+ All Categories
Home > Documents > Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric...

Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric...

Date post: 10-Aug-2019
Category:
Upload: dodiep
View: 217 times
Download: 0 times
Share this document with a friend
17
Allosteric Modulator Discovery: From Serendipity to Structure- Based Design Shaoyong Lu, ,,§ Xinheng He, ,§ Duan Ni, ,§ and Jian Zhang* ,,Key Laboratory of Cell Dierentiation and Apoptosis of Chinese Ministry of Education, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China ABSTRACT: Allosteric modulators bound to structurally diverse allosteric sites can achieve better pharmacological advantages than orthosteric ligands. The discovery of allosteric modulators, however, has been traditionally serendipitous, owing to the complex nature of allosteric modulation. Recent advances in the understanding of allosteric regulatory mechanisms and remarkable availability of structural data of allosteric proteins and modulators, as well as their complexes, have greatly contributed to the development of various computational approaches to guide the structure-based discovery of allosteric modulators. This Perspective rst outlines the evolution of the allosteric concept and describes the advantages and hurdles facing allosteric modulator discovery. The current available computational approaches, together with experimental approaches aiming to highlight allosteric studies, are then highlighted, emphasizing successful examples with their combined applications. We aimed to increase the awareness of the feasibility of the structure-based discovery of allosteric modulators using an integrated computational and experimental paradigm. 1. INTRODUCTION Allostery is an inherent property of biomacromolecules in which topographically distinct binding sites within a protein are functionally coupled and can communicate over a distance. 13 Typically, the allosteric signal propagation from allosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as eector (ions, small molecules, proteins, DNA, and RNA) binding, point mutations, and posttranslational modications. 4,5 The exis- tence of allosteric modulation in the control of biomacromo- lecule function allows for exquisite control of innumerable biological processes, ranging from enzyme catalysis, gene expression, and cellular dierentiation to metabolism and homeostasis. 610 Because of the omnipresence of allosteric control over normal cellular processes, allosteric modulation has thus been regarded as the second secret of life. 11 This notion has been well established as a fundamental biological phenomenon for insights into cellular functions and dis- eases. 5,12 From a pharmaceutical standpoint, protein allostery provides a promising, novel opportunity for the development of innovative therapeutics. Owing to the nonoverlap of allosteric and orthosteric sites, allosteric modulators, by attaching to allosteric sites, did not compete with endogenous or exogenous ligands bound to orthosteric sites. This feature guarantees the cooperative regulation of protein function by both allosteric modulators and orthosteric ligands in individual proteins. 1315 Moreover, the much more diversied allosteric sites relative to the highly conserved orthosteric sites endow allosteric modulators with potentially enhanced selectivity and reduced toxicity than orthosteric ligands. 1618 Despite the potential advantages of allosteric therapeutics, allosteric modulator discovery has encountered a key challenge. Over the years, most allosteric modulators have been discovered serendipitously by high-throughput screening experiments. 19 Such unexpected ndings manifest a lack of a comprehensive understanding of allosteric interactions and their eects on the molecular mechanism of proteins. Fortunately, structural data pertaining to allosteric proteins and their allosteric sites and modulators have become more available in the past few years. 20 Such data are benecial to develop computational approaches to predict allosteric interactions and mechanisms and subsequently screen allosteric modulators for the intended allosteric sites. Mounting evidence suggests that allosteric modulators can be Received: November 11, 2018 Published: February 28, 2019 Perspective pubs.acs.org/jmc Cite This: J. Med. Chem. XXXX, XXX, XXX-XXX © XXXX American Chemical Society A DOI: 10.1021/acs.jmedchem.8b01749 J. Med. Chem. XXXX, XXX, XXXXXX Downloaded via UNIV LEIPZIG on March 11, 2019 at 11:55:39 (UTC). See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Transcript
Page 1: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

Allosteric Modulator Discovery: From Serendipity to Structure-Based DesignShaoyong Lu,†,‡,§ Xinheng He,†,§ Duan Ni,†,§ and Jian Zhang*,†,‡

†Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Clinical and Fundamental ResearchCenter, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China‡Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China

ABSTRACT: Allosteric modulators bound to structurally diverse allosteric sites can achieve better pharmacological advantagesthan orthosteric ligands. The discovery of allosteric modulators, however, has been traditionally serendipitous, owing to thecomplex nature of allosteric modulation. Recent advances in the understanding of allosteric regulatory mechanisms andremarkable availability of structural data of allosteric proteins and modulators, as well as their complexes, have greatlycontributed to the development of various computational approaches to guide the structure-based discovery of allostericmodulators. This Perspective first outlines the evolution of the allosteric concept and describes the advantages and hurdlesfacing allosteric modulator discovery. The current available computational approaches, together with experimental approachesaiming to highlight allosteric studies, are then highlighted, emphasizing successful examples with their combined applications.We aimed to increase the awareness of the feasibility of the structure-based discovery of allosteric modulators using anintegrated computational and experimental paradigm.

1. INTRODUCTION

Allostery is an inherent property of biomacromolecules inwhich topographically distinct binding sites within a proteinare functionally coupled and can communicate over adistance.1−3 Typically, the allosteric signal propagation fromallosteric to functional, orthosteric sites is elicited byperturbations at the allosteric site such as effector (ions,small molecules, proteins, DNA, and RNA) binding, pointmutations, and posttranslational modifications.4,5 The exis-tence of allosteric modulation in the control of biomacromo-lecule function allows for exquisite control of innumerablebiological processes, ranging from enzyme catalysis, geneexpression, and cellular differentiation to metabolism andhomeostasis.6−10 Because of the omnipresence of allostericcontrol over normal cellular processes, allosteric modulationhas thus been regarded as the “second secret of life”.11 Thisnotion has been well established as a fundamental biologicalphenomenon for insights into cellular functions and dis-eases.5,12

From a pharmaceutical standpoint, protein allostery providesa promising, novel opportunity for the development ofinnovative therapeutics. Owing to the nonoverlap of allostericand orthosteric sites, allosteric modulators, by attaching toallosteric sites, did not compete with endogenous or exogenous

ligands bound to orthosteric sites. This feature guarantees thecooperative regulation of protein function by both allostericmodulators and orthosteric ligands in individual proteins.13−15

Moreover, the much more diversified allosteric sites relative tothe highly conserved orthosteric sites endow allostericmodulators with potentially enhanced selectivity and reducedtoxicity than orthosteric ligands.16−18

Despite the potential advantages of allosteric therapeutics,allosteric modulator discovery has encountered a keychallenge. Over the years, most allosteric modulators havebeen discovered serendipitously by high-throughput screeningexperiments.19 Such unexpected findings manifest a lack of acomprehensive understanding of allosteric interactions andtheir effects on the molecular mechanism of proteins.Fortunately, structural data pertaining to allosteric proteinsand their allosteric sites and modulators have become moreavailable in the past few years.20 Such data are beneficial todevelop computational approaches to predict allostericinteractions and mechanisms and subsequently screenallosteric modulators for the intended allosteric sites.Mounting evidence suggests that allosteric modulators can be

Received: November 11, 2018Published: February 28, 2019

Perspective

pubs.acs.org/jmcCite This: J. Med. Chem. XXXX, XXX, XXX−XXX

© XXXX American Chemical Society A DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

Dow

nloa

ded

via

UN

IV L

EIP

ZIG

on

Mar

ch 1

1, 2

019

at 1

1:55

:39

(UT

C).

Se

e ht

tps:

//pub

s.ac

s.or

g/sh

arin

ggui

delin

es f

or o

ptio

ns o

n ho

w to

legi

timat

ely

shar

e pu

blis

hed

artic

les.

Page 2: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

discovered in conjunction with computational and exper-imental methods, indicative of the advent of structure-basedallosteric modulator discovery.21−27

In this Perspective, we briefly overview the evolution of theallosteric concept and describe the advantages and hurdlesfacing allosteric modulator discovery. Importantly, we elucidatethe currently available computational methods that comple-ment experimental methods and provide powerful tools tofacilitate allosteric modulator discovery. We mainly focus onthe identification of allosteric modulators guided by computa-tional methods, followed by experimental validations. Weoutline some guidelines to increase the awareness of thestructure-based discovery of allosteric modulators using anintegrated computational and experimental paradigm. Finally,we discuss the future directions that can contribute to allostericmodulator discovery.

2. PAST AND PRESENT OF THE ALLOSTERICCONCEPT

The pioneering work by Bohr in 1904 uncovered an allostericphenomenon where the binding of carbon dioxide influencesthe binding affinity of oxygen to hemoglobin. This well-recognized phenomenon describing the cooperative binding ofmolecules to two distinct sites is known as the “Bohr effect”,which is currently named the “allosteric effect”.5

In 1961, Monod and Jacob first proposed the concept ofallostery to account for the mechanism of feedback inhibitionof enzyme activity in which “the inhibitor is not a stericanalogue of the substrate”.28 Later, in the 1960s, two classicmodels of allosteric interactions were created to explain thecooperative binding of regulatory molecules; one is the“concerted” or MWC (Monod−Wyman−Changeux)(1965)29 model, and the other is the “sequential” or KNF(Koshland−Nemethy−Filmer) (1966)30 model. Both modelsunderscore the role of conformational changes in the allostericregulation of two end-state structures.In 1984, Cooper and Dryden proposed a theoretical model

named “dynamics-driven allostery” by which allostery occurswithout significant conformational changes in the averagestructure, revealing the importance of the entropic contribu-tion to allostery.31 Advances in solution nuclear magneticresonance (NMR) spectroscopy32 have substantially contrib-uted to probe “dynamics/entropy-driven allostery” through thecharacterization of protein internal motions such as in thecatabolite activator protein (CAP),33,34 PDZ domains,35

calmodulin,36−38 and protein kinase A.39 Dramatically,Frederick et al. found a linear relationship between the proteinconformational entropy and change in the overall bindingentropy,37 revealing the important role of conformationalentropy in the recognition of proteins.40 They furtherdeveloped an empirical, quantitative method to describe therelationship between changes in conformational dynamicscharacterized by the motion of side chains and changes inprotein conformational entropy.41,42

In 1999, Nussinov and co-workers proposed a “conforma-tional selection and population shift” model that explains howallostery works from the viewpoint of free energy landscapetheory.43−45 This model broadens the concept of allosteryfrom two structural states to conformational ensembles ofmultiple states. In the same year, Ranganathan and co-workersproposed an “allosteric networks” model by which signaltransmission between distinct sites of proteins is achieved by anetwork of physically interconnected residues.46 This struc-

tural view of allostery has been demonstrated by numerousstudies using high-resolution structures.47−50

Recently, Hilser and co-workers have proposed an“ensemble allosteric” model to explain the origin ofallostery.51−53 The Hilser’s model is similar to the Nussinov’smodel in the explanation of the allosteric origin. Both modelsassume that all possible conformations of a protein ensembleare populated in terms of their respective energies, and thebinding of allosteric effectors to the protein reshapes the freeenergy landscape. However, Hilser’s model further extendsNussinov’s model to account for allostery in intrinsicallydisordered proteins; the former provides a framework thatunifies the illustrations of allostery in structured, dynamic, anddisordered systems.As mentioned above, allosteric concepts and models have

advanced over 50 years since the word “allostery” was coinedby Monod and Jacob.28 It is obvious that the concept ofallostery has evolved from the two-state model, the staticstructural model to the dynamic ensemble model.54−56

Importantly, some models have been applied to developcomputational methods to predict allosteric sites andmutations, discover allosteric modulators, assess allostericinteractions, and investigate allosteric mechanisms, all ofwhich will be elaborated in the following sections.

3. ADVANTAGES AND CHALLENGES FACINGALLOSTERIC MODULATOR DISCOVERY

3.1. Advantages. Allosteric modulators harbor severaldistinct advantages over orthosteric ligands that occupy aprotein’s active site. The advantageous characteristics ofallosteric modulators can be witnessed by their high specificityand low adverse effects.57−59 G-protein-coupled receptors(GPCRs) and protein kinases are the two most important drugtargets in therapeutic pharmacology. The highly evolutionarilyconserved endogenous orthosteric binding sites represent a keychallenge to develop selective orthosteric drugs for GPCRs andprotein kinases. It is recognized that orthosteric ligands of anintended GPCR or protein kinase frequently suffer from cross-reactivities with its homologous proteins, leading to unwantedside effects and off-target toxicity.16,60 As an alternative,targeting the structural diversity and topological difference ofallosteric sites enables one to surmount two major obstaclesencountered by orthosteric ligands.61,62 For instance, pyr-azolopyridone-based compound 10 (1) bound to the allostericsite at the C-lobe of the kinase exhibits excellent selectivity forp38α (IC50 = 1.2 μM) against three closely related structures,p38β, p38γ, and p38δ (IC50 > 40 μM) (Figure 1), due to theresidue differences within this site.63

Allosteric modulators have a spatiotemporal basis inspecificity. They, together with endogenous ligands, exerttheir cooperative actions by the fine-tuned modulation of theprotein function rather than by shutting off or turning onendogenous physiological signaling.13 This feature can result inincreased safety in the event of an overdose of allostericdrugs.16

Importantly, allosteric modulators have the potential tocombat drug-resistant mutations situated at orthosteric sites inpatients. For example, the “gatekeeper mutation” T315I ofBCR-ABL1 causes resistance to a set of clinically approvedorthosteric drugs such as imatinib, bosutinib, nilotinib, anddasatinib.64 Treatment of the T315I mutant with an allostericinhibitor ABL001 (asciminib) bound to the myristoyl pocket

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

B

Page 3: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

at the C-lobe of the kinase can overcome drug resistance(Figure 2).65,66

A protein exists in an ensemble consisting of numerous,distinct conformational states that may be involved in multiplesignaling pathways.9,67 An allosteric modulator binding to aunique conformation of the protein can achieve biasedsignaling.13 For example, G-protein- or β-arrestin-mediateddownstream signaling cascades of GPCRs can be activated bydifferent conformational states of receptor−ligand com-plexes.68

Remarkably, allosteric regulation also provides a new avenuefor drugging “undruggable” targets that are difficult to targetpharmacologically.69,70 This category of proteins is intractableto drugs because of strong substrate binding, the lack of deepbinding sites, or large and flat protein−protein interaction(PPI) interfaces.71,72 Under these circumstances, allostericmodulators can bypass direct competition with orthosteric sitesor PPI interfaces and bind alternative sites to fine-tune proteinactivities. Furthermore, cryptic (or hidden) allosteric sites canbe captured from the minor or intermediate conformations of

the protein and exploited for drug design, if the evidentbinding sites are invisible in the solved crystal structures.73−75

Currently, many previously considered “undruggable” targetshave proven to be treatable through allosteric regulatorymodules, including K-Ras,76,77 transcription factors (MYC78

and nuclear factor-κB79), the SH2-kinase domain PPI interfaceof BCR-ABL,80 phosphatases (SHP2,81 PTP1B,82 andPP2A83), hypoxia inducible factor-2,84 and STAT3.85

The explosive growth in the number of allosteric modulatorsin recent years has demonstrated advantages to the explorationof allostery in drug discovery.86 However, allosteric modulatordiscovery also poses significant challenges, which may berelieved by the deeper understanding of the molecular detailsof allosteric mechanisms and structural and biochemicalproperties of allosteric proteins and modulators.

