+ All Categories
Home > Documents > Druggable protein–protein interactions – from hot spots to hot segments

Druggable protein–protein interactions – from hot spots to hot segments

Date post: 21-Dec-2016
Category:
Upload: ora
View: 215 times
Download: 0 times
Share this document with a friend
8
Druggable proteinprotein interactions from hot spots to hot segments Nir London 1 , Barak Raveh 2 and Ora Schueler-Furman 3 ProteinProtein Interactions (PPIs) mediate numerous biological functions. As such, the inhibition of specific PPIs has tremendous therapeutic value. The notion that these interactions are ‘undruggable’ has petered out with the emergence of more and more successful examples of PPI inhibitors, expanding considerably the scope of potential drug targets. The accumulated data on successes in the inhibition of PPIs allow us to analyze the features that are required for such inhibition. Whereas it has been suggested and shown that targeting hot spots at PPI interfaces is a good strategy to achieve inhibition, in this review we focus on the notion that the most amenable interactions for inhibition are those that are mediated by a ‘hot segment’, a continuous epitope that contributes the majority of the binding energy. This criterion is both useful in guiding future target selection efforts, and in suggesting immediate inhibitory candidates the dominant peptidic segment that mediates the targeted interaction. Addresses 1 Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA 2 Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA 3 Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Hadassah Medical School, The Hebrew University, POB 12272, Jerusalem 91120, Israel Corresponding author: Schueler-Furman, Ora ([email protected]) Current Opinion in Chemical Biology 2013, 17:952959 This review comes from a themed issue Synthetic biomolecules Edited by Shang-Cheng Hung and Derek N Woolfson For a complete overview see the Issue and the Editorial Available online 31st October 2013 1367-5931/$ see front matter, # 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.cbpa.2013.10.011 Introduction Accurate communication between different proteins is critical to the proper functioning of any living cell. The interaction between protein partners not only establishes macromolecular machineries such as ribosomes, poly- merases and proteasomes, but also mediates transient signaling pathways, and timely regulatory processes. ProteinProtein Interactions (PPIs) can be mediated by the classical interaction between two globular protein domains. Alternatively, as much as 40% of the PPIs are estimated to involve peptidedomain interactions, through the binding of a short peptidic linear binding motif to a globular protein domain [1], or even by the co- folding of these linear motifs within two unstructured regions [2]. For some time, protein-protein interfaces were considered relatively flat and featureless (inspired among others by the seminal analysis of proteinprotein interfaces by Thornton in 1996 [3]), and therefore hard to ‘drug’ using small molecules that prefer well-defined binding pockets [4]. However, from early on it was also shown that even seemingly featureless proteinprotein interfaces contain ‘hot spot’ residues [57]. These often involve large amino acids such as Tyrosine, Arginine and Tryptophan that bind in small pockets across the interface and contribute the major part of the binding interaction energy. Additional studies that characterized proteinprotein interfaces revealed a higher-level organization of these interfaces [8]. The prevalence of hot spot residues in both domaindomain and peptidedomain interfaces [9 ] made it theoretically feasible to disrupt proteinprotein (and peptide-protein) interactions with small molecules that target their binding sites. In recent years, the number of successful attempts at inhibiting PPIs with small mol- ecules has increased considerably (and also been exten- sively reviewed by e.g. [1013,14 ] and many others). In this review, we survey distinctive features of proteinprotein interfaces that are amenable for inhibition, and posit that these druggable PPIs are often dominated by ‘hot segments’, an extension of the concept of hot spots to a continuous binding epitope that dominates the interaction [15 ]. The prevalence of hot segments in druggable PPIs highlights peptides and their derivatives as a suitable class of PPI inhibitors. We discuss various techniques for further optimization that can propel these starting points into potential drugs. Characteristics of successfully inhibited PPIs Following the accumulation of examples of small mol- ecules that inhibit PPIs, databases such as TIMBAL [16], 2P2I [17] and iPPI-DB [18] have been dedicated to the collection of small molecule PPI inhibitors and relevant structural data on these PPIs. The 2P2I database organizes inhibited PPIs in two classes: peptidedomain interactions and domaindomain interactions [19 ] (see Figure 1 and Table 1). A structural comparison of these complexes to other heterodimeric complexes suggested that the druggable 2P2I interfaces involve a much smaller buried surface area, are more hydrophobic, and do not show major conformational changes upon binding. Inter- estingly, we previously found that these features, in Available online at www.sciencedirect.com ScienceDirect Current Opinion in Chemical Biology 2013, 17:952959 www.sciencedirect.com
Transcript
Page 1: Druggable protein–protein interactions – from hot spots to hot segments

Druggable protein–protein interactions – from hot spotsto hot segmentsNir London1, Barak Raveh2 and Ora Schueler-Furman3

Available online at www.sciencedirect.com

ScienceDirect

Protein–Protein Interactions (PPIs) mediate numerous

biological functions. As such, the inhibition of specific PPIs has

tremendous therapeutic value. The notion that these

interactions are ‘undruggable’ has petered out with the

emergence of more and more successful examples of PPI

inhibitors, expanding considerably the scope of potential drug

targets. The accumulated data on successes in the inhibition of

PPIs allow us to analyze the features that are required for such

inhibition. Whereas it has been suggested and shown that

targeting hot spots at PPI interfaces is a good strategy to

achieve inhibition, in this review we focus on the notion that the

most amenable interactions for inhibition are those that are

mediated by a ‘hot segment’, a continuous epitope that

contributes the majority of the binding energy. This criterion is

both useful in guiding future target selection efforts, and in

suggesting immediate inhibitory candidates – the dominant

peptidic segment that mediates the targeted interaction.

