Drug Discovery Today: Technologies

Vol. 10, No. 4 2013

Editors-in-Chief Kelvin Lam – Blue Sky Biotech, Inc., Worcester, MA Henk Timmerman – Vrije Universiteit, The Netherlands DRUG DISCOVERY

TODAY

TECHNOLOGIES

Scaffold hopping

De novo design – hop(p)ing against hope Gisbert Schneider Swiss Federal Institute of Technology (ETH), Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zu¨rich, Switzerland

Current trends in computational de novo design provide a fresh approach to ‘scaffold-hopping’ in drug

Section editor: D. Lloyd – Trinity College, Dublin, Ireland.

discovery. The methodological repertoire is no longer limited to receptor-based methods, but specifically ligand-based techniques that consider multiple properties in parallel, including the synthetic feasibility of the computer-generated molecules and their polypharmacology, provide innovative ideas for the discovery of new chemical entities. The concept of fragment-based and virtual reaction-driven design enables rapid compound optimization from scratch with a manageable complexity of the search. Starting from known drugs as a reference, such algorithms suggest drug-like molecules with motivated scaffold variations, and advanced mathematical models of structure-activity landscapes and multi-objective design techniques have created new opportunities for hit and lead finding.

Introduction De novo design aims to generate novel bioactive molecular entities ‘from scratch’. Ideally, such computer-designed molecules have drug-like properties, potently interact with a target macromolecule in the low nanomolar range, lack adverse effects, are orally bioavailable, free of intellectual property rights, and may be easily synthesized from cheap starting material. Evidently, such a catalog of demands and expectations is unrealistic. What one can expect from de novo design software solutions, however, is some degree of structural novelty, modest bioactivity, and chemical feasibility of the designed compounds [1]. This has not always been so. E-mail address: G. Schneider ([email protected]) 1740-6749/$ ß 2012 Elsevier Ltd. All rights reserved.

After several remarkable but salutary failures in the pioneering days, it quickly became evident that while constructing molecules atom-by-atom might in fact lead to novel chemical entities that fill a binding cavity to a desired extent in a desired binding pose and form potentially strong interactions with a receptor structure, there is a high risk that these artificial compounds cannot be realized by chemical synthesis in a straightforward fashion or possess adverse physicochemical properties. Contemporary computer-assisted de novo design algorithms largely rely on molecular fragments as the basic building blocks to come up with appealing molecular frameworks rather than fully optimized potential ligands [2]. This development is partly owed to the success of fragment-based hit finding by high-throughput NMR and Xray methods and demonstrates how our current understanding of computational drug design has co-evolved with advancing technological developments [3,4]. Equally important, it has been realized that a desired or observed pharmacological effect often is the result of a compound’s activity on multiple targets. Explicit consideration of potential polypharmacology has become viable in drug design [5,6], enabled by the availability of large functionally annotated compound databases [7,8]. It therefore is a worthwhile exercise to try to de novo generate new chemical entities (NCEs) that exhibit a desired target and property profile. A promising approach is to use a known drug as a reference structure that serves as a template for virtual compound assembly (ligand- or template-based design). In this context, the term ‘scaffold-hopping’ is closely related and in some way linked with computer-assisted molecular design [9]. The first joint occurrence of the mesh words ‘de novo design’ and ‘drug’ in the context of computational chemistry dates back to a

http://dx.doi.org/10.1016/j.ddtec.2012.06.001

e453

Drug Discovery Today: Technologies | Scaffold hopping

Vol. 10, No. 4 2013

50 40

“scaffold-hopping”

30 N 20

“de novo design” & drug

10 0 2011

2006

1999

1991

Drug Discovery Today: Technologies

Figure 1. Number of publications per year with the keywords {‘de novo design’ and drug} (blue bars), and ‘scaffold-hopping’ (stacked orange bars) mentioned in the topic descriptions according to Web of Science (Thomson Reuters, New York, 2011).

