Ligand efficiency as a guide in fragment hit selection and optimization



Similar documents
STRUCTURE-GUIDED, FRAGMENT-BASED LEAD GENERATION FOR ONCOLOGY TARGETS

Ligand efficiency indices for effective drug discovery

How To Understand Protein-Protein Interaction And Inhibitors

Data Visualization in Cheminformatics. Simon Xi Computational Sciences CoE Pfizer Cambridge

Ligand Efficiency and Lipophilicity

Malaria Journal. Open Access RESEARCH. Samuel Ayodele Egieyeh 1,2, James Syce 2, Sarel F. Malan 2 and Alan Christoffels 1*

Alterações empresariais sustentadas pelo conceito de engenharia do Produto Patrício Soares da Silva, MD, PhD

Accelerating Lead Generation: Emerging Technologies and Strategies

Strategies and tactics for optimizing the Hit-to-Lead process and beyond A computational chemistry perspective

Hit discovery and hit-to-lead approaches

Consensus Scoring to Improve the Predictive Power of in-silico Screening for Drug Design

Christian Lemmen HYDE: Scoring for Lead Optimization

How To Understand The Chemistry Of A 2D Structure

Scoring Functions and Docking. Keith Davies Treweren Consultants Ltd 26 October 2005

De novo design in the cloud from mining big data to clinical candidate

Thermodynamics guided lead discovery and optimization

Cheminformatics and Pharmacophore Modeling, Together at Last

QSAR. The following lecture has drawn many examples from the online lectures by H. Kubinyi

The Open PHACTS Discovery Platform Semantic data integration for Medicinal Chemists

Exploiting the Pathogen box

Consideration of Molecular Weight during Compound Selection in Virtual Target-Based Database Screening

A decade of fragment-based drug design: strategic advances and lessons learned

Nuclear Structure. particle relative charge relative mass proton +1 1 atomic mass unit neutron 0 1 atomic mass unit electron -1 negligible mass

Chem 115 POGIL Worksheet - Week 4 Moles & Stoichiometry

Infrared Spectroscopy: Theory

Integrating Medicinal Chemistry and Computational Chemistry: The Molecular Forecaster Approach

Molecular Spectroscopy

Nathan Brown. The Application of Consensus Modelling and Genetic Algorithms to Interpretable Discriminant Analysis.

FRAGMENT-BASED LEAD DISCOVERY

Cheminformatics and its Role in the Modern Drug Discovery Process

Review of Chemical Equilibrium 7.51 September free [A] (µm)

Separation of Amino Acids by Paper Chromatography

LEAD GENERATION APPROACHES IN DRUG DISCOVERY

Diabetes and Drug Development

Introduction to Netlogo: A Newton s Law of Gravity Simulation

Use of Predictive ADME in Library Profiling and Lead Optimization

HIGHLIGHTS OF THE SOCIETY FOR MEDICINES RESEARCH SYMPOSIUM: APPROACHES TO LEAD GENERATION

Assessing Checking the the reliability of protein-ligand structures

Lead generation and lead optimisation:

Spectrophotometry and the Beer-Lambert Law: An Important Analytical Technique in Chemistry

Biacore X BIACORE. The versatile high sensitivity system

Lead optimization services

Physicochemical Properties of Drugs

C o m p u te r M o d e lin g o f M o le c u la r E le c tro n ic S tru c tu re

Chapter 1: Chemistry: Measurements and Methods

How To Understand Protein Ligand Interaction

THE CAMBRIDGE CRYSTALLOGRAPHIC DATA CENTRE (CCDC)

History of the Atom & Atomic Theory

Keystone Exams: Chemistry Assessment Anchors and Eligible Content. Pennsylvania Department of Education

Computational Tools for Medicinal Chemists Increasing the Dimensions of Drug Discovery. Dr Robert Scoffin CEO

Use the Force! Noncovalent Molecular Forces

For convenience, we may consider an atom in two parts: the nucleus and the electrons.

10.7 Kinetic Molecular Theory Kinetic Molecular Theory. Kinetic Molecular Theory. Kinetic Molecular Theory. Kinetic Molecular Theory

We use Reaxys intensively for hit identification, hit-to-lead and lead optimization.

State of the art in reactive magnetron sputtering

Vacuum Evaporation Recap

What is a weak hydrogen bond?