Addresses1 Department of Pharmaceutical Chemistry, University of California, San

Francisco, San Francisco, CA 94158, USA2 Department of Bioengineering and Therapeutic Sciences, University of

California, San Francisco, San Francisco, CA 94158, USA3 Department of Microbiology and Molecular Genetics, Institute for

Medical Research Israel-Canada, Hadassah Medical School, The

Hebrew University, POB 12272, Jerusalem 91120, Israel

Corresponding author: Schueler-Furman, Ora ([email protected])

Current Opinion in Chemical Biology 2013, 17:952–959

This review comes from a themed issue Synthetic biomolecules

Edited by Shang-Cheng Hung and Derek N Woolfson

For a complete overview see the Issue and the Editorial

Available online 31st October 2013

1367-5931/$ – see front matter, # 2013 Elsevier Ltd. All rights

reserved.

http://dx.doi.org/10.1016/j.cbpa.2013.10.011

IntroductionAccurate communication between different proteins is

critical to the proper functioning of any living cell. The

interaction between protein partners not only establishes

macromolecular machineries such as ribosomes, poly-

merases and proteasomes, but also mediates transient

signaling pathways, and timely regulatory processes.

Protein–Protein Interactions (PPIs) can be mediated by

the classical interaction between two globular protein

domains. Alternatively, as much as 40% of the PPIs are

estimated to involve peptide–domain interactions,

through the binding of a short peptidic linear binding

Current Opinion in Chemical Biology 2013, 17:952–959

motif to a globular protein domain [1], or even by the co-

folding of these linear motifs within two unstructured

regions [2].

For some time, protein-protein interfaces were considered

relatively flat and featureless (inspired among others by the

seminal analysis of protein–protein interfaces by Thornton

in 1996 [3]), and therefore hard to ‘drug’ using small

molecules that prefer well-defined binding pockets [4].

However, from early on it was also shown that even

seemingly featureless protein–protein interfaces contain

‘hot spot’ residues [5–7]. These often involve large amino

acids such as Tyrosine, Arginine and Tryptophan that bind

in small pockets across the interface and contribute the

major part of the binding interaction energy. Additional

studies that characterized protein–protein interfaces

revealed a higher-level organization of these interfaces

[8]. The prevalence of hot spot residues in both

domain–domain and peptide–domain interfaces [9�] made

it theoretically feasible to disrupt protein–protein (and

peptide-protein) interactions with small molecules that

target their binding sites. In recent years, the number of

successful attempts at inhibiting PPIs with small mol-

ecules has increased considerably (and also been exten-

sively reviewed by e.g. [10–13,14��] and many others). In

this review, we survey distinctive features of protein–protein interfaces that are amenable for inhibition, and

posit that these druggable PPIs are often dominated by ‘hot

segments’, an extension of the concept of hot spots to a

continuous binding epitope that dominates the interaction

[15��]. The prevalence of hot segments in druggable PPIs

highlights peptides and their derivatives as a suitable class

of PPI inhibitors. We discuss various techniques for further

optimization that can propel these starting points into

potential drugs.

Characteristics of successfully inhibited PPIsFollowing the accumulation of examples of small mol-

ecules that inhibit PPIs, databases such as TIMBAL [16],

2P2I [17] and iPPI-DB [18] have been dedicated to the

collection of small molecule PPI inhibitors and relevant

structural data on these PPIs. The 2P2I database

organizes inhibited PPIs in two classes: peptide–domain

interactions and domain–domain interactions [19�] (see

Figure 1 and Table 1). A structural comparison of these

complexes to other heterodimeric complexes suggested

that the druggable 2P2I interfaces involve a much smaller

buried surface area, are more hydrophobic, and do not

show major conformational changes upon binding. Inter-

estingly, we previously found that these features, in

www.sciencedirect.com

Page 2: Druggable protein–protein interactions – from hot spots to hot segments

Hot spot segments define druggable protein interactions London, Raveh and Schueler-Furman 953

Figure 1

Protein-Protein Interactions

Domain-Domain Domain-Peptide

Estimated15-40%

Estimated>50%

No DominantHot Segment

DominantHot Segment

Amenable to inhibitionby:

&Small

molecules

Peptides

Current Opinion in Chemical Biology

Protein–protein interactions that are peptide-mediated or contain an

interface hot-segment are suitable targets for inhibition. A substantial

fraction of interactions (estimated up to 40% [1]) are mediated by a linear

peptide stretch (domain–peptide interactions). Among the remaining

domain–domain interactions, about half of the protein–protein

interactions with solved structures contain one contiguous interface hot

segment that contributes the dominant part of binding energy (as

estimated using the PeptiDerive protocol [15��], see text).

particular the latter, are also characteristic of peptide–protein interactions [9�]. Another structural analysis of

inhibited PPIs divided the interactions into four classes,

based on whether they are narrow/wide and tight/loose

[14��]. Among these, the narrow (surface area <2500 A2)

and tight (Kd < 200 nM) PPIs are more amenable to

inhibition. In turn, the inhibition of narrow and loose

interactions often involves conformational changes and

allosteric effects. Since these are more difficult to model

and design, they are usually identified in a posterioristructural analyses of inhibitors detected by high through-

put screening (HTS) approaches [14��].

A large share of PPIs are dominated by a hotsegmentAs evident from the large proportion of peptide-

mediated interactions that are targeted by small mol-

ecule inhibitors (see Table 1), this prevalent form of

interaction seems particularly suitable for inhibition.

Peptide-mediated interactions are obviously dominated

by a continuous binding epitope (namely, the peptide).