seminal article by Moon and Howe from 1991 (Fig. 1) [10]. Ever since, the term ‘de novo design’ has been used on a regular basis to refer to computer-assisted molecular design studies. Notably, there seems to be no significant increase in publications per year, although after an initial small wave of papers published in the mid 1990s, we are currently witnessing a renewed interest in this field, possibly motivated by improved techniques and increased demand [11,12]. This hypothesis is clearly mirrored in the remarkable increase in publications on ‘scaffold-hopping’, especially after 2006, with the inaugural publication dating back to our own work from 1999 [13]. It seems that finding meaningful bioisosters and scaffold replacements has become feasible by systematic molecular de novo design, and is not a product of pure chance. In fact, the field has been reviewed recently presenting numerous success stories [14,15]. We here discuss some aspects of de novo design that require further attention so that the full potential of this scientific approach may be exploited for future drug discovery.

The odds are good but the goods are odd From a mere statistical point of view, generating new NCEs from scratch with a clearly defined activity and property profile should be borderline impossible. Even model-based computational methods allow us to construct by far too many virtual compounds with drug-like properties than could ever be exhaustively screened in silico or in vitro. To be successful in this enterprise and increase our chance of finding potent compounds among a small set of selected candidates, we have to take smart shortcuts by focusing on promising regions in chemical space – and accept imperfection. This blunt statement simply means that any knowledge about focal points of interest for a computational search for NCEs, for example, by considering privileged substructures and biophoric molecular patterns in a virtual screen, is likely to reduce the novelty of the designs. While we can expect to find e454

www.drugdiscoverytoday.com

appealing new compounds by interpolating between reference points (e.g. representations of known drugs and their binding sites) it is unlikely to discover never-before-seen molecular structures that pass our current definition (and perception) of pharmaceutically active agents. Herein lies a dilemma of both ligand- and receptor/structure-based de novo drug designs. From a medicinal chemistry perspective, computationally assembled and virtually screened candidate compounds have always had a certain tendency to be either awkward or boring: one either finds obvious analogs or unexpected similarities to given reference drugs. With the introduction of fragment- and reaction-based approaches to virtual compound assembly and the predictive functions used for compound scoring, the potential synthetic accessibility of candidate structures can be controlled better, although these methods might also come at the cost of reduced structural novelty as they are based on known chemistry. Estimations of the number of theoretically feasible drug-like compounds that could be generated using such a combinatorial technique range between 1015 and 1018 compounds – still a sufficiently large part of chemical space that should allow for finding attractive NCEs [16,17]. Traditionally, receptor-based in situ fragment assembly has been grounded on the assumption that fragment-contributions to the ligand binding energy are additive [18]. While this principle holds if the binding mode and orientation of the individual fragments are only marginally perturbed in the elaborated product, there are growing numbers of reports of unexpected binding modes and non-additive, that is, nonlinear, fragment contributions [19,20]. Non-additivity of DG contributions of individual fragments can lead to marked discrepancies between the sum of fragment affinities and the final ligand (Fig. 2) [21,22]. In order to achieve more reliable estimations of binding energies of virtually assembled compounds, it will be mandatory to account for the effect of linker elements during the construction process. Possibly, the evaluation of the full ligand product is obligatory. Consequently, despite the appeal of fast algorithmic solutions for fragment-based exhaustive or global combinatorial product evaluation that implement the additivity principle, the actual practical applicability of these techniques can hardly be assessed a priori. As a workaround, both stochastic and deterministic local optimization strategies that score the full product have become a main design strategy. Although these approaches do not guarantee finding the globally best compound, there is ample evidence that they identify new bioactive ligands. In fact, a trade-off between conservative and exploratory design can be helpful in hit identification. For example, our ligand-based fragment-growing tool DOGS can be tuned to a desired ratio of scaffold exploitation/exploration during candidate compound assembly [23]. It also features a simplistic deterministic approach to estimating the score contribution of linker parts that originate from

