Affinity selection-mass spectrometry screening techniques for small molecule drug discovery

https://doi.org/10.1016/j.cbpa.2007.07.011Get rights and content

Affinity selection-mass spectrometry (AS-MS) techniques assess the binding of candidate molecules to immobilized or soluble receptors, and these methods are gaining acceptance in high throughput screening laboratories as valuable complements to traditional drug discovery technologies. A diversity of receptor types have been evaluated by AS-MS, including those that are difficult to screen using traditional biochemical approaches. AS-MS techniques that couple liquid chromatography-MS with size-based separation methods, such as ultrafiltration, gel permeation, or size-exclusion chromatography, are particularly amenable to the demands of MS-based screening and have demonstrated the greatest success across a broad range of drug targets. MS measurements of receptor function have many of the same advantages as AS-MS screening and are increasingly used for drug discovery as well.

Introduction

Mass spectrometry (MS) is one of the most powerful analytical techniques in modern science, and it plays a key role in nearly every stage of the drug development process [1]. Recent advances in MS instrument design, especially the development of electrospray ionization (ESI), have extended the reach of MS to the lead discovery stages of drug development as well. Modern pharmaceutical discovery relies increasingly on target-based screening techniques to identify new lead compounds for receptors that have known or suspected involvement in a disease pathway. High-throughput screening (HTS) methods that use MS as the detection system are of particular interest because of the exquisite sensitivity and unique selectivity possible with MS. In some screening formats, the selectivity achieved by MS enables direct analysis of compound mixtures such as combinatorial libraries and unpurified natural products extracts – an advantage that is not easily achieved using other analytical methods. MS-based screening is primarily implemented in two ways: by monitoring the functional output of a receptor-dependent biochemical reaction, or by using affinity-based methods that directly assess binding of a candidate molecule to its target receptor. Because of their expanding popularity, both of these styles of MS-based drug discovery have been the subject of several timely reviews, special issues of topical journals, and the focus of at least two recent books [2, 3••, 4, 5, 6, 7]. Herein we report on the recent contributions to the field of MS-based drug discovery, with special emphasis on lead identification methods that couple an affinity selection step with MS confirmation of receptor–ligand binding.

Section snippets

Affinity selection-MS: direct detection of receptor–ligand complexes

Affinity selection-MS (AS-MS) methods directly or indirectly measure binding of small molecules to their biomolecular target, and all varieties of AS-MS include the following steps: (i) an affinity selection stage, where the protein is equilibrated with one or more potential ligands, allowing the protein to form a complex with any compound capable of binding; (ii) the resulting receptor–ligand complexes are separated from non-binding mixture components; and (iii) ligands are identified by MS or

Immobilized ligand or receptor

In part to enable the use of non-volatile buffers in the receptor–ligand binding reaction, indirect AS-MS screening methods have been developed that couple a separation technique with ESI-MS detection. The simplest variants are those where one binding partner, either the receptor or the small molecule ligand, is immobilized on a solid support. Familiar non-MS methods infer receptor–ligand interactions by measuring changes in surface plasmon resonance (Biacore) or wavelength shift (Corning's

MS measurements of receptor function

MS-based techniques that monitor functional assays have advantages that parallel those of AS-MS. Target-based functional assays identify lead compounds by measuring the outcome of a biomolecule-mediated transformation, such as production or depletion of the starting materials or products of an enzymatic reaction, or displacement of a labeled marker compound from a biomolecular receptor. These measurements are traditionally done using radiochemical methods, fluorescence readouts, or UV

Conclusions and future directions

As shown in this report, the pharmaceutical industry is enthusiastically using MS-based HTS methods to discover and characterize new lead compounds, and we anticipate these applications will continue and expand in the future. Presently, no one-size-fits-all AS-MS solution is expected to satisfy all future screening demands, and no commercial product specifically tailored for AS-MS screening is available to interested researchers. Rather, a diversity of methods is implemented by different

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We thank Satish Jindal, Jerry Shipps, Arshad Siddiqui, and our colleagues at SPRI for critical review of this manuscript.

References (63)

  • W.A. Korfmacher

    Principles and applications of LC–MS in new drug discovery

    Drug Discov Today

    (2005)
  • Lee MS (Ed): Integrated Strategies for Drug Discovery Using Mass Spectrometry, Hoboken, NJ: Wiley-Interscience;...
  • Wanner K, Höfner G (Eds): Mass Spectrometry in Medicinal Chemistry, Weinheim: Wiley-VCH;...
  • S.M. Schermann et al.

    Mass spectrometry-based approaches to protein–ligand interactions

    Expert Rev Proteomics

    (2005)
  • G. Deng et al.

    Applications of mass spectrometry in early stages of target based drug discovery

    J Pharm Biomed Anal

    (2006)
  • K.F. Geoghegan et al.

