throbber
Irreversible Protein Kinase Inhibitors: Balancing the Benefits and
`Risks
`Tjeerd Barf* and Allard Kaptein
`
`Perspective
`
`pubs.acs.org/jmc
`
`Drug Discovery Unit, Covalution Pharma BV, Ravenstein, The Netherlands
`
`■ INTRODUCTION
`
`In the relatively young but expanding field of irreversible kinase
`inhibitor drug discovery, there are two main developments that
`are of central importance. First, the patients have watched the
`first wave of low molecular weight protein kinase inhibitors
`becoming available to them over the past decade. These kinase
`inhibitors have been approved by the U.S. Food and Drug
`Administration (FDA) for
`therapeutic use in oncology
`indications and constitute an important addition to the arsenal
`of drugs to combat cancer (Table 1). Without exception, these
`marketed protein kinase inhibitors have been identified and
`developed using conventional approaches,
`i.e.,
`reversible
`inhibitors that (partly) occlude the ATP pocket in the catalytic
`domain of the kinase. Although protein kinases are regarded as
`an attractive drug target family, it took (and still takes) huge
`efforts to master the human kinome, which comprises more
`than 500 protein kinases. The search for clinically applicable
`kinase inhibitors that target the highly conserved ATP pocket
`has been thwarted by a couple of well-known hurdles.
`Selectivity, cellular potency, and an increasingly crowded
`intellectual property arena are major points of attention.
`The second important development is the renewed interest
`in covalent binding drugs (reviewed by Potashman and Duggan
`and by Singh et al).1,2 This recent revival results from a better
`understanding of the benefits of the covalent binding principle
`and the approval of effective and safe covalent drugs.
`Historically, drug discoverers have been taught to stay away
`from small molecular entities that harbor reactive electrophilic
`groups because these used to be equivalent to promiscuity.3
`Promiscuous hits that relied on reactive groups were tradition-
`ally hard to optimize toward leads, since these were more than
`often interfering with the biochemical assay rather than truly
`modifying the activity of the target of interest.4 Even if the
`target modulation was real,
`indiscriminant
`reactivity was
`believed to trigger
`insurmountable toxic events that may
`surface in late stage clinical
`trials when larger patient
`“suicide
`populations are involved. As a consequence,
`inhibitors”, “warheads”, and covalent
`irreversible inhibitors
`developed a negative flavor over time and became almost
`synonymous with toxicity in some organizations. The
`skepticism toward irreversible drugs may evaporate as more
`examples of
`irreversible drugs progress clinically that
`demonstrate good efficacy and safety margins.
`In a nutshell, the therapeutic applicability or the success of
`irreversible binding kinase inhibitors is dependent on whether
`or not the covalent bond can be confined solely to the protein
`kinase of interest. So this approach is in essence a story about
`two T’s: treatment and toxicity. When relying on the covalent
`binding principle,
`it
`is important
`to discern adduct-based
`toxicity and adduct-based treatment, since the adducting
`
`molecular entities in question obey overlapping fundamental
`rules in terms of reactivity. The only key difference is the nature
`and the function of the proteins that are covalently modified.
`Covalent kinase inhibitors with well-balanced recognition and
`reactivity should provide efficacy, selectivity, and ultimately the
`safety margins that are required for regulatory approval. If we
`strike the right balance, a third “T” will give enough comfort:
`therapeutic window.
`This Perspective aims to give a comprehensive account from
`a medicinal chemist's point of view on the progress of
`irreversible kinase inhibitor drug discovery. The “state of the
`art” is reviewed by means of reported irreversible kinase
`inhibitors profiles and their chemical structures. The potential
`upsides and pitfalls that are associated with this concept are
`highlighted to provide a general understanding of
`the
`differences with respect to conventional drug discovery, as
`well as the future potential of this approach.
