You are on page 1of 15

REVIEWS

Flying under the radar: the new wave


of BCR–ABL inhibitors
Alfonso Quintás-Cardama, Hagop Kantarjian and Jorge Cortes
Abstract | The introduction of the BCR–ABL kinase inhibitor imatinib mesylate (Gleevec;
Novartis) revolutionized the treatment of chronic myeloid leukaemia (CML). However, most
patients with CML receiving imatinib still harbour molecular residual disease and some
develop resistance associated with ABL kinase domain mutations. The second-generation
BCR–ABL inhibitors nilotinib (Tasigna; Novartis) and dasatinib (Sprycel; Bristol–Myers Squibb)
have shown significant activity after imatinib failure in clinical trials, but still face similar
obstacles to imatinib, including negligible activity against the frequent BCR–ABL T315I
mutation and modest effects in advanced phases of CML. Various medicinal chemistry efforts,
in part aided by structural studies of the ABL kinase–imatinib complex have resulted in the
synthesis of a new generation of BCR–ABL inhibitors, some of which have shown encouraging
preliminary activity in clinical trials, including against T315I mutants. Here, we discuss these
emerging therapies, which have the potential to improve the outcome of patients with CML.

Chronic myeloid leukaemia (or chronic myelogenous targets (FIG. 1). In this respect, the remarkable clinical
leukaemia; CML), which is characterized by increased success of the ABL tyrosine kinase inhibitor (TKI)4,5
and unregulated proliferation of predominantly myeloid imatinib mesylate (formerly STI571 or CGP 5148B;
cells in the bone marrow, occurs most commonly in the Gleevec, Novartis) in the treatment of patients with
middle-aged and elderly and accounts for 15–20% of all CML has highlighted the potential of molecularly tar-
cases of adult leukaemia in Western populations. The geted anticancer therapies, and has sparked the exten-
underlying cause of CML is a characteristic reciprocal sive development and use of such agents in cancers in
translocation between chromosomes 9 and 22, which general6,7.
cytogenetically results in the Philadelphia chromo- Protein kinases such as ABL have evolved highly
some (Ph) and molecularly gives rise to the chimeric specialized mechanisms for transitioning between
BCR–ABL1 gene1,2. In CML, the protein product of this active and inactive states, and crystal structures of inac-
hybrid gene is a 210 kDa constitutively active protein tive kinases have revealed a remarkable plasticity in
kinase containing 902 or 927 amino acids of BCR fused the kinase domain, facilitating the adoption of distinct
to exons 2–11 of ABL1,2. In addition, the BCR–ABL1 conformations8. Imatinib, a 2‑phenylaminopyrimidine
transcript is present in approximately 25% of patients derivative, binds to the activation loop of ABL kinase
with B‑cell acute lymphoblastic leukaemia (B-ALL). Of outside of a highly conserved ATP binding site, which
these patients with B-ALL, two-thirds express a splic- traps the kinase in an inactive conformation9. In doing
ing variant of BCR–ABL1 that gives rise to a 190-kDa so, imatinib inhibits activity of the kinase and induces a
Department of Leukemia, BCR–ABL protein, whereas the remaining patients complete cytogenetic response (CCyR) in more than 80%
Unit 428, The University express a 210-kDa product. It has been suggested that of patients with CML in chronic phase10. Noteworthy are
of Texas M. D. Anderson
patients with CML carrying p190BCR–ABL1 may have a the annual rates of disease progression to the acceler-
Cancer Center, 1515
Holcombe Blvd, Houston, worse prognosis than those expressing the classical ated or blastic phases among patients in chronic phase
Texas 77030, USA. p210BCR–ABL1 isoform3. after 5 years of follow-up — being 1.5%, 2.8%, 1.6%,
Correspondence to A.Q.-C. BCR–ABL kinase drives the pathogenesis of BCR– 0.9% and 0.6% over the respective years10. However, the
e‑mail: ABL1-positive leukaemia through the phosphorylation initial optimism following these results was tempered by
aquintas@mdanderson.org
doi:10.1038/nrd2324
and activation of a broad range of downstream substrates the realization that the majority of patients with CML
Published online 14 that play critical roles in cellular signal transduction and receiving imatinib retain residual leukaemic cells that
September 2007 transformation, and thus represent potential therapeutic were detectable by molecular techniques10, and that any

834 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

to which imatinib binds9,13,18,19. The impaired interaction


between imatinib and ABL kinase as a consequence of
Y177 mutations within the ABL kinase is best exemplified by
the T315I mutation, which has frequently been detected
BCR ABL in patients with imatinib-resistant CML20, but also in
some patients with imatinib-naive CML13. This threo-
GRB2 P
P
nine residue at position 315, also called the gatekeeper,
STAT
SOS GAB2 is located near the ABL catalytic domain in the centre
of the imatinib binding site, which controls access to a
RAS SFK hydrophobic region of the enzymatic active site. Different
?
(HCK, LYN) BCR–ABL mutants have been associated with differ-
SHP2 PI3K
ent degrees of fitness (oncogenicity) relative to that of
JNK wild-type BCR–ABL. Gain-of-fitness has been shown to
P
AKT ICSBP correlate with increased kinase activity of the enzyme
and enhanced transformation potential. A notable excep-
JUN
tion is the T315I mutation, which displays enhanced
oncogenicity despite reduced kinase activity for classi-
cal ABL substrates21. Using mass-spectrometry-based
phosphotyrosine profiling techniques, it has been shown
BCL2 STAT5 that T315I is associated with a specific phosphorylation
BCLX
pattern in the phosphate binding loop of BCR–ABL21.
Nucleus This might explain the distinct oncogenic potential of
Figure 1 | Oncogenic signalling of BCR–ABL kinase. This highly simplified scheme T315I relative to wild-type BCR–ABL and the closely
illustrates some of the more important BCR–ABL signalling pathways. Phosphorylation at related T315A mutation, which at the same time unveils
Nature
the Y177 residue of BCR generates a high-affinity docking site Reviews | Drug Discovery a previously unknown role of T315I in substrate recog-
for growth-factor
receptor-bound protein 2 (GRB2), which in turn binds the scaffolding adaptor GRB2- nition21. Mutations at other positions within the ABL
associated binding protein 2 (GAB2), as well as SOS (a guanine-nucleotide exchanger of kinase other than T315I weaken the binding of imatinib
RAS), resulting in RAS–MAPK activation (leading to BCL2 gene transcription). to various degrees, which in some cases is enough to
On phosphorylation by BCR–ABL, GAB2 recruits SHP2 (also known as PTPN11) and phos- confer clinical resistance.
phatidylinositol-3 kinase (PI3K), which leads to AKT activation. BCR–ABL also activates
The T315I mutation poses a formidable therapeutic
other signalling pathways such as signal transducer and activator of transcription 5
(STAT5), leading to BCLX gene (also known as BCL2L1) transcription and the SRC family
challenge as not only does it mediate complete resist-
kinases (SFKs) LYN and HCK. By contrast, BCR–ABL represses interferon consensus ance to imatinib, but also to many of the next generation
sequence binding protein (ICSBP) transcription through an unknown mechanism, which of ABL kinase inhibitors, including dasatinib (Sprycel;
releases the ISCBP-mediated inhibition of BCL2 and BCLX gene transcription. Notably, Bristol–Myers Squibb) and the imatinib-related com-
downregulation of ICSBP transcripts has been consistently documented in patients with pound nilotinib (Tasigna; Novartis). Although less
chronic myeloid leukaemia (CML). So, the net effect of BCR–ABL kinase activation is the frequent than ABL kinase domain mutations, other
promotion of cell proliferation and survival through activation of the RAS, SHP2 and mechanisms linked to ABL TKI resistance have been
PI3K–AKT signalling pathways that lead to increased BCL2 and BCLX expression and identified in vivo, such as BCR–ABL1 gene amplifica-
inhibition of ICSBP transcription. Small molecules targeting BCR–ABL kinase represent tion, overexpression of the BCR–ABL protein, activa-
the current mainstay of CML therapy. Pointed arrows indicate direct interactions and/or
tion of SRC family kinases (SFKs) 17,22 and transporters
activations. Blunt-ended arrows indicate inhibitory effects. P, phosphate.
involved in drug efflux, suggesting that targeting this
group of proteins could be a useful characteristic for
agents to treat CML.
response observed in patients in the more advanced The need for new strategies to treat imatinib-resistant
stages of CML (accelerated and blastic phases) are CML has stimulated considerable efforts to develop novel
typically short-lived11. Moreover, patients treated with BCR–ABL inhibitors. Two such agents — dasatinib and
imatinib may eventually develop resistance, particularly nilotinib — have already demonstrated success against
those treated in the accelerated or blastic phases. imatinib-resistant CML in clinical trials. Furthermore,
A pervading theme regarding resistance to ABL TKI propelled by advances in structural biology that have
therapy in CML is the development of point mutations aided rational inhibitor design (BOX 1), a plethora of
within the kinase domain of BCR–ABL1. The frequency novel compounds have proved highly active against
of BCR–ABL1 mutations in imatinib-resistant patients the BCR–ABL protein kinase in preclinical studies,
ranges from 40–90% depending on the CML phase and and some have already shown promising results in pre-
the methodology of detection and definition of resist- liminary clinical studies in patients with CML. Several
ance12–17. To date, more than 50 distinct point mutations of these agents inhibit the kinase activity of BCR–ABL
encoding single amino-acid substitutions in the kinase through mechanisms of action other than interference
domain of the BCR–ABL1 gene have been detected in with the ATP binding site of the kinase while preserving
patients with imatinib-resistant CML. At the protein their specificity for BCR–ABL. This may provide a more
level, these point mutations result in distorted configu- favourable toxicity profile in vivo and overcome mecha-
rations of the ABL kinase–imatinib interface. The result nisms of resistance to imatinib, dasatanib and nilotinib,
is that ABL is unable to adopt the inactive conformation including the T315I mutation in some cases.