PERMEABILITY MEASUREMENT

Supporting Information

Chem 115 POGIL Worksheet - Week 4 Moles & Stoichiometry Answers

Molecular basis of sweet taste in dipeptide taste ligands*

PERIODIC TABLE OF GROUPS OF ELEMENTS Elements can be classified using two different schemes.

m/z

標 準 納 期 2 週 間 を 誇 るKINOMEscan 受 託 試 験 : 報 告 書 から 抽 出 できる 更 なるデータ? 日 本 事 業 担 当 大 嶺 青 河 DiscoveRx Corporation

Proton Nuclear Magnetic Resonance ( 1 H-NMR) Spectroscopy

Lipid membrane physics

Molecular Docking. - Computational prediction of the structure of receptor-ligand complexes. Receptor: Protein Ligand: Protein or Small Molecule

Strategies for Identifying Students at Risk for USMLE Step 1 Failure

Eudendron: an Innovative Biotech Start-up

Chemistry 13: States of Matter

Methods for the Investigation of Protein-Ligand Complexes

Call 2014: High throughput screening of therapeutic molecules and rare diseases

Electronic Medical Record Customization and the Impact Upon Chart Completion Rates

Biomolecular Modelling

Name Lab #3: Solubility of Organic Compounds Objectives: Introduction: soluble insoluble partially soluble miscible immiscible

BBSRC TECHNOLOGY STRATEGY: TECHNOLOGIES NEEDED BY RESEARCH KNOWLEDGE PROVIDERS

AP CHEMISTRY 2009 SCORING GUIDELINES

Corporate Presentation November, 2013

electron does not become part of the compound; one electron goes in but two electrons come out.

SEMICONDUCTOR I: Doping, semiconductor statistics (REF: Sze, McKelvey, and Kittel)

Return to Lab Menu. Stoichiometry Exploring the Reaction between Baking Soda and Vinegar

Gold (Genetic Optimization for Ligand Docking) G. Jones et al. 1996

Chapter 4: Chemical and Solution Stoichiometry

CHEM 36 General Chemistry EXAM #1 February 13, 2002

The Mole Notes. There are many ways to or measure things. In Chemistry we also have special ways to count and measure things, one of which is the.

Chapter 14 - Acids and Bases

Unfolding and Aggregation of mabs Application Note NT-PR-005

Figure 1. A typical Laboratory Thermometer graduated in C.

Floyd Brownewell, Ph.D. Albany Molecular Research, Inc. 21 Corporate Circle PO Box Albany, NY

Molecular Formula Determination

Chapter 13 Solution Dynamics. An Introduction to Chemistry by Mark Bishop

Hot Spot Analysis for Driving the Development of Hits into Leads in Fragment-Based Drug Discovery

Combinatorial Chemistry and solid phase synthesis seminar and laboratory course

Welcome to Week 7. Chapter 11 Lead Optimization Introduction. Starting week seven video. Introduction to Chapter 11. Feedback cycle video

Lecture 11 Enzymes: Kinetics

Why? Intermolecular Forces. Intermolecular Forces. Chapter 12 IM Forces and Liquids. Covalent Bonding Forces for Comparison of Magnitude

1 The water molecule and hydrogen bonds in water

Molecular descriptors and chemometrics: a powerful combined tool for pharmaceutical, toxicological and environmental problems.

Robert Birdsall, Eoin Cosgrave, Henry Shion, and Weibin Chen Waters Corporation, Milford, MA, USA APPLICATION BENEFITS INTRODUCTION WATERS SOLUTIONS

Pesticide Analysis by Mass Spectrometry

Transcription:

Drug Discovery Today: Technologies Vol. 7, o. 3 2010 DRUG DISCVERY TDAY TECLGIES Editors-in-Chief Kelvin Lam arvard University, USA enk Timmerman Vrije Universiteit, The etherlands Fragment based drug discovery Ligand efficiency as a guide in fragment hit selection and optimization Sabine Schultes 1, Chris de Graaf 2, Eric E.J. aaksma 1,2, *, Iwan J.P. de Esch 2, Rob Leurs 2, liver Krämer 1 1 Boehringer Ingelheim RCV Gmb & Co. KG, Department of Medicinal Chemistry, Dr. Boehringer-Gasse 5-11, Vienna, Austria 2 Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 V Amsterdam, The etherlands Fragment-based screening (FBS) has become an established approach for hit identification. Starting points identified by FBS, are small fragments that require substantial modification to become leads. As fragments are different from classical hits a process tailored for fragment evolution is required. Scores for ligand efficiency have been proposed as guides for this process. ere we review how these have been applied to guide the selection and optimization of fragment hits. Introduction A common approach for hit identification is high-throughput screening (TS) [1]. In TS a large number of compounds (10 6 ) are screened to assess biological activity against a target. evertheless, considering the theoretically large chemical space of drug-like compounds [2], the probability of finding hits is relatively low [3]. This has led to the development of alternative approaches such as FBS and fragment-based drug discovery (FBDD) [4 15]. Advantages of FBS are (a) a more efficient sampling due to the smaller chemical space of fragment-sized compounds [16,17] and (b) a higher probability of fragments possessing good complementarity with the target [18]. Both aspects are likely to be the cause for the higher hit rates which are typically observed for FBS in comparison to TS [10]. owever, fragment hits have lower affinities towards the target. As a consequence, more effort has to be spent on *Corresponding author: Eric E.J. aaksma (eric.haaksma@boehringer-ingelheim.com) Section editor: Iwan de Esch Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The etherlands optimization to obtain lead compounds with an acceptable affinity. Strategies have been proposed to guide and evaluate this process. These strategies aim at the efficient optimization of fragments while maintaining their generally good physicochemical properties. In this review we discuss various efficiency indices and how to best leverage them in FBDD projects. Considerations during hit selection and optimization Traditionally, affinity is the first aspect considered for hit selection and optimization. owever, affinity alone can be misleading as it is often found to be linked with molecular size. Thus a focus on affinity leads to a bias towards a selection of bigger compounds. In addition, optimization of affinity during subsequent stages of drug discovery typically leads to a further increase in molecular weight (MW) [19]. Moreover, affinity is often optimized through the introduction of lipophilic groups, as these contribute favourably to the hydrophobic effect without the need for specific interactions with the target. This contrasts with polar groups, which need to establish very good interactions with the target to compensate the desolvation penalty. For this reason, polar groups are often used to improve solubility rather than affinity [20]. This phenomenon is reflected in the general trend towards generation of not only bigger but also more lipophilic compounds during the hit optimization process [19]. 1740-6749/$ ß 2010 Elsevier Ltd. All rights reserved. DI: 10.1016/j.ddtec.2010.11.003 e157

