Abstract
A powerful early approach to evaluating the druggability of proteins involved determining the hit rate in NMR-based screening of a library of small compounds. Here we show that a computational analog of this method, based on mapping proteins using small molecules as probes, can reliably reproduce druggability results from NMR-based screening, and can provide a more meaningful assessment in cases where the two approaches disagree. We apply the method to a large set of proteins. The results show that, because the method is based on the biophysics of binding rather than on empirical parameterization, meaningful information can be gained about classes of proteins and classes of compounds beyond those resembling validated targets and conventionally druglike ligands. In particular, the method identifies targets that, while not druggable by druglike compounds, may become druggable using compound classes such as macrocycles or other large molecules beyond the rule-of-five limit.
Keywords: drug discovery, ligand design, binding hot spot, drug target protein, macrocyclic compounds, inhibitors of protein-protein interaction
INTRODUCTION
Among the distinct proteins encoded by the human proteome, it had been estimated that only about 3,000 belong to structural families for which small molecule (i.e. synthetic organic) drugs have been developed.1 Many biologically compelling drug targets belong to other protein families that lack such empirical proof of principle that they can be inhibited by small molecule drugs. Consequently, approaches to assessing the druggability of potential drug targets – i.e. the likelihood of being able to identify a druglike small ligand that can modulate the activity of the target – have emerged as an important tool for target-based drug discovery. We note that throughout this paper, we call a small compound “druglike” if it satisfies Lipinski's rule of five (no more than 5 hydrogen bond donors, no more than 10 hydrogen bond acceptors, a molecular mass less than 500 g/mol, and an octanol-water partition coefficient not greater than 5).2 There are two major approaches to determining the druggability of a protein. One is based on empirical analysis of known target/drug associations,1,3,4 which can be applied only when the protein in question belongs to a well-studied family that includes close homologs for which reliable druggability information is available. The other is based solely on analysis of the structure of the target protein.5–17 For novel targets, from families for which no drugs or advanced drug candidates are known, one has to rely on the latter method of structure based predictive models. This approach was pioneered by Hajduk et al.,7 who re-examined the results of NMR-based fragment screening for a diverse set of 23 proteins, and observed a high correlation between the fragment hit rate and the ability to identify high-affinity (KD < 300 nM) druglike ligands. They used these data to derive a regression-based model to predict the likely fragment hit rate for untested targets, based on structural properties of the protein binding site such as total and apolar surface area and pocket compactness, generating for each a predicted “druggability score” defined as the logarithm of the predicted NMR hit rate. The regression formula provided a good approximation of the original hit rates and, as we will show, resulted in fairly accurate prediction of druggability. Another important contribution came from Cheng et al.,8 who used the curvature and the lipophilic surface area of pockets to predict the maximal affinity achievable by passively absorbed drugs. Despite its simplicity, the model also provided good discrimination of druggable binding sites from those labeled as difficult or undruggable, but was tested only on a limited number of proteins.
To further improve druggability prediction, a variety of methods have been published in the last six years, most of them trained on the growing number of protein targets with some level of druggability information available. 5,10–12,14,16 For method validation, Schmidtke and Barril compiled a large set of protein targets with associated degrees of druggability.11 Krasowski et al.12 added further proteins to the training set, and develop the DrugPred druggability score. All these methods are based on the analysis of various pocket properties, including hydrophobicity, size, compactness, hydrogen bond donor and acceptor surface areas, and amino acid composition in pockets. Thus, evaluating the druggability of proteins with respect to conventional druglike compounds is now a mature technology that can perform such assessments with some reliability. However, this technology has considerable limitations. Apart from the NMR based fragment screening by Hajduk et al.,7 all methods correlate pocket properties with druggability data available for traditional targets, and therefore in essence simply evaluate whether the novel target in question contains a binding pocket that in some sense resembles pockets on known druggable targets that bind conventional druglike ligands. Moreover, the methods provide a single druggability scale that attempts to capture both the expected affinity that can be achieved, as well as the druglikeness – and implicitly, the potential for oral biovalability – of ligands that will potentially bind to the site. A general shortcoming of all these methods, therefore, is that they are poorly equipped to evaluate the druggability of proteins with surface topologies that substantially differ from the relatively small number of protein families for which drugs or high affinity druglike ligands exist, such as protein-protein interaction (PPI) targets, or to assess the potential of any target to bind ligands that do not conform to conventional definitions of druglikeness. Moreover, because these methods are parameterized rather than being based on biophysical principles, and return only a monolithic measure of druggability, they provide little insight into why a particular target is or is not druggable, and therefore into the prospects that strong binding might be achievable by an appropriately chosen nontraditional drug chemotype such as a natural product, a macrocyclic compound,18–21 a covalent inhibitor,22–24 or a peptide-derived foldamer.25–28
It is additionally noteworthy that, among the above methods, only Seco et al.9 explicitly takes into account the potential for conformational flexibility of the protein to affect the druggability of a binding site. The other methods base their analysis on static X-ray crystal structures. Several of the studies did, however, test the robustness of their results to variations in protein conformation, by comparing the results obtained after computationally perturbing the structure10 or by comparing the results obtained for different crystal structures of the same target protein,5,8,11,12 and generally reported that the ability to identify druggable sites was not greatly sensitive to such conformational effects. Other work has suggested, however, that conformational adaptivity at the binding site can substantially affect druggability, in cases where the alternative conformations that are sampled through thermal motions persist for a sufficient period of time.29
In this perspective we first discuss the advantages of the fragment screening approach used by Hajduk et al.,7 and introduce an analog of the method that uses a computational algorithm rather than experimental NMR data for fragment screening. The computational approach was used to evaluate some of the “historical” druggability results published during the last decade, including the 23 proteins considered in the NMR based screening study,6,7 and the additional set of 72 proteins previously studied by the regression model, based on the NMR results and developed by the same authors.7 We also studied the protein set selected by Cheng et al.,8 and some of the more contentious results from the DrugPred algorithm of Krasowski et al.12 As will be shown, the method can reliably reproduce earlier druggability results. Moreover, evaluation of the small number of cases in which the original assessment was uncertain or was subsequently proved incorrect shows that the method presented here can in fact provide an even more meaningful assessment of druggability. We additionally show that biophysics based computational approaches (as opposed to empirical approaches that are parameterized against a particular training set of protein-ligand complexes) are now sufficiently reliable that they can be used to address the potential for highly challenging or atypical drug targets, such as protein-protein interfaces, to be addressed using non-canonical drug chemotypes such as synthetic macrocycles. These methods therefore point the way to expanding the range of drug targets that can be addressed, by using chemotypes that are known to be pharmaceutically relevant but that fall outside of conventional druglike chemical space. We also provide detailed discussion of the druggability of six targets of high pharmaceutical interest for which the prospects of success have been controversial.
Experimental fragment based analysis of druggability
As already mentioned, the structure based analysis of druggability was introduced in a landmark paper by Hajduk and co-workers,7 who re-examined the results of NMR-based fragment screening experiments, and observed a high correlation between the fragment hit rate and the ability to identify high-affinity druglike inhibitors. This early method has two major advantages over more recent approaches. First, by determining the binding locations of fragment-sized compounds, the screening based method automatically identifies the binding site. In contrast, the prediction methods described in the introduction are based on the analysis of pocket properties, primarily size, shape, complexity, and hydrophobicity, and thus have first to select the binding site residues. As noticed by Rarey and co-workers,17 pocket descriptors depend heavily on correctly selecting the region of the protein surface that is most likely to form a binding site. This is a nontrivial task for a ligand-free protein, and is also complicated by potential conformational differences between ligand-bound and ligand-free structures. Second, as Hajduk et al.7 were aware, their method actually identifies binding hot spots, i.e., regions of the binding site that contribute a disproportionate amount of the binding free energy, 30,31 and hence are primarily responsible for ligand binding, in contrast to other methods that base their predictions on global pocket properties without regard for the extent to which sites within the pocket can contribute significant binding energy.
The concept of hot spots was originally introduced in the context of mutating interface residues to alanine in protein-protein or protein-peptide interfaces30,32–34. On the basis of this method, a residue is considered a hot spot if its mutation to alanine gives rise to a substantial drop in binding affinity. The concept was extended to the binding of small molecules by Ringe and co-workers, who used X-ray crystallography to determine the structure of the target protein soaked in aqueous solutions of 6–8 organic solvents. By superimposing the structures in these so-called Multiple Solvent Crystal Structure (MSCS) experiments, regions that bind multiple different probes were detected35,36. While individual probes were able to bind at a number of locations, their clusters indicated binding hot spots. In their NMR based experiments, Hajduk et al.7 incubated the target protein with a series of small organic probes, generically referred to as “fragments”, and used the perturbations in residue chemical shifts to identify residues that participate in small molecule binding, thereby also finding the binding hot spots. While results of the MSCS method requiring the determination of X-ray structures in multiple solvents were reported only for a few proteins,36–40 Hajduk et al.7 analyzed the results of NMR-based fragment screening for 23 proteins. These results convincingly demonstrated that the small fragment “probe” ligands cluster at hot spots, and that the hit rate predicts the druggability of the site,7,35 thereby initiating the development of structure based druggability conditions.
Computational fragment screening by FTMap
Based on the above results from the Hajduk7 and Ringe35 groups, NMR and X-ray based fragment screening provides information on druggability, and is now broadly used in pharmaceutical research.41,42 However, the experiments are expensive, and may be hampered by the requirement that the target protein be expressed and purified in relatively large quantities for experimental analysis.17 Thus, it is desirable to replace these experimental methods with in silico approaches to evaluating druggability, particularly when exploring novel targets. Hajduk et al.7 accomplished this goal by constructing a regression equation to express the NMR hit rates in terms of pocket properties. In this paper we describe druggability conditions based on direct simulation of the X-ray and NMR-based screening experiments. In fact, modeling the weakly specific binding of small and rigid ligands to proteins is not very difficult, and hence computational methods offer attractive alternatives.43,44 We use computational solvent mapping, implemented as the FTMap algorithm.45 The method places small molecular probes of various sizes and shapes on a dense grid around the protein, finds favorable positions using empirical energy functions, refines the bound positions while accounting for side chain and probe flexibility, clusters the conformations, and ranks the clusters on the basis of the average energy. All ligands and crystallographic water molecules are removed prior to mapping, and the probes are initially distributed over the entire protein surface without any assumption as to the location of the binding site(s). As has been extensively validated, the regions that bind multiple low energy probe clusters, called consensus cluster (CC) sites, identify the locations of the binding energy hot spots.40,45–50 The CCs are ranked on the basis of the number of probe clusters contained, which we have shown corresponds to relative energetic importance,51 and are denoted as CC1, CC2, and so on. The CC with the highest number of probe clusters in the binding site of interest is defined as the primary hot spot, whereas other CCs with fewer probe clusters are secondary hot spots.
FTMap has been implemented as a server (http://ftmap.bu.edu/), which currently uses 16 small molecules as probes (ethanol, isopropanol, isobutanol, acetone, acetaldehyde, dimethyl ether, cyclohexane, ethane, acetonitrile, urea, methylamine, phenol, benzaldehyde, benzene, acetamide, and N,N-dimethylformamide). The server and its potential applications were recently discussed in detail.52 We explored the use of substantially larger probe libraries, but since this did not affect druggability predictions, we returned to the 16 compounds whose aqueous solutions have been used previously in mapping experiments based on NMR53 or X-ray techniques35–38,54. Since most probes include both polar and nonpolar moieties, our mapping results show that the highest number of probes cluster in pockets that have a mosaic-like pattern of hydrophobic and polar regions, enabling the binding of many different compounds in a variety of poses.48
Druggability analysis by FTMap
As will be described, we have mapped and analyzed over 150 ligand-protein complexes, as well as the corresponding ligand-free proteins. To establish objective FTMap-based criteria for evaluating the druggability of proteins, we set out to benchmark our results by using the 16 probe types used in the FTMap server. For most target proteins, we mapped both the highest resolution ligand-free and ligand-bound structures available in the Protein Data Bank (PDB).55 As explained below, generally druggability can best be determined by mapping the ligand-free structure; although analysis of ligand-bound structure provides additional information, in most cases it is used only to indicate the binding location of a known ligand. Based on the analysis of this large set of proteins we have observed that the hot spots of druggable proteins satisfy conditions on (1) strength, (2) connectivity or compactness, and (3) the maximum dimension of the hot spot region. We describe the origin of these conditions in turn.
As already mentioned, Hajduk et al. observed that the hit rate in fragment screening is a predictor of druggability.7 In agreement with this observation, we have previously established that, when using 16 probes for the mapping, the sites that are known to be druggable invariably contain a strong hot spot containing 16 or more probe clusters.48 Indeed, among the 121 drug targets considered in earlier studies by Hajduk et al.7 and Cheng et al.,8 and also examined by FTMap here, only in a single case was there a druggable target with known, potent ligands that had fewer than 16 probe clusters in the strongest hot spot at the drug binding site (Figure S1), whereas we found 21 such targets among those which experimental evidence suggests are not druggable (Figure S2). Since FTMap clusters probe positions using pairwise 4Å root mean square deviation (RMSD) as the clustering radius, the probe clusters are relatively small, and thus a consensus cluster (CC) can include several clusters of the same probe type. Thus, the requirement of 16 probe clusters does not imply that a druggable site has to bind all 16 different probes, although very frequently this is the case. FTMap analysis of the targets considered herein indicates that the requirement for a hot spot of at least 16 probe clusters is the most important condition for druggability: without it high affinity binding is essentially impossible (Figure S1).
The second condition, requiring connectivity or compactness, is based on the observation that a druggable site should also include one or more secondary hot spots that are close enough to the primary hot spot to be reached by a drug sized molecule.48 For the current work we attempted to establish a quantitative threshold for how close these additional hot spots must be to support the strong binding of a druglike ligand, so we could include this measure as part of an objective metric for site druggability. We therefore analyzed the separation between hot spots seen for a large set of druggable sites that bind druglike ligands with molecular weight not exceeding 500 g/mol, as shown in Figure S3. The results show that the mean distance between the centers of CCs in druggable sites (i.e., the points with the highest density of probe atoms within the consensus clusters) is between 5 and 6 Å. However, to avoid missing any druggable sites we designated 8 Å as the maximum allowed distance between connected hot spots, which captures over 99% of the druggable sites. In evaluating druggability, we used this criterion for hot spot proximity to characterize the ensemble of hot spots that constitute a particular binding site. Specifically, we first selected the primary hot spot in a given site (not necessarily CC1), to form the kernel of the ensemble. The ensemble was then expanded by iteratively adding all secondary hot spots within 8 Å from any hot spot already in the ensemble, until no further expansion was possible. In this process, secondary hot spots with less than three probe clusters were ignored.
The third criterion involving the size of the hot spot region is based on the observation that a binding site can be too small to be easily druggable. It is well known that very small binding sites, and the fragment-sized ligands that bind to them, tend to be promiscuous.56–60 Indeed, this fact is implicit in the observations that experimental fragment libraries containing only 1000–2000 compounds typically provide multiple hits when screened against essentially any druggable target, and that a druggable site is characterized by the observation of a high fragment hit rate indicating that each site can bind many different fragment structures.56,57 Thus, while a small site containing one or more strong CCs can be sufficient to enable strong ligand binding, for the binding to be selective it appears also necessary that the site be sufficiently large and topologically complex that a ligand that is complementary to this site is unlikely to bind as strongly to unrelated sites.42 As the simplest way to capture the extent of the binding site – or, at least, that portion of the binding site that is relevant for the generation of binding energy with the ligand – we simply report the longest dimension of the CC ensemble, which we defined as the distance between the two most widely separated probe atoms across the ensemble of probe clusters. In the following analysis we found that the maximum extent of the binding site should exceed 10 Å for a site to be considered druggable. Indeed, among the druggable targets that have known druglike ligands we found only 3 violations of this rule (Figure S4).
Based on the three properties described above, we classify target sites on proteins into four broad classes of druggability (Table 1).
Table 1.
Classification of Protein Druggability based on FTMap Results
| Druggability class | Strength (S) (Number of probe clusters) | Center-to-center distance (CD), Å | Maximum dimension (MD), Å | Druggability subclass | Notation |
|---|---|---|---|---|---|
| Druggable | S ≥ 16 | CD < 8 | MD ≥ 10 | Druggable using druglike compounds | D |
| Not druggable | S < 13 | Any CD | Any MD | Not druggable due to weak hot spots | |
| Not druggable | Any S | Any CD | MD < 7 | Not druggable due to small hot spot ensemble | N |
| Non-Canonically Druggable - Large | S ≥ 16 | CD ≥ 8 | MD ≥ 10 | Druggable only by large chemotype such as macrocycle or foldamer | D*L |
| Non-Canonically Druggable - Small | S ≥ 16 | CD < 8 | 7 ≤ MD < 10 | Druggable only by peptide, macrocycle, or charged compound | D*S |
| Borderline Druggable | 13 ≤ S < 16 | CD < 8 | MD ≥ 10 | At most micromolar affinity using druglike compounds | B |
| Borderline Non-Canonically Druggable - Large | 13 ≤ S < 16 | CD ≥ 8 | MD ≥ 10 | Micromolar affinity by large chemotype such as macrocycle or foldamer | B*L |
| Borderline Non-Canonically Druggable - Small | 13 ≤ S < 16 | CD < 8 | 7 ≤ MD < 10 | Micromolar affinity by peptide, macrocycle, or charged compound | B*S |
Druggable. A binding site is categorized as “Druggable” with respect to conventional, druglike (e.g. Rule of Five compliant)2 compounds if it meets the following criteria: (a) It includes a strong primary hot spot with at least 16 probe clusters, denoted as S ≥ 16. (b) One or more secondary hot spots are connected to the primary hot spot with a center-to-center distance (CD) < 8 Å. (c) The maximum dimension (MD) of the connected hot spot ensemble is at least 10 Å, i.e., MD ≥ 10 Å (Table 1).
Not Druggable. If the primary hot spot is very weak (S < 13), or the hot spot ensemble is very small (MD < 7 Å), a site is not druggable irrespective of the compounds used.
Non-canonically Druggable – Large. If a sufficient number of sufficiently strong hot spots are present (S ≥ 16) but the hot spot ensemble is too sparse, requiring a center-to-center distance exceeding 8 Å to reach a secondary hot spot from the primary one, then small druglike compounds cannot bind with sufficient affinity. However, the evidence indicates that such sites generally can bind strongly to larger but still pharmaceutically relevant compounds such as macrocycles61,62 or peptide/peptidomimetic foldamers.63,64 As will be discussed, it is possible that a subset of hot spots can satisfy the conditions CD < 8 Å, S ≥ 16 and MD ≥ 10 Å, but there exist additional strong hot spots beyond the ensemble. In such cases the target is druggable using druglike molecules, but there is the potential for binding affinity to be improved if the size of the compounds is increased to reach all hot spots, although doing so will likely require using compounds that are large and so lie outside of conventional druglike chemical space.
Non-canonically Druggable – Small. Another class of sites is encountered when the hot spot ensemble is small, but not very small (7 Å ≤ MD < 10 Å). Such sites may be druggable using ligands with a charged group that is not fully desolvated upon binding. Alternatively, druggability can be achieved by large and thus not druglike compounds (peptides,65 peptidomimetics,64 or macrocycles61) that extend beyond the small hot spot ensemble to interact with large surface regions without any strong hot spot. As it will be demonstrated, while such interactions are weak, they still contribute to the binding free energy.