`
`■ EFFECT OF RESIDENCE TIME ON THE
`THERAPEUTIC WINDOW
`Residence Time and Efficacy. A kinase inhibitor will only
`be efficacious when it
`is modulating the action of
`the
`physiological kinase and can only do so when the inhibitor is
`bound. This general paradigm was effectively captured by
`Copeland and co-workers, who described the potential
`advantages of
`long residence time in terms of duration of
`action and target selectivity.5 The equilibrium dissociation
`the drug−target binary complex as
`constant (Kd) for
`determined in a closed (in vitro) system is a more accurate
`measure than the IC50 or Ki. Yet the equilibrium setting is
`different in vivo, since the drug concentrations are no longer
`constant, and the efficacy is dependent on the on-rate (kon) and
`even more importantly the off-rate constant (koff). The
`importance of the related occupancy time or half-life for the
`protein−drug complex was corroborated in the context of
`pharmacological agents for whom longer
`residence times
`seemed to be beneficial
`in terms of biological or clinical
`efficacy.6,7 In this respect, kinase inhibitors that exert covalent
`and irreversible binding can achieve the ultimate physiological
`goal: Efficacy is maintained until the target kinase is physically
`restored by the body to physiologically relevant levels (Figure
`1).8 In other words, the pharmacodynamics will be a function
`of the de novo synthesis rate of the target protein rather than
`the trough level of the compound.
`Unlike for kinase inhibitors with a reversible binding mode,
`we anticipate that not all ADME parameters need to be as
`efficiently optimized for covalent
`inhibitors. The general
`
`Received: March 7, 2012
`Published: May 23, 2012
`
`© 2012 American Chemical Society
`
`6243
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`Downloaded via Sharisse Geiger on November 21, 2022 at 14:09:42 (UTC).
`
`See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 1 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`Perspective
`
`Table 1. FDA Approved Protein Kinase Inhibitors (as of March 2012)
`
`generic (brand) name
`imatinib (Gleevec)
`gefitinib (Iressa)
`erlotinib (Tarceva)
`sorafenib (Nexavar)
`sunitinib (Sutent)
`dasatinib (Sprycel)
`nilotinib (Tasigna)
`lapatinib (Tykerb)
`pazopanib (Votrient)
`vandetanib (Caprelsa)
`vemurafinib (Zelboraf)
`crizotinib (Xalkori)
`ruxolitinib (Jakafi)
`axitinib (Inlyta)
`
`year of approval
`2001
`2003
`2004
`2005
`2006
`2006
`2007
`2007
`2009
`2011
`2011
`2011
`2011
`2012
`
`company
`Novartis
`AstraZeneca
`Genetech, OSI
`Bayer, Onyx
`Pfizer
`Bristol-Myers Squibb
`Novartis
`GlaxoSmithKline
`GlaxoSmithKline
`AstraZeneca
`Roche, Plexxicon
`Pfizer
`Incyte
`Pfizer
`
`indication
`chronic myeloid leukemia (CML)
`non-small-cell lung carcinoma (NSCLC)
`NSCLC, pancreatic cancer
`hepatocellular carcinoma, renal cell carcinoma (RCC)
`gastrointestinal stromal tumor (GIST), RCC
`CML
`CML
`breast cancer
`RCC
`thyroid cancer
`CML
`NSCLC (ALK +ve)
`myelofibrosis
`RCC
`
`target kinase
`Abl, c-Kit, PDGFRα/β
`EGFR
`EGFR
`Raf, VEGFR2/3, c-Kit, PDGFRβ
`c-Kit, VEGFR, PDGFR, FLT3
`Abl, c-Kit, PDGFR, Src
`Abl, c-Kit, PDGFR, Src, ephrin
`EGFR, ErbB2
`VEGFR, PDGFRα/β, c-Kit
`VEGFR, EGFR, RET
`Abl, c-Kit, PDGFR, Src, ephrin
`ALK, MET
`JAK1/2
`VEGFR, PDGFRβ, c-Kit
`
`Figure 1. For conventional drugs, the pharmacodynamic effect is driven by the pharmacokinetics of the drug (left). For irreversible covalent drugs,
`the pharmacodynamic effect is driven by the turnover rate (de novo synthesis) of the protein target (right). The red hatched area is drug exposure
`not required for efficacy.