nature reviews | drug discovery volume 6 | october 2007 | 835


© 2007 Nature Publishing Group
REVIEWS

Box 1 | Rational design of kinase inhibitors which participates in hydrogen-bond interactions with
both Ile360 and His361, increases affinity for the inac-
Protein kinases are molecular switches that have evolved highly specialized tive conformation of wild-type BCR–ABL by 20-fold
conformational plasticity for transitioning between inactive and active states. to 30-fold, while similar activity against mast/stem-cell
The protein kinase complement of the human genome (that is, kinome) encompasses
growth factor receptor (KIT; IC50 of 60 nM) and platelet-
more than 500 protein kinases. Several members of the serine/threonine and the
derived growth factor receptor (PDGFR; IC50 of 57 nM)
tyrosine family of protein kinases play crucial roles in cancer. To date, 90 genes
encoding tyrosine kinases have been identified, of which 58 encode receptors and is maintained32.
32 encode non-receptor tyrosine kinases123. Many, albeit not all, protein tyrosine Dasatinib is a multikinase inhibitor with potent activ-
kinases are involved in cellular-signalling pathways — the deregulation of which has ity against BCR–ABL kinase (IC50 <1 nM) and SFKs (IC50
been implicated in the development of tumorigenesis124. of 0.2–1.1 nM)31 that has been approved by the US Food
The highly conserved kinase domain of protein kinases consists of a bi-lobed and Drug Administration (FDA) for the treatment of
structure in which Mg-ATP is located in a deep cleft between the N‑terminal and patients with CML following failure or with intolerance
C‑terminal lobes125. Most tyrosine kinase inhibitors (TKIs) have been designed to target to imatinib therapy. Unlike other ABL or SFK inhibitors,
the ATP binding site of the kinase in the active conformation. TKIs acting in this manner
dasatinib was originally designed as an immunosuppres-
have been termed type I kinase inhibitors. By contrast, type II kinase inhibitors bind
preferentially to an inactive conformation of the kinase, locking it in that state and
sant. A potentially advantageous feature of dasatinib
preventing its activation125. In addition to the ATP binding cleft, type II inhibitors bind over imatinib and nilotinib is its ability to bind ABL
an adjacent hydrophobic pocket created by the activation loop, in which the with greater affinity, due, at least in part, to it recogniz-
phenylalanine of the conserved motif DFG (Asp-Phe-Gly) swings more than 10 Å away ing multiple states of the enzyme25,31,33. The resolution
from its position (DFG-out) in the kinase active conformation125,126. TKIs targeting this of the co-crystal structure of dasatinib in complex with
hydrophobic pocket (for example, imatinib, BIRB‑796) bind a group of amino acids that the active conformation of ABL appears to support this
are less conserved than those surrounding the ATP binding site, which has been hypothesis34,35. As a result, the number of BCR–ABL1
proposed as an ideal target for the design of highly selective type II TKIs (FIG. 3). This mutants that confer resistance to dasatinib is limited
concept of high TKI selectivity is further supported by the fact that many kinases
almost exclusively to direct contact sites20,36.
cannot adopt an inactive conformation128. The DFG-out conformation has been solved
crystallographically for a select group of kinases, including ABL; mast/stem-cell growth A potential limitation of these compounds, par-
factor receptor (KIT); Aurora A (AURKA; also known as STK6); epidermal growth- ticularly of dasatinib, is that their increased potency
factor receptor (EGFR); p38 (also known as MAPK14); KDR; and BRAF125. So, type II may be associated with untoward off-target toxicities,
TKIs rationally designed in this manner could have high affinity with a restricted which probably relate to their inhibitory activity against
spectrum of inhibitory activity against various oncogenic protein kinases expressed in a broader range of protein kinases than imatinib. For
haematological malignancies and solid tumours. instance, dasatinib, in addition to inhibiting SFKs in the
An alternative approach to obtain highly selective TKIs, which may be used when subnanomolar range, is also a potent inhibitor of the
the cancer (for example, chronic myeloid leukaemia) is resistant to ATP-competitive
KIT, PDGFR and ephrin receptor (EPHA2) tyrosine
compounds, is to synthesize compounds with affinity for binding sites that,
although physically distant from the ATP binding site, regulate the kinase activity127. kinases, which are directly implicated in haematopoi-
A prime example of such an approach is the development of ‘substrate competitive’ esis, control of tissue interstitial-fluid pressure and ang-
compounds that block the binding of the kinase to its substrate, thereby preventing iogenesis. These effects may provide the physiological
substrate phosphorylation (FIG. 3). explanation for some of the toxicities associated with
dasatinib therapy, such as myelosuppression and pleural
effusion. However, dasatinib-mediated SFK inhibition
With the excitement generated by the success of might be beneficial in cases of imatinib-resistant CML
dasatinib and nilotinib, many of these new agents have that are due to overexpression of SFKs such as LYN22, as
been ‘flying under the radar screen’. In this article, we well as in Ph-positive B-ALL in which SFKs seem to play
summarize these recent advances in the discovery and an important pathogenetic role.
development of kinase inhibitors for CML, and discuss Recently, imatinib-mediated ABL kinase inhibi-
the potential of these agents as a more effective treat- tion has been linked to the potential development of
ment for CML. Ideally, the clinical development and full cardiotoxicity in patients with CML37. This contention
approval of these agents should be expedited by the suc- has been supported by experimental models in which
cess of their immediate predecessors so that they could imatinib-treated mice develop left-ventricular contrac-
become available to patients who may benefit from their tile dysfunction. This toxic myopathy appears second-
use and to researchers to continue their endeavours on ary to imatinib-induced activation of the endoplasmic
the ever more realistic enterprise of curing CML. reticulum stress response and cell death37. Although only
12 out of 1,995 (0.6%) patients with CML treated with
The second generation of BCR–ABL inhibitors imatinib in six clinical trials developed congestive heart
The second-generation BCR–ABL inhibitors nilotinib failure possibly related to imatinib exposure38, patients
and dasatinib have proved highly efficacious in patients receiving therapy with the highly potent ABL kinase
with CML following failure of imatinib therapy 23–30 inhibitors nilotinib and dasatinib must be monitored
(TABLE 1). Both agents have demonstrated remarkable for this complication.
activity against most imatinib-resistant ABL mutants,
except for the T315I mutation24,25,31,32. Therapeutic targeting of SFKs in CML
Nilotinib is a phenylaminopyrimidine derivative The SFKs comprise nine cytoplasmic structurally
developed by the reconciliation of the crystal structures homologous non-receptor intracellular tyrosine kinases
of imatinib and ABL kinase in a complex32. Replacing (SRC, FYN, YES, BLK, YRK, FGR, HCK, LCK and
the N‑methylpiperazine ring in the imatinib molecule, LYN)39. Some SFKs are ubiquitously expressed, whereas

836 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

Table 1 | Haematological and cytogenetic responses to nilotinib and dasatinib* LYN and FGR — the three main SFKs expressed in early
haematopoetic progenitor and myeloid cells — can
ABL- CML No. Response rate (%) Refs efficiently induce a CML-like myeloproliferative disor-
kinase phase patients
CHR Cytogenetic der43. Second, mice with CML-like disease responded
inhibitor
Major Complete to imatinib but not treatment with CGP76030, a TKI
that inhibits all SFKs, suggesting that SFKs other than
Nilotinib Chronic 316 74 52 34 28
HCK, LYN and FGR do not compensate for the lack
Accelerated 64 23 36 22 29 of activity of these three. However, HCK, LYN and
Blastic and 161 28 NR NR 30 FGR are required for BCR–ABL1-induced B‑ALL.
Ph+ ALL (120+41) Overall, these results suggest that BCR–ABL kinase
Dasatinib Chronic 387 90 52 39 26 uses different signalling pathways to induce CML and
Accelerated 107 39 33 24 23 BCR–ABL1-positive B‑ALL43.
Patients with the BCR–ABL1 T315I mutation are
Blastic 116 26 38 33 27 resistant to the potent ABL and SFK inhibitor dasatinib,
(myeloid)
which suggests that pure SFK inhibition will have neg-
Blastic 46 33 57 54 27 ligible therapeutic activity in most CML cases. Further
(lymphoid)
supporting this hypothesis is the fact that patients
*These are results of Phase II studies of patients with Philadelphia chromosome (Ph)-positive with CML who relapse during dasatinib therapy usu-
leukaemia resistant or intolerant to imatinib therapy. ALL, acute lymphoblastic leukaemia;
CHR, complete haematological response (normalization of peripheral blood counts and ally express dasatinib-resistant BCR–ABL1 mutations.
disappearance of all signs and symptoms related to leukaemia for at least 4 weeks); complete However, overexpression and/or activation of the SFKs
cytogenetic response, 0% Ph-positive cells; CML, chronic myeloid leukaemia; major cytogenetic
response, 0–35% Ph-positive cells; NR, not reported.
HCK and LYN has been implicated in some cases of
CML progression to blast phase and imatinib resist-
ance22,49–51. Although these results point towards a causa-
others display tissue-specific expression patterns 39. tive role of SFKs in BCR–ABL-independent resistance,
Importantly, the SFKs LCK and FYN are linked with the it remains unknown whether the concomitant inhibi-
co-receptors CD4 and CD8 and have a crucial role in tion of BCR–ABL and SFKs with dasatinib will lead to
mediating T-cell-receptor signal transduction in clonal improved responses in patients overexpressing SFKs.
lymphocytes40. Moreover, it has been shown that HCK phosphorylates
Experiments with SRC-dominant-negative mutants the activation loop of ABL kinase in vitro and blocks the
suggest that SFKs are involved in the proliferation binding of imatinib9.
of BCR–ABL1-expressing cell lines39,41,42. BCR–ABL The kinase domain of ABL can readily adopt a
kinase activates the SFKs LYN, HCK and FGR43. In conformation that closely resembles that of the inac-
fact, multiple domains of BCR–ABL have been shown tive SFKs52 (FIG. 2). Not surprisingly, ATP-competitive
to interact with HCK and LYN, leading to their activa- compounds originally developed as SFK inhibitors
tion. The formation of the HCK–BCR–ABL complex frequently exert potent inhibition of ABL kinase owing
alone appears to be sufficient for activation of SFKs, to the striking resemblance between the catalytically
with neither process being dependent on the activity of active state of both protein kinases18,53. This similarity,
ABL kinase44,45. This is supported by the fact that SFKs and the important role of SFKs in BCR–ABL signalling
remain active following imatinib inhibition of BCR–ABL and imatinib-resistance, has led to the development of
kinase activity in leukaemic cells. SFK activation may an array of small-molecule TKIs with overlapping activ-
promote phosphorylation of the Tyr177 binding site for ity against both ABL and SFKs and enhanced activity
growth-factor receptor-bound protein 2 (GRB2) in the against imatinib-resistant BCR–ABL kinase mutant
BCR moiety of BCR–ABL42,44. GRB2 recruits the linker isoforms (TABLE 2).
protein GRB2-associated binding protein 2 (GAB2), as
well as SOS, a guanine-nucleotide exchanger of RAS. Dual ABL and SFK inhibitors
The complex BCR–ABL–GRB2–SOS promotes RAS– Bosutinib. Bosutinib (also known as SKI 606) is an
mitogen-activated protein kinase (MAPK) activation, orally available 4‑anilino‑3-quinolinecarbonitrile deriv-
which results in BCL2 gene transcription. Importantly, ative54 that has potent dual SFK and ABL tyrosine kinase
BCR–ABL-induced GAB2 phosphorylation facilitates inhibitory activity55. Bosutinib has demonstrated potent
SHP2 (also known as PTPN11) recruitment and activa- antiproliferative activity against three BCR–ABL1-posi-
tion of the phosphatidylinositol-3 kinase (PI3K)/AKT tive cell lines, including LAMA84R, in which resistance
pathway. On activation, HCK activates signal transducer is caused by BCR–ABL1 gene amplification, and K562R
and activator of transcription 5 (STAT5), which in turn and KCL22R, in which the underlying mechanism of
modulates gene transcription by binding to cognate resistance has not yet been defined55. Furthermore,
DNA sequences43,46 (FIG. 1). HCK-mediated STAT5 acti- bosutinib proved active in vitro against cells transfected
vation leads to upregulation of the expression of cyclin with wild-type BCR–ABL1 and several clinically impor-
D1, which results in the cell-cycle progression from G1 tant mutant isoforms, except for T315I55 (TABLE 2). The
to S phase47,48. in vivo activity of bosutinib was demonstrated in human
A role for SFKs in the pathogenesis of CML has been KU812 xenografts in nude mice and in syngeneic
challenged by recent findings. First, transduction of BCR–ABL1 wild-type and mutant Ba/F3 xenografts55.
BCR–ABL1 into bone marrow from mice lacking HCK, In contrast to imatinib and dasatinib, bosutinib exerts

