6
Available online at www.sciencedirect.com Drug target deconvolution by chemical proteomics Manfred Raida Drug target deconvolution is a process where the action of a drug, a small molecule, is characterised by identifying the proteins binding the drug and initiating the biological effect. The biological relevant target has to be extracted, or deconvoluted, from a list of proteins identified in such an approach. Beside the medically desired action of the drug, the identification of other proteins binding the drug can help to identify side effects and toxicity at a very early stage of drug development. The current approach to identify the proteins binding to the drug is an affinity-enrichment based approach, where the drug molecule is immobilised to a matrix through a linker and the proteins binding to the drug are identified by proteomics. Address Experimental Therapeutics Centre, A*STAR, 31 Biopolis Way, Nanos L3- 01, Singapore, 138669, Singapore Corresponding author: Raida, Manfred ([email protected]) Current Opinion in Chemical Biology 2011, 15:570–575 This review comes from a themed issue on Next Generation Therapeutics Edited by Alex Matter and Thomas H. Keller Available online 18th July 2011 1367-5931/$ see front matter # 2011 Elsevier Ltd. All rights reserved. DOI 10.1016/j.cbpa.2011.06.016 Introduction Affinity chromatography using small molecules immobil- ised through a linker to a solid matrix has been used over several decades to enrich and purify proteins. Back in 1968 and later in 1985 this approach was used to identify the protein binding to Isoquinoline H9 [1,2]. While in these early years the identification of the protein was the major bottleneck, today state-of-the-art mass spectrom- etry and available protein and genome databases enable a fast and sensitive identification of the proteins. This makes the method relevant for the identification of proteins that bind to drug candidates and other com- pounds of interest. In the past few years, the major focus in drug target deconvolution was on kinase inhibitors. Through the identification of the protein targets of a drug or a drug candidate, it becomes possible to understand side effects and toxicity in an early state of development, which should help to reduce the attrition rate in development. Even after 50 years, the teratology of Thalidomide (Contergan) [3,4] was explained by chemical proteomics [5 ]. By analysing kinase inhibitors in clinical use, the targets of these drugs could be identified by chemical proteomics and side effects and toxicity rationalised [617,18 ]. Besides the kinase inhibitors, other examples are cGTP-binding proteins [19,20], transcription factors and other proteins involved in nucleic acid processing [21], serine hydrolases [22], cysteine proteases [23,24] aspartyl proteases [25], metalloproteases [26], glycosi- dases [27,28], histone deacetylases [29,30], nitrilases [31,32] and oxireductases [33,34] and epigenetic enzymes as potential targets for cancer treatment [35 ]. There are some limitations to the identification of the relevant targets out of a large number of identified proteins, such as unspecific binding of background proteins, the risk of missing the interesting target owing to low abundance, and the risk of missing the target owing to sample processing steps. In any case, the identified target has to be validated by independent biological experiments, as shown for a tankyrase inhibitor [36 ]. Another important aspect that is crucial for a chemical proteomics approach is the orientation of the drug in the binding pocket of the target and the off-target protein. Parkinson drugs, for example, bind in different manner to the target protein and to the off-target or the liver-toxicity causing protein [37 ]. In the past years some methods for chemical proteomics have been developed that will be introduced here. Direct deconvolution of drug targets by affinity probes The direct way to identify a molecular target of a drug is the use of a chemical derivative of the drug to be immobilised on a solid matrix, as agarose, sepharose or magnetic beads [6,9,12,15 ,18 ,38,39 ,4042]. Incubating complex protein mixtures, as cell, tissue or organ lysates with the matrix-bound drug captures the target proteins. There are some restrictions to this approach, first of all, the drug molecule must retain the biological activity, a linker may interfere with the structure-activity relationship (SAR) of the native molecule. In many cases the attachment of a linker requires the complete new synthesis of the molecule and an attachment of linkers to different positions to avoid destruction of the SAR. The use of a linker attached to different sites of the molecule can give further insight into the drug actions as one compound can have different orientations in binding pockets in different Current Opinion in Chemical Biology 2011, 15:570575 www.sciencedirect.com

Drug target deconvolution by chemical proteomics

Embed Size (px)

Citation preview

Page 1: Drug target deconvolution by chemical proteomics

Available online at www.sciencedirect.com

Drug target deconvolution by chemical proteomicsManfred Raida

Drug target deconvolution is a process where the action of a

drug, a small molecule, is characterised by identifying the

proteins binding the drug and initiating the biological effect. The

biological relevant target has to be extracted, or deconvoluted,

from a list of proteins identified in such an approach. Beside the

medically desired action of the drug, the identification of other

proteins binding the drug can help to identify side effects and

toxicity at a very early stage of drug development. The current

approach to identify the proteins binding to the drug is an

affinity-enrichment based approach, where the drug molecule

is immobilised to a matrix through a linker and the proteins

binding to the drug are identified by proteomics.