`
`requirements for absorption and distribution characteristics
`remain relatively unchanged with respect to the conventional
`approach. It is still advantageous to have good oral availability
`and rapid distribution, but in the case of an irreversible kinase
`inhibitor it would be favorable to clear the drug from the body
`once the target kinase is maximally occupied. Thus, the in vitro
`and in vivo criteria for optimization cycles with respect to
`metabolism and excretion are very much different and should
`accentuate a swift clearance of the drug.
`Residence Time and Toxicity. Drug-safety-related issues
`pose a serious problem to the pharmaceutical industry and have
`been a major contributing factor to attrition rates in the drug
`trajectories.9 As discussed above,
`since no
`development
`excessive circulating levels are required to maintain efficacy
`with irreversible binding drugs, rapid clearance should lead to a
`lower propensity for off-target related adverse effects. But this
`can only be achieved when off-target protein−drug interactions
`are short-lived because of the noncovalent nature of binding.
`On the other hand, toxic events can also be associated with
`covalent binding, and this can be either on-target or off-target
`related. If the toxicity is a direct consequence of on-target
`modulation, then irreversible inhibition might exacerbate the
`adverse effect potential. In this case, the clinical benefit should
`outweigh the adverse event. Alternatively, prolonged residence
`times can occur if collateral targets are being adducted by the
`potential drug. In that case, a high degree of in vivo selectivity
`can only be accomplished when the off-target turnover rate is
`(much) faster than the on-target resynthesis rate.
`For irreversible kinase inhibitors, the target selectivity thus
`should not only be discussed in terms of reversible binding to
`closely related kinases with respect
`to primary sequence
`homology. Especially kinases that (also) share the site of
`covalent modification are highly relevant to include in the
`selectivity panel. A good level of selectivity for those off-target
`
`kinases is suggestive of a high degree of overall selectivity
`within the human kinome, as well as for other protein classes.
`Also, potential adverse effects due to covalent inhibition of
`specific off-target kinases can be designed out early in the
`optimization cycles to yield a better therapeutic window.
`Much of the aversion to irreversible binders is predominantly
`the result of the negative experiences with reactive metabolites
`that could bind covalently to a variety of proteins.10,11 In such a
`case, the parent drugs are metabolically bioactivated to reactive
`species that potentially can bind covalently to all sorts of
`macromolecules. Next to the predictable intrinsic toxicity, this
`may lead to unpredictable idiosyncratic toxicity (IDT) in late
`stage clinical
`testing and beyond. In order to exclude or
`minimize the propensity of reactive metabolite formation,
`potential drugs are usually evaluated preclinically for (i)
`gluthathione (GSH) adduct formation and depletion when
`incubated with recombinant enzymes or human liver tissue
`preparations and (ii) magnitude of covalent protein binding in
`liver microsomal fractions and in rodents. It is obvious that
`these models are highly relevant
`for the optimization of
`irreversible kinase inhibitors as well. The advantage with
`irreversible kinase inhibitors in this respect is that the parent
`molecules are the reactive species, which should have a better
`predictive power for the reactivity assays and body retention
`models involved.
`Recently, the occurrence of IDT or hypersensitivity reaction
`has been linked by Nakayama et al. to the magnitude of
`covalent and irreversible binding to off-targets and the daily
`dose required in patients.12 A classification system to assess the
`risk of IDT was proposed based on covalent binding to human
`hepatocytes as the best predictor, in combination with the daily
`dose.
`that has to be considered for
`Another potential event
`covalent and irreversible kinase inhibitors is hapten formation,
`
`6244
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 2 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`i.e., triggering an immune response to the adducted protein.13
`Experiences with reactive compounds and reactive metabolites
`indicate that additional danger signals (e.g., mitochondrial
`damage) are required next to the hapten mechanism to induce
`an immune response (for a review, see Zhang et al).14 Except
`for the β-lactam antibiotics, absence of reports of
`immune
`responses for recently marketed irreversible binding drugs does
`not support this concern as a general problem for covalent
`drugs.