It is important to note that druggability is not an on-off property. In particular, for the categories (1), (3), and (4), a site where the primary hot spot is somewhat weaker than the threshold (S ≤ 13 < 16), we use the term “borderline” druggable (Table 1). For such sites ligands can be found that bind with micromolar affinity, but attempts to increase potency usually fail. More generally, the strengths and arrangement of hot spots provide information beyond the simple four categories above. As an example, we discuss here the analysis of druggability for the FK506 Binding Protein (FKBP), which binds 8-deethyl-8-[but-3-enyl]-ascomycin (FK506), a macrocyclic compound with a molecular weight (MW) of 804 g/mol (Figure 1A).66 Later we will also study ABT737, a well-known regulator of apopotosis by inhibiting the interaction between the Bcl-xL protein and the BAK peptide (Figure 1B).67 Figure 1C shows the results of mapping a ligand-free structure of FKBP (PDB ID 2ppn). As mentioned, each hot spot is identified by a consensus cluster (CC). In all our figures, each probe cluster contained in a CC is represented by the single lowest energy probe structure to avoid the overcrowding of the figure. All probes in a CC have the same color, using the color code as follows: CC1 cyan, CC2 magenta, CC3 yellow, CC4 salmon, CC5 white, CC6 blue, CC7 orange, and CC8 green. Figure 1C also shows the location of the bound FK506 (taken from the structure 1fkj) superimposed for reference. The ligand binding site contains three connected hot spots. These were the top-ranked hot spot, CC1, which contained 18 probe clusters, plus two secondary CCs, CC3 (14 probe clusters) and CC6 (7 probe clusters). The center-to-center distance (CD) between CC1 and CC3 is 6.3 Å, and the maximum dimension of the ensemble with the two hot spots is 10.6 Å. Thus, according to our condition, the site would be druggable when using a drug that binds only to these two hot spots. However, since CC1 is not much stronger than the required S=16, and the MD value is close to the required threshold of 10 Å, the affinity would be moderate. Since the CD distance between CC1 and CC6 is 7.2 Å, CC6 can be also added to the hot spot example used for the binding. Adding the third hot spot CC6 (7 probe clusters) increases MD to 12.3 Å. According to Table 1, all three hot spots in this extended ensemble still can be reached using druglike compounds. In fact, FKBP has been co-crystallized with smaller but still potent inhibitors. Examples are PDB ID 1fkh (MW=453, Ki = 7 nM),68 and PDB ID 1f40 (MW=360.5, Ki = 7.5 nM).69 However, the inhibitors that reach the three hot spots are fairly long, and connecting the two far ends by forming a macrocycle in FK506 improves the affinity to Ki = 0.2 nM.66 This example also demonstrates that while the analysis of druggability could be simplified by integrating the three different requirements into a single druggability measure, considering the three requirements separately can more explicitly show problems that may lead to the lack of druggability, as well as explore opportunities for using non-canonical compounds as drugs.
Figure 1.
Using hot spot properties to assess druggability. (A) The structure of FK506, considered in the example of druggability analysis by FTMap. (B) The structure of ABT737, a well-known example of protein-protein interaction inhibitors. (C) Mapping the ligand-free FK506 binding protein (PDB ID 2ppn, shown as surface), with bound FK506 (salmon sticks, from the structure with PDB ID 1fkj) superimposed for reference. The consensus clusters shown are CC1 (cyan, 18 probe clusters), CC3 (yellow, 14 probe clusters), and CC6 (blue, 7 probe clusters). The center-to-center distance (CD) between CC1 and CC3 is 6.3 Å, and the maximum dimension of the ensemble of the two hot spots is 10.6 Å. Thus, the sub-set of these two hot spots is druggable on its own, but would bind small ligands with only moderate potency. Using all three hot spots increases MD to 12.3 Å, and allows for high affinity binding. (D) Distribution of the parameters used for assessing druggability of the 23 protein targets in the Hajduk et al.7 data set. (E) Top view of the distribution shown in Figure 1D to show the separation of different druggability classes. Note that the non-canonically druggable – large (D*L) category is defined by the large (CD ≥ 8Å) distance between the centers of the hot spots. (F) Right view of the distribution shown in Figure 1D. The non-canonically druggable – small (D*S) category is defined by the small (MD < 10Å) dimension of the entire hot spot ensemble.
If several structures of a target are available, an important question is which structure to use for the analysis. Frequently druggability needs to be established before discovering any ligand, motivating preference for unbound structures. If multiple unbound structures are available, it is useful to map all, and consider the one that results in the highest level of druggability, assuming that ligands can be optimized to take advantage of the available hot spots. While ligand binding may affect their location and relative strength,70,71 hot spots are generally less sensitive to conformational changes than binding sites are,48 and the analysis of the ligand-free structures provides valid druggability assessment for most targets. However, the binding sites in the unliganded structures of some proteins may be completely closed, or ligand binding may create new hot spots, and hence it is generally advisable to also map ligand-bound structures when they become available in the process of drug discovery. This additional information frequently shows if there is a choice between a large and hence non-druglike ligand that reaches all major hot spots, or a smaller and less potent druglike ligand that can take advantage of only the main hot spot and some of the weaker hot spots around it.
Druggability results from NMR based screening versus predicted by FTMap
Table 2 shows the analysis of the 23 proteins in the test set from Hajduk et al.7 using the three FTMap criteria. In two cases, PTP1B (T19 in Table 2) and survivin (T22), the protein was found to contain two distinct binding sites that were characterized separately, resulting in a total of 25 binding sites. Druggability results are also shown graphically in Figures 1D, 1E, and 1F. Table 2 list the short names of the targets and specifies their binding sites as given by Hajduk et al.7 While we quote the Protein Data Bank (PDB) entry as given in the original publication,7 for our analysis we mapped both the highest resolution ligand-free and ligand-bound structures currently available in the PDB.55 As discussed, in most cases druggability can be determined by mapping the ligand-free structure alone, and the ligand-bound structure is needed only to judge whether the hot spots are located in the appropriate binding site for the purpose of retrospective validation with respect to experimentally identified ligands. For the protein PAK4 (T16 in Table 2) no structure was found in the PDB, and hence the druggability of this target is not discussed. The affinities shown in Table 2 are the ones given by Hajduk et al.,7 but we also list (in italics) if more potent compounds have subsequently been reported. Table S1 is a copy of Table 2, but its footnotes provide detailed information on targets and their affinities, with references. Additional references may not be listed if the value is given in PDBbind,72 BindingDB,73 or BindingMOAD.74 Table 2 also shows the experimental and predicted druggability scores (DS) calculated by Hajduk et al.,7 which represent the logarithm of the actual or predicted hit rate in an NMR-based fragment screen. These authors observed that most of the targets for which high-affinity, druglike leads could be identified had DS values greater than -1.0, whereas DS values below -1.5 indicated proteins without such ligands. Consequently, a protein pocket with DS > −1.0 was considered to have “high” druggability, while “low” druggability was assigned for targets with DS <−1.5. The DS value may be close to −2.0 for very undruggable proteins. Protein pockets with intermediate values (−1.5 <DS < −1.0) were assigned as having “moderate” druggability.
Table 2.
Druggability results from NMR based screening versus predicted by FTMap
| Target | Site | Structure7 | PDB | Affinity nM | Consensus sites | D1 | Druggability Score (DS)7 | Distances, Å | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|
|
|
|
|||||||||||
| U | B | Exp. | Pred. | To further CCs | Max. Dim. | |||||||
| 1 | AK | Adenosine | Internal | 1bx4 | 2i6b | <10 68 |
1bx4: 1(19), 8(4) 2a6b: 1(21), 6(6), 7(5) |
D | −0.66 | −0.42 | 1bx4: 7.3 2a6b: 6.5, 6.8 |
1bx4:11.5 2a6b:11.7 |
| 2 | Akt-PH | IP3 | 1h10 | 1unr | 1h10 | >1000 6300 |
1unr: 3(14), 4(10) 1h10: 5(9), 6(8) |
B | −1.91 | −1.98 | 1unr: 7.8 1h10: 7.0 |
1unr: 14.0 1h10:11.0 |
| 3 | Bcl-xL | Bak | Internal | 1r2d | 2yxj | <200 60 |
1r2d: 1(18), 2(17), 5(9) 2yxj: 1(28), 2(25), 4(9), 5(8) |
D | −0.11 | −0.64 | 1r2d: 7.4, 5.8, 2yxj: 6.3, 7.7, 7.0 |
1r2d: 12.2 2yxj: 16.5 |
| 4 | XIAP Bir3 | Pept. | 1g3f | -- | 2opz | 28 | 2opz: 1(23), 7(5) | D | −1.03 | −0.72 | 2opz: 3.1 | 2opz:10.6 |
| 5 | CMPK | CMP | Internal | 1cke | 2cmk | --- | 1cke: 1(20), 3(10) 2cmk: 1(19), 4(10) |
D | −1.13 | −0.81 | 1cke: 5.3 2cmk: 4.7 |
1cke: 14.1 2cmk: 11.0 |
| 6 | E2-31 | DNA | Internal | 1dhm | 1jj4 | >1000 60,000 |
1dhm: 5(12), 6(7) 1jj4: 4(11) |
N | −0.71 | −0.72 | 1dhm: 5.4 1jj4: - |
1dhm: 9.9 1jj4: 4.8 |
| 7 | ErmAM | SAH | 1qam | 1qam | 1qan | >1000 1100 |
1qam: 1(25), 3(16) 1qan: 2(15), 3(12), 5(8) |
D | −1.01 | −0.87 | 1qam: 5.6 1qan: 5.3, 7.8 |
1qam:11.7 1qan: 15.8 |
| 8 | FBP | DNA | 1j4w | -- | 1j4w | >1000 | 1j4w: 1(18), 2(15) | D*S | −1.61 | −1.04 | 1j4w: 5.1 | 1j4w: 9.8 |
| 9 | FKBP | FK506 | 1fkj | 2ppn | 1fkj | <10 2.3 |
2ppn: 1(18), 3(14), 6(7) 1fkj: 1(34), 2(16) |
D | −0.03 | −0.24 | 2ppn: 6.3, 7.2 1fkj: 7.9 |
2ppn: 11.7 1fkj: 10.7 |
| 10 | HI-0065 | ADP | 1fl9 | 1fl9 | 1htw | >1000 10,000 |
1fl9: 3(13), 5(8) 1htw: 2(19) |
B | −0.82 | −1.28 | 1fl9: 6.2 1htw: - |
1fl9: 11.1 1htw: 7.9 |
| 11 | LCK | PTyr | 1lkl | 3hck | 1lkl | 300 | 3hck: 3(18), 5(9), 6(7) 1lkl: 6(9), 8(3) |
D | −0.21 | −1.07 | 3hck: 6.6. 7.6 1lkl: 6.2 |
3hck: 11.2 1lkl: 9.3 |
| 12 | LFA | IDAS | 1rd4 | 1lfa | 1rd4 | <50 18.3 |
1lfa: 1(31), 4(12) 1rd4: 1(18), 2(13), 3(12), 7(6), 8(6) |
D | −0.40 | −0.35 | 1lfa: 5.6 1rd4: 5.7, 6.0, 4.7, 3.7 |
1lfa: 11.6 1rd4: 13.2 |
| 13 | MDM2 | p53 | 1ycr | 1z1m | 1rv1 | <100 | 1z1m: 1(21), 2(21) 1rv1: 1(21), 2(20), 3(19) |
D | −0.49 | −0.35 | 1z1m: 5.6 1rv1: 5.7, 4.7 |
1z1m:10.6 1rv1: 14.8 |
| 14 | MurA | UDPNAG | 1uae | 1ejd | 1uae | 250 | 1ejd: 2(22), 4(10), 5(8) 1uae: 1(21), 2(12), 3(11), 4(8) |
D | −1.38 | −1.44 | 1ejd: 4.3, .8, 5.9, 7.2 |
1ejd: 13.5 1uae: 16.0 |
| 15 | MurI | Glu | Internal | 1b73 | 1b74 | <20 | 1b73: 1(30), 2(14), 3(12), 4(11), 5(9), 6(8), 7(7) 1b74: 1(20), 3(14), 4(12), 6(9) |
D | −1.93 | −2.00 | 1b73: 5.3, 7.5, 5.3, 7.1, 3.0, 4.9 1b74: 4.0, 7.6, 3.8, |
1b73: 15.8 1b74: 14.4 |
| 16 | PAK4 | ATP | Inter. | --- | --- | --- | -0.78 | -0.63 | ---- | ---- | ||
| 17 | Pin1 WW domain | Pept. | 1i8h | 1i6c | 1i8g 1i8h |
>1000 | 1i6c: 1(28), 2(18), 4(13) 1i8h: 1(20), 6(11), 7(4) |
D*L | −0.94 | −1.49 | 1i6c: 8.0, 4.6 1i8h: 6.3, 5.6 |
1i6c: 14.0 1i8h: 13.7 |
| 18 | PDZ-PSD95 | Pept. | 1iu0 | 1iu2 | 1rgr | <300 | 1iu2: 1(22), 5(11) 1rgr: 1(22), 6(7) |
D*L | −2.00 | −1.99 | 1iu2: 9.8 1rgr: 5.9 |
1iu2: 12.6 1rgr: 8.7 |
| 19 | PTP1B | Cat. pY | 1ph0 | 3a5j | 1pty | <10 | 3a5j: 1(20), 2(16) 1pty: 1(13), 7(7) |
D | −0.68 | −1.15 | 3a5j: 6.8 1pty: 5.3 |
3a5j: 11.0 1pty: 9.0 |
| 19 | PTP1B | non-cat. pY | 1ph0 | 3a5j | 1pty | >1000 | 3a5j: 4(9), 7(7), 8(7) 1pty: 8(7), 9)7) |
N | −1.77 | −1.66 | 3a5j: 5.0, 6.2 1pty: 5.9 |
3a5j: 11.4 1pty: 10.2 |
| 20 | SARS N-term | RNA | Internal | 1ssk | ---- | >1000 ~106 |
none | N | −1.93 | −1.92 | -- | -- |
| 21 | SCD | Subst. | 1g4k | 1cqr | 1g4k | <10 | 1cqr: 1(26), 5(7) 1g4k: 1(20), 2(17), 5(9), 7(4) |
D | −0.09 | −0.55 | 1cqr: 4.7 1g4k: 4.6, 4.8, 3.2 |
1cqr: 12.3 1g4k: 13.0 |
| 22 | Survivin | Bir3 | 1e31 | 1e31 | 3uih | >1000 | 1e31: 7(5) 3uih: 8(5) |
N | −1.97 | −1.99 | 1e31: --- 3uih: --- |
1e31: 4.3 3uih: 4.1 |
| 22 | Survivin | other | 1e31 | 1e31 | ----- | <300 | 1(24), 2(24) | D*L | −0.45 | −0.35 | 1e31: 8.3 | 1e31: 13.9 |
| 23 | UK | Pept. | 1fv9 | 2o8t | 1fv9 | <50 | 2o8t: 1(29), 2(26) 1fv9: 1(18), 2(15), 5(9) |
D | −0.40 | −0.81 | 2o8t: 6.0 1fv9: 4.3, 2.1 |
2o8t: 17.8 1fv9: 13.0 |
Druggability class based on FTMap analysis, as described in Table 1.
According to the FTMap results, 14 of the 25 distinct binding sites in this test set satisfy all three criteria for druggability: i.e. the top-ranking CC in the cluster contains at least 16 probe clusters, the ensemble contains at least two CCs with a center-to-center distance of less than 8 Å, and the maximum dimension of the ensemble is at least 10 Å. These 14 sites are therefore classified as druggable in our analysis, indicating a strong likelihood of finding a druglike small molecule ligand that binds strongly to the site. Such ligands are known for 12 of these 14 targets (see Table S1). The exceptions are cytidine monophosphate kinase (CMPK, T5) with only micromolar ligands identified so far,75 and rRNA methyltransferase ErmC (ErmAM, T7), for which the best known inhibitor has IC50~1100 nM.76 Two targets, Akt-PH (T2) and HI-0065 (T10), are predicted as borderline druggable, and have micromolar ligands.77,78 Four sites (E2-31, the non-catalytic phosphotyrosyl binding site of PTP1B, the N-terminal domain of SARS, and the Bir3 domain of survivin) are classified as not druggable, in each case because of a weak main hot spot (S <13). According to FTMap, three targets (the WW domain of Pin1, PDZ-PSD95, and the “other” site of survivin) belong to the D*L category, and thus achieving substantial potency requires peptide-like or macrocyclic ligands. PZD-PSD95 (T18) was considered not druggable by Hajduk et al.7 (DS=-2.00), but FTMap identifies two hot spots in the site that are 9.8 Å apart (Figure 2A), and hence based on the FTMap results this site can be targeted by peptides or macrocycles that are large enough to reach the two hot spots.79 In fact, PZD-PSD95 binds cyclic peptides with IC50 ~ 1000 nM in a solvent exposed shallow pocket.79 The two main hot spots are also too far apart in the peptide binding site of Pin1 WW domain (T17). We note that based on the PDB code (1ih8) given by Hajduk et al.,7 we consider here the WW domain of Pin1 that has only peptides as known ligands,80 rather than the peptidyl prolyl cis/trans isomerase (PPIase) domain that is the target of more recent studies.81–83 Survivin (T22 in Table 2) has two binding sites of interest. As shown in Figure 2B, in contrast to XIAP Bir3, the Smac peptide binding site of this Bir3 domain includes only CC7 with 5 probe clusters, and hence is not druggable. However, the “other” site in Table 2, close to the dimer interface, includes two strong hot spots that are 8.3 Å apart, almost within the reach of druglike ligands. Inhibitors that binds to both hot spots have Kd values as low as 37 nM.84 Finally, based on the FTMap results, one target, FUSE-binding protein (FBP, T8 in Table 2) is in the D*S category due to the small size of the hot spot ensemble. In fact, the only known inhibitors of DNA binding to FBP are charged and of high micromolar affinity.85
Figure 2.
Hot spots in two protein-protein interaction targets, belonging to the druggability class D*L. (A) Mapping of the ligand free PDZ-1 domain of PSD-95 (PDB ID 1iu2, shown as surface), with a cyclic peptide (from PDB ID 1rgr, shown as yellow stick model) superimposed for reference. The two hot spots in the interface, CC1 (cyan, 22 probe clusters) and CC5 (white, 11 probe clusters), are at 9.8Å from each other. (B) Mapping of the ligand-free survivin dimer (PDB ID 1e31, shown as cyan and green cartoon), with the Smac/DIABLO peptide (from PDB ID 3uih, shown as yellow cartoon) superimposed for reference. The only hot spot in the survivin/Smac interface is CC7 (orange, 5 probe clusters), and thus the peptide binding site is not druggable. A novel site is defined by the strong hot spots CC1 (cyan, 24 probe clusters) and CC2 (magenta, also 24 probe clusters) at the dimerization interface. Since the center-to-center distance between CC1 and CC2 is 8.3Å, ligands that bind to both are over the 500 g/mol MW limit.