nature reviews | drug discovery volume 6 | october 2007 | 837


© 2007 Nature Publishing Group
REVIEWS

a activity of bosutinib has been explored in a Phase I/II


SRC kinase family Core P SRC, YES, FYN, study involving 18 patients with CML in chronic phase
SH3 SH2 Kinase domain LYN, LCK, BLK, who were resistant or intolerant to imatinib57. Bosutinib
Phosphorylated tail HCK, FGR, YRK was given as a single dose at 400 mg (n = 3), 500 mg
Myristate/palmitate
(n = 3) or 600 mg (n = 12) daily. Side effects were mini-
mal with the most frequent being mild to moderate
ABL kinase family
diarrhoea (87%), nausea (33%) and vomiting (20%). In
1b contrast to dasatinib58, no pleural effusion or pulmo-
Myristate SH3 SH2 Kinase domain ABL, ARG nary oedema was observed with bosutinib and these
1a Last exon region events have been rarely reported with either imatinib
or nilotinib59, perhaps as a result of the minor activity
BCR SH3 SH2 Kinase domain BCR–ABL of these agents against PDGFR55. The extent of PDGFR
inhibition may play a crucial role in the development of
pleural effusion, as this occurrence has been reported
b ABL kinase SH2–kinase more frequently in patients receiving higher dasatinib
linker Kinase
domain doses on a twice-daily schedule60. The dose-limiting
SH3 toxicity of bosutinib occurred at 600 mg daily in the
domain
form of severe rash. Of the seven patients who received
bosutinib for more than 12 weeks, three had CCyRs and
PD166326
H2N P-loop one had minimal cytogenetic response. Responders har-
Y412
boured various BCR–ABL1 mutations, but no patient
SH3–SH2 with the T315I mutation responded. A dose of 500 mg
connector Activation daily was selected as the dose for the Phase II portion
loop of the study, which is currently recruiting patients in all
phases of CML and BCR–ABL1-positive B‑ALL57. On
the basis of these results, bosutinib is currently being
tested in patients with CML who have become resistant
SH2 to imatinib or the second-generation TKIs nilotinib and
domain Myristoyl group
α-helix dasatinib.
COOH
Figure 2 | Structural organization of BCR–ABL and SFKs. SRC family kinases (SFKs) INNO‑406. INNO‑406 (also known as NS-187) (TABLE 2)
regulate signal transduction pathways involved in cell growth, differentiation and is a 3‑substituted benzamide derivative obtained through
survival. a | The common central core of SFKs and BCR–ABLNature Reviewsof| Drug
is composed Discovery
a tyrosine the introduction of various hydrophobic substituents at
kinase or SRC-homology 1 (SH1) domain, which is preceded by an SH2 domain and an
the phenyl ring adjacent to the piperazinylmethyl group
SH3 domain. These three domains are conserved in terms of their sequence homology
(42% between ABL and SRC) and arrangement129. By contrast, regions upstream of the
of imatinib61. INNO‑406 is a dual-specificity ABL and
SH3 domain and downstream of the kinase domain are variable. The NH2-terminus in the LYN kinase inhibitor that is 25–55-times more potent
ABL family of kinases is the ‘Cap’ region, which is present in different splice variants. The than imatinib62 against ABL. In addition, INNO‑406
homology region in SFK is the N‑terminal membrane-localization domain (also referred inhibited the in vitro growth of cells producing a wide
to as the SH4 domain), which anchors the protein to membranes through myristoyl and/ range of mutant BCR–ABL oncoproteins63, and pro-
or palmitoyl groups. b | A ribbon representation of the ABL kinase in complex with the longed the survival of Balb/c nu/nu mice engrafted with
ABL and SFK inhibitor PD166326 (Ref. 53). The catalytically active state of ABL kinase BaF3/E255K, Y253F, G250E, Q252H, M351T or H396P
closely resembles the active form of SFKs and is often potently inhibited by ATP- mutant cells62, but not with T315I62,63. INNO‑406 inhib-
competitive molecules originally developed as SFK inhibitors. Dual ABL and SFK ited 4 out of the 79 tyrosine kinases tested, including
inhibitors have proved active against chronic myeloid leukaemia cells exhibiting various
ABL, ABL-related gene (ARG; also known as ABL2) and
forms of imatinib resistance, including BCR–ABL1 overexpression, SFK activation and
several BCR–ABL kinase domain mutations.
FYN, but not PDGFRα/β, SRC, BLK or YES at 0.1 µM,
a concentration that adequately inhibits ABL kinase62.
Furthermore, INNO‑406 potently inhibited LYN kinase
(IC50 of 19 nM), which has been implicated in BCR–ABL-
no significant inhibition of KIT or PDGFR, which may independent resistance22, and so INNO-406 might have
result in a safer toxicity profile in vivo55. The superior further potential in imatinib-resistant CML62.
spectrum of activity of bosutinib with respect to imat- Like imatinib, INNO‑406 is a substrate of
inib may be attributed to its ability to bind both inac- P‑glycoprotein61. As a result, the concentration of both
tive and intermediate conformations of BCR–ABL 55. agents in the CNS is approximately 10% of that detected
Importantly, bosutinib is significantly more potent than in plasma64–67. However, unlike imatinib, in a mouse
imatinib in inhibiting BCR–ABL tyrosine kinase in both model of Ph-positive CNS leukaemia, this concentra-
primitive and committed CD34 +CD38– progenitors tion of INNO‑406 is sufficient to inhibit the growth of
from untreated CML patients. Unlike imatinib, bosuti- BCR–ABL1-positive leukaemic cells producing wild-
nib therapy did not result in a compensatory increase in type as well as multiple BCR–ABL1 mutant isoforms,
the important BCR–ABL downstream effector p42/44 except for T315I. Cyclosporine A, a P‑glycoprotein
MAPK, which could potentially be beneficial in target- inhibitor, administered before INNO‑406 signifi-
ing malignant stem cells56. The preliminary clinical cantly enhanced the influx of this TKI into the CNS,

838 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

Table 2 | Imatinib compared with a selection of promising dual ABL/SFK inhibitors in clinical and preclinical development*
Compound Chemical structure Chemical Potency against Potency Comments Refs
class BCR–ABL against
SFKs
Imatinib H H N 2-phenyl- • Inhibits p210 IC50 > • Standard therapy 5
(Gleevec; N N N N
amino- BCR–ABL1-positive 100 mM for patients newly
Novartis) pyrimidine 32D and MO7e cells diagnosed with
N O with IC50 ~300 nM CML based on
Phase III data

Bosutinib Cl Cl 4-anilino-3- • Inhibits WT, Y253F IC50 < • DLT in Phase I 54–57
quinoline- and D276G BCR– 10 nM studies occurred
HN O
carbonitrile ABL1 mutants with at 600 mg daily
derivative IC50 values of 13–40 • Undergoing
O CN
nM and E255K with Phase II studies
IC50 of 394 nM in CML after
N O N • Not active against imatinib failure
N T315I

INNO-406 CF3 Benzamide • Potent inhibitor IC50 of 19 • Reaches 61–64


derivative of WT BCR–ABL nM against concentrations
H H N N (IC50 of 5.8 nM) LYN kinase in CSF enough to
N N N and a wide array but no inhibit multiple
of mutants, except activity BCR–ABL1
N O T315I against mutants
• Active against the BLK, SRC • Undergoing
WT BCR–ABL1- or YES clinical evaluation
positive K562 in Phase I studies
N N
(IC50of 11 nM) and
293T (IC50 of 22 nM)
cell lines
AZD0530 O Anilino- • IC50 of 30 nM in IC50 of 1–5 • Highly selective, 70,71
O quinazoline kinase-based assays nM; active orally bioavailable
O derivative • IC50 of 220 nM in in a SRC- • Unknown activity
H human K562 cells transfected against BCR–ABL1
O N
expressing WT NIH3T3 mutants
N N
Cl BCR–ABL1 tumour • Evaluated in Phase I
xenograft studies in healthy
N
O N in athymic volunteers
mice • DLTs: diarrhoea
and vomiting
*Continued in TABLE 3. IC50, concentration of an inhibitor required for 50% inhibition of its target; CML, chronic myeloid leukaemia; CSF, cerebrospinal fluid;
DLT, dose-limiting toxicity; SFKs, SRC family kinases; WT, wild-type.

resulting in improved activity against Ph-positive have evidence of clinical response. Dose escalation is
leukaemic cells in vivo64. Approximately 6% of adult ongoing to identify a recommended Phase II dose.
patients in lymphoid blast phase CML have evidence
of CNS involvement. However, this incidence is close AZD0530. AZD0530 (TABLE 2) is a TKI obtained from a
to 12% in patients with BCR–ABL1-positive leukaemia novel series of anilinoquinazolines substituted at the C5
who relapse during imatinib monotherapy68. In view of position of the quinazoline ring, which imparts excellent
the scarce data available regarding the actual concentra- selectivity towards SFKs while preserving activity against
tions of nilotinib or dasatinib in the CNS at the dose ABL70,71. AZD0530 inhibited the cell proliferation of the
schedules currently used in clinical practice, INNO‑406 SRC-transfected mouse NIH3T3 cells with an IC50 value
represents a promising alternative for the treatment of 76 nM70. This drug was well-tolerated in Phase I trials
of CNS BCR–ABL1-positive leukaemia. In a Phase I in healthy individuals at a dose of 500 mg72. The activity
study, 11 patients with CML (8 in chronic phase, 2 in of AZD0530 against BCR–ABL1-resistant mutants has
accelerated phase and 1 in blast phase), and 3 patients not yet been reported.
with BCR–ABL1-positive ALL, who had failed prior
imatinib therapy, received INNO‑406 after imatinib 2,6,9-trisubstituted purine derivatives. AP23464 and
failure at doses ranging between 30 mg and 240 mg its analogue AP23848 (TABLE 3) are ATP-based 2,6,9-
daily69. INNO‑406 was well-tolerated. Three out of six trisubstituted purine derivatives conceptually designed
patients — two in chronic phase and one in accelerated as inhibitors of bone resorption and metastatic spread,
phase — who received INNO‑406 for more than 4 weeks respectively73. Compound optimization involved an

nature reviews | drug discovery volume 6 | october 2007 | 839


© 2007 Nature Publishing Group
REVIEWS

iterative design approach using molecular docking to which may allow PD166326 to retain activity against cer-
an SRC model, followed by compound synthesis and tain imatinib-resistant mutations18. Indeed, treatment of
SRC-kinase inhibition assays74. Both agents are potent syngeneic mice with PD166326 was more effective than
ATP-competitive dual inhibitors of ABL and SFKs75. imatinib in controlling Ph-positive leukaemia express-
Although both compounds bind to the active form of ing the imatinib-resistant BCR–ABL1 mutants H396P
SRC kinase, the affinity of AP23464 for this target is and M351T80. Recently, PD166326 was shown to inhibit
substantially higher than that of AP23848. This differ- multiple imatinib-resistant BCR–ABL1 mutants76, but not
ence has been attributed to the presence of the bulky T315I76,80 (TABLE 3). Furthermore, the yield of PD166326-
hydroxyphenethyl group located at the N9 position of resistant BCR–ABL1-positive colonies in a cell-based
AP23464 (Ref. 73). AP23464 and AP23848 inhibit the screen was consistently lower than that for imatinib76,81,
proliferation of BaF3 cells transformed with BCR–ABL1 with the most frequently recovered mutant clones being
with IC50 values of 13.4 nM and 5.4 nM, respectively76. G250E, T315I, F317L, E355G, E255K/V and Y253H76.
Similarly, AP23464 and AP23848 inhibit SRC kinase The remarkable in vivo activity of PD166326 provides a
with respective IC50 values of 67 nM and 0.45 nM73. The solid rationale to develop this TKI in human CML studies.
co-crystal structure of AP23464 and SRC reveals that the However, its limited water solubility and the development
DFG (Asp-Phe-Gly) motif at the start of the activation of tubulointerstitial nephritis in several PD166326-treated
loop is distinctly open and catalytically favourable73. In mice at autopsy at the 50 mg per kg twice-daily dose may
one study, the growth of Ba/F3 mouse pro-B-cell lines represent potential hurdles that may hamper the clinical
producing either wild-type BCR–ABL1 or the commonly development of this promising drug80.
observed clinical mutants Q252H, Y253F, E255K, M351T Other pyrido[2,3‑d]-pyrimidine dual ABL and SFK
and H396P was inhibited by AP23464 with IC50 values inhibitors include PD173955 and PD180970 (TABLE 3).
ranging from 8 nM to 94 nM75, resulting in remarkable PD173955 is a potent BCR–ABL kinase inhibitor with
apoptosis of Ba/F3 cells expressing wild-type or mutated IC50 values of 1–2 nM82. A model of PD173955 interact-
BCR–ABL1, but not parental Ba/F3 cells75. AP23464 ing with both the open and closed conformations of ABL
demonstrated potent activity against multiple imatinib- kinase suggests that PD173955 can nestle into the ABL
resistant BCR–ABL1 mutants76 (TABLE 3). However, pro- binding pocket, avoiding major steric clashes with the
liferation of parental Ba/F3 cells (IC50 >8 µM) and Ba/F3 protein, although minor adjustments of the P‑loop are
cells expressing BCR–ABL1 T315I (IC50 >9 µM) was not necessary18. PD180970 potently inhibited the BCR–ABL
significantly inhibited by AP23464 (Ref. 75). In combina- kinase (IC50 of 5 nM)83, inducing apoptosis of CD34-posi-
tion with imatinib, AP23464 suppressed drug resistance, tive leukaemic cells from patients with imatinib-resistant
except for the BCR–ABL1 T315I mutation76. CML in blast phase83,84, and of cells harbouring clinically
Based on its preliminary favourable pharmacokinet- relevant mutations84 without having any apparent effects
ics and similar effect on haematopoietic colony forma- on cell proliferation of the BCR–ABL1-negative HL60
tion compared with AP23464, AP23848 was selected for cell line83. However, the relative insolubility of PD180970
in vivo testing77. Treatment of female DBA/2 mice that may hamper the clinical development of this TKI in
had previously been injected subcutaneously with syn- BCR–ABL1-positive leukaemia83.
geneic P815 mouse mastocytoma cells with AP23848,
resulted in the inhibition of activation-loop mutant KIT Other dual ABL and SFK inhibitors. Two groups of
phosphorylation77. Although the serum concentration of BCR–ABL kinase inhibitors have been recently identi-
AP23848 after 1 hour of treatment was 1,117 nM, this fied based on a synthetic route involving the 1,3-dipolar
dropped to an average of 70 nM at 8 hours, indicating a cycloaddition of diazoketones with activated acetylenes,
short plasma half-life of approximately 2 hours77. Based followed by heat-induced cycloreversion to yield furans,
on the activity of imatinib on BCR–ABL1-expressing whose planar architecture permits the drug to reach deep
tumours, in which tumour regression requires continu- within the ATP pocket without disturbing the confor-
ous kinase inhibition, the identification of 2,6,9-trisubsti- mation of the ABL kinase domain85. Two of the most
tuted purine derivatives with superior pharmacokinetics promising compounds, the acetylanes AC22 and K1P,
compared with AP23848 is warranted before the activ- inhibited the growth of 32Dtetp210 BCR–ABL cells with
ity of such compounds is assessed in clinical trials in IC50 values of 1 µM and also exhibited inhibitory activity
patients with CML. against the SFKs LYN and FYN85. Further studies will
be necessary to determine whether these compounds
Pyrido[2,3‑d]-pyrimidines. PD166326 (TABLE 3) is the may be effective in the presence of ABL kinase point
most potent compound of a series of ATP-competitive mutations.
pyrido[2,3‑d]pyrimidine TKIs originally characterized A series of compounds initially characterized as
as inhibitors of fibroblast growth factor receptor (FGFR) specific SFK inhibitors have also been shown to inhibit
and PDGFR78. PD166326 potently inhibited K562 prolif- ABL kinase. The pyrazolo[3,4‑d]pyrimidine derivative
eration with an IC50 of 0.3 nM79, and has a relatively long PP1 inhibited SRC (IC50 of 170 nM) and HCK (IC50 of
half-life (8.4 hours) at steady state when administered 20 nM), whereas the related compound PP2 potently
orally to Balb/c mice, suggesting that once‑a-day dosing inhibited HCK (IC50 of 5 nM)86. PP1 and PP2 inhib-
would be feasible in humans80. Crystallographic studies ited BCR–ABL kinase phosphorylation in intact p210
predict that PD166326 interacts with 11 amino acids of the BCR–ABL1-positive FDCP1 cells with an IC50 of 1 µM87.
BCR–ABL kinase, compared with at least 21 for imatinib, Like PP1, the 5,7-diphenyl-pyrrolo[2,3‑d]pyrimidine