`Reactive Metabolite Considerations. Considerations
`that have been used historically within the pharmaceutical
`industry to assess the fate of conventional drugs with suspected
`reactive metabolites can provide guidance for
`irreversible
`binding kinase inhibitors as well.10 It
`is obvious that
`the
`“avoidance strategy”,
`i.e., complete elimination of reactive
`groups, cannot be applied on warhead-containing drugs, since
`the pharmacological effect is largely dependent on the reactive
`group. Perhaps it would be better to develop a reversible
`therapy for less severe disorders, especially if one can rationalize
`that reversible drugs should work equally well. Also,
`the
`anticipated human dose (for any drug) has to be considered, as
`the body burden has to be kept to a minimum (vide infra). In
`addition, a disease that needs acute dosing rather than chronic
`treatment might be more appropriate to engage with
`irreversible binding drugs. These and other questions need to
`be addressed before a covalent (kinase) inhibitor program can
`be progressed toward clinical development. Obviously, the key
`issue here is the benefit/risk ratio, and serious life threatening
`diseases may be particularly justified in this scenario.
`reactive
`One has to keep in mind though that most
`metabolites are by no means
`tuned, and they can be
`uncontrollable like “unguided missiles”. In contrast, deliberately
`designed irreversible kinase inhibitors can be subjected to safety
`assays during optimization cycles
`to ensure selection of
`compounds that meet stringent safety prediction criteria. In
`some other instances, bioactivation is even required to generate
`the active drug, such as in the case of proton pump inhibitors
`like omeprazole and P2Y12 receptor antagonist clopidogrel.1 No
`matter the type of covalent approach involved, a decision tree
`that incorporates several safety assessment parameters will be
`required to mitigate the risks.
`In addition,
`the level of
`acceptance that has been set for the magnitude of covalent
`binding of reactive metabolites in vitro and in vivo also applies
`for covalent binding parent drugs.
`
`■ THE CONCEPT APPLIED TO KINASES
`
`the members belonging to the
`The question is whether
`superfamily of protein kinases are suitable targets at all for
`irreversible inhibition. First of all, the replenishment of the
`target kinase has to be sluggish enough to allow drug dosing
`once or twice a day. This has to be checked on a kinase-by-
`kinase basis. Second, unlike for instance the enzyme families of
`(cysteine) proteases or phosphatases, kinases lack a catalytically
`active amino acid residue that is key to the function of a
`particular enzyme by virtue of its overt nucleophilicity. The
`relatively unreactive cysteines, serines, threonines, and lysines
`in kinases may have to be exposed to excessive reactivity to
`ensure covalent bond formation. There are plenty of examples
`of overly reactive electrophiles
`that despite the best of
`intentions, in retrospect never had a chance to make it into a
`safe drug. Nevertheless, these prototypical molecules definitely
`marked the beginning of this emerging platform.