More surprisingly however, this holds also for domain–domain interactions (e.g. interactions between two glob-

ular protein partners): these too are often dominated by

one hot segment.

www.sciencedirect.com

We have previously developed a protocol that, based on a

solved complex structure, identifies the contiguous pep-

tide epitope within a protein that contributes most to

binding (Rosetta PeptiDerive [15��]). In a systematic

analysis of standard benchmarks of domain–domain inter-

actions using PeptiDerive, we estimated that more than

50% of those interactions are mediated by one hot seg-

ment that contributes the majority of the binding inter-

action energy [15��] (Figure 1). Application of the same

methodology to the entire Protein Data Bank (PDB; [20])

has led Teyra et al. [21�] to estimate that almost 700

protein families are dominated by a hot segment at the

interface of the protein–protein interaction. In a similar

study that was restricted to a-helix mediated interactions,

Jochim and Arora [22�] highlighted a set of �400 com-

plexes that contain a dominant a-helix at the interface.

Hot segments are good predictors of PPIdruggabilityTo examine the relation between druggability and the

existence of hot segments, we apply here the aforemen-

tioned PeptiDerive protocol to the druggable protein–protein complexes enlisted in the 2P2I database. The

results suggest that indeed, one contiguous peptide dom-

inates the interaction in practically all of these reported

cases (Table 1). Trivially, for all 7 peptide-protein inter-

actions, one short dominant epitope contributes 64–99%

of the estimated total interaction energy. Less trivially so,

however, PPIs of the second domain–domain class are

also dominated by a single short continuous epitope that

contributes 43–71% of the interaction energy. Remark-

ably, in all cases, we found that this epitope binds to the

same site that is targeted by a known small molecule

inhibitor (Figure 2). We conclude that it is the presence of

a dominant hot segment at a protein–protein interface

that often renders this PPI druggable.

Additional computational approaches for theprediction of PPI druggabilityAccumulating knowledge about druggable PPIs has

spurred the development of computational approaches

to predict the ‘druggability’ of a query PPI. The most

straightforward approach is to detect hot spot residues,

either by experiment [5] or by computation [23]. This

usually requires a solved structure of the PPI, or a very

accurate model. Such models may be built using protein

docking protocols of PPIs [13] or peptide-docking proto-

cols [24].

The structure of a protein complex is not always available,

but luckily it is also not always necessary in order to

determine the druggability of an interaction. Using

‘pocket biased’ conformational sampling of unbound

proteins on a subset of 2P2I, Johnson and Karanicolas

find that druggable protein interaction sites display low-

energy pocket-containing conformations that are not

found elsewhere on the protein surface [25��].

Current Opinion in Chemical Biology 2013, 17:952–959

Page 3: Druggable protein–protein interactions – from hot spots to hot segments

954 Synthetic biomolecules

Table 1

Hot segments in the 2P2I database of druggable interactions. The interactions are divided into domain-peptide and domain–domain

interaction, and sorted according to % of binding energy contributed by the hot segment

Class Partner 1 Partner 2 Hot segmenta Energy (REU)b % of total

binding energy

PDB

Domain–peptide

Menin MLL 4–13 �28.7 99 4gq6

Xdm2 p53 17–26 �16.6 95 1ycq

ZipA FtsZ 3–12 �18.8 95 1f47

Mdm4 p53 19–28 �17.4 91 3dab

Mdm2 p53 19–28 �17.5 86 1ycr

Bcl-xL Bak 573–582 �14.6 79 1bxl

Bcl-2 Bax 62–71 �22.0 64 2xa0

Domain–domain

IN LEDGF 360–369 �15.4 71 2b4j

XIAP Smac 1–10 �15.8 57 1g73

TNFR1Ac TNFb 44–53 �8.0 54 1tnr

IL2 IL2Ra 34–43 �13.9 50 1z92

TNFa (B)d TNFa (A) 91–100 �8.6 48 1tnf

HPV E2 HPV E1 452–461 �9.5 45 1tue

XIAP-Bir3 Casp9 316–325 �16.7 45 1nw9

TNFa (A)d TNFa (B) 113–122 �7.8 43 1tnf

a A continuous stretch of 10 amino acids derived from Partner 2 that contributes a dominant part of the estimated binding energy. Partner 1 is the

receptor against which a small molecule was designed. For cases where Partner 1 contains a corresponding continuous stretch that contributes even

more to binding affinity, the PDB code is underlined. In these cases a small molecule (or peptide) might be designed to bind to Partner 2.b As estimated by the PeptiDerive protocol (see [15��]; REU: Rosetta Energy Units).c We should note that in this case, the small molecule inhibitor discovered against the interaction is a covalent modifier, and while it does show

overlap with the predicted dominant peptide, the reversible binding of the molecule is quite weak (40–100 mM).d TNFa forms a symmetric homotrimer; due to small structural differences peptides derived from different monomers have slightly different predicted

binding energies. In this case the most dominant peptide derived from chain A does not overlap with the PPI inhibitor, while the dominant peptide

derived from chain B does.

Computational solvent mapping of apo structures of 15

PPI target proteins [26��] showed that the druggable sites

combine subsites with properties such as concavity and a

mixture of hydrophobicity and polarity that confer a

tendency to bind organic compounds, unique from other

surface regions.

Residues that participate in protein binding in one

protein and in small-molecule (or peptide) binding in

its homologue could be expected to be part of drug-

gable sites. Two studies analyzed such ‘bi-functional’

[27] or ‘multibinding site’ residues [28]. Surprisingly,

while the former found these residues to be less con-

served, the latter found them to be more conserved than

other (mono-functional) interface residues. Nonethe-

less, both approaches seem to indicate that these sites

are prevalent (more than 8000 proteins were annotated

with ‘bi-functional’ residues in the human proteome

[29]).