Vol. 10, No. 4 2013

Drug Discovery Today: Technologies | Scaffold hopping

H N

O S

O

N

H N

O +

S

N H

ΔGfragment1 = -11.4 kJ mol-1

N

Cl

ΔGfragment2 = -24.2 kJ mol-1

O

O

S O

N H

S Cl

ΔGligand = -49.6 kJ mol-1

Tyr99 Gln192 Thr98 Ser195

S4 Phe174

Cys191

Trp215 S1

Val213 Ala190

Tyr228

Asp189 Drug Discovery Today: Technologies

Figure 2. Example of fragment super-additivity [58]: the experimentally determined free energy of binding DG of the product, a potent factor Xa inhibitor (Ki = 2 nM), exceeds the sum of the individual fragment contributions by 14 kJ  mol 1. A single bond was added as linker (filled arrowhead). The cartoon shows the enzyme–ligand complex (PDB ID: 4a7i). Several interacting residue side-chains are highlighted.

O

STEP 1 De novo design

HO O HO O

N

N H

STEP 2 Virtual screening

259,829 unique structures

9,416 candidates

NH2

Lisinopril

STEP 3 Framework analysis

Drug Discovery Today: Technologies

Figure 3. Analysis of molecular frameworks resulting from ligand-based de novo design using the ACE inhibitor Lisinopril as template. The software Flux was employed to generate a large pool of designs (Step 1), which were condensed to promising candidates using a random forest model predicting ACE inhibition (Step 2), and subjected to scaffold analysis (Step 3). The ten most frequently generated molecular frameworks are presented. All of these frameworks are also found in known ACE inhibitors. The molecular framework of Lisinopril is highlighted in magenta.

www.drugdiscoverytoday.com

e455

Drug Discovery Today: Technologies | Scaffold hopping

Vol. 10, No. 4 2013

synthesis reactions. Similarly, evolutionary design algorithms are easily adaptable to produce desired scaffold diversity of the products [24,25]. Instead of accounting for synthetic accessibility by explicit reaction-based compound construction, one can also rely on software that has especially been designed for synthesis planning. For example, the computer programs CAESA [26], SYLVIA [27], and RouteDesigner [28] propose synthesis plans for de novo generated compounds post hoc. Fragment-based molecular design intrinsically follows the concept of scaffold-hopping, which aims at identifying scaffold replacements in a known drug so that an NCE with the same or improved properties as the reference drug emerges. Intentionally such an approach mimics compound design in medicinal chemistry, where a human mind conceives of new

(a)

molecular structures by drawing ideas from personal but individually unique knowledge. One needs to accept that any medicinal chemist’s choice is influenced and biased towards preferred regions of chemical space by years of training, and that such preferences affect the innovation potential of human-designed compounds – just like computational methods do. For the inclined user, own expertise may be complemented by computer-assisted scaffold-hopping and ultimately de novo design to come up with surprising but acceptable suggestions that can be actually realized in the laboratory and support hit and lead structure identification in drug discovery. A worked example of de novo design for the purpose of scaffold-hopping is presented in Fig. 3. In this pilot study [29], we applied our software Flux [30,31] to generating

(b) DOGS

+

Building blocks

Reactions

De novo design

Template Comparative (“homology”) model of Plk1

O

Pocket-derived pharmacophore model

N

H2N

+ O

N

O

Virtual screening of compound database Reductive amination Potent type-II Plk1 inhibitor O O

N H

O

O Cl

N H

O O

N N

N

Template

Designed compound Drug Discovery Today: Technologies

Figure 4. From receptor structure to a potent inhibitor by virtual screening (a), and conservative scaffold-hopping by fragment-based de novo design (b). The template structure for de novo design was obtained in a receptor-based virtual screen for Plk1 inhibitors [59]. It served as input for the software DOGS, which suggested a variation of the template together with a plausible synthesis route. The designed compound also strongly inhibits Plk1 activity [17]. The frameworks of both molecules are shown in blue color. They differ in the size of the rings and the linker in the eastern part of the structures.