840 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

Table 3 | Imatinib compared with a selection of promising dual ABL/SFK inhibitors in clinical and preclinical development*
Compound Chemical structure Chemical Potency against Potency Comments Refs
class BCR–ABL against SFKs
AP23464 O 2,6,9- • IC50 of 13.4 nM IC50 of 67 nM • Additive/synergistic 73–76
P trisubstituted against WT with imatinib
purine BCR–ABL • Active against
HN
derivative 58 imatinib-resistant
BCR–ABL1 mutants,
N
N with IC50 values ranging
from 3 nM (E282D) to
N N 201 nM (A269V)
• Not active against
T315I
OH • In preclinical
development
AP23848 2,6,9- • IC50 of 5.4 nM IC50 of • Like AP23464, 73–76
O trisubstituted against WT 0.45 nM still in preclinical
P purine BCR–ABL development
derivative
HN

N
N

N N

OH

PD166326 Cl Pyrido[2,3- • Inhibits cell IC50 ~5 nM • Inhibits 57 imatinib- 18,76,


d]pyrimidine proliferation against resistant BCR–ABL1 78–81
N derivative of BCR–ABL1- SRC and LYN mutants, with IC50 values
positive cell line kinases ranging from 1 nm
HO Cl
N N N O K562 with IC50 (I502M) to 199 nM (L248R)
H of 0.3 nM • In preclinical development
CH3

PD173955 Cl Pyrido[2,3- • IC50 of 1–2 nM IC50 <5 nM • Binds both the active 18,82
d]pyrimidine and inactive
N derivative configurations of ABL
H3C Cl
kinase
S N N N O • In preclinical
H
CH3 development

PD180970 Cl Pyrido[2,3- • Inhibits tyrosine IC50 of 0.8 nM • Inhibits BCR–ABL1 83,84


d]pyrimidine phosphorylation against SRC E255K, A380T, H396P
F
N derivative of p210-BCR– kinase and Y253F, but not
Cl
ABL kinase in T315I
H3C N N N O K562 cells with • In preclinical
H
CH3 IC50 of 170 nM development
*Continued from TABLE 2. IC50, concentration of an inhibitor required for 50% inhibition of its target; SFKs, SRC family kinases; WT, wild-type.

derivative CGP76030 had moderate activity against ABL targeting BCR–ABL motifs that are remote from the
kinase88. Approximately 25–50 µM of PP1 or 5–10 µM kinase domain (FIG. 3). Such compounds may potentially
of CGP76030 was necessary to inhibit substrate phos- be unaffected by mutations of the kinase domain that
phorylation by BCR–ABL in COS7 and 32D BCR–ABL1- make BCR–ABL1-positive leukaemic cells resistant to
positive cells88. However, PP1 and CGP76030 blocked imatinib.
cell growth and survival in COS7 cells expressing
various imatinib-resistant BCR–ABL1 mutants, except GNF‑2. An alternative approach to the discovery of new
for T315I88. However, further investigation is required BCR–ABL inhibitors that differ to the traditional ATP-
to characterize the specificity and safety of these com- competitive molecules is the use of high-throughput
pounds before they can be introduced in the clinic. differential cytotoxicity assays. Using such an approach,
Adrian et al. compared the antiproliferative activity of
Non-ATP-competitive BCR–ABL kinase inhibitors approximately 50,000 compounds, representing 30 differ-
All TKIs currently used for the treatment of CML com- ent heterocyclic scaffolds, against non-transformed and
pete with ATP for the binding site of the BCR–ABL BCR–ABL1–transformed cells89. As a result, some novel
oncoprotein. An alternative strategy to ABL kinase 4,6-disubstituted pyrimidine compounds were found
inhibition involves the development of small molecules to have similar inhibitory activity to imatinib. Among

nature reviews | drug discovery volume 6 | october 2007 | 841


© 2007 Nature Publishing Group
REVIEWS

O values of 752 nM and 3,269 nM, respectively. Notably,


NH2 HN CO2H GNF‑2 did not affect the viability of Ba/F3 cells express-
O ing BCR–ABL1 mutated at sites that directly contact
CH3 O O O imatinib (Thr315 and Phe317), or at positions Gly250
O S and Gln252 within the P-loop at concentrations up to
N
ON012380 5–10 µM89.
O O
N
H
N GNF‑2 is the leading compound of a new class of
GNF-2 putative allosteric ABL kinase inhibitors. Although the
Allosteric mechanism of action of GNF‑2 is not yet fully character-
site Substrate ized, its exceptional selectivity for BCR–ABL suggests that
site this agent may be devoid of off-target effects and, there-
BCR ABL
ATP fore, of significant toxicity. Hence, GNF‑2 may represent
pocket the first truly targeted antileukaemia drug. Results from
Imatinib (Gleevec)
N clinical trials of GNF‑2 in CML are eagerly awaited.
H H
N N N N
ON012380. ON012380 is a small-molecule TKI unre-
N O
lated to ATP or other purine and pyrimidine nucleosides,
with approximately tenfold higher activity than imatinib
against BCR–ABL kinase90. In kinase inhibition assays,
N
ON012380 did not significantly affect the maximal velocity
Figure 3 | Different modes of BCR–ABL binding. Imatinib behaves as an ATP-competitive of BCR–ABL despite the fact that the Km values were
130
inhibitor of the ABL tyrosine kinase with a Ki value of 85 nMNature Reviewsbinds
. Imatinib | DrugtoDiscovery
the ATP significantly increased, suggesting that this compound
site and an adjacent small hydrophobic pocket to effectively lock the kinase in an acts as a competitive inhibitor. Thus, it is believed that
inactive conformation that prevents the transfer of phosphate from ATP to target ON012380 blocks the substrate binding site of BCR–ABL
substrates. By this mechanism imatinib induces cytogenetic responses in the majority of
rather than the ATP binding site. Unlike imatinib, it may
patients with chronic myeloid leukaemia in the chronic phase with a remarkably
benign toxicity profile. The development of mutations within the ABL kinase domain
therefore be unaffected by mutations in the tyrosine
has prompted the development of other BCR–ABL inhibitors that have unique modes kinase domain90 (FIG. 3, TABLE 4). ON012380 competes
of action. One such agent, ON012380, blocks the substrate binding site of ABL rather with natural substrates of BCR–ABL such as CRKL, but
than the ATP pocket, inhibiting this kinase with an IC50 value of 10 nM. Alternatively, not with ATP90. In doing so, ON012380 induced cell
other regulatory sites on BCR–ABL are potential targets for small-molecule inhibitors. death of BCR–ABL1-positive CML cells with a potency
ABL kinase has a myristoyl binding pocket at the C lobe. Myristoylation stabilizes ABL greater than tenfold that of imatinib and caused regres-
in its inactive conformation. Although BCR–ABL is not myristoylated, it is expected sion of leukaemias induced by intravenous injection of
that compounds that bind to the myristoyl binding pocket can also lock the kinase in 32DcI3 cells expressing BCR–ABL T315I kinase90. After
its inactive configuration. GNF‑2 ((3 [6 [[4-(Trifluoromethoxy)phenyl]amino] 7 and 14 days, mice treated with ON012380 showed
4-pyrimidinyl)benzamide) binds to this myristoyl pocket, resulting in potent ABL kinase
significantly lower counts of leukaemic cells expressing
inhibition with exceptional enzymatic selectivity.
the T315I mutant than imatinib-treated or saline-treated
mice. Doses of 300 mg per kg of ON012380 produced
no signs of toxicity in mice. Interestingly, this agent also
them, GNF‑2 — (3‑[6‑[[4-(trifluoromethoxy)phenyl]a inhibits the activity of the SFK LYN in the nanomolar
mino]‑4-pyrimidinyl]benzamide) — was identified as a range (IC50 of 85 nM), and so is capable of overcoming
highly selective inhibitor of BCR–ABL1-dependent cell resistance conferred by LYN overexpression90. Although
proliferation that retains potency against most clinically ON012380 has shown remarkable in vitro activity, the
relevant imatinib-resistant BCR–ABL mutants. GNF‑2 is safety and activity of ON012380 has yet to be explored
the first compound shown to inhibit BCR–ABL kinase in clinical trials.
by binding to the autoregulatory myristate binding cleft
of BCR–ABL located at the N‑terminus, spatially distant ON01910. ON01910 is a small-molecule substrate-com-
from the active site of ABL kinase, which results in stabi- petitive inhibitor of human polo-like kinase 1 (PLK1),
lization of the protein in an inactive state89 (FIG. 3). This a member of the Polo family of protein kinases. PLK1
mode of action probably contributes to the synergistic plays an important role in numerous aspects of cell-cycle
effect between GNF‑2 and imatinib. GNF‑2 does not progression, including centrosome maturation, mitotic
compete with any ABL substrate, and inhibits the pro- spindle assembly and activation of the anaphase-pro-
liferation of BCR–ABL1-expressing cells in a selective, moting complex91. Interestingly, ON01910 also inhibits
non-ATP-competitive manner with an IC50 of 138 nM, several tyrosine kinases, including wild-type BCR–ABL
but proved inactive against the catalytic domain of ABL (IC50 of 32 nM) and the SFKs SRC (IC50 of 155 nM) and
kinase. The selectivity of GNF‑2 was further supported FYN (IC50 of 182 nM) 92. Phase I trials of ON01910 in
by its lack of activity against a panel of 63 kinases, includ- advanced metastatic cancers are in progress.
ing FLT3, KIT, PDGFR, LCK and SRC. GNF‑2 inhibited
the growth of Ba/F3 cells expressing BCR–ABL1 E255V BCR–ABL T315I kinase inhibitors
or Y253H with IC50 values of 268 nM and 194 nM, The BCR–ABL1 T315I mutation, which can be detected
respectively. However, Ba/F3 cells carrying BCR–ABL1 in 10–20% of patients with CML after failure of imatinib
M351T or H396P were less sensitive to GNF‑2, with IC50 therapy11,14, confers resistance to all currently approved

842 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

Table 4 | Selected compounds with activity against BCR–ABL T315I kinase


Compound Chemical structure Chemical Potency Potency Comments Refs
class against BCR– against BCR–
ABL WT ABL T315I
ON012380 Vinyl IC50 <10 nM IC50 <10 nM Potent inhibitor 86
HN CO2H
sulphone of LYN kinase
(IC50 of 85 nM);
O
O synergistic with
O O
S
imatinib

