ArticlePDF AvailableLiterature Review

25 Years of Small Molecular Weight Kinase Inhibitors: Potentials and Limitations

Authors:

Abstract

Deregulation of protein and lipid kinases of these kinase activities lead to a variety of pathologies ranging from cancer to inflammatory diseases, diabetes, infectious diseases, cardiovascular disorders, cell growth and survival. Protein kinase and lipid kinases represent, therefore, an important target for the pharmaceutical industry. In fact, approximately one third of all protein targets under investigation in the pharmaceutical industry are protein or lipid kinases. To date 27 kinase inhibitors that have been approved, which, with few exceptions, are mainly for oncological indications and are directed against only a handful of protein and lipid kinases, leaving 70% of the kinome untapped. Despite of these successes the major challenges in kinase drug discovery the development of kinase inhibitors with outstanding selectivity, the identification and validation of driver kinase(s) in diseases and the emerging resistance to the inhibition of key target kinases remain major challenges. This review will provide an insight into protein and lipid kinase drug discovery with respect to achievements, binding modes of inhibitors and novel avenues for the generation of second generation kinase inhibitors to treat cancers. The American Society for Pharmacology and Experimental Therapeutics.
1521-0111/87/5/766775$25.00 http://dx.doi.org/10.1124/mol.114.095489
MOLECULAR PHARMACOLOGY Mol Pharmacol 87:766775, May 2015
Copyright ª2015 by The American Society for Pharmacology and Experimental Therapeutics
MINIREVIEW
25 Years of Small Molecular Weight Kinase Inhibitors: Potentials
and Limitations
Doriano Fabbro
PIQUR Therapeutics AG, Basel, Switzerland
Received August 20, 2014; accepted December 30, 2014
ABSTRACT
Deregulation of protein and lipid kinase activities leads to a variety
of pathologies, ranging from cancer inflammatory diseases, dia-
betes, infectious diseases, and cardiovascular disorders. Protein
kinases and lipid kinases represent, therefore, an important target
for the pharmaceutical industry. In fact, approximately one-third of
all protein targets under investigation in the pharmaceutical in-
dustry are protein or lipid kinases. To date, 30 kinase inhibitors
have been approved, which, with few exceptions, are mainly for
oncological indications and directed against only a handful of
protein and lipid kinases, leaving 70% of the kinome untapped.
Despite these successes in kinase drug discovery, the develop-
ment of kinase inhibitors with outstanding selectivity, identification
and validation of driver kinase(s) in diseases, and the emerging
problem of resistance to the inhibition of key target kinases remain
major challenges. This minireview provides an insight into protein
and lipid kinase drug discovery with respect to achievements,
binding modes of inhibitors, and novel avenues for the generation
of second-generation kinase inhibitors to treat cancers.
Introduction
Protein and lipid kinases represent, after GTP protein
coupled receptors and proteases, one of the most important
target classes for treating human disorders. A set of divergent
kinases play essential roles in eukaryotic signaling. These
include various protein kinases like BRAF, ABL, ALK, as
well as kinases that are able to phosphorylate phosphatidyl-
inositol (PI) like PI3Ks and PI4Ks (Engelman, 2009; Courtney
et al., 2010). Many human malignancies are associated with
deregulated activation of protein or lipid kinases due to
mutations, chromosomal rearrangements, and/or gene am-
plification (Hanahan and Weinberg, 2000; Hunter, 2000;
Blume-Jensen and Hunter, 2001; Cohen, 2002; Hahn and
Weinberg, 2002; Weinstein, 2002; Bardelli et al., 2003; Levitzki,
2003; Vieth et al., 2004, 2005; Luo et al., 2009; Fedorov et al.,
2010; Fabbro et al., 2011).
The human kinome contains typical, atypical protein
kinases, as well as pseudo-kinases (Manning et al., 2002;
http://kinase.com/kinbase). The latter makes up 10% of human
protein kinases and are either inactive or only weakly active
due to the lack of at least one of three motifs in the catalytic
domain that are essential for catalysis (Boudeau et al., 2006;
Kannan and Taylor, 2008). Despite lacking the ability to
phosphorylate substrates, pseudo-kinases are still very impor-
tant in regulating diverse cellular processes, as indicated by the
regulation of LKB1 by STRAD and the activation of JAK2 by
V617F on its JH2 pseudo-kinase domain (Boudeau et al., 2006;
Kannan and Taylor, 2008).