Address

Experimental Therapeutics Centre, A*STAR, 31 Biopolis Way, Nanos L3-

01, Singapore, 138669, Singapore

Corresponding author: Raida, Manfred ([email protected])

Current Opinion in Chemical Biology 2011, 15:570–575

This review comes from a themed issue on

Next Generation Therapeutics

Edited by Alex Matter and Thomas H. Keller

Available online 18th July 2011

1367-5931/$ – see front matter

# 2011 Elsevier Ltd. All rights reserved.

DOI 10.1016/j.cbpa.2011.06.016

IntroductionAffinity chromatography using small molecules immobil-

ised through a linker to a solid matrix has been used over

several decades to enrich and purify proteins. Back in

1968 and later in 1985 this approach was used to identify

the protein binding to Isoquinoline H9 [1,2]. While in

these early years the identification of the protein was the

major bottleneck, today state-of-the-art mass spectrom-

etry and available protein and genome databases enable a

fast and sensitive identification of the proteins. This

makes the method relevant for the identification of

proteins that bind to drug candidates and other com-

pounds of interest. In the past few years, the major focus

in drug target deconvolution was on kinase inhibitors.

Through the identification of the protein targets of a drug

or a drug candidate, it becomes possible to understand side

effects and toxicity in an early state of development, which

should help to reduce the attrition rate in development.

Even after 50 years, the teratology of Thalidomide

Current Opinion in Chemical Biology 2011, 15:570–575

(Contergan) [3,4] was explained by chemical proteomics

[5�].

By analysing kinase inhibitors in clinical use, the targets

of these drugs could be identified by chemical proteomics

and side effects and toxicity rationalised [6–17,18��].Besides the kinase inhibitors, other examples are

cGTP-binding proteins [19,20], transcription factors

and other proteins involved in nucleic acid processing

[21], serine hydrolases [22], cysteine proteases [23,24]

aspartyl proteases [25], metalloproteases [26], glycosi-

dases [27,28], histone deacetylases [29,30], nitrilases

[31,32] and oxireductases [33,34] and epigenetic enzymes

as potential targets for cancer treatment [35��].

There are some limitations to the identification of the

relevant targets out of a large number of identified

proteins, such as unspecific binding of background

proteins, the risk of missing the interesting target owing

to low abundance, and the risk of missing the target owing

to sample processing steps. In any case, the identified

target has to be validated by independent biological

experiments, as shown for a tankyrase inhibitor [36�].

Another important aspect that is crucial for a chemical

proteomics approach is the orientation of the drug in the

binding pocket of the target and the off-target protein.

Parkinson drugs, for example, bind in different manner to

the target protein and to the off-target or the liver-toxicity

causing protein [37��].

In the past years some methods for chemical proteomics

have been developed that will be introduced here.

Direct deconvolution of drug targets byaffinity probesThe direct way to identify a molecular target of a drug is the

use of a chemical derivative of the drug to be immobilised

on a solid matrix, as agarose, sepharose or magnetic

beads [6,9,12,15��,18��,38,39�,40–42]. Incubating complex

protein mixtures, as cell, tissue or organ lysates with the

matrix-bound drug captures the target proteins. There are

some restrictions to this approach, first of all, the drug

molecule must retain the biological activity, a linker may

interfere with the structure-activity relationship (SAR) of

the native molecule. In many cases the attachment of a

linker requires the complete new synthesis of the molecule

and an attachment of linkers to different positions to avoid

destruction of the SAR. The use of a linker attached

to different sites of the molecule can give further

insight into the drug actions as one compound can have

different orientations in binding pockets in different

www.sciencedirect.com

Page 2: Drug target deconvolution by chemical proteomics

Drug target deconvolution Raida 571

Figure 1

Current Opinion in Chemical Biology

Principle of direct chemical proteomics: The drug bound to a solid matrix through a linker is incubated with the lysate and target proteins identified

after removal of unbound proteins by extensive washing.

proteins, resulting in side effects and toxicity beside the

desired biological action [37��]. Matrix and linker have to

be selected to have little or no unspecific binding of

proteins, a control with beads with linker but no drug

molecule has to be done for every experiment to identify

unspecific binders. This direct approach works generally

for kD’s below 1 mM. Another problem is the less specific

binding of a high abundant protein that also interferes with

the binding to the real, often low abundant target. The lysis

conditions are also of high importance, if the target is a

membrane bound protein, a nuclear or a cytosolic protein;

different conditions have to be applied. The chemical

proteomics approach has been shown applicable also to

membrane proteins [43] (Figure 1).