  O O

MK-0457 4,6 diamino IC50 of 10 nM IC50 of 30 nM Potent Aurora, FLT3 and 91–93
pyrimidine JAK2 kinase inhibitor;
N currently under
HN N evaluation in Phase I
H H
N studies in relapsed
N
AML, JAK2V617F-
N N S
O positive MPD and BCR-
ABL1 T315I-positive
N leukaemia
H3C

BIRB-796 Diaryl urea IC50 of 5.3 µM IC50 of 5.3 µM Undergoing clinical 104
O evaluation in patients
O N with inflammatory
N O diseases; in CML, IC50
N N N
H H not reached even at
20 µM for Ba/F3 cells
expressing BCR-ABL1
T315I

AP23846 O 2,6,9- IC50 of 500 nM IC50 of 297 nM Potent inhibitor of FLT3 72


P trisubstituted (IC50 of 10 nM) and SFKs
purine (IC50 of 0.1–13 nM);
derivatives however, it inhibits
HN
both parental and
N
N BCR-ABL1-positive
Ba/F3 cells
N N

AML, acute myeloid leukaemia; CML, chronic myeloid leukaemia; MPD, myeloproliferative disorder; SFKs, SRC family kinases.

agents for the treatment of CML, including nilotinib and the ABL T315I mutant isoform, with IC50 values of
dasatinib24,25,31–33. Several avenues have been pursued in 10 nM and 30 nM, respectively95. Furthermore, MK‑0457
an attempt to overcome T315I-mediated TKI resist- inhibited the viability of Ba/F3 cells transformed by
ance, including the modelling of novel small-molecule wild-type, Y253F or T315I mutants of BCR–ABL96,97
inhibitors amenable to the accommodation of structural (IC50 ~300 nM). A recent comparison of the AURKA–
constraints imposed by this mutant, and the develop- MK‑0457 co-crystal structure at 2.9 Å resolution and that
ment of agents that, like the above referenced ON12380, of imatinib bound to ABL, revealed that these small mol-
target binding sites outside the ATP binding domain of ecules exploit non-overlapping sets of interactions with
BCR–ABL. Alternatively, several TKIs designed to target their respective kinases98. MK‑0457 exploits a lipophilic
other kinases have shown off-target effects that include pocket that is only available in a closed conformation of
the inhibition of BCR–ABL T315I kinase (TABLE 4). AURKA. As this pocket appears to be available in both
wild-type ABL and ABL T315I, MK‑0457 is therefore
Aurora kinase inhibitors. The human Aurora kinases able to bind to both enzymes and inhibit their activity,
(AURKs) — AURKA (also known as STK6), AURKB with Kis of 30 nM and 42 nM, respectively98. Another
and AURKC — are essential for proliferation and correct key difference when compared with imatinib, which
progression of cells through the mitotic phase of the cell penetrates deeply into the ABL kinase domain, is that
cycle93 (BOX 2). AURKA and AURKB are overexpressed MK‑0457 anchors itself firmly at the hinge region and
or gene amplified in several human malignancies. engages Asp381 at a much more superficial level within
MK‑0457 (formerly VX‑680) is an Aurora kinase inhibi- the kinase domain99. This allows MK‑0457 to overcome
tor (AKI) that has potent activity against tumour growth the potential steric constraints imposed by the mutant
in xenograft leukaemia models94 (TABLE 4). In enzyme- ABL T315I kinase. MK‑0457 has shown activity against
activity assays, MK‑0457 inhibited wild-type ABL and BCR–ABL1 T315I-positive leukaemia in three patients

nature reviews | drug discovery volume 6 | october 2007 | 843


© 2007 Nature Publishing Group
REVIEWS

when given as a 5‑day continuous intravenous infu- Other T315I inhibitors. The structural basis for the resist-
sion at doses from 12 mg per m2 per hour at 2–3‑week ance of BCR–ABL T315I to the 2,6,9-trisubstituted purine
intervals100. In a recent Phase I study in 11 patients with derivative AP23464 lies in the inability of the phenol ring
BCR–ABL1 T315I-positive refractory CML, therapy attached to N9 of AP23464 to access the deep hydropho-
with MK‑0457 resulted in one major haematological bic pocket of the ABL kinase active site76. To overcome
response, four minor haematological responses and this physical constraint, a series of AP23464 analogues
four cytogenetic responses (one CCyR, two partial and have been developed with modifications at the N9 posi-
one minimal)101. This activity was accompanied by the tion. Two such analogues, AP23848 and AP23980, were
inhibition of CRKL phosphorylation, a widely accepted marginally effective, with IC50 values of 6.4 µM and 5 µM,
surrogate of ABL kinase activity. MK‑0457 steady-state respectively. However, AP23846, owing to the replace-
plasma concentrations were ≥1 µM at a dose level of ≥20 ment of the phenol moiety with the ethyl group of N9,
mg per m2 per hour, a concentration higher than that showed remarkable potency against BCR–ABL T315I76
predicted to inhibit ABL T315I kinase101. Ongoing stud- and SFKs109 (TABLE 4). When the ethyl group at N9 was
ies will define the adequate dose schedule of MK‑0457 substituted with hydrogen the resultant compound,
for patients with AML and CML. Combination trials of AP23980, lacked any inhibitory potency against BCR–
MK‑0457 with dasatinib or nilotinib are planned. ABL T315I76. However, AP23846 inhibited the prolifera-
VE‑465, an AKI chemically related to MK‑0457, tion of Ba/F3 cells transformed by wild-type BCR–ABL1
inhibited CRKL phosphorylation in Ba/F3 cells express- and BCR–ABL1 T315I, but also the proliferation of the
ing wild-type BCR–ABL1 (IC50 of 2.0 µM) and BCR– parental Ba/F3 cells, thus indicating that off-target effects
ABL1 T315I (IC50 of 3.5 µM), was additive with imatinib may be, at least in part, responsible for the activity of this
against K562 cells, and prolonged survival of nude mice compound76.
injected with Ba/F3 cells expressing T315I102. AT9283, It has recently been shown that the benzotriazine
KW2449 and PHA‑739358 (Ref. 103) are other AKIs derivative TG100598 is a potent inhibitor of wild-type
with activity against the T315I mutation currently under ABL (IC50 of 0.4 nM) and BCR–ABL T315I (IC50 of
evaluation in Phase I/II trials for patients with refractory 3.4 nM) kinases, as well as SRC kinase (IC50 of 3.6 nM)110.
leukaemia, including BCR–ABL1 T315I-positive CML. TG101114 has demonstrated comparable potency to
that of TG100598 against T315I, but superior phar-
BIRB‑796. BIRB‑796 is a picomolar-level inhibitor macokinetic properties with increased in vivo efficacy
of p38 MAPK (also known as MAPK14)104,105, which against BCR–ABL T315I tumours, when given orally to
was discovered using combinatorial lead-optimization a xenograft SCID (severe combined immunodeficiency)
strategies based on a simple biaryl urea compound104 mouse model111. Interestingly, TG101114 has led to the
(TABLE 4). Initial reports suggested that BIRB‑796 binds evolution of a new class of BCR–ABL inhibitors led by
selectively to ABL T315I (Kd of 41 nM)95,106 compared TG101477, a compound that contains the dasatinib
with wild-type ABL (Kd of 1,500 nM) or any of the five thiazole core. TG101477 not only specifically inhib-
other imatinib-resistant ABL variants (Kd of 2,200 to ited BCR–ABL1 T315I-positive cells that were isolated
>10,000 nM)106. Although high concentrations of this from patients with CML with equal potency to that of
compound are necessary to inhibit autophosphorylation TG101114, but also showed increased selectivity com-
of this mutant in Ba/F3 cells (IC50 of 1–2 µM), its selectiv- pared with dasatinib, inhibiting only 6 out of the 76 tested
ity against ABL T315I suggested a targeted application kinases at 500 nM111.
of this agent for the treatment of patients bearing the Homoharringtonine, a pro-apoptotic cephalotaxine
imatinib-insensitive T315I mutation95. However, when ester derived from the evergreen tree Cephalotaxus har-
tested in assays that directly measure inhibition rather ringtonia k. koch var harringtonia, has recently been
than binding, BIRB‑796 was found to be a moderately shown to inhibit the proliferation of imatinib-resistant
effective inhibitor of Ba/F3 cells expressing BCR–ABL cells expressing the T315I mutation, both in vitro and
T315I (IC50 of 2−3 µM)95,107, with negligible activity in vivo112,113. These results provided the rationale for an
(IC50 >10 µM) in parental and BCR–ABL1-positive Ba/F3 ongoing Phase II multicentre study designed to test the
cells95,107. Notwithstanding the quantitative discrepancy efficacy of homoharringtonine in patients with CML
of the activity of BIRB‑796 in kinase-based and cell-based harbouring the T315I mutation after imatinib failure.
assays, this TKI is proof-of-concept that it is possible to In summary, these data indicate that the structural
inhibit BCR–ABL T315I with ATP-competitive com- constraints posed by BCR–ABL T315I kinase can be
pounds, and supports medicinal chemistry efforts aimed overcome by direct ATP-competitive inhibitors and by
at developing more effective compounds against T315I. agents with mechanisms of action other than ABL kinase
inhibition. However, the lack of specificity of some of
XL228. XL228 is a multikinase inibitor with potent these drugs may result in untoward toxic effects that may
activity against ABL, SFKs and insulin-like growth factor hinder their use in the clinic.
type 1 receptor (IGF1R). In preclinical studies, XL228
has shown potent activity against the T315I mutation108. Lessons learned from BCR–ABL inhibition
A Phase I dose-finding study of intravenous XL228 The BCR–ABL oncoprotein has proved to be an ideal
is currently underway to evaluate the safety, pharma- target for validation of the clinical utility of TKIs in can-
cokinetics and pharmacodynamics of this compound in cer10. Two major factors have been paramount in the
patients with CML or BCR–ABL1-positive B‑ALL. success of TKIs in CML: the identification of a critical

844 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

target, the BCR–ABL protein, which is sufficient to cause single molecular defect in a protein kinase, such as
CML5, and the development of an appropriate initial lead ALK-positive anaplastic large-cell lymphoma115, mul-
compound, imatinib114. tiple endocrine neoplasia with mutations in RET116
Given our current ability to design specific TKIs, the or gastrointestinal stromal tumours (GIST) carrying
most important issue for the application of molecularly gain-of-function mutations of KIT117. Although specific
targeted therapies to other cancers is the identifica- TKIs will probably provide their maximal therapeutic
tion of targets for drug development. In addition to benefit when their target kinase is the main causative
CML, other malignancies can also be ascribed to a abnormality, such agents may prove efficacious, either
alone or in combination, in genetically complex cancers
that have no obvious unique critical molecular drivers.
Box 2 | Inhibition of human Aurora kinases A prime example of the latter is the activity of the TKIs
The human Aurora kinases (AURKA, AURKB and AURKC) are key mitotic regulators that gefitinib (Iressa; AstraZeneca) and erlotinib (Tarceva;
are required for genomic stability93,128. Auroras are frequently overexpressed in cancer, OSI Pharmaceuticals) in non-small cell lung cancer
which is thought to be important for tumour formation and/or progression. (NSCLC) carrying mutant variants of EGFR118,119.
a | The domain structure, number of amino acids, and percentage of sequence homology Several tools used in the development of BCR–ABL
of human Auroras are depicted. Aurora kinases contain an NH2-terminal regulatory inhibitors for CML will aid researchers in improving the
domain (pale orange) and a catalytic kinase domain (grey). Phosphorylation at threonine process of designing and optimizing efficacious TKIs for
residues T287/T288 (AURKA), T232 (AURKB) or T195 (AURKC) within the activating other human cancers, including liquid chromatography–
T‑loops is required for kinase activity. Two sequences, the destruction box (D-Box) and mass spectrometry (LC–MS) for screening of libraries of
the D‑Box-activating domain (DAD/A-Box), promote degradation at the end of mitosis.
inhibitors that bind to different activation states of target
These features have been confirmed in AURKA and AURKB. However, the structure of
AURKC is inferred by sequence alignment and appears to lack the A‑Box. AURKA
protein kinases; high-throughput medicinal chemistry
localizes to the duplicated centrosomes and to the spindle poles in mitosis. AURKA methods (for example, multicomponent reactions, solid-
overexpression compromises spindle-check-point function and inhibits cytokinesis. phase parallel synthesis and diversity oriented synthe-
AURKB relocates at the onset of anaphase from the chromosomal centrosome to the sis) coupled with structural biology of protein kinases
microtubules that interdigitate at the spindle equator, regulating chromosome for the rapid detection, profiling and optimization of
condensation by phosphorylating histone H3 (Ref. 93). Aurora kinase inhibitors (AKIs) do lead compounds; exploration of signal-transduction
not inhibit cell-cycle progression. Rather, cells exposed to AKIs undergo mitosis with pathways through LC‑MS and other global phospho-
normal kinetics and subsequently either re-replicate their genomes, resulting in proteome strategies to identify ‘phospho-partners’ and
polyploidy, or undergo apoptosis in a pseudo G1 state. b | This depicts the structure of ‘phospho-signatures’ linked to the target tyrosine kinase;
ABL kinase domain bound to MK‑0457, which inhibits all Aurora kinase homologues and
and murine cancer models to predict the in vivo activity
the FLT3 kinase that is frequently mutated in acute myeloid leukaemia. Moreover,
MK‑0457 potently inhibits BCR–ABL T315I kinase, which renders chronic myeloid
of targeted agents.
leukaemia cells insensitive to imatinib, nilotinib and dasatinib. c | The Y‑shaped structure Last, the realization of the full potential of any given
of MK‑0457 avoids close encounter with leucine in Aurora kinases and isoleucine in TKI will require addressing the emergence of drug
BCR–ABL T315 at the gatekeeper position. The four hydrogen bonds within the ABL resistance. Based on the CML experience showing
kinase are depicted (distances in Å). that a mutation of the conserved gatekeeper threonine
a T287/T288 D-box residue (T315I) prevents the binding of all clinically
A-box
approved ABL TKIs, structurally related mutations
AURKA 403 have been described in other kinases such as KIT
132 383 (T670I) 120 in GIST; EGFR (T790M) 121 in NSCLC;
A-box T232 D-box 57% and FIP1L1-PDGFRα (T674I)122 in hypereosinophilic
syndrome that play central roles in the pathogenesis
AURKB 344 60% of these conditions. Spurred by the discovery of TKIs
76 327 against BCR–ABL T315I in CML, several compounds
T195 D-box 75% are being investigated as potential inhibitors of gate-
keeper mutants in NSCLC and GIST95. Undoubtedly,
AURKC 306 the headway made by CML investigators will continue
39 290 to inspire the advance of molecularly targeted therapies
for a wide spectrum of human cancers associated with
b MK-0457/ABL kinase c protein-kinase deregulation.