Finally, a database of interface residue clusters that

provide Small-Molecule Inhibitor Starting Points

(SMISPs) – not necessarily continuous – was used in

an orthogonal machine learning based approach [30] to

train a classifier to identify druggable sites on protein

structures. Application of this approach also estimated

that 48% of the non-redundant PPIs in the PDB would be

amenable for small-molecule inhibition.

Current Opinion in Chemical Biology 2013, 17:952–959

Peptide based inhibitors of PPIsTraditionally, the pharmaceutical industry has focused on

the discovery of small molecules to inhibit PPIs. How-

ever, chemical analyses of successful small molecule PPI

inhibitors suggest that these do not comply with classic

drug-like properties: they are larger, more hydrophobic,

mediate less hydrogen bonds, and have lower ligand

efficiency values [16,31��,32]. Whereas small molecules

seem more appropriate for targeting isolated hot spot

binding sites, peptides may very well be the natural

choice for competing for binding with hot segments.

Peptides may also be readily subjected to large-scale

proteomic methods that are not appropriate for small

molecules, and thanks to their size and increased infor-

mation content, they may be expected to bind with higher

specificity.

Discovery and design approaches

Perhaps the most naıve, yet effective method to discover

peptidic PPI inhibitors is their derivation from the inter-

face of their native PPIs. The use of peptides derived

from proteins to modulate interactions has been exten-

sively used to, for example, investigate G protein sig-

naling [12��], apoptosis [33], cancer related targets such as

survivin [34], angiogenesis [35], host–pathogen inter-

actions [36–38] and countless other systems. It is worth

noting that the discovery of these derived peptides has

mostly been based on the primary sequences of the

www.sciencedirect.com

Page 4: Druggable protein–protein interactions – from hot spots to hot segments

Hot spot segments define druggable protein interactions London, Raveh and Schueler-Furman 955

Figure 2

(a) (b) (c)

(d) (e) (f)

Current Opinion in Chemical Biology

Small molecule PPI inhibitors overlap in space with a continuous dominant peptide at the interaction interface. Examples of dominant peptides

detected at the interfaces of protein complex structures are compared to the structures of bound small molecule PPI inhibitors. The protein target for

the inhibitor (Partner 1 in Table 1) is given in surface representation. The interacting protein (Partner 2) is depicted as cartoon. The dominant peptide

detected at the interface by PeptiDerive [15��] is colored red. The small molecule PPI inhibitor is colored purple. The PDB IDs for the structures of the

PPIs are reported in Table 1. Here we include the PDB IDs of the small molecule complexes in parentheses. (A) The complex between Bcl-xL and a

peptide from Bak (1ysi) represents the class of peptide-protein interactions in which trivially the dominant peptide that is derived by the crystal

structure corresponds to the inhibitor binding site. (B–F) Non-obvious multiple epitope interfaces: here too the dominant peptide overlaps with the

small molecule inhibitor: (B) XIAP-Bir3 interaction with Casp9 (1tfq); (C) Human papilloma virus (HPV) proteins E2–E1 interaction (1r6n); (D) LEDGF-

Integrase interaction (3lpt); (E) IL2/IL2Ra interaction (1m48), and (F) TNFa trimer disrupter (2az5).

targets and only rarely on structural models (e.g. by using

peptide arrays to test all overlapping peptides in a protein

sequence for binding [39], or by identification of com-

mon/unique peptide sequences among a set of proteins to

inhibit a general/specific interaction [40,41]).

Display methods are a good source for the discovery of

usually completely novel binding sequences. Phage dis-

play, in particular, has been the source of numerous

peptide PPI inhibitors [42]. Lastly, computational design

of peptide binders is increasingly showing promise as a

source of new inhibitory peptide sequences [43�,44,45,

46�,47,48].

Stabilization and optimization

Short and isolated peptides may be conformationally

unstable, susceptible to proteolysis, and deficient in their

bioavailability [49]. Consequently, a swath of stabiliz-

ation, optimization and mimicry strategies has been

developed for rigidifying and improving the bioactivity

of peptide molecules.

Secondary structure stabilization

a-Helices play a major role in PPIs. An analysis of PPIs in

the PDB found that 62% of protein complexes feature an

a-helix at their interface [22�]. 20% of dominant peptides

at PPI interfaces also adopt a helical conformation [15��].For these reasons, stabilization of a-helices holds great

www.sciencedirect.com

promise for peptide based PPI inhibition. Two of the

main approaches for helix stabilization are helix ‘staples’

(Figure 3A; recently reviewed by Verdine and Hilinski

[50]), and Hydrogen Bond Surrogate (HBS) helices

(Figure 3B; comprehensively reviewed in Patgiri et al.[51]). b-Hairpins are also common interface motifs

(Figure 3C), and as such, b-hairpin mimetics can be

useful for inhibition of PPIs [52,53].

For coiled epitopes that do not adopt a well-defined

secondary structure, peptide cyclization methods can be

used to rigidify the peptide conformation and protect it

against proteolysis (Figures 3D and E). Cyclization can

be mediated by head-to-tail covalent linkage of the

peptide backbone or terminal residues [54,55], disulfide

bridging by pairs of cysteine residues, or other side-chain

tethering techniques (e.g. [56]). Like linear peptides,

libraries of cyclic peptides can be screened for activity

using standard proteomic screens such as reverse yeast

two-hybrid and phage display assays (e.g. [55,57–59]),

and combined with, for example, antibody competition

assays [59,60].

Lastly, peptide based molecules such as peptoids [61,62]

(Figure 3F) and foldamers [63] represent a more chem-

istry-centered direction. These take advantage of pepti-

dic properties such as specificity and compatibility with

protein binding, while at the same time they address

Current Opinion in Chemical Biology 2013, 17:952–959

Page 5: Druggable protein–protein interactions – from hot spots to hot segments

956 Synthetic biomolecules

Figure 3

(a)

(d) (e) (f)

(b) (c)

Current Opinion in Chemical Biology

Peptide stabilization approaches. Examples of various peptide stabilization technologies. For clarity, only the peptide backbones and non-peptidic

moieties are displayed. PDB IDs are given in parentheses. (A) An i � i+4 hydrocarbon stapled helix designed to bind Estrogen Receptor b (2yjd) [72].

