1-s2.0-S1367593109000453-main

download 1-s2.0-S1367593109000453-main

of 12

Transcript of 1-s2.0-S1367593109000453-main

  • 7/29/2019 1-s2.0-S1367593109000453-main

    1/12

    Available online at www.sciencedirect.com

    The therapeutic potential of phosphatase inhibitorsViktor V Vintonyak1, Andrey P Antonchick1, Daniel Rauh3 and HerbertWaldmann1,2

    Protein phosphatases (PPs) constitute a large family of

    enzymes, which are crucial modulators of cellular

    phosphorylation events. Malfunction in PP activity has been

    associated with human diseases, including diabetes, obesity,

    cancer, and neurodegenerative and autoimmune disorders,

    and makes this class of enzymes attractive targets for chemical

    biology and medicinal chemistry research. A number of

    strategies are currently explored for the identification and

    development of various classes of PP modulators and have

    resulted in a plethora of chemically distinct inhibitors. Limited

    selectivity and adverse pharmacological properties of PP

    inhibitors are still major bottlenecks for further clinical

    development and resulted in only a few molecular entitiescurrently in clinical trials.

    Addresses1 Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11,

    D-44227 Dortmund, Germany2 Technical University Dortmund, Otto-Hahn-Strasse 6, D-44221Dortmund, Germany3 Chemical Genomics Centre of the Max Planck Society, Otto-Hahn-Strasse 15, D-44227 Dortmund, Germany

    Corresponding author: Rauh, Daniel ([email protected]) and

    Waldmann, Herbert ([email protected])

    Current Opinion in Chemical Biology 2009, 13:272283

    This review comes from a themed issue on

    Next-generation therapeutics

    Edited by Karl-Heinz Altmann and Dario Neri

    Available online 4th May 2009

    1367-5931/$ see front matter# 2009 Elsevier Ltd. All rights reserved.

    DOI 10.1016/j.cbpa.2009.03.021

    IntroductionPhosphorylation and dephosphorylation are among the

    most important modifications by which nature modulatesprotein function in nearly all biological systems to trans-

    duce information between distinct cellular sites. The

    specific transfer of theg-phosphate from ATP to substrate

    proteins is catalyzed by protein kinases (PKs) and can

    lead to conformational changes, promote proteinprotein

    interactions, or turn on/off enzymatic activity to allow

    cells to translate a wide variety of environmental signals

    into functional changes. Protein phosphatases (PPs) are

    defined by their ability to catalyze the hydrolysis of

    phosphates to restore the protein substrate to its depho-sphorylated state and can be considered as physiological

    counterparts of PKs. The balanced and highly dynamic

    interplay between these two enzyme classes is crucial for

    the control of cell signaling cascades. To achieve the

    essential temporal and spatial specificity in modulating

    enzyme activity in signaling events, nature often uses the

    compartmentalization of PK and PP activities. In con-

    sequence, aberrantly regulated PKs and PPs play causa-

    tive roles in diseases such as cancer, diabetes, and

    neurological and autoimmune disorders, making these

    enzymes an important set of therapeutic targets across

    almost all disease areas [1,24,5,6]. PPs are classified

    by their substrate specificities into serine/threonine-phos-

    phatase (STP), protein histidine-phosphatase (PHP),protein tyrosine-phosphatase (PTP) and dual-specific

    phosphatases (DSPs) which catalyze the dephosphoryla-

    tion of both serine/threonine or tyrosine residues. The

    detailed understanding of the different catalytic mech-

    anisms, substrate specificities, and associated confor-

    mational changes in phosphatase structure has provento be crucial for the design and development of potent

    and selective inhibitors. STPs have metal ions in their

    catalytic center (zinc, manganese, or magnesium) which

    are coordinated by side chains of His and Asp amino acids.

    A highly polarized water molecule bridges the two metal

    centers and can act as a nucleophile attacking the phos-

    phate group to initiate its hydrolysis. PTPs follow adifferent mechanism and do not require metal ions. A

    structural characteristic crucial for the design of most PTP

    inhibitors is a deep pocket to accommodate the pTyr of

    the substrate. A WPD-loop (Trp-Pro-Asp) closes on this

    site, regulates catalysis by its Asp residue, and allows

    proper substrate recognition. In the closed substrate

    bound state, the Asp forms a hydrogen bond to the

    phenolic oxygen of the pTyr and polarizes this site for

    the nucleophilic attack of the adjacent catalytic cysteine.

    This Cys is positioned at the N-terminus of a long helix in

    the middle of the catalytic domain. The helix induces adipole and thereby alters the properties of the Cys thio-

    late side chain. The phosphate gets transferred onto the

    Cys to form a thiophosphate, which is subsequentlyhydrolyzed by a water molecule. DSPs follow the same

    mechanism but possess a shallower substrate pocket to

    accommodate pThr and pSer.

    The human genome encodes for a total of 518 kinases,

    including 90 tyrosine kinases. Similarly, more than 130

    PPs are encoded in the human genome, including 107

    tyrosine phosphatases [7]. This represents a fairly

    balanced complement of PTKs and PTPs and suggests

    an equal partnership of these enzymes in regulating

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

    mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.cbpa.2009.03.021http://dx.doi.org/10.1016/j.cbpa.2009.03.021mailto:[email protected]:[email protected]
  • 7/29/2019 1-s2.0-S1367593109000453-main

    2/12

    protein function through tyrosine phosphorylation. Themajority of protein phosphorylation, however, occurs on

    serines and threonines and is catalyzed by the remaining

    428 protein serine/threonine kinases. The fewer protein

    STPsimply thatthese enzymes mustdisplaymuch broader

    substrate specificities than the complementary kinases.The catalytic domains of PPs are highly conserved and

    differences in substrate specificity are largely determined

    by regulatory domains or subunits. Somecatalytic domains,

    in particular those of the protein STPs, can interact with a

    large variety of regulatory subunits and are controlled by

    post-translational modifications, proteinprotein inter-

    actions, or heteromeric compositions. The incorporation

    of catalytic subunits in multidomain complexes explains

    why eukaryotes contain fewer genes that encode for PPs

    than genes that encodefor PKs. When theholoenzymesare

    considered, the numbers of PKs and PPs are more or less

    balanced [3,810]. PPs are often kinase substrates them-

    selves, underlining their essential regulatory role in sig-

    naling cascades. As a consequence, many diseases have

    beenshown to be associated withregulatory malfunction of

    PPs and the acceptance of PPs as equal in importance to

    PKs as potential drug targets has been supposed over the

    last decade. In this review we will focus on recent progress

    in the development of inhibitors directed against clinically

    relevant PPs (Table 1).

    Clinically relevant phosphatasesPTP1B

    Acquired Type 2 diabetes and obesity often can be

    considered as lifestyle-related diseases of civilization

    and are linked to insulin resistance and loss of proper

    glucose homeostasis. Type 2 diabetes and obesity areoften coupled in human and can lead to the metabolic

    syndrome, which dramatically increases the risk of cardi-

    ovascular incidences and decreases lifespan. Some pre-

    dictions for the year 2015 estimate 70% of the population

    of the western hemisphere to be overweight and 40% of

    these to be obese. The social impact and costs associated

    with this health risk will be substantial and drugs regulat-ing metabolic disorders are in high demand.

    The discovery of the receptor tyrosine phosphatasePTP1B as a negative regulator of the insulin and leptin

    receptor pathways [11,12,13] and animal studies demon-

    strating that PTP1B-deficient mice show an enhanced

    insulin sensitivity, improved glycemic control, and resist-

    ance to high fat diet induced obesity [14

    ,15] made thisenzyme an attractive drug target. Although the molecular

    causality of insulin resistance and obesity is not fully

    understood, the hormone insulin induces autophosphor-

    ylation of its receptor and thereby triggers a kinase

    cascade that finally induces synthesis of the short-term

    energy storage glycogen and synthesis of fatty acids and

    proteins. Dephosphorylation of the receptor by PTP1B

    leads to its inactivation and shut-down additional down-

    stream processes. The pharmacological blockage of

    PTP1B should therefore counteract insulin resistance.

    At the latest when Abbott demonstrated that antisense

    oligonucleotides (ASOs) designed to downregulate

    expression of PTP1B normalized blood glucose and

    improved insulin sensitivity without changing the regular

    diet of mice [16], nearly every major pharmaceutical

    company launched a program for the identification of

    potent PTP1B inhibitors. Besides its central role in the

    insulin cascade PTP1B is also involved in a variety of

    clinically relevant pathways [17,18] and is overexpressed

    or upregulated in human breast, colon, and ovarian can-

    cers [1921]. However, the discovery of clinically useful

    PTP1B inhibitors has proven to be very difficult because

    of limited inhibitor selectivity and low bioavailability

    [4,5,22].

    Structure-based design and active-site inhibitors

    Structure-based design is widely applied to phosphataseinhibitor research and best exemplified by more than 80

    complex crystal structures of PTP1B deposited in the

    Protein Data Base (PDB) (Figure 1a). One of the first

    crystal structures of PTP1B (pdb code: 1pty) featured an

    enzymatically dead mutant variant (catalytic Cys mutated

    to Ser) in complex with phosphotyrosine. The substrate is

    coordinated in the active site of the phosphatase by anetwork of hydrogen bonds. Interestingly, a second pTyr

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 273

    Table 1

    Selected phosphatases as target proteins

    Family Phosphatases Disease, therapeutic approachSerine/t hre onine phospha tases PP1, PP2A Tumor suppression, malignancy

    PP2B, PP2C Cystic fibrosis, immune suppression, asthma, cardiovascular

    Tyrosine phosphatases PTP1B Diabetes, obesity

    MptpA/B Tuberculosis

    CD45 Alzheimers disease, autoimmune disease, inflammation,

    organ transplantationSHP-1/2 Neuron protection, obesity, Noonan syndrome, leukemia,

    regulation of RAS/MAP-kinases

    Dual-specific phosphatases VHR Regulation of MAP-kinases

    Cdc25 Cell cycle progression, tumor therapy (various human cancers)

    PRL-1/2/3 Promoting tumor cells, leukemia, Hodgkins disease, prostate cancer

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283

  • 7/29/2019 1-s2.0-S1367593109000453-main

    3/12

    molecule was identified in an adjacent, noncatalytic site

    and stimulated ideas to fuse the two moieties by a suitable

    linker which resulted in bidentate inhibitors with

    dramatically increased affinities (Figure 1b). This second

    site has since then been highly important in lead structureoptimization for PTP1B. Fragment-based approaches

    both by protein X-ray crystallography and NMR tech-

    niques were employed to identify, for example, small

    organic acids as binders to this second site and resulted in

    an impressive array of potent nM inhibitors when fused to

    phosphomimics (Figure 2).

    Most active-site-directed PTP inhibitors reported to date

    are nonhydrolyzable pTyr mimics that take advantage of

    the positively charged active site. However, this con-

    served mode of action reduces selectivity and often

    results in the potent inhibition of multiple phosphatases.

    Most pTyr mimics possess a high charge density, which in

    turn is often detrimental to cell penetration. The most

    useful and potent pTyr surrogate devised for PTP1B thus

    far contains the nonhydrolyzable difluorophosphono-

    methylphenylalanine (F2Pmp) group [23]. The fluorine

    atoms have been proposed to interact via hydrogen bonds

    with NH-groups of the PTP and to increase the binding

    affinity by several orders of magnitude. Compound 1a

    identified from a combinatorial chemistry approach dis-

    plays a Ki value of 2.4 nM for PTP1B and exhibits notableselectivity in favor of PTP1B against a panel of PTPs

    including Cdc25A, SHP-2, and VHR [24]. Because of its

    high polarity compound 1a is not cell permeable and

    shows no cellular activity. To enhance penetration across

    membranes, derivatives of 1a were prepared either by

    coupling to a cell permeable (D)-Arg8-peptide tag (1b)[25] or to a highly lipophilic fatty acid (1c) [26]. Com-

    pound 1b showed a significant improvement in leptin-

    dependent suppression of food intake in leptin-resistant

    rats [27]. Furthermore, the concept of a prodrug was

    utilized in 2 [28].

    Several strategies have been applied to identify pTyr

    mimics with more favorable pharmacological properties.

    One such approach, referred to Breakaway Tethering,

    introduces a Cys residue on the surface of the enzyme,

    which acts as a tethering point. The modified enzyme is

    274 Next-generation therapeutics

    Figure 1

    Inhibitors in complex with the catalytic domain of PTP1B. (a) 88 PTP1B-inhibitor complexes found in the PDB and aligned to the first reportedPTP1B crystal structure (PDB code: 1pty). Most inhibitors target the phosphate binding sites and mimic substrate binding. (b) Close-up

    view of the active site shows binding of the pTyr of the substrate peptide (gray sticks) to the N-terminal end of the central helix (blue) to

    position the phosphate in close proximity to the catalytic Cys215 (ball and sticks). The WPD-loop (yellow) controls access to the active

    site. Binding of a second pTyr molecule (blue ball and sticks) adjacent to the active site stimulated the design of molecules that bind toboth pTyr sites and resulted in the development of the potent bidentate inhibitor 8 (green) (PDB code: 2qbp). (c) In inactive PTP1B

    conformations the C-terminal helix (orange) adopts an extended orientation and exposes a new cavity (green). Allosteric modulators

    such as 3 inhibit PTP1B activity by binding to this allosteric site and preventing the WPD-loop from adopting a catalytically competent

    conformation. The inhibitors bind around the side chain of Phe280 and form favorable pp interactions. Figures were prepared with PyMol(http://www.pymol.org).

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

    http://www.pymol.org/http://www.pymol.org/
  • 7/29/2019 1-s2.0-S1367593109000453-main

    4/12

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 275

    Figure 2

    Structures of selected PTP1B and Cdc25 inhibitors, IC50 or Ki values are given.

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283

  • 7/29/2019 1-s2.0-S1367593109000453-main

    5/12

    incubated with a library of disulfide-containing small-molecule fragments under reducing conditions, which

    promote thiol exchange and formation of an SS bond

    between the ligand and the surface Cys residue [ 29].

    This tethering approach in combination with mass spec-

    trometry allows for capturing compounds that displayeven weak affinity for the target protein. Using this

    approach Erlanson et al. identified a novel PTP1B inhibi-

    tor with enhanced bioavailability.

    In a more recent analysis 1,2,5-thiadiazolidin-3-one-1,1-

    dioxide was identified as a novel phosphomimic and the

    isothiazolidinone group was utilized in the synthesis of the

    peptide-based inhibitor 4, which shows an IC50 of 40 nM[30]. In addition, using the samemimic, nonpeptide-based

    compound 5 was identified, which displays high potency in

    biochemical assays (IC50 of 35 nM). The same compound

    also shows cellular activity and increases the level of

    phosphorylation on the insulin receptor (2.8-fold) at

    80 mM [31]. More recently, Zhang et al. identified aryl

    diketoacids 6 as novel pTyr surrogates and showed that

    neutral amide-linked aryl diketoacid dimers 7 also exhibitPTP inhibitory activity. Detailed studies by enzyme

    kinetics and protein X-ray crystallography revealed that

    these derivatives stabilize PTP1B in its inactive, WPD-

    loop open conformation and act as noncompetitive inhibi-

    tors [32]. Using a high-throughput screening approach a

    series of monocyclic thiophenes were identified as PTP1B

    inhibitors [33]. Further optimization resulted in the de-

    velopment of the potent inhibitor 8. Introduction of a

    tetrazole ring or 1,2,5-thiadiazolidine-3-one-1,1-dioxide

    as a carboxylate mimic led to the discovery of two unique

    starting points that improved cell permeability andincreased potency up to 300 nM [34]. Additional examples

    for active-site-directed PTP1B inhibitors include arylben-

    zonaphthothiophenes and arylbenzonaphthofurans. Both

    scaffolds were shown to improve insulin sensitivity in

    rodents. One compound from these efforts, ertiprotafib

    (9), progressed to clinical trials for the treatment of Type 2

    diabetes. Development was discontinued in phase IIbecause of insufficient efficacy and strong unwanted side

    effects [35]. Although great progress has been made to

    increase potency of PTP1B inhibitors, initial successes

    were compromised by unwanted cross reactivity with T-

    cell protein tyrosine phosphatase (TCPTP) (77%

    sequence identity with PTP1B) and the discovery thatTCPTP knockout mice are born healthy but die after

    about four weeks. Even more disturbing was the finding

    that parallel knockoutof TCPTP andPTP1B turnedout to

    be lethal. The design and synthesis of selective PTP1B

    inhibitors that are less potent against TCPTP can be

    considered a particular challenge in current PTP1B med-

    icinal chemistry research.

    Allosteric inhibitors

    An alternative approach to overcome the current limita-tions in selectivity and bioavailability of active-site-

    directed phosphatase inhibitors was presented in 2004when researchers at Sunesis reported an allosteric site

    located at the back of the phosphatase about 20 A distant

    to the catalytic site. The crystal structure of PTP1B in

    complex with the allosteric inhibitor 3 (pdb code: 1t4j)

    proved that the ligand stabilizes the inactive phosphataseconformation by preventing the WPD-loop from adopting

    a catalytically competent conformation [36] and

    revealed a novel but general mechanism to inhibit tyro-

    sine phosphatases (Figure 1c). In the allosteric site, the

    inhibitor binds close to a central Phe residue and forms

    favorable pp interactions. Interestingly, the structural

    homolog TCPTP holds a Cys residue at this position and

    3 binds only with reduced affinity. Future work will show

    if the selectivity problem can be tackled adequately by

    this new mode of action. Another allosteric inhibitor of

    PTP1B, trodusquemine (MSI-1436) (10), is being devel-

    oped by Genaera Corp. for the potential treatment of

    Type 2 diabetes and obesity and proceeded to phase Ib

    [37]. Two previous phase I studies showed that single

    doses of10 administered to more than 60 subjects were

    well-tolerated with an acceptable adverse event profile,

    produced dose-dependent weight loss and improved

    insulin sensitivity. Genaera Corp. expects to verify the

    clinical potential and positive efficacy results of this drug

    in ongoing clinical trials [38].

    Meanwhile a new clinical focus is on ASOs which are

    directed against PTP1B. The main advantage of oligo-

    nucleotides compared to small-molecular-weight inhibi-

    tors is their genetic selectivity for PTP1B without perturbing

    TCPTP function. Several PTP1B ASOs have been devel-

    oped by ISIS Pharmaceuticals Inc. and a second gener-ation ASO ISIS-113715 is currently in phase II clinical

    trials for the treatment of Type 2 diabetes.

    Cancer-related PTPs

    A number of PTPs have been identified as critical onco-

    genes in human malignancy and are considered potential

    drug targets for the development of novel anticancertherapeutics [5,6,39,40].

    Cdc25

    Cdc25s belong to the family of DSPs and regulate cyclin-

    dependent kinases (Cdks), which are the key participants

    in cell division induced in response to extracellular signalsincluding growth factors. Cdc25 phosphatases are also the

    key components of the checkpoint pathway and are

    inactivated or degraded to induce cell cycle arrest in

    response to DNA damage, leading to DNA repair or

    apoptosis [41]. Dysregulation of these processes can

    contribute to genomic instability and lead to cancer. In

    humans, the three isoforms Cdc25A, Cdc25B, and

    Cdc25C seem to have overlapping substrate specificities

    for the different Cdkcyclin complexes [42]. Cdc25A and

    Cdc25B overexpression has been reported in varioushuman cancers, including breast, ovarian, prostate, lung,

    276 Next-generation therapeutics

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

  • 7/29/2019 1-s2.0-S1367593109000453-main

    6/12

    colorectal, pancreatic, gastric, thyroid, hepatocellular, andneuroblastoma and often put in context with more aggres-

    sive tumors and poorer clinical outcome [41]. The

    inhibition of Cdc25 phosphatases may represent a novel

    approach for the development of anticancer therapeutics.

    Two recent reviews by Garuti et al. [43] and Contour-Galcera et al. [44] provide a comprehensive overview of

    current Cdc25 inhibitor development. The most potent

    Cdc25 inhibitor reported to date is the naphthoquinone

    NSC95397 (11) with IC50 values of 22, 125, and 56.9 nMfor Cdc25A, Cdc25B, and Cdc25C, respectively [45]. In

    addition, it displayed significant inhibition when tested

    against human and murine carcinoma cells and blocked

    the G2/M phase transition during the cell cycle. Another

    potent compound is NSC663284 (12), which inhibits

    Cdc25s in vitro in an irreversible, time-dependent manner

    with Ki values in the mid nM range for all Cdc25 isoforms.

    It arrests cells in the G1 and G2/M phases of the cell cycle

    and induces significant growth inhibition of human breast

    cancer cell lines [46]. Contour-Galcera et al. recently

    reported the identification of a new thiazoloquinone,

    BN82685 (13), which inhibits Cdc25 in biochemicalassays (IC50 = 250, 250, and 170 nM for Cdc25A, Cdc25B,

    and Cdc25C, respectively) and is active against human

    xenografts [47]. In addition, low concentrations of

    BN82685 in combination with the mitotic inhibitor pacli-

    taxel (Taxol) inhibit the proliferation of colon cancer cells

    [48]. Most quinone-based Cdc25 inhibitors reported to

    date are irreversible binders and act via arylation of the

    nucleophilic catalytic Cys. The redox properties of qui-

    nones can also generate reactive oxygen species (ROS),

    which may cause toxicity to normal tissues and thus

    reduce therapeutic potential. In order to overcome thisproblem, Carr et al. recently synthesized a series of non-

    quinone sulfone analogs of vitamin K3, H32 (14) [49].

    Such compounds preferentially inhibit Cdc25 by rever-

    sibly binding to the catalytic cysteine and lead to G1

    phase arrest during the cell cycle in Hep3B cells. An

    alternative strategy in the design of Cdc25 inhibitors is

    the utilization of phosphate surrogates, which anchor theligand in the active site. Dysidiolide (15a), a sesterterpe-

    noid isolated from the Caribbean sponge Dysidea etheria

    was the first natural product reported to be active on

    Cdc25 (inhibition of Cdc25A with an IC50 of 9.4 mM). The

    g-hydroxybutenolide moiety is thought to mimic the

    substrate phosphate. Facilitating 15a as a biologicallyvalidated starting point, we used solid-phase synthesis

    for the generation of a small collection of dysidiolide

    analogs [50,51] and were able to identify the inhibitors

    of Cdc25C that are more potent than the parent natural

    product 15a. Analogs 15b and 15c displayed IC50 values of

    5.1 and 0.8 mM, respectively. Moreover, these analogs

    proved considerable biological activity in cytotoxicity

    assays employing different cancer cell lines. Using the

    structures of dysidiolide and vitamin D3 as starting points

    Shimazawa et al. designed several potent Cdc25 inhibi-tors. Compound 16 inhibits Cdc25A and Cdc25B with

    IC50 values of 0.44 and 1.9 mM, respectively [52]. Recently2-methoxyestradiol (2-ME) was reported as a potent,

    selective, and relatively nontoxic inhibitor of hepatoma

    growth both in vitro and in vivo. It was suggested that 2-

    ME binds to the catalytic site Cys [53]. Another class of

    Cdc25 inhibitors is maleic anhydride derivatives bearing afatty acid chain at the C-4 position [54]. Compound 17

    inhibits Cdc25s with IC50 values in the low mM range and

    induces G0/G1 phase accumulation with subsequent

    inhibition of Cdk2 activity. Moreover, apoptosis was

    triggered in the presence of17 within a 48-hour treatment

    without oxidative burst. N-Arylmaleimide derivatives are

    potent electrophiles and reagents for thiol-selective

    modifications and were introduced as a novel class of

    Cdc25 inhibitors [55]. PM-20 (18) is selective for Cdc25A

    with an IC50 value of 1 mM. Furthermore, PM-20 inhibits

    the growth of several human tumor cells, especially

    Hep3B cells with an IC50 of 0.7 mM. More recently several

    Cdc25 phosphatase inhibitors with micromolar activities

    were discovered from structure-based virtual screening

    [56]. The most active of them was compound 19, which

    inhibits Cdc25A and Cdc25B with IC50 of 0.8 and 2.0 mM,respectively.

    SHP-2

    SHP-2 is a nonreceptor PTP that mediates cell signaling

    by growth factors and cytokines acting via the RAS/MAP-

    kinase pathway [40]. Consistent with its overall role in cell

    signaling, mutations of the SHP-2 gene (PTPN11) can

    cause hyperactivation of its catalytic activity and have

    been identified in the Noonan syndrome, a developmen-

    tal disorder that is frequently associated with short stature

    [57], and in various childhood leukemia [58]. The inci-dence of the Noonan syndrome is relatively frequent and

    affects 1 in 1002500 children, while leukemia accounts

    for 2% of adult cancers and 1/3 of childhood cancers.

    Because mutation associated SHP-2 overactivation seems

    to be the cause of a great portion of these incidents,

    pharmacological modulation of SHP-2 activity represents

    an attractive way to prevent disease development inpatients bearing these mutations. The exploration of

    selective SHP-2 inhibitors not only is thought to be of

    use for the treatment of cancer but may also become the

    basis for future treatments of infectious diseases. The

    pentavalent antimony derivative sodium stibogluconate,

    a known agent against leishmaniasis, has recently beenfound to inhibit SHP-2 activity [59]. Moreover, sodium

    stibogluconate is the first SHP-1/2 inhibitor that reached

    the clinic. In September 2006 VioQuest Pharmaceuticals

    initiated a phase I/IIa clinical trial for sodium stiboglu-

    conate as chemotherapeutic in patients with advanced

    malignancies. Although SHP-2 represents an attractive

    target for the treatment of cancer, only a few SHP-2

    inhibitors are known from the literature. Recently the

    design and synthesis of a compound collection containing

    SHP-2 inhibitors inspired by furanodictines and the con-cept of biology-oriented synthesis was reported [60].

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 277

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283

  • 7/29/2019 1-s2.0-S1367593109000453-main

    7/12

    Inhibitor 20 (Figure 3) exhibits an IC50 of 2.5 mM against

    SHP-2. A screening initiative at the National Cancer

    Institute resulted in the identification of NSC-87877

    (21) [61] and NSC-117199 (22) [62] which inhibit SHP-

    2 with IC50 values of 0.32 and 47 mM, respectively. On the

    basis of the oxindole 22 a focused library was designed

    and 23 was identified as a selective inhibitor of SHP-2

    over SHP-1 and PTP1B with an IC50 value of 0.8 mM for

    278 Next-generation therapeutics

    Figure 3

    Structures of selected inhibitors of SHP-2, PRLs, MptpA, MptpB, and CD45. IC50 or Ki values are given.

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

  • 7/29/2019 1-s2.0-S1367593109000453-main

    8/12

    SHP-2 [62]. Recently, Birchmeier et al. performed an in

    silico screen of low-molecular-weight compounds that

    may bind to the catalytic site of SHP-2. This analysis

    resulted in the discovery of PHPS1 (24) as a potent and

    cell-permeable inhibitor, which is selective for SHP-2

    over SHP-1 and PTP1B [63]. PHPS1 efficiently inhibitsthe activation of Erk1/2 by a leukemia-associated mutant

    variant of SHP-2 and blocks growth of a variety of human

    tumor cell lines. In addition, Geronikaki et al. reported the

    synthesis and biological evaluation of thiazolidin-4-one

    derivatives as a novel class of SHP-2 inhibitors [64].

    Compound 25 exhibited the best inhibitory activity with

    a Ki of 11.7 mM.

    PRL

    Members of the phosphatase of regenerating liver (PRL)

    subfamily are DSPs, consisting of PRL-1, PRL-2, and

    PRL-3, and represent an intriguing group of potential

    target proteinsfor the treatment of various cancers [6,65].

    PRL-3overexpression correlates withmetastasis in many

    malignancies and several recent reports suggest that

    PRLs may play key causal roles in promoting tumor cellmotility and invasion. PRL-3 expression is upregulated

    in the tumors of colorectal cancers metastasized to liver

    [66], brain, lung, or ovary [67] and is also elevated in

    pediatric acute myeloid leukemia, Hodgkins lymphoma

    cells, and prostate cancer. In addition, alteration of PRL-

    1 expression in a number of cancer cell lines leads to

    changes in cell adherence and invasive properties. The

    genetic knockdown of PRL-3 with interfering RNA in

    cancer cells can abrogate cell motility and the ability to

    metastasize in a mouse model [68]. However, the dis-

    covery of PRL phosphatase inhibitors has lagged behindthe extensive pharmacological and structural studies on

    this target protein and only a few inhibitor classes have

    been reported so far. The bis-benzamidine derivative

    and antileishmaniasis drug pentamidine (26) were

    reported to inhibit all three PRL isoforms in vitro and

    induced tumor shrinkage in a melanoma mousexenograft

    model [69]. In addition, recent studies by Lee et al.

    showed that the combination of pentamidine with the

    phenothiazine antipsychotic agent chlorpromazine

    exerts synergistic antiproliferative effects. Pentamidine

    treatment resulted in chromosomal segregation defects

    and delayed progression through mitosis, which is con-

    sistent with the inhibition of PRL [70]. Another class ofPRL-3 inhibitors was identified by screening of chemical

    libraries by Ahn et al. Benzylidene rhodanine derivatives

    showed good inhibitory activity against PRL-3. Com-

    pound 27 was the most active with an IC50 of 0.9 mM in

    vitro and showed reduced invasion in cell-based assays

    [71]. In addition, biflavonoids isolated from young

    branches of Taxus cuspidata inhibit PRL-3 with IC50values in the low mM range [72]. More recently Park

    et al. identified 12 novel inhibitors of PRL-3 by means of

    virtual screening and docking simulations [73]. Thediscovered inhibitors revealed structural diversity but

    low inhibitory potency with IC50 values ranging from10 to 50 mM.

    MptpA and MptpB

    Tuberculosis (TB) continues to be a major cause of

    morbidity and mortality throughout the world. Accordingto the World Health Organization, one-third of the

    worlds population is infected with Mycobacterium tuber-

    culosis [74] and about 35 million people are expected to

    die from TB in the first 20 years of this century. Because

    of the increasing occurrence of drug-resistant mycobac-

    teria and the need of the extended use of current drugs,

    new targets and drugs for therapeutic interventions are in

    high demand. M. tuberculosis protein tyrosine phosphatase

    A (MptpA) and MptpB are two enzymes secreted by

    growing mycobacteria and believed to mediate M. tuber-

    culosis survival in host macrophages by the dephosphor-

    ylation of proteins that are involved in interferon

    signaling, which represents a crucial pathway of the

    immune system [75]. The genetic knockout of MptpB

    suppressed growth of M. tuberculosis in activated macro-

    phages and guinea pigs [76] and suggests that MptpA/Bcould qualify as potential drug targets in the treatment of

    TB. In addition, inhibitors might also prove useful as

    probe molecules in chemical biology approaches to dis-

    sect the role of MptpA/B phosphatases in hostpathogen

    invasion. Recently the identification of MptpA inhibitors

    from screening natural-product inspired compound

    libraries was reported [77]. Compound 28 is an analog

    of roseophilin, a natural product found in Streptomyces

    species and proved to be an inhibitor of MptpA with

    an IC50 value of 0.9 mM. Utilization of fragment-based

    library design resulted in the discovery of several novelclasses of MptpA inhibitors, among which compound 29

    was the most active one, exhibiting a Ki value of 1.6 mM.

    Additional MptpA inhibitors are based on the indolizine-

    1-carbonitrile scaffold [78]. Using biology-oriented syn-

    thesis (BIOS) as an efficient approach to the discovery of

    new compound classes for medicinal chemistry and

    chemical biology research, also inhibitors of MptpB wereidentified [60,79]. The indole derivatives 30, 31, and 32

    were at least 100-fold more selective for MptpB and

    displayed IC50 values in the low mM and high nM range.

    Alber et al. reported the development of a potent and

    selective (oxalylamino-methylene)-thiophene sulfona-

    mide inhibitor for MptpB (33) (OMTS) [80

    ]. OMTS(33) has an IC50 value of 440 50 nM and >60-fold

    specificity for MptpB over six human PTPs. The crystal

    structure of MptpB in complex with OMTS revealed

    substantial structural rearrangements of the enzyme, with

    some residues shifting >27 A relative to the MptpB:PO4complex. In addition, extensive contacts with the cata-

    lytic loop provided a potential basis for inhibitor selec-

    tivity. More recently, Ellman et al. developed a substrate-

    based fragment-based approach termed substrate activity

    screening (SAS) to identify novel PTP inhibitors withsubmicromolar inhibitory activities [81]. Application of

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 279

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283

  • 7/29/2019 1-s2.0-S1367593109000453-main

    9/12

    this method to MtptB resulted in the discovery of theisoxazole-based inhibitor 34. With a Ki of 220 nM com-

    pound 34 is the most potent MptpB inhibitor known from

    literature to date. Moreover 34 was shown to be selective

    for mycobacterial (MptpA) against a panel and human

    PTPs (VHR, TCPTP, CD45, and LAR). Recently, thediscovery of a new class of MptpB inhibitors was facili-

    tated by the previously mentioned BIOS approach. A

    stereoselective solid-phase synthesis of macroline deriva-

    tives yielded 120 natural product analogs. Kinetic studies

    and extensive NMR spectroscopy suggest that inhibitors

    identified from these macrolines inhibit MptpB not via

    the substrate-binding site, but rather via an allosteric

    mechanism yet to be identified [82]. Compounds 35

    and 36 selectively inhibit MptpB with IC50 values of

    7.0 and 9.6 mM, respectively and did not show inhibition

    of other phosphatases (MptpA, VE-PTP, PTP1B, TC-

    PTPN2, and Cdc25A) up to a tested concentration of

    100 mM. More recently, Terenzi et al. reported the identi-

    fication of synthetic chalcones as a new class of MptpA

    inhibitors [83]. Compound 37 turned out to be the most

    active representative with an IC50 value of 8.4 mM.

    CD45

    CD45 is the first and the best characterized transmem-

    brane phosphatase. It is expressed in hematopoietic cells

    and plays a crucial role in T-cell receptor mediated

    signaling. It regulates the phosphorylation status and

    thereby activity of Src-family protein tyrosine kinases

    and their substrates [84]. CD45 has gathered particular

    attention because its inhibition by antibodies blocks T-

    cell activation in vitro and graft rejection in mice [85].

    These studies highlight the potential value of selectiveinhibitors of CD45 in the treatment of autoimmune

    diseases and transplant rejection. Point mutations in

    the CD45 gene have been associated with autoimmune

    diseases such as multiple sclerosis [85] and autoimmune

    hepatitis. In addition the negative regulation of cytokine

    receptor signaling by CD45 could rationalize the loss of

    CD45 activity that has been observed in several cancers,such as leukemia. CD45 has been correlated with the

    proliferation of myeloma cells and could therefore be a

    potential target for the treatment of multiple myelomas

    [86]. In addition, recently it was reported that targeted

    radiotherapy with monoclonal CD45 antibodies induces

    apoptosis and breaks b-irradiation-resistance and doxor-ubicin-resistance in leukemia cells [87]. In spite of the

    potential therapeutic utility of CD45-specific inhibitors,

    comparatively little has been reported in the literature on

    their development. The first comprehensive review on

    the development of CD45 inhibitors was composed by

    Lee and Burke [88]. 9,10-Phenanthrenediones (e.g. 38)

    and 1,2-naphthalenediones have been reported by

    researchers from AstraZeneca as potent inhibitors of

    CD45 [89]. Simple structural modifications resulted in

    the identification of compounds that also inhibit T-cellproliferation at low mM concentrations and are selective

    for CD45 over other PTPs including PTP1B. A variety ofbenzimidazole derivatives have been identified through

    high-throughput screening as potent CD45 inhibitors and

    led to the development of TU-572 (39), which inhibits

    CD45 with an IC50 of 0.28 mM [90]. In addition, 2-amino-

    2-thioxoacetamide derivatives were also reported to inhi-bit CD45 [91]. However, the most active compound 40

    exhibited only moderate activity with an IC50 of 29 mM.

    ConclusionsBecause of the physiological role of phosphatases as

    antagonists of kinase activity, there is a high therapeutic

    potential in targeting these enzymes. Fostered by the

    enthusiasm and strong indications provided by initial

    PTP1B inhibitor development, the discovery of phospha-

    tase-modulating agents has progressed steadily in the past

    years and resulted in the generation of a variety of potent

    inhibitors. However, because most of the chemical scaf-

    folds that qualify as PP-inhibitors embody phosphate

    mimics, several challenges remain in current phospha-

    tase-related medicinal chemistry endeavors. These in-

    clude limited selectivity, limited cell permeability, and

    insufficient pharmacological properties. As a direct con-

    sequence, only a few PP inhibitors have entered clinical

    trials and new inhibitor classes are in high demand. In

    addition, relatively few phosphatases are being explored

    chemically and new targets may be more promising

    although general limitations such as polarity of active-

    site-directed inhibitors prevail. Because PPs often func-

    tion in multienzyme complexes, targeting proteinprotein interactions either by stabilizers or disruptors

    might offer a suitable strategy to overcome the current

    limitations of small molecules directed against the activesite of the enzyme. The very recent development of short

    interfering RNAs (siRNAs) for therapeutic approaches

    [92,93] may be a viable new alternative in targeting

    phosphatases. Interfering RNAs lead to the degradation

    of messenger RNA and facilitate nucleic acids as drugsagainst otherwise challenging drug targets [94]. Recently,

    Sirna Therapeutic Inc. claimed to have developed a

    RNAi directed against PTP1B that potently reduces

    cellular PTP1B expression levels [95]. However, at this

    point only time will tell whether any of the discussed

    therapeutic approaches will succeed. As soon as the first

    phosphatase-targeted drug will reach the market a flood of

    new development programs is to be expected just as forkinases at the end of the 1980s.

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

    of special interest

    of outstanding interest

    1.

    Tonks NK: Protein tyrosine phosphatases: from genes, tofunction, to disease. Nat Rev Mol Cell Biol 2006, 7:833-846.

    This is an excellent review on the current understanding of the biologicalrole and function of PTPs.

    280 Next-generation therapeutics

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

  • 7/29/2019 1-s2.0-S1367593109000453-main

    10/12

    2. Zhang ZY: Protein tyrosine phosphatases: prospects fortherapeutics. Curr Opin Chem Biol 2001, 5:416-423.

    3. Barford D, Das AK, Egloff MP: The structure and mechanism ofprotein phosphatases: insights into catalysis and regulation.Annu Rev Biophys Biomol Struct 1998, 27:133-164.

    4. Kasibhatla B, Wos J, Peters KG: Targeting protein tyrosinephosphatase to enhance insulin action for the potentialtreatment of diabetes. Curr Opin Invest Drugs 2007,8:805-813.

    5.

    Bialy L, Waldmann H: Inhibitors of protein tyrosinephosphatases: next-generation drugs? Angew Chem Int EdEngl 2005, 44:3814-3839.

    This review describes in detail various strategies that were applied in thedevelopment of small-molecule PTP inhibitors.

    6. Yi T, Lindner D: The role and target potential of proteintyrosine phosphatases in cancer. Curr Oncol Rep 2008,10:114-121.

    7.

    Alonso A, Sasin J, Bottini N, Friedberg I, Osterman A, Godzik A,Hunter T, Dixon J, Mustelin T: Protein tyrosine phosphatases inthe human genome. Cell 2004, 117:699-711.

    This paper describes the classification of PTPs based on the humangenome.

    8. Gallego M, Virshup DM: Protein serine/threoninephosphatases: life, death, and sleeping. Curr Opin Cell Biol2005, 17:197-202.

    9. Perrotti D, Neviani P: Protein phosphatase 2A (PP2A), adruggable tumor suppressor in Ph1(+) leukemias. CancerMetastasis Rev 2008, 27:159-168.

    10. Bollen M, Beullens M: Signaling by protein phosphatases in thenucleus. Trends Cell Biol 2002, 12:138-145.

    11. Asante-Appiah E, Kennedy BP: Protein tyrosine phosphatases:the quest for negative regulators of insulin action.Am J PhysiolEndocrinol Metab 2003, 284:E663-670.

    12. Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, Haj F,Wang Y, Minokoshi Y, Kim YB, Elmquist JK, Tartaglia LA, Kahn BBet al.: PTP1B regulates leptin signal transduction in vivo. DevCell 2002, 2:489-495.

    13. Bence KK, Delibegovic M, Xue B, Gorgun CZ, Hotamisligil GS,Neel BG, Kahn BB: Neuronal PTP1B regulates body weight,adiposity and leptin action. Nat Med 2006, 12:917-924.

    This work demonstrates that PTP1B regulates body mass and adiposityprimarily through processes in the CNS as well via the regulation of leptinproduction in adipocytes.

    14.

    Elchebly M, Payette P, Michaliszyn E, Cromlish W, Collins S,Loy AL, Normandin D, Cheng A, Himms-Hagen J, Chan CC et al.:Increased insulin sensitivity and obesity resistance in micelacking the protein tyrosine phosphatase-1B gene. Science1999, 283:1544-1548.

    This is an important first study that demonstrated the role of PTP1B ininsulin resistance and obesity using gene knockout technologies. Thefound phenotype generated much interest in PTP1B as a drug target forType 2 diabetes and obesity.

    15. Klaman LD,Boss O, PeroniOD, KimJK, Martino JL,Zabolotny JM,Moghal N, Lubkin M, Kim YB, Sharpe AH et al.: Increased energyexpenditure, decreased adiposity, and tissue-specific insulin

    sensitivity in protein-tyrosine phosphatase 1B-deficient mice.Mol Cell Biol 2000, 20:5479-5489.

    16. Zinker BA, Rondinone CM, Trevillyan JM, Gum RJ, Clampit JE,Waring JF, Xie N, Wilcox D, Jacobson P, Frost L et al.: PTP1Bantisense oligonucleotide lowers PTP1B protein, normalizesblood glucose, and improves insulin sensitivity in diabeticmice. Proc Natl Acad Sci U S A 2002, 99:11357-11362.

    17. Julien SG, Dube N, Read M, Penney J, Paquet M, Han Y,Kennedy BP, Muller WJ, Tremblay ML: Protein tyrosinephosphatase 1B deficiency or inhibition delaysErbB2-inducedmammary tumorigenesis and protects from lung metastasis.Nat Genet 2007, 39:338-346.

    18. Dube N, Tremblay ML: Beyond the metabolic function ofPTP1B. Cell Cycle 2004, 3:550-553.

    19. Bentires-Alj M, Neel BG: Protein-tyrosine phosphatase 1B isrequired for HER2/Neu-induced breast cancer. Cancer Res2007, 67:2420-2424.

    20. Tonks NK, Muthuswamy SK: A brake becomes an accelerator:PTP1Ba new therapeutic target for breast cancer. CancerCell 2007, 11:214-216.

    21. Zhu S,Bjorge JD, FujitaDJ: PTP1B contributesto the oncogenicproperties of colon cancer cells through Srcactivation. CancerRes 2007, 67:10129-10137.

    22. Zhang S, Zhang ZY: PTP1B as a drug target: recentdevelopments in PTP1B inhibitor discovery. Drug Discov Today2007, 12:373-381.

    23. Burke TR Jr, Kole HK, Roller PP: Potent inhibition of insulinreceptor dephosphorylation by a hexamer peptide containingthe phosphotyrosyl mimetic F2Pmp. Biochem Biophys ResCommun 1994, 204:129-134.

    24. Shen K, Keng YF, Wu L, Guo XL, Lawrence DS, Zhang ZY:Acquisition of a specific and potent PTP1B inhibitor from anovel combinatorial library and screening procedure. J BiolChem 2001, 276:47311-47319.

    25. LeeSY, Liang F, GuoXL, Xie L, Cahill SM, Blumenstein M, Yang H,Lawrence DS,Zhang ZY: Design, construction, and intracellularactivation of an intramolecularly self-silenced signaltransduction inhibitor. Angew Chem Int Ed Engl 2005,44:4242-4244.

    26. Xie L, Lee SY, Andersen JN, Waters S, Shen K, Guo XL, Moller NP,Olefsky JM, Lawrence DS, Zhang ZY: Cellular effects of smallmolecule PTP1B inhibitors on insulin signaling. Biochemistry2003, 42:12792-12804.

    27. Morrison CD, White CL, Wang Z, Lee SY, Lawrence DS,Cefalu WT, Zhang ZY, Gettys TW: Increased hypothalamicprotein tyrosine phosphatase 1B contributes to leptinresistance with age. Endocrinology2007, 148:433-440.

    28. Boutselis IG,Yu X, Zhang ZY, BorchRF: Synthesisand cell-basedactivity of a potent and selective protein tyrosine phosphatase1B inhibitor prodrug. J Med Chem 2007, 50:856-864.

    29.

    Erlanson DA, McDowell RS, He MM, Randal M, Simmons RL,Kung J, Waight A, Hansen SK: Discovery of a newphosphotyrosine mimetic for PTP1B using breakaway

    tethering. J Am Chem Soc 2003, 125:5602-5603.The authors report on an extension of the tethering technology break-away tethering. This approach was successfully applied to the antidia-betic protein PTP1B for thediscovery of pTyr mimetics,whichbelong to anew molecular class with a different binding mode.

    30. Yue EW, Wayland B, Douty B, Crawley ML, McLaughlin E,Takvorian A, Wasserman Z, Bower MJ, Wei M, Li Y et al.:Isothiazolidinone heterocycles as inhibitors of protein tyrosinephosphatases:synthesis and structureactivity relationships ofa peptide scaffold. Bioorg Med Chem 2006, 14:5833-5849.

    31. Combs AP, Zhu W, Crawley ML, Glass B, Polam P, Sparks RB,Modi D, Takvorian A, McLaughlin E, Yue EW et al.: Potentbenzimidazole sulfonamide protein tyrosine phosphatase 1Binhibitors containing the heterocyclic (S)-isothiazolidinonephosphotyrosine mimetic. J Med Chem 2006, 49:3774-3789.

    32. Liu S, Zeng LF, Wu L, Yu X, Xue T, Gunawan AM, Long YQ,Zhang ZY: Targeting inactive enzyme conformation: aryldiketoacid derivatives as a new class of PTP1B inhibitors.J Am Chem Soc 2008, 130:17075-17084.

    33. Wan ZK, Lee J, Xu W, Erbe DV, Joseph-McCarthy D, Follows BC,Zhang YL: Monocyclic thiophenes as protein tyrosinephosphatase 1B inhibitors: capturing interactions with Asp48.Bioorg Med Chem Lett 2006, 16:4941-4945.

    34. Wan ZK,Follows B,KirincichS, WilsonD, BinnunE, Xu W,Joseph-McCarthy D, Wu J, Smith M, Zhang YL et al.: Probing acidreplacements of thiophene PTP1B inhibitors. Bioorg MedChem Lett 2007, 17:2913-2920.

    35. Erbe DV, Wang S, Zhang YL, Harding K, Kung L, Tam M, Stolz L,Xing Y, Furey S, Qadri A et al.: Ertiprotafib improves glycemiccontrol and lowers lipids via multiple mechanisms. MolPharmacol 2005, 67:69-77.

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 281

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283

  • 7/29/2019 1-s2.0-S1367593109000453-main

    11/12

    36.

    Wiesmann C, Barr KJ, Kung J, Zhu J, Erlanson DA, Shen W,Fahr BJ, Zhong M, Taylor L, Randal M et al.: Allosteric inhibitionof protein tyrosine phosphatase 1B. Nat Struct Mol Biol 2004,11:730-737.

    This paper provides insights into the use of allosteric PP inhibitors tobattle selectivity issues.

    37. Takahashi N, Qi Y, Patel HR, Ahima RS: A novel aminosterol

    reverses diabetes and fatty liver disease in obese mice .J Hepatol 2004, 41:391-398.

    38. Genaera_Corp.: Genaera BeginsPhase1b Trial of Trodusquemine(MSI-1436) in Overweight and Obese Type 2 Diabetics. PressRelease (2009) February.

    39. Jiang ZX, Zhang ZY: Targeting PTPs with small moleculeinhibitors in cancer treatment. Cancer Metastasis Rev 2008,27:263-272.

    40. Ostman A, Hellberg C, Bohmer FD: Protein-tyrosinephosphatases and cancer. Nat Rev Cancer 2006,6:307-320.

    41.

    Boutros R, Lobjois V, Ducommun B: CDC25 phosphatases incancer cells: key players?Good targets?. Nat RevCancer2007,7:495-507.

    This comprehensive review describes the role of Cdc25 phosphatases inboth normal and abnormal cell proliferation, provides a critical assess-ment of the current data on Cdc25 overexpression in cancer, anddiscusses both current and future therapeutic strategies for targetingCdc25 activity in cancer treatment.

    42. Rudolph J: Inhibiting transient proteinprotein interactions:lessons from the Cdc25 protein tyrosine phosphatases. NatRev Cancer 2007, 7:202-211.

    43. Garuti L, Roberti M, Pizzirani D: Synthetic small moleculeCdc25 phosphatases inhibitors. Curr Med Chem 2008,15:573-580.

    44. Contour-Galcera MO, Sidhu A, Prevost G, Bigg D, Ducommun B:Whats new on CDC25 phosphatase inhibitors. Pharmacol Ther2007, 115:1-12.

    45. Lazo JS, Nemoto K, Pestell KE, Cooley K, Southwick EC,Mitchell DA, Furey W, Gussio R, Zaharevitz DW, Joo B et al.:Identification of a potent and selective pharmacophore forCdc25 dual specificity phosphatase inhibitors. Mol Pharmacol2002, 61:720-728.

    46. Pu L, Amoscato AA, Bier ME, Lazo JS: Dual G1 and G2 phaseinhibition by a novel, selective Cdc25 inhibitor 6-chloro-7-(2-morpholin-4-ylethylamino)-quinoline-5,8-dione . J Biol Chem2002, 277:46877-46885.

    47. Brezak MC, Quaranta M, Contour-Galcera MO, Lavergne O,Mondesert O, Auvray P, Kasprzyk PG, Prevost GP, Ducommun B:Inhibition of human tumor cell growth in vivo by an orallybioavailable inhibitor of CDC25phosphatases. MolCancer Ther2005, 4:1378-1387.

    48. Cazales M, Boutros R, Brezak MC, Chaumeron S, Prevost G,Ducommun B: Pharmacologic inhibition of CDC25phosphatases impairs interphase microtubule dynamics andmitotic spindle assembly. Mol Cancer Ther 2007, 6:318-325.

    49. Kar S, Wang M, Ham SW, Carr BI: H32, a non-quinone sulfoneanalog of vitamin K3, inhibits human hepatoma cell growth byinhibiting Cdc25 and activating ERK. Cancer Biol Ther 2006,5:1340-1347.

    50. Brohm D, Philippe N, Metzger S, Bhargava A, Muller O, Lieb F,Waldmann H: Solid-phase synthesis of dysidiolide-derivedprotein phosphatase inhibitors. J Am Chem Soc 2002,124:13171-13178.

    51.

    Brohm D, Metzger S, Bhargava A, Muller O, Lieb F, Waldmann H:Natural products are biologically validated starting points instructural space for compound library development: solid-phase synthesis of dysidiolide-derived phosphataseinhibitors. Angew Chem Int Ed Engl 2002, 41:307-311.

    This paper demonstrated that the synthesis of natural product derivedlibraries executed on a polymeric support and employing a variety ofwidely differing synthetic transformations is feasible and that it can beused for the delivery of potent biologically active compounds with highfrequency.

    52. Shimazawa R, Suzuki T, Dodo K, Shirai R: Design andsynthesis of dysidiolide analogs from vitamin D3: novelclass of Cdc25A inhibitors. Bioorg Med Chem Lett 2004,14:3291-3294.

    53. Kar S, Wang M, Carr BI: 2-Methoxyestradiol inhibitshepatocellular carcinoma cell growth by inhibiting Cdc25 andinducing cell cycle arrest and apoptosis. Cancer Chemother

    Pharmacol 2008, 62:831-840.54. Brault L, Bagrel D: Activity of novel Cdc25 inhibitors and

    preliminary evaluation of their potentiation ofchemotherapeutic drugs in human breast cancer cells. Life Sci2008, 82:315-323.

    55. Kar S, Wang M, Yao W, Michejda CJ, Carr BI: PM-20, a novelinhibitor of Cdc25A, induces extracellular signal-regulatedkinase 1/2 phosphorylation and inhibits hepatocellularcarcinoma growth in vitro and in vivo . Mol Cancer Ther 2006,5:1511-1519.

    56. Park H, Bahn YJ, Jung SK, Jeong DG, Lee SH, Seo I, Yoon TS,Kim SJ, Ryu SE: Discovery of novel Cdc25 phosphataseinhibitors with micromolar activity based on thestructure-based virtual screening. J Med Chem 2008,51:5533-5541.

    57. Tartaglia M, Mehler EL, Goldberg R, Zampino G, Brunner HG,

    Kremer H, van der Burgt I, Crosby AH, Ion A, Jeffery S et al.:Mutations in PTPN11, encoding the protein tyrosinephosphatase SHP-2, cause Noonan syndrome. Nat Genet2001, 29:465-468.

    58. Tartaglia M, Niemeyer CM, Fragale A, Song X, Buechner J, Jung A,Hahlen K, Hasle H, Licht JD, Gelb BD: Somatic mutations inPTPN11 in juvenile myelomonocytic leukemia,myelodysplastic syndromes and acute myeloid leukemia. NatGenet 2003, 34:148-150.

    59. Pathak MK, Yi T: Sodium stibogluconate is a potent inhibitor ofprotein tyrosine phosphatases and augments cytokineresponses in hemopoietic cell lines. J Immunol 2001,167:3391-3397.

    60.

    Noren-Muller A, Reis-Correa I Jr, Prinz H, Rosenbaum C,Saxena K, Schwalbe HJ, Vestweber D, Cagna G, Schunk S,Schwarz O et al.: Discovery of protein phosphatase inhibitorclasses by biology-oriented synthesis. ProcNatl AcadSci U S A

    2006, 103:10606-10611.In this work biochemical screening has been applied for the discovery ofunprecedented and selective phosphatase inhibitor classes and demon-strates that biology-oriented synthesis is an efficient approach to thediscovery of new compound classes for medicinal chemistry and che-mical biology research.

    61. Chen L, Sung SS, Yip ML, Lawrence HR, Ren Y, Guida WC,Sebti SM,LawrenceNJ, Wu J: Discovery of a novel shp2 proteintyrosine phosphatase inhibitor. Mol Pharmacol 2006,70:562-570.

    62. Lawrence HR,Pireddu R, Chen L, LuoY, Sung SS,Szymanski AM,Yip ML, Guida WC, Sebti SM, Wu J et al.: Inhibitors of Srchomology-2 domaincontaining proteintyrosine phosphatase-2 (Shp2) based on oxindole scaffolds. J Med Chem 2008,51:4948-4956.

    63. Hellmuth K, Grosskopf S, Lum CT, Wurtele M, Roder N, vonKries JP, Rosario M, Rademann J, Birchmeier W: Specificinhibitors of the protein tyrosine phosphatase Shp2 identifiedby high-throughput docking. Proc Natl Acad Sci U S A 2008,105:7275-7280.

    64. Geronikaki A, Eleftheriou P, Vicini P, Alam I, Dixit A, Saxena AK: 2-Thiazolylimino/heteroarylimino-5-arylidene-4-thiazolidinonesas new agents with SHP-2 inhibitory action. J Med Chem 2008,51:5221-5228.

    65. Stephens BJ,Han H, Gokhale V, VonHoff DD: PRL phosphatasesas potential molecular targets in cancer. MolCancer Ther2005,4:1653-1661.

    66. Saha S, Bardelli A, Buckhaults P, Velculescu VE, Rago C, StCroix B, Romans KE, Choti MA, Lengauer C, Kinzler KW et al.: Aphosphatase associated with metastasis of colorectal cancer.Science 2001, 294:1343-1346.

    282 Next-generation therapeutics

    Current Opinion in Chemical Biology 2009, 13:272283 www.sciencedirect.com

  • 7/29/2019 1-s2.0-S1367593109000453-main

    12/12

    67. Bardelli A, Saha S, Sager JA, Romans KE, Xin B, Markowitz SD,Lengauer C, Velculescu VE, Kinzler KW, Vogelstein B: PRL-3expression in metastatic cancers. Clin Cancer Res 2003,9:5607-5615.

    68. Rouleau C, Roy A, St Martin T, Dufault MR, Boutin P, Liu D,Zhang M, Puorro-Radzwill K, Rulli L, Reczek D et al.: Proteintyrosine phosphatase PRL-3 in malignant cells and endothelial

    cells: expression and function. Mol Cancer Ther 2006,5:219-229.

    69. Pathak MK, Dhawan D, Lindner DJ, Borden EC, Farver C, Yi T:Pentamidine is an inhibitor of PRL phosphatases withanticancer activity. Mol Cancer Ther 2002, 1:1255-1264.

    70. Lee MS, Johansen L, Zhang Y, Wilson A, Keegan M, Avery W,Elliott P, Borisy AA, Keith CT: The novel combination ofchlorpromazine and pentamidine exerts synergisticantiproliferative effects through dual mitotic action. CancerRes 2007, 67:11359-11367.

    71. Ahn JH, Kim SJ, Park WS, Cho SY, Ha JD, Kim SS, Kang SK,Jeong DG, Jung SK, Lee SH et al.: Synthesis and biologicalevaluation of rhodanine derivativesas PRL-3 inhibitors. BioorgMed Chem Lett 2006, 16:2996-2999.

    72. Choi SK, Oh HM, Lee SK, Jeong DG, Ryu SE, Son KH, Han DC,Sung ND, Baek NI, Kwon BM: Biflavonoids inhibitedphosphatase of regenerating liver-3 (PRL-3). Nat Prod Res2006, 20:341-346.

    73. Park H, Jung SK, Jeong DG, Ryu SE, Kim SJ: Discovery of novelPRL-3 inhibitors based on the structure-based virtualscreening. Bioorg Med Chem Lett 2008, 18:2250-2255.

    74. Stewart GR, Robertson BD, Young DB: Tuberculosis: a problemwith persistence. Nat Rev Microbiol 2003, 1:97-105.

    75.

    Koul A, Herget T, Klebl B, Ullrich A: Interplay betweenmycobacteria and host signalling pathways. Nat Rev Microbiol2004, 2:189-202.

    This review highlights strategies employed by mycobacteria to subverthost-cell signaling and identifies key molecules involved in these pro-cesses as potential targets for new antimycobacterial therapies.

    76. Singh R, Rao V, Shakila H, Gupta R, Khera A, Dhar N, Singh A,Koul A, Singh Y, Naseema M et al.: Disruption of mptpB impairsthe ability of Mycobacterium tuberculosis to survive in guineapigs. Mol Microbiol 2003, 50:751-762.

    77. Manger M, Scheck M, Prinz H, von Kries JP, Langer T, Saxena K,Schwalbe H, Furstner A, Rademann J, Waldmann H: Discovery of

    Mycobacterium tuberculosis protein tyrosine phosphatase A(MptpA) inhibitors based on natural products and a fragment-based approach. Chembiochem 2005, 6:1749-1753.

    78. Weide T, Arve L, Prinz H, Waldmann H, Kessler H: 3-Substitutedindolizine-1-carbonitrile derivatives as phosphataseinhibitors. Bioorg Med Chem Lett 2006, 16:59-63.

    79. Correa IR Jr,Noren-Muller A, AmbrosiHD, JakupovicS, SaxenaK,Schwalbe H, Kaiser M, Waldmann H: Identification of inhibitorsfor mycobacterial protein tyrosine phosphatase B (MptpB) bybiology-oriented synthesis (BIOS). Chem Asian J 2007,2:1109-1126.

    80.

    Grundner C, Perrin D, Hooft van Huijsduijnen R, Swinnen D,Gonzalez J, Gee CL, Wells TN, Alber T: Structural basis forselective inhibition of Mycobacterium tuberculosis protein

    tyrosine phosphatase PtpB. Structure 2007, 15:499-509.The development of a potent, selective inhibitor of MptpB was reported.The inhibitor, showed an IC50 of 440 nM and >60-fold specificity forMptpB over six human PTPs.

    81.

    Soellner MB, Rawls KA, Grundner C, Alber T, Ellman JA:Fragment-based substrate activity screening method for the

    identification of potent inhibitors of the Mycobacteriumtuberculosis phosphatase PtpB. J Am Chem Soc 2007,129:9613-9615.

    In this study a new substrate-based fragment approach for the identifica-tion of novel PTP inhibitors was developed. Application of this method toMptpB resulted in the development of the most potent MptpB inhibitorreported to date.

    82. Noren-Muller A, Wilk W, Saxena K, Schwalbe H, Kaiser M,Waldmann H: Discovery of a new class of inhibitors ofMycobacterium tuberculosis protein tyrosine phosphatase Bby biology-oriented synthesis. Angew Chem Int Ed Engl 2008,47:5973-5977.

    83. Chiaradia LD,Mascarello A, Purificacao M, Vernal J, Cordeiro MN,Zenteno ME, Villarino A, Nunes RJ, Yunes RA, Terenzi H:Synthetic chalcones as efficient inhibitors of Mycobacteriumtuberculosis protein tyrosine phosphatase PtpA. Bioorg MedChem Lett 2008, 18:6227-6230.

    84. Hermiston ML, Xu Z, Weiss A: CD45: a critical regulator ofsignaling thresholds in immune cells. Annu Rev Immunol 2003,21:107-137.

    85. Jacobsen M, Schweer D, Ziegler A, Gaber R, Schock S,Schwinzer R, Wonigeit K, Lindert RB, Kantarci O, Schaefer-Klein Jet al.: A point mutation in PTPRC is associated with thedevelopment of multiple sclerosis. Nat Genet2000, 26:495-499.

    86. Bataille R, Robillard N, Pellat-Deceunynck C, Amiot M: A cellularmodel for myeloma cell growth and maturation based on anintraclonal CD45 hierarchy. Immunol Rev 2003, 194:105-111.

    87. Friesen C, Glatting G, Koop B, Schwarz K, Morgenstern A,Apostolidis C, Debatin KM, Reske SN: Breakingchemoresistance and radioresistance with [213Bi]anti-CD45antibodies in leukemia cells. Cancer Res 2007, 67:1950-1958.

    88. Lee K, Burke TR Jr: CD45 protein-tyrosine phosphataseinhibitor development. Curr Top Med Chem 2003, 3:797-807.

    89. Urbanek RA, Suchard SJ, Steelman GB, Knappenberger KS,Sygowski LA, Veale CA, Chapdelaine MJ: Potent reversibleinhibitors of the protein tyrosine phosphatase CD45. J MedChem 2001, 44:1777-1793.

    90. Hamaguchi T, Takahashi A, Kagamizono T, Manaka A, Sato M,Osada H: Synthesis and characterization of a potent andselective proteintyrosine phosphatase inhibitor. 2. Bioorg Med

    Chem Lett 2000, 10:2657-2660.

    91. Kobayashi-Matsunaga Y, Ishii T, Hamaguchi T, Osada H, Sato M:Synthesis and characterization of a novel protein tyrosinephosphatase inhibitor, 2-(cyclobutylamino)-N-(2-furylmethyl)-2-thioxoacetamide. Lett Drug Des Discov 2005, 2:224-227.

    92.

    Castanotto D, Rossi JJ: The promises and pitfalls of RNA-interference-based therapeutics. Nature 2009, 457:426-433.

    This is an excellentreview which summarizes therecent progress in RNA-interference-based therapeutics development.

    93.

    WhiteheadKA, Langer R, AndersonDG: Knocking downbarriers:advances in siRNA delivery. Nat Rev Drug Discov 2009,8:129-138.

    Theauthorsprovidedan updateon theprogressof RNAi therapeuticsandhighlight novel synthetic materials for the encapsulation and intracellulardelivery of nucleic acids.

    94. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R,

    Donoghue M, Elbashir S, Geick A, Hadwiger P, Harborth J et al.:Therapeutic silencing of an endogenous gene by systemicadministration of modified siRNAs. Nature 2004, 432:173-178.

    95. Montalibet J, Kennedy BP: Therapeutic strategies for targetingPTP1B in diabetes. Drug Discov Today: Ther Strat2005,2:129-135.

    Therapeutic potential of phosphatase inhibitors Vintonyak et al. 283

    www.sciencedirect.com Current Opinion in Chemical Biology 2009, 13:272283