e456

www.drugdiscoverytoday.com

Vol. 10, No. 4 2013

Drug Discovery Today: Technologies | Scaffold hopping

compounds that mimic the reference compound Lisinopril, a potent inhibitor of angiotensin converting enzyme (ACE). Flux used a selection of 10,497 drug-derived virtual building blocks and RECAP-type [32] connectivity for fragment-based molecular design, guided by an evolutionary algorithm (a (1, l) evolution strategy). The design objective was to optimize two-dimensional pharmacophore similarity (CATS descriptor) between the template (here: Lisinopril) and the virtually generated compounds. We condensed the initially large number of more than 250,000 suggestions by virtual screening for potential ACE inhibitors using a random forest [33] classifier model. The remaining 9416 candidate molecules featured a total of 2582 different scaffolds (graph frameworks). Only 55 scaffolds appeared more than 100 times in the final compound set, indicating strong algorithmic scaffold exploration. It is of note that all of the ten most frequent scaffolds are contained in known ACE inhibitors, which clearly mirrors chemotypes memorized by the classifier model. This exercise demonstrates that (i) useful molecular frameworks can be obtained by fragment-based de novo design, and (ii) recurring scaffold motifs might be of particular interest for further analysis. At the same time it becomes apparent that meaningful extrapolation from the domain of training or reference compounds is unlikely when (Q)SAR models are used for candidate compound triaging. An example of a minor scaffold variation is the potent typeII inhibitor of Polo-like kinase 1 (Plk1) which was generated by ligand-based de novo design using the DOGS method, and synthesized according to the reaction scheme proposed by

the software (Fig. 4) [17]. In analogy to biophysical fragmentfinding and extension [34], DOGS grows new compounds from a small start fragment so that virtual products are obtained that possess high pharmacophoric feature similarity to the template. This study suggests that conservative ligandbased de novo design may be applied to exploring the structural neighborhood of known actives in the absence of a known receptor structure. However, when reliable receptor models are available, structural optimization by small incremental modifications could also be performed by computationally more demanding free energy estimation methods like BOMB [35].

Against all odds – but take a look at it now! The central idea of scaffold-hopping is to find structurally different chemotypes with isofunctional bioactivity to a given template. As the actually relevant set of functiondetermining molecular features (substructures, pharmacophore points) is often unknown a priori, virtual screening and de novo design alike are typically performed with a permissive pharmacophore model to ensure sufficiently large scaffold variability among the matching compounds. The degree of ‘fuzziness’ or permissiveness of the model critically influences scaffold diversity in the hit lists (Fig. 5) [36]. Of note, the overlap between individual hit lists is not necessarily large, but occurrences of the same compounds in multiple screens (or consensus scoring approaches) argues for at least partially shared bioactivity (‘Authority Effect’) [37,38]. This observation is extremely useful for de novo design, as it

O

N

LIQUID

O Cl

N

S

Cl O N

Model 1

Cl

Clopidogrel Top 50 compounds LIQUID

O

N

Model 1

Model 2

N

35

S

Model 2

14 1

5

44

O LIQUID

30

N

N Model 3

Model 3 Drug Discovery Today: Technologies

Figure 5. Similarity searching with a conservative (model 1) or increasingly permissive pharmacophore hypotheses (models 2 and 3) results in distinctive virtual hit lists with limited overlap. Here, clopidogrel served as the template (query; left panel), and the software LIQUID [60] computed probabilistic pharmacophoric feature densities (green: lipophilic, red: hydrogen-bond acceptor). The top-scoring hits are shown together with their respective LIQUID pharmacophore model. The only compound retrieved from the screening compound pool by all three pharmacophore hypotheses is clopidogrel itself (right panel).