The approach described above uses the immobilised drug

on a solid matrix; a biphasic system of the solubilised

proteins and the solid phase bound drug molecules. Pore

size of the matrix may prevent larger proteins to reach the

drug molecules and reduce the binding, but may also

capture proteins in an unspecific way. Another approach is

the use of the drug molecule connected to a sorting

function with a defined specificity, like a biotin group,

through a linker. In this case the drug and the protein

mixture are incubated in solution, only after binding the

target molecules to the drug, they are affinity captured

through the biotin residue [44,45].

The deconvolution of the potential drug targets [46��] is

done in consecutive steps, experiments are repeated at

least two times and only proteins identified in both

experiments are considered to be valid. Next the frequent

hitters, proteins seen repeatedly in independent exper-

iments using unrelated drugs or with matrices without

immobilised drugs, are removed from the list. Then

highly abundant proteins that are present in many cell

www.sciencedirect.com

lines, the so-called ‘core-proteome’ proteins [47], are

removed from the list. These two steps always carry

the risk that the target is removed because it falls into

one of the exclusion criteria. The last step is the com-

petition, an independent experiment to identify the

relevant protine. The lysate is pre-incubated with the

molecule of interest in solution, and then the binding to

the immobilised compound is carried out. Proteins com-

peted from binding to the immobilised compound are

then probably the most relevant targets.

Usually the proteins purified by the affinity step are then

extracted from the matrix under denaturing conditions

and separated by gel-electrophoresis. The bands are

submitted to in-gel tryptic digestion and the proteins

identified by consecutive liquid-chromatography tan-

dem-mass spectrometry (LC–MS/MS) followed by data-

base searches. Questions of transient phosphorylation in

relation of a drug action on pathways can be addressed by

using specific enrichments of the phosphorylated pep-

tides [15��,46��].

In the past years this approach has been shown to be

applicable for a number of drugs and drug like molecules.

The major targets have been kinases, as their involve-

ment in cancer has been described for many cases. But

even for successful drugs with a well-defined target

profile, as Imatinib or Dasatinib, off-targets have been

identified [17,48–51]. The use of a single immobilised

kinase inhibitor [52��,53] allows the capturing of specific

target proteins while a combination of kinase inhibitors,

for example ‘Kinobeads’, allows the affinity capturing of

over 300 kinases from cell lysates [15��,16,18��]. While in

the first approach the specific targets of the inhibitor are

identified, the second approach is required for the com-

petition approach described below.

Current Opinion in Chemical Biology 2011, 15:570–575

Page 3: Drug target deconvolution by chemical proteomics

572 Next Generation Therapeutics

Figure 2

Current Opinion in Chemical Biology

Principle of competition chemical proteomics: The lysate is pre-incubated with the drug or with molecules in development that block the binding

pocket and prevent the target protein from binding to the immobilised compound.

Figure 3S

tand

ard

No

com

petit

ion

Com

petit

ion

Current Opinion in Chemical Biology

Virtual 1D SDS-gel of proteins affinity purified by the direct and the

competition approach. The bands marked in red are not detected in the

competition approach, therefore suggested as target candidates, while

the other proteins are unspecific bound or background.

Chemical proteomics has not only been applied to lysates

but also to living cells [54�].

A further development of this approach is the use of a

photo-reactive group close to the molecule [37��,55,56].

This allows for the identification of enzymes that

modify the compound and releases it, or use the com-

pound through intermediate binding to modify other

proteins, for examples as substrates as in the case of

proteases, epigenetic enzymes or kinases. The cell

lysate is incubated with a construct containing the

selective group, the drug moiety, a photo-reactive

group, and a sorting group or function, in most cases

a biotin. After binding to the drug moiety, the proteins

are covalently captured by the photo-reactive after UV

irradiation and the drug-protein construct is affinity

purified by the biotin group and identified as described

above.