Protein kinases have a bilobal architecture, with one N-terminal
lobe with mainly b-sheets and one C-terminal domain with
ahelices. The two lobes are connected by the hinge region,
which lines the ATP-binding site, which is targeted by the
majority of small molecular weight kinase inhibitors (Cohen,
2002; Levitzki, 2003; Vieth et al., 2004, 2005; Cowan-Jacob,
2006; Fabbro et al., 2011). In past decades, 29 small-molecule
kinase inhibitors have been approved for clinical use, mainly
for oncological indications. Despite these successes, three major
challenges in kinase drug discovery remain:
1. Kinase inhibitors with outstanding selectivity are likely
to become important not only for minimizing side effects
and allowing chronic treatment of nonlife-threatening
diseases, but also to better understand the on- and off-
target pharmacology of kinase inhibitors (Noble et al.,
2004; Fabbro et al., 2011; Cowan-Jacob et al., 2014).
2. Identification and validation of the driver kinase(s) in
human malignancies and disorders (Weinstein, 2002;
Luo et al., 2009) by genome-wide screening for kinase
amplifications/translocations/mutations in conjunction
dx.doi.org/10.1124/mol.114.095489.
ABBREVIATIONS: CML, chronic myeloid leukemia; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; myr,
myristate; PI, phosphatidyl-inositol; PX-866, 1E,4S,4aR,5R,6aS,9aR)-5-(acetyloxy)-1-[(di-2-propen-1-ylamino)methylene]-4,4a,5,6,6a,8,9,9a-octa-
hydro-11-hydroxy-4-(methoxymethyl)-4a,6a-dimethyl-cyclopenta[5,6]naphtho[1,2-c]pyran-2,7,10(1H)-trione; SSR128129E, benzoic acid, 2-amino-
5-[(1-methoxy-2-methyl-3-indolizinyl)carbonyl]-, sodium salt.
766
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
with proteomic technologies. These studies have re-
vealed how the mutational status of kinases may be
associated with various cancer conditions (Hunter,
2000; Blume-Jensen and Hunter, 2001; Cohen, 2002;
Bardelli et al., 2003; Sawyers, 2004; Vieth et al., 2004;
Takano et al., 2005; Ventura and Nebreda, 2006; Wolf-
Yadlin et al., 2006; Ali and Ali, 2007; Engelman et al.,
2007; Greenman et al., 2007; Thomas et al., 2007).
3. Emerging resistance to the inhibition of key target
kinases. Multiple mechanisms of resistance have been
and are being elucidated to improve the efficacy of these
types of targeted therapies.
In this review, we will update the current status of kinase
drug discovery and discuss strategies of how to override these
various types of resistances, including compounds capable of
circumventing the target related drug resistance (Lombardo
et al., 2004; Weisberg et al., 2005; Adrian et al., 2006; Quintas-
Cardama et al., 2007; Engelman et al., 2008b; Zhang et al.,
2010).
Current Status of Protein and Lipid Kinase Drug
Discovery
Protein Kinase Domain. Thanks to the many available
structures of kinase domains, we have a reasonable under-
standing of the various structural elements that are required for
the phosphorylation reaction (Cowan-Jacob et al., 2014). Protein
kinase domains consist of a small, mostly b-structured N-lobe
connected by a short hinge fragment to a larger a-helical
C-lobe. ATP binds in the cleft between the N- and C-terminal
lobes of the kinase domain where the adenine group of ATP is
sandwiched between hydrophobic residues and makes contact
via hydrogen bonds to the hinge (Nolen et al., 2004; Taylor and
Kornev, 2011; Fabbro et al., 2011). Just N-terminal of the
hinge, deep in the ATP pocket, is an important residue called
the gatekeeper, which is often mutated in kinase alleles
resistant to inhibitors. The gatekeeper controls the access to
the hydrophobic back pocketof the kinase (Nolen et al., 2004;
Kornev et al., 2006; Cowan-Jacob et al., 2009; Taylor and
Kornev, 2011; Moebitz and Fabbro, 2012). An outstanding
structural element of the ATP-binding sites is the activation
loop (also called the activation segment or A-loop), with its
N-terminal Asp-Phe-Gly (DFG) motif, whose orientation is piv-
otal for the access of the protein substrate sites (Nolen et al.,
2004; Cowan-Jacob, 2006). Other elements required for catalysis
are the C-helix, which contains the Glu that forms a salt bridge
with the active site Lys (the Ala-X-Lys motif in the b3-strand),
thereby anchoring and orienting the ATP, P-loop (a Gly-rich loop
also known as the G-loop), which contributes to coordination of
the phosphates of ATP, catalytic loop (the Y/HRD motif in the
b6/b7-strand), where Asp functions as a base acceptor for the
proton transfer, and finally, the abovementioned DFG motif,
where Asp binds the Mg
21
ions that coordinate the b-and
g-phosphates of ATP in the ATP-binding cleft (Hanks and
Hunter, 1995; Nolen et al., 2004; Cowan-Jacob, 2006; Taylor
and Kornev, 2011).
Approved Kinase Inhibitors. Since the approval of
fasudil in 1995, sirolimus in 1999, and imatinib in 2001, the
number of approved kinase inhibitors has increased to 30, with
many others still in the preclinical state (Table 1). More than
130 kinase inhibitors are reported to be in phase 13 clinical
trials (http://www.clinicaltrials.gov/) (Vieth et al., 2005). It is
beyond the scope of this review to discuss all of the protein
kinase inhibitors that are in preclinical or clinical development.
Although there are many more kinase inhibitors in development,
it should be emphasized that all of the mentioned approved and
advanced kinase inhibitors do not cover more than 1015% of
the whole kinome (Fedorov et al., 2010).
With a few exceptions, like the rapalogs (everolimus and
temsirolimus) and trametinib, all of the small molecular
weight kinase inhibitors are directed toward the ATP-binding
site. The conservation of ATP binding in the human kinome
causes these ATP mimics to often crossreact with many other
different kinases, resulting in compounds with promiscuous
profiles like, for example, dasatinib (Lombardo et al., 2004) or
sunitinib (Motzer et al., 2006; Faivre et al., 2007). The relative
restrictions imposed by the ATP pharmacophore make it
increasingly difficult to operate in a free intellectual property
space and has increased the interest in identifying inhibitors
that do not compete with ATP (Fabbro et al., 2011; Moebitz and
Fabbro, 2012; Cowan-Jacob et al., 2014).
Most of the approved kinase drugs are active against more
than one type of cancer. Only a few of them have been used for
the treatment of nononcological indications, namely, tofacitinib
for rheumatoid arthritis, nitedanib for idiopathic pulmonary
fibrosis, everolimus for organ rejection of the heart and kidney,
and fasudil for cerebral vasospasm (Table 1).
Also, most of the approved kinase drugs are active against
more than one type of cancer. On the other hand, there are
multiple kinase drugs for one single indication. For example
imatinib, nilotinib, dasatinib, bosutinib, and ponatinib have
all been approved for chronic myeloid leukemia (CML), while
sorafenib, sunitinib, everolimus, temsirolimus, axitinib, or
pazopanib are indicated for various stages of renal cell cancer.
Ceritinib and crizotinib are indicated for nonsmall-cell lung
cancer with ALK translocations, while gefitinib, erlotinib, and
afatinib are indicated for nonsmall-cell lung cancer with an
activated epidermal growth factor receptor (EGFR). Vandetanib
and cabozantinib are used for the treatment of medullary thyroid
carcinoma, while imatinib, sunitinib, and regorafenib are in-
dicated for gastrointestinal tumors. Finally, vemurafenib alone
or the combination of dabrafenib with trametinib is indicated
for metastatic melanoma with BRAFV600 mutations (Table 1).
The actual landscape of kinase inhibitor drugs developed
over the last two decades shows that only a handful of protein
kinases (about 10% of the kinome) have been successfully targeted
with inhibitors, which mainly target oncological indications. It
should be emphasized that selective protein kinase inhibitors
will not only be important for the treatment of diseases but also
as useful reagents to better understand the on- and off-target
kinase biology (Robert et al., 2005; Force et al., 2007; Knapp
et al., 2013).
Binding Modes of Kinase Inhibitors
Kinase inhibitors show different modes of binding to kinases.
In principle, one can divide kinase inhibitors into those that bind
covalently or reversibly to the kinase (Zhang et al., 2009; Cowan-
Jacob et al., 2014).
Covalent Inhibitors
Inhibitors that bind covalently may have reversible or ir-
reversible binding modes, depending on the reactivity of the
Kinase Inhibitors 767
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
war-head (Cowan-Jacob, 2006; Zhang et al., 2009; Liu et al.,
2013). Many inhibitors bind covalently to the ATP site to form
a bond to a Cys residue in or around the active site, preventing
the binding of ATP to the protein kinase (Zhang et al., 2009;
Liu et al., 2013). The most prominent example is the recently
approved BTK inhibitor ibrutinib and the EGFR inhibi-
tor afatinib, which targets the Cys in the bottom part of the
ATP-binding site [receptor protein tyrosine kinase (RTK)]
(Zhou et al., 2009, 2011; Wiestner, 2013; Akinleye et al., 2014).
More covalent inhibitors targeting these cysteines are being
developed to target the most resistant form of the EGFR RTK
that emerges upon treatment with noncovalent EGFR in-
hibitors (Zhang et al., 2009; Zhou et al., 2011; Liu et al., 2013).
An early discovered type of covalent binder is exemplified by
Wortmannin (or its derivative PX-866 [1E,4S,4aR,5R,6aS,
9aR)-5-(acetyloxy)-1-[(di-2-propen-1-ylamino)methylene]-4,4a,
5,6,6a,8,9,9a-octahydro-11-hydroxy-4-(methoxymethyl)-4a,
6a-dimethyl-cyclopenta[5,6]naphtho[1,2-c]pyran-2,7,10(1H)-trione]),
TABLE 1
Approved kinase inhibitors as of October 2014
The 30 kinase inhibitors approved to date are shown with generic name, compound code, trade name, primary indications, year of approval, company, and mode of binding. The
chemical structures and biochemical profiles of the 30 approved kinase inhibitors can be viewed in the IUPHAR database (http://www.guidetopharmacology.org/GRAC/
LigandListForward?type=Approved&database=all). The approved kinase inhbitors include fasudil (HA-1077), sirolimus, imatinib (Glivec), gefitinib (Iressa), erlotinib
(Tarceva), lapatinib (Tykerb), sorafenib (Nexavar), sunitinib (Sutent), dasatinib (Sprycel), nilotinib (Tasigna), torisel (Temsirolimus), everolimus (Rad001) as Afinitor, Zortress,
Certican, and Votubia, crizotinib (Xalcori), vandetanib (Caprelsa), ruxolitinib (Jakafi), vemurafenib (Zelboraf), axitinib (Inlyta), regorafenib (Stivarga), pazopanib (Votrient),
tofacitnib (Xeljanz), cabozantinib (Cometriq), ponatinib (Iclusig), bosutinib (Bosulif), dabrafenib (Tafinlar), trametinib (Mekinist), afatinib (Gilotrif), ibrutinib (Imbruvica),
ceritinib (Zykadia), idelalisib (Zydelig), and nintedanib (Vargatef). All compounds are commercially available.
Generic Name (Compound Code, Trade Names) Kinase Target Disease Company (Year, Type)
Fasudil (HA-1077) ROCK Cerebral vasospasm, PAH Asahi Kasei (1995,
type 1)
Sirolimus (Rapamune) mTOR Kidney transplants Pfizer, Wyeth (1999,
type 3)
Imatinib (STI571, Glivec, Gleevec) ABL, PDGFR, KIT CML, Ph+ B-ALL, CMML,
HES, GIST
Novartis (2001, type 2)
Gefitinib (ZD1839, Iressa) EGFR NSCLC AZ (2003, type 1)
Erlotinib (OSI-774,Tarceva) EGFR NSCLC, pancreatic cancer Roche, OSI (2004, type 1)
Sorafenib (BAY 43-9006, Nexavar) VEGFR2, PDGFR, KIT, FLT3,
BRAF
RCC, HCC Bayer, Onyx (2005, type 2)
Sunitinib (SU11248, Sutent) VEGFR, KIT, PDGFR, RET,
CSF1R, FLT3
RCC, imatinib-resistant GIST Pfizer (2006, type 1)
Lapatinib (GW2016, Tykerb) EGFR, ERBB2 BC GSK (2007, type 1.5)