(B) Hydrogen Bond Surrogate (HBS) seeding a 10 amino-acid helix [73]. (C) A b-hairpin peptide solved in complex with CXCR4 (3odu) [74]. (D) A

backbone cyclic, disulfide constrained, protease inhibitor peptide (1sfi) [75] (E) A bi-cyclic human urokinase-type plasminogen activator, discovered by

screening a combinatorial library (3qn7) [76]. (F) A five-mer peptoid [77]. Note that the non-canonical amino-acid side-chains, (R)-N-1-cyclohexylethyl,

are connected to the backbone nitrogen rather than the Ca atom.

typical peptide-related challenges and improve, for

example, bio-stability and permeability.

Summary and conclusionsTargeting PPIs with small molecules has come a long

way. Computational discovery methods [64] together

with major advances in experimental techniques, such

as fragment based drug design [65], allow efficient dis-

covery of hits against this challenging class of targets, and

have led to an ever increasing number of PPI inhibitors

that are making their way to the clinic [66]. These

successes, in combination with the accumulation of struc-

tural data about these interactions, make it possible to

elucidate basic features of protein interactions that can be

used to guide and improve target selection and inhibitor

design.

In this review, we have focused on hot segments –continuous peptide stretches that dominate the PPI –as starting points for PPI inhibition. These hot segments

seem to be a general feature of druggable interfaces

(Table 1), as supported by our observation that a range

of small-molecule PPI inhibitors, discovered by a variety

of methods, overlap with these suggested hot segments

in the binding site (Figure 2). That said, we found that

such a predominant peptide is present in just over half of

the analyzed complex structures [15��] (Figure 1), but

Current Opinion in Chemical Biology 2013, 17:952–959

missing from the rest. This remaining portion of pre-

dominantly non-continuous interfaces might prove more

challenging for inhibition. The same may hold for ‘tight

& wide’ interactions (according to the classification of

Smith and Gestwicki [14��]). Targeting these more

challenging interfaces might require development and

improvement of alternative approaches, for example the

combination of several non-continuous epitopes (e.g.

[67]), or approaches not directly targeting the PPI inter-

face, such as allosteric modulators [68] or peptides that

shift the oligomeric state of the target [69,70]. Another

challenge is encountered by competitive peptide based

inhibition in the context of signal transduction, where

the peptide that competes with the original partner

might as well bind and induce the same functional

response as the full protein (e.g. [12��]).

The use of peptides to modulate PPIs goes way back in

time. In nature, living organisms rely on naturally occur-

ring peptides to inhibit or compete with and regulate

interactions. As an example, the Extracellular Death

Factor (EDF) peptide prevents the binding of the MazE

antitoxin to the toxin MazF, thereby activating this

enzyme and leading to cell death. Our modeling studies

indicate that this is achieved by EDF binding to the same

site as the toxin [71]. Similarly, BH3 domain peptides

[33] in anti-apoptotic proteins compete with internal

www.sciencedirect.com

Page 6: Druggable protein–protein interactions – from hot spots to hot segments

Hot spot segments define druggable protein interactions London, Raveh and Schueler-Furman 957

interactions of a helix with the same binding groove in

apoptotic proteins such as Bcl-xL, thereby preventing

apoptosis [33]. Finally, peptide motifs in regulatory

regions of a range of kinases mediate interactions both

between and within proteins, by for example occluding

the active site, and serving as a scaffold to bind target

proteins [40,41].

Successful inhibition of protein–protein interactions not

only holds great promise, but also great challenges. PPIs

that are dominated by a hot segment might well represent

the lowest and ripest of these high-hanging fruits, and

peptide-based inhibitors might be one ladder on the way

to pick them.

AcknowledgementsThis work was funded by the Israel Science Foundation, founded by theIsrael Academy of Science and Humanities (grant number 319/11) and theUSA-Israel Binational Science Foundation (grant number 2009418) toOSF. NL was supported by an EMBO long-term fellowship (ALTF 1121-2011).

References and recommended readingPapers of particular interest, published within the period of review,have been highlighted as:

� of special interest�� of outstanding interest

1. Petsalaki E, Russell RB: Peptide-mediated interactions inbiological systems: new discoveries and applications. CurrOpin Biotechnol 2008, 19:344-350.

2. Tompa P, Fuxreiter M, Oldfield CJ, Simon I, Dunker AK,Uversky VN: Close encounters of the third kind: disordereddomains and the interactions of proteins. Bioessays 2009,31:328-335.

3. Jones S, Thornton JM: Principles of protein–proteininteractions. Proc Natl Acad Sci U S A 1996, 93:13-20.

4. Hopkins AL, Groom CR: The druggable genome. Nat Rev DrugDiscov 2002, 1:727-730.

5. Clackson T, Wells JA: A hot spot of binding energy in ahormone–receptor interface. Science 1995, 267:383-386.

6. Bogan AA, Thorn KS: Anatomy of hot spots in proteininterfaces. J Mol Biol 1998, 280:1-9.

7. DeLano WL: Unraveling hot spots in binding interfaces:progress and challenges. Curr Opin Struct Biol 2002, 12:14-20.

8. Reichmann D, Rahat O, Cohen M, Neuvirth H, Schreiber G: Themolecular architecture of protein–protein binding sites. CurrOpin Struct Biol 2007, 17:67-76.

9.�

London N, Movshovitz-Attias D, Schueler-Furman O: Thestructural basis of peptide-protein binding strategies.Structure 2010, 18:188-199.

This study compared structural features of protein–protein interactionswith peptide-mediated interactions. The main relevant finding to thisreview is that hotspots are prevalent also in peptide–protein interactionsand can be used as starting point for inhibitor design.

10. Fletcher S, Hamilton AD: Protein surface recognition andproteomimetics: mimics of protein surface structure andfunction. Curr Opin Chem Biol 2005, 9:632-638.

11. Arkin MR, Whitty A: The road less traveled: modulating signaltransduction enzymes by inhibiting their protein–proteininteractions. Curr Opin Chem Biol 2009, 13:284-290.

12.��

Shpakov AO: Signal protein-derived peptides as functionalprobes and regulators of intracellular signaling. J Amino Acids2011:656051.

www.sciencedirect.com

Very detailed and comprehensive review on the use of peptides tounderstand biological interactions using intervening peptides and smallmolecules derivatives.

13. Bienstock RJ: Computational drug design targeting protein–protein interactions. Curr Pharm Des 2012, 18:1240-1254.

14.��

Smith MC, Gestwicki JE: Features of protein–proteininteractions that translate into potent inhibitors: topology,surface area and affinity. Expert Rev Mol Med 2012, 14:e16.

Thorough and insightful analysis of known small molecule PPI inhibitorsand the features of the inhibited interfaces.

15.��

London N, Raveh B, Movshovitz-Attias D, Schueler-Furman O:Can self-inhibitory peptides be derived from the interfaces ofglobular protein–protein interactions? Proteins 2010,78:3140-3149.

This study describes the development and application of PeptiDerive:Starting from a solved protein complex structure, this protocol identifies‘‘hot segments’’, that is, the continuous peptide stretch within a protein thatcontributes most to binding (computed, estimated DDG). Analysis of thesepeptides indicates that for over 50% of the protein interactions in 2 bench-marks of protein complex structures, one such peptide contributes most ofthe binding energy. See also references [21�,22�] that have applied this or asimilar protocol to a larger set of protein complex structures. Current large-scale analyses of the structural and functional features of these peptides areunderway, and a dedicated Webserver is currently being set up.

16. Higueruelo AP, Schreyer A, Bickerton GR, Pitt WR, Groom CR,Blundell TL: Atomic interactions and profile of small moleculesdisrupting protein–protein interfaces: the TIMBAL database.Chem Biol Drug Des 2009, 74:457-467.

17. Basse MJ, Betzi S, Bourgeas R, Bouzidi S, Chetrit B, Hamon V,Morelli X, Roche P: 2P2Idb: a structural database dedicated toorthosteric modulation of protein–protein interactions. NucleicAcids Res 2013, 41:D824-D827.

18. Labbe CM, Laconde G, Kuenemann MA, Villoutreix BO,Sperandio O: iPPI-DB: a manually curated and interactivedatabase of small non-peptide inhibitors of protein–proteininteractions. Drug Discov Today 2013.

19.�

Bourgeas R, Basse MJ, Morelli X, Roche P: Atomic analysis ofprotein–protein interfaces with known inhibitors: the 2P2Idatabase. PLoS ONE 2010, 5:e9598.

Databases of structures of inhibited interactions provides a good startingpoint to identify critical features for successful inhibition (see references[16–18]). This study analyses one of these databases: 2P2I. In particular, itdivides the set into peptide-mediated and protein domain–domain inter-actions. See also reference [31��].

20. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H,Shindyalov IN, Bourne PE: The Protein Data Bank. Nucleic AcidsRes 2000, 28:235-242.

21.�

Teyra J, Sidhu SS, Kim PM: Elucidation of the bindingpreferences of peptide recognition modules: SH3 and PDZdomains. FEBS Lett 2012, 586:2631-2637.

Large scale analysis of hot segments. See reference [15��].

22.�

Jochim AL, Arora PS: Systematic analysis of helical proteininterfaces reveals targets for synthetic inhibitors. ACS ChemBiol 2010, 5:919-923.

Large scale analysis of hot segments. See reference [15��].

23. Potapov V, Cohen M, Schreiber G: Assessing computationalmethods for predicting protein stability upon mutation: goodon average but not in the details. Protein Eng Des Sel 2009,22:553-560.

24. London N, Raveh B, Schueler-Furman O: Peptide docking andstructure-based characterization of peptide binding: fromknowledge to know-how. Curr Opin Struct Biol 2013 http://dx.doi.org/10.1016/j.sbi.2013.07.006.

25.��

Johnson DK, Karanicolas J: Druggable protein interaction sitesare more predisposed to surface pocket formation than therest of the protein surface. PLoS Comput Biol 2013, 9:e1002951.

References [25��,26��] describe approaches to identify druggable sites onthe free protein structures. This reference presents a very nice computa-tional approach that is based on the reduced stability and increasedstructural dynamics of these regions. This dynamic fluctuation allows toidentify the open binding site conformation. See reference [26��] foradditional approach.

Current Opinion in Chemical Biology 2013, 17:952–959

Page 7: Druggable protein–protein interactions – from hot spots to hot segments

958 Synthetic biomolecules

26.��

Kozakov D, Hall DR, Chuang GY, Cencic R, Brenke R, Grove LE,Beglov D, Pelletier J, Whitty A, Vajda S: Structural conservationof druggable hot spots in protein–protein interfaces. Proc NatlAcad Sci U S A 2011, 108:13528-13533.

References [25��,26��] describe approaches to identify druggable sites onthe free protein structures. This reference presents another complemen-tary approach based on computational solvent mapping: druggable sitesare identified where solvent molecules cluster. As reference [25��], thisstudy is also focussed on application to the free, apo structure.

27. Davis FP, Sali A: The overlap of small molecule and proteinbinding sites within families of protein structures. PLoSComput Biol 2010, 6:e1000668.

28. Thangudu RR, Bryant SH, Panchenko AR, Madej T: Modulatingprotein–protein interactions with small molecules: theimportance of binding hotspots. J Mol Biol 2012, 415:443-453.

29. Davis FP: Proteome-wide prediction of overlapping smallmolecule and protein binding sites using structure. Mol Biosyst2011, 7:545-557.

30. Koes DR, Camacho CJ: Small-molecule inhibitor starting pointslearned from protein–protein interaction inhibitor structure.Bioinformatics 2012, 28:784-791.

31.��

Morelli X, Bourgeas R, Roche P: Chemical and structurallessons from recent successes in protein–protein interactioninhibition (2P2I). Curr Opin Chem Biol 2011, 15:475-481.

Comprehensive review on the inhibition of PPIs. As in reference [19�], theinteractions are classified according to their size and affinity (small/largeand tight/loose), and successful examples are described along failuresthat highlight the current challenges in the field.

32. Wells JA, McClendon CL: Reaching for high-hanging fruit indrug discovery at protein–protein interfaces. Nature 2007,450:1001-1009.

33. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S,Korsmeyer SJ: Distinct BH3 domains either sensitize oractivate mitochondrial apoptosis, serving as prototype cancertherapeutics. Cancer Cell 2002, 2:183-192.

34. Plescia J, Salz W, Xia F, Pennati M, Zaffaroni N, Daidone MG,Meli M, Dohi T, Fortugno P, Nefedova Y et al.: Rational design ofshepherdin, a novel anticancer agent. Cancer Cell 2005, 7:457-468.

35. Rosca EV, Koskimaki JE, Rivera CG, Pandey NB, Tamiz AP,Popel AS: Anti-angiogenic peptides for cancer therapeutics.Curr Pharm Biotechnol 2011, 12:1101-1116.

36. Galdiero S, Falanga A, Tarallo R, Russo L, Galdiero E, Cantisani M,Morelli G, Galdiero M: Peptide inhibitors against herpes simplexvirus infections. J Pept Sci 2013, 19:148-158.

37. Maes M, Loyter A, Friedler A: Peptides that inhibit HIV-1integrase by blocking its protein–protein interactions. FEBS J2012, 279:2795-2809.

38. Castel G, Chteoui M, Heyd B, Tordo N: Phage display ofcombinatorial peptide libraries: application to antiviralresearch. Molecules 2011, 16:3499-3518.

39. Katz C, Levy-Beladev L, Rotem-Bamberger S, Rito T, Rudiger SG,Friedler A: Studying protein–protein interactions using peptidearrays. Chem Soc Rev 2011, 40:2131-2145.

40. Mochly-Rosen D, Khaner H, Lopez J, Smith BL: Intracellularreceptors for activated protein kinase C. Identificationof a binding site for the enzyme. J Biol Chem 1991,266:14866-14868.

41. Mochly-Rosen D, Das K, Grimes KV: Protein kinase C, an elusivetherapeutic target? Nat Rev Drug Discov 2012, 11:937-957.

42. Sidhu SS, Fairbrother WJ, Deshayes K: Exploring protein–protein interactions with phage display. Chembiochem 2003,4:14-25.

43.�

Sammond DW, Bosch DE, Butterfoss GL, Purbeck C, Machius M,Siderovski DP, Kuhlman B: Computational design of thesequence and structure of a protein-binding peptide. J AmChem Soc 2011, 133:4190-4192.

One of a few examples of the successful design of a peptide binders tothe target.

Current Opinion in Chemical Biology 2013, 17:952–959

44. Lee J, Sammond DW, Fiorini Z, Saludes JP, Resch MG, Hao B,Wang W, Yin H, Liu X: Computationally designed peptideinhibitors of the ubiquitin E3 Ligase SCF(Fbx4). Chembiochem2013.

45. Rubinstein M, Niv MY: Peptidic modulators of protein–proteininteractions: progress and challenges in computationaldesign. Biopolymers 2009, 91:505-513.

46.�

Vanhee P, van der Sloot AM, Verschueren E, Serrano L,Rousseau F, Schymkowitz J: Computational design of peptideligands. Trends Biotechnol 2011, 29:231-239.

This is a comprehensive review on the computational design of peptideligands, their derivatives, and experimental high throughput techniques.

47. Chandra D, Morrison CJ, Woo J, Cramer S, Karande P: Design ofpeptide affinity ligands for S-protein: a comparison ofcombinatorial and de novo design strategies. Mol Divers 2013,17:357-369.

48. Donsky E, Wolfson HJ: PepCrawler: a fast RRT-based algorithmfor high-resolution refinement and binding affinity estimationof peptide inhibitors. Bioinformatics 2011, 27:2836-2842.

49. Adessi C, Soto C: Converting a peptide into a drug: strategiesto improve stability and bioavailability. Curr Med Chem 2002,9:963-978.

50. Verdine GL, Hilinski GJ: Stapled peptides for intracellular drugtargets. Methods Enzymol 2012, 503:3-33.

51. Patgiri A, Jochim AL, Arora PS: A hydrogen bond surrogateapproach for stabilization of short peptide sequences inalpha-helical conformation. Acc Chem Res 2008, 41:1289-1300.

52. Obrecht D, Chevalier E, Moehle K, Robinson JA: b-Hairpinprotein epitope mimetic technology in drug discovery. DrugDisc Today: Technol 2012, 9:e63-e69.

53. Robinson JA: Beta-hairpin peptidomimetics: design,structures and biological activities. Acc Chem Res 2008,41:1278-1288.