The competition approachAffinity capturing of proteins using immobilised com-

pounds usually results in a long list of proteins identified

by mass spectrometry. Easily several hundreds of proteins

are identified [36�] in one experiment, selecting the real

target candidate from this list requires a deconvolution

process as described above [40]. After removing frequent

hitters, the core proteome, and proteins found only once

in repeated experiments the next step is to repeat the

experiment with the lysate pre-incubated with the

soluble form of the immobilised compound or with a

known competitor. Competing for the binding pocket of

the target protein, the soluble compound will prevent or

reduce the binding to the immobilised compound

[36�,57]. Only proteins competed by the compound in

solution are considered as real target candidates, further

verification has to be done by biological experiments

(Figure 2).

Current Opinion in Chemical Biology 2011, 15:570–575

But beside the target verification, the competition

approach offers more possibilities in the drug develop-

ment process (Figure 3).

Often it is not easily possible to attach a linker to the

molecule without destroying the SAR. For many natural

www.sciencedirect.com

Page 4: Drug target deconvolution by chemical proteomics

Drug target deconvolution Raida 573

compounds, the chemical re-synthesis itself alone and

with linker is impossible or is very time consuming and

man-power consuming. In many cases the linker has to

be attached to the molecule on different locations to

ensure capturing all potential targets. This has only

been shown for the Parkinson drugs [37��]. Very few

natural compounds have been used in chemical proteo-

mics; only Staurosporine, a fungal pan-kinase inhibitor,

has been immobilised to capture kinases [18��,58]. But

some of the natural compounds may also become future

drugs or leads and their target protein have to be

identified. The unbiased approach cannot be done in

competition experiments; the analysis has to be limited

to a class of potential target proteins. For these classes,

relative unspecific binders have to be immobilised to

cover as many as possible members of the enzyme class.

Using a set of pan-kinase inhibitors immobilised to the

matrix and incubating cell lysates with the candidate

molecule, proteins are competed from binding to the

immobilised compounds. Pre-incubation of the lysate

with increasing concentrations of a compound leads to a

concentration-dependent competition, which allows the

determination of IC50 values and the selectivity and

specificity [15��]. The main limitation is the use of the

immobilised inhibitors, which do not cover the entire

human kinome. In case of kinase inhibitors, this

approach allowed determining the specificity of drugs

in development and in clinics and a binding constant

that roughly describes the KD of the soluble compound.

The competition approach has been described for

kinase inhibitors and for epigenetic enzyme inhibitors

[15��,35��].

LimitationsThe described process has several limitations. First of all,

the drug must maintain its SAR, the linker has to be

attached without interfering with the binding to the

protein target. To overcome this limitation, the linker

can be attached to different sites of the molecule, but

requiring often-difficult chemistry approaches. Another

limitation is the preparation of the cell or tissue lysate,

the protein target can be missed owing to the preparation

conditions, for example membrane-spanning protein

targets are not solubilised. The major problem is the

identification of the real target from the large number of

proteins identified in a drug-target deconvolution exper-

iment, often exceeding 500 proteins. Only competition

experiments can help to identify the correct protein

target. In case of a low-expressed protein it may be

missed owing to sensitivity reasons or may be masked

by a high abundant background protein. This case can

happen if 1-dimensional gel electrophoresis is used to

separate the proteins prior to mass spectrometric identi-

fication.

Another case is the binding of the drug to more than one

target with low affinity, leading to a synergistic effect. In

www.sciencedirect.com

this case the binding may be too weak for an affinity

capturing of the protein targets.

Another major limitation is the still slow mass spectro-

metric identification of the proteins; an average exper-

iment can take up to several weeks for a full analysis. In

the current situation, no development is in sight to

increase the speed without losing analytical depth and

sensitivity.

Current developments and outlookSince the first publication of targeted affinity purification

of proteins using immobilised small molecule [2] the

process is still under development, the methods have

to be adapted for each molecule and cell or tissue source.

The number of drug-molecules for which the side effects

are not understood is large, chemical proteomics can help

to understand these.

The availability of faster and more sensitive mass spec-

trometers will help to identify low abundant proteins, but

also requires a cleaner sample preparation to avoid con-

taminations.

In combination with methods for protein structure

analysis, as NMR or X-ray crystallography of proteins

chemical proteomics will increase the knowledge of the

orientation of a small molecule in a binding pocket. This

increased knowledge can help to design better probes for

the identification of the protein targets.

SummaryDrug target deconvolution, chemical proteomics, chemo-

gentics, the identification of a drug target and the identi-

fication of the off-targets to explain side effects and

toxicity is becoming an essential step in early develop-

ment of a new drug and in understanding marketed drugs.

Currently the techniques are still under development, far

away from high-throughput approaches and can only be

applied to a limited number of molecules. But it will

become a major tool to support chemistry to develop more

targeted drugs in future and to identify potential side

effects and toxicity risks in early preclinical states.