The FTMap results generally agree well with the assessment based on experimental and predicted druggability scores given by Hajduk et al.,7 as well as with the actual success or failure to identify a high affinity druglike ligand that binds to the site in question. We have substantive differences with the DS value from Hajduk et al. only for 3 of the 25 sites. Moreover, the available experimental data suggest that, in these three cases, the druggability assignment based on FTMap is in fact the correct one. Specifically, the DNA binding site of the human papillomavirus (HPV) E2-31 protein (Target T6 in Table 1) is considered druggable by Hajduk et al.,7 but not by FTMap. The best inhibitor of the HPV E2 DNA binding site in the literature has an IC50 value of approximately 10 μM,86 and due to the consistent lack of success in finding more potent ligands for this site efforts have been largely redirected to target the DNA rather than the protein.87,88 Moreover, the majority of DNA binding sites of proteins have limited druggability.6 FTMap also differs from the DS method in predicting that the adenosine diphosphate (ADP) binding site of the hypothetical bacterial ATPase HI-0065, also known as YjeE (T10 in Table 1), is at most borderline druggable. HI-0065 was targeted by the Abbott group,78 but the best compound identified by NMR based screening and optimization had a KD value of about 5 μM. We note that both HPV E2-31 and HI-0065 have strong binding hot spots close to, but not overlapping with, the ligand binding site, which may have resulted in a misleadingly high NMR fragment hit rates. The third deviation between FTMap and the earlier results occurs for the glutamate racemase MurI that was rated as not druggable by Hajduk et al.,7 although they reported that 4-substituted D-glutamic acid analogs (not considered druglike) bind with nanomolar affinity.89 It is not clear why the NMR based screening did not result in hits. According to FTMap this target is druggable, emphasizing that our condition does not directly account for the lack of druglikeness due to the need for a charged ligand, unless the hot spot region is too small. In this particular case we note that a close homolog, glutamate racemase from Helicobacter pylori, has potent and druglike non-competitive inhibitors that bind to the corresponding site.90,91
Comparison of FTMap results obtained using the ligand-bound structure rather than the unbound structure of the protein (Table 2) shows that small differences between the structures can affect the strength and location of hot spots, but that such changes impact druggability assessment only for three targets, HI-0065, LCK and PTP1B, all having strongly charged ligands with a phosphate group (see footnotes to Table S1). It appears that the binding of such ligands tighten the binding pocket, which then binds fewer probe clusters. Thus, the mapping of ligand-free proteins generally provides valid druggability information, emphasizing that druggability is an inherent property of a protein. Nevertheless, as will be further discussed, it may be useful to also map ligand-bound protein structures if available, since in some cases ligand binding may open additional subsites and create additional hot spots, favoring druggability.
Validation of FTMap druggability assessment against extended 72-protein set of Hajduk et al.7
In addition to analyzing the 23 drug targets for which experimental NMR fragment screening results were available, Hajduk et al.7 calculated druggability indices for a set of 72 proteins based on co-crystal structures with ligands. They did this by constructing a regression equation to calculate a druggability score based on geometric features of the binding site, including the total and apolar surface area, compactness, and the first and third principal moments of the pocket, which they parameterized using the results of NMR fragment screening of the original 23 protein test set. The 72-protein set includes 35 proteins for which high affinity, druglike molecules have been reported, plus 37 proteins for which the known ligands were either not high affinity or not druglike. Table S2 compares the results of FTMap analysis of this extended 72-protein set to the druggability scores (DS) reported by Hajduk et al.7
For each of these 72 targets we mapped the highest resolution ligand-free structure available in the PDB. Bound structures are listed only to specify the binding site of interest. Among the 35 proteins that were considered by Hajduk et al. to have druggability, the FTMap results disagree with the high DS value only for two targets, cytochrome c peroxidase (Target T17) and leghemoglobin (Target T32). Target T17, given by PDB ID 1ccg,7 is actually the His175Gly mutant of the cytochrome c peroxidase, which binds imidazole in a cavity created by the removal of His175 side chain, but with a Kd value of only 2.7 mM.92 FTMap finds this imidazole binding site not druggable. Although a number of strong hot spots exist in the large cavity of the protein, these do not overlap with the bound imidazole position. The problem with leghemoglobin (T32) is that the binding site is formed in the process of ligand binding, and hence only the bound structure, which was studied by Hajduk et al.,7 reveals the druggability of this target. Since we generally consider the mapping of the ligand-free structure to be more predictive of druggability, this may appear as a shortcoming of our approach. However, leghemoglobin is not a drug target and its binding pocket is extremely small, accommodating just a nicotinate with molecular weight 122 g/mol. 93 As we have discussed, such small ligands are unlikely to be drugs, and in fact the binding of nicotinate is weak (KD = 1400 nM).93
Among the remaining 37 structures that did not have high affinity druglike ligands, the agreement between FTMap and DS druggability predictions was also generally good, however we again see some differences. Phosphoserine phosphatase (T36) is rated as druggable by Hajduk et al.7 (DS=−0.808), but only borderline druggable by FTMap. No potent inhibitor is indicated by Hajduk et al.,7 nor has such a compound been reported in the subsequent literature. In the apo structure of flavodoxin (T37, DS=−0.857), the side chains of W57 and Y94 completely close down the flavin mononucleotide binding site,94 and hence FTMap does not find a hot spot there. As mentioned, proteins for which the druggability of the bound conformation is so greatly different from that of the apo-protein are very rare; we have found only two cases (leghemoglobin and flavodoxin) among the more than 150 proteins we have studied, and these two are not drug targets. FTMap confirms druggability for most other targets with DS > −1.2 (Targets T38 through T45), and apart from inositol monophosphatase (T44) these proteins indeed have known potent ligands, as does sialidase (T46) with DS=−1.201 (Table S2). Targets PI phospholipase (T47) and enterotoxin (T48), with lower DS values, we rate as borderline druggable, and phosphofructokinase (T49) as not druggable. Consistent with these ratings, none of these proteins has any known potent ligand. The druggability of targets with DS values between −1.334 and −1.5 is somewhat undefined by Hajduk et al.7 FTMap shows substantial variation in this range, and for several proteins our results are supported by binding data. In particular, protein kinase Cδ (PKCδ, T50 in Table S2) is in the D*S category, and it binds phorbol-12,13-dibutyrate, a non-druglike compound with MW = 504.6 g/mol, with nanomolar Kd.95 FTMap confirms the druggability of the GTP binding site of Ras p21 (T52 in Table S2). Guanylate kinase (T53) is not druggable, whereas based on FTMap chloramphenicol acetyltransferase (T54) is in the D*S category, and indeed is known to bind fusidic acid with Ki = 5400 nM.96 FTMap predicts to be druggable seven targets (aldehyde ferredoxin oxidoreductase, galactose binding protein, cytidine deaminase, trichosanthin, achromobacter protease, diaminopimelic acid dehydrogenase, and glutamine methyltransferase) that were assigned a low druggability index (DS < −1.5) by Hajduk et al.7 In three cases the FTMap assignment is supported by known affinity data. These three are the galactose binding protein (T61), which binds galactose with Kd=400 nM,97 cytidine deaminase (T62), which has potent inhibitors;98 and achromobacter protease (T64), which has inhibitors with Ki = 6.5 nM.99 We could not find data confirming druggability of the remaining four targets, aldehyde ferredoxin oxidoreductase (T60), trichosanthin (T63), diaminopimelic acid dehydrogenase (T66), and glutamine methyltransferase (T71), but in three cases – T60, T63, and T66 – the proteins are known to bind (nondruglike) small substrates or cofactors with relatively high affinity (see Table S2). In the same range (i.e., with DS< −1.5) we predict four targets in the D*S category. Two of these, periplasmic glucose/galactose receptor (T69) and IP phosphatase (T70), are known to bind charged ligands with nanomolar affinity (Table S2). We have no information on the other two, deoxynucleoside monophosphate kinase (T56) and glucoamylase (T58). Finally, Table S2 has one target, pp60Src SH2 domain (T57), with DS <−1.5 that FTMap places in the D*L category. This domain is known to bind a peptidomimetic ligand with IC50=1800 nM.100
As shown, we were able to find binding data for a substantial fraction of targets that were predicted to be druggable by FTMap, but had DS <−1.5 assigned by Hajduk et al.7 This poses the question why such low DS values were predicted. The most likely reason is that the regression formula for DS is based on the properties of pockets rather than of the hot spots, and this may underestimate potential binding capacity for pockets that have somewhat unusual shape. Indeed, targets T55, T60, and T61 all have completely buried ligand binding sites with no solvent accessibility in the ligand-bound structures considered by Hajduk et al.,7 which likely contributed to their low DS values.7 However, FTMap can still place probes in such internal cavities, and the proteins are likely to have alternative conformations with more open pockets. Trichosanthin (T63) and glutamine methyltransferase (T71) have deep and narrow binding pockets, whereas the binding site in diaminopimelic acid dehydrogenase (T66) is largely solvent exposed, and it seems that the simple regression formula by Hajduk et al.7 underestimates the druggability for such unusually shaped pockets.
Validation of FTMap druggability assessment against the test set of Cheng et al. 8
A biophysical model developed by Cheng et al.8 accounts for lipophilic surface area and curvature of binding sites to compute a score that corresponds to the maximal affinity predicted for a passively absorbed oral drug (MAPPOD). MAPPOD is based on the hypothesis that the maximal achievable binding free energy is largely due to desolvation, and models the desolvation contributions from target and ligand in terms of nonpolar surface areas with coefficients that depend on surface curvature. The expression does not have an explicit polar desolvation term, since it is assumed that the electrostatic interactions and the desolvation energies largely cancel each other.8 This model was used to assess the druggability of 27 binding sites. The value of this test set is that all sites are of pharmaceutical interest, in contrast to later test sets that included many proteins without this property. The 27 target sites were classified as druggable (17), “difficult” (6), or undruggable (4). Nearly all targets classified as druggable or difficult had associated orally bioavailable, marketed drugs whereas the ‘undruggable’ targets did not, despite considerable efforts within the industry.
In Table S3 we compare the FTMap results to the maximal affinity (MAPPOD) values predicted by Cheng et al.8 that were recently reviewed.101 Both ligand-free and ligand-bound structures were mapped, but as mentioned we consider the unbound results more relevant for assessing druggability. The agreement with the results of Cheng et al.8 is generally good. For the 23 druggable or difficult targets we have different results only for the binding site of COX-2 (Target T6 in Table S3), which FTMap places into the D*S category due to the small size of the hot spot ensemble, predicting that potent ligands binding to this site must have a charged or highly polar group not fully desolvated upon binding. Indeed, all strong COX-2 inhibitors seem to have either a carboxyl or a terminal sulfonamide group.102 Cheng et al.8 examined six targets that they considered ‘difficult’, namely ACE-1 (T1), HIV RT nucleotide site (T13), IMPDH (T16), neuraminidase (T18), penicillin binding protein (T22), and thrombin (T23), because they required highly polar molecules that were either processed from prodrugs or relied on active transport mechanisms. Thrombin (T23) is predicted to be druggable both by Cheng et al.8 and by FTMap, and now has direct (i.e., not prodrug) oral inhibitors.103,104 With 15 probe clusters, the penicillin binding protein (T22) is borderline druggable by FTMap, and ligand binding substantially strengthens the hot spot. This protein binds piperacillin,105 an approved but not orally bioavailable drug (MW= 517.5 g/mol), with an IC50 of 166 nM.106 FTMap predicts that ACE-1 (T1), HIV RT nucleotide site (T13), IMPDH (T16), and neuraminidase (T18) are all druggable, though the primary hot spot in IMPDH is relatively weak, with only 17 probe clusters, and ACE-1 and neuraminidase have strong but small hot spots (MD < 11.0). Both ACE-1 and neuraminidase have approved drugs that include a carboxyl group but are not pro-drugs, for example N2-[(1S)-1-carboxy-3-phenylpropyl]-L-lysyl-L-proline (lisinopril)107 for ACE-1, and (2R,3R,4S)-3-acetamido-4-(diaminomethylideneamino)-2-[(1R,2R)-1,2,3-trihydroxypropyl]-3,4-dihydro-2H-pyran-6-carboxylic acid (zanamivir)108 for influenza neuraminidase. These results confirm that ACE-1 and neuraminidase are correctly classified as druggable, but also show that a small hot spot is likely to require a charged ligand. Two targets, Factor Xa (T10) and HMG CoA reductase (T15), were placed on the border of the druggable region by Cheng et al.13 Factor Xa is categorized by FTMap as D*L, and it has druglike inhibitors close to the upper limit on size. For example, the inhibitor methyl(2R,3R)-3-([[3'-(aminomethyl)biphenyl-4-yl]carbonyl]amino)-2-(3-carbamimidoylbenzyl)butanoate (RPR128515),109 bound in the structure 1ezq, has the molecular weight of 458.6 g/mol.109 Both Cheng et al.8 and FTMap underestimate the druggability of HMG CoA reductase, which has potent statin inhibitors,110 but is considered only borderline druggable by FTMap (Table S3). This is due to the fact that statins occupy only a portion of the large ligand binding site. While this is enough to block access of the substrate to the active site of the enzyme, the unoccupied region also includes a number of hot spots, thus resulting in fewer probe clusters overlapping with the inhibitor.110
Cheng et al. 8 classify four targets as not druggable: cathepsin K (T24), Caspase 1 (T25), the active site of HIV integrase (T26), and PTP1B. Cathepsin K (T24) and the catalytic site of the HIV integrase are predicted to be druggable by FTMap. This finding is supported by data that became available after the publication of the analysis by Cheng et al.8 Cathepsin K has acrylic cyanamide-based inhibitors with Ki values as low as 0.003 nM.111 The osteoporosis drug N-(1-cyanocyclopropyl)-4-fluoro-N2-[(1S)-2,2,2-trifluoro-1-[4'-(methylsulfonyl)biphenyl-4-yl]ethyl]-L-leucinamide (Odanacatib),112 in Phase III clinical trial by Merck, is a potent cathepsin K inhibitor, and current studies focus on safety issues rather than affinity or oral bioavailability.113 For HIV integrase there exist several FDA approved orally bioavalable inhibitors such as N-(4-fluorobenzyl)-5-hydroxy-1-methyl-2-(2-[[(5-methyl-1,3,4-oxadiazol-2-yl)carbonyl]amino]-2- propanyl)-6-oxo-1,6-dihydro-4-pyrimidinecarboxamide (raltegravir)114 with low nanomolar IC50 values. These “newly” druggable sites emphasize the importance of assessing druggability using biophysical rather than purely statistical methods. However, we agree with Cheng et al.8 that the other two targets are very difficult. FTMap results categorize Caspase 1 as borderline druggable at best, due to the weak hot spots and the small size of the hot spot region. Indeed, although there are peptidomimetic inhibitors containing a carboxyl group with Ki values as low as 5 nM, their oral bioavailability is limited,115 and based on our mapping results it is unlikely that more druglike inhibitors will be found. The druggability of PTP1B will be discussed in details later in this paper, and hence is not considered here.
Validation of FTMap druggability assessment against DrugPred
Krasowski et al.12 compiled a non-redundant data set containing crystal structures of 71 druggable and 44 less druggable proteins. This data set was used to train a structure-based druggability predictor called DrugPred. We did not apply FTMap to this entire set because, in contrast to the set by Cheng et al.8, it includes many proteins that have not been targets of substantive screening campaigns. However, we mapped the targets for which DrugPred clearly failed. These included six targets (DPP-IV, urokinase plasminogen activator, factor Xa, human thymidine phosphorylase, thiamine pyrophosphokinase, and thymidine kinase) that were included as druggable in either training or validation sets by Krasowski et al.12 but were predicted as undruggable by DrugPred, and two proteins (peptide deformylase and CDP-D-glucose synthase) that were included as undruggable, but were predicted to be druggable (see Table S4). These analyses were performed to obtain more information on the likely druggability of these targets, and to explore whether the FTMap results differ from those of DrudPred. Of the six targets that were considered to have false negative predictions,12 FTMap correctly predicts two (DPP-IV and urokinase plasminogen activator) to be druggable. As already shown, factor Xa is categorized as D*L, but it still binds druglike ligands, albeit at the upper limit of molecular weight.109 Human thymidine phosphorylase is classified as B*S, and it is known to require charged ligands or prodrugs for nanomolar affinity.116 The two remaining false negatives (thiamine pyrophosphokinase and thymidine kinase) are predicted as not druggable also by FTMap. While we did not find any evidence that thiamine pyrophosphokinase has potent inhibitors, the thymidine kinase of the herpes simplex virus has inhibitors with Ki values as low as 5 nM.117 According to FTMap, the thymidine kinase binding site has only weak hot spots both in apo and holo structures (Table S4), making this target the sole real failure of our method. The apparent reason is that the thymidine kinase inhibitors are planar molecules that bind in an extremely narrow and fully buried pocket that can accommodate only the few probes that are also planar. This problem is very rare, and at this point we have no solution to avoid it. The last two targets in Table S4 were considered false positives by Krasowski et al.12 and were also predicted to be druggable by FTMap. Of these, peptide deformylase now has low nanomolar inhibitors.118 CDP-D-glucose synthase binds cytidine-5'-triphosphate with Ki = 35 μM,119 but it has not been considered a drug target and hence its druggability has not been established experimentally.
TARGETTING CONVENTIONALLY UNDRUGGABLE TARGETS WITH NON- CANONICAL DRUG CHEMOTYPES
Because the FTMAP algorithm evaluates the binding potential of protein surface sites based on structural and physicochemical principles, rather than by empirical parameterization using a particular test set of target proteins, the method has the potential to provide information on the druggability of sites that differ in structure or properties from those that previously have been successfully targeted in drug discovery efforts. We therefore evaluated whether the FTMap method can shed light on precisely why sites that were not assessed to be druggable failed to qualify. This information is potentially useful in deciding whether the site might be targeted by a non-canonical drug chemotype, such as a peptide, a macrocycle, or a covalent inhibitor, or whether the target is likely intractable to all approaches currently available for small molecule drug discovery. Of the targets in Table 2 that were rated as not druggable, four of them – E2-31, the non-catalytic site of PTP1B, the SARS N-terminal domain, and the bir3 site of survivin – simply lack a hot spot of sufficient strength to serve as a strong “anchor site” for ligand binding. Such targets are unlikely to bind strongly to any small ligand, and so we consider them to likely be undruggable by any such means. However, PDZ-PSD95 and the WW domain of Pin1 have strong hot spots that are positioned too far apart to be easily spanned by a ligand of typical drug size, and in the “other” site of survivin the separation of hot spots also slightly exceeds 8 Å. In these cases, one might expect that these sites will not be conventionally druggable, but might bind strongly to a somewhat larger ligand such as a peptide-derived foldamer or a large macrocycle, and hence are classified as non-canonically druggable – large, denoted by D*L. Other examples include the pp60Src SH2 domain, elongation factor TU, the catalytic domain of protein phosphatase-1, and the site on interleukin-2 (IL-2) that binds the IL-2 receptor α-chain. In each of these cases, FTMap analysis indicates that the binding site contains a sufficient number of strong hot spots to support high affinity binding of a ligand, but these hot spots are spaced too far apart to be encompassed by a ligand the size of a typical drug, and thus the site requires a larger chemotype to achieve strong binding (see Table S1–S6 for details). Furthermore, we have recently reported and further discuss below that the binding sites on proteins binding natural product macrocycles of MW > 600 g/mol differ from conventional drug binding sites in having hot spots that are spaced further apart, supporting the notion that large ligands such as large macrocycles can bind to sites that have sufficiently strong hot spots but fail the standard druggability criterion due to the spacing between the hot spots being too large.61
Druggability of proteins binding macrocyclic compounds
It is generally recognized that macrocyclic compounds can be useful to expand the class of druggable targets, and provide additional options against DNA, RNA and carbohydrate binding proteins and protein-protein interactions.18,20,21 In Table 3 we show druggability results for a number of proteins that bind macrocyclic natural products with high affinity (except for actin, where the binding is at best micromolar). Additional details on individual targets are given in Table S5. These targets include the already discussed FK506-binding protein (Figure 1C), which FTMap indicates is druggable even using druglike compounds as confirmed by published experimental data,68,69 albeit not with the very high affinity of the macrocyclic FK506 (Figure 1A).66 We find a similar result for a number of other targets in Table 3. Cyclophilin A, pancreatic elastase, chitinase, and HSP90 are all predicted to be conventionally druggable and indeed, in addition to their potent nanomolar macrocyclic inhibitors, these proteins are known to also have somewhat less potent non-macrocyclic inhibitors (see Table S5). However, in the case of three proteins, elongation factor TU, protein phosphatase 1, and the biotin carboxylase (BC) domain of human Acetyl-CoA Carboxylase 2, the hot spots are too far from each other to render the sites conventionally druggable, and hence these sites can be targeted only using larger compounds such as macrocycles or peptide foldamers.120 According to FTMap, actin is not druggable by any means. In fact, although the protein has been co-crystallized with several large macrocycles and the compounds have biological activity, the binding is very weak in all cases (Table S5). These results support our previous suggestion61 that sites that fall outside the FTMap criteria for conventional druggability for the sole reason that the hot spots are spaced too far apart might be good targets for macrocyclic compounds or other larger ligands. For this reason, we classify such sites as non-canonically druggable D*L (Table 1), to indicate that they are not druggable using conventional druglike compounds, but could be druggable using other, larger but still pharmaceutically relevant drug chemotypes.