Future directions
2.72 2.62 The pressing need for the development of novel agents
Thr315 capable of overcoming mechanisms of resistance to
gatekeeper
DFG 3.08 clinically approved TKIs, and a better understanding
motif of the structural biology of ABL kinase, has guided the
3.04 synthesis of an array of compounds with activity against
Pro396
Asp381 BCR–ABL1-positive leukaemic cells, and in some cases
with remarkably improved kinase specificity. The
rather limited approach of targeting the ABL kinase
domain directly has been recently challenged with the
design of two particularly promising agents, GNF‑2

nature reviews | drug discovery Nature Reviews | Drug Discovery volume 6 | october 2007 | 845
© 2007 Nature Publishing Group
REVIEWS

and ON012380, directed against ABL domains that are speculate that the combination of potent dual ABL and
distant from the ATP binding site. Because of their distinct SFK inhibitors (for example, bosutinib) with TKIs that
mode of BCR–ABL inhibition, these agents hold great target BCR–ABL motifs other than the kinase domain
promise regarding the inhibition of ABL kinase domain (for example, GNF‑2), might provide a powerful strat-
mutants and, owing to their enhanced kinase selectivity, egy to simultaneously inhibit multiple non-overlapping
may be associated with benign toxicity profiles. BCR–ABL-related elements. The clinical development
Furthermore, ON012380 inhibits the highly insensitive of these combinations must be preceded by a systematic
mutant T315I, which mediates TKI resistance in a sig- effort to track their effects on molecular targets in vitro.
nificant proportion of patients with CML. Other agents, The speed with which BCR–ABL inhibitors have
such as the AKI MK‑0457, have also shown remarkable been developed has exceeded expectations and fuels
activity against T315I. This encouraging preliminary optimism regarding the achievement of the final goal of
activity warrants investigation in larger series of patients curing CML. To accomplish this goal it is essential that
and a longer follow-up will be required to define the the pace of scientific discoveries is matched with more
durability of these responses. agile and flexible policies regarding drug approval by the
To date, ABL kinase inhibition has not yielded cures pertinent regulatory bodies. The swiftness with which
in patients with CML. The general consensus is that the FDA approved imatinib in 2001 has rapidly been
combination approaches will be required to eradicate outpaced. Current regulations must be implemented to
BCR–ABL1-positive leukaemic clones. Although the enable highly promising BCR–ABL inhibitors to become
clinical relevance of the novel BCR–ABL kinase inhibi- available sooner to patients with CML, even when other
tors described here has yet to be proved, it is tempting to active compounds may already be available.

1. Shtivelman, E., Lifshitz, B., Gale, R. P. & Canaani, E. 14. Gorre, M. E. et al. Clinical resistance to STI‑571 cancer 25. Talpaz, M. et al. Dasatinib in imatinib-resistant
Fused transcript of ABL and BCR genes in chronic therapy caused by BCR–ABL gene mutation or Philadelphia chromosome-positive leukemias. N. Engl.
myelogenous leukaemia. Nature 315, 550–554 amplification. Science 293, 876–880 (2001). J. Med. 354, 2531–2541 (2006).
(1985). The first description of the occurrence of imatinib A description of the Phase I trial of dasatinib in
2. Ben-Neriah, Y., Daley, G. Q., Mes-Masson, A. M., resistance in patients owing to point mutations within patients with CML exhibiting imatinib resistance.
Witte, O. N. & Baltimore, D. The chronic myelogenous the kinase domain of ABL. The realization of this The favourable toxicity profile and significant
leukemia-specific p210 protein is the product of the mechanism of resistance spurred the development of clinical activity shown in this trial facilitated the
BCR/ABL hybrid gene. Science 233, 212–214 (1986). a second generation of ABL-kinase inhibitors, best approval of dasatinib by the FDA for the treatment
3. Ravandi, F. et al. Chronic myelogenous leukaemia with represented by nilotinib and dasatinib. of imatinib-resistant CML.
p185(BCR/ABL) expression: characteristics and clinical 15. Lowenberg, B. Minimal residual disease in chronic 26. Hochhaus, A. et al. Dasatinib induces notable
significance. Br. J. Haematol. 107, 581–586 (1999). myeloid leukemia. N. Engl. J. Med. 349, 1399–1401 hematologic and cytogenetic responses in chronic-
4. Daley, G. Q., Van Etten, R. A. & Baltimore, D. (2003). phase chronic myeloid leukemia after failure of
Induction of chronic myelogenous leukemia in mice by 16. Corbin, A. S., La Rosee, P., Stoffregen, E. P., Druker, imatinib therapy. Blood 109, 2303–2309 (2007).
the p210 Bcr/Abl gene of the Philadelphia B. J. & Deininger, M. W. Several BCR–ABL kinase 27. Cortes, J. et al. Dasatinib induces complete
chromosome. Science 247, 824–830 (1990). domain mutants associated with imatinib mesylate hematologic and cytogenetic responses in patients
5. Lugo, T. G., Pendergast, A. M., Muller, A. J. & Witte, resistance remain sensitive to imatinib. Blood 101, with imatinib-resistant or -intolerant chronic myeloid
O. N. Tyrosine kinase activity and transformation 4611–4614 (2003). leukemia in blast crisis. Blood 109, 3207–3213
potency of BCR–ABL oncogene products. Science 17. Gambacorti-Passerini, C. B. et al. Molecular (2006).
247, 1079–1082 (1990). mechanisms of resistance to imatinib in 28. le Coutre, P. et al. A Phase II study of nilotinib, a novel
6. Druker, B. J. et al. Effects of a selective inhibitor of the Philadelphia‑chromosome‑positive leukaemias. Lancet tyrosine kinase inhibitor administered to imatinib-
Abl tyrosine kinase on the growth of BCR–ABL Oncol. 4, 75–85 (2003). resistant and -intolerant patients with chronic
positive cells. Nature Med. 2, 561–566 (1996). 18. Nagar, B. et al. Crystal structures of the kinase domain myelogenous leukemia (CML) in chronic phase (CP).
7. Druker, B. J. et al. Efficacy and safety of a specific of c‑Abl in complex with the small molecule inhibitors Blood 108, Abstract 165 (2006).
inhibitor of the BCR–ABL tyrosine kinase in chronic PD173955 and imatinib (STI‑571). Cancer Res. 62, 29. Kantarjian, H. et al. A Phase II study of nilotinib a
myeloid leukemia. N. Engl. J. Med. 344, 1031–1037 4236–4243 (2002). novel tyrosine kinase inhibitor administered to
(2001). 19. Hantschel, O. et al. A myristoyl/phosphotyrosine imatinib-resistant or intolerant patients with chronic
8. Huse, M. & Kuriyan, J. The conformational plasticity of switch regulates c‑Abl. Cell 112, 845–857 (2003). myelogenous leukemia (CML) in accelerated phase
protein kinases. Cell 109, 275–282 (2002). 20. Azam, M., Latek, R. R. & Daley, G. Q. Mechanisms of (AP). Blood 108, Abstract 2169 (2006).
9. Schindler, T. et al. Structural mechanism for STI‑571 autoinhibition and STI‑571/imatinib resistance 30. Ottmann, O. et al. A Phase II study of nilotinib, a novel
inhibition of abelson tyrosine kinase. Science 289, revealed by mutagenesis of BCR–ABL. Cell 112, tyrosine kinase inhibitor administered to imatinib
1938–1942 (2000). 831–843 (2003). resistant or intolerant patients with chronic
This paper describes the crystal structure of the A description of an in vitro screen of randomly myelogenous leukemia (CML) in blast crisis (BC) or
catalytic domain of ABL in complex with a variant mutagenized BCR–ABL that identified all the major relapsed/refractory Ph+ acute lymphoblastic leukemia
of imatinib, and underscores the importance of the ABL kinase mutations previously detected in (ALL). Blood 108, Abstract 1862 (2006).
adoption of an inactive conformation by ABL kinase patients with CML resistant to imatinib as well as 31. Lombardo, L. J. et al. Discovery of
for imatinib binding. numerous others, highlighting the heterogeneity of N‑(2‑chloro‑6‑methyl‑ phenyl)‑2‑(6‑(4‑(2-hydroxyethyl)-
10. Druker, B. J. et al. Five-year follow-up of patients mutations of ABL kinase. piperazin‑1‑yl)‑2‑methylpyrimidin‑4‑
receiving imatinib for chronic myeloid leukemia. 21. Skaggs, B. J. et al. Phosphorylation of the ATP-binding ylamino)thiazole‑5‑carboxamide (BMS‑354825), a
N. Engl. J. Med. 355, 2408–2417 (2006). loop directs oncogenicity of drug-resistant BCR–ABL dual Src/Abl kinase inhibitor with potent antitumor
A critical report on the activity and safety of the mutants. Proc. Natl Acad. Sci. USA 103, 19466– activity in preclinical assays. J. Med. Chem. 47,
ABL-kinase inhibitor imatinib in patients with CML 19471 (2006). 6658–6661 (2004).
after 5 years of follow-up. 22. Donato, N. J. et al. BCR–ABL independence and LYN A description of the synthesis and characterization
11. Druker, B. J. et al. Activity of a specific inhibitor of the kinase overexpression in chronic myelogenous of the second-generation dual ABL and SFK
BCR–ABL tyrosine kinase in the blast crisis of chronic leukemia cells selected for resistance to STI571. Blood inhibitor dasatinib, which proved highly active and
myeloid leukemia and acute lymphoblastic leukemia 101, 690–698 (2003). safe in a K562 xenograft model of CML.
with the Philadelphia chromosome. N. Engl. J. Med. 23. Guilhot, F. et al. Dasatinib induces significant 32. Weisberg, E. et al. Characterization of AMN107, a
344, 1038–1042 (2001). hematologic and cytogenetic responses in patients with selective inhibitor of native and mutant BCR–ABL.
12. Hochhaus, A. & La Rosee, P. Imatinib therapy in imatinib-resistant or -intolerant chronic myeloid Cancer Cell 7, 129–141 (2005).
chronic myelogenous leukemia: strategies to avoid and leukemia in accelerated phase. Blood 109, 4143–4150 The first characterization of the second-generation
overcome resistance. Leukemia 18, 1321–1331 (2007). ABL kinase inhibitor, nilotinib, endowed with
(2004). 24. Kantarjian, H. et al. Nilotinib in imatinib-resistant CML significantly higher potency against ABL kinase
13. Shah, N. P. et al. Multiple BCR–ABL kinase domain and Philadelphia chromosome-positive ALL. N. Engl. J. than imatinib and the ability to override imatinib
mutations confer polyclonal resistance to the tyrosine Med. 354, 2542–2551 (2006). resistance conferred by ABL kinase mutations.
kinase inhibitor imatinib (STI571) in chronic phase This paper describes the results of a clinical trial 33. Shah, N. P. et al. Overriding imatinib resistance with a
and blast crisis chronic myeloid leukemia. Cancer Cell testing nilotinib in patients with imatinib-resistant novel ABL kinase inhibitor. Science 305, 399–401
2, 117–125 (2002). CML. (2004).