54. Conlan BF, Gillon AD, Craik DJ, Anderson MA: Circular proteinsand mechanisms of cyclization. Biopolymers 2010, 94:573-583.

55. Tavassoli A, Benkovic SJ: Split-intein mediated circular ligationused in the synthesis of cyclic peptide libraries in E. coli. NatProtoc 2007, 2:1126-1133.

56. Angelini A, Morales-Sanfrutos J, Diderich P, Chen S,Heinis C: Bicyclization and tethering to albuminyields long-acting peptide antagonists. J Med Chem 2012,55:10187-10197.

57. Horswill AR, Benkovic SJ: Identifying small-moleculemodulators of protein–protein interactions. Curr Protoc ProteinSci 2006. Chapter 19:Unit 19 15.

58. Joo SH, Xiao Q, Ling Y, Gopishetty B, Pei D: High-throughputsequence determination of cyclic peptide library members bypartial Edman degradation/mass spectrometry. J Am ChemSoc 2006, 128:13000-13009.

59. Hall PR, Hjelle B, Njus H, Ye C, Bondu-Hawkins V, Brown DC,Kilpatrick KA, Larson RS: Phage display selection of cyclicpeptides that inhibit Andes virus infection. J Virol 2009,83:8965-8969.

60. Ramanujam P, Tan WS, Nathan S, Yusoff K: Novel peptides thatinhibit the propagation of Newcastle disease virus. Arch Virol2002, 147:981-993.

61. Dohm MT, Kapoor R, Barron AE: Peptoids: bio-inspiredpolymers as potential pharmaceuticals. Curr Pharm Des 2011,17:2732-2747.

62. Butterfoss GL, Yoo B, Jaworski JN, Chorny I, Dill KA,Zuckermann RN, Bonneau R, Kirshenbaum K, Voelz VA: De novostructure prediction and experimental characterization offolded peptoid oligomers. Proc Natl Acad Sci U S A 2012,109:14320-14325.

63. Goodman CM, Choi S, Shandler S, DeGrado WF: Foldamers asversatile frameworks for the design and evolution of function.Nat Chem Biol 2007, 3:252-262.

www.sciencedirect.com

Page 8: Druggable protein–protein interactions – from hot spots to hot segments

Hot spot segments define druggable protein interactions London, Raveh and Schueler-Furman 959

64. Kruger DM, Jessen G, Gohlke H: How good are state-of-the-artdocking tools in predicting ligand binding modes in protein–protein interfaces? J Chem Inf Model 2012, 52:2807-2811.

65. Valkov E, Sharpe T, Marsh M, Greive S, Hyvonen M: Targetingprotein–protein interactions and fragment-based drugdiscovery. Top Curr Chem 2012, 317:145-179.

66. Mullard A: Protein–protein interaction inhibitors get into thegroove. Nat Rev Drug Discov 2012, 11:173-175.

67. Eichler J: Peptides as protein binding site mimetics. Curr OpinChem Biol 2008, 12:707-713.

68. Nussinov R, Tsai CJ: Allostery in disease and in drug discovery.Cell 2013, 153:293-305.

69. Hayouka Z, Rosenbluh J, Levin A, Loya S, Lebendiker M,Veprintsev D, Kotler M, Hizi A, Loyter A, Friedler A: Inhibiting HIV-1 integrase by shifting its oligomerization equilibrium. ProcNatl Acad Sci U S A 2007, 104:8316-8321.

70. Gabizon R, Brandt T, Sukenik S, Lahav N, Lebendiker M,Shalev DE, Veprintsev D, Friedler A: Specific recognition of p53tetramers by peptides derived from p53 interacting proteins.PLoS ONE 2012, 7:e38060.

71. Belitsky M, Avshalom H, Erental A, Yelin I, Kumar S, London N,Sperber M, Schueler-Furman O, Engelberg-Kulka H: TheEscherichia coli extracellular death factor EDF induces theendoribonucleolytic activities of the toxins MazF and ChpBK.Mol Cell 2011, 41:625-635.

www.sciencedirect.com

72. Phillips C, Roberts LR, Schade M, Bazin R, Bent A, Davies NL,Moore R, Pannifer AD, Pickford AR, Prior SH et al.: Design andstructure of stapled peptides binding to estrogen receptors. JAm Chem Soc 2011, 133:9696-9699.

73. Liu J, Wang D, Zheng Q, Lu M, Arora PS: Atomicstructure of a short alpha-helix stabilized by a mainchain hydrogen-bond surrogate. J Am Chem Soc 2008,130:4334-4337.

74. Wu B, Chien EY, Mol CD, Fenalti G, Liu W, Katritch V,Abagyan R, Brooun A, Wells P, Bi FC et al.: Structuresof the CXCR4 chemokine GPCR with small-moleculeand cyclic peptide antagonists. Science 2010, 330:1066-1071.

75. Luckett S, Garcia RS, Barker JJ, Konarev AV, Shewry PR,Clarke AR, Brady RL: High-resolution structure of a potent,cyclic proteinase inhibitor from sunflower seeds. J Mol Biol1999, 290:525-533.

76. Angelini A, Cendron L, Chen S, Touati J, Winter G, Zanotti G,Heinis C: Bicyclic peptide inhibitor reveals large contactinterface with a protease target. ACS Chem Biol 2012, 7:817-821.

77. Wu CW, Kirshenbaum K, Sanborn TJ, Patch JA, Huang K,Dill KA, Zuckermann RN, Barron AE: Structural andspectroscopic studies of peptoid oligomers withalpha-chiral aliphatic side chains. J Am Chem Soc 2003,125:13525-13530.

Current Opinion in Chemical Biology 2013, 17:952–959


Recommended