Table 3.
Druggability of proteins binding macrocyclic compounds
| Protein name | PDB | Hot spots | D1 | Comment | ||
|---|---|---|---|---|---|---|
|
| ||||||
| U | B | |||||
| 1 | Cyclophilin A | 2cpl | 1cwa 1sfa |
1(23), 3(15), 5(6) | D | CC1 and CC5 are overlapping, CC5 to CC3 = 5.8 Å, MD=14.0 Å, druggable by druglike compounds. |
| 2 | FKBP | 2ppn | 2tke 2dg3 |
1(18), 3(14), 6(7), 7(6) | D | CC1, CC3, and CC7 are close and make it already druggable by druglike compounds (MD=10.5 Å); CC1 to CC6 = 7.2 Å. |
| 3 | Pancreatic elastase | 1lvy | 1okx | 2(19), 5(8), 6(6), 7(5) | D | CC2 is elongated, MD=9.5 Å (borderline), but CC2 to CC7 =6 Å, CC6 to CC4= 5.3 Å, CC2 to CC6 7.2 Å, thus all are connected. |
| 4 | Elongation factor TU | 1efc | 1dt8 | 1(21), 4(12), 6(5) | D*L | CC1 and CC6 are close but too small (9.0 Å); CC1 to CC4 =11.5 Å, too far. Druggable only using a long ligand (peptide) or a macrocycle. |
| 5 | Protein phosphatase 1 | 3egg | 3e7a | 2(16), 6(7) | D*L | CC2 to CC6 12.9 Å. Too far, druggable only using either a macrocycle or a very elongated ligand. |
| 6 | Type I signal peptidase | 1kn9 | 1t7d | 2(18) | D*S | CC2 is elongated, but MD = 10.0 Å. Small and weak hot spot. |
| 7 | Chitinase | 1guv | 1waw 1wb0 |
1(23), 4(10), 5(6), 6(6) | D | Overlapping hot spots, total MD=12.6 Å. Also druggable using druglike ligands |
| 8 | Actin | 1nwk | 1qz5 | 3(14) | N | Inherently not druggable, additional surface area is needed. |
| 2q0u | 2(16), 7(4) | CC2 to CC7= 8.0 Å, too weak hot spots, at most borderline. | ||||
| 2q0r | 10(2) | Inherently not druggable, additional surface area is needed. | ||||
| 9 | AcetylCoA Carboxylase |
3glk | 3gid | 3(15), 4(9), 5(8), 6(6) | B*L | CC3 and CC6 are close, but MD=9.0 Å only. CC5 is close to CC4, but the distance between the CC3 and CC5 is 9.0 Å, too large. Druggable only using a macrocycle or very long compound. |
| 10 | HSP90 (closed) | 1yer | 1yet 3inw |
1(23), 3(10), 5(8), 6(8) | D | CC1, CC3, and CC5 provide druggability, MD=14.5 Å; CC1 to CC6 is 8.9 Å, too far; macrocycle is useful, but not necessary. |
| HSP90 (open) | 1yes | 1yet 3inw |
1(22), 2(13), 3(12), 5(9) | D | CC1, CC2, and CC5 provide druggability, MD=11.4 Å; CC2 to CC3 = 7.67 Å, also part of the hot spot ensemble. | |
Druggability class based on FTMap analysis, as described in Table 1.
Druggability of protein-protein interaction targets
A substantial number of PPI interfaces are biologically compelling targets for drug discovery, and in some cases small molecules are known to inhibit the interactions of two proteins with moderate to high potency.121,122 The druggability of such targets is still poorly understood, however, and PPI target sites are generally considered to be highly challenging if not undruggable. The problem in assessing the druggability of PPI targets is that protein-protein interfaces generally have much smaller pockets than traditional drug targets, and it is difficult to determine which one, if any, will be able to bind a small compound with sufficient affinity to modulate the interaction at pharmacologically relevant doses.123 We recently showed that FTMap can identify the most important ligandable interface binding sites based on analysis of the unbound protein structure.48 Here we review these results in the light of our current and more extensive druggability analysis (Table S6). The PPI targets T1 through T8 of Table S6 (Bcl-xL, XIAP Bir3, MDM2, Pin1, PDZ-PSD95, the survivin Bir3 domain, the “other” site on survivin. and urokinase) are already in the Hajduk et al.7 data set of Table 1, and we have previously studied the druggability of targets T9 through T12 (IL-2, HPV-11 E2, TNF-α, and stromelysin 1 catalytic domain).48 Targets T13, T14 and T15 (ZipA, K-Ras, and HIV integrase) will be described in more detail in the next section. Based on the mapping results collected in Table S6, five of the 15 PPI targets (Bcl-xL, XIAP Bir3, MDM2, urokinase, and HPV-11 E2) appear to be druggable using druglike compounds. However, the binding of small ligands in protein-protein interfaces may introduce conformational changes that affect the location and relative strength of the hot spots. While these changes do not affect the predicted druggability, they may impact the class of compounds required for strong binding. For example, in the ligand-free structure of Bcl-xL the three hot spots are close to each other, indicating druggability. Indeed, it has been reported that fragments of known inhibitors all bind in the pocket that binds L578 of the BAK peptide.124 Upon binding of the inhibitor ABT737, shown in Figure 1B,125 the hot spots move farther apart (Table S6). Nevertheless, Bcl-xL is assessed to be druggable also on the basis of mapping the ligand bound structure, since the distance from CC1 to CC5 is 6.3 Å, from CC5 to CC4 is 7.7 Å, and from CC4 to CC2 is 7.0 Å. However, to achieve high potency, ABT737 must extend from CC1 all the way to CC2, a distance of 11.3 Å, and this requires long molecules with MW >650 g/mol.48,126,127 In XIAP Bir3, the two hot spots are close to each other, and several potent monomeric inhibitors have been developed.128 However, the hot spot region is quite small (MD=10.6 Å), thus the XIAP Bir3 is close to the D*S category. Accordingly, the inhibitors are druglike but relatively large. For example, the inhibitor (2S)-1-[(2S)-2-cyclohexyl-2-[[(2S)-2-(methylamino)propanoyl]amino]acetyl]-N-(4-phenylthiadiazol-5-yl)pyrrolidine-2-carboxamide (GDC-0152)128 has the MW of 498.2, and recently macrocyclic inhibitors have also been developed.129 Mapping results for MDM2 are similar: the protein has strong hot spots close to each other, but the region is small (MD=10.6 Å), and the inhibitors in the Nutlin family130 and related compounds spread beyond the hot spots, resulting in MW of over 600 g/mol.131 However, MD exceeds the threshold of 10.0 Å, and more recent optimizations succeeded to yield inhibitors with MW below 600 g/mol, and even slightly below 500 g/mol.132,133 Urokinase is druggable, and druglike nanomolar inhibitors are known.134 HPV-11 E2 is also druggable but the hot spot region is small (MD=10.8 Å), and known inhibitors with high nanomolar affinity have MW > 500 g/mol.135 Based on FTMap, three targets (the Smac peptide binding site of survivin, the FtsZ interface of ZipA, and the receptor binding interface of TNF-α) are not druggable with noncovalent inhibitors under any circumstances.48 In fact, all three interactions have been targets of extensive screening campaigns that did not produce any potent compounds. The SOS-binding interface of KRAS, which is discussed in greater detail below, is borderline druggable, and the best noncovalent inhibitor found so far has KD = 190 μM.136 Three targets in Table 6 (the WW domain of Pin1, PDZ-PSD95, the interface of IL-2 with IL-2Rα) contain strong hot spots but with spacing that exceeds 8 Å, and thus fall into the Non-Canonically Druggable–Large (D*L) category, and are druggable only using large (peptide-like or macrocyclic) ligands. Since the 8.3 Å distance between the two strong hot spots in the “other” site of survivin barely puts this target into the D*L category, relatively small (though not <500 g/mol) potent ligands have been developed.84 In the unbound structure of the stromelysin 1 catalytic domain the ligand binding pocket is partially closed, and since the hot spot ensemble is small, FTMap predicts druggability in the D*S category. We also mapped the structure complexed with the inhibitor 5-methyl-5-(4-phenoxyphenyl)pyrimidine-2,4,6(1H,3H,5H)-trione (PDB ID 1g4k),137 which showed a more extended hot spot region and hence suggested a higher level of druggability. However, it appears that the most potent inhibitor still has only Ki =192 nM.137 Although we have only a limited number of examples, the ones considered here suggest that the even the druggable PPI targets frequently require the use of compounds lying outside of traditional druglike chemical space.
DRUGGABILITY ANALYSIS OF SELECTED HIGH VALUE TARGETS
In this final section we apply our analysis to a number of targets that have been of high pharmaceutical interest and for which the question of druggability is not fully settled. Most of the targets considered in this section are complex and undergo significant conformational change upon ligand binding, which may affect the strength and location of hot spots. Therefore it is important to recall that based on our experience drawn from the analysis of more than 150 proteins, as described above, for druggability it is necessary to identify appropriate hot spots in ligand-free protein structures, and it is the hot spot properties of the unliganded proteins that provide the most valuable information for establishing druggability. As discussed, the analysis of ligand bound structures can, however, be useful for understanding the changes in the relative importance of the binding energy hot spots upon ligand binding. In addition, as shown for PTP1B, the analysis of hot spots can also indicate that a target that can bind small ligands with high affinity can nevertheless be undruggable because any such ligands will not be able to satisfy additional requirements for specificity and/or for membrane permeability. We additionally note that, even for targets with good druggability, lead and drug discovery can be challenging when pursued by particular approaches. For example, although high druggability generally correlates with the observation of a high hit rate in fragment-based screening,7 whether the resulting fragment hits will have sufficiently robust binding modes to serve as good starting points for lead discovery depends additionally on how compact or diffuse the main hot spot is and how fully the fragment occupies it.138 This topic is beyond the scope of the current article, however, where we are concerned with whether high affinity binding can occur, rather than with differences between druggable sites in the technical ease or difficulty in identifying a good ligand.
Cell division protein ZipA
The ZipA/FtsZ protein-protein interaction has been considered as a potential target for antibacterial agents.121 We have mapped three different structures of ZipA: unliganded, co-crystallized with a peptide fragment of FtsZ,139 and co-crystallized with the indoloquinolizin inhibitor DB01967 (1), shown in Figure 3A.140 In all three structures FTMap finds only three weak hot spots at the protein-peptide interface (Table 4 and Figure 3C). Although ZipA has strong hot spots elsewhere, these are at least 11 Å distant from the interface. Thus, we conclude that the interface between ZipA and FtsZ is not a druggable target. In fact, the KD of the interaction between ZipA and a 17-mer peptide of FtsZ has been measured to be ~ 7 μM,141 and efforts to identify and optimize small molecule PPI inhibitors for this system, including HTS of 250,000 compounds, resulted only in inhibitors with KD ≥ 12 μM.140,142–144
Figure 3.
Druggability assessment for important targets. (A) Structure of the weak inhibitor 1 of the ZipA/FtsZ protein-protein interaction. (B) Structure of the PTP1B inhibitor 2. (C) Mapping of the ligand free ZipA protein (PDB ID 1f46), with inhibitor 1 (from PDB ID 1s1s) superimposed for reference. The consensus clusters shown are CC8 (green, 6 probe clusters), CC9 (blue, 5 clusters), and CC10 (orange, 3 clusters). We also show the strongest hot spot CC1 (cyan, 22 clusters), which is located outside the ZipA/FtsZ interface. CC4 (salmon, 9 clusters) is also too far from the interface. (D) Mapping of the apurinic/apyrimidinic endonuclease 1 (APE1) structure from PDB ID 1de9, co-crystallized with abasic DNA (fragment of the DNA is shown as blue sticks for reference), and an Mn2+ ion (cyan sphere). As always, the DNA and the ion were removed prior to the mapping. The hot spots shown are CC1 (cyan, 16 clusters), CC2 (magenta, 16 clusters), and CC3 (yellow, 13 clusters). (E) Mapping of the ligand free protein tyrosine phosphatase 1B (PTP1B, PDB ID 3a5j), with two bound phosphotyrosine (pTyr) molecules superimposed for reference (from PDB ID 1pty). The hot spots shown are CC1 (cyan, 20 clusters) at the catalytic PTyr site, CC4 (salmon, 9 clusters), CC7 (orange, 7 clusters), and CC8 (green, 7 clusters) close to the site that binds the non-catalytic pTyr. The hot spots CC2 (magenta, 16 clusters) and CC7 (orange, 7 clusters) are closed in any ligand-bound structure. (F) Same as Figure 3E, but with the bound structure of the potent inhibitor 2 shown for reference from the structure with PDB ID 1q6t.
Table 4.
Detailed analysis of important targets
| Target | Site(s) | PDB | Consensus sites | D1 | Comment | ||
|---|---|---|---|---|---|---|---|
|
| |||||||
| U | B | ||||||
| 1 | ZipA | FtsZ peptide binding site | 1f46 | 8(6), 9(5), 11(3) | N | All hot spots in the ZipA:FtsZ interface are very weak. There is a strong hot spots, 1(22) and 2(16), but are more than 10 Å from any hot spot in the interface. | |
| 1f47 | 5(13), 6(6), 7(3) | ||||||
| 1s1s | 4(10), 8(4), 10(2) | ||||||
| 2 | APE-1 | A: PO3 of abasic nucleotide B: sugar of abasic nucleotide U: PO3 of two nucl. upstream Y: at Y128 |
1bix | A: 1(18), B: 2(16); U: 5(10) | D | For the ensemble formed by CC1 and CC2, MD=11.6Å, druggable but weak; CC5 from CC2 8 Å, CC5 is closed in DNA bound structures. | |
| 1e9n | A: 4(15), B: 2(17), U: 6(4) | ||||||
| 1de9 | A: 1(16), B: 2(16), Y: 3(13) | With bound DNA and Mn2+; CC3 is close to CC1, not present in the DNA-free structures, MD=14.5 Å. | |||||
| 1dew | A: 3(14), B: 1(22), Y: 7(5) | With bound DNA, no Mn2+; MD=14.4 Å. | |||||
| 3 | PTP1B | A site: catalytic pTyr | 3a5j | A: 1(20), 2(16); B: 4(9), 7(7), 8(7) | D | A and B sites are too far (9.0 Å), but A is druggable on its own. | |
| 1pa1 | A: 1(21), 2(16); B: 8(4), 10(4) | ||||||
| B site: non-catalytic pTyr | 1pty | A: 1(13), 7(7); B: 8(7), 9(7) | Co-crystallized with 2 pTyr amino acids; A site contracts upon pTtyr binding due to the move of loop 180–184. | ||||
| C site: Tyr46, Arg47 & Asp48 | 2veu | A: 1(16), 10(3); B: 3(12), 6(7), 9(4) | IC50=100 nM, not selective; inhibitor binds only at A &C; A to B=9.1 Å is too far; MW=604.6. | ||||
| 4i8n | A: 1(26); B: 3(10), 8(6) | Binds at A & and the weaker hot spot of B, not selective; A to B= 6.4 Å, MW=483.5. | |||||
| 2vew | A: 1(25); B: 4(9), 6(7) | IC50=64 nM, binds A & the weaker hot spot of B & part of C, A to B= 6.6 Å, MW=584.6. | |||||
| 2fjn | A: 1(15), 6(9); B: 5(11), 7(7) | IC50=39 nM; A & one hot spot of B & part of C, A to B= 7.0 Å; MW=603.6. | |||||
| 2qbp | A: 1(18), 5(8); B: 4(10), 9(5) | Ki=4 nM, Both A & B; both hot spots in B are used, A to B= 7.7 Å; MW=609.5 | |||||
| 1q6t | A: 1(20), 10(3); B: 5(11), 9(5) | IC50=7 nM; pTyr analog, both A & B & some C; A to B= 7.7 Å; MW=826.8. | |||||
| 1pxh | A: 1(20), 9(5); B: 5(7), 7(7) | Ki=1.4 nM; binds at A & weaker hot spot of B & C, A to B= 6.6 Å; MW=873.2. | |||||
| 4 | 14-3-3 protein | S1: pS/T binding S2: pS/T-1 S3: pS/T+1 S4: fusicoccin S5: pS/T-X-COOH |
1yz5 | S1: 1(21); S2: 4(12); S3: 7(5); S4: 2(16), 5(11) | D*L | CC1 to CC4 8.2 Å; all other hot spots are even further: CC1 to CC2 10.7 Å, CC1 to CC5 9.7 Å; | |
| 3p1n | S2: 2(14); S3: 1(29); S4: 3(13); S5; 5(10) | Bound phosphate group moves the side chain of Arg129 and closes down the pS/T pocket; S3 is strong. | |||||
| 4dhu | S2: 4(13), 8(3); S3: 5(11); S4: 1(18), 3(160, 7(5) | The small inhibitor places its phosphate group into the pS/T pocket (S1 site), which becomes closed. | |||||
| 5 | KRAS | G: GTP/GDP S1: Site 1 S2: Site 2 (SOS interface) S3: Site 3 S4: Site 4 S5: Site 5 S6: not in Raf or SOS interface |
3gft | G: 1(24), 7(6), 9(5); S2: 2(13); S3: 4(9), 7(6); S5: 6(7), 8(6) | B*S | Inhibitor binding site S2 is a weak and isolated hot spot; MD=5.0 Å. | |
| 2pmx | G 1(25), 3(11), 7(5); S2: 2(12), S3: 8(5); S5: 6(9) | S2 is slightly weaker, but still there. | |||||
| 4epr | G: 1(26), 4(10), 5(9); S3: 7(6); S5: 3(11) | S2 does not include any hot spot. | |||||
| 4dst | G: 1(19), 5(6), 8(3); S2: 3(17), S3: 6(5), 7(4); S5: 2(19), | S2 stronger, but still isolated, MD=6.7Å. | |||||
| 4epw | G: 2(17), 4(12); S1: 7(5), S2: 1(21), 8(3); S5: 3(14), 5(9) | S2 becomes the strongest hot spot region, MD=10.9 Å but the secondary hot spot is weak, KD=420 μM. | |||||
| 4epy | G: 2(16), 3(12), 4(10), 5(8), S2: 1(20), 8(5); S3: 10(3); S5: 6(7), 9(4) | There are no hot spots for further extension. Adding different groups to the end of the inhibitor, the best KD value achieved is 190 μM. | |||||
| Mapping with masked GTP site, i.e., F site is not available to probes | 3gft | S2: 4(14), S6: 1(19), S3: 7(5), S5: 2(18) | Strongest site is the one identified by MSCS; S2 site is essentially invariant, remains weak. | ||||
| 4dst | S2: 2(17), S3: 5(11), 6(9); S5: 1(23), | Almost no change relative to free GTP site. | |||||
| 4epw | S2: 1(17), 4(13); S3: 3(14), S5: 2(15) | The secondary site in S2 appears to be stronger, but S2 is still only borderline druggable. | |||||
| 4epy | S2: 1(16), 3(14); S3: 2(16), 7(9), S5: 4(11) | The same as 4epw; MD=11.4 Å. | |||||
| 6 | HIV Integrase | LEDGF/p75 site | 1bi4 | 1(18), 5(9) | D*S | Ligand binding site is a deep pocket, hot spot at the bottom; rather small hot spot ensemble, MD=9.6 Å; Predicted to require charged ligand for potency. | |
| 3avn | 1(20) | Bound to LEDGF peptide; elongated hot spot, but still only MD=9.2 Å. | |||||
| 2b4j | 1(19) | Bound to LEDGF, MD=8.9 Å. | |||||
| 3lpu | 3(8), 9(3) | Inhibitor bound, MD=9.5 Å; related inhibitor with EC50= 69 nM, all ligands are carboxylic acid derivatives. | |||||
Druggability class based on FTMap analysis, as described in Table 1.
Apurinic/apyrimidinic (AP) endonuclease 1 (APE1)
APE1 is an essential protein that operates in the base excision repair (BER) pathway and is responsible for ≥ 95% of the total AP endonuclease activity in human cells. BER is a major pathway that copes with DNA damage induced by several cancer causing agents, including ionizing radiation and temozolomide. Overexpression of APE1 and enhanced AP endonuclease activity have been linked to increased resistance of tumor cells to treatment with monofunctional alkylators, implicating inhibition of APE1 as a valid strategy for cancer therapy.145 We analyzed two different unbound structures of APE1 by FTMap (Table 4). The results show two main hot spots at the locations that bind the phosphate group (site A) of the abasic nucleotide, close to the divalent metal ion in the active site, and the sugar ring (site B) of the same nucleotide (Table 4). In ligand-free structures there is also a weak hot spot (site U) at the phosphate group of the nucleotide two positions upstream from the abasic group, but the site is far (8 Å) from site B, and closes down upon DNA binding. The mapping of DNA bound structures reveals another hot spot (site Y), formed by turning the side chain of Tyr128 out of the active site. This site is somewhat stronger when a bound Mn2+ ion is present (Figure 3D). However, the two main hot spots remain weak, and although utilizing the latter hot spot may provide additional potency, there is nothing else near the small active site.
Thus, we consider the active site of APE-1 to be druggable, but very weak, close to borderline druggability. In spite of recent hopes,146 information available in the literature fully confirms this prediction. As described in an excellent review,147 the reproducibility and biological value of a few potent compounds reported in early studies remain questionable, and therefore APE1 was targeted in a number of more recent screens, which collectively returned over 50 compounds. However, even the best inhibitors had only low micromolar IC50 values. Almost all compounds share a relatively small size, an aromatic hydrophobic central core that most likely binds at site B, and a negative ionizable group, frequently a carboxylate, which is thus predicted to bind at site A and to interact with the metal. Based on the weak druggability of the active site we believe it will be difficult to get much higher potency, and APE1 selectivity may also become an issue.147 However, it is possible that larger compounds, primarily macrocycles, will be able to take advantage of the additional U and Y sites, improving both potency and selectivity.
Protein tyrosine phosphatase 1B (PTP1B)
PTP1B is a highly validated drug target, but with a very long history of failures to identify potent and selective inhibitors. The enzyme acts as a negative regulator in insulin signaling pathways. Inhibition of PTP1B activity exhibits potential for enhancing insulin action by prolonging the phosphorylated state of the insulin receptor.148 A large variety of potent PTP1B inhibitors have been developed such as 2 shown in Figure 3B with IC50 =7nM,149 and it is known that the primary challenges are cell permeability and selectivity.150 The challenge of permeability is a consequence of the highly polar nature of the active site. Achieving selectivity is difficult because PTP1B and the most highly related tyrosine phosphatase, T-cell PTP (TCPTP), share 72% sequence identity overall and 94% identity for the active site residues. We analyzed two different unbound PTP1B structures by FTMap (Table 4). The results show that top two hot spots in the ligand-free PTP1B structures are located at the catalytic site (A site), with additional, weaker hot spots at a second, non-catalytic site for phosphotyrosine binding (B site) (Table 4 and Figure 3E). We agree with Hajduk et al.7 that the A site is druggable on its own, i.e., it binds potent (albeit charged) ligands. As seen for other ligands with a phosphate group, the structure of PTP1B in complex with phosphotyrosines (PDB entry 1pty) shows that the binding changes the conformation of the loop comprising residues 180–184, which tightens the pocket, but the site can open up to accommodate larger inhibitors. All inhibitors must interact with the charged A site because it is the main source of binding affinity. Attempts have been made to replace the phosphate group with 1,1-dioxido-3-oxoisothiazolidin in site A and expand the inhibitor toward residues Tyr46, Arg47, and Asp 48, defined as the C site, without any strong hot spots. Such inhibitors (e.g., PDB entry 2veu) have IC50 values between 100 and 2900 nM, but MW >600 g/mol.151 Alternatively, a number of inhibitors have been developed that have the phosphate group in site A, but expand into the less polar B site, thereby improving both affinity and cell permeability. To reach even the weaker hot spot at the B site, around 7 Å away from the A site, requires a molecule with MW >500 g/mol. In addition, the residues that differ between PTP1B and TCPTP (Phe52Tyr and Ala27Ser) lie at the distal end of the B site. Thus, in order to improve selectivity, inhibitors first have to bridge from the A site to the first hot spot of the B site (7.7 Å), and then continue to the second, stronger hot spot of the latter site (another 6 Å). Adding interactions with the C site further improves binding, and such compounds reach an IC50 of 5 nM and seven-fold selectivity over TCPTP.149 However, inhibitor 2 of this type from PDB entry 1q6,149 shown in Figure 3B, still has the charged phosphate group and the high MW of 826.76 g/mol. Thus, we conclude that PTP1B is formally druggable, in the sense that very potent inhibitors can be and have been developed. However, the analysis of the hot spot strengths and locations, and the residue differences between PTP1B and its close homolog TCPTP, show that the requirements of high affinity, selectivity, and cell permeability cannot be simultaneously satisfied by an ordinary druglike compound.
14-3-3 protein
The 14-3-3 family of phosphoserine/threonine-recognition proteins engage multiple nodes in signaling networks that control diverse physiological and pathophysiological functions and have emerged as promising therapeutic targets for such diseases as cancer and neurodegenerative disorders.152 14-3-3 proteins have been co-crystallized with several phosphoserine or phosphothreonine (pS/T) containing peptides.153 Figure 4A shows such a phosphopeptide, ERα 591–595 (3), with phosphothreonine at position 594, and Figure 4B shows the compound named fusicoccin (4), originally identified as a fungal toxin, that stabilizes the binding of 3. 154 The 14-3-3 protein forms a weak homodimer, but each monomer can bind 3 independently in a well-defined elongated site with a deep pS/T binding cavity, together with 4 (Figure 4D). The protein has been recently co-crystallized with the inhibitor PubChem17588196 (5), shown in Figure 4C.155 Figure 4E shows the resulting complex.155 Although the IC50 value of 5 is only 5 μM, it was shown to disrupt the interaction of 14-3-3 with aminopeptidase N in a cellular assay.155
Figure 4.
Druggability assessment for 14-3-3 protein. (A) Structure of the phosphothreonine peptide 3 that was co-cystallyzed with 14-3-3. (B) Structure of fusicoccin (4), a compound that binds to 14-3-3 and stabilizes the binding of 3. (C) Structure of inhibitor 5. (D) Mapping of the ligand free structure of 14-3-3 (PDB ID 1yz5). For reference we superimpose the bound compounds 3 and 4, shown as green and yellow sticks, respectively, from PDB ID 4jdd. The hot spots shown are CC1 (cyan, 21 clusters) at the deep pocket that binds 3, CC2 (magenta, 16 clusters) and CC5 (white, 11 clusters) at the region that binds 4. Two more hot spots are close to 3: CC4 (salmon, 12 clusters), and CC7 (orange, 5 clusters). (E) Same as Figure 4D, but with inhibitor 5 (magenta).
As shown in Table 4 and Figure 4D, the strongest hot spot of the ligand-free 14-3-3 protein is in the pS/T binding pocket, with additional hot spots, both at the side chains adjacent to the bound 3, and in other regions in the binding site. The binding of the phosphate is clearly an important part of the interactions with peptides and small inhibitors. However, the binding of the phosphate group, both in the peptides and the inhibitors, attracts the side chain of Arg129 into the pS/T binding pocket, eliminating the hot spot at that location (Table 4). This example emphasizes that the analysis of ligand-free protein structure, which here predicts the site to be druggable, provides useful druggability information, whereas restricting consideration to pocket properties in ligand-bound structures can be misleading when the protein binds charged ligands. Indeed, the inhibitors are located at the strongest hot spot, CC1, of the unliganded protein, rather than those present in the ligand-bound structure (Figure 4E). The hot spot closest to CC1 is CC5, but even that distance is 8.2 Å; all other hot spots are even further, and are not used by the small inhibitor. However, the S2 and S3 sites at the pS/T-1 and pS/T+1 positions, respectively, remain relatively strong in all structures, suggesting that higher potency but larger inhibitors can most likely be found.
KRAS
The Ras (rat sarcoma) protein family members are all low molecular weight GTP-binding proteins that play a role in regulating cell differentiation, proliferation, and survival. KRAS, a key member of the Ras family, is an attractive cancer target, as frequent point mutations in the KRAS gene render the protein constitutively active.156 However, even though the structure and function of RAS have been studied intensively for nearly 30 years, it has remained elusive as a drug target.40 The most obvious target site would be the deep pocket where GTP and GDP bind, but these nucleotides bind with nanomolar affinity,157 and are present at very high concentrations in cells. Therefore we mapped the protein both with the GTP/GDP site free, and also with the site masked to prevent probe binding (Table 4). There have been many attempts to find alternative sites to target, with recent successes by two groups, who reported small molecules binding to a pocket in the surface of KRAS that interacts with the SOS protein 136,158, referred to as Site 2 159. Studying KRAS is complicated by the fact that the protein acts as a molecular switch with complex dynamics between multiple conformational states.40 We analyzed three different unbound structures by FTMap. As shown in Table 4, in mapping KRAS the GTP/GDP site dominates, if the site is not masked, with several strong hot spots at this location. In two of the three unbound KRAS structures Site 2 in the KRAS/SOS interface had the second strongest hot spots. However, the hot spot has at most 13 probe clusters and has a small size (MD=5.0 Å), and hence based on the FTMap criteria, this site is classified as B*S. Nevertheless, the KRAS/SOS interface has been targeted in recent campaigns, and two groups independently identified series of druglike inhibitors binding at Site 2,136,158 such as CHEMBL2086796 (6)136 shown in Figure 5A. To show the hot spot in the unliganded KRAS structure we superimpose a small ligand, benzamidine (Figure 5C). As the size of ligands increases, the number of probes in the main hot spot increases to 20, and second hot spot also opens in the structure co-crystallized with 6, but it is very weak and includes at most 5 probe clusters, (Figure 5D). In addition, the maximum dimension of the hot spot region increases to 10.9 Å, and based on these values the site would be considered druggable. However, as our analyses of other targets shows, the mapping of the ligand-free structure usually provides more valid assessment of druggability that the mapping of the ligand-bound state, and based on the former the interface is only borderline druggable (B*S). This prediction is supported by the fact that, in spite of major efforts, the best-known non- covalent inhibitor binding at this site has a KD value of 190 μM, and is positively charged at neutral pH.136 The hot spots are even weaker if the GTP/GDP site is masked (Table 4), so the weakness of the hot spots at Site 2 is not due to the binding of probes elsewhere.
Figure 5.
Druggability assessment for KRAS. (A) Structure of 6, one of the highest affinity inhibitors binding to KRAS at the interface with SOS. (B) Structure of the inhibitor 7 binding covalently to KRAS(G12C). (C) Mapping of the ligand-free KRAS (PDB ID 3gft). The cavities with probes bound are the GTP/GDP binding site, with part of CC1 (cyan, 24 clusters) visible, and the targeted hot spot CC2 (magenta, 13 clusters) in Site 2, located in the KRAS/SOS interface. For reference we superimpose a small ligand, benzamidine (shown as yellow sticks, from the structure with PDB ID 4dso), binding to Site 2. (D) Mapping of the KRAS structure from its complex with inhibitor 6 from PDB ID 4epy (KD=340 μM, superimposed as magenta sticks). Due to the binding of 6, the strongest hot spot CC1 (cyan, 20 clusters) is now in the KRAS/SOS interface, and the interface includes a second weak hot spot CC8 (green, 7 clusters) located close to CC1. (E) The mapping of the ligand-free KRAS (PDB ID 3gft) places CC4 (salmon, 9 clusters) at the S3 site, in the pocket binding the KRAS(G12C) inhibitor 7 that allosterically controls GTP affinity and effector interactions. Inhibitor 7 from PDB ID 4lv6 is superimposed for reference, and is shown as green sticks. Notice that in this closed conformation of KRAS, the loop 58 to 65 would clash with 7, and thus prevents its binding. For reference the figure also shows CC2 (magenta) and the benzamidine ligand (yellow) on the other side of the protein. (F) Homology model of KRAS based on a HRAS structure (PDB ID 4q21) to model the open state. In this structure the opening of the 58–65 loop creates a stronger hot spot, CC2 (magenta, 16 clusters). Inhibitor 7 from PDB ID 4lv6 is shown again for reference. Although the site in this open structure binds 16 probe clusters, the maximum dimension of CC2 is only 5.1 Å, thus the site is not druggable without the covalent interaction with G12C.
We note that the mapping of the ligand-free KRAS structure places CC4 (salmon, 9 clusters) at the S3 site, in the pocket that binds the recently developed covalent KRAS(G12C) inhibitors such as SCHEMBL15337743 (7), shown in Figure 5B.160 In this closed conformation of KRAS, the loop 58 to 65 would clash with the allosteric inhibitor 7, and thus prevents binding (Figure 5E). In spite of this unfavorable geometry, FTMap places a hot spot at the allosteric site. To show that the opening of the site would allow for the binding of 7, we constructed a homology model of the KRAS open state based on an open structure of HRAS. Figure 5F shows the mapping of the resulting structure with 7 superimposed. As expected, the opening of the 58–65 loop creates a stronger hot spot, CC2 (magenta, 16 clusters). However, the maximum dimension of this CC2 is only 5.1 A, and thus the site is not druggable without covalent protein-ligand interaction.160
HIV integrase, LEDGF/p75 interaction site
Integration of HIV-1 DNA is required to maintain the viral DNA in the infected cell, and hence the integrase (IN) is an attractive target for HIV treatment. Two different mechanisms have been explored for IN inhibition. The design and discovery of IN inhibitors were first focused at targeting the catalytic site of IN, with the goal of achieving an effect on strand transfer.161 Cheng et al.8 considered this site to be undruggable, whereas based on FTMap we predicted it to be druggable (Table S3). In fact, since the publication of the work by Cheng et al.,8 several compounds targeting the active site, called integrase strand transfer inhibitors (INSTIs), have been approved.162 Since INSTI resistance evolves readily, a proposed alternative approach is to disrupt the interaction between IN and the transcriptional coactivator protein LEDGF/p75, which results in inhibition through an allosteric mechanism.161,162 According to FTMap, the interface region of IN has a strong hot spot, but the hot spot ensemble is small (MD=9.6 Å), and thus we classify the site as Non-Canonically Druggable – Small (D*S), requiring charged inhibitors. In agreement with this prediction, the recently discovered potent inhibitors called LEDGINs include a carboxyl group.163 An example of this class is (2S)-2-(6-chloro-2-methyl-4-phenylquinolin-3-yl)pentanoic acid (PubChem45281242),161 which binds to IN with EC50 = 69 nM. Note that the binding of the charged ligands reduces the hot spot, similarly to the 14-3-3 protein, again emphasizing that the ligand-free structure is likely to provide more accurate druggability prediction.
CONCLUSIONS
We show here that the druggability of binding sites on proteins can be evaluated with good reliability based on fragment hit rates, whether obtained experimentally (e.g., by NMR) or computationally using the FTMap algorithm, and establish clear FTMap-based criteria for druggability. Importantly, because the FTMap method is based on the biophysics of binding rather than on empirical parameterization to a particular training set of protein-drug complexes, meaningful information can be gained about classes of proteins and classes of compounds beyond those resembling validated targets and conventionally druglike ligands. In particular, the information provided by the mapping identifies targets that, while not druggable by druglike ligands, may become druggable using pharmaceutically attractive compound classes such as macrocycles or other large molecules that fall outside conventional definitions of druglikeness such as the Rule-of-Five. Application of the method to ZipA and PTP-1B show why these targets are not druggable. In contrast, results for other targets of high current biomedical interest, including apurinic/apyrimidinic endonuclease 1 (APE1), the SOS interaction site of KRAS, the phosphoserine/threonine-recognition protein 14-3-3, and the LEDGF/p75 interaction site of HIV integrase, show that in these cases it may be necessary to consider ligands lying outside conventional druglike chemical space, and provide guidance as to what compound classes are most likely to succeed. Overall, due to the progress that has been made in recent years across multiple research groups and institutions, we believe that evaluation of protein druggability has entered an era in which, for the majority of cases, the prospects for identifying a pharmaceutically useful small molecule ligand can now be ascertained with good confidence based only on the X-ray crystal structure of the unliganded protein. These advances substantially lower the technical risk associated with launching a drug discovery project involving a novel protein target, and provide valuable information concerning which approaches and chemotypes are most likely to succeed against a given site.
Supplementary Material
Acknowledgments
This investigation was supported by grants GM064700 to SV and GM094551 to AW and SV from the National Institute of General Medical Sciences.