`
`Perspective
`
`Non-ATP Competitiveness. There is one major upside
`that can specifically be exploited in the kinase field. Non-ATP
`competitive inhibitors offer distinct advantages to conventional
`ATP competitive binders, especially in terms of efficacy and
`selectivity.15,16 The highly conserved nature of the ATP binding
`pocket poses a challenge in the identification of selective kinase
`inhibitors, but more important is that high intracellular ATP
`concentrations affect the cellular potency of ATP competitive
`kinase inhibitors under physiological conditions.17 Besides
`allosteric binders of protein kinases, blockade of the ATP
`pocket with an irreversible inhibitor has emerged as an
`attractive and alternative strategy to achieve non-ATP
`competitive inhibition of kinase-mediated signaling.18 As a
`consequence, caution should be taken with the interpretation
`and/or comparison of the biochemical and functional IC50
`values, since irreversible inhibitors usually become more potent
`over time. The selection of the length of preincubation time
`will
`thus
`influence the observed value, unless maximal
`inhibition is achieved instantly. The inactivation of the kinase
`involves two steps: formation of a reversible complex based on
`affinity (Ki) followed by the covalent bond formation between
`the inhibitor and the kinase (kinact).19 The first step depends on
`the ATP level used in the various assays reported. The second
`step is primarily influenced by the inherent reactivity of the
`electrophilic group, as well as the distance to the nucleophilic
`trap. The better the juxtaposition of the reacting partners, the
`better the inactivation step. As the kinase is effectively removed
`from the equilibrium with ATP and substrate, a shift in IC50 is
`expected and, as demonstrated in Figure 2, the inhibitor shows
`an increasingly non-ATP competitive character as
`time
`progresses. Consequently, this would be beneficial for protein
`kinases with a low Km,ATP in particular.
`Many research groups have already embarked intentionally,
`or unintentionally, on the search for
`irreversible kinase
`inhibitors (partly reviewed by Garuti et al).20 In some instances,
`these inhibitors have become invaluable assets in the target
`
`Figure 2. IC50 determination depends on the ATP concentration and
`preincubation time for an irreversible kinase inhibitor (left), whereas
`an ATP competitive kinase inhibitor
`is only affected by ATP
`concentration (right). Data are from an unpublished kinase inhibitor
`study included for illustration purposes only.
`
`6245
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 3 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`Perspective
`
`Figure 3. Examples of lysine trapping kinase inhibitors.
`
`Figure 4. Schematic representation of available cysteines in or near the ATP binding pocket of kinases in the active conformation (based on the
`crystal structure of interleukin 2 tyrosine kinase, PDB code 1SM2). The spheres indicate the locations of cysteines that are in principle accessible for
`covalent modification: group 1, glycine-rich- or P-loop (dark blue); group 2, the roof of the ATP binding pocket (pink); group 3, hinge region and
`front pocket (yellow); group 4, DFG-motif neighboring cysteine (light blue). Group 5 (in dark red) denotes the cluster of cysteines that resides on
`the activation loop. The gatekeeper group is shown in orange. Adapted by permission from Macmillan Publishers Ltd.: Nature Reviews Cancer
`(http://www.nature.com/nrc/index.html), Copyright 2009.38
`
`validation stage. Others were deliberately designed to be the
`drugs of the future. The selection of examples that is reviewed
`in this Perspective covers both and in addition is primarily
`based on the diversity of cysteine locations within the known
`druggable binding regions of kinases. The electrophilic portions
`of these inhibitors (highlighted in blue in this Perspective) are
`either being trapped by the highly conserved lysine or cysteines
`at various positions of the kinases. To our knowledge, there are
`no reports to date that describe covalent kinase inhibitors that
`adduct other amino acid residues, such as serine or threonine.
`Lysine-Trapping Covalent Kinase Inhibitors. Because of
`the well-conserved nature and its abundant presence, the lysine
`involved in the phosphate transfer machinery is not the most
`ideal candidate amino acid for trapping electrophilic inhib-
`itors.21 This particular lysine resides in virtually every ATP-
`binding pocket of the known human kinases. Nevertheless, a
`couple of lysine trapping covalent inhibitors have appeared in
`the first reported is ATP analogue 5′-
`literature. One of
`fluorosulfonylbenzoyl adenosine (FSBA, 1), a tool frequently
`used as an affinity label for kinases like EGFR,22 p38γ,23 and
`brain PI4K (Figure 3).24 As per example, Fox and co-workers
`used 1 for analysis of kinetic mechanisms and ATP-binding site
`reactivity toward p38γ and found it to bind irreversibly to the
`unphosphorylated and active form.23 Digest maps showed that
`Lys56 of p38γ was selectively and covalently modified by 1.
`The use of 1 has been reported in more than 200 publications,
`underscoring the usefulness of this particular chemical probe.
