+ All Categories
Home > Documents > Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition...

Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition...

Date post: 12-Jul-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
6
Immune-modulating enzyme indoleamine 2,3-dioxygenase is effectively inhibited by targeting its apo-form Micah T. Nelp a , Patrick A. Kates a , John T. Hunt b , John A. Newitt c , Aaron Balog d , Derrick Maley b , Xiao Zhu c , Lynn Abell e , Alban Allentoff f , Robert Borzilleri d , Hal A. Lewis c , Zeyu Lin e , Steven P. Seitz d , Chunhong Yan c , and John T. Groves a,1 a Department of Chemistry, Princeton University, Princeton, NJ 08544; b Immuno-oncology Biology, Bristol-Myers Squibb Co., Princeton, NJ 08543; c Molecular Discovery Technologies, Bristol-Myers Squibb Co., Princeton, NJ 08543; d Department of Discovery Chemistry, Bristol-Myers Squibb Co., Princeton, NJ 08543; e Leads Discovery and Optimization, Bristol-Myers Squibb Co., Princeton, NJ 08543; and f Department of Radiochemistry, Bristol-Myers Squibb Co., Princeton, NJ 08543 Contributed by John T. Groves, February 12, 2018 (sent for review November 3, 2017; reviewed by Emma L. Raven and Syun-Ru Yeh) For cancer cells to survive and proliferate, they must escape normal immune destruction. One mechanism by which this is accomplished is through immune suppression effected by up-regulation of indole- amine 2,3-dioxygenase (IDO1), a heme enzyme that catalyzes the oxidation of tryptophan to N-formylkynurenine. On deformylation, kynurenine and downstream metabolites suppress T cell function. The importance of this immunosuppressive mechanism has spurred intense interest in the development of clinical IDO1 inhibitors. Herein, we describe the mechanism by which a class of compounds effectively and specifically inhibits IDO1 by targeting its apo- form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzymeheme dissocia- tion, especially in the ferric form. X-ray crystal structures of the inhibitorenzyme complexes show that heme is displaced from the enzyme and blocked from rebinding by these compounds. The results reveal that apo-IDO1 serves as a unique target for inhibition and that heme lability plays an important role in posttranslational regulation. IDO1 | heme | cancer | kynurenine M ulticellular life is tasked with the immensely complex pro- cess of clearing foreign and aberrant cells while preventing autoimmunity. This finely tuned balance is often mediated by en- zymes involved in central metabolism, reflecting the ancient origin and strong selective pressure of immune regulation (1). Indoleamine 2,3-dioxygenase (IDO1) is one such enzyme that oxidizes the es- sential amino acid tryptophan to produce N-formylkynurenine, which is further hydrolyzed to kynurenine (2, 3). This enzyme is present in many tissues and is up-regulated in response to in- flammation, specifically the presence of cytokines, such as IFN-γ, as well as bacterial lipopolysaccharides (4, 5). Cells expressing IDO1 are able to deplete the inflamed environment of the metaboli- cally expensive substrate, tryptophan, inhibiting the proliferation of immune targets (6, 7). IDO1 is also able to serve as an immunosuppressive enzyme. In a seminal paper, IDO1, which was known to be highly expressed in placental tissue, was shown to be essential for the protection of embryos from maternal immune responses. An IDO1 inhibitor, 1-methyl-tryptophan, caused rejection of fetuses capable of pro- voking the maternal immune response, whereas fetuses closely related to the mother, and thus less likely to provoke an immune response, survived (8). This remarkable ability of a single enzyme to mediate such complex immune behavior has since been studied in depth, revealing a rich variety of mechanisms by which this en- zyme affects immune regulation. Expression of IDO1 inactivates surrounding immune cells through the combined effects of low tryptophan and high concentrations of kynurenine (1, 9). T cells are especially sensitive to low tryptophan concentrations, where they undergo cell cycle arrest (10). Addi- tionally, the downstream metabolites of the product of IDO1 are potent activators of the aryl hydrocarbon receptor through which apoptosis of immune cells can be initiated (11). IDO1 has further been shown to provide protection from oxidative stress caused by inflammatory processes (12). The importance of IDO1 in precise immune regulation is high- lighted by its effects in a variety of processes and disease states, including autoimmune disorders, response to infection, tolerance in transplantation, HIV infection, and blood pressure regulation (1315). This regulation/dysregulation is perhaps most sinisterly manifested in cancer cells that co-opt the immunosuppressive ability of IDO1 to evade immune destruction (1618). The transcriptional control of IDO1 in cancers is often altered through mutation of the Bin1 repressor that allows for vastly up-regulated levels of IDO1 (19). Tumors displaying high IDO1 activity are associated with poor prognoses (20). Accordingly, IDO1 is a prime target in the treat- ment of almost all cancers for which IDO1 inhibition could restore the ability of the immune system to remove these cancer cells on its own or in combination with other treatments (17). The immuno- suppressive effects of IDO1 are also implicated in persistent Significance Indoleamine 2,3-dioxygenase (IDO1) is a heme protein that cata- lyzes the dioxygenation of tryptophan. Cells expressing this ac- tivity are able to profoundly alter their surrounding environment to suppress the immune response. Cancer cells exploit this path- way to avoid immune-mediated destruction. Through a range of kinetic, structural, and cellular assays, we show that two classes of highly selective inhibitors of IDO1 act by competing with heme binding to apo-IDO1. This shows that IDO1 is dynamically bound to its heme cofactor in what is likely a critical step in the regula- tion of this enzyme. These results have elucidated a previously undiscovered role for the ubiquitous heme cofactor in immune regulation, and it suggests that other heme proteins in biology may be similarly regulated. Author contributions: M.T.N., P.A.K., J.T.H., J.A.N., A.B., D.M., X.Z., L.A., A.A., R.B., H.A.L., S.P.S., C.Y., and J.T.G. designed research; M.T.N., P.A.K., J.A.N., A.B., D.M., X.Z., A.A., H.A.L., Z.L., S.P.S., and C.Y. performed research; M.T.N., P.A.K., J.T.H., J.A.N., A.B., D.M., X.Z., L.A., A.A., R.B., H.A.L., Z.L., S.P.S., C.Y., and J.T.G. analyzed data; and M.T.N. and J.T.G. wrote the paper. Reviewers: E.L.R., University of Leicester; and S.-R.Y., Albert Einstein College of Medicine. Conflict of interest statement: M.T.N., P.A.K., and J.T.G. declare no conflict of interest. X.Z. and L.A. are former employees of Bristol-Myers Squibb Co. J.T.H., J.A.N., A.B., D.M., A.A., R.B., H.L., Z.L., S.P.S., and C.Y. are employees of Bristol-Myers Squibb Co. This open access article is distributed under Creative Commons Attribution-NonCommercial- NoDerivatives License 4.0 (CC BY-NC-ND). Data deposition: The atomic coordinates and structure factors have been deposited in the Protein Data Bank, www.wwpdb.org [PDB ID codes 6AZU (holoenzyme), 6AZV (1), and 6AZW (3)]. 1 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1719190115/-/DCSupplemental. Published online March 12, 2018. www.pnas.org/cgi/doi/10.1073/pnas.1719190115 PNAS | March 27, 2018 | vol. 115 | no. 13 | 32493254 CHEMISTRY BIOCHEMISTRY Downloaded by guest on August 20, 2020
Transcript
Page 1: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