www.drugdiscoverytoday.com

e457

Drug Discovery Today: Technologies | Scaffold hopping

Vol. 10, No. 4 2013

COBRA

Factor Xa

PPARγ

H-bond donors

TPSA

alogP

COX

combine Drug Discovery Today: Technologies

Figure 6. Property landscapes of a collection of 11,230 drugs and lead compounds (COBRA v10.3) [61]. Molecules were represented by a 210dimensional pharmacophore descriptor (CATS2D v2) and projected to two dimensions using stochastic neighbor embedding [62] (SNE). Surfaces were computed using our software tool LiSARD [46]. Red color indicates undesired areas in chemical space, green color denotes regions containing compounds with desired features. Color intensity correlates with prediction confidence. Upper panel: The 2D distribution of the compound collection (left), and corresponding activity islands formed by factor Xa, PPARg, and cyclooxygenase (COX) ligands. Lower panel: Computed property landscapes for the number of hydrogen-bond donors, total polar surface area (TPSA), and lipophilicity (a log P); the combined landscape (TPSA + a log P) shown on the right indicates a pronounced preferred target area (arrow) for de novo design with regard to these two properties. Note that the combined landscape is tilted and slightly enlarged compared to the other visualizations to highlight hills and valleys. Physicochemical compound properties were computed using the software implementations in MOE (v2011, The Chemical Computing Group, Montreal, Canada).

provides a rationale for scoring and analyzing the numerous suggestions that can be obtained from a compound generator. Although the numbers of high-scoring molecules drop away rapidly as more selection criteria are applied, there is motivated hope that the fraction of actives increases. The general optimization paradigm requires acceptably smooth fitness landscapes [39,40], which seems to be fulfilled by using fragments (as opposed to atoms) as molecular building blocks for de novo design. Can we also predict the effect of seemingly minor structural modification on molecular properties, including affinity (e.g. ‘activity cliffs’), physicochemistry (e.g. aqueous solubility), and pharmacokinetics (e.g. oxidative metabolism)? Matched Molecular Pairs Analysis (MMPA) [41] has lately raised great interest in medicinal chemistry, and might also support de novo design in this regard [42,43]. Multi-objective optimization provides another concept for computer-assisted drug design that helps navigate in chemical space under the constraint of partially conflicting objective functions [44]. Focusing on compounds with sub-optimal but still acceptable properties might actually be key to success in de novo design, simply due to the fact that all predictive models are error-prone when applied to an individual molecule. Graphical visualization of the optimization progress in models of the underlying fitness landscape e458

www.drugdiscoverytoday.com

can help steer into the right direction and avoid artifact designs caused by over-optimization [45]. As an illustration, the graphical combination of two objective functions is presented in Fig. 6. The resulting fitness landscape was generated to implicitly address several design criteria while not being limited to compound potency as the sole driving force [46]. Similar visualizations with different underlying landscape models have been developed recently [47,48]. The appropriate mathematical framework is available to enable multidimensional optimization of synthetically accessible compounds as the next realistic step in practical computerassisted drug discovery. This encouraging development gives new hope for successful NCE identification by fragmentbased de novo design.

Conclusion: back to the future! One may wonder why computational de novo design has become fashionable again [49,50]. Although the basic ideas were conceived approximately three decades ago, its essential ingredients have remained the same – but with a new twist, admittedly. We have come a long way ever since, and have learned from earlier failure. Contemporary approaches include fragment-based compound assembly, a variety of scoring schemes that allow for nonlinear contributions,

Vol. 10, No. 4 2013

explicit or implicit consideration of synthetic feasibility of the designs, and multi-objective optimization addressing multiple targets and properties. Existing software solutions focus on each of these features to a different extent, and it is only a matter of time and effort to implement working and applicable de novo design tools that actually suggest synthesizable compounds exhibiting desired polypharmacology [51]. Proven scaffold-hopping potential might actually be key to successful identification of NCEs, and ligand-based de novo methods offer themselves as premier tools for rapid analog design. In future applications, de novo compound generators could also be directly linked to patent databases to identify most promising candidate scaffolds and potential activity profiles. Another idea is to compare designed compounds to databases such as ZINC [52], which contain physically available substances with known synthetic accessibility. This could flag up compounds that share the scaffold (both as a framework and more particularly with specific heteroatoms) with a design, to obtain confidence to the feasibility of the design and a potential shortcut to synthetic work. While there has been notable progress during the past years, several limiting factors still prevent de novo design approaches from being overly successful [1]. For example, water molecules modify the size, shape and pharmacophoric features of a ligand-binding pocket, thereby critically affecting the diversity of chemotypes that can be accommodated [53]. Mancera and coworkers were among the first to investigate the role that water molecules, specifically hydration sites in the receptor cavity, can have on the structural interpretation of ligand-derived pharmacophore models [54]. Since water molecules are known to influence molecular diversity in receptor-based design [55], their importance clearly needs to be recognized in the context of ligand-based approaches. Another major limitation originates from insufficient consideration of entropic effects upon ligand binding and consequently the still largely unsolved scoring problem [56], although recent developments of ab initio approaches, like the fragment molecular orbital method, point towards progress in this regard for structure-based de novo design [57]. In the author’s view, the most critical part in computational drug design has always been its actual practical realization, so that rapid feedback loops enable adaptive learning. As long as coupling computer-based design to immediate automated synthesis and testing will remain a future (but certainly attainable) goal, teams of dedicated modelers and steadfast chemists will have to do the hard work. Despite all enthusiasm, we should not forget that any computational prediction is error-prone, and there currently are too few worked examples to allow for a sound statistical assessment of success rates in de novo design. This fact alone should encourage us to find out.

Drug Discovery Today: Technologies | Scaffold hopping

Acknowledgements The author is grateful to his coworkers and collaborating partners for realizing exciting molecular design studies. Dr. David Lloyd is thanked for helpful comments on the manuscript. The work presented was financially supported by ETH ¨ rich, the Swiss National Science Foundation (SNSF, grant Zu 205321-134783), Deutsche Forschungsgemeinschaft (DFG, ¨ rich. The Chegrant FOR1406_TP4), and OPO-Foundation Zu mical Computing Group Inc. provided a research license of the software MOE. The funding sources had no such involvement in this study.

References 1 2

3 4

5 6 7

8

9 10

11 12 13

14 15 16 17 18 19 20 21

22

Schneider, G. (2010) Virtual screening: an endless staircase? Nat Rev. Drug Discov. 9, 273–276 Sheng, C. and Zhang, W. (2012) Fragment informatics and computational fragment-based drug design: an overview and update. Med. Res. Rev. doi:10.1002/med.21255 (in press) Law, R. et al. (2009) The multiple roles of computational chemistry in fragment-based drug design. J. Comput. Aided Mol. Des. 23, 459–473 Hajduk, P.J. and Greer, J. (2007) A decade of fragment-based drug design: strategic advances and lessons learned. Nat. Rev. Drug Discov. 6, 211– 219 Garcia-Serna, R. and Mestres, J. (2010) Anticipating drug side effects by comparative pharmacology. Expert Opin. Drug Metab. Toxicol. 6, 1253–1263 Merino, A. et al. (2010) Drug profiling: knowing where it hits. Drug Discov. Today 15, 749–756 Gleeson, M.P. et al. (2011) Probing the links between in vitro potency, ADMET and physicochemical parameters. Nat. Rev. Drug Discov. 10, 197–208 Metz, J.T. and Hajduk, P.J. (2010) Rational approaches to targeted polypharmacology: creating and navigating protein–ligand interaction networks. Curr. Opin. Chem. Biol. 14, 498–504 Hartenfeller, M. and Schneider, G. (2011) Enabling future drug discovery by de novo design. WIREs Comp. Mol. Sci. 1, 742–759 Moon, J.B. and Howe, W.J. (1991) Computer design of bioactive molecules: a method for receptor-based de novo ligand design. Proteins 11, 314–328 Schneider, G. and Fechner, U. (2005) Computer-based de novo design of drug-like molecules. Nat. Rev. Drug Discov. 4, 649–663 Hartenfeller, M. and Schneider, G. (2011) De novo drug design. Methods Mol. Biol. 672, 299–323 Schneider, G. et al. (1999) ‘Scaffold-hopping’ by topological pharmacophore search: a contribution to virtual screening. Angew. Chem. Int. Ed. 38, 2894–2896 Schneider, G. (2012) Designing the molecular future. J. Comput. Aided Mol. Des. 26, 115–120 Langdon, S.R. et al. (2010) Bioisosteric replacement and scaffold hopping in lead generation and optimization. Mol. Inform. 29, 366–385 Rarey, M. and Stahl, M. (2001) Similarity searching in large combinatorial chemistry spaces. J. Comput. Aided Mol. Des. 15, 497–520 Schneider, G. et al. (2011) Reaction-driven de novo design, synthesis and testing of potential type II kinase inhibitors. Future Med. Chem. 3, 415–424 Hajduk, P.J. (2006) Fragment-based drug design: how big is too big? J. Med. Chem. 49, 6972–6976 Ichihara, O. et al. (2011) Compound design by fragment-linking. Mol. Inform. 30, 298–306 Babaoglu, K. and Shoichet, B.K. (2006) Deconstructing fragment-based inhibitor discovery. Nat. Chem. Biol. 2, 720–723 Murray, C.W. and Verdonk, M.L. (2002) The consequences of translational and rotational entropy lost by small molecules on binding to proteins. J. Comput. Aided Mol. Des. 16, 741–753 Hubbard, R.E. et al. (2007) Informatics and modeling challenges in fragment-based drug discovery. Curr. Opin. Drug Discov. Dev. 10, 289–297 www.drugdiscoverytoday.com

e459

Drug Discovery Today: Technologies | Scaffold hopping

23 24 25

26

27 28

29

30 31

32

33

34 35

36

37

38 39 40 41

Hartenfeller, M. et al. (2012) DOGS: reaction-driven de novo design of bioactive compounds. PLoS Comput. Biol. 8, e1002380 Schneider, G. et al. (2009) Voyages to the (un)known: adaptive design of bioactive compounds. Trends Biotechnol. 27, 18–26 Hiss, J.A. et al. (2010) Concepts and applications of natural computing techniques in de novo drug and peptide design. Curr. Pharm. Des. 16, 1656– 1665 Gillett, V.J. (1995) SPROUT, HIPPO and CAESA: tools for de novo structure generation and estimation of synthetic accessibility. Perspect. Drug Discov. Des. 3, 34–50 Zaliani, A. et al. (2009) Second-generation de novo design: a view from a medicinal chemist perspective. J. Comput. Aided Mol. Des. 23, 593–602 Law, J. et al. (2009) Route designer: a retrosynthetic analysis tool utilizing automated retrosynthetic rule generation. J. Chem. Inform. Model. 49, 593–602 Krueger, B.A. et al. (2009) Scaffold-hopping potential of fragment-based de novo design: the chances and limits of variation. Comb. Chem. High Throughput Screen. 12, 383–396 Fechner, U. and Schneider, G. (2006) Flux (1): a virtual synthesis scheme for fragment-based de novo design. J. Chem. Inform. Model. 46, 699–707 Fechner, U. and Schneider, G. (2007) Flux (2): comparison of molecular mutation and crossover operators for ligand-based de novo design. J. Chem. Inform. Model. 47, 656–667 Lewell, X.Q. et al. (1998) RECAP – retrosynthetic combinatorial analysis procedure: a powerful new technique for identifying privileged molecular fragments with useful applications in combinatorial chemistry. J. Chem. Inform. Comput. Sci. 38, 511–522 Svetnik, V. et al. (2003) Random forest: a classification and regression tool for compound classification and QSAR modeling. J. Chem. Inform. Comput. Sci. 43, 1947–1958 Schmidt, M.F. and Rademann, J. (2009) Dynamic template-assisted strategies in fragment-based drug discovery. Trends Biotechnol. 27, 512–521 Jorgensen, W.L. et al. (2006) Computer-aided design of non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorg. Med. Chem. Lett. 16, 663–667 Renner, S. and Schneider, G. (2004) Fuzzy pharmacophore models from molecular alignments for correlation-vector-based virtual screening. J. Med. Chem. 47, 4653–4664 Holliday, J.D. et al. (2011) Multiple search methods for similarity-based virtual screening: analysis of search overlap and precision. J. Cheminform. 3, 29 Spoerri, A. (2008) Authority and ranking effects in data fusion. J. Am. Soc. Inform. Sci. Technol. 59, 450–460 Schneider, G. and So, S.S. (2001) Adaptive Systems in Drug Design. Landes Bioscience Guha, R. (2011) The ups and downs of structure-activity landscapes. Methods Mol. Biol. 672, 101–117 Hussain, J. and Rea, C. (2010) Computationally efficient algorithm to identify matched molecular pairs (MMPs) in large data sets. J. Chem. Inform. Model. 50, 339–348

e460

www.drugdiscoverytoday.com

Vol. 10, No. 4 2013

42 43 44 45

46

47

48

49 50 51 52

53 54

55

56 57

58

59 60 61 62

¨ ller, K. (2012) The power of MMPA and a teaching lesson in medicinal Mu chemistry. J. Med. Chem. 55, 1815–1816 Griffen, E. et al. (2011) Matched molecular pairs as a medicinal chemistry tool. J. Med. Chem. 54, 7739–7750 Nicolaou, C.A. et al. (2012) Multi-objective optimization methods in de novo drug design. Mini Rev. Med. Chem. (in press) Reutlinger, M. and Schneider, G. (2012) Nonlinear dimensionality reduction and mapping of compound libraries for drug discovery. J. Mol. Graphics Modell. 34, 108–117 Reutlinger, M. et al. (2011) Neighborhood-preserving visualization of adaptive structure-activity landscapes and application to drug discovery. Angew. Chem. Int. Ed. 50, 11633–11636 Dimova, D. et al. (2011) Design of multitarget activity landscapes that capture hierarchical activity cliff distributions. J. Chem. Inform. Model. 51, 258–266 Medina-Franco, J.L. et al. (2011) Multitarget structure-activity relationships characterized by activity-difference maps and consensus similarity measure. J. Chem. Inform.Inform. Model. 51, 2427–2439 Stouch, T.R. (ed.) (2012) The Next 25 Years. J. Comput-Aided Mol. Des. 26 Mauser, H. and Guba, W. (2008) Recent developments in de novo design and scaffold hopping. Curr. Opin. Drug Discov. Dev. 11, 365–374 Keiser, M.J. et al. (2007) Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 25, 197–206 Irwin, J.J. and Shoichet, B.K. (2005) ZINC – a free database of commercially available compounds for virtual screening. J. Chem. Inform. Model. 45, 177–182 Mancera, R.L. (2007) Molecular modeling of hydration in drug design. Curr. Opin. Drug Discov. Dev. 10, 275–280 Lloyd, D.G. et al. (2004) The effect of tightly bound water molecules on the structural interpretation of ligand-derived pharmacophore models. J. Comput. Aided Mol. Des. 18, 89–100 Garcı´a-Sosa, A.T. and Mancera, R.L. (2006) The effect of a tightly bound water molecule on scaffold diversity in the computer-aided de novo ligand design of CDK2 inhibitors. J. Mol. Model. 12, 422–431 Blaney, J. (2012) A very short history of structure-based design: how did we get here and where do we need to go? J Comput. Aided Mol. Des. 26, 13–14 Mazanetz, M.P. et al. (2011) Prediction of cyclin-dependent kinase 2 inhibitor potency using the fragment molecular orbital method. J. Cheminform. 3, 2 Nazare´, M. et al. (2012) Fragment deconstruction of small, potent factor Xa inhibitors: exploring the superadditivity energetics of fragment linking in protein–ligand complexes. Angew. Chem. Int. Ed. 51, 905–911 Keppner, S. et al. (2009) Identification and validation of a potent type II inhibitor of inactive polo-like kinase 1. ChemMedChem 4, 1806–1809 Tanrikulu, Y. et al. (2007) Scaffold hopping by fuzzy pharmacophores and its application to RNA targets. Chembiochem 8, 1932–1936 Schneider, P. and Schneider, G. (2003) Collection of bioactive reference compounds for focused library design. QSAR Comb. Sci. 22, 713–718 Hinton, G. and Roweis, S. (2002) Stochastic neighbor embedding. Adv. Neural Inform. Proc. Syst. 15, 833–840

De novo design - hop(p)ing against hope.

Current trends in computational de novo design provide a fresh approach to 'scaffold-hopping' in drug discovery. The methodological repertoire is no l...
1MB Sizes 3 Downloads 0 Views