References and recommended readingPapers of particular interest, published within the annual period ofreview, have been highlighted as:

� of special interest

�� of outstanding interest

1. Inagaki M, Watanabe M, Hidaka H: N-(2-aminoethyl)-5-isoquinolinesulfonamide, a newly synthesized protein kinaseinhibitor, functions as a ligand in affinity chromatography.Purification of ca2+-activated, phospholipid-dependentand other protein kinases. J Biol Chem 1985,260:2922-2925.

2. Cuatrecasas P, Wilchek M, Anfinsen CB: Selective enzymepurification by affinity chromatography. Proc Natl Acad Sci USA1968, 61:636-643.

Current Opinion in Chemical Biology 2011, 15:570–575

Page 5: Drug target deconvolution by chemical proteomics

574 Next Generation Therapeutics

3. Speirs AL: Drug-induced malformations in the fetus. Am Heart J1962, 64:717-718.

4. Burgio GR: The thalidomide disaster briefly revisited. Eur JPediatr 1981, 136:229-230.

5.�

Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y,Yamaguchi Y, Handa H: Identification of a primary target ofthalidomide teratogenicity. Science 2010, 327:1345-1350.

The authors describe the identification of the teratogenic target ofthaladomide. By using chemoproteomics, proteins were identifed whichare important for early embryonal development.

6. Brehmer D, Greff Z, Godl K, Blencke S, Kurtenbach A, Weber M,Muller S, Klebl B, Cotten M, Keri G et al.: Cellular targets ofgefitinib. Cancer Res 2005, 65:379-382.

7. Daub H: Characterisation of kinase-selective inhibitors bychemical proteomics. Biochim Biophys Acta 2005,1754:183-190.

8. Daub H: Kinase inhibitors: narrowing down the real targets. NatChem Biol 2010, 6:249-250.

9. Breitkopf SB, Oppermann FS, Keri G, Grammel M, Daub H:Proteomics analysis of cellular imatinib targets and theircandidate downstream effectors. J Proteome Res 2010,9:6033-6043.

10. Daub H, Godl K, Brehmer D, Klebl B, Muller G: Evaluation ofkinase inhibitor selectivity by chemical proteomics. Assay DrugDev Technol 2004, 2:215-224.

11. Godl K, Daub H: Proteomic analysis of kinase inhibitorselectivity and function. Cell Cycle 2004, 3:393-395.

12. Godl K, Gruss OJ, Eickhoff J, Wissing J, Blencke S, Weber M,Degen H, Brehmer D, Orfi L, Horvath Z et al.: Proteomiccharacterization of the angiogenesis inhibitor su6668 revealsmultiple impacts on cellular kinase signaling. Cancer Res 2005,65:6919-6926.

13. Godl K, Wissing J, Kurtenbach A, Habenberger P, Blencke S,Gutbrod H, Salassidis K, Stein-Gerlach M, Missio A, Cotten M,Daub H: An efficient proteomics method to identify the cellulartargets of protein kinase inhibitors. Proc Natl Acad Sci USA2003, 100:15434-15439.

14. Wissing J, Godl K, Brehmer D, Blencke S, Weber M,Habenberger P, Stein-Gerlach M, Missio A, Cotten M, Muller S,Daub H: Chemical proteomic analysis reveals alternativemodes of action for pyrido[2,3-d]pyrimidine kinase inhibitors.Mol Cell Proteomics 2004, 3:1181-1193.

15.��

Bantscheff M, Eberhard D, Abraham Y, Bastuck S, Boesche M,Hobson S, Mathieson T, Perrin J, Raida M, Rau C et al.:Quantitative chemical proteomics reveals mechanisms ofaction of clinical abl kinase inhibitors. Nat Biotechnol 2007,25:1035-1044.

This publication describes a chemical proteomics approach to profile theinteraction of small molecules with hundreds of endogenously expressedprotein kinases and purine-binding proteins. The use of several immo-bilized pan-kinase inhibitors enables the screening of multiple kinases inone analysis. By competition with drugs in clinics, the specificity andselectivity could be determined.

16. Bantscheff M, Hopf C, Kruse U, Drewes G: Proteomics-basedstrategies in kinase drug discovery. Ernst Schering Found SympProc 2007, 3:1-28.

17. Bantscheff M, Scholten A, Heck AJ: Revealing promiscuousdrug-target interactions by chemical proteomics. Drug DiscovToday 2009, 14:1021-1029.