    Biochemical applications of mass spectrometry in pharmaceutical drug discovery

    Mass Spectrom Rev

    (2005)
  • Hofstadler SA, Lee M (Eds): International Journal of Mass Spectrometry, Special Issue: Drug Discovery: Elsevier;...
  • M.T. Cancilla et al.

    Tethering: fragment-based drug discovery by mass spectrometry

  • D.A. Erlanson et al.

    Making drugs on proteins: site-directed ligand discovery for fragment-based lead assembly

    Curr Opin Chem Biol

    (2004)
  • B. Ganem et al.

    Observation of noncovalent enzyme-substrate and enzyme-product complexes by ion-spray mass spectrometry

    J Am Chem Soc

    (1991)
  • B. Ganem et al.

    Going gently into flight: analyzing noncovalent interactions by mass spectrometry

    Bioorg Med Chem

    (2003)
  • A. Sinz

    Investigation of Protein–ligand Interactions by Mass Spectrometry

    Chem Med Chem

    (2007)
  • S.A. Hofstadler et al.

    Applications of ESI-MS in drug discovery: interrogation of noncovalent complexes

    Nat Rev Drug Discov

    (2006)
  • S. Zhang et al.

    Chip-based nanoelectrospray mass spectrometry for protein characterization

    Expert Rev Proteomics

    (2004)
  • S. Zhang et al.

    Quantitative determination of noncovalent binding interactions using automated nanoelectrospray mass spectrometry

    Anal Chem

    (2003)
  • Y. Xie et al.

    Top-down ESI-ECD-FT-ICR mass spectrometry localizes noncovalent protein–ligand binding sites

    J Am Chem Soc

    (2006)
  • M.A. Raji et al.

    Development of an ESI-MS screening method for evaluating binding affinity between integrin fragments and RGD-based peptides

    Int J Mass Spectrom

    (2007)
  • S.M. Clark et al.

    Determination of ligand-protein dissociation constants by electrospray mass spectrometry-based diffusion measurements

    Anal Chem

    (2004)
  • S.M. Clark et al.

    Screening for noncovalent ligand-receptor interactions by electrospray ionization mass spectrometry-based diffusion measurements

    Anal Chem

    (2004)
  • G.M. Makara et al.

    Improving success rates for lead generation using affinity binding technologies

    Curr Opin Biotechnol

    (2005)
  • B.T. Cunningham et al.

    Microplate-based, label-free detection of biomolecular interactions: applications in proteomics

    Expert Rev Proteomics

    (2006)
  • E.S. Ng et al.

    High-throughput screening for enzyme inhibitors using frontal affinity chromatography with liquid chromatography and mass spectrometry

    Anal Chem

    (2005)
  • N. Chan et al.

    Frontal affinity chromatography – mass spectrometry for ligand discovery and characterization

  • T.R. Besanger et al.

    Immobilized enzyme reactor chromatography: Optimization of protein retention and enzyme activity in monolithic silica stationary phases

    Anal Chim Acta

    (2006)
  • T.R. Besanger et al.

    Monolithic membrane-receptor columns: Optimization of column performance for frontal affinity chromatography/mass spectrometry applications

    Anal Chim Acta

    (2006)
  • J.J. Slon-Usakiewicz et al.

    Global kinase screening. Applications of frontal affinity chromatography coupled to mass spectrometry in drug discovery

    Anal Chem

    (2005)
  • J.J. Slon-Usakiewicz et al.

    Frontal affinity chromatography with MS detection (FAC-MS) in drug discovery

    Drug Discov Today

    (2005)
  • J.J. Slon-Usakiewicz et al.

    Frontal affinity chromatography with MS detection of EphB2 tyrosine kinase receptor. 1. Comparison with conventional ELISA

    J Med Chem

    (2004)
  • V.P. Gullo et al.

    Drug discovery from natural products

    J Ind Microbiol Biotechnol

    (2006)
  • C. Schou et al.

    Recent applications of affinity interactions in capillary electrophoresis

    Electrophoresis

    (2006)
  • P. Kovarik et al.

    Capillary-scale frontal affinity chromatography/MALDI tandem mass spectrometry using protein-doped monolithic silica columns

    Anal Chem

    (2005)
  • Cited by (196)

    • Recent discovery of tyrosinase inhibitors in traditional Chinese medicines and screening methods

      2023, Journal of Ethnopharmacology
      Citation Excerpt :

      According to the forms of tyrosinase, bioaffinity-based screening techniques can be classified into two modes, free enzyme-based and immobilized-based screenings (L.L. Li et al., 2016). It is worth mentioning that free enzyme-based affinity methods (e. g. Ultrafiltration-high performance liquid chromatography-mass spectrometry (UF-HPLC-MS)) do not need to immobilize enzymes and can avoid the alteration of targets’ properties caused by the introduction of chemical linkage in enzyme immobilization (Annis et al., 2007). In UF-HPLC-MS, molecules are incubated with target proteins in solution to form complexes that stay upon the filter membrane, while the unbound small molecules can pass through.

    View all citing articles on Scopus
    View full text