Dasatinib (BM-354825,Sprycel) ABL, PDGFR, KIT, SRC CML BMS (2007, type 1)
Nilotinib (AMN107,Tasigna) ABL, PDGFR, KIT CML Novartis (2007, type 2)
Everolimus (Rad001, Certican, Zortress,
Afinitor, Votubia)
mTOR RCC, SEGA, transplantation Novartis (2009, type 3)
Temsirolimus (CCI-779, Torisel) mTOR RCC Pfizer, Wyeth (2009,
type 3)
Crizotinib (PF-02341066, Xalcori) MET and ALK NSCLC with ALK translocations Pfizer (2011, type 1)
Vandetanib (ZD6474, Caprelsa) RET, VEGFR1, VEGFR2, FGFR,
EGFR
MTC AZ (2011, type 1)
Ruxolitinib (INC424, Jakafi) JAK2 IMF with JAK2V617F mutations Novartis, Incyte (2011,
type 1)
Vemurafenib (PLX4032, RG7204,
Zelboraf)
BRAF Metastatic melanoma with
BRAFV600E mutations
Roche, Plexxikon (2011,
type 2)
Axitinib (AG013736, Inlyta) VEGFR, KIT, PDGFR, RET,
CSF1R, FLT3
RCC Pfizer (2012, type 1)
Regorafenib (BAY 73-4506, Stivarga) VEGFR2, Tie2 CRC, GIST Bayer (2012, type 2)
Pazopanib (GW-786034, Votrient) VEGFR, PDGFR, KIT RCC GSK (2012, type 1)
Tofacitinib (CP-690550, Xeljanz,
tasocitinib)
JAK3 RA Pfizer (2012, type 1)
Cabozantinib (XL184, BMS907351,
Cometriq)
VEGFR2, PDGFR, KIT, FLT3 MTC Exelexis (2012, type 1)
Ponatinib (AP24534, Iclusig) ABL Imatinib-resistant CML with
T315I mutations
Ariad (2012, type 1)
Bosutinib (SKI-606, Bosulif) ABL CML resistant/intolerant to therapy Pfizer (2012, type 1)
Dabrafenib (Tafinlar) BRAF Metastatic melanoma with
BRAFV600E mutations
GSK (2013, type 2)
Trametinib (Mekinist) MEK Metastatic melanoma with
BRAFV600E mutations
GSK (2013, type 3)
Afatnib (Gilotrif, Tomtovok, Tovok) EGFR NSCLC with EGFR-activating
mutations
BI (2013, covalent)
Ibrutinib (PCI-32765, Imbruvica) BTK MCL, CLL Janssen, Pharmacyclic
(2013, covalent)
Ceritinib (LDK378, Zykadia) ALK NSCLC with ALK translocations Novartis (2014, type 1)
Idelalisib (CAL101, GS1101, Zydelig) PI3Kdelta CLL, FL and SLL Gilead, Calistoga, ICOS
(2014, type 1)
Nintedanib (BIBF 1120, Vargatef,
intedanib)
VEGFR, PDGFR, FGFR Idiopathic pulmonary fibrosis BI (2014, type 1)
AZ, Astra-Zeneca; BC, breast cancer; BI, Boehringer-Ingelheim; BMS, Bristol-Myers Squibb; CLL, chronic lymphocytic leukemia; CMML, chronic monomyelocytic
leukemia; CRC, colorectal cancer; FL, follicular lymphoma; HCC, hepatocellular carcinoma; HES, hyper-eosinophilic syndrome; IMF, interstitial myelofibrosis; GIST,
gastrointestinal stromal tumor; GSK, Glaxo-Wellcome; MCL, mantle cell lymphoma; MTC, medullary thyroid cancer; PAH, Pulmonary Arterial Hpertension; Ph+ B-ALL,
Philadelphia chromosome positive B-cell acute lymphoblastic leukemia; RA, rheumatoid arthritis; RCC, renal clear cell carcinoma; SEGA, subependymal giant cell
astrocytoma; SLL, small lymphocytic leukemia.
768 Fabbro
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
which binds to the active site Lys in the ATP-binding site of the
PI3K (Wymann et al., 1996; Ihle et al., 2004). Various covalent
inhibitors targeting different cysteines in various kinases have
been reported (Liu et al., 2013; Kwiatkowski et al., 2014).
Reversible (Noncovalent) Inhibitors
Noncovalent kinase inhibitors can be further classified into
those which either bind or do not bind to the hinge region of
the kinase, leading to the classification of type 13 reversible
kinase inhibitors (Liu and Gray, 2006; Zhang et al., 2009).
These have been briefly summarized below.