846 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group
REVIEWS

This paper characterizes dasatinib, a second- 56. Konig, H. et al. The dual Src/Abl kinase inhibitor 76. Azam, M. et al. Activity of dual SRC-ABL inhibitors
generation ABL kinase inhibitor, which is 300-fold SKI‑606 effectively inhibits BCR–ABL kinase activity highlights the role of BCR/ABL kinase dynamics in
more potent than imatinib against ABL kinase and and reduces proliferation of CML primitive progenitor drug resistance. Proc. Natl Acad. Sci. USA 103,
overcomes imatinib resistance due to most point cells. Blood 108, Abstract 1370 (2006). 9244–9249 (2006).
mutations within the kinase domain of ABL. 57. Cortes, J. et al. A phase 1/2 study of SKI‑606, a dual 77. Corbin, A. S. et al. In vitro and in vivo activity of ATP-
34. Gambacorti-Passerini, C., Gasser, M., Ahmed, S., inhibitor of Src and Abl kinases, in adult patients with based kinase inhibitors AP23464 and AP23848
Assouline, S. & Scapozza, L. Abl inhibitor BMS354825 Philadelphia chromosome positive (Ph+) chronic against activation-loop mutants of Kit. Blood 106,
binding mode in Abelson kinase revealed by molecular myelogenous leukemia (CML) or acute lymphocytic 227–234 (2005).
docking studies. Leukemia 19, 1267–1269 (2005). leukemia (ALL) relapsed, refractory or intolerant of 78. Hamby, J. M. et al. Structure–activity relationships for
35. Tokarski, J. S. et al. The structure of Dasatinib imatinib. Blood 108, Abstract 168 (2006). a novel series of pyrido[2,3‑d]pyrimidine tyrosine
(BMS‑354825) bound to activated ABL kinase 58. Quintas-Cardama, A. et al. Pleural effusion in patients kinase inhibitors. J. Med. Chem. 40, 2296–2303
domain elucidates its inhibitory activity against (pts) with chronic myelogenous leukemia (CML) (1997).
imatinib-resistant ABL mutants. Cancer Res 66, treated with dasatinib after imatinib failure. Blood 79. Huron, D. R. et al. A novel pyridopyrimidine inhibitor
5790–7 (2006). 108, Abstract 2164 (2006). of abl kinase is a picomolar inhibitor of BCR–
36. Burgess, M. R., Skaggs, B. J., Shah, N. P., Lee, F. Y. & 59. Sawyers, C. L. et al. Imatinib induces hematologic and ABL‑driven K562 cells and is effective against STI571-
Sawyers, C. L. Comparative analysis of two clinically cytogenetic responses in patients with chronic resistant BCR–ABL mutants. Clin. Cancer Res. 9,
active BCR–ABL kinase inhibitors reveals the role of myelogenous leukemia in myeloid blast crisis: results 1267–1273 (2003).
conformation-specific binding in resistance. Proc. Natl of a Phase II study. Blood 99, 3530–3539 (2002). 80. Wolff, N. C. et al. PD166326, a novel tyrosine kinase
Acad. Sci. USA 102, 3395–3400 (2005). 60. Shah, N. et al. Dasatinib 50 mg or 70 mg BID inhibitor, has greater antileukemic activity than
37. Kerkela, R. et al. Cardiotoxicity of the cancer compared to 100 mg or 140 mg QD in patients with imatinib mesylate in a murine model of chronic
therapeutic agent imatinib mesylate. Nature Med. 12, CML in chronic phase (CP) who are resistant or myeloid leukemia. Blood 105, 3995–4003 (2005).
908–916 (2006). intolerant to imatinib: One-year results of CA180034. 81. von Bubnoff, N., Barwisch, S., Speicher, M. R.,
38. Hatfield, A., Owen, S. & Pilot, P. R. In reply to J. Clin. Oncol. 25 (Suppl. 20), 7004 (2007). Peschel, C. & Duyster, J. A cell-based screening
‘Cardiotoxicity of the cancer therapeutic agent 61. Kimura, S., Niwa, T., Hirabayashi, K. & Maekawa, T. strategy that predicts mutations in oncogenic tyrosine
imatinib mesylate’. Nature Med. 13, 15–16 (2007). Development of NS‑187, a potent and selective dual kinases: implications for clinical resistance in targeted
39. Abram, C. L. & Courtneidge, S. A. Src family tyrosine BCR–ABL/Lyn tyrosine kinase inhibitor. Cancer cancer treatment. Cell Cycle 4, 400–406 (2005).
kinases and growth factor signaling. Exp. Cell Res. Chemother. Pharmacol. 58 (Suppl. 7), 55–61 (2006). 82. Wisniewski, D. et al. Characterization of potent
254, 1–13 (2000). 62. Kimura, S. et al. NS‑187, a potent and selective dual inhibitors of the BCR–ABL and the c‑kit receptor
40. Straus, D. B. & Weiss, A. Genetic evidence for the BCR–ABL/Lyn tyrosine kinase inhibitor, is a novel tyrosine kinases. Cancer Res. 62, 4244–4255
involvement of the lck tyrosine kinase in signal agent for imatinib-resistant leukemia. Blood 106, (2002).
transduction through the T cell antigen receptor. Cell 3948–3954 (2005). 83. Dorsey, J. F., Jove, R., Kraker, A. J. & Wu, J. The
70, 585–593 (1992). 63. Naito, H. et al. In vivo antiproliferative effect of pyrido[2,3‑d]pyrimidine derivative PD180970 inhibits
41. Stanglmaier, M., Warmuth, M., Kleinlein, I., Reis, S. & NS‑187, a dual BCR–ABL/Lyn tyrosine kinase p210BCR–Abl tyrosine kinase and induces apoptosis
Hallek, M. The interaction of the BCR–ABL tyrosine inhibitor, on leukemic cells harbouring Abl kinase of K562 leukemic cells. Cancer Res. 60, 3127–3131
kinase with the Src kinase Hck is mediated by multiple domain mutations. Leuk. Res. 30, 1443–1446 (2000).
binding domains. Leukemia 17, 283–289 (2003). (2006). 84. La Rosee, P., Corbin, A. S., Stoffregen, E. P.,
42. Danhauser-Riedl, S., Warmuth, M., Druker, B. J., 64. Yokota, A. et al. INNO‑406, a novel BCR–ABL/Lyn Deininger, M. W. & Druker, B. J. Activity of the
Emmerich, B. & Hallek, M. Activation of Src kinases dual tyrosine kinase inhibitor, suppresses the growth BCR–ABL kinase inhibitor PD180970 against
p53/56lyn and p59hck by p210bcr/abl in myeloid of Ph+ leukemia cells in the central nervous system, clinically relevant BCR–ABL isoforms that cause
cells. Cancer Res. 56, 3589–3596 (1996). and cyclosporine A augments its in vivo activity. Blood resistance to imatinib mesylate (Gleevec, STI571).
43. Hu, Y. et al. Requirement of Src kinases Lyn, Hck and 109, 306–314 (2007). Cancer Res. 62, 7149–7153 (2002).
Fgr for BCR–ABL1‑induced B‑lymphoblastic leukemia 65. Petzer, A. L. et al. Low concentrations of STI571 in the 85. Lerma, E. I. et al. Novel compounds with
but not chronic myeloid leukemia. Nature Genet. 36, cerebrospinal fluid: a case report. Br. J. Haematol. antiproliferative activity against imatinib-resistant cell
453–461 (2004). 117, 623–625 (2002). lines. Mol. Cancer Ther. 6, 655–666 (2007).
44. Warmuth, M. et al. The Src family kinase Hck 66. Takayama, N., Sato, N., O’Brien, S. G., Ikeda, Y. & 86. Hanke, J. H. et al. Discovery of a novel, potent, and
interacts with BCR–ABL by a kinase-independent Okamoto, S. Imatinib mesylate has limited activity Src family-selective tyrosine kinase inhibitor. Study of
mechanism and phosphorylates the Grb2-binding site against the central nervous system involvement of Lck- and FynT-dependent T cell activation. J. Biol.
of Bcr. J. Biol. Chem. 272, 33260–33270 (1997). Philadelphia chromosome-positive acute lymphoblastic Chem. 271, 695–701 (1996).
45. Klejman, A. et al. The Src family kinase Hck couples leukaemia due to poor penetration into cerebrospinal 87. Tatton, L., Morley, G. M., Chopra, R. & Khwaja, A. The
BCR/ABL to STAT5 activation in myeloid leukemia fluid. Br. J. Haematol. 119, 106–108 (2002). Src-selective kinase inhibitor PP1 also inhibits Kit and
cells. EMBO J. 21, 5766–5774 (2002). 67. Neville, K. et al. Plasma and cerebrospinal fluid BCR–ABL tyrosine kinases. J. Biol. Chem. 278,
46. Bhatia, R. et al. Persistence of malignant pharmacokinetics of imatinib after administration to 4847–4853 (2003).
hematopoietic progenitors in chronic myelogenous nonhuman primates. Clin. Cancer Res. 10, 2525–2529 88. Warmuth, M. et al. Dual-specific Src and Abl kinase
leukemia patients in complete cytogenetic remission (2004). inhibitors, PP1 and CGP76030, inhibit growth and
following imatinib mesylate treatment. Blood 101, 68. Pfeifer, H. et al. Risk and prognosis of central nervous survival of cells expressing imatinib mesylate-resistant
4701–4707 (2003). system leukemia in patients with Philadelphia BCR–ABL kinases. Blood 101, 664–672 (2003).
47. Kerkhoff, E. & Rapp, U. R. Cell cycle targets of Ras/Raf chromosome-positive acute leukemias treated with 89. Adrian, F. J. et al. Allosteric inhibitors of BCR–ABL-
signalling. Oncogene 17, 1457–1462 (1998). imatinib mesylate. Clin. Cancer Res. 9, 4674–4681 dependent cell proliferation. Nature Chem. Biol. 2,
48. Tetsuya N, T. K., Kazuhide M, et al. STAT5 as a (2003). 95–102 (2006).
molecular regulator of proliferation, differentiation 69. Kantarjian, H. et al. A Phase I study of INNO‑406, a The first description of a novel class of BCR–ABL
and apoptosis in hematopoietic cells. EMBO J. 18, dual inhibitor of Abl/Lyn kinases, in adult patients with inhibitors, identified using an unbiased differential
4754–4765 (1999). imatinib-resistant or intolerant Philadelphia cytotoxicity screen of a combinatorial kinase-
49. Dai, Y., Rahmani, M., Corey, S. J., Dent, P. & Grant, S. chromosome positive (Ph+) leukemias. Proc. Am. Soc. directed heterocycle library. One such compound,
A Bcr/Abl-independent, Lyn-dependent form of Cancer Res. Abstract 2637 (2007). GNF‑2, inhibits specifically BCR–ABL kinase
imatinib mesylate (STI‑571) resistance is associated 70. Hennequin, L. F. et al. N‑(5‑chloro‑1,3‑benzodioxol‑4- activity through an allosteric non-ATP competitive
with altered expression of Bcl‑2. J. Biol. Chem. 279, yl)‑7‑[2‑(4‑methylpiperazin‑1-yl)ethoxy]‑5‑ mechanism.
34227–34239 (2004). (tetrahydro‑2H‑pyran‑4‑yloxy)quinazolin‑4‑amine, 90. Gumireddy, K. et al. A non‑ATP‑competitive inhibitor
50. Donato, N. J. et al. Imatinib mesylate resistance a novel, highly selective, orally available, dual-specific of BCR–ABL overrides imatinib resistance. Proc. Natl
through BCR–ABL independence in chronic c‑Src/Abl kinase inhibitor. J. Med. Chem. 49, Acad. Sci. USA 102, 1992–1997 (2005).
myelogenous leukemia. Cancer Res. 64, 672–677 6465–6488 (2006). This paper describes the activity of ON012380, a
(2004). 71. Hennequin, L. et al. The discovery of AZD0530: a compound that inhibits BCR–ABL in a substrate-
51. Hofmann, W. K. et al. Relation between resistance of novel, oral, highly selective and dual-specific inhibitor competitive manner. More importantly, ON012380
Philadelphia‑chromosome‑positive acute of the Src and Abl family kinases. Proc. Am. Assoc. was found to induce apoptosis of all of the known
lymphoblastic leukaemia to the tyrosine kinase Cancer Res. 46, 595 (2005). imatinib-resistant mutants at concentrations of
inhibitor STI571 and gene-expression profiles: a gene- 72. Lockton, J. Phase I ascending single and multiple dose <10 nM concentration in vitro.
expression study. Lancet 359, 481–486 (2002). studies to assess the safety, tolerability and 91. Lane, H. A. & Nigg, E. A. Antibody microinjection
52. Levinson, N. M. et al. A Src-like inactive conformation pharmacokinetics of AZD0530, a highly selective, dual reveals an essential role for human polo-like kinase 1
in the abl tyrosine kinase domain. PLoS Biol. 4, e144 specific Src/Abl inhibitor. J. Clin. Oncol. 23, (Suppl. 1), (Plk1) in the functional maturation of mitotic
(2006). 3125 (2005). centrosomes. J. Cell Biol. 135, 1701–1713 (1996).
53. Nagar, B. et al. Structural basis for the autoinhibition 73. Dalgarno, D. et al. Structural basis of Src tyrosine 92. Gumireddy, K. et al. ON01910, a non‑ATP‑competitive
of c‑Abl tyrosine kinase. Cell 112, 859–871 (2003). kinase inhibition with a new class of potent and small molecule inhibitor of Plk1, is a potent anticancer
54. Golas, J. M. et al. SKI‑606, a 4‑anilino‑3- selective trisubstituted purine-based compounds. agent. Cancer Cell 7, 275–286 (2005).
quinolinecarbonitrile dual inhibitor of Src and Abl Chem. Biol. Drug Des. 67, 46–57 (2006). 93. Carmena, M. & Earnshaw, W. C. The cellular
kinases, is a potent antiproliferative agent against 74. Wang, Y. et al. Bone-targeted 2,6,9-trisubstituted geography of aurora kinases. Nature Rev. Mol. Cell
chronic myelogenous leukemia cells in culture and purines: novel inhibitors of Src tyrosine kinase for the Biol. 4, 842–854 (2003).
causes regression of K562 xenografts in nude mice. treatment of bone diseases. Bioorg. Med. Chem. Lett. 94. Harrington, E. A. et al. VX‑680, a potent and selective
Cancer Res. 63, 375–381 (2003). 13, 3067–3070 (2003). small-molecule inhibitor of the Aurora kinases,
55. Puttini, M. et al. In vitro and in vivo activity of 75. O’Hare, T. et al. Inhibition of wild-type and mutant suppresses tumor growth in vivo. Nature Med. 10,
SKI‑606, a novel Src-Abl inhibitor, against imatinib- BCR–ABL by AP23464, a potent ATP-based 262–267 (2004).
resistant BCR–ABL+ neoplastic cells. Cancer Res. 66, oncogenic protein kinase inhibitor: implications for This paper reports the discovery of the highly
11314–11322 (2006). CML. Blood 104, 2532–2539 (2004). potent and selective small-molecule inhibitor of

