a MST KD

(M)1.5 0.44 0.13

HIF-2a ITC KD

(M)0.44 1.25 0.66

HIF-2a SPA IC50

(M)1.0 0.96 0.49

Extensive Characterization of Initial HIF-2α Initial Lead Series

• Basis for regulation of protein-protein

interaction6:

• Small molecule binds to HIF-2α PAS-B cavity

→ Conformational change

‒ HIF dimerization disrupted

→ Gene transcription inactive

AssayArcus

Compound 1Arcus

Compound 2Arcus

Compound 3Arcus

Compound 4MK-6482

(PT2977, competitor)7

HIF-2a 786-O Luc. IC50 (nM) 61.5 9.4 15.5 4.2 22.8

786-O Reporter

(control, nM)>10,000 >10,000 >10,000 >10,000 > 10,000

HIF-2a SPA IC50

(nM)64.0 22.7 14.9 14.6 30.4

VEGF Secretion IC50

(nM)n.d. 75.9 51.1 21.1 93.1

Arcus Compound 3 Selectively Inhibits HIF-2α Gene Transcription

Pharmacokinetic Characterization of Compound 3

21% O2DMSO

1% O2DMSO

10 1 0.1 10 1 0.10

2

4

6

8

Re

lati

ve

PD

K1

Compound 3

(M)

MK-6482

(M)

21% O2DMSO

1% O2DMSO

10 1 0.1 10 1 0.10

20

40

60

80

100

Re

lati

ve

PG

K1

Compound 3

(M)

MK-6482

(M)

******** * *** * *

HIF-2α Target Genes

HIF-1α Target Genes

ARNT PAS-B

HIF-2a PAS-B

ARNT PAS-B

iHIF-2αiHIF-2α

[Stable HIF-2α/ARNT Complex][Inhibitor induced dissociation of

HIF-2α/ARNT complex]

+

the cavity and induce a

subtle conformation

change of the HIF-2α

PAS-B domain, which, in

turn, results in

destabilization of the HIF-

2α/ARNT complex.6

-9 -8 -7 -6 -5 -4

0

50

100

Log 10 [Compound] (M)

Pe

rce

nt

of

Co

ntr

ol

-9 -8 -7 -6 -5 -4

0

25

50

75

100

125

Log 10 [Compound] (M)

Pe

rce

nt

of

Co

ntr

ol

-8 -7 -6 -5 -4 -3

-50

0

50

100

150

Transform of A0306327-003_SPA

Log 10 [Compound] (M)

PO

C (

%)

Series 2

Series 1

Series 3

Panels A-D:

PDB-3F1P

HIF-2α / ARNT

Preclinical Species Pharmacokinetics

SpeciesCL

(L/h/kg)Vss

(L/kg)T1/2

(h)F

(%)

Rat 1.94 3.04 1.3 79

Dog 0.26 1.25 3.7 78

Table 5. Summary of experimental PK parameters in rat and dog. Rats were dosed

0.25 mg/kg IV in DMAC:Ethanol:Propylene Glycol:Saline (10:10:30:50) and 2 mg/kg

PO in PEG400:Vitamin E TPGS (95:5). Dogs were dosed 0.33 mg/kg IV in

DMA/PG/water (1:1:1) and 2 mg/kg PO in 1% HMPC.

Hepatocyte Stability

Mouse Rat Dog Human

T1/2

(min)21 100 340 950

CLint

(µL/min/106 cells)33 6.9 2.1 0.7

Table 3. Summary of hepatocyte stability in various species.

CYP Isoform

2C8 2C9 2C19 2D6 3A4

IC50 (µM) >40 39.0 16.2 >40 >40

Table 4. Compound was evaluated in vitro for its potential to inhibit major human

drug metabolizing enzymes of the cytochrome P450 family.

VEGF Secretion BA

Figure 4. A) Representative VEGF dose-response curve of optimized Arcus

HIF-2α inhibitors. MK-6482 included for comparison.7,8 B) X-ray co-crystal

structure of prototypical series 1 analog, Arcus Compound 1 (orange spheres), bound

to HIF-2α/ARNT complex confirms inhibitor binding to PAS-B domain hydrophobic cavity.

Figure 2. X-ray structure of HIF-2α/ARNT complex.

EPO PAI1 (Serpine1)

PGK1PDK1

Table 2. Potency of select series 1 inhibitors. HIF/control reporter and SPA assays performed as described in Table 1. VEGF

Protein Secretion Assay - 786-O cells were treated with inhibitors for 48 hours at 37 C 5% CO2 (Media replaced with fresh after

24 hr). VEGF in the cell supernatant was quantified by AlphaLISA (Perkin Elmer).

HIF-2α 786-O Luc.

HIF-2α PAS-B

ARNT PAS-B

❖ Preclinical and clinical evidence suggests that HIF-2α inhibition is a

valid approach to destroy tumor cells, particularly in clear cell renal

carcinoma (ccRCC)1,2.

❖ Arcus Biosciences is developing novel HIF-2α-specific small-

molecule inhibitors and investigating the biology of HIF-2α in various

cancer and non-cancer cell subsets.

❖ Here we describe the application of a pharmacophore mapping and

structure-based design approach to discover multiple novel series of

HIF-2α inhibitors which are characterized via a collection of in vitro

assays. Highly optimized inhibitors exhibit low-nanomolar potency

against HIF-2α and a favorable pharmacokinetic profile.