`
`Bell and co-workers discovered a pyrrole-5-carboxaldehyde
`class of type 1 insulin-like growth factor receptor (IGF-1R)
`inhibitors with modest activity but good selectivity over a small
`panel of other kinases.25 These aldehyde-based inhibitors were
`shown to bind in the ATP-binding pocket of IGF-1R in a
`covalent but reversible manner via imine formation between
`Lys1003 and the aldehyde functionality. Reduction of the imine
`adduct with sodium borohydride irreversibly inactivated the
`kinase, enabling a tryptic digest and X-ray crystallography
`examination. A representative example is inhibitor 2, which
`demonstrated an IC50 of 0.49 μM for IGF-1R.
`A frequently studied lipid kinase inhibitor is represented by
`antiproliferative agent wortmannin (3, Figure 3). This PI3K
`inhibitor modifies the ATP pocket covalently and irreversibly
`via trapping of conserved Lys802 (p110α numbering).26
`Analogues of 3 inhibit PI3K (reported IC50 values in the low
`nanomolar range) by virtue of an activated furan ring as a fairly
`unusual electrophile, which opens upon nucleophilic attack of
`the side chain of Lys802.27 SAR studies on analogues of 3
`demonstrated that omission (or modification) of this furan ring
`results in significant reduction in inhibitory activity. An X-ray
`crystallographic structure revealed the exact nature of
`the
`binding mode of 3, and clear density was observed for the
`primary amine linking to the opened furane ring.28 Later on, 3
`was also shown to inhibit the protein kinase Plk1 (IC50 = 24
`nM), shedding some doubt on the pharmacology that was
`previously attributed to PI3K inhibition.29 Subsequently, it was
`
`6246
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 4 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`established that 3 also inhibits Plk3 by covalent modification of
`the conserved lysine in the ATP binding pocket.30 In general,
`deliberately designed covalent drugs that target the invariant
`lysine in kinases are not to be expected. However, uniquely
`positioned lysines elsewhere in or near the ATP binding pocket
`may well be suitable for a covalent inhibitor approach.
`Cysteine-Trapping Covalent Kinase Inhibitors. Unlike
`cysteines
`in kinases, cysteines
`involved in the catalytic
`for instance, protein tyrosine phosphatases31
`machinery in,
`and cysteine proteases32 are part of very specific environments
`made up of neighboring amino acids and ternary architecture of
`these particular proteins. Part of the active site architecture is
`the position of
`these cysteines, residing frequently at
`the
`“bottom” of one or more α-helixes. Backbone NH groups point
`downward generating a net positive charge that stabilizes the
`sulfhydryl anion and augments nucleophilicity.33 The pKa of a
`thiol in a typical cysteine residue is ∼8.5, whereas the presence
`of a polar or positively charged environment can decrease the
`pKa up to 5 log units. In many protein families, cysteines
`contribute to maintaining ternary protein structures via
`disulfide bond formation.
`In the ATP-binding pocket of
`kinases,
`reduced cysteines generally do not seem overly
`nucleophilic and the side chain functionality may have a
`rudimentary role at most. There are, however, several recent
`reports that suggest that reaction of a cysteine thiol can regulate
`and control even protein kinase function,
`for instance, by
`means of redox signaling34,35 or S-alkylation with endogenous
`electrophiles.36,37 For this very same reason, reducing agents
`such as dithiothreitol (DTT) or dithioerythritol (DTE) are
`indispensable ingredients in biochemical assays for kinases, in
`order to keep the enzyme in an active and sensitive state. This
`also supports the notion that these cysteines are sensitive
`enough for electrophilic modification by designer drugs.
`Recently, Zhang et al. have suggested a very useful
`classification system that originally binned the cysteines into
`four groups according to the relative positions in the ATP
`binding pocket (Figure 4).38 It is justified to add at least two
`additional groups to this initial classification,
`involving the
`cysteines located at the gatekeeper position and a fifth group.