Abbreviations Used
- PTP-1B
protein tyrosine phosphatase-1B
- APE1
apurinic/apyrimidinic endonuclease 1
- SOS
son of sevenless
- LEDGF/p75
lens epithelium-derived growth factor
- KRAS
Kirsten rat sarcoma viral oncogene homolog
- MSCS
Multiple Solvent Crystal Structures
- CC
consensus cluster
- PDB
Protein Data Bank
- RMSD
root mean square deviation
- CD
center-to-center distance
- MD
maximum distance
- DS
druggability score
Biographies
Dima Kozakov received an M.S. in Applied Mathematics and Physics in Moscow Institute of Physics and Technology, and PhD in Biomedical Engineering at Boston University. Currently he is Assistant Professor of Applied Mathematics & Statistics at Stony Brook University. Dr. Kozakov has been working on the development of algorithms and software for molecular modeling and drug design. He is the primary architect of ClusPro v2 protein docking server, currently the best automatic server according to latest evaluation round in the blind protein docking assessment CAPRI. ClusPro has more than 10,000 users, and run more than 100,000 jobs in the last three years. Dr. Kozakov is also primary developer of FTMap approach, which was used in this work. The FTMap server currently has more than 2000 users.
David R. Hall received a Ph.D. in Biomedical Engineering from Boston University in 2012. Since 2012, he has led Acpharis, a computer-aided drug design company specializing in computational solvent mapping and protein-protein docking. In this role, he has led a long-term collaboration with Biogen on evaluating the binding sites of proteins using computational solvent mapping.
Raeanne Napoleon received a Ph.D. in Theoretical Physical Chemistry from Boston University in 2012. She received her B.S. (cum laude) from the University of the Sciences in Philadelphia, PA, in 2005. She is currently Assistant Professor of Chemistry at Santa Barbara City College, where she focuses on curriculum development in STEM education, with an emphasis on both chemistry and nanoscience disciplines, and promoting women in STEM. She also continues to work on computational simulations of protein systems and interfacial condensed phase simulations.
Christine Yueh received a BS in Chemical Engineering from UC Santa Barbara in 2010. She is currently a PhD candidate in the Biomedical Engineering program at Boston University, where her research involves using computational modeling to study protein-protein and protein-small ligand interactions.
Adrian Whitty received a B.Sc. in Chemistry from King’s College London in 1985, followed by M.S. and Ph.D. degrees in Chemistry from the University of Illinois at Chicago. After completing postdoctoral work with William P. Jencks FRS at Brandeis University, he joined the biopharmaceutical company Biogen, where he worked in the Department of Drug Discovery, rising to the position of Director of Physical Biochemistry. In 2008 he moved to Boston University, where he is an Associate Professor in the Department of Chemistry. His research interests include protein-protein and protein-ligand binding, especially as related to the discovery of small molecule inhibitors of protein-protein interactions, and also the quantitative, mechanistic analysis of growth factor receptor activation and signaling.
Sandor Vajda received an M.S. in Applied Mathematics and Ph.D. in chemistry in Budapest, Hungary. He held research positions in the Department of Engineering, University of Warwick, England, and the Department of Chemistry, Princeton University. He was a faculty member in Hungary and at the Mount Sinai School of Medicine, New York, NY. Currently he is Professor of Biomedical Engineering and Chemistry at Boston University. He also directs the Biomolecular Engineering Research Center at BU. Dr. Vajda has been active in method development for modeling of biological macromolecules, with emphasis on molecular interactions and drug design. In 2004 he and his graduate students founded SolMap Pharmaceuticals, a start-up company focusing on fragment-based drug design. SolMap was acquired by Forma Therapeutics, Cambridge, MA, in 2008.
Footnotes
Supporting Information Available: Additional tables with notes and references. This material is available free of charge via the Internet at http://pubs.acs.org.
Notes
D.R.H. is a full-time employee of Acpharis, Inc. The company offers software similar to the FTMap program that was used in this paper. D.K. and S.V. own Acpharis stock. However, the FTMap software and server are free for use.
References
- 1.Hopkins AL, Groom CR. The druggable genome. Nat Rev Drug Discov. 2002;1:727–30. doi: 10.1038/nrd892. [DOI] [PubMed] [Google Scholar]
- 2.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3–26. doi: 10.1016/s0169-409x(00)00129-0. [DOI] [PubMed] [Google Scholar]
- 3.Russ AP, Lampel S. The druggable genome: An update. Drug Discov Today. 2005;10:1607–10. doi: 10.1016/S1359-6446(05)03666-4. [DOI] [PubMed] [Google Scholar]
- 4.Johns MA, Russ A, Fu HA. Current drug targets and the druggable genome. Chemical Genomics. 2012:320–31. [Google Scholar]
- 5.Sheridan RP, Maiorov VN, Holloway MK, Cornell WD, Gao YD. Drug-like density: A method of quantifying the "bindability" of a protein target based on a very large set of pockets and drug-like ligands from the Protein Data Bank. J Chem Inf Model. 50:2029–40. doi: 10.1021/ci100312t. [DOI] [PubMed] [Google Scholar]
- 6.Hajduk PJ, Huth JR, Tse C. Predicting protein druggability. Drug Discov Today. 2005;10:1675–82. doi: 10.1016/S1359-6446(05)03624-X. [DOI] [PubMed] [Google Scholar]
- 7.Hajduk PJ, Huth JR, Fesik SW. Druggability indices for protein targets derived from NMR-based screening data. J Med Chem. 2005;48:2518–25. doi: 10.1021/jm049131r. [DOI] [PubMed] [Google Scholar]
- 8.Cheng AC, Coleman RG, Smyth KT, Cao Q, Soulard P, Caffrey DR, Salzberg AC, Huang ES. Structure-based maximal affinity model predicts small-molecule druggability. Nat Biotechnol. 2007;25:71–5. doi: 10.1038/nbt1273. [DOI] [PubMed] [Google Scholar]
- 9.Seco J, Luque FJ, Barril X. Binding site detection and druggability index from first principles. J Med Chem. 2009;52:2363–71. doi: 10.1021/jm801385d. [DOI] [PubMed] [Google Scholar]
- 10.Halgren TA. Identifying and characterizing binding sites and assessing druggability. J Chem Inf Model. 2009;49:377–89. doi: 10.1021/ci800324m. [DOI] [PubMed] [Google Scholar]
- 11.Schmidtke P, Barril X. Understanding and predicting druggability. A high-throughput method for detection of drug binding sites. J Med Chem. 2010;53:5858–67. doi: 10.1021/jm100574m. [DOI] [PubMed] [Google Scholar]
- 12.Krasowski A, Muthas D, Sarkar A, Schmitt S, Brenk R. Drugpred: A structure-based approach to predict protein druggability developed using an extensive nonredundant data set. J Chem Inf Model. 2011;51:2829–42. doi: 10.1021/ci200266d. [DOI] [PubMed] [Google Scholar]
- 13.Fauman EB, Rai BK, Huang ES. Structure-based druggability assessment - identifying suitable targets for small molecule therapeutics. Curr Opin Chem Biol. 2011;15:463–8. doi: 10.1016/j.cbpa.2011.05.020. [DOI] [PubMed] [Google Scholar]
- 14.Volkamer A, Kuhn D, Rippmann F, Rarey M. Dogsitescorer: A web server for automatic binding site prediction, analysis and druggability assessment. Bioinformatics. 2012;28:2074–5. doi: 10.1093/bioinformatics/bts310. [DOI] [PubMed] [Google Scholar]
- 15.Nisius B, Sha F, Gohlke H. Structure-based computational analysis of protein binding sites for function and druggability prediction. J Biotechnol. 2012;159:123–34. doi: 10.1016/j.jbiotec.2011.12.005. [DOI] [PubMed] [Google Scholar]
- 16.Perola E, Herman L, Weiss J. Development of a rule-based method for the assessment of protein druggability. J Chem Inf Model. 2012;52:1027–38. doi: 10.1021/ci200613b. [DOI] [PubMed] [Google Scholar]
- 17.Volkamer A, Kuhn D, Grombacher T, Rippmann F, Rarey M. Combining global and local measures for structure-based druggability predictions. J Chem Inf Model. 2012;52:360–72. doi: 10.1021/ci200454v. [DOI] [PubMed] [Google Scholar]
- 18.Driggers EM, Hale SP, Lee J, Terrett NK. The exploration of macrocycles for drug discovery--an underexploited structural class. Nat Rev Drug Discov. 2008;7:608–24. doi: 10.1038/nrd2590. [DOI] [PubMed] [Google Scholar]
- 19.Wessjohann LA, Ruijter E, Garcia-Rivera D, Brandt W. What can a chemist learn from nature's macrocycles?--A brief, conceptual view. Mol Divers. 2005;9:171–86. doi: 10.1007/s11030-005-1314-x. [DOI] [PubMed] [Google Scholar]
- 20.Marsault E, Peterson ML. Macrocycles are great cycles: Applications, opportunities, and challenges of synthetic macrocycles in drug discovery. J Med Chem. 2011;54:1961–2004. doi: 10.1021/jm1012374. [DOI] [PubMed] [Google Scholar]
- 21.Mallinson J, Collins I. Macrocycles in new drug discovery. Fut Med Chem. 2012;4:1409–38. doi: 10.4155/fmc.12.93. [DOI] [PubMed] [Google Scholar]
- 22.Kalgutkar AS, Dalvie DK. Drug discovery for a new generation of covalent drugs. Exp Opin Drug Disc. 2012;7:561–81. doi: 10.1517/17460441.2012.688744. [DOI] [PubMed] [Google Scholar]
- 23.Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10:307–17. doi: 10.1038/nrd3410. [DOI] [PubMed] [Google Scholar]
- 24.Way JC. Covalent modification as a strategy to block protein-protein interactions with small-molecule drugs. Curr Opin Chem Biol. 2000;4:40–6. doi: 10.1016/s1367-5931(99)00049-6. [DOI] [PubMed] [Google Scholar]
- 25.Edwards TA, Wilson AJ. Helix-mediated protein--protein interactions as targets for intervention using foldamers. Amino Acids. 2011;41:743–54. doi: 10.1007/s00726-011-0880-8. [DOI] [PubMed] [Google Scholar]
- 26.Murray JK, Gellman SH. Targeting protein–protein interactions: Lessons from p53/MDM2. Biopolymers. 2007;88:657–86. doi: 10.1002/bip.20741. [DOI] [PubMed] [Google Scholar]
- 27.Raj M, Bullock BN, Arora PS. Plucking the high hanging fruit: A systematic approach for targeting protein-protein interactions. Bioorg Med Chem. 2013;21:4051–7. doi: 10.1016/j.bmc.2012.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Walensky LD, Kung AL, Escher I, Malia TJ, Barbuto S, Wright RD, Wagner G, Verdine GL, Korsmeyer SJ. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science. 2004;305:1466–70. doi: 10.1126/science.1099191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Brown SP, Hajduk PJ. Effects of conformational dynamics on predicted protein druggability. ChemMedChem. 2006;1:70–2. doi: 10.1002/cmdc.200500013. [DOI] [PubMed] [Google Scholar]
- 30.Clackson T, Wells JA. A hot spot of binding energy in a hormone-receptor interface. Science. 1995;267:383–6. doi: 10.1126/science.7529940. [DOI] [PubMed] [Google Scholar]
- 31.DeLano WL. Unraveling hot spots in binding interfaces: Progress and challenges. Curr Opin Struct Biol. 2002;12:14–20. doi: 10.1016/s0959-440x(02)00283-x. [DOI] [PubMed] [Google Scholar]
- 32.Keskin O, Ma BY, Nussinov R. Hot regions in protein-protein interactions: The organization and contribution of structurally conserved hot spot residues. J Mol Biol. 2005;345:1281–94. doi: 10.1016/j.jmb.2004.10.077. [DOI] [PubMed] [Google Scholar]
- 33.Bogan AA, Thorn KS. Anatomy of hot spots in protein interfaces. J Mol Biol. 1998;280:1–9. doi: 10.1006/jmbi.1998.1843. [DOI] [PubMed] [Google Scholar]
- 34.Kortemme T, Baker D. A simple physical model for binding energy hot spots in protein-protein complexes. Proc Natl Acad Sci U S A. 2002;99:14116–21. doi: 10.1073/pnas.202485799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mattos C, Ringe D. Locating and characterizing binding sites on proteins. Nat Biotechnol. 1996;14:595–9. doi: 10.1038/nbt0596-595. [DOI] [PubMed] [Google Scholar]
- 36.Allen KN, Bellamacina CR, Ding XC, Jeffery CJ, Mattos C, Petsko GA, Ringe D. An experimental approach to mapping the binding surfaces of crystalline proteins. J Phys Chem-US. 1996;100:2605–11. [Google Scholar]
- 37.English AC, Done SH, Caves LS, Groom CR, Hubbard RE. Locating interaction sites on proteins: The crystal structure of thermolysin soaked in 2% to 100% isopropanol. Proteins. 1999;37:628–40. [PubMed] [Google Scholar]
- 38.English AC, Groom CR, Hubbard RE. Experimental and computational mapping of the binding surface of a crystalline protein. Prot Eng. 2001;14:47–59. doi: 10.1093/protein/14.1.47. [DOI] [PubMed] [Google Scholar]
- 39.Dechene M, Wink G, Smith M, Swartz P, Mattos C. Multiple solvent crystal structures of ribonuclease A: An assessment of the method. Proteins. 2009;76:861–81. doi: 10.1002/prot.22393. [DOI] [PubMed] [Google Scholar]
- 40.Buhrman G, O'Connor C, Zerbe B, Kearney BM, Napoleon R, Kovrigina EA, Vajda S, Kozakov D, Kovrigin EL, Mattos C. Analysis of binding site hot spots on the surface of Ras GTPase. J Mol Biol. 2011;413:773–89. doi: 10.1016/j.jmb.2011.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Edfeldt FNB, Folmer RHA, Breeze AL. Fragment screening to predict druggability (ligandability) and lead discovery success. Drug Discov Today. 2011;16:284–287. doi: 10.1016/j.drudis.2011.02.002. [DOI] [PubMed] [Google Scholar]
- 42.Barelier S, Krimm I. Ligand specificity, privileged substructures and protein druggability from fragment-based screening. Curr Opin Chem Biol. 2011;15:469–74. doi: 10.1016/j.cbpa.2011.02.020. [DOI] [PubMed] [Google Scholar]
- 43.Dennis S, Kortvelyesi T, Vajda S. Computational mapping identifies the binding sites of organic solvents on proteins. Proc Natl Acad Sci U S A. 2002;99:4290–5. doi: 10.1073/pnas.062398499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Landon MR, Lancia DR, Jr, Yu J, Thiel SC, Vajda S. Identification of hot spots within druggable binding regions by computational solvent mapping of proteins. J Med Chem. 2007;50:1231–40. doi: 10.1021/jm061134b. [DOI] [PubMed] [Google Scholar]
- 45.Brenke R, Kozakov D, Chuang GY, Beglov D, Hall D, Landon MR, Mattos C, Vajda S. Fragment-based identification of druggable 'hot spots' of proteins using Fourier domain correlation techniques. Bioinformatics. 2009;25:621–7. doi: 10.1093/bioinformatics/btp036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Landon MR, Lieberman RL, Hoang QQ, Ju S, Caaveiro JM, Orwig SD, Kozakov D, Brenke R, Chuang GY, Beglov D, Vajda S, Petsko GA, Ringe D. Detection of ligand binding hot spots on protein surfaces via fragment-based methods: Application to DJ-1 and glucocerebrosidase. J Comput Aid Mol Des. 2009;23:491–500. doi: 10.1007/s10822-009-9283-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kozakov D, Chuang GY, Beglov D, Vajda S. Where does amantadine bind to the influenza virus M2 proton channel? Trends Biochem Sci. 2010;35:471–5. doi: 10.1016/j.tibs.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kozakov D, Hall DR, Chuang GY, Cencic R, Brenke R, Grove LE, Beglov D, Pelletier J, Whitty A, Vajda S. Structural conservation of druggable hot spots in protein-protein interfaces. Proc Natl Acad Sci U S A. 2011;108:13528–33. doi: 10.1073/pnas.1101835108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hall DH, Grove LE, Yueh C, Ngan CH, Kozakov D, Vajda S. Robust identification of binding hot spots using continuum electrostatics: Application to hen egg-white lysozyme. J Am Chem Soc. 2011;133:20668–71. doi: 10.1021/ja207914y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hall DR, Ngan CH, Zerbe BS, Kozakov D, Vajda S. Hot spot analysis for driving the development of hits into leads in fragment-based drug discovery. J Chem Inf Model. 2012;52:199–209. doi: 10.1021/ci200468p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Golden MS, Cote SM, Sayeg M, Zerbe BS, Villar EA, Beglov D, Sazinsky SL, Georgiadis RM, Vajda S, Kozakov D, Whitty A. Comprehensive experimental and computational analysis of binding energy hot spots at the NF-κB essential modulator/IKKβ, protein-protein interface. J Am Chem Soc. 2013;135:6242–56. doi: 10.1021/ja400914z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kozakov D, Grove LE, Hall DR, Bohnuud T, Mottarella SE, Luo L, Xia B, Beglov D, Vajda S. The FTMap family of web servers for determining and characterizing ligand-binding hot spots of proteins. Nat Protoc. 2015;10:733–55. doi: 10.1038/nprot.2015.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liepinsh E, Otting G. Organic solvents identify specific ligand binding sites on protein surfaces. Nat Biotechnol. 1997;15:264–8. doi: 10.1038/nbt0397-264. [DOI] [PubMed] [Google Scholar]
- 54.Mattos C, Bellamacina CR, Peisach E, Pereira A, Vitkup D, Petsko GA, Ringe D. Multiple solvent crystal structures: Probing binding sites, plasticity and hydration. J Mol Biol. 2006;357:1471–82. doi: 10.1016/j.jmb.2006.01.039. [DOI] [PubMed] [Google Scholar]
- 55.Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN, Bourne PE. The Protein Data Bank. Nucl Acids Res. 2000;28:235–42. doi: 10.1093/nar/28.1.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Murray CW, Rees DC. The rise of fragment-based drug discovery. Nat Chem. 2009;1:187–192. doi: 10.1038/nchem.217. [DOI] [PubMed] [Google Scholar]
- 57.Chen Y, Shoichet BK. Molecular docking and ligand specificity in fragment-based inhibitor discovery. Nat Chem Biol. 2009;5:358–64. doi: 10.1038/nchembio.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Warr WA. Fragment-based drug discovery. J Comput Aid Mol Des. 2009;23:453–8. doi: 10.1007/s10822-009-9292-1. [DOI] [PubMed] [Google Scholar]
- 59.Congreve M, Chessari G, Tisi D, Woodhead AJ. Recent developments in fragment-based drug discovery. J Med Chem. 2008;51:3661–80. doi: 10.1021/jm8000373. [DOI] [PubMed] [Google Scholar]
- 60.Carr RA, Congreve M, Murray CW, Rees DC. Fragment-based lead discovery: Leads by design. Drug Discov Today. 2005;10:987–92. doi: 10.1016/S1359-6446(05)03511-7. [DOI] [PubMed] [Google Scholar]
- 61.Villar EA, Beglov D, Chennamadhavuni S, Porco JA, Kozakov D, Vajda S, Whitty A. How proteins bind macrocycles. Nat Chem Biol. 2014;10:723–31. doi: 10.1038/nchembio.1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ganesan A. The impact of natural products upon modern drug discovery. Curr Opin Chem Biol. 2008;12:306–17. doi: 10.1016/j.cbpa.2008.03.016. [DOI] [PubMed] [Google Scholar]
- 63.Goodman CM, Choi S, Shandler S, DeGrado WF. Foldamers as versatile frameworks for the design and evolution of function. Nat Chem Biol. 2007;3:252–262. doi: 10.1038/nchembio876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zuckermann RN, Kodadek T. Peptoids as potential therapeutics. Curr Opin Mol Ther. 2009;11:299–307. [PubMed] [Google Scholar]
- 65.Dietrich U, Durr R, Koch J. Peptides as drugs: From screening to application. Curr Pharm Biotechnol. 2013;14:501–12. doi: 10.2174/13892010113149990205. [DOI] [PubMed] [Google Scholar]
- 66.Wilson KP, Yamashita MM, Sintchak MD, Rotstein SH, Murcko MA, Boger J, Thomson JA, Fitzgibbon MJ, Black JR, Navia MA. Comparative x-ray structures of the major binding-protein for the immunosuppressant FK506 (tacrolimus) in unliganded form and in complex with FK506 and rapamycin. Acta Crystallogr D. 1995;51:511–21. doi: 10.1107/S0907444994014514. [DOI] [PubMed] [Google Scholar]
- 67.Sattler M, Liang H, Nettesheim D, Meadows RP, Harlan JE, Eberstadt M, Yoon HS, Shuker SB, Chang BS, Minn AJ, Thompson CB, Fesik SW. Structure of Bcl- (L)-BAK peptide complex: Recognition between regulators of apoptosis. Science. 1997;275:983–6. doi: 10.1126/science.275.5302.983. [DOI] [PubMed] [Google Scholar]
- 68.Holt DA, Luengo JI, Yamashita DS, Oh HJ, Konialian AL, Yen HK, Rozamus LW, Brandt M, Bossard MJ, Levy MA, Eggleston DS, Liang J, Schultz LW, Stout TJ, Clardy J. Design, synthesis, and kinetic evaluation of high-affinity FKBP ligands and the X-ray crystal-structures of their complexes with FKBP12. J Am Chem Soc. 1993;115:9925–38. [Google Scholar]
- 69.Sich C, Improta S, Cowley DJ, Guenet C, Merly JP, Teufel M, Saudek V. Solution structure of a neurotrophic ligand bound to FKBP12 and its effects on protein dynamics. Eur J Biochem. 2000;267:5342–54. doi: 10.1046/j.1432-1327.2000.01551.x. [DOI] [PubMed] [Google Scholar]
- 70.Silberstein M, Dennis S, Brown L, Kortvelyesi T, Clodfelter K, Vajda S. Identification of substrate binding sites in enzymes by computational solvent mapping. J Mol Biol. 2003;332:1095–113. doi: 10.1016/j.jmb.2003.08.019. [DOI] [PubMed] [Google Scholar]
- 71.Bohnuud T, Kozakov D, Vajda S. Evidence of conformational selection driving the formation of ligand binding sites in protein-protein interfaces. PLoS Comput Biol. 2014;10:e1003872. doi: 10.1371/journal.pcbi.1003872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wang R, Fang X, Lu Y, Wang S. The PDBbind database: Collection of binding affinities for protein-ligand complexes with known three-dimensional structures. J Med Chem. 2004;47:2977–80. doi: 10.1021/jm030580l. [DOI] [PubMed] [Google Scholar]
- 73.Liu T, Lin Y, Wen X, Jorissen RN, Gilson MK, Binding DB. A web-accessible database of experimentally determined protein-ligand binding affinities. Nucl Acids Res. 2007;35:D198–201. doi: 10.1093/nar/gkl999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Benson ML, Smith RD, Khazanov NA, Dimcheff B, Beaver J, Dresslar P, Nerothin J, Carlson HA. Binding MOAD, a high-quality protein-ligand database. Nucl Acids Res. 2008;36:D674–8. doi: 10.1093/nar/gkm911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Topalis D, Kumamoto H, Velasco MFA, Dugue L, Haouz A, Alexandre JAC, Gallois-Montbrun S, Alzari PM, Pochet S, Agrofoglio LA, Deville-Bonne D. Nucleotide binding to human UMP-CMP kinase using fluorescent derivatives - a screening based on affinity for the UMP-CMP binding site. FEBS J. 2007;274:3704–14. doi: 10.1111/j.1742-4658.2007.05902.x. [DOI] [PubMed] [Google Scholar]
- 76.Mashhoon N, Pruss C, Carroll M, Johnson PH, Reich NO. Selective inhibitors of bacterial DNA adenine methyltransferases. J Biomol Screen. 2006;11:497–510. doi: 10.1177/1087057106287933. [DOI] [PubMed] [Google Scholar]
- 77.Miao BC, Skidan I, Yang JS, Lugovskoy A, Reibarkh M, Long K, Brazell T, Durugkar KA, Maki J, Ramana CV, Schaffhausen B, Wagner G, Torchilin V, Yuan JY, Degterev A. Small molecule inhibition of phosphatidylinositol-3,4,5-triphosphate (pip3) binding to pleckstrin homology domains. Proc Natl Acad Sci U S A. 2010;107:20126–31. doi: 10.1073/pnas.1004522107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lerner CG, Hajduk PJ, Wagner R, Wagenaar FL, Woodall C, Gu YG, Searle XB, Florjancic AS, Zhang T, Clark RF, Cooper CS, Mack JC, Yu L, Cai M, Betz SF, Chovan LE, McCall JO, Black-Schaefer CL, Kakavas SJ, Schurdak ME, Comess KM, Walter KA, Edalji R, Dorwin SA, Smith RA, Hebert EJ, Harlan JE, Metzger RE, Merta PJ, Baranowski JL, Coen ML, Thornewell SJ, Shivakumar AG, Saiki AY, Soni N, Bui M, Balli DJ, Sanders WJ, Nilius AM, Holzman TF, Fesik SW, Beutel BA. From bacterial genomes to novel antibacterial agents: Discovery, characterization, and antibacterial activity of compounds that bind to HI0065 (YjeE) from haemophilus influenzae. Chem Biol Drug Des. 2007;69:395–404. doi: 10.1111/j.1747-0285.2007.00521.x. [DOI] [PubMed] [Google Scholar]
- 79.Piserchio A, Salinas GD, Li T, Marshall J, Spaller MR, Mierke DF. Targeting specific PDZ domains of PSD-95: Structural basis for enhanced affinity and enzymatic stability of a cyclic peptide. Chem Biol. 2004;11:469–73. doi: 10.1016/j.chembiol.2004.03.013. [DOI] [PubMed] [Google Scholar]
- 80.Wintjens R, Wieruszeski JM, Drobecq H, Rousselot-Pailley P, Buee L, Lippens G, Landrieu I. 1H NMR study on the binding of Pin1 Trp-Trp domain with phosphothreonine peptides. J Biol Chem. 2001;276:25150–6. doi: 10.1074/jbc.M010327200. [DOI] [PubMed] [Google Scholar]
- 81.Guo CX, Hou XJ, Dong LM, Dagostino E, Greasley S, Ferre R, Marakovits J, Johnson MC, Matthews D, Mroczkowski B, Parge H, VanArsdale T, Popoff I, Piraino J, Margosiak S, Thomson J, Los G, Murray BW. Structure-based design of novel human Pin1 inhibitors (I) Bioorg Med Chem Lett. 2009;19:5613–6. doi: 10.1016/j.bmcl.2009.08.034. [DOI] [PubMed] [Google Scholar]
- 82.Dong LM, Marakovits J, Hou XJ, Guo CX, Greasley S, Dagostino E, Ferre R, Johnson MC, Kraynov E, Thomson J, Pathak V, Murray BW. Structure-based design of novel human Pin1 inhibitors (II) Bioorg Med Chem Lett. 2010;20:2210–4. doi: 10.1016/j.bmcl.2010.02.033. [DOI] [PubMed] [Google Scholar]
- 83.Guo CX, Hou XJ, Dong LM, Marakovits J, Greasley S, Dagostino E, Ferre R, Johnson MC, Humphries PS, Li HT, Paderes GD, Piraino J, Kraynov E, Murray BW. Structure-based design of novel human Pin1 inhibitors (III): Optimizing affinity beyond the phosphate recognition pocket. Bioorg Med Chem Lett. 2014;24:4187–91. doi: 10.1016/j.bmcl.2014.07.044. [DOI] [PubMed] [Google Scholar]
- 84.Wendt MD, Sun CH, Kunzer A, Sauer D, Sarris K, Hoff E, Yu LP, Nettesheim DG, Chen J, Jin S, Comess KM, Fan YH, Anderson SN, Isaac B, Olejniczak ET, Hajduk PJ, Rosenberg SH, Elmore SW. Discovery of a novel small molecule binding site of human survivin. Bioorg Med Chem Lett. 2007;17:3122–9. doi: 10.1016/j.bmcl.2007.03.042. [DOI] [PubMed] [Google Scholar]
- 85.Huth JR, Yu L, Collins I, Mack J, Mendoza R, Isaac B, Braddock DT, Muchmore SW, Comess KM, Fesik SW, Clore GM, Levens D, Hajduk PJ. NMR-driven discovery of benzoylanthranilic acid inhibitors of far upstream element binding protein binding to the human oncogene c-myc promoter. J Med Chem. 2004;47:4851–7. doi: 10.1021/jm0497803. [DOI] [PubMed] [Google Scholar]
- 86.Hajduk PJ, Dinges J, Miknis GF, Merlock M, Middleton T, Kempf DJ, Egan DA, Walter KA, Robins TS, Shuker SB, Holzman TF, Fesik SW. NMR-based discovery of lead inhibitors that block DNA binding of the human papillomavirus E2 protein. J Med Chem. 1997;40:3144–50. doi: 10.1021/jm9703404. [DOI] [PubMed] [Google Scholar]
- 87.Schaal TD, Mallet WG, McMinn DL, Nguyen NV, Sopko MM, John S, Parekh BS. Inhibition of human papilloma virus E2 DNA binding protein by covalently linked polyamides. Nucl Acids Res. 2003;31:1282–91. doi: 10.1093/nar/gkg206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kurg R, Langel U, Ustav M. Inhibition of the bovine papillomavirus E2 protein activity by peptide nucleic acid. Virus Res. 2000;66:39–50. doi: 10.1016/s0168-1702(99)00124-0. [DOI] [PubMed] [Google Scholar]
- 89.de Dios A, Prieto L, Martin JA, Rubio A, Ezquerra J, Tebbe M, Lopez de Uralde B, Martin J, Sanchez A, LeTourneau DL, McGee JE, Boylan C, Parr TR, Jr, Smith MC. 4-substituted D-glutamic acid analogues: The first potent inhibitors of glutamate racemase (MurI) enzyme with antibacterial activity. J Med Chem. 2002;45:4559–70. doi: 10.1021/jm020901d. [DOI] [PubMed] [Google Scholar]
- 90.Geng B, Breault G, Comita-Prevoir J, Petrichko R, Eyermann C, Lundqvist T, Doig P, Gorseth E, Noonan B. Exploring 9-benzyl purines as inhibitors of glutamate racemase (MurI) gram-positive bacteria. Bioorg Med Chem Lett. 2008;18:4368–4372. doi: 10.1016/j.bmcl.2008.06.068. [DOI] [PubMed] [Google Scholar]
- 91.Breault GA, Comita-Prevoir J, Eyermann CJ, Geng B, Petrichko R, Doig P, Gorseth E, Noonan B. Exploring 8-benzyl pteridine-6,7-diones as inhibitors of glutamate racemase (MurI) in gram-positive bacteria. Bioorg Med Chem Lett. 2008;18:6100–6103. doi: 10.1016/j.bmcl.2008.10.022. [DOI] [PubMed] [Google Scholar]
- 92.McRee DE, Jensen GM, Fitzgerald MM, Siegel HA, Goodin DB. Construction of a bisaquo heme enzyme and binding by exogenous ligands. Proc Natl Acad Sci U S A. 1994;91:12847–51. doi: 10.1073/pnas.91.26.12847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Appleby CA, Wittenbe Ba, Wittenbe Jb. Nicotinic-acid as a ligand affecting leghemoglobin structure and oxygen reactivity. Proc Natl Acad Sci U S A. 1973;70:564–568. doi: 10.1073/pnas.70.2.564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Maldonado S, Lostao A, Irun MP, Fernandez-Recio J, Genzor CG, Gonzalez EB, Rubio JA, Luquita A, Daoudi F, Sancho J. Apoflavodoxin: Structure, stability, and FMN binding. Biochimie. 1998;80:813–20. doi: 10.1016/s0300-9084(00)88876-8. [DOI] [PubMed] [Google Scholar]
- 95.Irie K, Oie K, Nakahara A, Yanai Y, Ohigashi H, Wender PA, Fukuda H, Konishi H, Kikkawa U. Molecular basis for protein kinase c isozyme-selective binding: The synthesis, folding, and phorbol ester binding of the cysteine-rich domains of all protein kinase c isozymes. J Am Chem Soc. 1998;120:9159–67. [Google Scholar]
- 96.Murray IA, Cann PA, Day PJ, Derrick JP, Sutcliffe MJ, Shaw WV, Leslie AGW. Steroid recognition by chloramphenicol acetyltransferase - engineering and structural-analysis of a high-affinity fusidic acid-binding site. J Mol Biol. 1995;254:993–1005. doi: 10.1006/jmbi.1995.0671. [DOI] [PubMed] [Google Scholar]
- 97.Vyas MN, Vyas NK, Quiocho FA. Crystallographic analysis of the epimeric and anomeric specificity of the periplasmic transport chemosensory protein-receptor for D-glucose and D-galactose. Biochemistry. 1994;33:4762–8. doi: 10.1021/bi00182a003. [DOI] [PubMed] [Google Scholar]
- 98.Chung SJ, Fromme JC, Verdine GL. Structure of human cytidine deaminase bound to a potent inhibitor. J Med Chem. 2005;48:658–60. doi: 10.1021/jm0496279. [DOI] [PubMed] [Google Scholar]
- 99.Masaki T, Tanaka T, Tsunasawa S, Sakiyama F, Soejima M. Inhibition of achromobacter protease-I by lysinal derivatives. Biosci Biotech Bioch. 1992;56:1604–7. doi: 10.1271/bbb.56.1604. [DOI] [PubMed] [Google Scholar]
- 100.Lunney EA, Para KS, Rubin JR, Humblet C, Fergus JH, Marks JS, Sawyer TK. Structure-based design of a novel series of nonpeptide ligands that bind to the pp60src SH2 domain. J Am Chem Soc. 1997;119:12471–6. [Google Scholar]
- 101.Fauman EB, Rai BK, Huang ES. Structure-based druggability assessment--identifying suitable targets for small molecule therapeutics. Curr Opin Chem Biol. 2011;15:463–8. doi: 10.1016/j.cbpa.2011.05.020. [DOI] [PubMed] [Google Scholar]
- 102.Schonthal AH. Direct non-cyclooxygenase-2 targets of celecoxib and their potential relevance for cancer therapy. Br J Cancer. 2007;97:1465–8. doi: 10.1038/sj.bjc.6604049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Stangier J, Clemens A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin Appl Thrombosis/Hemostasis. 2009;15(Suppl 1):9S–16S. doi: 10.1177/1076029609343004. [DOI] [PubMed] [Google Scholar]
- 104.Kong Y, Chen H, Wang YQ, Meng L, Wei JF. Direct thrombin inhibitors: Patents 2002–2012 (review) Mol Med Rep. 2014;9:1506–14. doi: 10.3892/mmr.2014.2025. [DOI] [PubMed] [Google Scholar]
- 105.Fu KP, Neu HC. Piperacillin, a new penicillin active against many bacteria resistant to other penicillins. Antimicrob Agents Chemother. 1978;13:358–67. doi: 10.1128/aac.13.3.358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.van Berkel SS, Nettleship JE, Leung IK, Brem J, Choi H, Stuart DI, Claridge TD, McDonough MA, Owens RJ, Ren J, Schofield CJ. Binding of (5s)-penicilloic acid to penicillin binding protein 3. ACS Chem Biol. 2013;8:2112–6. doi: 10.1021/cb400200h. [DOI] [PubMed] [Google Scholar]
- 107.Patchett AA, Harris E, Tristram EW, Wyvratt MJ, Wu MT, Taub D, Peterson ER, Ikeler TJ, ten Broeke J, Payne LG, Ondeyka DL, Thorsett ED, Greenlee WJ, Lohr NS, Hoffsommer RD, Joshua H, Ruyle WV, Rothrock JW, Aster SD, Maycock AL, Robinson FM, Hirschmann R, Sweet CS, Ulm EH, Gross DM, Vassil TC, Stone CA. A new class of angiotensin-converting enzyme inhibitors. Nature. 1980;288:280–3. doi: 10.1038/288280a0. [DOI] [PubMed] [Google Scholar]
- 108.Gubareva LV, Kaiser L, Hayden FG. Influenza virus neuraminidase inhibitors. Lancet. 2000;355:827–35. doi: 10.1016/S0140-6736(99)11433-8. [DOI] [PubMed] [Google Scholar]
- 109.Maignan S, Guilloteau JP, Pouzieux S, Choi-Sledeski YM, Becker MR, Klein SI, Ewing WR, Pauls HW, Spada AP, Mikol V. Crystal structures of human factor Xa complexed with potent inhibitors. J Med Chem. 2000;43:3226–32. doi: 10.1021/jm000940u. [DOI] [PubMed] [Google Scholar]
- 110.Istvan ES, Deisenhofer J. Structural mechanism for statin inhibition of HMG-CoA reductase. Science. 2001;292:1160–4. doi: 10.1126/science.1059344. [DOI] [PubMed] [Google Scholar]
- 111.Barrett DG, Deaton DN, Hassell AM, McFadyen RB, Miller AB, Miller LR, Payne JA, Shewchuk LM, Willard DH, Jr, Wright LL. Acyclic cyanamide-based inhibitors of cathepsin K. Bioorg Med Chem Lett. 2005;15:3039–43. doi: 10.1016/j.bmcl.2005.04.032. [DOI] [PubMed] [Google Scholar]
- 112.Gauthier JY, Chauret N, Cromlish W, Desmarais S, Duong le T, Falgueyret JP, Kimmel DB, Lamontagne S, Leger S, LeRiche T, Li CS, Masse F, McKay DJ, Nicoll-Griffith DA, Oballa RM, Palmer JT, Percival MD, Riendeau D, Robichaud J, Rodan GA, Rodan SB, Seto C, Therien M, Truong VL, Venuti MC, Wesolowski G, Young RN, Zamboni R, Black WC. The discovery of odanacatib (MK-0822), a selective inhibitor of cathepsin K. Bioorg Med Chem Lett. 2008;18:923–8. doi: 10.1016/j.bmcl.2007.12.047. [DOI] [PubMed] [Google Scholar]
- 113.Brixen K, Chapurlat R, Cheung AM, Keaveny TM, Fuerst T, Engelke K, Recker R, Dardzinski B, Verbruggen N, Ather S, Rosenberg E, de Papp AE. Bone density, turnover, and estimated strength in postmenopausal women treated with odanacatib: A randomized trial. J Clin Endocrinol Metab. 2013;98:571–80. doi: 10.1210/jc.2012-2972. [DOI] [PubMed] [Google Scholar]
- 114.Summa V, Petrocchi A, Bonelli F, Crescenzi B, Donghi M, Ferrara M, Fiore F, Gardelli C, Gonzalez Paz O, Hazuda DJ, Jones P, Kinzel O, Laufer R, Monteagudo E, Muraglia E, Nizi E, Orvieto F, Pace P, Pescatore G, Scarpelli R, Stillmock K, Witmer MV, Rowley M. Discovery of raltegravir, a potent, selective orally bioavailable HIV-integrase inhibitor for the treatment of HIV-AIDS infection. J Med Chem. 2008;51:5843–55. doi: 10.1021/jm800245z. [DOI] [PubMed] [Google Scholar]
- 115.Leung D, Abbenante G, Fairlie DP. Protease inhibitors: Current status and future prospects. J Med Chem. 2000;43:305–41. doi: 10.1021/jm990412m. [DOI] [PubMed] [Google Scholar]
- 116.Reigan P, Edwards PN, Gbaj A, Cole C, Barry ST, Page KM, Ashton SE, Luke RWA, Douglas KT, Stratford IJ, Jaffar M, Bryce RA, Freeman S. Aminoimidazolylmethyluracil analogues as potent inhibitors of thymidine phosphorylase and their bioreductive nitroimidazolyl prodrugs. J Med Chem. 2005;48:392–402. doi: 10.1021/jm049494r. [DOI] [PubMed] [Google Scholar]
- 117.Manikowski A, Verri A, Lossani A, Gebhardt BM, Gambino J, Focher F, Spadari S, Wright GE. Inhibition of herpes simplex virus thymidine kinases by 2-phenylamino-6-oxopurines and related compounds: Structure–activity relationships and antiherpetic activity in vivo. J Med Chem. 2005;48:3919–29. doi: 10.1021/jm049059x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Clements JM, Beckett RP, Brown A, Catlin G, Lobell M, Palan S, Thomas W, Whittaker M, Wood S, Salama S, Baker PJ, Rodgers HF, Barynin V, Rice DW, Hunter MG. Antibiotic activity and characterization of BB-3497, a novel peptide deformylase inhibitor. Antimicr Agents Chemother. 2001;45:563–70. doi: 10.1128/AAC.45.2.563-570.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Koropatkin NM, Cleland WW, Holden HM. Kinetic and structural analysis of alpha-d-glucose-1-phosphate cytidylyltransferase from salmonella typhi. J Biol Chem. 2005;280:10774–80. doi: 10.1074/jbc.M414111200. [DOI] [PubMed] [Google Scholar]
- 120.Heffron SE, Jurnak F. Structure of an EF-Tu complex with a thiazolyl peptide antibiotic determined at 2. 35 Å resolution: Atomic basis for GE2270A inhibition of EF-Tu. Biochemistry. 2000;39:37–45. doi: 10.1021/bi9913597. [DOI] [PubMed] [Google Scholar]
- 121.Wells JA, McClendon CL. Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature. 2007;450:1001–9. doi: 10.1038/nature06526. [DOI] [PubMed] [Google Scholar]
- 122.Whitty A, Kumaravel G. Between a rock and a hard place? Nat Chem Biol. 2006;2:112–8. doi: 10.1038/nchembio0306-112. [DOI] [PubMed] [Google Scholar]
- 123.Fuller JC, Burgoyne NJ, Jackson RM. Predicting druggable binding sites at the protein-protein interface. Drug Discov Today. 2009;14:155–61. doi: 10.1016/j.drudis.2008.10.009. [DOI] [PubMed] [Google Scholar]
- 124.Barelier S, Pons J, Marcillat O, Lancelin JM, Krimm I. Fragment-based deconstruction of Bcl-x(L) inhibitors. J Med Chem. 2010;53:2577–88. doi: 10.1021/jm100009z. [DOI] [PubMed] [Google Scholar]
- 125.Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, Bruncko M, Deckwerth TL, Dinges J, Hajduk PJ, Joseph MK, Kitada S, Korsmeyer SJ, Kunzer AR, Letai A, Li C, Mitten MJ, Nettesheim DG, Ng S, Nimmer PM, O'Connor JM, Oleksijew A, Petros AM, Reed JC, Shen W, Tahir SK, Thompson CB, Tomaselli KJ, Wang B, Wendt MD, Zhang H, Fesik SW, Rosenberg SH. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435:677–81. doi: 10.1038/nature03579. [DOI] [PubMed] [Google Scholar]
- 126.Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, Johnson EF, Marsh KC, Mitten MJ, Nimmer P, Roberts L, Tahir SK, Xiao Y, Yang X, Zhang H, Fesik S, Rosenberg SH, Elmore SW. Abt-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–8. doi: 10.1158/0008-5472.CAN-07-5836. [DOI] [PubMed] [Google Scholar]
- 127.Tao ZF, Hasvold L, Wang L, Wang X, Petros AM, Park CH, Boghaert ER, Catron ND, Chen J, Colman PM, Czabotar PE, Deshayes K, Fairbrother WJ, Flygare JA, Hymowitz SG, Jin S, Judge RA, Koehler MF, Kovar PJ, Lessene G, Mitten MJ, Ndubaku CO, Nimmer P, Purkey HE, Oleksijew A, Phillips DC, Sleebs BE, Smith BJ, Smith ML, Tahir SK, Watson KG, Xiao Y, Xue J, Zhang H, Zobel K, Rosenberg SH, Tse C, Leverson JD, Elmore SW, Souers AJ. Discovery of a potent and selective bcl-xl inhibitor with in vivo activity. ACS Med Chem Lett. 2014;5:1088–93. doi: 10.1021/ml5001867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Flygare JA, Beresini M, Budha N, Chan H, Chan IT, Cheeti S, Cohen F, Deshayes K, Doerner K, Eckhardt SG, Elliott LO, Feng BN, Franklin MC, Reisner SF, Gazzard L, Halladay J, Hymowitz SG, La H, LoRusso P, Maurer B, Murray L, Plise E, Quan C, Stephan JP, Young SG, Tom J, Tsui V, Um J, Varfolomeev E, Vucic D, Wagner AJ, Wallweber HJA, Wang L, Ware J, Wen ZY, Wong H, Wong JM, Wong M, Wong S, Yu R, Zobel K, Fairbrother WJ. Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152) J Med Chem. 2012;55:4101–13. doi: 10.1021/jm300060k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Seigal BA, Connors WH, Fraley A, Borzilleri RM, Carter PH, Emanuel SL, Fargnoli J, Kim K, Lei M, Naglich JG, Pokross ME, Posy SL, Shen H, Surti N, Talbott R, Zhang Y, Terrett NK. The discovery of macrocyclic XIAP antagonists from a DNA-programmed chemistry library, and their optimization to give lead compounds with in vivo antitumor activity. J Med Chem. 2015;58:2855–61. doi: 10.1021/jm501892g. [DOI] [PubMed] [Google Scholar]
- 130.Vassilev LT. MDM2 inhibitors for cancer therapy. Trends Mol Med. 2007;13:23–31. doi: 10.1016/j.molmed.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 131.Zhao Y, Aguilar A, Bernard D, Wang S. Small-molecule inhibitors of the MDM2-p53 protein-protein interaction (MDM2 inhibitors) in clinical trials for cancer treatment. J Med Chem. 2015;58:1038–52. doi: 10.1021/jm501092z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sun D, Li Z, Rew Y, Gribble M, Bartberger MD, Beck HP, Canon J, Chen A, Chen X, Chow D, Deignan J, Duquette J, Eksterowicz J, Fisher B, Fox BM, Fu J, Gonzalez AZ, Gonzalez-Lopez De Turiso F, Houze JB, Huang X, Jiang M, Jin L, Kayser F, Liu JJ, Lo MC, Long AM, Lucas B, McGee LR, McIntosh J, Mihalic J, Oliner JD, Osgood T, Peterson ML, Roveto P, Saiki AY, Shaffer P, Toteva M, Wang Y, Wang YC, Wortman S, Yakowec P, Yan X, Ye Q, Yu D, Yu M, Zhao X, Zhou J, Zhu J, Olson SH, Medina JC. Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development. J Med Chem. 2014;57:1454–72. doi: 10.1021/jm401753e. [DOI] [PubMed] [Google Scholar]
- 133.Yu M, Wang Y, Zhu J, Bartberger MD, Canon J, Chen A, Chow D, Eksterowicz J, Fox B, Fu J, Gribble M, Huang X, Li Z, Liu JJ, Lo MC, McMinn D, Oliner JD, Osgood T, Rew Y, Saiki AY, Shaffer P, Yan X, Ye Q, Yu D, Zhao X, Zhou J, Olson SH, Medina JC, Sun D. Discovery of potent and simplified piperidinone-based inhibitors of the MDM2-p53 interaction. ACS Med Chem Lett. 2014;5:894–9. doi: 10.1021/ml500142b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wendt MD, Rockway TW, Geyer A, McClellan W, Weitzberg M, Zhao X, Mantei R, Nienaber VL, Stewart K, Klinghofer V, Giranda VL. Identification of novel binding interactions in the development of potent, selective 2-naphthamidine inhibitors of urokinase. Synthesis, structural analysis, and SAR of N-phenyl amide 6-substitution. J Med Chem. 2004;47:303–24. doi: 10.1021/jm0300072. [DOI] [PubMed] [Google Scholar]
- 135.D'Abramo CM, Archambault J. Small molecule inhibitors of human papillomavirus protein - protein interactions. The Open Virology J. 2011;5:80–95. doi: 10.2174/1874357901105010080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Sun Q, Burke JP, Phan J, Burns MC, Olejniczak ET, Waterson AG, Lee T, Rossanese OW, Fesik SW. Discovery of small molecules that bind to K-Ras and inhibit SOS-mediated activation. Angew Chem Int Edit. 2012;51:6140–3. doi: 10.1002/anie.201201358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Pikul S, Dunham KM, Almstead NG, De B, Natchus MG, Taiwo YO, Williams LE, Hynd BA, Hsieh LC, Janusz MJ, Gu F, Mieling GE. Heterocycle-based MMP inhibitors with P2' substituents. Bioorg Med Chem Lett. 2001;11:1009–13. doi: 10.1016/s0960-894x(01)00137-8. [DOI] [PubMed] [Google Scholar]
- 138.Kozakov D, Hall DR, Jehle S, Luo L, Ochiana SO, Jones EV, Pollastri M, Allen KN, Whitty A, Vajda S. Ligand deconstruction: Why some fragment binding positions are conserved and others are not. Proc Natl Acad Sci U S A. 2015;112:E2585–94. doi: 10.1073/pnas.1501567112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mosyak L, Zhang Y, Glasfeld E, Haney S, Stahl M, Seehra J, Somers WS. The bacterial cell-division protein ZipA and its interaction with an FtsZ fragment revealed by X-ray crystallography. The EMBO J. 2000;19:3179–91. doi: 10.1093/emboj/19.13.3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Jennings LD, Foreman KW, Rush TS, 3rd, Tsao DH, Mosyak L, Li Y, Sukhdeo MN, Ding W, Dushin EG, Kenny CH, Moghazeh SL, Petersen PJ, Ruzin AV, Tuckman M, Sutherland AG. Design and synthesis of indolo[2 3-a]quinolizin-7-one inhibitors of the ZipA-FtsZ interaction. Bioorg Med Chem Lett. 2004;14:1427–31. doi: 10.1016/j.bmcl.2004.01.028. [DOI] [PubMed] [Google Scholar]
- 141.Kenny CH, Ding WD, Kelleher K, Benard S, Dushin EG, Sutherland AG, Mosyak L, Kriz R, Ellestad G. Development of a fluorescence polarization assay to screen for inhibitors of the FtsZ/ZipA interaction. Anal Biochem. 2003;323:224–33. doi: 10.1016/j.ab.2003.08.033. [DOI] [PubMed] [Google Scholar]
- 142.Tsao DH, Sutherland AG, Jennings LD, Li Y, Rush TS, 3rd, Alvarez JC, Ding W, Dushin EG, Dushin RG, Haney SA, Kenny CH, Malakian AK, Nilakantan R, Mosyak L. Discovery of novel inhibitors of the ZipA/FtsZ complex by NMR fragment screening coupled with structure-based design. Bioorg Med Chem. 2006;14:7953–61. doi: 10.1016/j.bmc.2006.07.050. [DOI] [PubMed] [Google Scholar]
- 143.Jennings LD, Foreman KW, Rush TS, Tsao DHH, Mosyak L, Kincaid SL, Sukhdeo MN, Sutherland AG, Ding WD, Kenny CH, Sabus CL, Liu HL, Dushin EG, Moghazeh SL, Labthavikul P, Petersen PJ, Tuckman M, Haney SA, Ruzin AV. Combinatorial synthesis of substituted 3-(2-indolyl)piperidines and 2-phenyl indoles as inhibitors of ZipA-FtsZ interaction. Bioorg Med Chem. 2004;12:5115–31. doi: 10.1016/j.bmc.2004.07.031. [DOI] [PubMed] [Google Scholar]
- 144.Rush TS, Grant JA, Mosyak L, Nicholls A. A shape-based 3-D scaffold hopping method and its application to a bacterial protein-protein interaction. J Med Chem. 2005;48:1489–95. doi: 10.1021/jm040163o. [DOI] [PubMed] [Google Scholar]
- 145.Rai G, Vyjayanti VN, Dorjsuren D, Simeonov A, Jadhav A, Wilson DM, 3rd, Maloney DJ. Synthesis biological evaluation and structure-activity relationships of a novel class of apurinic/apyrimidinic endonuclease 1 inhibitors. J Med Chem. 2012;55:3101–12. doi: 10.1021/jm201537d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Abbotts R, Madhusudan S. Human AP endonuclease 1 (APE1): From mechanistic insights to druggable target in cancer. Cancer Treat Rev. 2010;36:425–35. doi: 10.1016/j.ctrv.2009.12.006. [DOI] [PubMed] [Google Scholar]
- 147.Wilson DM, Simeonov A. Small molecule inhibitors of DNA repair nuclease activities of APE1. Cell Mol Life Sci. 2010;67:3621–31. doi: 10.1007/s00018-010-0488-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Patel D, Jain M, Shah SR, Bahekar R, Jadav P, Joharapurkar A, Dhanesha N, Shaikh M, Sairam KV, Kapadnis P. Discovery of potent, selective and orally bioavailable triaryl-sulfonamide based PTP1B inhibitors. Bioorg Med Chem Lett. 2012;22:1111–7. doi: 10.1016/j.bmcl.2011.11.122. [DOI] [PubMed] [Google Scholar]
- 149.Scapin G, Patel SB, Becker JW, Wang QP, Desponts C, Waddleton D, Skorey K, Cromlish W, Bayly C, Therien M, Gauthier JY, Li CS, Lau CK, Ramachandran C, Kennedy BP, Asante-Appiah E. The structural basis for the selectivity of benzotriazole inhibitors of PTP1B. Biochemistry. 2003;42:11451–9. doi: 10.1021/bi035098j. [DOI] [PubMed] [Google Scholar]
- 150.Barr AJ. Protein tyrosine phosphatases as drug targets: Strategies and challenges of inhibitor development. Fut Med Chem. 2010;2:1563–76. doi: 10.4155/fmc.10.241. [DOI] [PubMed] [Google Scholar]
- 151.Douty B, Wayland B, Ala PJ, Bower MJ, Pruitt J, Bostrom L, Wei M, Klabe R, Gonneville L, Wynn R, Burn TC, Liu PCC, Combs AP, Yue EW. Isothiazolidinone inhibitors of PTP1B containing imidazoles and imidazolines. Bioorg Med Chem Lett. 2008;18:66–71. doi: 10.1016/j.bmcl.2007.11.012. [DOI] [PubMed] [Google Scholar]
- 152.Zhao J, Du YH, Horton JR, Upadhyay AK, Lou B, Bai Y, Zhang X, Du LP, Li MY, Wang BH, Zhang LX, Barbieri JT, Khuri FR, Cheng XD, Fu HA. Discovery and structural characterization of a small molecule 14-3-3 protein-protein interaction inhibitor. Proc Natl Acad Sci U S A. 2011;108:16212–6. doi: 10.1073/pnas.1100012108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gardino AK, Smerdon SJ, Yaffe MB. Structural determinants of 14-3-3 binding specificities and regulation of subcellular localization of 14-3-3-ligand complexes: A comparison of the x-ray crystal structures of all human 14-3-3 isoforms. Semin Cancer Biol. 2006;16:173–82. doi: 10.1016/j.semcancer.2006.03.007. [DOI] [PubMed] [Google Scholar]
- 154.De Vries-van Leeuwen IJ, da Costa Pereira D, Flach KD, Piersma SR, Haase C, Bier D, Yalcin Z, Michalides R, Feenstra KA, Jimenez CR, de Greef TF, Brunsveld L, Ottmann C, Zwart W, de Boer AH. Interaction of 14-3-3 proteins with the estrogen receptor alpha f domain provides a drug target interface. Proc Natl Acad Sci U S A. 2013;110:8894–9. doi: 10.1073/pnas.1220809110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Thiel P, Roglin L, Meissner N, Hennig S, Kohlbacher O, Ottmann C. Virtual screening and experimental validation reveal novel small-molecule inhibitors of 14-3-3 protein-protein interactions. Chem Commun (Camb) 2013;49:8468–70. doi: 10.1039/c3cc44612c. [DOI] [PubMed] [Google Scholar]
- 156.Wang Y, Kaiser CE, Frett B, Li HY. Targeting mutant kras for anticancer therapeutics: A review of novel small molecule modulators. J Med Chem. 2013;56:5219–30. doi: 10.1021/jm3017706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Feig LA, Cooper GM. Inhibition of NIH-3T3 cell-proliferation by a mutant Ras protein with preferential affinity for GDP. Mol Cell Biol. 1988;8:3235–43. doi: 10.1128/mcb.8.8.3235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Maurer T, Garrenton LS, Oh A, Pitts K, Anderson DJ, Skelton NJ, Fauber BP, Pan B, Malek S, Stokoe D, Ludlam MJ, Bowman KK, Wu J, Giannetti AM, Starovasnik MA, Mellman I, Jackson PK, Rudolph J, Wang W, Fang G. Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. Proc Natl Acad Sci U S A. 2012;109:5299–304. doi: 10.1073/pnas.1116510109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wang WR, Fang GW, Rudolph J. Ras inhibition via direct Ras binding-is there a path forward? Bioorg Med Chem Lett. 2012;22:5766–76. doi: 10.1016/j.bmcl.2012.07.082. [DOI] [PubMed] [Google Scholar]
- 160.Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51. doi: 10.1038/nature12796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Christ F, Debyser Z. The LEDGF/p75 integrase interaction, a novel target for anti-HIV therapy. Virology. 2013;435:102–9. doi: 10.1016/j.virol.2012.09.033. [DOI] [PubMed] [Google Scholar]
- 162.Malet I, Calvez V, Marcelin AG. The future of integrase inhibitors of HIV-1. Curr Opin Virol. 2012;2:580–7. doi: 10.1016/j.coviro.2012.08.005. [DOI] [PubMed] [Google Scholar]
- 163.Demeulemeester J, Chaltin P, Marchand A, De Maeyer M, Debyser Z, Christ F. LEDGINs, non-catalytic site inhibitors of HIV-1 integrase: A patent review (2006 - 2014) Exp Opin Therap Patents. 2014;24:609–32. doi: 10.1517/13543776.2014.898753. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