Drug Discovery Today: Technologies Fragment based drug discovery Vol. 7, o. 3 2010 Table 1. Comparison of fragment-like and drug-like compounds Type of compound Fragment-like Drug-like Rule Rule-of-Three * [34] Rule-of-Five [21] Thresholds MW <300 500 c log P 3 5 -bond donors 3 5 -bond acceptors 3 10 Typical values pic 50 4.4 ** 8 A 15 ** 38 Ligand efficiencies LE 0.38 0.29 FQ 0.55 0.81 * The authors suggest number of rotatable bonds 3 and polar surface area 60 as additional useful criteria. ** Median values taken from the literature examples (see supplementary material). Ultimately, affinity for the target is not the only aspect that has to be considered during drug discovery. To enter later stages of drug development, a compound needs to have suitable physicochemical properties. Lipinski et al. [21] studied the properties of oral drugs that managed to enter clinical Phase II. The study resulted in the Rule-of-Five stating that poor absorption or permeation is more likely if more than one of the relevant parameters (see Table 1) are outside the range typically observed for drug-like compounds. The Rule-of-Five has had a strong influence on the drug discovery process. Good physicochemical properties help to reduce the attrition rate at later stages towards to market [22]. A recent study showed that almost all ADMET parameters deteriorate with either increasing MW and/or log P [23]. These studies emphasize the importance for selecting appropriate hits and monitoring MW and lipophilicity in addition to affinity during hit optimization. Ligand efficiency scores Ligand efficiency To escape the affinity-biased selection and optimization towards larger ligands the focus should be directed towards the generation of compounds that use their atoms most efficiently. To estimate the efficiency of compounds, opkins et al. [24] recommended to assess binding affinity in relation to the number of heavy atoms in a molecule and introduced the term ligand efficiency (LE) LE ¼ DG A where DG is the free energy of binding and A is the number of non-hydrogen atoms of the ligand. Instead of considering the affinity of the whole compound, the average affinity contribution per atom is taken into account. This provides a way to compare the affinity of molecules corrected for their size. Abad-Zapatero and Metz [25] introduced the binding efficiency index (BEI) defined as BEI = pic 50 /MW as an alternative metric. Prioritizing hits according to their LE allows also smaller low affinity compounds to be attractive for further optimization. Group efficiency Verdonk and Rees [26] introduced group efficiency (GE) as a metric to compare the quality of added groups. It is defined as GE ¼ DDG DA DDG ¼ DGðBÞ DDGðAÞ DA ¼ AðBÞ AðAÞ where the affinity gained by molecule B, through the introduction of additional non-hydrogen atoms DA to molecule A, is expressed as the difference of the free energies of binding ( DDG). The group efficiency describes the average affinity gain contributed by each atom of an added group. nly the addition of groups with the same (or a better) group efficiency, compared to the LE of the initial molecule A, will allow to maintain (or increase) the LE during compound optimization. Fit quality Reynolds et al. systematically investigated the size-dependence of LE [27]. In their study, the binding affinity data and corresponding LEs taken from the BindingDB [28] were plotted against the number on non-hydrogen atoms. Altogether, over 8000 ligands for 28 targets were considered. The result of their study is that the maximum observed ligand efficiency decreases with molecular size. The authors conclude that LE cannot be evaluated independent of the molecular size. To enable a size-independent comparison of ligands they derived a scaling function (LE_Scale) by fitting an exponential function to the maximal ligand efficiency values observed for a given A count. Dividing the actual observed ligand efficiency by the calculated maximal achievable ligand efficiency (the scaling function) results in a scaled ligand efficiency called fit quality (FQ): LE FQ ¼ LE Scale Different ways to obtain the scaling function are published [29 31]. evertheless, independent of how the scaling function is derived, FQ values near one indicate near optimal ligand binding. Ligand-lipophilicity efficiency LE and corresponding FQ are useful for optimizing affinity with respect to molecular size. owever, to achieve optimal e158

Vol. 7, o. 3 2010 Drug Discovery Today: Technologies Fragment based drug discovery ADMET properties molecular size and lipophilicity are important factors to consider. If lipophilicity is too high, the likelihood of a compound to bind to multiple targets increases [32]. To facilitate optimization of affinity with respect to lipophilicity, Leeson and Springthorpe [32] defined the ligand-lipophilicity efficiency (LLE): LLE ¼ pic 50 clogp igh LLE favours compounds that gain a lot of their affinity through directed interactions thus making the interaction with the receptor more specific. While one can say that LLE describes how efficient a ligand exploits its lipophilicity, no explicit measure of molecular size is used. Therefore, a lipophilicity corrected LE is needed to enable optimization of affinity without the extensive use of lipophilic nonspecific interactions. Keseru and Makara [19] proposed not only LELP = log P/LE as monitoring function to achieve that goal, but also other ways to combine molecular size and lipophilicity into a single efficiency measurement are being discussed [33]. Application of ligand efficiency scores to FBDD Clinical candidates are generally preferred to be Rule-of-Five compliant with a special focus on lipophilicity. To achieve this, the starting restrictions for fragments should be obviously stronger. Congreve et al. [34] studied fragment hits that could be successfully optimized into potent leads, and noticed they have particular physicochemical properties. These properties and congruent thresholds are summarized as the Rule-of-Three (see Table 1). To stay within these thresholds has been suggested as a criterion for fragment library design [34]. For fragment hit selection, LE has become a widely accepted metric. In general, it is best to start with a fragment that shows a high LE because in most cases LE decreases during optimization. Looking at the examples in the literature [5 8,14] and following the evolution of LE, there are fewer examples where LE could be maintained or even increased. In the majority of examples (70%, see suppl. material), LE decreases during fragment optimization (by either fragment linking or growth). Therefore, starting with a highly efficient fragment hit makes it easier to optimize the fragment into a drug-like compound. In general, an orally available clinical candidate possesses a potency of better than 10 nm and, if Rule-of-Five -compliant, a maximal molecular weight of 500 Da (which equals, on average, 38 A). This means that a LE of at least 0.29 kcal mol 1 A 1 needs to be maintained during hit optimization. Screening only Rule-of-Three -compliant fragments ensures the identification of good starting points for optimization if an affinity in the range of 1 mm can be achieved. For a fragment hit with less than 300 Da (that equals on average 23A) this would result in a LE of at least 0.36 kcal mol 1 A 1. Looking at examples from the literature (see suppl. material) such a high affinity cannot always be achieved (the median pic 50 is 4.35), but because the average fragment hit is also smaller than 23 A (median number of A is 15), the median LE of fragment hits considered here is 0.38 kcal mol 1 A 1. Therefore, some loss of LE during optimization is acceptable in most cases. This is illustrated in the example (a) in Fig. 1: the LE score of the initial fragment hit is quite high (0.59). Although, LE decreases during fragment growth, the final potent compound (IC 50 = 3 nm) reaches a LE score of 0.42. This is still a high value which is significantly above the suggested value of about 0.3. Another possible scenario is illustrated by example (b): The LE score can be maintained throughout the optimization process. This is achieved through the introduction of groups that have GEs comparable to LE of the starting compound. As long as the initial fragment hit has a LE score > 0.3, also this strategy can lead to potent drug-like compounds. Example (c) shows one of the rare cases where LE is increased during fragment growth. Although the LE of the starting fragment is below 0.3, it was possible to significantly improve the affinity by introducing an additional group to finally reach a potent drug-like compound. Revisiting example (a) illustrates the usefulness of FQ (in addition to LE) for fragment optimization. Although LE is decreasing, FQ is maintained, indicating that fragment optimization is on the right track. In general, the goal should be to either maintain or increase FQ during fragment assembly to reach a near optimal affinity for the final compound. LLE provides a way to evaluate the affinity of a compound with respect to its lipophilicity. The challenge is to increase potency without increasing lipophilicity at the same time. As lipophilicity is the major factor for promiscuity of compounds, LLE optimized compounds should be more selective. It is suggested to target a LLE in a range of 5 7 or even higher [32]. In example (a) LLE is increased during optimization. The final compound reaches a LLE of 7.3 which is even above the suggested range of 5 7. In combination with the acceptable LE of 0.42 this indicates that this compound was successfully optimized. Comparing the c log P values of the compounds reveals that lipophilicity was kept fairly constant during fragment growth. This means that affinity is mainly gained by the introduction of groups making specific directed interactions. In the other two examples, (b) and (c) LLE is increasing during optimization but none of the compounds reaches a LLE above 5. In these cases the gain of affinity is accompanied by an increase of lipophilicity. In this respect, optimization was not as optimal as in the first example. e159

Drug Discovery Today: Technologies Fragment based drug discovery Vol. 7, o. 3 2010 (a) GE=0.38 GE=0.29 A=14 MW=184.20 clogp=1.89 IC 50 =0.91µM LE=0.59 FQ=0.80 LLE=4.15 A=18 A=23 A=28 MW=241.25 MW=303.3 MW=381.44 clogp=1.13 clogp=2.98 clogp=1.21 IC 50 =0.07µM IC 50 =0.0059µM IC 50 =0.003µM LE=0.55 FQ=0.83 LLE=6.02 LE=0.49 FQ=0.86 LLE=5.25 LE=0.42 FQ=0.85 LLE=7.31 (b) GE=0.53 GE=0.41 A=12 MW=168.20 clogp=0.97 IC 50 =60µM LE=0.48 FQ=0.61 LLE=3.25 A=18 MW=244.30 clogp=2.75 IC 50 =0.27µM LE=0.50 FQ=0.76 LLE=3.82 2 A=21 MW=289.29 clogp=2.69 IC 50 =0.033µM LE=0.48 FQ=0.81 LLE=4.79 (c) 2 A=15 MW=200.24 clogp=2.09 IC 50 =1300µM LE=0.26 FQ=0.36 LLE=0.80 GE=0.37 Cl A=31 MW=441.89 clogp=4.05 IC 50 =0.065µM LE=0.32 FQ=0.71 LLE=3.14 F Drug Discovery Today: Technologies Figure 1. Examples of successful fragment-based drug discovery campaigns (a) Aurora [38], (b) PDE4B [39], and (c) p38a [40,41]. Conclusions FBDD, as illustrated in Table 1, typically starts with a Rule-of- Three -compliant fragment and ideally ends up with a potent Rule-of-Five -compliant candidate compound. Colleagues from Astex proposed that an efficient fragment growth is one where LE can be maintained [4]. This goal is further supported by a study which concluded that a linear increase of binding affinity with molecular mass is possible [35]. If maintenance of LE at an acceptable level (LE 0.3) can be achieved, FQ scores will rise during fragment growth and the affinity of the final compound will approach a near optimal affinity. At the same time, retrospective studies show that in most cases LE scores decrease during fragment assembly [5,8]. Still, an acceptable affinity of the final compound can be reached if FQ can be maintained at a high level (FQ 0.8). Therefore, Bembenek et al. [36] suggested that, unlike LE alone, the FQ score can be used as a measure of efficiency across the entire optimization process from initial fragment hit to optimized clinical candidate. Another mentionable guide to maintain the good physicochemical properties of fragment hits is to consider LLE during FBDD. Lipinski states in the Rule-of-Five that the c log P, which is used to calculate the LLE, should be smaller e160

Vol. 7, o. 3 2010 Drug Discovery Today: Technologies Fragment based drug discovery Table 2. Summary of ligand efficiency scores to be considered during FBDD Parameter Definition Focus during fragment hit selection leads to Ligand efficiency LE = DDG/A Bias towards smaller compounds Fit quality FQ = LE/LE_Scale Size independent selection of efficient compounds Ligand-lipophilicity efficiency LLE = pic 50 log P Selection of more specific compounds Recommended range for fragment hits LE 0.3 Aim during fragment optimization Try to maintain (decrease acceptable for starting fragments with LE 0.3) FQ 0.8 Maintain at high level or increase to FQ 1 LLE 3 Maintain at high level or increase to LLE > 5 7 than 5. owever, a 10 nm compound with an acceptable c log P of 5 will have a LLE of 3. This is much smaller than the suggested range of 5 7 [32]. To achieve a LLE in this range, the c log P must be smaller than 3. This is in agreement with a recent study which showed that there is an increased risk of adverse outcome with c log P > 3[37]. Table 2 summarizes the ligand efficiency scores that should be considered during FBDD. Both LE and FQ have been very helpful in guiding the selection and optimization of fragment hits. In addition, LLE is expected to become increasingly popular to ensure an increase of affinity more than lipophilicity. Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.ddtec.2010.11.003. References 1 Goodnow, R.A., Jr (2006) it and lead identification: integrated technology-based approaches. Drug Discov. Today: Technol. 3, 367 375 2 Bohacek, R.S. et al. (1996) The art and practice of structure-based drug design: a molecular modeling perspective. Med. Res. Rev. 16, 3 50 3 Dobson, C.M. (2004) Chemical space and biology. ature 432, 824 828 4 Carr, R.A. et al. (2005) Fragment-based lead discovery: leads by design. Drug Discov. Today 10, 987 992 5 Congreve, M. et al. (2008) Recent developments in fragment-based drug discovery. J. Med. Chem. 51, 3661 3680 6 de Kloe, G.E. et al. (2009) Transforming fragments into candidates: small becomes big in medicinal chemistry. Drug Discov. Today 14, 630 646 7 Erlanson, D.A. et al. (2004) Fragment-based drug discovery. J. Med. Chem. 47, 3463 3482 8 Erlanson, D.A. (2006) Fragment-based lead discovery: a chemical update. Curr. pin. Biotechnol. 17, 643 652 9 ajduk, P.J. (2006) SAR by MR: putting the pieces together. Mol. Interv. 6, 266 272 10 ajduk, P.J. and Greer, J. (2007) A decade of fragment-based drug design: strategic advances and lessons learned. at. Rev. Drug Discov. 6, 211 219 11 Jhoti,. et al. (2007) Fragment-based screening using X-ray crystallography and MR spectroscopy. Curr. pin. Chem. Biol. 11, 485 493 12 Leach, A.R. et al. (2006) Fragment screening: an introduction. Mol. Biosyst. 2, 430 446 13 rita, M. et al. (2010) Advances in fragment-based drug discovery platforms. Expert pin. Drug Discov. 4, 1125 1144 14 Rees, D.C. et al. (2004) Fragment-based lead discovery. at. Rev. Drug Discov. 3, 660 672 15 Zartler, E.R. and Shapiro, M.J. (2005) Fragonomics: fragment-based drug discovery. Curr. pin. Chem. Biol. 9, 366 370 16 Fink, T. et al. (2005) Virtual exploration of the small-molecule chemical universe below 160 Daltons. Angew. Chem. Int. Ed. Engl. 44, 1504 1508 17 Fink, T. and Reymond, J.L. (2007) Virtual exploration of the chemical universe up to 11 atoms of C,,, F: assembly of 26. 4 million structures (110. 