3.2. Challenges. A key step for allosteric modulatordiscovery is the identification of bona fide allosteric sites in aprotein. Unlike orthosteric ligands that occupy a conserved,known orthosteric site, allosteric sites are less evolutionallyconserved than orthosteric sites (vide infra).87,88 Theoretically,any binding pocket in a protein spatially distinct from theorthosteric site can be regarded as a latent allosteric site.Moreover, the regulatory effect of the latent allosteric site onthe orthosteric site is difficult to determine. These disadvan-tages have severely impeded the allosteric modulator discoveryprocess over a long period. Fortunately, the ample availabilityof structural complexes of allosteric protein−modulatorinteraction provided by X-ray crystallography and NMRspectroscopy in recent years has partially addressed thisquandary.20 Many computational methods (see below) havebeen developed to predict allosteric sites based on thecharacteristics of known allosteric sites and the underlyingmechanisms of allosteric regulation,89,90 facilitating thestructure-based discovery of allosteric modulators.Challenges can also stem from the emergence of drug-

resistant mutations at allosteric sites, the same situation facedby orthosteric drugs. For example, in the case of Ba/F3 celllines expressing BCR-ABL1 variants, single-point mutations(A337V, P465S, V468F, and I502L) at the allosteric myristoylsite in the C-lobe of the kinase confer resistance to theallosteric inhibitor ABL001 (Figure 3).65 Furthermore, acombination of ABL001 and an orthosteric drug nilotinibcan inhibit the mutant more effectively than each agentalone.66 This additive effect suggests the possibility of asynergistic effect of allosteric and orthosteric drugs in thetreatment of human diseases. A large body of evidence suggeststhat allosteric sites have evolved under lower evolutionarypressure compared with orthosteric sites, implying thatallosteric mutations occur more frequently than orthostericmutations.87,88 Indeed, more than 20 disease-associatedmutations at the allosteric regulator binding domain ofpyruvate kinase have been observed owing to the evolutionarilyless conserved allosteric site.91 Additionally, mutations canoccur at the allosteric communication pathways beyondallosteric sites, altering the protein dynamics and causingindirect resistance to allosteric modulators.47 Such mutationsare very difficult to identify experimentally because of thecomplexity of allosteric networks in proteins.Additionally, allosteric modulators show significant species

differences owing to the low evolutionary conservation ofallosteric sites.5 For instance, the actions of allostericcandidates are effective in recombinant human receptors invitro. However, when using rodent receptors or models to

Figure 1. Surface representation of the X-ray crystal structure of p38αcomplexed to an allosteric compound 10 (1) (PDB code 3NEW).The orthosteric ATP binding site and allosteric site at the C-lobe ofthe kinase are highlighted in blue and red, respectively. Multiplesequence alignments of allosteric site residues in p38α, p38β, p38γ,and p38δ are shown, with identical and similarity residues marked byblack and gray, respectively.

Figure 2. Surface representation of the X-ray crystal structure of BCR-ABL1 complexed to both orthosteric drug imatinib and the allostericinhibitor ABL001 (asciminib) (PDB code 5MO4), with orthostericand allosteric sites highlighted in blue and red, respectively.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

C

Page 4: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

examine drug effects, it is possible to obtain the oppositeresult.92

In general, allosteric modulators have lower binding affinitythan orthosteric ligands93 and often encounter “flat,” non-tractable structure−activity relationships.16 Furthermore, allos-teric modulators often have lower aqueous solubility thanorthosteric ligands.16 These properties can lead not only to thedifficulty of advancing allosteric modulators as drug candidatesfor clinical testing but also to the crystallization of allostericprotein−modulator complexes.

4. METHODS TO INVESTIGATE ALLOSTERY4.1. Experimental Approaches. X-ray crystallography,

the most frequently used experimental approach in structuralbiology, has provided insights into structural and mechanisticinvestigations of biologically important macromolecules.94

Advances in X-ray crystallographic technology in the pastdecade have substantially contributed to determine thestructures of challenging macromolecules such as GPCRs,95

the proteasome,96 the maltose transporter,97 and eukaryoticexosomes.98 Remarkably, recent breakthroughs in the field ofsingle-particle cryoelectron microscopy (cryo-EM) haveenabled the characterization of the conformation of largemacromolecular assemblies such as viruses,99 cytoskeletalproteins,100 and spliceosomes101 at near-atomic resolution,providing potential applications in drug discovery.102 Both X-ray crystallography and cryo-EM can provide detailed essentialinformation about the three-dimensional (3D) structures ofligand-bound (holo) and unbound (apo) forms. Thesemethods have enabled a structural view of allostery. In anallosterically regulated protein, the conformational change atthe orthosteric site that occurs upon modulator binding can begenerally uncovered by structural comparison of the holo andapo forms of the protein. By contrast, in the case of dynamics-driven allosteric proteins such as CAP33,34 and PDZdomains,35 the orthosteric site and whole protein exhibit nostructural change between the holo and apo forms. In fact, X-ray and cryo-EM crystal structures are static average snapshotsof macromolecules. However, these molecules can interconvertalong many distinct conformations under physiological

conditions to mediate different biological functions. The lackof conformational change based on the static crystal structureoccasionally limits the usefulness of X-ray crystallography andcryo-EM in studies of allostery at the molecular level.In nature, allostery is a dynamic process. NMR spectroscopy,

on the other hand, can probe the dynamic processes ofbiomolecules on a wide range of ps−ms time scales andobserve conformational states in solution that are sparselypopulated, yielding detailed dynamical information that is verysuitable for the characterization of their allosteric mecha-nisms.103−106 It can be used to study allosterically regulatedproteins with significant conformational changes when boundto allosteric modulators. Most notably, it is particularly suitedto explore protein allostery when conformational change of theprotein is rarely detected in response to allosteric modulatorbinding, thus providing direct experimental evidence toestablish the dynamics or ensemble view of allostery. As anexample, using chemical shift and relaxation dispersion NMRanalyses, Tzeng and Kalodimos revealed that one cAMPmolecule binding to the first homodimeric CAP has a minoreffect on the fast dynamics of CAP, while the binding ofanother cAMP molecule to the second homodimeric CAPquenches such dynamics, further highlighting the role ofconformational entropy in the allosteric regulation of CAP,which was validated by ITC measurements.34 Traditionally,solution NMR spectroscopy has focused on relatively smallproteins. Recent advances in the amide-/methyl-TROSYapproaches, isotope labeling, and pulse sequence techniques107

have made it possible to characterize large-molecular-weightproteins such as the 20S proteasome core particle108 andmolecular chaperones such as HSP90109 and GroEL.110

X-ray crystallography, cryo-EM, and NMR spectroscopy canprovide direct experimental proof for allosteric modulation ofbiological macromolecule function. In addition to these directapproaches, a few indirect experimental approaches have beendeveloped to explore allostery in a particular protein. Theseinclude site-directed mutagenesis such as disulfide trapping (ortethering)111 and alanine scanning,112 fluorescent113,114 andphotoaffinity115 labeling, and hydrogen/deuterium exchange(HDX) mass spectrometry.116

4.2. Computational Approaches. It is typically time-consuming and frequently unsatisfactory to determineallosteric sites in proteins and the mechanism of allosteryusing experimental approaches such as X-ray crystallographyand NMR because of the complex nature of allostery inbiological processes. As a viable alternative to experimentalmethods, computational approaches for the study of allosteryprovide robust tools to aid in drug design, which has been anarea of intensive research.21,89,90,117 For example, AlloStericDatabase (ASD)20,118,119 and ASBench120 benchmark are twomajor developments that have facilitated the development ofcomputational methods to predict allosteric sites. A largeamount of the state-of-the-art computational methods haveemerged, facilitated by ASD and ASBench data in the past fewyears.21,89,90,117 Most critically, many of the theoreticalpredictions have been confirmed by experimental observations,which will be described in the following section, suggesting thepotential usefulness of computational methods in structure-based drug discovery. As such, a combined computational andexperimental approach should be become a new strategy inallosteric modulator discovery.Table 1 summarizes the current availability of representative

computational allosteric prediction methods. Such methods

Figure 3. Cartoon representation of the X-ray crystal structure ofBCR-ABL1. The drug-resistant mutation residues at the orthostericsite (Thr315) and allosteric site (Ala337, Pro465, Val468, and Ile502)are depicted by sphere representations.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

D

Page 5: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

include the prediction of allosteric sites, allosteric mutations,allosteric communication in proteins, assessment of allostericprotein−modulator interactions, and virtual screening ofallosteric modulators. Remarkably, most computationalmethods focus on the identification of allosteric sites becausethe determination of the location of allosteric sites is the firststep for structure-based allosteric modulator discovery. Awealth of predictive models for allosteric sites have beencreated based on different methodologies, including structure-based methods (e.g., Allosite,121 AllositePro,122 ALLO,123 anda random forest model124), evolution-based methods (e.g.,statistical coupling analysis (SCA)125), normal model analysis-based methods (e.g., AlloPred126 and PARS127), dynamics-based methods (a two-state Go model,128 ExProSE,129

molecular dynamics (MD) simulations with enhanced

conformational sampling,130 and AllosMod131), energy-basedperturbation methods (e.g., reverse perturbation),132 andcorrelation-based methods (e.g., CorrSite).133 Moreover,some of these programs, such as Allosite,121 PARS,127

AllosMod,131 and CorrSite,133 have been deployed as Webservers, facilitating medicinal chemists to find allostericmodulators. In addition to allosteric site prediction, Webservers or tools such as MCPath,134 SPACER,135 DynOmicsENM,136 AlloSigMA,137 AlloMAPS,138 and AlloDriver139 forallosteric communication and mutation analysis, STRESS140

for allosteric hotspot residues prediction, Alloscore141 forallosteric interaction evaluation, and CovCys142 for covalentallosteric modulator design have also been developed.

Table 1. Databases of Structures Containing Allosteric Sites and Software To Detect and Predict Them

name ref Web server available content, methods, and applications

databaseASD 20, 118, 119 http://mdl.shsmu.edu.cn/

ASDcollection of experimentally determined allosteric proteins, modulators, andpathways

benchmarkASBench 120 http://mdl.shsmu.edu.cn/

asbenchtwo benchmark sets of allosteric sites: “core set” and “core-diversity set”

allosteric site identificationAllosite 121 http://mdl.shsmu.edu.cn/

ASTdetection of allosteric sites based on a structure-based machine learning method

AllositePro 122 no detection of allosteric sites based on a combined structure- and normal-modeanalysis-based method

ALLO 123 no detection of allosteric sites based on a structure-based machine learning methodrandom forest model 124 no detection of allosteric sites based on a structure-based machine learning methodSCA 125 no detection of allosteric sites based on an evolutionary methodAlloPred 126 no detection of allosteric sites based on the normal-mode analysis methodPARS 127 http://bioinf.uab.cat/pars detection of allosteric sites based on the normal-mode analysis methodtwo-state Go model 128 no detection of allosteric sites based on an ensemble generated by coarse-grained

simulationsExProSE 129 no detection of allosteric sites based on an ensemble generated by a distance

geometry-based methodAllosMod 131 http://modbase.compbio.

ucsf.edu/allosmoddetection of allosteric sites and allosteric mechanism research from MDsimulations based on a modeled energy landscape

reverse perturbation approach 132 no detection of allosteric sites based on analysis of energetics of reverse allostericcommunication from orthosteric to allosteric sites

CorrSite 133, 142 http://www.pkumdl.cn/cavityplus

detection of allosteric sites based on motion correlation analysis of allosteric andorthosteric sites

allosteric hotspot residues predictionSTRESS 140 http://stress.molmovdb.org Prediction of allosteric hotspot residues based on models of conformational

change generated by Monte Carlo simulationsallosteric communication and mutation analysisMCPath 134 http://safir.prc.boun.edu.

tr/clbet_serverprediction of allosteric communication and allosteric residues based on anensemble generated by Monte Carlo path simulation

SPACER 135 http://allostery.bii.a-star.edu.sg

prediction of allosteric communication and allosteric sites based on the normal-mode analysis method

DynOmics ENM 136 http://dyn.life.nthu.edu.tw/oENM

prediction of allosteric communication and functional residues based on elasticnetwork model

AlloSigMA 137 http://allosigma.bii.a-star.edu.sg/home

assessment of the energetics of allosteric communication induced by ligandbinding and mutation

AlloMAPS 138 http://allomaps.bii.a-star.edu.sg

evaluation of allosteric effects of mutations and prediction of latent regulatoryexosites