18.��

Kruse U, Pallasch CP, Bantscheff M, Eberhard D, Frenzel L,Ghidelli S, Maier SK, Werner T, Wendtner CM, Drewes G:Chemoproteomics-based kinome profiling and targetdeconvolution of clinical multi-kinase inhibitors in primarychronic lymphocytic leukemia cells. Leukemia 2011, 25:89-100.

This publication describes the potential use of kinase profiling usingchemical proteomics for the development of treatments of chromiclymphotic leukemia (CLL). This is an application of the affinity matrixcarrying several pan-kinase inhibitors.

19. Scholten A, Poh MK, van Veen TA, van Breukelen B, Vos MA,Heck AJ: Analysis of the cgmp/camp interactome using a

Current Opinion in Chemical Biology 2011, 15:570–575

chemical proteomics approach in mammalian heart tissuevalidates sphingosine kinase type 1-interacting protein as agenuine and highly abundant akap. J Proteome Res 2006,5:1435-1447.

20. Hanke SE, Bertinetti D, Badel A, Schweinsberg S, Genieser HG,Herberg FW: Cyclic nucleotides as affinity tools:phosphorothioate camp analogues address specific pkasubproteomes. Nat Biotechnol 2011, 4:294-301.

21. Lin Y, Tang X, Zhu Y, Shu T, Han X: Identification of parp-1 asone of the transcription factors binding to the repressorelement in the promoter region of cox-2. Arch Biochem Biophys2011, 505:123-129.

22. Patricelli MP, Giang DK, Stamp LM, Burbaum JJ: Directvisualization of serine hydrolase activities in complexproteomes using fluorescent active site-directed probes.Proteomics 2001, 1:1067-1071.

23. Greenbaum D, Medzihradszky KF, Burlingame A, Bogyo M:Epoxide electrophiles as activity-dependent cysteineprotease profiling and discovery tools. Chem Biol 2000,7:569-581.

24. Kato D, Boatright KM, Berger AB, Nazif T, Blum G, Ryan C,Chehade KA, Salvesen GS, Bogyo M: Activity-based probes thattarget diverse cysteine protease families. Nat Chem Biol 2005,1:33-38.

25. Li YM, Xu M, Lai MT, Huang Q, Castro JL, DiMuzio-Mower J,Harrison T, Lellis C, Nadin A, Neduvelil JG et al.: Photoactivatedgamma-secretase inhibitors directed to the active sitecovalently label presenilin 1. Nature 2000, 405:689-694.

26. Sieber SA, Niessen S, Hoover HS, Cravatt BF: Proteomicprofiling of metalloprotease activities with cocktails of active-site probes. Nat Chem Biol 2006, 2:274-281.

27. Hekmat O, Kim YW, Williams SJ, He S, Withers SG: Active-sitepeptide ‘‘fingerprinting’’ of glycosidases in complex mixturesby mass spectrometry. Discovery of a novel retaining beta-1,4-glycanase in cellulomonas fimi. J Biol Chem 2005,280:35126-35135.

28. Vocadlo DJ, Hang HC, Kim EJ, Hanover JA, Bertozzi CR: Achemical approach for identifying o-glcnac-modified proteinsin cells. Proc Natl Acad Sci USA 2003, 100:9116-9121.

29. Salisbury CM, Cravatt BF: Optimization of activity-basedprobes for proteomic profiling of histone deacetylasecomplexes. J Am Chem Soc 2008, 130:2184-2194.

30. Salisbury CM, Cravatt BF: Activity-based probes for proteomicprofiling of histone deacetylase complexes. Proc Natl Acad SciUSA 2007, 104:1171-1176.

31. Barglow KT, Cravatt BF: Substrate mimicry in an activity-basedprobe that targets the nitrilase family of enzymes. Angew ChemInt Ed Engl 2006, 45:7408-7411.

32. Barglow KT, Saikatendu KS, Bracey MH, Huey R, Morris GM,Olson AJ, Stevens RC, Cravatt BF: Functional proteomic andstructural insights into molecular recognition in the nitrilasefamily enzymes. Biochemistry 2008, 47:13514-13523.

33. Adam GC, Sorensen EJ, Cravatt BF: Proteomic profiling ofmechanistically distinct enzyme classes using a commonchemotype. Nat Biotechnol 2002, 20:805-809.

34. Barglow KT, Cravatt BF: Discovering disease-associatedenzymes by proteome reactivity profiling. Chem Biol 2004,11:1523-1531.