9 million stereoisomers) and analysis for new ring systems, stereochemistry, physicochemical properties, compound classes, and drug discovery. J. Chem. Inf. Model. 47, 342 353 18 ann, M.M. et al. (2001) Molecular complexity and its impact on the probability of finding leads for drug discovery. J. Chem. Inf. Comput. Sci. 41, 856 864 19 Keseru, G.M. and Makara, G.M. (2009) The influence of lead discovery strategies on the properties of drug candidates. at. Rev. Drug Discov. 8, 203 212 20 Ruben, A.J. et al. (2006) vercoming roadblocks in lead optimization: a thermodynamic perspective. Chem. Biol. Drug Des. 67, 2 4 21 Lipinski, C.A. et al. (1997) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Del. Rev. 23, 3 25 22 Wenlock, M.C. et al. (2003) A comparison of physiochemical property profiles of development and marketed oral drugs. J. Med. Chem. 46, 1250 1256 23 Gleeson, M.P. (2008) Generation of a set of simple, interpretable ADMET rules of thumb. J. Med. Chem. 51, 817 834 24 opkins, A.L. et al. (2004) Ligand efficiency: a useful metric for lead selection. Drug Discov. Today 9, 430 431 25 Abad-Zapatero, C. and Metz, J.T. (2005) Ligand efficiency indices as guideposts for drug discovery. Drug Discov. Today 10, 464 469 26 Verdonk, M.L. and Rees, D.C. (2008) Group efficiency: a guideline for hitsto-leads chemistry. ChemMedChem 3, 1179 1180 27 Reynolds, C.. et al. (2007) The role of molecular size in ligand efficiency. Bioorg. Med. Chem. Lett. 17, 4258 4261 28 Goodnow, J.J. et al. (1959) The role of past events in problem solving. J. Exp. Psychol. 58, 456 461 29 Reynolds, C.. et al. (2008) Ligand binding efficiency: trends, physical basis, and implications. J. Med. Chem. 51, 2432 2438 30 issink, J.W. (2009) Simple size-independent measure of ligand efficiency. J. Chem. Inf. Model. 49, 1617 1622 31 rita, M. et al. (2009) Golden Ratio indices in fragment-based drug discovery. Drug Discov. Today 14, 321 328 32 Leeson, P.D. and Springthorpe, B. (2007) The influence of drug-like concepts on decision-making in medicinal chemistry. at. Rev. Drug Discov. 6, 881 890 33 Kenny P. Scaling potency by lipophilicity and molecular size. http://fbddlit.blogspot.com/2009/06/scaling-potency-by-lipophilicity-and.html. 2009. Ref Type: Internet Communication 34 Congreve, M. et al. (2003) A rule of three for fragment-based lead discovery? Drug Discov. Today 8, 876 877 35 ajduk, P.J. (2006) Fragment-based drug design: how big is too big? J. Med. Chem. 49, 6972 6976 36 Bembenek, S.D. et al. (2009) Ligand efficiency and fragment-based drug discovery. Drug Discov. Today 14, 278 283 37 ughes, J.D. et al. (2008) Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorg. Med. Chem. Lett. 18, 4872 4875 e161

Drug Discovery Today: Technologies Fragment based drug discovery Vol. 7, o. 3 2010 38 oward, S. et al. (2009) Fragment-based discovery of the pyrazol-4-yl urea (AT9283), a multitargeted kinase inhibitor with potent aurora kinase activity. J. Med. Chem. 52, 379 388 39 Card, G.L. et al. (2005) A family of phosphodiesterase inhibitors discovered by cocrystallography and scaffold-based drug design. at. Biotechnol. 23, 201 207 40 Gill, A. et al. (2005) The discovery of novel protein kinase inhibitors by using fragment-based high-throughput X-ray crystallography. Chembiochem 6, 506 512 41 Gill, A.L. et al. (2005) Identification of novel p38a MAP kinase inhibitors using fragment-based lead generation. J. Med. Chem. 48, 414 426 e162