AlloDriver 139 no analysis of clinic high-throughput data on allosteric mutationsallosteric interaction scoringAlloscore 141 http://mdl.shsmu.edu.cn/

alloscoreprediction of binding affinities of allosteric protein−modulator interactions basedon a machine learning method

allosteric modulator screening and designAlloFinder 26 http://mdl.shsmu.edu.cn/

ALFautomatic discovery of allosteric modulators and allosteric mechanism research

CovCys 142 http://www.pkumdl.cn/cavityplus

automatic detection of druggable cysteine residues for allosteric covalent drugdesign

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

E

Page 6: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

5. PRINCIPLES FOR ALLOSTERIC MODULATORDISCOVERY

The latest release of ASD (Nov 2018) contains ∼1800allosteric proteins regardless of species (Figure 4), which aremainly distributed among eight protein families: kinases,GPCRs, transcription factors, ion channels, oxidoreductases,lyases, hydrolases, and transferases (Figure 5). At the nascent

stage of allosteric research, the allosteric regulatory phenomenawere observed in multimeric proteins with hemoglobinrepresenting the prototype of this class. Owing to the progressin chemical and structural biology techniques, allostericbinding has been observed in monomeric proteins such asGPCRs, kinases, and enzymes, greatly expanding the repertoireof available allosteric drug targets.The number of allosteric modulators has been rapidly

increasing in recent years due to the growing investment of thescientific community in allosteric modulator discovery (Figure6). ASD now contains ∼80 000 allosteric modulators thattarget more than 1300 proteins. Among the allostericmodulators, five chemical entities have been approved by theU.S. FDA as marketed drugs, including three GPCR(Cinacalcet, Maraviroc, and Plerixafor) and two kinase(Gleevec and Mekinist) allosteric drugs. Additionally, manypromising candidates are now entering clinical trials.5.1. Characteristics of Allosteric Sites. The molecular

evolution of allosteric and orthosteric sites is different. Kim

and co-workers analyzed the sequence conservation ofallosteric and orthosteric sites in 56 allosterically regulatedenzymes.88 The results showed that allosteric site residues(average conservation score = 0.58) are significantly lessconserved than orthosteric site residues (average conservationscore = 0.94; P = 1.3 × 10−67). Similarly, comparing thesequence conservation of ATP binding sites in the allostericand orthosteric data sets, we found that residues in theorthosteric ATP binding site (average conservation score =0.63) are less conserved than residues in the orthosteric ATPbinding site (average conservation score = 0.83; P = 1.2 ×10−3).87 Furthermore, the shapes calculated by the pocketsimilarity score (PS-score) between the allosteric andorthosteric ATP binding sites are distinct. The global structuralsimilarity of the allosteric ATP binding site is lower (averagePS-score = 0.3) than that of the orthosteric ATP binding site(average PS-score = 0.6).87 Collectively, these data areconsistent with the higher target selectivity observed forallosteric modulators relative to orthosteric ligands.The physicochemical properties of allosteric and orthosteric

sites in an individual protein were further explored bycomparison of the amino acid compositions of the two setsof binding sites curated from ASD version 3.0.20 The resultsshowed that hydrophobic residues such as isoleucine andtyrosine are highly enriched in allosteric sites (Figure 7). Bycontrast, polar residues such as aspartic acid, glutamine,histidine, and glycine are highly enriched in orthosteric sites.These data suggest that allosteric sites are more hydrophobicthan orthosteric sites.

5.2. Characteristics of Allosteric Modulators. On thebasis of the allosteric modulators from ASD version 3.0 andorthosteric ligands from ChEMBL, Carlson and co-workerscompared the physicochemical properties of allostericmodulators and orthosteric ligands.143 The results showedthat allosteric modulators are more rigid and aromatic thanorthosteric ligands, in accordance with previous studies byOverington and co-workers.93 This principle can be applied tohelp select potential allosteric modulators for a protein aftervirtual screening; that is, in cases where it is known there aredifferences in polarity between orthosteric and allosteric sites,

Figure 4. Total number of X-ray or NMR structures of allosteric proteins and yearly growth of total structures in the AlloSteric Database (ASD).Representative structures of allosteric proteins are shown.

Figure 5. Major class distribution of the structures of allostericproteins in ASD.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

F

Page 7: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

choosing compounds that are rigid and more aromatic canincrease the likelihood of occupying an allosteric site.

6. EXAMPLES OF THE STRUCTURE-BASED DESIGNOF ALLOSTERIC MODULATORS

Due to the recent progress in the molecular understanding ofthe allosteric process in recent years, many computationalapproaches have been developed for various aspects ofallosteric applications. Such approaches proved successful in

finding putative allosteric modulators, confirmed by subse-quent crystallographic/NMR or biological experiments. In thissection, we will present some representative examples of usingcombined computational and experimental tactics to identifyallosteric modulators.

6.1. SIRT6 Allosteric Activators. Sirtuin 6 (SIRT6), amember of the human sirtuin family (SIRT1−7), is a histonedeacetylase that transfers the acetyl group from the lysine sidechain of a protein or peptide substrate to nicotinamide adeninedinucleotide (NAD+).144 The regulation of SIRT6 has beenclosely associated with multiple biological processes, encom-passing DNA damage repair, organ metabolism, aging, andtumorigenesis.145 Accumulating evidence strongly indicatesthat SIRT6 knockout mice are accompanied by a severepremature aging syndrome, while mice containing SIRT6overexpression display a dramatically longer lifespan. They alsoshow massive apoptosis in various cancer cells but not innormal cells.146−148 Pharmacological activation of SIRT6 bysmall-molecule compounds may, therefore, provide a potentialnew avenue in age- and cancer-related treatment.149,150

To increase SIRT6 catalytic activity, the design ofcompounds to directly compete with endogenous NAD+ is

Figure 6. Yearly growth of allosteric modulators in ASD. The chemical structures of the marketed allosteric drugs are indicated in the chart.

Figure 7. Residue compositions in the orthosteric (light blue) andallosteric (blue) sites calculated from ASD version 3.0.

Figure 8. Workflow of the rational discovery of SIRT6 allosteric activators. The potential allosteric site of SIRT6 was predicted using Allosite(http://mdl.shsmu.edu.cn/AST). SIRT6 is shown as surface colored in gray, with the orthosteric and predicted allosteric sites highlighted in blueand red, respectively.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

G

Page 8: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

infeasible because NAD+ is the SIRT6 cofactor involved in thecatalytic reaction. As such, it is advisable and effective to targetallosteric sites outside the NAD+ orthosteric site. However, theallosteric sites of SIRT6 were unavailable at the onset of ourstudies on SIRT6 allosteric activators in 2013.To address this issue, we employed our previously

developed structure-based Allosite121 approach to computa-tionally predict potential allosteric sites (Figure 8). Theprediction informed us of a potential allosteric site formed byresidues Phe82 and Phe86 adjacent to the N-terminus ofSIRT6. On the basis of this predicted allosteric site, virtualscreening of seven commercial chemical libraries (SPECS,ChemBridge, ChemDiv, Enamine, InterBioScreen, Life Chem-icals, and Maybridge) consisting of more than 5 000 000compounds was performed. According to the principle thatallosteric modulators are rigid and more aromatic thanorthosteric ligands, we chose and purchased 20 rigid andaromatic compounds from the top-ranked compoundscalculated by our Alloscore141 approach to assess allostericinteractions. After experimental testing, two initial hits fromthe SPECS chemical library, AN-988/40889624 (2) and AH-487/41802661 (3) (Figure 7), were found to dose-depend-ently activate SIRT6 deacetylation, with half-maximal effectiveconcentration values of 173.8 ± 1.3 μM and 217.6 ± 1.1 μM,respectively.25 To improve the activation potency of the hits,medicinal chemistry optimizations of 3 led to two activatorsMDL-800 (4) and MDL-801 (5) (Figure 8), both of whichsignificantly enhanced the deacetylation activity of SIRT6, withEC50 values of 10.3 ± 0.3 μM and 5.7 ± 0.3 μM, respectively.Crystallographic studies further confirmed that 5 binds to theallosteric site located at the surface exit of the long SIRT6channel pocket but not to the substrate site of SIRT6. Thisallosteric site is formed by the N-terminal residues Val3-Leu9,Val70, Glu74, Phe82, Phe86, Val153, and Met157 (Figure9),25 in agreement with the computationally predictedallosteric site by Allosite121 and supporting the feasibility ofAllosite in the identification of as-yet-unknown allosteric sites.Cellular and animal model studies on the growth inhibitory

effects of human hepatocellular carcinoma (HCC) furtherdemonstrated that allosteric SIRT6 activators can suppress theproliferation of HCC.25 These findings should provide astarting point to facilitate the exploration of allosteric SIRT6activators as therapeutic agents or as probe compounds tounderstand SIRT6 biology.

6.2. STAT3 Allosteric Inhibitors. Signal transducer andactivator of transcription 3 (STAT3) play an important role inmany biological processes, including differentiation, survival,proliferation, and angiogenesis.151 However, constitutive oraberrant activation of STAT3 has been linked to malignanttransformation and tumorigenesis.152 As such, inhibition ofSTAT3 activity represents an attractive strategy for anticancerdrug discovery.153

STAT3 is a multidomain protein consisting of six distinctfunctional domains (Figure 10). Many STAT3 inhibitors werepreviously developed that targeted to the Src homology 2(SH2) domain and, to a lesser extent, the DNA-bindingdomain (DBD).154−157 Such inhibitors often encounter poorpharmacokinetic properties and cytotoxicity during clinicaltrials, making them ineffective drug candidates.158 Unlike theSH2 domain and DBD, inhibitors targeted to other domainssuch as the coiled-coil domain (CCD) are scarce. The CCDcan be regarded as an allosteric domain to regulate STAT3functional activity.85

On the basis of the STAT3 CCD, we used our developedAlloFinder26 approach to screen in silico for potential allostericinhibitors. This approach can automatically predict allostericsites by Allosite,121 perform molecular docking by AutoDockVina,159 and rank the docked poses by Alloscore.141 We choseand purchased the top 15 hits for bioassays; these hits arebound to the predicted allosteric site formed by residuesAsp171, Asn175, Gln202, and Met213 (Figure 10). Afterbiological testing, we observed that the small-moleculecompound K116 (AH-034/11936955) (6) from the SPECSchemical library can bind to STAT3 CCD, with a Kd value of3.22 ± 0.85 μM.26 Site-directed mutagenesis experimentsshowed that both double mutants (N175G/K177G andQ212A/M213G) decreased the binding affinity of 6 to themutants by ∼10-fold compared with wild-type STAT3.26

These data support the possibility of the discovery of STAT3allosteric inhibitors by the AlloFinder approach. However, afurther crystallographic structural complex is required toconfirm the allosteric inhibition of STAT3 by 6 directly.Currently, medicinal chemistry optimization of 6 is in progress,and structural determination of STAT3 in complex with 6derivatives will be performed soon.

6.3. GPX4 Allosteric Activators. By analysis of thecorrelations between allosteric sites and correspondingorthosteric sites in monomer or oligomeric allosteric proteinsusing a Gaussian network model, Lai and co-workers foundthat the motions of allosteric and orthosteric sites in proteinsare highly correlated.133 According to this principle, theydeveloped a correlation analysis method named CorrSite133 topredict potential allosteric sites in proteins.Glutathione peroxidase 4 (GPX4) controls many biological

processes, including cell membrane repair, inflammationsuppression, and ferroptosis inhibition.160 UpregulatingGPX4 enzyme activity has a clinical benefit in the treatmentof inflammation- and ferroptosis-associated diseases.161,162 Assuch, the development of GPX4 activators can provide apharmacological benefit.

Figure 9. Detailed interactions between MDL-801 and SIRT6 at theallosteric site determined by the cocrystal structure (PDB code5Y2F). SIRT6 is shown in gray, and MDL-801 is depicted by stickswith carbon atoms colored in yellow. The key residues within theallosteric site of MDL-801 are represented by sticks with carbonatoms colored in cyan.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

H

Page 9: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

To discover GPX4 activators, Lai and co-workers recentlyhave combined the computational methods CAVITY163 andCorrSite133 to predict a potential allosteric site in GPX4.27 Thepredicted allosteric site, with a CorreSite score of 1.49 (cavitieswith CorreSite scores of >0.5 are considered potentialallosteric sites by the CorrSite method), is located at theopposite side of the protein from the substrate binding site(Figure 11). This allosteric site is formed by three acidic

residues (Asp21, Asp23, and Asp101), two basic residues(Lys31 and Lys90), and seven hydrophobic residues (Ile22,Ala93, Ala94, Val98, Phe100, Met102, and Phe103). Virtualscreening of the SPECS chemical library was then performedbased on the predicted allosteric site, and 251 compoundswere selected for biological testing with ≥1 protein−ligandhydrogen bond and good hydrophobic interactions. Amongthese compounds, compound 1 (7) (Figure 11) was observedto dose-dependently increase GPX4 activity to 260% at anactivator concentration of 500 μM in the cell-free assay, with aKd value of 63 ± 5 μM. On the basis of the modeling of CPX4-7 protein−ligand interactions, SAR studies of 7 were furtherperformed to generate the strongest compound, 1d4 (8)(Figure 11), which significantly increased GPX4 activity to150% at 20 μM in the cell-free assay and 61 μM in cell extracts.

A double mutant (D21A/D23A) and two single mutants(D21A and D23A) showed reduced activation of GPX4 inresponse to both 7 and 8, confirming the predicted allostericsite by the CorrSite133 approach.