`Group 5 covers cysteines residing at various locations in the
`activation loop. Some cysteines are more unique than others,
`and there are at least 200 kinases that seem tractable for a
`covalent approach. Given the known conformational flexibility
`of kinases, this number may be even higher. Structural analyses
`of the different conformations, including the active kinase state
`and inactive “DFG-out” and “C-helix-out” conformations,
`unveil additional cysteines that could be targeted.39 These
`cumulative analyses generate a fingerprint of the kinome active
`site toward cysteines, and the resulting “cysteinome” is of great
`value for the design of covalent kinase inhibitors. There are
`now several examples of covalent kinase inhibitors that target
`this region, and in the following sections the several groups are
`reviewed. The order of appearance is dictated by the group
`numbering as indicated in Figure 4.
`Group 1 Cysteines. So far, there is only one report that
`describes
`the identification of covalent kinase inhibitors
`targeting a cysteine on the lip of the glycine-rich loop (Figure
`4). An irreversible group 1B inhibitor
`is represented by
`fibroblast growth factor receptor (FGFR) inhibitor 4 (FIIN-1,
`Figure 5), which was identified via a structure-based approach
`using available reversible FGFR inhibitors.40 The FGFR family
`consists of four members, FGFR1−4 and plays an important
`role in tumor formation and progression as well as wound
`
`Perspective
`
`Figure 5. (Top) Chemical structure of FGFR inhibitor 4. (Bottom)
`The human kinome and the group 1B cysteines. Adapted illustration
`reproduced with permission from AAAS (Manning, G.; Whyte, D. B.;
`Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase
`complement of the human genome. Science 2002, 298, 1912−1934)
`and courtesy of Cell Signaling Technology, Inc. (www.cellsignal.
`com).42
`
`healing.41 Compound 4 is a biochemically potent acrylamide-
`based inhibitor of FGFR1, FGFR2, and FGFR3 (IC50 values of
`9.2, 6.2, and 11.9 nM, respectively) and is somewhat less potent
`on FGFR4 (IC50 = 189 nM), with decent selectivity over the
`other group 1B cysteine kinases, such as Src, YES, and TNK1
`(Figure 5). The reversible propionyl analogue of 4 was 24- to
`100-fold less potent in FGFR1 and FGFR3 harboring cells,
`respectively. With the aid of a biotinylated version of 4 (Figure
`17), specific covalent wild type (WT) FGFR1 labeling was
`demonstrated in transfected HEK293 cells whereas the C486S
`mutant was barely modified. This is in line with the binding
`model that strongly suggests that 4 forms a covalent bond with
`FGFR1 via Cys486.
`Group 2 Cysteines. Within this cluster, subgroup 2B
`comprises only 11 kinase members and has been the sole
`subject of proven covalent kinase targeting within the group 2
`cysteines. Cohen et al. were the first to exploit this particular
`cysteine as a selectivity filter in addition to the threonine
`gatekeeper residue in p90 ribosomal protein S6 kinase (RSK).43
`The four closely related RSKs (RSK1−4) all have the P-loop
`cysteine, whereas RSK3 has
`the sterically demanding
`
`6247
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 5 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`Perspective
`
`Figure 6. (Left) Human kinome and the group 2B cysteines. Adapted illustration reproduced with permission from AAAS (Manning, G.; Whyte, D.
`B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, 1912−1934) and courtesy of
`Cell Signaling Technology, Inc. (www.cellsignal.com).42 (Right) Chemical structures of group 2B inhibitor 5−8 and group 2C kinase inhibitor 9.