35.��

Bantscheff M, Hopf C, Savitski MM, Dittmann A, Grandi P,Michon AM, Schlegl J, Abraham Y, Becher I, Bergamini G et al.:Chemoproteomics profiling of hdac inhibitors revealsselective targeting of hdac complexes. Nat Biotechnol 2011,29:255-265.

Using a matrix with a set of immobilized HDAC inhibitors, the authorscreate a profiling tool for the identification of the action of small moleculeson the histone modification enzyme complexes. As HDAC complexes getmore into interest as therapeutic targets, this approach will allow thetargeted development of new inhibitors and the screening of potentialcandidates using competition approaches.

www.sciencedirect.com

Page 6: Drug target deconvolution by chemical proteomics

Drug target deconvolution Raida 575

36.�

Huang SM, Mishina YM, Liu S, Cheung A, Stegmeier F,Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S et al.:Tankyrase inhibition stabilizes axin and antagonizes wntsignalling. Nature 2009, 461:614-620.

Wnt-signalling is involved in early development as well as in manycancers. In this publication, chemical proteomics was used to identifythe target of a small molecule, the Tankyrase 1, which plays an importantrole in the downstream signaling of Wnt. Beside the chemical proteomics,this publication describes the underlying biology in detail.

37.��

Fischer JJ, Michaelis S, Schrey AK, Graebner OG, Glinski M,Dreger M, Kroll F, Koester H: Capture compound massspectrometry sheds light on the molecular mechanisms ofliver toxicity of two parkinson drugs. Toxicol Sci 2010, 113:243-253.

The orientation of a drug in the binding pockets of different protein targetscan be different. This publication addresses this issue by attaching alinker to different sites of two drugs in clinics. Beside the therapeuticeffect, these drugs show different toxic effects. It could be shown that anattached linker that prevents binding to the therapeutic target allows thedrug to bind to other proteins, which explains the toxic effect.

38. Brehmer D, Godl K, Zech B, Wissing J, Daub H: Proteome-wideidentification of cellular targets affected bybisindolylmaleimide-type protein kinase c inhibitors. Mol CellProteomics 2004, 3:490-500.

39.�

Raijmakers R, Dadvar P, Pelletier S, Gouw J, Rumpel K, Heck AJ:Target profiling of a small library of phosphodiesterase 5(pde5) inhibitors using chemical proteomics. ChemMedChem2010, 5:1927-1936.

Inhibitors of phosphodiesterase 5 (PDE5) are drugs in clinics. Beside theirwell-defined effects, the targets beside PDE5 are not fully characterized.The authors characterize the protein targets of the inhibitors by chemicalproteomics and give inside into their biological activities.

40. Rix U, Superti-Furga G: Target profiling of small molecules bychemical proteomics. Nat Chem Biol 2009, 5:616-624.

41. Knockaert M, Gray N, Damiens E, Chang YT, Grellier P, Grant K,Fergusson D, Mottram J, Soete M, Dubremetz JF et al.:Intracellular targets of cyclin-dependent kinase inhibitors:identification by affinity chromatography using immobilisedinhibitors. Chem Biol 2000, 7:411-422.

42. Peters EC, Gray NS: Chemical proteomics identifiesunanticipated targets of clinical kinase inhibitors. ACS ChemBiol 2007, 2:661-664.

43. Hensley K, Christov A, Kamat S, Zhang XC, Jackson KW, Snow S,Post J: Proteomic identification of binding partners for thebrain metabolite lanthionine ketimine (lk) and documentationof lk effects on microglia and motoneuron cell cultures. JNeurosci 2010, 30:2979-2988.

44. Lenz T, Poot P, Grabner O, Glinski M, Weinhold E, Dreger M,Koster H: Profiling of methyltransferases and other s-adenosyl-l-homocysteine-binding proteins by capturecompound mass spectrometry (ccms). J Vis Exp 2010:46.

45. Dalhoff C, Huben M, Lenz T, Poot P, Nordhoff E, Koster H,Weinhold E: Synthesis of s-adenosyl-l-homocysteine capturecompounds for selective photoinduced isolation ofmethyltransferases. Chembiochem 2010, 11:256-265.

46.��

Li J, Rix U, Fang B, Bai Y, Edwards A, Colinge J, Bennett KL, Gao J,Song L, Eschrich S et al.: A chemical and phosphoproteomiccharacterization of dasatinib action in lung cancer. Nat ChemBiol 2010, 6:291-299.