6.4. Cathepsin K Allosteric Inhibitors. Evolution-basedprediction of allosteric sites exploit sequence alignments andother genetics analysis methods to identify conserved residuesthroughout evolution.164 SCA is a prototype of an evolution-based prediction method, which utilizes multiple sequencealignment (MSA) to uncover evolutionarily conserved net-works of residues within a protein family.46 Such networks,referred to as protein sectors, are proposed to mediateallosteric communications between functional orthostericsites and regulatory allosteric sites.46,165 Nevertheless, thepracticality of SCA has limitations.166−170 Major concernsinclude that this method is only applicable to residues in closephysical contacts that are immediately subject to evolutionselection167,169,170 and whether its prediction of the evolutio-narily constrained positions can completely reflect thethermodynamic or allosteric couplings within protein struc-tures.166

Despite its advantages and disadvantages, the potentialapplication of SCA has been exemplified in the discovery of thefirst low-molecular-weight allosteric inhibitor of cathepsin K,NSC13345 (9) (Figure 12).171 Novinec and co-workers firstconstructed an MSA of 1239 catalytic domains from thepapain-like cysteine peptidase family.171 The pairwisecorrelations between all position pairs within the alignmentwere revealed through the positional correlation matrixcalculations, which were then clustered using hierarchicalclustering. A continuous network surrounding the orthostericsite and stretching around cathepsin K (i.e., the protein sector)was identified through automated sector identification. Thissector comprises the coevolving residues in the protein familywith functional importance. A few potential allosteric siteswere subsequently detected using the AutoLigand172 cavityprediction method. 9 from the NCI Diversity Set IIIcompound library was identified as a potential hit. A crystalstructure of cathepsin K in complex with 9, solved todetermine the allosteric regulatory mechanism for its binding,showed that it was bound to a remote surface-exposedallosteric site (Figure 12), site 6. The latter mainly compriseshelix α10 and adjacent β sheets and loops. Within this site, 9makes hydrogen bonds with Gly113, Lys236, Arg237, and

Figure 10. Workflow of the rational discovery of a STAT3 allosteric inhibitor. An X-ray structure of STAT3 (residues 130−722) (PDB code1BG1) is colored by the domain. The predicted allosteric site at the coiled-coil domain (CCD), large-scale molecular docking of compoundlibraries at the CCD allosteric site, and allosteric interaction evaluation after docking were executed automatically by AlloFinder (http://mdl.shsmu.edu.cn/ALF). Enlarged portion shows the potential interactions between the allosteric inhibitor K116 (6) and STAT3, consistent withmutagenesis experiments.

Figure 11. Workflow of the rational discovery of GPX4 allostericactivators. The allosteric site of GPX4 was predicted using CAVITYand CorrSite (http://www.pkumdl.cn/cavityplus) based on thecrystal structure of GPX4 (PDB code 2OBI). The predicted allostericsite (red) is on the opposite side of the orthosteric site (blue). Thepredicted binding mode of 1d4 (8) represented by blue sticks toGPX4 is shown, with key residues within the binding site highlightedin orange by stick representations.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

I

Page 10: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

Gln313. Further biochemical assays revealed that 9 abolishedthe collagenolytic activity of cathepsin K, with an IC50 value of80 ± 30 μM. Together, these results suggest the possibility ofapplying an evolution-based allosteric method in the discoveryof allosteric modulators. In fact, this method has been appliedin other cases, such as the PDZ domain,173 G proteins,174 andTonB-dependent transporters.175

6.5. c-Src Allosteric Inhibitors. Protein allostery is rootedin protein dynamics.176 The proper understanding of allostericeffects and accurate prediction of allostery cannot be achievedunless the conformational dynamics and energetic landscape ofprotein structures are thoroughly dissected. MD simulationscan characterize both large-scale conformational changes inoverall structures and subtle alterations in residue orientations,as well as provide energetic insights into conformationaldynamics.177−180 Thus, MD-based approaches, coupled withsubsequent correlated motions and community analyses, canuncover allosteric sites coupled to orthosteric sites with a highaccuracy.181−184 Furthermore, MD simulations enable theproduction of complete dynamic biological processes, facilitat-ing the detection of the highly flexible or even transient, so-called “hidden” allosteric sites.73,74

Pande and co-workers performed 550 μs of massivelydistributed MD simulations and Markov state model (MSM)analyses to identify potential allosteric sites on the anticancerdrug target c-Src.185 Snapshot structures from simulatedtrajectories were clustered using MSM analysis, whichseparated the dynamic process into a series of discrete statesand calculated the population of each state and transitionsbetween them. Two intermediate states (I1 and I2) werethereby identified along the activation pathway of c-Src. A highsimilarity was observed between the I2 conformational stateand previously reported structure of an allosteric inhibitor(ANS) (10)-bound cyclin-dependent kinase 2 (CDK2).186 Inthese two complexes, the activation loop (A-loop) adopted anunfolded conformation, forming a potential allosteric site withthe C-helix and adjacent β-sheets (Figure 13A), which cannot

be found in both the active (Figure 13B and Figure 13C) andinactive (Figure 13D) states of c-Src. The binding of 10 to thec-Src was further confirmed by simulations of a 10-bound I2structure. 10 was bound to the pocket formed by the A-loop,C-helix, and β3, 5, 6 of c-Src, similar to that in the crystalstructure of the CDK2-10 complex (Figure 13A). Throughblocking the displacement of the C-helix, 10 allostericallyinhibited c-Src by trapping it in a partially active intermediatestate. Collectively, this study highlighted that large-scale MDsimulations can detect a novel, cryptic allosteric site in theintermediate state structure of c-Src. Due to their ability tocharacterize the kinetic, thermodynamic, and structuralfeatures of the protein ensembles, MD simulations, coupledwith subsequent MSM analysis, have become a popular, robusttool for allosteric site and modulator discovery.187−190

7. CONCLUSIONS AND PERSPECTIVESAllostery underlies a myriad of biological processes thatregulate protein functions, where binding of an effector at theallosteric site affects catalytic processing at an orthosteric site.From a medicinal chemistry perspective, allostery represents aneffective tool for the medicinal chemist to drug “undruggable”proteins or alternatively to expand the repertoire of theavailable drug targets. Allosteric drugs differ from orthostericdrugs in the mode of drug action, with the possibility ofachieving higher selectivity, fewer adverse effects, and lowertoxicity than orthosteric drugs. Allosterism in drug discoveryhas, therefore, emerged as a promising strategy to developefficient and safe therapeutic agents. Because the nature of

Figure 12. Structural overview of cathepsin K in complex with itsallosteric inhibitor NSC13345 (9) and substrate TCO (PDB code5J94). The backbone structure of cathepsin K is shown in gray. Theallosteric site (site 6) and orthosteric site are highlighted in red andblue, respectively. The upper panel shows the chemical structure of 9,and the lower panel shows the detailed interactions between 9 andcathepsin K. Hydrogen bonds are displayed in red dashed lines, andresidues within the allosteric site are represented by sticks. SubstrateTCO is manually docked into the orthosteric site based on thesuperposition of PDB code 5J94 with PDB code 1Q6K. Figure 13. (A) Chemical structure of ANS (10) and structural

overview of CDK2 in complex with 10 (PDB code 3PXF). (B) Thebinding site of 10 is in a closed conformation in the active state of c-Src (PDB code 1Y57). (C) Structural comparison between the 10-bound CDK2 (yellow) and active state of c-Src (blue) shows that theallosteric site is in a closed conformation mainly due to thedisplacements of the C-helix and A-loop. (D) The binding site of10 is also in a closed conformation in the inactive state of c-Src (PDBcode 2SRC). The C-helix, A-loop, and β-sheets involved in thebinding site of 10 are colored in orange, pink, and cyan, respectively.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

J

Page 11: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

allosteric modulation is complicated, allosteric modulatorswere traditionally discovered serendipitously by high-through-put screening.Over the past decade, understanding of the basic

mechanisms of allosteric modulation, technology advances inthe experimental approaches to study allostery, and remarkableavailability of structural data have contributed to thedevelopment of various computational approaches to predictallosteric phenomena.191 Such predictive models were createdbased on the structure-, evolution-, or dynamics-basedmethods to detect and discover allosteric sites andmodulators.89,90 Some of these computational predictionshave been confirmed by experimental observations, high-lighting the possibility of computational approaches in thediscovery of allosteric modulators.Despite decades of allosteric research and the development

of many models for allosteric applications, allosteric modulatordiscovery continues to face challenges. One of the biggestchallenges originates from identifying putative allosteric sites.Particularly, in the case of cryptic allosteric sites that areinvisible in apo crystal structures but can emerge in theconformational transition of proteins such as in holo crystalstructures, current structure- and evolution-based allostericcomputational methods cannot detect such sites in the apostructures.73 Large-scale MD simulations, coupled with MSManalyses, can provide a potential opportunity to identify crypticallosteric sites, but such simulations with an effectiveconformational sampling are extremely time-consuming.192,193

Alternatively, using normal-mode analysis to rapidly capturethe conformational ensemble of a protein and then employingcomputational approaches to identify cryptic allosteric sites canoffer guidelines to help to circumvent this dilemma.Despite the current understanding of the general character-

istics of known allosteric sites and modulators, the guidelinesfor the structural modifications of initial allosteric hits toincrease affinities (hit-to-lead optimization) are lacking.Because the properties of allosteric sites and modulators differfrom the corresponding orthosteric sites and ligands, softwaretraditionally used for orthosteric drug design faces challengesin guiding allosteric modulator design. There is a strongdemand for insights into the unique protein−modulatorinteractions underlying allosteric binding. Our previouslycreated ASBench120 benchmark of high-quality data ofallosteric protein−modulator complexes can assist in uncover-ing interactions useful for designing allosteric-active com-pounds.Many reports of analyses of allostery on the single-molecule

level have emerged, leaving the role of allostery at the cellularlevel unexplored.194 Allostery is a fundamental cellularphenomenon. Thus, it is required to develop multiscaleapproaches that unite computational and experimentalmethods to explore allosteric effects on cellular networks.The system-centric method will identify new targets tobroaden the allosteric modulator discovery scope.195−197 Forexample, the allo-network drug concept has recently beenproposed to design allosteric modulators in system-based drugdesign.198

Overall, recently developed computational allosteric pre-diction approaches have shown some success in designingallosteric modulators for known protein targets. Thesetechniques, coupled with advances in experimental approachesto understand allosteric activation or inhibition, indicate that

allosteric modulator discovery has emerged as a new strategyfor drug design.

■ AUTHOR INFORMATIONCorresponding Author*Phone: +86-21-63846590-776922. Fax: +86-21-64154900. E-mail: [email protected] Zhang: 0000-0002-6558-791XAuthor Contributions§S.L., X.H., and D.N. contributed equally to this work.NotesThe authors declare no competing financial interest.BiographiesShaoyong Lu received his undergraduate Applied Chemistry degreefrom Hangzhou Normal University in 2007. Thereafter, he obtained aPh.D. degree in Chemistry from Zhejiang University, China, in 2012.He then continued with postdoctoral research at Shanghai Jiao TongUniversity, School of Medicine, in Professor Jian Zhang’s laboratoryand is now an Associate Professor. During 2014−2015, he studied as aVisiting Scholar in Professor Ruth Nussinov’s group at the NationalCancer InstituteFrederick. His main interests include the study ofallosteric protein−ligand interactions and the development ofallosteric methods and their applications.

Xinheng He is an undergraduate student from the Discipline ofBiomedical Science, Shanghai Jiao Tong University, School ofMedicine, in Professor Jian Zhang’s group. He mainly focuses onallosteric mechanism research and allosteric drug design.

Duan Ni is an undergraduate student from the Discipline ofBiomedical Science, Shanghai Jiao Tong University, School ofMedicine, in Professor Jian Zhang’s group. He mainly focuses onusing computational methods to investigate the allosteric mechanismsand predict allosteric sites.

Jian Zhang received a B.M. degree in Pharmacology in 2002 fromPeking University and a Ph.D. in 2007 from Shanghai Institute ofMateria Medica, Chinese Academy of Sciences. After receiving hisPh.D., he moved to the University of Michigan to conductpostdoctoral research in Professor Shaomeng Wang’s laboratory. In2009, he joined Shanghai Jiao Tong University, School of Medicine,where he is a full-time researcher and doctoral supervisor. Now, he isalso the director of the Medicinal Bioinformatics Center. His fields ofresearch include first-in-class drug discovery and chemical biology thatmainly pertains to the repertoire of allostery. His Web site providesfurther details: http://mdl.shsmu.edu.cn/.

■ ACKNOWLEDGMENTSThis work was supported by the National Natural ScienceFoundation of China (Grants 21778037, 81603023, 81322046,91753117, 81473137), the National Basic Research Programof China (973 Program) (Grant 2015CB910403), and theInnovation Team “Diagnosis and Treatment of MajorCardiovascular Diseases” of High-Level Universities inShanghai.

■ ABBREVIATIONS USEDA-loop, activation loop; ASD, AlloSteric Database; CAP,catabolite activator protein; cryo-EM, cryoelectron micros-copy; CCD, coiled-coil domain; CDK2, cyclin-dependentkinase 2; 3D, three-dimensional; DBD, DNA-binding domain;GPX4, glutathione peroxidase 4; GPCR, G-protein-coupled

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

K

Page 12: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

receptor; HCC, human hepatocellular carcinoma; HDX,hydrogen/deuterium exchange; KNF, Koshland−Nemethy−Filmer; MSM, Markov state model; MD, molecular dynamics;MWC, Monod−Wyman−Changeux; MSA, multiple sequencealignment; NAD+, nicotinamide adenine dinucleotide; NMR,nuclear magnetic resonance; PPI, protein−protein interaction;STAT3, signal transducer and activator of transcription 3;SH2, Src homology 2; SCA, statistical coupling analysis;SIRT6, sirtuin 6

■ REFERENCES(1) Ribeiro, A. A. S. T.; Ortiz, V. A Chemical Perspective onAllostery. Chem. Rev. 2016, 116, 6488−6502.(2) Changeux, J.-P. The Concept of Allosteric Modulation: AnOverview. Drug Discovery Today: Technol. 2013, 10, e223−e228.(3) Dokholyan, N. V. Controlling Allosteric Networks in Proteins.Chem. Rev. 2016, 116, 6463−6487.(4) Nussinov, R.; Tsai, C. J.; Xin, F.; Radivojac, P. Allosteric Post-Translational Modification Codes. Trends Biochem. Sci. 2012, 37,447−455.(5) Nussinov, R.; Tsai, C. J. Allostery in Disease and in DrugDiscovery. Cell 2013, 153, 293−305.(6) Goodey, N. M.; Benkovic, S. J. Allosteric Regulation andCatalysis Emerge via a Common Route. Nat. Chem. Biol. 2008, 4,474−482.(7) Nussinov, R.; Ma, B.; Tsai, C. J.; Csermely, P. AllostericConformational Barcodes Direct Signaling in the Cell. Structure 2013,21, 1509−1521.(8) Changeux, J.-P. Allosteric Receptors: From Electric Organ toCognition. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 1−38.(9) Lu, S.; Jang, H.; Muratcioglu, S.; Gursoy, A.; Keskin, O.;Nussinov, R.; Zhang, J. Ras Conformational Ensembles, Allostery, andSignaling. Chem. Rev. 2016, 116, 6607−6665.(10) Lisi, G. P.; Loria, J. P. Allostery in Enzyme Catalysis. Curr.Opin. Struct. Biol. 2017, 47, 123−130.(11) Fenton, A. W. Allostery: An Illustrated Definition for the“Second Secret of Life”. Trends Biochem. Sci. 2008, 33, 420−425.(12) Changeux, J. P.; Christopoulos, A. Allosteric Modulation as aUnifying Mechanism for Receptor Function and Regulation. Cell2016, 166, 1084−1102.(13) Wootten, D.; Christopoulos, A.; Sexton, P. M. EmergingParadigms in GPCR Allostery: Implications for Drug Discovery. Nat.Rev. Drug Discovery 2013, 12, 630−644.(14) Rettenmaier, T. J.; Sadowsky, J. D.; Thomsen, N. D.; Chen, S.C.; Doak, A. K.; Arkin, M. R.; Wells, J. A. A Small-Molecule Mimic ofa Peptide Docking Motif Inhibits the Protein Kinase PDK1. Proc.Natl. Acad. Sci. U. S. A. 2014, 111, 18590−18595.(15) Zhang, J.; Adrian, F. J.; Jahnke, W.; Cowan-Jacob, S. W.; Li, A.G.; Iacob, R. E.; Sim, T.; Powers, J.; Dierks, C.; Sun, F.; Guo, G. R.;Ding, Q.; Okram, B.; Choi, Y.; Wojciechowski, A.; Deng, X.; Liu, G.;Fendrich, G.; Strauss, A.; Vajpai, N.; Grzesiek, S.; Tuntland, T.; Liu,Y.; Bursulaya, B.; Azam, M.; Manley, P. W.; Engen, J. R.; Daley, G. Q.;Warmuth, M.; Gray, N. S. Targeting Bcr-Abl by Combining Allostericwith ATP-Binding-Site Inhibitors. Nature 2010, 463, 501−506.(16) Wenthur, C. J.; Gentry, P. R.; Mathews, T. P.; Lindsley, C. W.Drugs for Allosteric Sites on Receptors. Annu. Rev. Pharmacol. Toxicol.2014, 54, 165−184.(17) De Smet, F.; Christopoulos, A.; Carmeliet, P. AllostericTargeting of Receptor Tyrosine Kinases. Nat. Biotechnol. 2014, 32,1113−1120.(18) Hauser, A. S.; Attwood, M. M.; Rask-Andersen, M.; Schioth, H.B.; Gloriam, D. E. Trends in GPCR Drug Discovery: New Agents,Targets and Indications. Nat. Rev. Drug Discovery 2017, 16, 829−842.(19) Hardy, J. A.; Wells, J. A. Searching for New Allosteric Sites inEnzymes. Curr. Opin. Struct. Biol. 2004, 14, 706−715.(20) Shen, Q.; Wang, G.; Li, S.; Liu, X.; Lu, S.; Chen, Z.; Song, K.;Yan, J.; Geng, L.; Huang, Z.; Huang, W.; Chen, G.; Zhang, J. ASD

v3.0: Unraveling Allosteric Regulation with Structural Mechanismsand Biological Networks. Nucleic Acids Res. 2016, 44, D527−D535.(21) Wagner, J. R.; Lee, C. T.; Durrant, J. D.; Malmstrom, R. D.;Feher, V. A.; Amaro, R. E. Emerging Computational Methods for theRational Discovery of Allosteric Drugs. Chem. Rev. 2016, 116, 6370−6390.(22) Lu, S.; Zhang, J. Designed Covalent Allosteric Modulators: AnEmerging Paradigm in Drug Discovery. Drug Discovery Today 2017,22, 447−453.(23) Perna, A. M.; Reisen, F.; Schmidt, T. P.; Geppert, T.; Pillong,M.; Weisel, M.; Hoy, B.; Simister, P. C.; Feller, S. M.; Wessler, S.;Schneider, G. Inhibiting Helicobacter Pylori HtrA Protease byAddressing a Computationally Predicted Allosteric Ligand BindingSite. Chem. Sci. 2014, 5, 3583−3590.(24) Hocker, H. J.; Cho, K.-J.; Chen, C.-Y. K.; Rambahal, N.;Sagineedu, S. R.; Shaari, K.; Stanslas, J.; Hancock, J. F.; Gorfe, A. A.Andrographolide Derivatives Inhibit Guanine Nucleotide Exchangeand Abrogate Oncogenic Ras Function. Proc. Natl. Acad. Sci. U. S. A.2013, 110, 10201−10206.(25) Huang, Z.; Zhao, J.; Deng, W.; Chen, Y.; Shang, J.; Song, K.;Zhang, L.; Wang, C.; Lu, S.; Yang, X.; He, B.; Min, J.; Hu, H.; Tan,M.; Xu, J.; Zhang, Q.; Zhong, J.; Sun, X.; Mao, Z.; Lin, H.; Xiao, M.;Chin, Y. E.; Jiang, H.; Xu, Y.; Chen, G.; Zhang, J. Identification of aCellularly Active SIRT6 Allosteric Activator. Nat. Chem. Biol. 2018,14, 1118−1126.(26) Huang, M.; Song, K.; Liu, X.; Lu, S.; Shen, Q.; Wang, R.; Gao,J.; Hong, Y.; Li, Q.; Ni, D.; Xu, J.; Chen, G.; Zhang, J. AlloFinder: AStrategy for Allosteric Modulator Discovery and AllosteromeAnalyses. Nucleic Acids Res. 2018, 46, W451−W458.(27) Li, C.; Deng, X.; Zhang, W.; Xie, X.; Conrad, M.; Liu, Y.;Angeli, J. P. F.; Lai, L. Novel Allosteric Activators for FerroptosisRegulator Glutathione Peroxidase 4. J. Med. Chem. 2019, 62, 266−275.(28) Monod, J.; Jacob, F. Telenomic Mechanisms in CellularMetabolism, Growth, and Differentiation. Cold Spring Harbor Symp.Quant. Biol. 1961, 26, 389−401.(29) Monod, J.; Wyman, J.; Changeux, J.-P. On the Nature ofAllosteric Transitions: A Plausible Model. J. Mol. Biol. 1965, 12, 88−188.(30) Koshland, D. E.; Nemethy, J. G.; Filmer, D. Comparison ofExperimental Binding Data and Theoretical Models in ProteinsContaining Subunits. Biochemistry 1966, 5, 365−385.(31) Cooper, A.; Dryden, D. T. Allostery without ConformationalChange. A Plausible Model. Eur. Biophys. J. 1984, 11, 103−109.(32) Igumenova, T. I.; Frederick, K. K.; Wand, A. J. Characterizationof the Fast Dynamics of Protein Amino Acid Side Chains Using NMRRelaxation in Solution. Chem. Rev. 2006, 106, 1672−1699.(33) Tzeng, S. R.; Kalodimos, C. G. Dynamic Activation of anAllosteric Regulatory Protein. Nature 2009, 462, 368−372.(34) Tzeng, S. R.; Kalodimos, C. G. Protein Activity Regulation byConformational Entropy. Nature 2012, 488, 236−240.(35) Petit, C. M.; Zhang, J.; Sapienza, P. J.; Fuentes, E. J.; Lee, A. L.Hidden Dynamic Allostery in a PDZ Domain. Proc. Natl. Acad. Sci. U.S. A. 2009, 106, 18249−18254.(36) Lundstrom, P.; Mulder, F. A.; Akke, M. Correlated Dynamics ofConsecutive Residues Reveal Transient and Cooperative Unfolding ofSecondary Structure in Proteins. Proc. Natl. Acad. Sci. U. S. A. 2005,102, 16984−16989.(37) Frederick, K. K.; Marlow, M. S.; Valentine, K. G.; Wand, A. J.Conformational Entropy in Molecular Recognition by Proteins.Nature 2007, 448, 325−329.(38) Lee, A. L.; Kinnear, S. A.; Wand, A. J. Redistribution and Lossof Side Chain Entropy upon Formation of a Calmodulin-PeptideComplex. Nat. Struct. Biol. 2000, 7, 72−77.(39) Kornev, A. P.; Taylor, S. S. Dynamics-Driven Allostery inProtein Kinases. Trends Biochem. Sci. 2015, 40, 628−647.(40) Wand, A. J. Bringing Disorder and Dynamics in ProteinAllostery into Focus. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 4278−4280.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

L

Page 13: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

(41) Marlow, M. S.; Dogan, J.; Frederick, K. K.; Valentine, K. G.;Wand, J. A. The Role of Conformational Entropy in MolecularRecognition by Calmodulin. Nat. Chem. Biol. 2010, 6, 352−358.(42) Kasinath, V.; Sharp, K. A.; Wand, A. J. Microscopic Insightsinto the NMR Relaxation-Based Protein Conformational EntropyMeter. J. Am. Chem. Soc. 2013, 135, 15092−15100.(43) Kumar, S.; Ma, B.; Tsai, C. J.; Wolfson, H.; Nussinov, R.Folding Funnels and Conformational Transitions via Hinge-BendingMotions. Cell Biochem. Biophys. 1999, 31, 141−164.(44) Ma, B.; Kumar, S.; Tsai, C. J.; Nussinov, R. Folding Funnelsand Binding Mechanisms. Protein Eng., Des. Sel. 1999, 12, 713−720.(45) Tsai, C. J.; Kumar, S.; Ma, B.; Nussinov, R. Folding Funnels,Binding Funnels, and Protein Function. Protein Sci. 1999, 8, 1181−1190.(46) Lockless, S. W.; Ranganathan, R. Evolutionarily ConservedPathways of Energetic Connectivity in Protein Families. Science 1999,286, 295−299.(47) del Sol, A.; Tsai, C. J.; Ma, B.; Nussinov, R. The Origin ofAllosteric Functional Modulation: Multiple Pre-Existing Pathways.Structure 2009, 17, 1042−1050.(48) Csermely, P.; Korcsmaros, T.; Kiss, H. J. M.; London, G.;Nussinov, R. Structure and Dynamics of Molecular Networks: ANovel Paradigm of Drug Discovery: A Comprehensive Review.Pharmacol. Ther. 2013, 138, 333−408.(49) Lu, S.; Deng, R.; Jiang, H.; Song, H.; Li, S.; Shen, Q.; Huang,W.; Nussinov, R.; Yu, J.; Zhang, J. The Mechanism of ATP-Dependent Allosteric Protection of Akt Kinase Phosphorylation.Structure 2015, 23, 1725−1734.(50) Changeux, J.-P.; Edelstein, S. J. Allosteric Mechanisms of SignalTransduction. Science 2005, 308, 1424−1428.(51) Hilser, V. J. An Ensemble View of Allostery. Science 2010, 327,653−654.(52) Motlagh, H. N.; Wrabl, J. O.; Li, J.; Hilser, V. J. The EnsembleNature of Allostery. Nature 2014, 508, 331−339.(53) Hilser, V. J.; Wrabl, J. O.; Motlagh, H. N. Structural andEnergetic Basis of Allostery. Annu. Rev. Biophys. 2012, 41, 585−609.(54) Liu, J.; Nussinov, R. Allostery: An Overview of Its History,Concepts, Methods, and Applications. PLoS Comput. Biol. 2016, 12,e1004966.(55) Tsai, C.-J.; Nussinov, R. A Unified View of “How AllosteryWorks”. PLoS Comput. Biol. 2014, 10, No. e1003394.(56) Cui, Q.; Karplus, M. Allostery and Cooperativity Revisited.Protein Sci. 2008, 17, 1295−1307.(57) Bagal, S. K.; Andrews, M.; Bechle, B. M.; Bian, J.; Bilsland, J.;Blakemore, D. C.; Braganza, J. F.; Bungay, P. J.; Corbett, M. S.;Cronin, C. N.; Cui, J. J.; Dias, R.; Flanagan, N. J.; Greasley, S. E.;Grimley, R.; James, K.; Johnson, E.; Kitching, L.; Kraus, M. L.;McAlpine, I.; Nagata, A.; Ninkovic, S.; Omoto, K.; Scales, S.; Skerratt,S. E.; Sun, J.; Tran-Dube, M.; Waldron, G. J.; Wang, F.; Warmus, J. S.Discovery of Potent, Selective, and Peripherally Restricted Pan-TrkKinase Inhibitors for the Treatment of Pain. J. Med. Chem. 2018, 61,6779−6800.(58) Kaniskan, H. U.; Eram, M. S.; Zhao, K.; Szewczyk, M. M.;Yang, X.; Schmidt, K.; Luo, X.; Xiao, S.; Dai, M.; He, F.; Zang, I.; Lin,Y.; Li, F.; Dobrovetsky, E.; Smil, D.; Min, S. J.; Lin-Jones, J.; Schapira,M.; Atadja, P.; Li, E.; Barsyte-Lovejoy, D.; Arrowsmith, C. H.; Brown,P. J.; Liu, F.; Yu, Z.; Vedadi, M.; Jin, J. Discovery of Potent andSelective Allosteric Inhibitors of Protein Arginine Methyltransferase 3(PRMT3). J. Med. Chem. 2018, 61, 1204−1217.(59) Ortiz Zacarías, N. V.; van Veldhoven, J. P. D.; Portner, L.; vanSpronsen, E.; Ullo, S.; Veenhuizen, M.; van der Velden, W. J. C.;Zweemer, A. J. M.; Kreekel, R. M.; Oenema, K.; Lenselink, E. B.;Heitman, L. H.; Ijzerman, A. P. Pyrrolone Derivatives as IntracellularAllosteric Modulators for Chemokine Receptors: Selective and Dual-Targeting Inhibitors of CC Chemokine Receptors 1 and 2. J. Med.Chem. 2018, 61, 9146−9161.(60) Davis, M. I.; Hunt, J. P.; Herrgard, S.; Ciceri, P.; Wodicka, L.M.; Pallares, G.; Hocker, M.; Treiber, D. K.; Zarrinkar, P. P.

Comprehensive Analysis of Kinase Inhibitor Selectivity. Nat.Biotechnol. 2011, 29, 1046−1051.(61) Wu, P.; Clausen, M. H.; Nielsen, T. E. Allosteric Small-Molecule Kinase Inhibitors. Pharmacol. Ther. 2015, 156, 59−68.(62) Lewis, J. A.; Lebois, E. P.; Lindsley, C. W. AllostericModulation of Kinases and GPCRs: Design Principles and StructuralDiversity. Curr. Opin. Chem. Biol. 2008, 12, 269−280.(63) Comess, K. M.; Sun, C.; Abad-Zapatero, C.; Goedken, E. R.;Gum, R. J.; Borhani, D. W.; Argiriadi, M.; Groebe, D. R.; Jia, Y.;Clampit, J. E.; Haasch, D. L.; Smith, H. T.; Wang, S.; Song, D.; Coen,M. L.; Cloutier, T. E.; Tang, H.; Cheng, X.; Quinn, C.; Liu, B.; Xin,Z.; Liu, G.; Fry, E. H.; Stoll, V.; Ng, T. I.; Banach, D.; Marcotte, D.;Burns, D. J.; Calderwood, D. J.; Hajduk, P. J. Discovery andCharacterization of Non-ATP Site Inhibitors of the MitogenActivated Protein (MAP) Kinases. ACS Chem. Biol. 2011, 6, 234−244.(64) Sherbenou, D. W.; Hantschel, O.; Kaupe, I.; Willis, S.; Bumm,T.; Turaga, L. P.; Lange, T.; Dao, K. H.; Press, R. D.; Druker, B. J.;Superti-Furga, G.; Deininger, M. W. BCR-ABL SH3-SH2 DomainMutations in Chronic Myeloid Leukemia Patients on Imatinib. Blood2010, 116, 3278−3285.(65) Schoepfer, J.; Jahnke, W.; Berellini, G.; Buonamici, S.; Cotesta,S.; Cowan-Jacob, S. W.; Dodd, S.; Drueckes, P.; Fabbro, D.; Gabriel,T.; Groell, J. M.; Grotzfeld, R. M.; Hassan, A. Q.; Henry, C.; Iyer, V.;Jones, D.; Lombardo, F.; Loo, A.; Manley, P. W.; Pelle, X.; Rummel,G.; Salem, B.; Warmuth, M.; Wylie, A. A.; Zoller, T.; Marzinzik, A. L.;Furet, P. Discovery of Asciminib (ABL001), an Allosteric Inhibitor ofthe Tyrosine Kinase Activity of BCR-ABL1. J. Med. Chem. 2018, 61,8120−8135.(66) Wylie, A. A.; Schoepfer, J.; Jahnke, W.; Cowan-Jacob, S. W.;Loo, A.; Furet, P.; Marzinzik, A. L.; Pelle, X.; Donovan, J.; Zhu, W.;Buonamici, S.; Hassan, A. Q.; Lombardo, F.; Iyer, V.; Palmer, M.;Berellini, G.; Dodd, S.; Thohan, S.; Bitter, H.; Branford, S.; Ross, D.M.; Hughes, T. P.; Petruzzelli, L.; Vanasse, K. G.; Warmuth, M.;Hofmann, F.; Keen, N. J.; Sellers, W. R. The Allosteric InhibitorABL001 Enables Dual Targeting of BCR-ABL1. Nature 2017, 543,733−737.(67) Wei, G.; Xi, W.; Nussinov, R.; Ma, B. Protein Ensembles: HowDoes Nature Harness Thermodynamic Fluctuations for Life? TheDiverse Functional Roles of Conformational Ensembles in the Cell.Chem. Rev. 2016, 116, 6516−6551.(68) Weis, W. I.; Kobilka, B. K. The Molecular Basis of G Protein−Coupled Receptor Activation. Annu. Rev. Biochem. 2018, 87, 897−919.(69) Lazo, J. S.; Sharlow, E. R. Drugging Undruggable MolecularCancer Targets. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 23−40.(70) Dang, C. V.; Reddy, E. P.; Shokat, K. M.; Soucek, L. Druggingthe “Undruggable” Cancer Targets. Nat. Rev. Cancer 2017, 17, 502−508.(71) Shao, H.; Li, X.; Moses, M. A.; Gilbert, L. A.; Kalyanaraman,C.; Young, Z. T.; Chernova, M.; Journey, S. N.; Weissman, J. S.;Hann, B.; Jacobson, M. P.; Neckers, L.; Gestwicki, J. E. Exploration ofBenzothiazole Rhodacyanines as Allosteric Inhibitors of Protein-Protein Interactions with Heat Shock Protein 70 (Hsp70). J. Med.Chem. 2018, 61, 6163−6177.(72) Pricer, R.; Gestwicki, J. E.; Mapp, A. K. From Fuzzy toFunction: The New Frontier of Protein-Protein Interactions. Acc.Chem. Res. 2017, 50, 584−589.(73) Lu, S.; Ji, M.; Ni, D.; Zhang, J. Discovery of Hidden AllostericSites as Novel Targets for Allosteric Drug Design. Drug DiscoveryToday 2018, 23, 359−365.(74) Brogi, S.; Ramunno, A.; Savi, L.; Chemi, G.; Alfano, G.;Pecorelli, A.; Pambianchi, E.; Galatello, P.; Compagnoni, G.; Focher,F.; Biamonti, G.; Valacchi, G.; Butini, S.; Gemma, S.; Campiani, G.;Brindisi, M. First Dual AK/GSK-3β Inhibitors Endowed withAntioxidant Properties as Multifunctional, Potential NeuroprotectiveAgents. Eur. J. Med. Chem. 2017, 138, 438−457.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

M

Page 14: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

(75) An, X.; Lu, S.; Song, K.; Shen, Q.; Huang, M.; Yao, X.; Liu, H.;Zhang, J. Are the Apo Proteins Suitable for the Rational Discovery ofAllosteric Drugs? J. Chem. Inf. Model. 2019, 59, 597−604.(76) Lu, S.; Jang, H.; Gu, S.; Zhang, J.; Nussinov, R. Drugging RasGTPase: A Comprehensive Mechanistic and Signaling StructuralView. Chem. Soc. Rev. 2016, 45, 4929−4952.(77) Cox, A. D.; Fesik, S. W.; Kimmelman, A. C.; Luo, J.; Der, C. J.Drugging the Undruggable RAS: Mission Possible? Nat. Rev. DrugDiscovery 2014, 13, 828−851.(78) Gustafson, W. C.; Meyerowitz, J. G.; Nekritz, E. A.; Chen, J.;Benes, C.; Charron, E.; Simonds, E. F.; Seeger, R.; Matthay, K. K.;Hertz, N. T.; Eilers, M.; Shokat, K. M.; Weiss, W. A. Drugging MYCNthrough an Allosteric Transition in Aurora Kinase A. Cancer Cell2014, 26, 414−427.(79) Liu, F.; Rehmani, I.; Esaki, S.; Fu, R.; Chen, L.; de Serrano, V.;Liu, A. Pirin Is an Iron-Dependent Redox Regulator of NF- B. Proc.Natl. Acad. Sci. U. S. A. 2013, 110, 9722−9727.(80) Krishnan, N.; Konidaris, K. F.; Gasser, G.; Tonks, N. K. APotent, Selective, and Orally Bioavailable Inhibitor of the Protein-Tyrosine Phosphatase PTP1B Improves Insulin and Leptin Signalingin Animal Models. J. Biol. Chem. 2018, 293, 1517−1525.(81) Xie, J.; Si, X.; Gu, S.; Wang, M.; Shen, J.; Li, H.; Li, D.; Fang,Y.; Liu, C.; Zhu, J. Allosteric Inhibitors of SHP2 with TherapeuticPotential for Cancer Treatment. J. Med. Chem. 2017, 60, 10205−10219.(82) Krishnan, N.; Koveal, D.; Miller, D. H.; Xue, B.; Akshinthala, S.D.; Kragelj, J.; Jensen, M. R.; Gauss, C. M.; Page, R.; Blackledge, M.;Muthuswamy, S. K.; Peti, W.; Tonks, N. K. Targeting the DisorderedC Terminus of PTP1B with an Allosteric Inhibitor. Nat. Chem. Biol.2014, 10, 558−566.(83) Jiang, L.; Stanevich, V.; Satyshur, K. A.; Kong, M.; Watkins, G.R.; Wadzinski, B. E.; Sengupta, R.; Xing, Y. Structural Basis of ProteinPhosphatase 2A Stable Latency. Nat. Commun. 2013, 4, 1611−1699.(84) Scheuermann, T. H.; Li, Q.; Ma, H. W.; Key, J.; Zhang, L.;Chen, R.; Garcia, J. A.; Naidoo, J.; Longgood, J.; Frantz, D. E.;Tambar, U. K.; Gardner, K. H.; Bruick, R. K. Allosteric Inhibition ofHypoxia Inducible Factor-2 with Small Molecules. Nat. Chem. Biol.2013, 9, 271−276.(85) Minus, M. B.; Liu, W.; Vohidov, F.; Kasembeli, M. M.; Long,X.; Krueger, M. J.; Stevens, A.; Kolosov, M. I.; Tweardy, D. J.; Sison,E. A. R.; Redell, M. S.; Ball, Z. T. Rhodium(II) Proximity-LabelingIdentifies a Novel Target Site on STAT3 for Inhibitors with PotentAnti-Leukemia Activity. Angew. Chem., Int. Ed. 2015, 54, 13085−13089.(86) Lu, S.; Zhang, J. Allosteric Modulators. Compr. Med. Chem. III2017, 2, 276−296.(87) Lu, S.; Huang, W.; Wang, Q.; Shen, Q.; Li, S.; Nussinov, R.;Zhang, J. The Structural Basis of ATP as an Allosteric Modulator.PLoS Comput. Biol. 2014, 10, No. e1003831.(88) Yang, J.-S.; Seo, S. W.; Jang, S.; Jung, G. Y.; Kim, S. RationalEngineering of Enzyme Allosteric Regulation through SequenceEvolution Analysis. PLoS Comput. Biol. 2012, 8, No. e1002612.(89) Lu, S.; Huang, W.; Zhang, J. Recent Computational Advancesin the Identification of Allosteric Sites in Proteins. Drug DiscoveryToday 2014, 19, 1595−1600.(90) Schueler-Furman, O.; Wodak, S. J. Computational Approachesto Investigating Allostery. Curr. Opin. Struct. Biol. 2016, 41, 159−171.(91) Pendergrass, D. C.; Williams, R.; Blair, J. B.; Fenton, A. W.Mining for Allosteric Information: Natural Mutations and PositionalSequence Conservation in Pyruvate Kinase. IUBMB Life 2006, 58,31−38.(92) Smith, N. J.; Milligan, G. Allostery at G Protein-CoupledReceptor Homo- and Heteromers: Uncharted PharmacologicalLandscapes. Pharmacol. Rev. 2010, 62, 701−725.(93) van Westen, G. J. P.; Gaulton, A.; Overington, J. P. Chemical,Target, and Bioactive Properties of Allosteric Modulation. PLoSComput. Biol. 2014, 10, No. e1003559.(94) Shi, Y. A Glimpse of Structural Biology through X-RayCrystallography. Cell 2014, 159, 995−1014.

(95) Lu, S.; Zhang, J. Small Molecule Allosteric Modulators of G-Protein-Coupled Receptors: Drug−Target Interactions. J. Med. Chem.2019, 62, 24−45.(96) Beck, F.; Unverdorben, P.; Bohn, S.; Schweitzer, A.; Pfeifer, G.;Sakata, E.; Nickell, S.; Plitzko, J. M.; Villa, E.; Baumeister, W.; Forster,F. Near-Atomic Resolution Structural Model of the Yeast 26SProteasome. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 14870−14875.(97) Chen, S.; Oldham, M. L.; Davidson, A. L.; Chen, J. CarbonCatabolite Repression of the Maltose Transporter Revealed by X-RayCrystallography. Nature 2013, 499, 364−368.(98) Zhou, L.; Hang, J.; Zhou, Y.; Wan, R.; Lu, G.; Yin, P.; Yan, C.;Shi, Y. Crystal Structures of the Lsm Complex Bound to the 3′ EndSequence of U6 Small Nuclear RNA. Nature 2014, 506, 116−120.(99) Stass, R.; Ilca, S. L.; Huiskonen, J. T. Beyond Structures ofHighly Symmetric Purified Viral Capsids by Cryo-EM. Curr. Opin.Struct. Biol. 2018, 52, 25−31.(100) Pospich, S.; Raunser, S. Single Particle Cryo-EM anOptimal Tool to Study Cytoskeletal Proteins. Curr. Opin. Struct. Biol.2018, 52, 16−24.(101) Fica, S. M.; Nagai, K. Cryo-Electron Microscopy Snapshots ofthe Spliceosome: Structural Insights into a Dynamic Ribonucleopro-tein Machine. Nat. Struct. Mol. Biol. 2017, 24, 791−799.(102) Renaud, J. P.; Chari, A.; Ciferri, C.; Liu, W. T.; Remigy, H.W.; Stark, H.; Wiesmann, C. Cryo-EM in Drug Discovery:Achievements, Limitations and Prospects. Nat. Rev. Drug Discovery2018, 17, 471−492.(103) Grutsch, S.; Bruschweiler, S.; Tollinger, M. NMR Methods toStudy Dynamic Allostery. PLoS Comput. Biol. 2016, 12, e1004620.(104) Tzeng, S. R.; Kalodimos, C. G. Protein Dynamics andAllostery: An NMR View. Curr. Opin. Struct. Biol. 2011, 21, 62−67.(105) Boulton, S.; Melacini, G. Advances in NMR Methods to MapAllosteric Sites: From Models to Translation. Chem. Rev. 2016, 116,6267−6304.(106) Lisi, G. P.; Loria, J. P. Solution NMR Spectroscopy for theStudy of Enzyme Allostery. Chem. Rev. 2016, 116, 6323−6369.(107) Jiang, Y.; Kalodimos, C. G. NMR Studies of Large Proteins. J.Mol. Biol. 2017, 429, 2667−2676.(108) Velyvis, A.; Kay, L. E. Measurement of Active Site IonizationEquilibria in the 670 kDa Proteasome Core Particle Using Methyl-TROSY NMR. J. Am. Chem. Soc. 2013, 135, 9259−9262.(109) Karagoz, G. E.; Duarte, A. M. S.; Akoury, E.; Ippel, H.;Biernat, J.; Moran Luengo, T.; Radli, M.; Didenko, T.; Nordhues, B.A.; Veprintsev, D. B.; Dickey, C. A.; Mandelkow, E.; Zweckstetter, M.;Boelens, R.; Madl, T.; Rudiger, S. G. D. Hsp90-Tau Complex RevealsMolecular Basis for Specificity in Chaperone Action. Cell 2014, 156,963−974.(110) Libich, D. S.; Fawzi, N. L.; Ying, J.; Clore, G. M. Probing theTransient Dark State of Substrate Binding to GroEL by Relaxation-Based Solution NMR. Proc. Natl. Acad. Sci. U. S. A. 2013, 110,11361−11366.(111) Ostrem, J. M.; Peters, U.; Sos, M. L.; Wells, J. A.; Shokat, K.M. K-Ras(G12C) Inhibitors Allosterically Control GTP Affinity andEffector Interactions. Nature 2013, 503, 548−551.(112) Boehlein, S. K.; Shaw, J. R.; Hannah, L. C.; Stewart, J. D.Probing Allosteric Binding Sites of the Maize Endosperm ADP-Glucose Pyrophosphorylase. Plant Physiol. 2010, 152, 85−95.(113) Schneider, R.; Becker, C.; Simard, J. R.; Getlik, M.; Bohlke,N.; Janning, P.; Rauh, D. Direct Binding Assay for the Detection ofType IV Allosteric Inhibitors of Abl. J. Am. Chem. Soc. 2012, 134,9138−9141.(114) Schneider, R.; Beumer, C.; Simard, J. R.; Grutter, C.; Rauh, D.Selective Detection of Allosteric Phosphatase Inhibitors. J. Am. Chem.Soc. 2013, 135, 6838−6841.(115) Magnaghi, P.; D’Alessio, R.; Valsasina, B.; Avanzi, N.; Rizzi, S.;Asa, D.; Gasparri, F.; Cozzi, L.; Cucchi, U.; Orrenius, C.; Polucci, P.;Ballinari, D.; Perrera, C.; Leone, A.; Cervi, G.; Casale, E.; Xiao, Y.;Wong, C.; Anderson, D. J.; Galvani, A.; Donati, D.; O’Brien, T.;Jackson, P. K.; Isacchi, A. Covalent and Allosteric Inhibitors of the

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

N

Page 15: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

ATPase VCP/p97 Induce Cancer Cell Death. Nat. Chem. Biol. 2013,9, 548−559.(116) de Vera, I. M. S.; Zheng, J.; Novick, S.; Shang, J.; Hughes, T.S.; Brust, R.; Munoz-Tello, P.; Gardner, W. J.; Marciano, D. P.; Kong,X.; Griffin, P. R.; Kojetin, D. J. Synergistic Regulation of Coregulator/Nuclear Receptor Interaction by Ligand and DNA. Structure 2017, 25,1506−1518.(117) Greener, J. G.; Sternberg, M. J. Structure-Based Prediction ofProtein Allostery. Curr. Opin. Struct. Biol. 2018, 50, 1−8.(118) Huang, Z.; Zhu, L.; Cao, Y.; Wu, G.; Liu, X.; Chen, Y.; Wang,Q.; Shi, T.; Zhao, Y.; Wang, Y.; Li, W.; Li, Y.; Chen, H.; Chen, G.;Zhang, J. ASD: A Comprehensive Database of Allosteric Proteins andModulators. Nucleic Acids Res. 2011, 39, D663−D669.(119) Huang, Z.; Mou, L.; Shen, Q.; Lu, S.; Li, C.; Liu, X.; Wang,G.; Li, S.; Geng, L.; Liu, Y.; Wu, J.; Chen, G.; Zhang, J. ASD v2.0:Updated Content and Novel Features Focusing on AllostericRegulation. Nucleic Acids Res. 2014, 42, D510−D516.(120) Huang, W.; Wang, G.; Shen, Q.; Liu, X.; Lu, S.; Geng, L.;Huang, Z.; Zhang, J. ASBench: Benchmarking Sets for AllostericDiscovery. Bioinformatics 2015, 31, 2598−2600.(121) Huang, W.; Lu, S.; Huang, Z.; Liu, X.; Mou, L.; Luo, Y.; Zhao,Y.; Liu, Y.; Chen, Z.; Hou, T.; Zhang, J. Allosite: A Method forPredicting Allosteric Sites. Bioinformatics 2013, 29, 2357−2359.(122) Song, K.; Liu, X.; Huang, W.; Lu, S.; Shen, Q.; Zhang, L.;Zhang, J. Improved Method for the Identification and Validation ofAllosteric Sites. J. Chem. Inf. Model. 2017, 57, 2358−2363.(123) Akbar, R.; Helms, V. ALLO: A Tool to Discriminate andPrioritize Allosteric Pockets. Chem. Biol. Drug Des. 2018, 91, 845−853.(124) Chen, A. S. Y.; Westwood, N. J.; Brear, P.; Rogers, G. W.;Mavridis, L.; Mitchell, J. B. O. A Random Forest Model for PredictingAllosteric and Functional Sites on Proteins. Mol. Inf. 2016, 35, 125−135.(125) Smock, R. G.; Rivoire, O.; Russ, W. P.; Swain, J. F.; Leibler, S.;Ranganathan, R.; Gierasch, L. M. An Interdomain Sector MediatingAllostery in Hsp70 Molecular Chaperones. Mol. Syst. Biol. 2010, 6, 1−10.(126) Greener, J. G.; Sternberg, M. J. AlloPred: Prediction ofAllosteric Pockets on Proteins Using Normal Mode PerturbationAnalysis. BMC Bioinf. 2015, 16, 335.(127) Panjkovich, A.; Daura, X. PARS: A Web Server for thePrediction of Protein Allosteric and Regulatory Sites. Bioinformatics2014, 30, 1314−1315.(128) Qi, Y.; Wang, Q.; Tang, B.; Lai, L. Identifying AllostericBinding Sites in Proteins with a Two-State Go? Model for NovelAllosteric Effector Discovery. J. Chem. Theory Comput. 2012, 8,2962−2971.(129) Greener, J. G.; Filippis, I.; Sternberg, M. J. E. PredictingProtein Dynamics and Allostery Using Multi-Protein Atomic DistanceConstraints. Structure 2017, 25, 546−558.(130) De Vivo, M.; Masetti, M.; Bottegoni, G.; Cavalli, A. Role ofMolecular Dynamics and Related Methods in Drug Discovery. J. Med.Chem. 2016, 59, 4035−4061.(131) Weinkam, P.; Pons, J.; Sali, A. Structure-Based Model ofAllostery Predicts Coupling between Distant Sites. Proc. Natl. Acad.Sci. U. S. A. 2012, 109, 4875−4880.(132) Tee, W.-V.; Guarnera, E.; Berezovsky, I. N. ReversingAllosteric Communication: From Detecting Allosteric Sites toInducing and Tuning Targeted Allosteric Response. PLoS Comput.Biol. 2018, 14, No. e1006228.(133) Ma, X.; Meng, H.; Lai, L. Motions of Allosteric andOrthosteric Ligand-Binding Sites in Proteins Are Highly Correlated.J. Chem. Inf. Model. 2016, 56, 1725−1733.(134) Kaya, C.; Armutlulu, A.; Ekesan, S.; Haliloglu, T. MCPath:Monte Carlo Path Generation Approach to Predict Likely AllostericPathways and Functional Residues. Nucleic Acids Res. 2013, 41,W249−W255.(135) Goncearenco, A.; Mitternacht, S.; Yong, T.; Eisenhaber, B.;Eisenhaber, F.; Berezovsky, I. N. SPACER: Server for Predicting

Allosteric Communication and Effects of Regulation. Nucleic AcidsRes. 2013, 41, 266−272.(136) Li, H.; Chang, Y. Y.; Lee, J. Y.; Bahar, I.; Yang, L. W.DynOmics: Dynamics of Structural Proteome and Beyond. NucleicAcids Res. 2017, 45, W374−W380.(137) Guarnera, E.; Tan, Z. W.; Zheng, Z.; Berezovsky, I. N.AlloSigMA: Allosteric Signaling and Mutation Analysis Server.Bioinformatics 2017, 33, 3996−3998.(138) Tan, Z. W.; Tee, W.-V.; Guarnera, E.; Booth, L.; Berezovsky, I.N. AlloMAPS: Allosteric Mutation Analysis and Polymorphism ofSignaling Database. Nucleic Acids Res. 2019, 47, D265−D270.(139) Shen, Q.; Cheng, F.; Song, H.; Lu, W.; Zhao, J.; An, X.; Liu,M.; Chen, G.; Zhao, Z.; Zhang, J. Proteome-Scale Investigation ofProtein Allosteric Regulation Perturbed by Somatic Mutations in7,000 Cancer Genomes. Am. J. Hum. Genet. 2017, 100, 5−20.(140) Clarke, D.; Sethi, A.; Li, S.; Kumar, S.; Chang, R. W. F.; Chen,J.; Gerstein, M. Identifying Allosteric Hotspots with Dynamics:Application to Inter- and Intra-Species Conservation. Structure 2016,24, 826−837.(141) Li, S.; Shen, Q.; Su, M.; Liu, X.; Lu, S.; Chen, Z.; Wang, R.;Zhang, J. Alloscore: A Method for Predicting Allosteric Ligand−protein Interactions. Bioinformatics 2016, 32, 1574−1576.(142) Xu, Y.; Wang, S.; Hu, Q.; Gao, S.; Ma, X.; Zhang, W.; Shen,Y.; Chen, F.; Lai, L.; Pei, J. CavityPlus: A Web Server for ProteinCavity Detection with Pharmacophore Modelling, Allosteric SiteIdentification and Covalent Ligand Binding Ability Prediction. NucleicAcids Res. 2018, 46, W374−W379.(143) Smith, R. D.; Lu, J.; Carlson, H. A. Are There PhysicochemicalDifferences between Allosteric and Competitive Ligands? PLoSComput. Biol. 2017, 13, No. e1005813.(144) Beauharnois, J. M.; Bolívar, B. E.; Welch, J. T. Sirtuin 6: AReview of Biological Effects and Potential Therapeutic Properties.Mol. BioSyst. 2013, 9, 1789−1806.(145) Tennen, R. I.; Chua, K. F. Chromatin Regulation and GenomeMaintenance by Mammalian SIRT6. Trends Biochem. Sci. 2011, 36,39−46.(146) Mostoslavsky, R.; Chua, K. F.; Lombard, D. B.; Pang, W. W.;Fischer, M. R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.;Murphy, M. M.; Mills, K. D.; Patel, P.; Hsu, J. T.; Hong, A. L.; Ford,E.; Cheng, H. L.; Kennedy, C.; Nunez, N.; Bronson, R.; Frendewey,D.; Auerbach, W.; Valenzuela, D.; Karow, M.; Hottiger, M. O.;Hursting, S.; Barrett, J. C.; Guarente, L.; Mulligan, R.; Demple, B.;Yancopoulos, G. D.; Alt, F. W. Genomic Instability and Aging-likePhenotype in the Absence of Mammalian SIRT6. Cell 2006, 124,315−329.(147) Kanfi, Y.; Naiman, S.; Amir, G.; Peshti, V.; Zinman, G.;Nahum, L.; Bar-Joseph, Z.; Cohen, H. Y. The Sirtuin SIRT6 RegulatesLifespan in Male Mice. Nature 2012, 483, 218−221.(148) Sebastian, C.; Zwaans, B. M. M.; Silberman, D. M.; Gymrek,M.; Goren, A.; Zhong, L.; Ram, O.; Truelove, J.; Guimaraes, A. R.;Toiber, D.; Cosentino, C.; Greenson, J. K.; MacDonald, A. I.;McGlynn, L.; Maxwell, F.; Edwards, J.; Giacosa, S.; Guccione, E.;Weissleder, R.; Bernstein, B. E.; Regev, A.; Shiels, P. G.; Lombard, D.B.; Mostoslavsky, R. The Histone Deacetylase SIRT6 Is a TumorSuppressor That Controls Cancer Metabolism. Cell 2012, 151, 1185−1199.(149) Schiedel, M.; Robaa, D.; Rumpf, T.; Sippl, W.; Jung, M. TheCurrent State of NAD+ -Dependent Histone Deacetylases (Sirtuins)as Novel Therapeutic Targets. Med. Res. Rev. 2018, 38, 147−200.(150) Sanchez-Fidalgo, S.; Villegas, I.; Sanchez-Hidalgo, M.; Alarconde la Lastra, C. Sirtuin Modulators: Mechanisms and PotentialClinical Implications. Curr. Med. Chem. 2012, 19, 2414−2441.(151) Wang, X.; Crowe, P. J.; Goldstein, D.; Yang, J. L. STAT3Inhibition, a Novel Approach to Enhancing Targeted Therapy inHuman Cancers (Review). Int. J. Oncol. 2012, 41, 1181−1191.(152) Zhao, C.; Li, H.; Lin, H. J.; Yang, S.; Lin, J.; Liang, G.Feedback Activation of STAT3 as a Cancer Drug-ResistanceMechanism. Trends Pharmacol. Sci. 2016, 37, 47−61.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