`
`methionine as a gatekeeper as opposed to threonine in the
`other three RSK kinase members. This was sufficient to develop
`a fluoromethylketone (FMK) derivative 5 as a highly potent
`and selective RSK1 and RSK2 inhibitor, with an IC50 value
`against RSK2 of 15 nM (Figure 6). Selective covalent labeling
`of WT (but not the C436V mutant) RSK2 was demonstrated
`by FMK-biotin in transfected HEK293 cells. Also, Michael
`acceptors such as an acrylate or acrylonitrile were appended
`from the same pyrrolopyrimidine scaffold, yielding irreversible
`RSK2 inhibitors with modest biochemical IC50 values.44 By
`contrast, the doubly activated electrophiles, as in cyanoacryla-
`mide 6, were 200-fold more potent (IC50 = 4 nM) in enzymatic
`assays than the monoactivated vinyl groups, yet in a fully
`reversible covalent binding mode. Owing to its greater
`reactivity, 6 demonstrated faster kinetics than 5 in cellular
`settings, but the duration of occupancy seemed to rely on
`protein resynthesis and was indistinguishable between the two
`inhibitors. Intriguingly, all RSK kinases also accommodate
`another cysteine which is located in the back of the ATP
`binding pocket, equivalent to Cys560 in RSK2 (group 4).45
`Potential trapping of derivatives of 5 by this cysteine was not
`investigated.
`NEK2 is a centrosomal kinase that interferes with the spindle
`assembly checkpoint, affecting mitosis. It harbors a cysteine
`(Cys22) in an identical position as the RSK family of kinases.46
`Henise and Taunton developed a reasonably potent (IC50 =
`770 nM) and cell permeable irreversible NEK2 inhibitor (7)
`based on an oxindole scaffold using a propynamide as the
`warhead. Selective covalent NEK2 inhibitors, such as 7, are
`important validation assets to investigate the consequences of
`intracellular inactivation of NEK2 on regulating the prolifer-
`ation of tumor cells overexpressing this kinase.
`The same specific cysteine on the P-loop of MEKK1 was
`previously identified as a glutathionylation site following
`
`oxidative stress.47 Bioactive nutrients harbor electrophilic
`isothiocyanates
`that
`intervene with stress-signaling kinase
`pathways. Reported activities include cell cycle effects and
`protection of experimental carcinogenesis in animal models.
`The SAPK/JNK signaling pathway was investigated by means
`of phenylethyl isothiocyanate (PEITC) 8, which was shown to
`inhibit WT MEKK1 (group 2B) but failed to intervene with the
`C1238V mutant in both biochemical and cellular assays.48 It
`was the second covalent kinase inhibitor reported for this
`subgroup, and although far from being druglike, it demonstrates
`the potential for covalent kinase inhibitors as research tools in
`the exploratory biology of kinase signaling pathways.
`One group 2C suspect has been reported, which is a p38α
`inhibitor with moderate biochemical potency (IC50 = 200 nM).
`Dialkynylimidazole derivative 9 did covalently modify p38α
`following prolonged incubation and as determined by ESI-
`MS.49 The authors propose trapping via an intricate cyclization
`and rearrangement mechanism involving both ethynyl groups
`of 9, but the exact mode of action and site of covalent binding
`were not confirmed. Cys39 could be adducted given its position
`in the roof of the ATP pocket of p38α and the known binding
`mode of the imidazole class of p38 inhibitors.50 However, the
`cysteine sulfhydryl group is directed away from the pocket
`itself, and as a consequence, this cysteine is unlikely to be
`involved in adduct formation.
`Group 3 Cysteines. The 3F cluster of the group 3 cysteine-
`containing kinases has received by far the most attention
`because of the clinical potential of kinase inhibitors belonging
`to this subgroup.18 In addition, the relatively high nucleophil-
`icity with respect to the other cysteines facilitates the reaction
`with all sorts of electrophilic agents. The pKa of the sulfhydryl
`group of this particular cysteine is most likely lower than
`average because of the position at the end of a C-lobe α-helix
`(Figure 4). The net positive charge through the backbone NH
`
`6248
`
`dx.doi.org/10.1021/jm3003203 | J. Med. Chem. 2012, 55, 6243−6262
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1030, p. 6 of 20
`
`

`

`Journal of Medicinal Chemistry
`
`thus making it more
`groups stabili

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket