Druggability

Last updated

Druggability is a term used in drug discovery to describe a biological target (such as a protein) that is known to or is predicted to bind with high affinity to a drug. Furthermore, by definition, the binding of the drug to a druggable target must alter the function of the target with a therapeutic benefit to the patient. The concept of druggability is most often restricted to small molecules (low molecular weight organic substances) [1] but also has been extended to include biologic medical products such as therapeutic monoclonal antibodies.

Contents

Drug discovery comprises a number of stages that lead from a biological hypothesis to an approved drug. Target identification is typically the starting point of the modern drug discovery process. Candidate targets may be selected based on a variety of experimental criteria. These criteria may include disease linkage (mutations in the protein are known to cause a disease), mechanistic rationale (for example, the protein is part of a regulatory pathway that is involved in the disease process), or genetic screens in model organisms. [2] Disease relevance alone however is insufficient for a protein to become a drug target. In addition, the target must be druggable.

Prediction of druggability

If a drug has already been identified for a target, that target is by definition druggable. If no known drugs bind to a target, then druggability is implied or predicted using different methods that rely on evolutionary relationships, 3D-structural properties or other descriptors. [3]

Precedence-based

A protein is predicted to be "druggable" if it is a member of a protein family [4] for which other members of the family are known to be targeted by drugs (i.e., "guilt" by association). While this is a useful approximation of druggability, this definition has limitations for two main reasons: (1) it highlights only historically successful proteins, ignoring the possibility of a perfectly druggable, but yet undrugged protein family; and (2) assumes that all protein family members are equally druggable.[ citation needed ]

Structure-based

This relies on the availability of experimentally determined 3D structures or high quality homology models. A number of methods exist for this assessment of druggability but all of them consist of three main components: [5] [6] [7] [8]

  1. Identifying cavities or pockets on the structure
  2. Calculating physicochemical and geometric properties of the pocket
  3. Assessing how these properties fit a training set of known druggable targets, typically using machine learning algorithms

Early work on introducing some of the parameters of structure-based druggability came from Abagyan and coworkers [9] and then Fesik and coworkers, [10] the latter by assessing the correlation of certain physicochemical parameters with hits from an NMR-based fragment screen. There has since been a number of publications reporting related methodologies. [5] [11] [12]

There are several commercial tools and databases for structure-based druggability assessment. A publicly available database of pre-calculated druggability assessments for all structural domains within the Protein Data Bank (PDB) is provided through the ChEMBL's DrugEBIlity portal. [13]

Structure-based druggability is usually used to identify suitable binding pocket for a small molecule; however, some studies have assessed 3D structures for the availability of grooves suitable for binding helical mimetics. [14] This is an increasingly popular approach in addressing the druggability of protein-protein interactions. [15]

Predictions based on other properties

As well as using 3D structure and family precedence, it is possible to estimate druggability using other properties of a protein such as features derived from the amino-acid sequence (feature-based druggability) [3] which is applicable to assessing small-molecule based druggability or biotherapeutic-based druggability or the properties of ligands or compounds known to bind the protein (Ligand-based druggability). [16] [17]

The importance of training sets

All methods for assessing druggability are highly dependent on the training sets used to develop them. This highlights an important caveat in all the methods discussed above: which is that they have learned from the successes so far. The training sets are typically either databases of curated drug targets; [18] [19] screened targets databases (ChEMBL, BindingDB, PubChem etc.); or on manually compiled sets of 3D structure known by the developers to be druggable. As training sets improve and expand, the boundaries of druggability may also be expanded.

Undruggable targets

About 3% of human proteins are known to be "mode of action" drug targets, i.e., proteins through which approved drugs act. [20] Another 7% of the human proteins interact with small molecule chemicals. [20] Based on DrugCentral, 1795 human proteins annotated to interact with 2455 approved drugs. [21]

Furthermore, it is estimated that only 10-15% of human proteins are disease modifying while only 10-15% are druggable (there is no correlation between the two), meaning that only between 1-2.25% of disease modifying proteins are likely to be druggable. Hence it appears that the number of new undiscovered drug targets is very limited. [22] [23] [24]

A potentially much larger percentage of proteins could be made druggable if protein–protein interactions could be disrupted by small molecules. However the majority of these interactions occur between relatively flat surfaces of the interacting protein partners and it is very difficult for small molecules to bind with high affinity to these surfaces. [25] [26] Hence these types of binding sites on proteins are generally thought to be undruggable but there has been some progress (by 2009) targeting these sites. [27] [28]

Chemoproteomics techniques have recently expanded the scope of what is deemed a druggable target through the identification of covalently modifiable sites across the proteome. [29]

Related Research Articles

<span class="mw-page-title-main">Allosteric regulation</span> Regulation of enzyme activity

In biochemistry, allosteric regulation is the regulation of an enzyme by binding an effector molecule at a site other than the enzyme's active site.

<span class="mw-page-title-main">Binding site</span> Molecule-specific coordinate bonding area in biological systems

In biochemistry and molecular biology, a binding site is a region on a macromolecule such as a protein that binds to another molecule with specificity. The binding partner of the macromolecule is often referred to as a ligand. Ligands may include other proteins, enzyme substrates, second messengers, hormones, or allosteric modulators. The binding event is often, but not always, accompanied by a conformational change that alters the protein's function. Binding to protein binding sites is most often reversible, but can also be covalent reversible or irreversible.

<span class="mw-page-title-main">Drug design</span> Invention of new medications based on knowledge of a biological target

Drug design, often referred to as rational drug design or simply rational design, is the inventive process of finding new medications based on the knowledge of a biological target. The drug is most commonly an organic small molecule that activates or inhibits the function of a biomolecule such as a protein, which in turn results in a therapeutic benefit to the patient. In the most basic sense, drug design involves the design of molecules that are complementary in shape and charge to the biomolecular target with which they interact and therefore will bind to it. Drug design frequently but not necessarily relies on computer modeling techniques. This type of modeling is sometimes referred to as computer-aided drug design. Finally, drug design that relies on the knowledge of the three-dimensional structure of the biomolecular target is known as structure-based drug design. In addition to small molecules, biopharmaceuticals including peptides and especially therapeutic antibodies are an increasingly important class of drugs and computational methods for improving the affinity, selectivity, and stability of these protein-based therapeutics have also been developed.

<span class="mw-page-title-main">Molecular recognition</span> Type of non-covalent bonding

The term molecular recognition refers to the specific interaction between two or more molecules through noncovalent bonding such as hydrogen bonding, metal coordination, hydrophobic forces, van der Waals forces, π-π interactions, halogen bonding, or resonant interaction effects. In addition to these direct interactions, solvents can play a dominant indirect role in driving molecular recognition in solution. The host and guest involved in molecular recognition exhibit molecular complementarity. Exceptions are molecular containers, including e.g. nanotubes, in which portals essentially control selectivity.

A biological target is anything within a living organism to which some other entity is directed and/or binds, resulting in a change in its behavior or function. Examples of common classes of biological targets are proteins and nucleic acids. The definition is context-dependent, and can refer to the biological target of a pharmacologically active drug compound, the receptor target of a hormone, or some other target of an external stimulus. Biological targets are most commonly proteins such as enzymes, ion channels, and receptors.

<span class="mw-page-title-main">Ligand (biochemistry)</span> Substance that forms a complex with a biomolecule

In biochemistry and pharmacology, a ligand is a substance that forms a complex with a biomolecule to serve a biological purpose. The etymology stems from Latin ligare, which means 'to bind'. In protein-ligand binding, the ligand is usually a molecule which produces a signal by binding to a site on a target protein. The binding typically results in a change of conformational isomerism (conformation) of the target protein. In DNA-ligand binding studies, the ligand can be a small molecule, ion, or protein which binds to the DNA double helix. The relationship between ligand and binding partner is a function of charge, hydrophobicity, and molecular structure.

<span class="mw-page-title-main">Docking (molecular)</span> Prediction method in molecular modeling

In the field of molecular modeling, docking is a method which predicts the preferred orientation of one molecule to a second when a ligand and a target are bound to each other to form a stable complex. Knowledge of the preferred orientation in turn may be used to predict the strength of association or binding affinity between two molecules using, for example, scoring functions.

The Structural Genomics Consortium (SGC) is a public-private-partnership focusing on elucidating the functions and disease relevance of all proteins encoded by the human genome, with an emphasis on those that are relatively understudied. The SGC places all its research output into the public domain without restriction and does not file for patents and continues to promote open science. Two recent publications revisit the case for open science. Founded in 2003, and modelled after the Single Nucleotide Polymorphism Database (dbSNP) Consortium, the SGC is a charitable company whose Members comprise organizations that contribute over $5,4M Euros to the SGC over a five-year period. The Board has one representative from each Member and an independent Chair, who serves one 5-year term. The current Chair is Anke Müller-Fahrnow (Germany), and previous Chairs have been Michael Morgan (U.K.), Wayne Hendrickson (U.S.A.), Markus Gruetter (Switzerland) and Tetsuyuki Maruyama (Japan). The founding and current CEO is Aled Edwards (Canada). The founding Members of the SGC Company were the Canadian Institutes of Health Research, Genome Canada, the Ontario Research Fund, GlaxoSmithKline and Wellcome Trust. The current Members comprise Bayer Pharma AG, Bristol Myers Squibb, Boehringer Ingelheim, the Eshelman Institute for Innovation, Genentech, Genome Canada, Janssen, Merck KGaA, Pfizer, and Takeda.

<span class="mw-page-title-main">Virtual screening</span>

Virtual screening (VS) is a computational technique used in drug discovery to search libraries of small molecules in order to identify those structures which are most likely to bind to a drug target, typically a protein receptor or enzyme.

In the fields of computational chemistry and molecular modelling, scoring functions are mathematical functions used to approximately predict the binding affinity between two molecules after they have been docked. Most commonly one of the molecules is a small organic compound such as a drug and the second is the drug's biological target such as a protein receptor. Scoring functions have also been developed to predict the strength of intermolecular interactions between two proteins or between protein and DNA.

Molecular binding is an attractive interaction between two molecules that results in a stable association in which the molecules are in close proximity to each other. It is formed when atoms or molecules bind together by sharing of electrons. It often, but not always, involves some chemical bonding.

Fragment-based lead discovery (FBLD) also known as fragment-based drug discovery (FBDD) is a method used for finding lead compounds as part of the drug discovery process. Fragments are small organic molecules which are small in size and low in molecular weight. It is based on identifying small chemical fragments, which may bind only weakly to the biological target, and then growing them or combining them to produce a lead with a higher affinity. FBLD can be compared with high-throughput screening (HTS). In HTS, libraries with up to millions of compounds, with molecular weights of around 500 Da, are screened, and nanomolar binding affinities are sought. In contrast, in the early phase of FBLD, libraries with a few thousand compounds with molecular weights of around 200 Da may be screened, and millimolar affinities can be considered useful. FBLD is a technique being used in research for discovering novel potent inhibitors. This methodology could help to design multitarget drugs for multiple diseases. The multitarget inhibitor approach is based on designing an inhibitor for the multiple targets. This type of drug design opens up new polypharmacological avenues for discovering innovative and effective therapies. Neurodegenerative diseases like Alzheimer’s (AD) and Parkinson’s, among others, also show rather complex etiopathologies. Multitarget inhibitors are more appropriate for addressing the complexity of AD and may provide new drugs for controlling the multifactorial nature of AD, stopping its progression.