O

Page 16: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

(153) Sgrignani, J.; Garofalo, M.; Matkovic, M.; Merulla, J.;Catapano, C. V.; Cavalli, A. Structural Biology of STAT3 and ItsImplications for Anticancer Therapies Development. Int. J. Mol. Sci.2018, 19, 1591.(154) Debnath, B.; Xu, S.; Neamati, N. Small Molecule Inhibitors ofSignal Transducer and Activator of Transcription 3 (stat3) Protein. J.Med. Chem. 2012, 55, 6645−6648.(155) Miklossy, G.; Youn, U. J.; Yue, P.; Zhang, M.; Chen, C. H.;Hilliard, T. S.; Paladino, D.; Li, Y.; Choi, J.; Sarkaria, J. N.; Kawakami,J. K.; Wongwiwatthananukit, S.; Chen, Y.; Sun, D.; Chang, L. C.;Turkson, J. Hirsutinolide Series Inhibit Stat3 Activity, Alter GCN1,MAP1B, Hsp105, G6PD, Vimentin, TrxR1, and Importin α-2Expression, and Induce Antitumor Effects against Human Glioma. J.Med. Chem. 2015, 58, 7734−7748.(156) Escobar, Z.; Bjartell, A.; Canesin, G.; Evans-Axelsson, S.;Sterner, O.; Hellsten, R.; Johansson, M. H. Preclinical Character-ization of 3β-(N-Acetyl L -Cysteine Methyl Ester)-2aβ,3-Dihydroga-liellalactone (GPA512), a Prodrug of a Direct STAT3 Inhibitor forthe Treatment of Prostate Cancer. J. Med. Chem. 2016, 59, 4551−4562.(157) Yu, W.; Li, C.; Zhang, W.; Xia, Y.; Li, S.; Lin, J.-y.; Yu, K.; Liu,M.; Yang, L.; Luo, J.; Chen, Y.; Sun, H.; Kong, L. Discovery of anOrally Selective Inhibitor of Signal Transducer and Activator ofTranscription 3 Using Advanced Multiple Ligand SimultaneousDocking. J. Med. Chem. 2017, 60, 2718−2731.(158) Beebe, J. D.; Liu, J. Y.; Zhang, J. T. Two Decades of Researchin Discovery of Anticancer Drugs Targeting STAT3, How Close AreWe? Pharmacol. Ther. 2018, 191, 74−91.(159) Trott, O.; Olson, A. J. AutoDock Vina: Improving the Speedand Accuracy of Docking with a New Scoring Function, EfficientOptimization, and Multithreading. J. Comput. Chem. 2010, 31, 455−461.(160) Savaskan, N. E.; Ufer, C.; Kuhn, H.; Borchert, A. MolecularBiology of Glutathione Peroxidase 4: From Genomic Structure toDevelopmental Expression and Neural Function. Biol. Chem. 2007,388, 1007−1017.(161) Yang, W. S.; Sriramaratnam, R.; Welsch, M. E.; Shimada, K.;Skouta, R.; Viswanathan, V. S.; Cheah, J. H.; Clemons, P. A.; Shamji,A. F.; Clish, C. B.; Brown, L. M.; Girotti, A. W.; Cornish, V. W.;Schreiber, S. L.; Stockwell, B. R. Regulation of Ferroptotic CancerCell Death by GPX4. Cell 2014, 156, 317−331.(162) Li, C.; Deng, X.; Xie, X.; Liu, Y.; Friedmann Angeli, J. P.; Lai,L. Activation of Glutathione Peroxidase 4 as a Novel Anti-Inflammatory Strategy. Front. Pharmacol. 2018, 9, 1−12.(163) Yuan, Y.; Pei, J.; Lai, L. LigBuilder 2: A Practical de NovoDrug Design Approach. J. Chem. Inf. Model. 2011, 51, 1083−1091.(164) Lee, J.; Natarajan, M.; Nashine, V. C.; Socolich, M.; Vo, T.;Russ, W. P.; Benkovic, S. J.; Ranganathan, R. Surface Sites forEngineering Allosteric Control in Proteins. Science 2008, 322, 438−442.(165) Reynolds, K. A.; McLaughlin, R. N.; Ranganathan, R. HotSpots for Allosteric Regulation on Protein Surfaces. Cell 2011, 147,1564−1575.(166) Fodor, A. A.; Aldrich, R. W. On Evolutionary Conservation ofThermodynamic Coupling in Proteins. J. Biol. Chem. 2004, 279,19046−19050.(167) Chi, C. N.; Elfstrom, L.; Shi, Y.; Snall, T.; Engstrom, A.;Jemth, P. Reassessing a Sparse Energetic Network within a SingleProtein Domain. Proc. Natl. Acad. Sci. U. S. A. 2008, 105, 4679−4684.(168) Liu, Z.; Chen, J.; Thirumalai, D. On the Accuracy of InferringEnergetic Coupling between Distant Sites in Protein Families fromEvolutionary Imprints: Illustrations Using Lattice Model. Proteins:Struct., Funct., Genet. 2009, 77, 823−831.(169) Livesay, D. R.; Kreth, K. E.; Fodor, A. A. A Critical Evaluationof Correlated Mutation Algorithms and Coevolution within AllostericMechanisms. Methods Mol. Biol. 2012, 796, 385−398.(170) Anishchenko, I.; Ovchinnikov, S.; Kamisetty, H.; Baker, D.Origins of Coevolution between Residues Distant in Protein 3DStructures. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 9122−9127.

(171) Novinec, M.; Korenc, M.; Caflisch, A.; Ranganathan, R.;Lenarcic, B.; Baici, A. A Novel Allosteric Mechanism in the CysteinePeptidase Cathepsin K Discovered by Computational Methods. Nat.Commun. 2014, 5, 3287.(172) Harris, R.; Olson, A. J.; Goodsell, D. S. Automated Predictionof Ligand-Binding Sites in Proteins. Proteins: Struct., Funct., Genet.2008, 70, 1506−1517.(173) McLaughlin, R. N.; Poelwijk, F. J.; Raman, A.; Gosal, W. S.;Ranganathan, R. The Spatial Architecture of Protein Function andAdaptation. Nature 2012, 491, 138−142.(174) Hatley, M. E.; Lockless, S. W.; Gibson, S. K.; Gilman, A. G.;Ranganathan, R. Allosteric Determinants in Guanine Nucleotide-Binding Proteins. Proc. Natl. Acad. Sci. U. S. A. 2003, 100, 14445−14450.(175) Ferguson, A. D.; Amezcua, C. A.; Halabi, N. M.; Chelliah, Y.;Rosen, M. K.; Ranganathan, R.; Deisenhofer, J. Signal TransductionPathway of TonB-Dependent Transporters. Proc. Natl. Acad. Sci. U. S.A. 2007, 104, 513−518.(176) Guo, J.; Zhou, H. X. Protein Allostery and ConformationalDynamics. Chem. Rev. 2016, 116, 6503−6515.(177) Perilla, J. R.; Goh, B. C.; Cassidy, C. K.; Liu, B.; Bernardi, R.C.; Rudack, T.; Yu, H.; Wu, Z.; Schulten, K. Molecular DynamicsSimulations of Large Macromolecular Complexes. Curr. Opin. Struct.Biol. 2015, 31, 64−74.(178) Elber, R. Simulations of Allosteric Transitions. Curr. Opin.Struct. Biol. 2011, 21, 167−172.(179) Ni, D.; Song, K.; Zhang, J.; Lu, S. Molecular DynamicsSimulations and Dynamic Network Analysis Reveal the AllostericUnbinding of Monobody to H-Ras Triggered by R135K Mutation.Int. J. Mol. Sci. 2017, 18, 2249.(180) Ni, D.; Liu, D.; Zhang, J.; Lu, S. Computational Insights intothe Interactions between Calmodulin and the c/nSH2 Domains ofp85α Regulatory Subunit of PI3Kα: Implication for PI3Kα Activationby Calmodulin. Int. J. Mol. Sci. 2018, 19, 151.(181) Stank, A.; Kokh, D. B.; Fuller, J. C.; Wade, R. C. ProteinBinding Pocket Dynamics. Acc. Chem. Res. 2016, 49, 809−815.(182) Dror, R. O.; Green, H. F.; Valant, C.; Borhani, D. W.;Valcourt, J. R.; Pan, A. C.; Arlow, D. H.; Canals, M.; Lane, J. R.;Rahmani, R.; Baell, J. B.; Sexton, P. M.; Christopoulos, A.; Shaw, D. E.Structural Basis for Modulation of a G-Protein-Coupled Receptor byAllosteric Drugs. Nature 2013, 503, 295−299.(183) Hertig, S.; Latorraca, N. R.; Dror, R. O. Revealing Atomic-Level Mechanisms of Protein Allostery with Molecular DynamicsSimulations. PLoS Comput. Biol. 2016, 12, No. e1004746.(184) Lu, S.; Banerjee, A.; Jang, H.; Zhang, J.; Gaponenko, V.;Nussinov, R. GTP Binding and Oncogenic Mutations May AttenuateHypervariable Region (HVR)-Catalytic Domain Interactions in SmallGTPase K-Ras4B, Exposing the Effector Binding Site. J. Biol. Chem.2015, 290, 28887−28900.(185) Shukla, D.; Meng, Y.; Roux, B.; Pande, V. S. ActivationPathway of Src Kinase Reveals Intermediate States as Targets forDrug Design. Nat. Commun. 2014, 5, 3397.(186) Betzi, S.; Alam, R.; Martin, M.; Lubbers, D. J.; Han, H.;Jakkaraj, S. R.; Georg, G. I.; Schonbrunn, E. Discovery of a PotentialAllosteric Ligand Binding Site in CDK2. ACS Chem. Biol. 2011, 6,492−501.(187) Sultan, M. M.; Kiss, G.; Pande, V. S. Towards Simple KineticModels of Functional Dynamics for a Kinase Subfamily. Nat. Chem.2018, 10, 903−909.(188) Husic, B. E.; Pande, V. S. Markov State Models: From an Artto a Science. J. Am. Chem. Soc. 2018, 140, 2386−2396.(189) Fronik, P.; Gaiser, B. I.; Sejer Pedersen, D. Bitopic Ligandsand Metastable Binding Sites: Opportunities for G Protein-CoupledReceptor (GPCR) Medicinal Chemistry. J. Med. Chem. 2017, 60,4126−4134.(190) Bowman, G. R.; Bolin, E. R.; Hart, K. M.; Maguire, B. C.;Marqusee, S. Discovery of Multiple Hidden Allosteric Sites byCombining Markov State Models and Experiments. Proc. Natl. Acad.Sci. U. S. A. 2015, 112, 2734−2739.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

P

Page 17: Allosteric Modulator Discovery: From Serendipity to ... fileallosteric to functional, orthosteric sites is elicited by perturbations at the allosteric site such as effector (ions,

(191) Lu, S.; Li, S.; Zhang, J. Harnessing Allostery: A NovelApproach to Drug Discovery. Med. Res. Rev. 2014, 34, 1242−1285.(192) Dror, R. O.; Dirks, R. M.; Grossman, J. P.; Xu, H.; Shaw, D. E.Biomolecular Simulation: A Computational Microscope for MolecularBiology. Annu. Rev. Biophys. 2012, 41, 429−452.(193) Lee, Y.; Basith, S.; Choi, S. Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled ReceptorsUtilizing Crystal Structures and Computational Simulations. J. Med.Chem. 2018, 61, 1−46.(194) Nussinov, R.; Tsai, C.-J.; Ma, B. The Underappreciated Roleof Allostery in the Cellular Network. Annu. Rev. Biophys. 2013, 42,169−189.(195) Pei, J.; Yin, N.; Ma, X.; Lai, L. Systems Biology Brings NewDimensions for Structure-Based Drug Design. J. Am. Chem. Soc. 2014,136, 11556−11565.(196) Nussinov, R.; Tsai, C.; Liu, J. Principles of AllostericInteractions in Cell Signaling. J. Am. Chem. Soc. 2014, 136, 17692−17701.(197) Csizmok, V.; Follis, A. V.; Kriwacki, R. W.; Forman-Kay, J. D.Dynamic Protein Interaction Networks and New Structural Paradigmsin Signaling. Chem. Rev. 2016, 116, 6424−6462.(198) Nussinov, R.; Tsai, C. J.; Csermely, P. Allo-Network Drugs:Harnessing Allostery in Cellular Networks. Trends Pharmacol. Sci.2011, 32, 686−693.

Journal of Medicinal Chemistry Perspective

DOI: 10.1021/acs.jmedchem.8b01749J. Med. Chem. XXXX, XXX, XXX−XXX

Q


Recommended