nature reviews | drug discovery volume 6 | october 2007 | 847


© 2007 Nature Publishing Group
REVIEWS

Aurora kinases MK‑0457 that blocks cell-cycle 107. O’Hare, T. & Druker, B. J. BIRB‑796 is not an effective 122. Cools, J. et al. A tyrosine kinase created by fusion of
progression and induces apoptosis in various ABL(T315I) inhibitor. Nature Biotech. 23, 1209–1210 the PDGFRA and FIP1L1 genes as a therapeutic target
in vivo xenograft models, including leukaemia, (2005). of imatinib in idiopathic hypereosinophilic syndrome.
colon and pancreatic tumours. 108. Zhang, W. Inhibition of the drug-resistant T315I N. Engl. J. Med. 348, 1201–1214 (2003).
95. Carter, T. A. et al. Inhibition of drug-resistant mutants mutant of BCR–ABL. Eur. J. Cancer Suppl. 4, 54 123. Robinson, D. R., Wu, Y. M. & Lin, S. F. The protein
of ABL, KIT, and EGF receptor kinases. Proc. Natl (2006). tyrosine kinase family of the human genome.
Acad. Sci. USA 102, 11011–11016 (2005). 109. Summy, J. M. et al. AP23846, a novel and highly Oncogene 19, 5548–5557 (2000).
96. Hoover, R. & Harding, M. W. Synergistic activity of the potent Src family kinase inhibitor, reduces vascular 124. Manning, G., Whyte, D. B., Martinez, R., Hunter, T. &
Aurora kinase inhibitor MK‑0457 (VX‑680) with endothelial growth factor and interleukin‑8 expression Sudarsanam, S. The protein kinase complement of the
idarubicin, Ara‑C, and inhibitors of BCR–ABL. Blood in human solid tumor cell lines and abrogates human genome. Science 298, 1912–1934 (2002).
108, Abstract 1384 (2006). downstream angiogenic processes. Mol. Cancer Ther. 125. Liu, Y. & Gray, N. S. Rational design of inhibitors that
97. Shah, N. et al. The most common dasatinib-resistant 4, 1900–1911 (2005). bind to inactive kinase conformations. Nature Chem.
BCR–ABL kinase domain mutations in patients with 110. Zhu, H. Enzymatic and cellular inhibition of BCR/ABL Biol. 2, 358–364 (2006).
chronic myeloid leukemia are sensitive to VX‑680: T315I by a novel src/ABL inhibitor. Proc. Am. Assoc. 126. Hubbard, S. R., Wei, L., Ellis, L. & Hendrickson, W. A.
rationale for early combination kinase inhibitor Cancer Res. 47, Abstract 3771 (2006). Crystal structure of the tyrosine kinase domain of the
therapy. Blood 108, Abstract 2175 (2006). 111. Zhu, H. Addressing BCR/ABL resistance: evolving human insulin receptor. Nature 372, 746–754 (1994).
98. Cheetham, G. M., Charlton, P. A., Golec, J. M. & dasatinib to a novel inhibitor of the T315I BCR/Abl 127. Blum, G., Gazit, A. & Levitzki, A. Substrate
Pollard, J. R. Structural basis for potent inhibition of mutation. Proc. Am. Assoc. Cancer Res. Abstract competitive inhibitors of IGF‑1 receptor kinase.
the Aurora kinases and a T315I multi-drug resistant 3254 (2007). Biochemistry 39, 15705–15712 (2000).
mutant form of ABL kinase by VX‑680. Cancer Lett. 112. Chen, R., Gandhi, V. & Plunkett, W. A sequential 128. Nigg, E. A. Mitotic kinases as regulators of cell
251, 323–329 (2007). blockade strategy for the design of combination division and its checkpoints. Nature Rev. Mol. Cell
99. Young, M. A. et al. Structure of the kinase domain of therapies to overcome oncogene addiction in Biol. 2, 21–32 (2001).
an imatinib-resistant Abl mutant in complex with the chronic myelogenous leukemia. Cancer Res. 66, 129. Superti-Furga, G. & Courtneidge, S. A. Structure-
Aurora kinase inhibitor VX‑680. Cancer Res. 66, 10959–10966 (2006). function relationships in Src family and related protein
1007–1014 (2006). 113. Legros, L. et al. BCR–ABLT315I transcript tyrosine kinases. Bioessays 17, 321–330 (1995).
This article describes the high-resolution crystal disappearance in an imatinib-resistant CML patient 130. Cowan-Jacob, S. W. et al. Imatinib (STI571) resistance
structure of the catalytic domain of a mutant treated with homoharringtonine: a new therapeutic in chronic myelogenous leukemia: molecular basis of
H396P ABL-kinase isoform in complex with the challenge? Leukemia 31 May 2007 (doi:10.1038/sj. the underlying mechanisms and potential strategies for
small-molecule inhibitor MK‑0457. MK‑0457 leu.2404772). treatment. Mini Rev. Med. Chem. 4, 285–299 (2004).
avoids the innermost cavity of the ABL, which 114. Buchdunger, E. et al. Inhibition of the Abl protein-
explains the effectiveness of this compound tyrosine kinase in vitro and in vivo by a Acknowledgements
against the T315I gatekeeper mutation. 2‑phenylaminopyrimidine derivative. Cancer Res. 56, A.Q.-C. is perennially indebted to E. Schiele, J. Lado-Abeal and
100. Giles, F. J. et al. MK‑0457, a novel kinase 100–104 (1996). L. Hayward for their ceaseless inspiration, and to D. L. Gibbons
inhibitor, is active in patients with chronic myeloid 115. Morris, S. W. et al. Fusion of a kinase gene, ALK, to a for his unremitting remaking/remodelling/rethinking. W.H.:
leukemia or acute lymphocytic leukemia with the nucleolar protein gene, NPM, in non-Hodgkin’s “I understand the fury in your words, but not the words”.
T315I BCR–ABL mutation. Blood 109, 500–502 lymphoma. Science 263, 1281–1284 (1994).
(2007). 116. Hofstra, R. M. et al. A mutation in the RET proto- Competing interests statement
101. Giles, F. et al. MK‑0457, a novel Aurora kinase and oncogene associated with multiple endocrine The authors declare competing financial interests: see web
BCR–ABL inhibitor, is active against BCR–ABL T315I neoplasia type 2B and sporadic medullary thyroid version for details.
mutant chronic myelogenous leukemia. Blood 108, carcinoma. Nature 367, 375–376 (1994).
Abstract 163 (2006). 117. Hirota, S. et al. Gain‑of‑function mutations of c‑KIT in
DATABASES
102. Tauchi, T., et al. Activity of a novel Aurora kinase human gastrointestinal stromal tumors. Science 279,
Entrez Gene:
inhibitor, VE‑465, against T315I mutant form of BCR– 577–580 (1998).
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene
ABL: in vitro and in vivo studies. Blood 108, Abstract 118. Paez, J. G. et al. EGFR mutations in lung cancer:
ABL1 | ABL2 | BCL2 | BCR
1358 (2006). correlation with clinical response to gefitinib therapy.
OMIM:
103. Modugno, M. et al. Targeted inhibition of the Science 304, 1497–1500 (2004).
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM
gatekeeper mutant T315I of BCR–ABL by an Aurora 119. Lynch, T. J. et al. Activating mutations in the
Analplastic large-cell lymphoma | chronic myeloid leukaemia |
kinase inhibitor. Proc. Am. Assoc. Cancer Res. epidermal growth factor receptor underlying
GIST | lung cancer
Abstract 3253 (2007). responsiveness of non‑small‑cell lung cancer to
UniProtKB: http://ca.expasy.org/sprot
104. Regan, J. et al. Pyrazole urea-based inhibitors of p38 gefitinib. N. Engl. J. Med. 350, 2129–2139
AURKA | AURKB | AURKC | BLK | FGR | FYN | HCK| LCK | LYN |
MAP kinase: from lead compound to clinical (2004).
SRC | YES
candidate. J. Med. Chem. 45, 2994–3008 (2002). 120. Tamborini, E. et al. A new mutation in the KIT ATP
105. Kuma, Y. et al. BIRB796 inhibits all p38 MAPK pocket causes acquired resistance to imatinib in a FURTHER INFORMATION
isoforms in vitro and in vivo. J. Biol. Chem. 280, gastrointestinal stromal tumor patient. M. D. Anderson Leukemia Department:
19472–19479 (2005). Gastroenterology 127, 294–299 (2004). http://www.mdanderson.org/departments/leukemia
106. Fabian, M. A. et al. A small molecule-kinase 121. Kobayashi, S. et al. EGFR mutation and resistance of
interaction map for clinical kinase inhibitors. Nature non‑small‑cell lung cancer to gefitinib. N. Engl. J. Med. All links are active in the online pdf
Biotech. 23, 329–336 (2005). 352, 786–792 (2005).

848 | october 2007 | volume 6 www.nature.com/reviews/drugdisc


© 2007 Nature Publishing Group

You might also like