<span class="mw-page-title-main">Cell surface receptor</span> Class of ligand activated receptors localized in surface of plama cell membrane

Cell surface receptors are receptors that are embedded in the plasma membrane of cells. They act in cell signaling by receiving extracellular molecules. They are specialized integral membrane proteins that allow communication between the cell and the extracellular space. The extracellular molecules may be hormones, neurotransmitters, cytokines, growth factors, cell adhesion molecules, or nutrients; they react with the receptor to induce changes in the metabolism and activity of a cell. In the process of signal transduction, ligand binding affects a cascading chemical change through the cell membrane.

<span class="mw-page-title-main">Stapled peptide</span>

A stapled peptide is a short peptide, typically in an alpha-helical conformation, that is constrained by a synthetic brace ("staple"). The staple is formed by a covalent linkage between two amino acid side-chains, forming a peptide macrocycle. Staples, generally speaking, refer to a covalent linkage of two previously independent entities. Peptides with multiple, tandem staples are sometimes referred to as stitched peptides. Among other applications, peptide stapling is notably used to enhance the pharmacologic performance of peptides.

<span class="mw-page-title-main">Targeted covalent inhibitors</span>

Targeted covalent inhibitors (TCIs) or Targeted covalent drugs are rationally designed inhibitors that bind and then bond to their target proteins. These inhibitors possess a bond-forming functional group of low chemical reactivity that, following binding to the target protein, is positioned to react rapidly with a proximate nucleophilic residue at the target site to form a bond.

Chemoproteomics entails a broad array of techniques used to identify and interrogate protein-small molecule interactions. Chemoproteomics complements phenotypic drug discovery, a paradigm that aims to discover lead compounds on the basis of alleviating a disease phenotype, as opposed to target-based drug discovery, in which lead compounds are designed to interact with predetermined disease-driving biological targets. As phenotypic drug discovery assays do not provide confirmation of a compound's mechanism of action, chemoproteomics provides valuable follow-up strategies to narrow down potential targets and eventually validate a molecule's mechanism of action. Chemoproteomics also attempts to address the inherent challenge of drug promiscuity in small molecule drug discovery by analyzing protein-small molecule interactions on a proteome-wide scale. A major goal of chemoproteomics is to characterize the interactome of drug candidates to gain insight into mechanisms of off-target toxicity and polypharmacology.

Molecular Operating Environment (MOE) is a drug discovery software platform that integrates visualization, modeling and simulations, as well as methodology development, in one package. MOE scientific applications are used by biologists, medicinal chemists and computational chemists in pharmaceutical, biotechnology and academic research. MOE runs on Windows, Linux, Unix, and macOS. Main application areas in MOE include structure-based design, fragment-based design, ligand-based design, pharmacophore discovery, medicinal chemistry applications, biologics applications, structural biology and bioinformatics, protein and antibody modeling, molecular modeling and simulations, virtual screening, cheminformatics & QSAR. The Scientific Vector Language (SVL) is the built-in command, scripting and application development language of MOE.

RNA-targeting small molecules represent a class of small molecules, organic compounds with traditional drug properties that can bind to RNA secondary or tertiary structures and alter translation patterns, localization, and degradation.

Molecular glue refers to a class of chemical compounds or molecules that play a crucial role in binding and stabilizing protein-protein interactions in biological systems. These molecules act as "glue" by enhancing the affinity between proteins, ultimately influencing various cellular processes. Molecular glue compounds have gained significant attention in the fields of drug discovery, chemical biology, and fundamental research due to their potential to modulate protein interactions, and thus, impact various cellular pathways. They have unlocked avenues in medicine previously thought to be “undruggable.”

Angela N. Koehler is an American biochemist who is the Karl Van Tassel (1925) Career Development Professor of Chemical Biology at the Broad Institute. Her research considers the development of chemical tools to understand transcriptional regulation, and the design of next-generation pharmaceuticals.