Immune-modulating enzyme indoleamine2,3-dioxygenase is effectively inhibitedby targeting its apo-formMicah T. Nelpa, Patrick A. Katesa, John T. Huntb, John A. Newittc, Aaron Balogd, Derrick Maleyb, Xiao Zhuc, Lynn Abelle,Alban Allentofff, Robert Borzillerid, Hal A. Lewisc, Zeyu Line, Steven P. Seitzd, Chunhong Yanc, and John T. Grovesa,1

aDepartment of Chemistry, Princeton University, Princeton, NJ 08544; bImmuno-oncology Biology, Bristol-Myers Squibb Co., Princeton, NJ 08543; cMolecularDiscovery Technologies, Bristol-Myers Squibb Co., Princeton, NJ 08543; dDepartment of Discovery Chemistry, Bristol-Myers Squibb Co., Princeton, NJ 08543;eLeads Discovery and Optimization, Bristol-Myers Squibb Co., Princeton, NJ 08543; and fDepartment of Radiochemistry, Bristol-Myers Squibb Co.,Princeton, NJ 08543

Contributed by John T. Groves, February 12, 2018 (sent for review November 3, 2017; reviewed by Emma L. Raven and Syun-Ru Yeh)

For cancer cells to survive and proliferate, they must escape normalimmune destruction. One mechanism by which this is accomplishedis through immune suppression effected by up-regulation of indole-amine 2,3-dioxygenase (IDO1), a heme enzyme that catalyzes theoxidation of tryptophan to N-formylkynurenine. On deformylation,kynurenine and downstream metabolites suppress T cell function.The importance of this immunosuppressive mechanism has spurredintense interest in the development of clinical IDO1 inhibitors.Herein, we describe the mechanism by which a class of compoundseffectively and specifically inhibits IDO1 by targeting its apo-form. We show that the in vitro kinetics of inhibition coincidewith an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially in the ferric form. X-ray crystal structures of theinhibitor–enzyme complexes show that heme is displaced fromthe enzyme and blocked from rebinding by these compounds.The results reveal that apo-IDO1 serves as a unique target forinhibition and that heme lability plays an important role inposttranslational regulation.

IDO1 | heme | cancer | kynurenine

Multicellular life is tasked with the immensely complex pro-cess of clearing foreign and aberrant cells while preventing

autoimmunity. This finely tuned balance is often mediated by en-zymes involved in central metabolism, reflecting the ancient originand strong selective pressure of immune regulation (1). Indoleamine2,3-dioxygenase (IDO1) is one such enzyme that oxidizes the es-sential amino acid tryptophan to produce N-formylkynurenine,which is further hydrolyzed to kynurenine (2, 3). This enzyme ispresent in many tissues and is up-regulated in response to in-flammation, specifically the presence of cytokines, such as IFN-γ,as well as bacterial lipopolysaccharides (4, 5). Cells expressing IDO1are able to deplete the inflamed environment of the metaboli-cally expensive substrate, tryptophan, inhibiting the proliferationof immune targets (6, 7).IDO1 is also able to serve as an immunosuppressive enzyme. In

a seminal paper, IDO1, which was known to be highly expressed inplacental tissue, was shown to be essential for the protection ofembryos from maternal immune responses. An IDO1 inhibitor,1-methyl-tryptophan, caused rejection of fetuses capable of pro-voking the maternal immune response, whereas fetuses closelyrelated to the mother, and thus less likely to provoke an immuneresponse, survived (8). This remarkable ability of a single enzymeto mediate such complex immune behavior has since been studiedin depth, revealing a rich variety of mechanisms by which this en-zyme affects immune regulation.Expression of IDO1 inactivates surrounding immune cells through

the combined effects of low tryptophan and high concentrations ofkynurenine (1, 9). T cells are especially sensitive to low tryptophanconcentrations, where they undergo cell cycle arrest (10). Addi-tionally, the downstream metabolites of the product of IDO1 arepotent activators of the aryl hydrocarbon receptor through which

apoptosis of immune cells can be initiated (11). IDO1 has furtherbeen shown to provide protection from oxidative stress caused byinflammatory processes (12).The importance of IDO1 in precise immune regulation is high-

lighted by its effects in a variety of processes and disease states,including autoimmune disorders, response to infection, tolerancein transplantation, HIV infection, and blood pressure regulation(13–15). This regulation/dysregulation is perhaps most sinisterlymanifested in cancer cells that co-opt the immunosuppressive abilityof IDO1 to evade immune destruction (16–18). The transcriptionalcontrol of IDO1 in cancers is often altered through mutation of theBin1 repressor that allows for vastly up-regulated levels of IDO1(19). Tumors displaying high IDO1 activity are associated with poorprognoses (20). Accordingly, IDO1 is a prime target in the treat-ment of almost all cancers for which IDO1 inhibition could restorethe ability of the immune system to remove these cancer cells on itsown or in combination with other treatments (17). The immuno-suppressive effects of IDO1 are also implicated in persistent