This publication describes a chemical proteomics approach to under-stand the action of a drug in clinics on signaling pathways active in lungcancer. The authors identified nearly 40 proteins as target of Dasatinib,mainly kinases. The effect of Dasatinib on the phosphorylation of proteinswas analyzed and gives insight into the drug action.

47. Burkard TR, Planyavsky M, Kaupe I, Breitwieser FP,Burckstummer T, Bennett KL, Superti-Furga G, Colinge J: Initial

www.sciencedirect.com

characterization of the human central proteome. BMC Syst Biol2011, 5:17.

48. Hantschel O, Rix U, Superti-Furga G: Target spectrum of the bcr-abl inhibitors imatinib, nilotinib and dasatinib. Leuk Lymphoma2008, 49:615-619.

49. Winger JA, Hantschel O, Superti-Furga G, Kuriyan J: Thestructure of the leukemia drug imatinib bound to humanquinone reductase 2 (nqo2). BMC Struct Biol 2009, 9:7.

50. Baumgartner C, Gleixner KV, Peter B, Ferenc V, Gruze A, RemsingRix LL, Bennett KL, Samorapoompichit P, Lee FY, Pickl WF et al.:Dasatinib inhibits the growth and survival of neoplastic humaneosinophils (eol-1) through targeting of fip1l1-pdgfralpha. ExpHematol 2008, 36:1244-1253.

51. Rix U, Hantschel O, Durnberger G, Remsing Rix LL, Planyavsky M,Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J et al.:Chemical proteomic profiles of the bcr-abl inhibitors imatinib,nilotinib, and dasatinib reveal novel kinase and nonkinasetargets. Blood 2007, 110:4055-4063.

52.��

Rix U, Remsing Rix LL, Terker AS, Fernbach NV, Hantschel O,Planyavsky M, Breitwieser FP, Herrmann H, Colinge J, Bennett KLet al.: A comprehensive target selectivity survey of the bcr-ablkinase inhibitor inno-406 by kinase profiling and chemicalproteomics in chronic myeloid leukemia cells. Leukemia 2010,24:44-50.

Resistance to drugs is one of the major problems in effective treatment ofcancers, often caused by point mutations in the targeted protein. Secondgeneration drugs may overcome these resistances by interacting withmutated target proteins and other targets. The authors analyse the targetprofile of a second generation drug by chemical proteomics and byfunctional assays using recombinant kinases. This will help to understandthe drug action and to develop new drugs with improved activities.

53. Hantschel O, Rix U, Schmidt U, Burckstummer T, Kneidinger M,Schutze G, Colinge J, Bennett KL, Ellmeier W, Valent P, Superti-Furga G: The btk tyrosine kinase is a major target of the bcr-ablinhibitor dasatinib. Proc Natl Acad Sci USA 2007,104:13283-13288.

54.�

Saxena C, Bonacci TM, Huss KL, Bloem LJ, Higgs RE, Hale JE:Capture of drug targets from live cells using a multipurposeimmuno-chemo-proteomics tool. J Proteome Res 2009,8:3951-3957.

Chemical proteomics generally uses cell or tissue lysates to identify thetargets of the small molecule. This publication describes a construct of asmall molecule, the drug, and a cell-permeable peptide with a fluoro-phore. This allows direct visualization of the drug in the cell, but also thecapturing of the drug with the protein bound to it. This approach allows towork with living cells rather than cell lysate to study the action of a drugand the binding to the targets.

55. Koster H, Little DP, Luan P, Muller R, Siddiqi SM, Marappan S,Yip P: Capture compound mass spectrometry: a technologyfor the investigation of small molecule protein interactions.Assay Drug Dev Technol 2007, 5:381-390.

56. Luo Y, Fischer JJ, Baessler OY, Schrey AK, Ungewiss J, Glinski M,Sefkow M, Dreger M, Koester H: Gdp-capture compound – anovel tool for the profiling of gtpases in pro- and eukaryotes bycapture compound mass spectrometry (ccms). J Proteomics2010, 73:815-819.

57. Terstappen GC, Schlupen C, Raggiaschi R, Gaviraghi G: Targetdeconvolution strategies in drug discovery. Nat Rev DrugDiscov 2007, 6:891-903.

58. Fischer JJ, Graebner Baessler OY, Dalhoff C, Michaelis S,Schrey AK, Ungewiss J, Andrich K, Jeske D, Kroll F, Glinski Met al.: Comprehensive identification of staurosporine-bindingkinases in the hepatocyte cell line hepg2 using capturecompound mass spectrometry (ccms). J Proteome Res 2010,9:806-817.

Current Opinion in Chemical Biology 2011, 15:570–575