References

  1. Owens J (2007). "Determining druggability". Nature Reviews Drug Discovery. 6 (3): 187. doi: 10.1038/nrd2275 .
  2. Dixon SJ, Stockwell BR (December 2009). "Identifying druggable disease-modifying gene products". Current Opinion in Chemical Biology. 13 (5–6): 549–555. doi:10.1016/j.cbpa.2009.08.003. PMC   2787993 . PMID   19740696.
  3. 1 2 Al-Lazikani B, Gaulton A, Paolini G, Lanfear J, Overington J, Hopkins A (2007). "The Molecular Basis of Predicting Druggability". In Wess G, Schreiber SL, Kapoor TM (eds.). Chemical Biology: From Small Molecules to Systems Biology and Drug Design. Vol. 1–3. Weinheim: Wiley-VCH. pp. 804–823. ISBN   978-3-527-31150-7.
  4. Hopkins AL, Groom CR (September 2002). "The druggable genome". Nature Reviews. Drug Discovery. 1 (9): 727–730. doi:10.1038/nrd892. PMID   12209152. S2CID   13166282.
  5. 1 2 Halgren TA (February 2009). "Identifying and characterizing binding sites and assessing druggability". Journal of Chemical Information and Modeling. 49 (2): 377–389. doi:10.1021/ci800324m. PMID   19434839.
  6. Nayal M, Honig B (June 2006). "On the nature of cavities on protein surfaces: application to the identification of drug-binding sites". Proteins. 63 (4): 892–906. doi:10.1002/prot.20897. PMID   16477622. S2CID   23887061.
  7. Seco J, Luque FJ, Barril X (April 2009). "Binding site detection and druggability index from first principles". Journal of Medicinal Chemistry. 52 (8): 2363–2371. doi:10.1021/jm801385d. PMID   19296650.
  8. Bakan A, Nevins N, Lakdawala AS, Bahar I (July 2012). "Druggability Assessment of Allosteric Proteins by Dynamics Simulations in the Presence of Probe Molecules". Journal of Chemical Theory and Computation. 8 (7): 2435–2447. doi:10.1021/ct300117j. PMC   3392909 . PMID   22798729.
  9. An J, Totrov M, Abagyan R (2004). "Comprehensive identification of "druggable" protein ligand binding sites". Genome Informatics. International Conference on Genome Informatics. 15 (2): 31–41. PMID   15706489.
  10. Hajduk PJ, Huth JR, Fesik SW (April 2005). "Druggability indices for protein targets derived from NMR-based screening data". Journal of Medicinal Chemistry. 48 (7): 2518–2525. doi:10.1021/jm049131r. PMID   15801841.
  11. Schmidtke P, Barril X (August 2010). "Understanding and predicting druggability. A high-throughput method for detection of drug binding sites". Journal of Medicinal Chemistry. 53 (15): 5858–5867. doi:10.1021/jm100574m. PMID   20684613.
  12. Gupta A, Gupta AK, Seshadri K (August 2009). "Structural models in the assessment of protein druggability based on HTS data". Journal of Computer-Aided Molecular Design. 23 (8): 583–592. Bibcode:2009JCAMD..23..583G. doi:10.1007/s10822-009-9279-y. PMID   19479324. S2CID   10718301.
  13. "DrugEBIlity Portal". ChEMBL. European Bioinformatics Institute.
  14. Jochim AL, Arora PS (October 2010). "Systematic analysis of helical protein interfaces reveals targets for synthetic inhibitors". ACS Chemical Biology. 5 (10): 919–923. doi:10.1021/cb1001747. PMC   2955827 . PMID   20712375.
  15. Kozakov D, Hall DR, Chuang GY, Cencic R, Brenke R, Grove LE, et al. (August 2011). "Structural conservation of druggable hot spots in protein-protein interfaces". Proceedings of the National Academy of Sciences of the United States of America. 108 (33): 13528–13533. Bibcode:2011PNAS..10813528K. doi: 10.1073/pnas.1101835108 . PMC   3158149 . PMID   21808046.