Significance

Indoleamine 2,3-dioxygenase (IDO1) is a heme protein that cata-lyzes the dioxygenation of tryptophan. Cells expressing this ac-tivity are able to profoundly alter their surrounding environmentto suppress the immune response. Cancer cells exploit this path-way to avoid immune-mediated destruction. Through a range ofkinetic, structural, and cellular assays, we show that two classes ofhighly selective inhibitors of IDO1 act by competing with hemebinding to apo-IDO1. This shows that IDO1 is dynamically boundto its heme cofactor in what is likely a critical step in the regula-tion of this enzyme. These results have elucidated a previouslyundiscovered role for the ubiquitous heme cofactor in immuneregulation, and it suggests that other heme proteins in biologymay be similarly regulated.

Author contributions: M.T.N., P.A.K., J.T.H., J.A.N., A.B., D.M., X.Z., L.A., A.A., R.B., H.A.L.,S.P.S., C.Y., and J.T.G. designed research; M.T.N., P.A.K., J.A.N., A.B., D.M., X.Z., A.A.,H.A.L., Z.L., S.P.S., and C.Y. performed research; M.T.N., P.A.K., J.T.H., J.A.N., A.B., D.M.,X.Z., L.A., A.A., R.B., H.A.L., Z.L., S.P.S., C.Y., and J.T.G. analyzed data; and M.T.N. andJ.T.G. wrote the paper.

Reviewers: E.L.R., University of Leicester; and S.-R.Y., Albert Einstein College of Medicine.

Conflict of interest statement: M.T.N., P.A.K., and J.T.G. declare no conflict of interest. X.Z.and L.A. are former employees of Bristol-Myers Squibb Co. J.T.H., J.A.N., A.B., D.M., A.A.,R.B., H.L., Z.L., S.P.S., and C.Y. are employees of Bristol-Myers Squibb Co.

This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

Data deposition: The atomic coordinates and structure factors have been deposited in theProtein Data Bank, www.wwpdb.org [PDB ID codes 6AZU (holoenzyme), 6AZV (1), and6AZW (3)].1To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1719190115/-/DCSupplemental.

Published online March 12, 2018.

www.pnas.org/cgi/doi/10.1073/pnas.1719190115 PNAS | March 27, 2018 | vol. 115 | no. 13 | 3249–3254

CHEM

ISTR

YBIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020

Page 2: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

bacterial infection, wherein IDO1 inhibition has been shown to aidin clearing the infection (21).The mechanism of IDO1 proceeds through a sequential oxygen

insertion reaction, where molecular oxygen binds to the ferrousheme and then adds across the C2-C3 double bond of tryptophanthrough an alkylperoxo intermediate to afford the epoxide withthe second atom of molecular oxygen left on the heme in a ferrylcompound II structure. This IDO1 ferryl is then proposed to at-tack the epoxide, breaking the oxygen–iron bond and reducing theiron back to the ferrous resting state (22–24). This surprising ferrylattack on an epoxide has not been fully explored, as it is not therate-determining step, but it is consonant with the recent rev-elations of the reactivity and versatility of compound II in otherenzymes (25–27).Typical of other heme proteins, IDO1 is vulnerable to com-

petitive inhibitors, such as substrate mimics or compounds withheme binding capabilities (28–31). This strategy, however, has suf-fered from a paucity of inhibitors capable of the specificity andnanomolar binding affinity desirable in a pharmaceutical (32). Ex-ceptions to this long-standing paradigm have been reported re-cently that bind to the enzyme without binding directly to theheme iron (33). We report here an examination of the uniqueinhibitory modes of two classes of inhibitors that display favor-able specificity and robust inhibition of IDO1 (HeLa cell IC50 =4.2 and 0.50 nM for compounds 1 and 2, respectively) (SI Ma-terials and Methods) (34).

Heme Cofactor of IDO1 Is LabileThe mechanism of IDO1 inhibition by compounds 1 and 2 (Fig.1A) was interrogated using activity assays with recombinanthuman IDO1. Compounds 1 and 2, the latter currently being in-vestigated in phase II clinical trials, are structurally disparate andlack any significant similarity to the substrate tryptophan or anyobvious heme binding moieties that could explain their mode ofaction (Fig. S1). In both cases, these compounds showed an in-triguingly slow onset of inhibition, which is inconsistent with asimple competitive mechanism (Fig. 1B). Both compounds alsoshowed a clear temperature dependence, only prompting in-hibition above 30 °C (Fig. 1C and Fig. S2).As a test for IDO1 heme loss as the key initiatory step of in-

hibition, IDO1 was incubated with apo-myoglobin, which is ca-pable of binding to free heme rapidly and essentially irreversibly,with a binding constant of 1014 M−1 (35). Remarkably, IDO1 withadded apo-myoglobin was seen to lose activity, interpreted as hemeloss to apo-myoglobin, with nearly the same time and temperaturedependence as it does with 1 and 2 (t1/2 = 17, 14, and 11 min,respectively). This behavior is in contrast to the steadily maintainedactivity of IDO1 incubated in the absence of inhibitor or apo-myoglobin, indicating that heme binding to IDO1 is a dynamic,reversible process (Fig. 1B).For a spectroscopic test of heme loss, IDO1 was incubated with

a modified version of sperm whale apo-myoglobin, H64YV68F,that, when bound to heme, possesses a unique green color (36). IDO1heme loss to apo-myoglobin can thus be conveniently monitored

A

N

OHN

X

H

H

YNH

O NH

NOH

O

1 2 X = Cl, Y = F3 X = CN, Y = H

C

0

40

80

120

12 20 28 36 44

% A

ctiv

ity o

f ID

O1

Not

Tre

ated

with

Inhi

bito

r

Temperature (°C) of Incubation with Inhibitors

Prior to Activity Assay

apomyoglobinCompound 1

Compound 2

B

0 10 20 30 400

0.37

0.75

1.1

1.5

Time (min) Incubated with InhibitorPrior to Activity Assay

apomyoglobin

Compound 1no inhibitorCompound 2

Act

ivity

of I

DO

1 af

ter

Incu

batio

n w

ith In

hibi

tor

(s-1)

Fig. 1. Time and temperature dependence of IDO1 inhibition. (A) Structure of three IDO1 inhibitors. (B) IDO1 (2.4 μM) was incubated with com-pounds 1 (20 μM), 2 (20 μM), or equine apomyoglobin (20 μM) at 37 °C for various times (x axis), after which this inhibition was tested using a standardactivity assay (y axis). (C ) IDO1 (2.4 μM) was incubated with compounds 1, 2, or equine apomyoglobin (20 μM) for 15 min at various temperatures(x axis), after which inhibition was tested using a standard activity assay following the rate of N-formylkynurenine production (y axis). Error barsrepresent SD. N ≥ 3.

0

0.4

0.8

1.2

300 400 500 600 700

Abs

orba

nce

Wavelength (nm)

0

0.04

500 600 700Wavelength (nm)

37 °C

40°C

30°C

25°C20°C15°C0

0.04

0.08

0 10 20 30 40

Nor

mal

ized

Abs

orba

nce

600

nm

Time (min)

A B

Fig. 2. Transfer of heme from IDO1 to apomyoglobin. (A) The UV-visiblespectra of heme transfer from IDO1 (5 μM) to apo-myoglobin H64YV68F(95 μM) over 40 min at 37 °C. (B) Heme dissociation from IDO1 (5 μM) to apo-myoglobin H64YV68F (95 μM) at various temperatures as indicated by theincrease in absorbance at 600 nm.

A

0

40

80

120

0 200 400

cpm

Time (min)

Compound 1 no cold chaseCompound 1 cold chase B

0

40

80

120

0 400 800 1200

cpm

Time (min)

Compound 2 no cold chaseCompound 2 cold chase

Fig. 3. Cold chase experiments with radiolabeled inhibitors. Radiolabeledversions of 1 (A) and 2 (B) were incubated with IDO1 followed by introductionof natural abundance 1 and 2, buffer exchange, and quantification of theresidual, IDO1-bound, 14C-labeled 1 and 2. Error bars represent SD. N = 2.

3250 | www.pnas.org/cgi/doi/10.1073/pnas.1719190115 Nelp et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020

Page 3: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

by an increase in absorbance at 600 nm. This confirmed that IDO1readily loses its heme cofactor (Fig. 2A and Fig. S3) and that thisunusual heme lability mirrors the temperature dependence of in-hibition by 1 and 2 (Fig. 2B). In this way, IDO1 heme loss was shownto be inhibitor-independent and likely the shared rate-determining

step of inhibition by 1 and 2, accounting for the striking similarity ofinhibitory profiles for these two distinct inhibitors.

1 and 2 Bind to IDO1 with Varying Off RatesThe mechanism by which these inhibitors exploit IDO1 hemelability was further explored using 14C-radiolabeled inhibitors incold chase experiments. To test whether and how strongly theseinhibitors bind to IDO1, the protein was incubated with 14C-labeled 1 and 2. These incubations then had natural abundanceversions of the inhibitors added, and at various times, the bufferand any unbound inhibitor were exchanged away from the proteinusing centrifugal concentrators. In this way, any residual inhibitormust be bound to IDO1 and can be quantified using its 14C ra-diation. This showed that IDO1 does bind to these compounds,but it does so with very large differences in off rate: t1/2 of 2 and50 min for 1 and 2, respectively (Fig. 3). This, despite nearly identicalinhibition kinetics, strongly supports heme loss to be the shared andrate-determining step of inhibition.

Inhibition Does Not Show a Linear Relationship withInhibitor ConcentrationThe concentration dependence of inhibition by 1 and 2 was thenexamined to illuminate any differences that may indicate a bindingevent before heme loss. In incubations preceding activity assays,the concentrations of the inhibitors, including apo-myoglobin, werevaried from 1 to 16 times that of IDO1, and despite these large dif-ferences, the rates of inhibition were nearly identical and certainlyfar from showing a linear relationship (Fig. 4A). These resultsshow that apo-IDO1 formed from intrinsic heme loss is the target

apo-myoglobinCompound 1Compound 2

A

0

0.4

0.8

1.2

0 10 20

Act

ivity

of I

DO

1 af

ter

Incu

batio

n w

ith In

hibi

tor

(s-1)

[Inhibitor]:[IDO1] in Incubation Prior to Activity Assay

B

0

0.4

0.8

1.2

0 10 20

Act

ivity

of I

DO

1 af

ter

Incu

batio

n w

ith In

hibi

tor

(s-1)

[Inhibitor]:[IDO1] in Incubation Prior to Activity Assay

Fig. 4. Concentration dependence of IDO1 inhibition. (A) IDO1 (2.4 μM)was incubated with various concentrations from 0 to 40 μM (x axis) ofcompounds 1, 2, or equine apomyoglobin at 37 °C, and for 15 min after,this inhibition was tested using a standard activity assay (y axis). (B) Theexperiment was repeated but with conditions that allow for turnover inthe incubation with IDO1 before the activity assay. In this case, IDO1 (2.4 μM)was incubated with 2.4–40 μM 1, 2, or equine apomyoglobin at 37 °C for15 min in the presence of 500 μM L-tryptophan, 10 mM ascorbate, 10 μMmethylene blue, and 10 μg/mL catalase. The rate of N-formylkynurenine pro-duction was then tested using a standard activity assay (y axis). Error barsrepresent SD. N ≥ 3.

Y126

S167

F214

H346

R343

F270

B

D

Y126

A264

F270

R343

H346F214

S167

C

AS263

A264

Fig. 5. Structure of IDO1–inhibitor complexes. (A) Superposition of the backbone atoms of the crystal structures of IDO1 holoenzyme (orange) over thatof IDO1/3 (blue). The overall structure of IDO1 undergoes little change on ligand binding. The loop corresponding to residues 260–265 is shifted, resultingin significant displacement (measured on Cα) of Ser-263 and Ala-264. Moreover, heme-coordinating residue His-346 is slightly rotated to interact with theligand. (B) Sphere representation of 1 overlaid with heme shows that they bind IDO1 in a mutually exclusive manner. Binding geometries are shown for 1in C and 3 in D. Structures were prepared from crystallographic coordinates using Maestro software to add hydrogens, determine protonation state ofamino acid side chains, and minimize heavy atom deviation to a maximum of 0.3 Å from their crystallographic coordinates. Helix S (residues 383–399) ishidden in C and D for clarity. Figs. S4 and S5 depict the pocket rendering of holo-IDO1 in comparison with inhibitor-bound structures (Fig. S4), the electrondensity maps of inhibitors 1 and 3 bound to apo-IDO1 (Fig. S5), and 2D ligand interaction maps for 1 and 3 (Fig. S5).

Nelp et al. PNAS | March 27, 2018 | vol. 115 | no. 13 | 3251

CHEM

ISTR

YBIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020

Page 4: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

for these inhibitors, which must act in some way to block hemefrom rebinding.

Inhibition Is Independent of IDO1 TurnoverThis mechanism predicts that inhibition is independent of activity.To further confirm this hypothesis, we incubated IDO1 with variousconcentrations of inhibitor and the natural substrate, L-tryptophan.Activity was then assessed subsequently. As with all activity exper-iments described herein, D-tryptophan was used as the substrate,which unlike L-tryptophan, does not cause substrate inhibitionand thus provides for a more convenient, repeatable means of de-termining activity (37–39). In these experiments, any L-tryptophannot converted to N-formylkynurenine will have been carried overfrom the incubations and acted as a substrate in addition to the10 mM D-tryptophan (maximum final concentration of 15 μM L-tryptophan with a 33× dilution from incubation with inhibitor toactivity assay solution). The inhibition profiles were nearly identical,regardless of turnover conditions during incubation with 1 and 2(Fig. 4B). This observation confirms that the mechanism of in-hibition is independent of turnover, supporting apo-IDO1 as thetarget for inhibition and its formation via heme dissociation as therate-limiting step for this process.

Crystal Structures of IDO1–Inhibitor Complexes Reveal TheirMode of ActionThe interaction of these inhibitors with IDO1 was clearly eluci-dated by X-ray crystallographic structures obtained from coc-rystallization with 1 and 3, the latter being a close analog of 2(Fig. 5). (Compound 1 is BMS-978587, 2 is BMS-986205, and 3 isBMS-116.) Both inhibitors bound in a manner that displaced theheme cofactor, with each occupying different, although overlapping,space in the vacated heme pocket (Figs. S4 and S5). Remarkably,the overall structure of IDO1 bound to each inhibitor was largelyunperturbed compared with a heme-containing IDO1 structure(0.61 and 0.44 Å rmsd from 2D0T for cocrystal structures with 1and 3, respectively). The carboxylate of 1 forms a hydrogen bondwith the backbone amide of Ala-264 and with His-346, whichordinarily coordinates with the heme iron on the proximal side.Binding of 1 also led to a shift in the flexible loop made of residues260–265, an event previously observed with the binding of phenyl-imidazole (40). The loop shift revealed the putative substratebinding site to the distal face of heme, where the phenylurea groupin 1 binds via edge-to-face π-interactions with Tyr-126 and hydrogenbonds with Ser-167. The quinoline of 3 occupies an additionalpocket made available by side-chain rearrangements of Phe-270,Phe-214, His-346, and Arg-343 that are stabilized by edge-to-faceπ-interactions with Phe-270 and a hydrogen bond with Arg-343.

Apo-IDO1 Is Present in CellsThe physiological relevance of apo-IDO1 and its potential as atarget for inhibition are supported by previous studies that haveshown that IDO1 exists in the apo-form within cells and is ca-pable of activation on addition of exogenous heme (41). To furthersupport apo-IDO1 as the authentic target of 1 and 2, additionalcellular assays were performed under conditions in which IDO1was overexpressed but heme concentrations were varied. Briefly,human ovarian cancer cells were treated with IFN-γ to induceIDO1 expression. The cells were then tested for IDO1 activity afteraddition of the ribosomal inhibitor cycloheximide to prevent in-terference from newly translated IDO1. Cellular IDO1 activity wasthen assessed after further incubation in the presence or absence ofadded heme. Despite evidence that IDO1 protein levels weresimilar (Fig. S6), the activity of those cells with added heme wasapproximately fivefold higher than that of cells not treated withadditional heme, indicating that at least 85% of IDO1 exists in

Fig. 6. Kynurenine production by human SKOV3 ovarian tumor cells. Leftdepicts results from cells stimulated for 24 h with IFN-γ in the absence ofadded hemin. Right depicts cells stimulated with IFN-γ in the presence of40 μM hemin. Blue bars depict kynurenine production after 24 h of IFN-γstimulation. Cells were washed for 1 h with fresh media containing cyclo-heximide to halt protein synthesis. Kynurenine production was measured24 h after addition of fresh media devoid of heme (pink) or containing40 μM hemin (dark blue). Error bars represent SD.

A

B

Fig. 7. Redox state dependence of heme dissociation from IDO1. IDO1 (1.1 μM)was incubated for 40 min at 37 °C in the presence of 10 μM 2. Heme disso-ciation from IDO1 was tracked by the loss of absorbance at the IDO1 λmax

of the Soret peak: 404 nm for ferric IDO1 and 425 nm for ferrous IDO1.(A) Spectra of ferric IDO1 at various times. Inset shows a comparison of theloss in absorbance for both ferric and ferrous IDO1 as a function of time.(B) Spectra of ferrous IDO1 at various times. IDO1 was reduced using 5 mMsodium dithionite in a nitrogen-flushed sealed cuvette, and the decay ofdithionite can be seen by its decreasing absorption below 375 nm. Inset is ascheme showing inhibitor binding to IDO1 in place of heme, causing theobserved reduction in absorbance.

3252 | www.pnas.org/cgi/doi/10.1073/pnas.1719190115 Nelp et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020

Page 5: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

these cells in an apo-form that is capable of activation by exogenouslyadded heme (Fig. 6). Thus, the target of these inhibitors as revealedin the crystal structures is confirmed to be not only physiologicallyrelevant but also, the predominant form of this enzyme in these cells.Having established that 1 and 2 compete with heme for apo-

IDO1, it was of interest to determine the effect of added heme inthe inhibition assays. When 40 μM heme was added to HeLa cellsconcurrently with stimulation of the cells with IFN-γ and inhibitionwas measured after 20 h of incubation, IC50 values were shifted lessthan twofold compared with the same experiment with no addedheme (1, IC50 ± heme = 7.1/4.2 nM; 2, IC50 ± heme = 1.1/0.5 nM).The lack of a dramatic heme-dependent potency shift for com-pound inhibition during the prolonged induction period couldresult from direct competition for newly synthesized apo-IDO1between inhibitor and heme, whereby the higher affinity and/orslower off rate of the inhibitors compensate for the higher hemeconcentration.

Heme Lability Is Dependent on Redox StateThe cellular results show that IDO1 heme loss is a major factorin its normal regulation. We found that IDO1 heme lability isstrongly dependent on the redox status of iron in the heme co-factor. Using compound 2 to capture apo-IDO1 as it forms fromheme loss, we found that ferrous IDO1, which is the resting stateof the enzyme in its active form, binds at least 10-fold more tightlyto heme than enzyme does in the ferric state, which is catalyticallyinactive (Fig. 7 and Fig. S7). This was measured by fitting the lossin absorbance at the corresponding Soret maxima to a single ex-ponential and comparing the half-lives. The kinetics of this lossshow that this process is complex and may involve multiple statesof heme binding, which will require additional study. The overalldifference in heme retention, however, is significant.A stretch of 20 amino acids missing from electron density of all

IDO1 structures has been predicted to form a “closed” state onsubstrate binding, effectively blocking the solvent-exposed face ofheme and introducing an intermolecular hydrogen bond with aheme propionate (42). It is, therefore, plausible that intracellulartryptophan concentrations also play a role in heme recruitmentand retention in IDO1. We tested heme loss from IDO1 to apo-myoglobin H64YV68F in the presence of 0–10 mM L-tryptophan(the Kd of L-tryptophan and ferric IDO1 is reported to be 5.8 mM)and found that the rate of heme loss was seen to decrease withincreasing substrate concentration (Fig. S8) (37). This effect onferric IDO1 may not be relevant, as intracellular concentrations oftryptophan are unlikely to approach this Kd (43); however, the Kdof ferrous IDO1 and L-tryptophan is significantly lower, 13 μM

(37), and therefore, it is possible that tryptophan exerts this hemeretention effect more strongly on the active, ferrous form of IDO1.

DiscussionThe goal of effective and specific inhibition of IDO1 has beenintensively pursued since the discovery that this enzyme is criticalto cancer’s evasion of immune responses (17). Whereas most in-hibitors to date act as competitive inhibitors either by mimickingthe substrate tryptophan or by binding to the heme cofactor (28), 1and 2 are capable of inhibiting IDO1 by taking advantage of anentirely separate strategy of competing with the heme cofactoritself in binding to the apo-form of the enzyme (Fig. 8). In theeffort to understand the mechanism of this inhibition, the im-portance of heme lability in IDO1 and its potential as a regulatorymechanism have been more fully revealed.The formation of such a large pool of apo-IDO1 is still not fully

understood, although it has previously been shown that IDO2, ahomolog of IDO1, is capable of down-regulating the activity ofIDO1 through heme transfer and sequestration (44), and it isknown that there is significant cross-talk between IDO1 and hemecatabolism through heme oxygenase-1, leading to loss of IDO1activity via heme starvation (45). It has also been shown thatferrous IDO1 can bind nitric oxide in the absence of tryptophan,inducing rupture of the heme iron-proximal histidine bond to formapo-IDO1 (46). This transformation bears similarity to the hemereceptor of soluble guanylyl cyclase that, although it is part of asignaling process and is not catalytic, uses heme lability in itsregulation and is a target for compounds capable of binding to itsapo-form (47, 48).We have here shown that heme lability is likely to play a signifi-

cant role in posttranslational control of IDO1 activity in cells, andthe fact that this lability is strongly dependent on the redox state ofthe heme provides a potential regulatory mechanism to controlIDO1 activity. Raven and coworkers (49) have shown that IDO1accumulates in the catalytically inactive ferric state during turnoverwhen tryptophan concentrations are low in vitro, and IDO1 is ca-pable of autoxidizing to the ferric state in vivo (50). Together, thesedata suggest that IDO1 is self-regulating; under low tryptophan con-ditions, activity can decrease immediately by conversion to the cata-lytically inactive ferric state from which it can proceed to the apo-formthrough heme loss (Fig. 8). Such a process would contribute to thelarge pool of apo-IDO1 that serves as the target for these inhibitors.Additionally, the complexity seen in the heme loss kinetics, a

fast phase followed by a slow phase evident in both assays of hemeloss to apomyoglobin H64YV68F and heme loss in the presence of2 (Figs. 2 and 7), suggests that a simple model of only holo- andapo-IDO1 may not fully encompass this heme loss phenomenon

Suppression

Fig. 8. Catalytic mechanism of tryptophan catabolism by IDO1 and its inhibition via heme dissociation.

Nelp et al. PNAS | March 27, 2018 | vol. 115 | no. 13 | 3253

CHEM

ISTR

YBIOCH

EMISTR

Y

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020

Page 6: Immune-modulating enzyme indoleamine 2,3 …form. We show that the in vitro kinetics of inhibition coincide with an unusually high rate of intrinsic enzyme–heme dissocia-tion, especially

and that there may be intermediate states of heme binding thatwill require additional study.In conclusion, we have shown that IDO1 readily loses heme in a

dynamic, reversible, and oxidation state-dependent manner. Tar-geting the pool of apo-IDO1 provides a means by which a class ofinhibitors is capable of effectively inhibiting IDO1 with the potentialto restore normal immune clearing of cancerous cells. These resultsalso reveal more fully the intricate metabolic control that can beachieved by modulating the affinity of IDO1 for its heme cofactorand suggest that many other such enzymes could be similarly reg-ulated and mechanistically rich.

Materials and MethodsCompounds 1, 2, and 3 were prepared at Bristol-Myers Squibb Co. by pro-cedures described in published patent applications (51, 52). The 14C-radio-labeled versions of 1 and 2 were provided by Bristol-Myers Squibb Co.

Protein was expressed and purified using standard methods. Cell-freeactivity assays were performed using standard methods with an Agilent8453 diode array spectrometer and temperature-controlled Fisher Scientificisotemp 1016s recirculating chiller. Cold chase experiments using 14C-radiolabeled 1 and 2 were performed as described in SI Materials andMethods. Crystallization conditions used to obtain inhibitor-bound andholo-IDO1 are described in SI Materials and Methods. Cell-based activityassays using HeLa and SKOV3 cells were performed as described in SIMaterials and Methods.

ACKNOWLEDGMENTS. We thank Dianlin Xie and Mian Gao for constructionof expression vectors; Frank Marsilio, Susan E. Kiefer, and Nicolas Szapiel forearly IDO expression and purification work; Yuval Blat, Hao Lu, and LitaiZhang for helpful discussions; Kathy Johnston and Joseph Naglich for assaydesign in Hela cells; and Alfred Lammens and Stefan Steinbacher (ProterosBiostructures GmbH) for cocrystal structural work with 3. P.A.K. thanks BMS,Inc. for fellowship support. Support of this work was provided by NIH Grant2R37 GM036298 (to J.T.G.).

1. Munn DH, Mellor AL (2013) Indoleamine 2,3 dioxygenase and metabolic control ofimmune responses. Trends Immunol 34:137–143.

2. Yamamoto S, Hayaishi O (1967) Tryptophan pyrrolase of rabbit intestine. D- and L-tryptophan-cleaving enzyme or enzymes. J Biol Chem 242:5260–5266.

3. Suzuki T, et al. (2003) Comparison of the sequences of Turbo and Sulculus indole-amine dioxygenase-like myoglobin genes. Gene 308:89–94.

4. Yamazaki F, Kuroiwa T, Takikawa O, Kido R (1985) Human indolylamine 2,3-dioxygenase. Itstissue distribution, and characterization of the placental enzyme. Biochem J 230:635–638.

5. Taylor MW, Feng GS (1991) Relationship between interferon-gamma, indoleamine2,3-dioxygenase, and tryptophan catabolism. FASEB J 5:2516–2522.

6. Däubener W, et al. (2001) Restriction of Toxoplasma gondii growth in human brainmicrovascular endothelial cells by activation of indoleamine 2,3-dioxygenase. InfectImmun 69:6527–6531.

7. Curti A, Trabanelli S, Salvestrini V, Baccarani M, Lemoli RM (2009) The role of in-doleamine 2,3-dioxygenase in the induction of immune tolerance: Focus on hema-tology. Blood 113:2394–2401.

8. Munn DH, et al. (1998) Prevention of allogeneic fetal rejection by tryptophan ca-tabolism. Science 281:1191–1193.

9. van Baren N, Van den Eynde BJ (2015) Tryptophan-degrading enzymes in tumoralimmune resistance. Front Immunol 6:34.

10. Munn DH, et al. (1999) Inhibition of T cell proliferation by macrophage tryptophancatabolism. J Exp Med 189:1363–1372.

11. Metz R, et al. (2012) IDO inhibits a tryptophan sufficiency signal that stimulatesmTOR: A novelIDO effector pathway targeted by D-1-methyl-tryptophan. OncoImmunology 1:1460–1468.

12. Keskin DB, Marshall B, Munn D, Mellor AL, Gearhart DA (2007) Decreased protein nitrationinmacrophages that overexpress indoleamine 2, 3-dioxygenase. Cell Mol Biol Lett 12:82–102.

13. Platten M, et al. (2005) Treatment of autoimmune neuroinflammation with a syn-thetic tryptophan metabolite. Science 310:850–855.

14. Wang Y, et al. (2010) Kynurenine is an endothelium-derived relaxing factor producedduring inflammation. Nat Med 16:279–285.

15. Favre D, et al. (2010) Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 altersthe balance of TH17 to regulatory T cells in HIV disease. Sci Transl Med 2:32ra36.

16. Munn DH, et al. (2002) Potential regulatory function of human dendritic cells ex-pressing indoleamine 2,3-dioxygenase. Science 297:1867–1870.

17. Uyttenhove C, et al. (2003) Evidence for a tumoral immune resistance mechanism basedon tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 9:1269–1274.

18. Muller AJ, et al. (2008) Chronic inflammation that facilitates tumor progression cre-ates local immune suppression by inducing indoleamine 2,3 dioxygenase. Proc NatlAcad Sci USA 105:17073–17078.

19. Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC (2005) In-hibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancersuppression gene Bin1, potentiates cancer chemotherapy. Nat Med 11:312–319.

20. Munn DH, Mellor AL (2016) IDO in the tumor microenvironment: Inflammation,counter-regulation, and tolerance. Trends Immunol 37:193–207.

21. Gautam US, et al. (2018) In vivo inhibition of tryptophan catabolism reorganizes thetuberculoma and augments immune-mediated control of Mycobacterium tubercu-losis. Proc Natl Acad Sci USA 115:E62–E71.

22. Lewis-Ballester A, et al. (2009) Evidence for a ferryl intermediate in a heme-baseddioxygenase. Proc Natl Acad Sci USA 106:17371–17376.

23. Basran J, et al. (2011) The mechanism of formation of N-formylkynurenine by hemedioxygenases. J Am Chem Soc 133:16251–16257.

24. Huang X, Groves JT (December 29, 2017) Oxygen activation and radical transformationsin heme proteins and metalloporphyrins. Chem Rev, 10.1021/acs.chemrev.7b00373.

25. Basran J, Booth ES, Lee M, Handa S, Raven EL (2016) Analysis of reaction intermedi-ates in tryptophan 2,3-dioxygenase: A comparison with indoleamine 2,3-dioxygenase.Biochemistry 55:6743–6750.

26. Wang X, Ullrich R, Hofrichter M, Groves JT (2015) Heme-thiolate ferryl of aromaticperoxygenase is basic and reactive. Proc Natl Acad Sci USA 112:3686–3691.

27. Moody PCE, Raven EL (January 12, 2018) The nature and reactivity of ferryl heme incompounds I and II. Acc Chem Res, 10.1021/acs.accounts.7b00463.

28. Röhrig UF, Majjigapu SR, Vogel P, Zoete V, Michielin O (2015) Challenges in the dis-covery of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. J Med Chem 58:9421–9437.

29. Lewis-Ballester A, et al. (2017) Structural insights into substrate and inhibitor bindingsites in human indoleamine 2,3-dioxygenase 1. Nat Commun 8:1693.

30. Malachowski WP, et al. (2016) O-alkylhydroxylamines as rationally-designed mechanism-based inhibitors of indoleamine 2,3-dioxygenase-1. Eur J Med Chem 108:564–576.

31. Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ (2017) Discovery ofIDO1 inhibitors: From bench to bedside. Cancer Res 77:6795–6811.

32. Markwalder JA, et al. (2017) Identification and optimization of a novel series of in-doleamine 2,3-dioxygenase inhibitors. Bioorg Med Chem Lett 27:582–585.

33. Crosignani S, et al. (2017) Discovery of a novel and selective indoleamine 2,3-dioxygenase(IDO-1) inhibitor 3-(5-Fluoro-1H-indol-3-yl)pyrrolidine-2,5-dione (EOS200271/PF-06840003)and its characterization as a potential clinical candidate. J Med Chem 60:9617–9629.

34. Williams DK, et al. (2018) Development of a series of novel o-phenylenediamine-basedindoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. Bioorg Med Chem Lett 28:732–736.

35. Hargrove MS, Barrick D, Olson JS (1996) The association rate constant for hemebinding to globin is independent of protein structure. Biochemistry 35:11293–11299.

36. Hargrove MS, et al. (1994) His64(E7) → Tyr apomyoglobin as a reagent for measuringrates of hemin dissociation. J Biol Chem 269:4207–4214.

37. Sono M, Taniguchi T, Watanabe Y, Hayaishi O (1980) Indoleamine 2,3-dioxygenase.Equilibrium studies of the tryptophan binding to the ferric, ferrous, and CO-boundenzymes. J Biol Chem 255:1339–1345.

38. Lu C, Lin Y, Yeh SR (2009) Inhibitory substrate binding site of human indoleamine 2,3-dioxygenase. J Am Chem Soc 131:12866–12867.

39. Efimov I, et al. (2012) The mechanism of substrate inhibition in human indoleamine2,3-dioxygenase. J Am Chem Soc 134:3034–3041.

40. Sugimoto H, et al. (2006) Crystal structure of human indoleamine 2,3-dioxygenase:Catalytic mechanism of O2 incorporation by a heme-containing dioxygenase. ProcNatl Acad Sci USA 103:2611–2616.

41. Thomas SR, et al. (2001) Antioxidants inhibit indoleamine 2,3-dioxygenase in IFN-γ-activated human macrophages: Posttranslational regulation by pyrrolidine di-thiocarbamate. J Immunol 166:6332–6340.

42. Álvarez L, et al. (2016) Structural study of a flexible active site loop in human indole-amine 2,3-dioxygenase and its functional implications. Biochemistry 55:2785–2793.

43. Heidemann R, Lütkemeyer D, Büntemeyer H, Lehmann J (1998) Effects of dissolved oxygenlevels and the role of extra- and intracellular amino acid concentrations upon themetabolismof mammalian cell lines during batch and continuous cultures. Cytotechnology 26:185–197.

44. Metz R, et al. (2014) IDO2 is critical for IDO1-mediated T-cell regulation and exerts anon-redundant function in inflammation. Int Immunol 26:357–367.

45. Hill M, et al. (2005) Heme oxygenase-1 inhibits rat and human breast cancer cellproliferation: Mutual cross inhibition with indoleamine 2,3-dioxygenase. FASEB J 19:1957–1968, and erratum (2006) 20:1573.

46. Samelson-Jones BJ, Yeh SR (2006) Interactions between nitric oxide and indoleamine2,3-dioxygenase. Biochemistry 45:8527–8538.

47. Martin F, et al. (2010) Structure of cinaciguat (BAY 58-2667) bound to Nostoc H-NOXdomain reveals insights into heme-mimetic activation of the soluble guanylyl cyclase.J Biol Chem 285:22651–22657.

48. Fritz BG, et al. (2011) Oxidation and loss of heme in soluble guanylyl cyclase fromManduca sexta. Biochemistry 50:5813–5815.

49. Booth ES, Basran J, Lee M, Handa S, Raven EL (2015) Substrate oxidation by indole-amine 2,3-dioxygenase: Evidence for a common reaction mechanism. J Biol Chem 290:30924–30930.

50. Vottero E, et al. (2006) Cytochrome b(5) is a major reductant in vivo of human in-doleamine 2,3-dioxygenase expressed in yeast. FEBS Lett 580:2265–2268.

51. Balog JA, et al. (2014) Patent Cooperation Treaty Int Appl WO 2014150677.52. Beck HP, et al. (2016) Patent Cooperation Treaty Int Appl WO 2016073774.

3254 | www.pnas.org/cgi/doi/10.1073/pnas.1719190115 Nelp et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 2

0, 2

020


Recommended