  16. Agüero F, Al-Lazikani B, Aslett M, Berriman M, Buckner FS, Campbell RK, et al. (November 2008). "Genomic-scale prioritization of drug targets: the TDR Targets database". Nature Reviews. Drug Discovery. 7 (11): 900–907. doi:10.1038/nrd2684. PMC   3184002 . PMID   18927591.
  17. Barelier S, Krimm I (August 2011). "Ligand specificity, privileged substructures and protein druggability from fragment-based screening". Current Opinion in Chemical Biology. 15 (4): 469–474. doi:10.1016/j.cbpa.2011.02.020. PMID   21411360.
  18. Overington JP, Al-Lazikani B, Hopkins AL (December 2006). "How many drug targets are there?". Nature Reviews. Drug Discovery. 5 (12): 993–996. doi:10.1038/nrd2199. PMID   17139284. S2CID   11979420.
  19. Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, et al. (January 2011). "DrugBank 3.0: a comprehensive resource for 'omics' research on drugs". Nucleic Acids Research. 39 (Database issue): D1035–D1041. doi:10.1093/nar/gkq1126. PMC   3013709 . PMID   21059682.
  20. 1 2 Oprea TI, Bologa CG, Brunak S, Campbell A, Gan GN, Gaulton A, et al. (May 2018). "Unexplored therapeutic opportunities in the human genome". Nature Reviews. Drug Discovery. 17 (5): 317–332. doi:10.1038/nrd.2018.14. PMC   6339563 . PMID   29472638.
  21. Halip L, Avram S, Curpan R, Borota A, Bora A, Bologa C, Oprea TI (December 2023). "Exploring DrugCentral: from molecular structures to clinical effects". Journal of Computer-Aided Molecular Design. 37 (12): 681–694. Bibcode:2023JCAMD..37..681H. doi:10.1007/s10822-023-00529-x. PMC   10692006 . PMID   37707619.
  22. Kwon B (2011-05-16). "Chemical biologist targets 'undruggable' proteins linked to cancer in quest for new cures". Brent Stockwell interview. Medical Xpress. Retrieved 2012-05-17.
  23. Stockwell BR (2011). The Quest for the Cure: The Science and Stories Behind the Next Generation of Medicines. New York: Columbia University Press. ISBN   978-0-231-15212-9.
  24. Stockwell B (October 2011). "Outsmarting Cancer. A biologist talks about what makes disease-causing proteins so difficult to target with drugs". Scientific American. 305 (4): 20. PMID   22106796.
  25. Buchwald P (October 2010). "Small-molecule protein-protein interaction inhibitors: therapeutic potential in light of molecular size, chemical space, and ligand binding efficiency considerations". IUBMB Life. 62 (10): 724–731. doi:10.1002/iub.383. PMID   20979208. S2CID   205970009.
  26. Morelli X, Bourgeas R, Roche P (August 2011). "Chemical and structural lessons from recent successes in protein-protein interaction inhibition (2P2I)". Current Opinion in Chemical Biology. 15 (4): 475–481. doi:10.1016/j.cbpa.2011.05.024. PMID   21684802.
  27. Verdine GL, Walensky LD (December 2007). "The challenge of drugging undruggable targets in cancer: lessons learned from targeting BCL-2 family members". Clinical Cancer Research. 13 (24): 7264–7270. doi:10.1158/1078-0432.CCR-07-2184. PMID   18094406. S2CID   7918779.
  28. Arkin MR, Whitty A (June 2009). "The road less traveled: modulating signal transduction enzymes by inhibiting their protein-protein interactions". Current Opinion in Chemical Biology. 13 (3): 284–290. doi:10.1016/j.cbpa.2009.05.125. PMID   19553156.
  29. Spradlin JN, Zhang E, Nomura DK (April 2021). "Reimagining Druggability Using Chemoproteomic Platforms". Accounts of Chemical Research. 54 (7): 1801–1813. doi:10.1021/acs.accounts.1c00065. PMID   33733731. S2CID   232303398.

Further reading