Type 1 and 1.5 Inhibitors. These inhibitors fulfill all of
the criteria of ATP mimetics by binding to the hinge and
targeting an active conformation of the kinase (often referred
to as DFG-in). In the active kinase, the A-loop adopts an open
conformation typical for the ATP-bound state of the kinase
where the Asp in the DFG motif coordinates the phosphates of
ATP, while Phe stabilizes helix C and A-loop for catalysis
(Nolen et al., 2004; Cowan-Jacob, 2006; Liu and Gray, 2006;
Zhang et al., 2009; Cowan-Jacob et al., 2014). Many of the type
1 inhibitors use variation in the size, shape, and polarity of
the gatekeeper residue to gain selectivity and knowledge about
the various inactive conformations of different kinases, which
allows one to combine these features (Liu and Gray, 2006;
Cowan-Jacob et al., 2014). The bias toward kinase inhibitors
that target the active conformation of the kinase stems from
the early days of kinase drug discovery, where an ATP mimetic
design, together with the use of enzymatic assays displaying
the highest level of activity, was used. Classic examples for this
type of approved kinase inhibitor class are gefitinib, erlotinib,
dasatinib, and sunitinib (Table 1).
The type 1.5 inhibitor is a subtype of the type 1 inhibitor
that binds to an inactive kinase conformation (Nolen et al.,
2004; Cowan-Jacob, 2006; Liu and Gray, 2006; Zhang et al.,
2009; Cowan-Jacob et al., 2014). Laptinib in the EGFR-RTK
adopts a DFG-in conformation, which is typical of an active
kinase, but helix C is pushed out by lapatinib, effectively
disrupting the ion pairing between the active site Lys and the
Glu from helix C (Wood et al., 2004). This type of binding to
the DFG-in inactive conformation, which is referred to as type
1.5 inhibition, has also been observed in other kinases (Cowan-
Jacob et al., 2014).
Although the structures of protein kinases in the active
ATP-bound state are very similar, specificity can be gained
through particular featuresintheATP-bindingsitesof
kinases. More selective type 1 inhibitors are those that, like
the type 1.5 inhibitors, use additional sites close to the ATP
binding, like the adjacent hydrophobic pockets whose entry is
regulated by the gatekeeper (Zuccotto et al., 2010). Other type
1 inhibitors interact with two different sites on the kinase, such
as the ATP site and the peptide-binding site like the bivalent/
bitopic inhibitors (Hill et al., 2012), macrocycles (Tao et al.,
2007), or some of the covalent inhibitors (Liu et al., 2013).
Type 2 Inhibitors. Type 2 kinase inhibitors bind to the
inactive, so-called DFG-out conformation, maintaining con-
tacts to the hinge region and usually displaying an ATP-
competitive behavior similar to type 1 inhibitors (Nolen et al.,
2004; Cowan-Jacob, 2006; Liu and Gray, 2006; Zhang et al.,
2009). The transition of the active DFG-in to the inactive DFG-
out conformation exposes an additional hydrophobic pocket
adjacent to the ATP site, which is used by type 2 inhibitors,
locking the kinase in the inactive conformation (Nolen et al.,
2004; Cowan-Jacob, 2006; Liu and Gray, 2006; Zhang et al.,
2009; Cowan-Jacob et al., 2014). Approved kinase inhibitors
binding to or stabilizing the DFG-out conformations are, for
example, imatinib, nilotinib, or sorafenib (Table 1).
In addition to DFG-out, combinations of different confor-
mational states of helix C, the A-loop, and/or P-loop can gen-
erate various inactive conformations of the kinase domain
(Cowan-Jacob, 2006; Cowan-Jacob et al., 2014). Each individ-
ual kinase has a preferred inactive conformation, dependingon
its phosphorylation state and regulatory mechanisms involving
structures outside the kinase domain (Cowan-Jacob et al.,
2014).
Type 3 Inhibitors. Type 3 kinase inhibitors are non-ATP
competitive (allosteric) kinase inhibitors that have no phys-
ical contact with the hinge. As they exploit binding sites and
regulatory mechanisms that are unique to a particular kinase,
they therefore show the highest degree of selectivity (Nolen
et al., 2004; Cowan-Jacob, 2006; Liu and Gray, 2006; Zhang
et al., 2009; Cowan-Jacob et al., 2014). The noncatalytic role of
kinases involving unique nonconserved interactions and which
increase the target space on the kinome have only recently
been appreciated (Rauch et al., 2011; Cowan-Jacob et al., 2014).
Type 3 inhibitors can either bind to the kinase domain (close to
or removed from the ATP site) or to sites that are located
outside of the kinase domain. These type 3 inhibitors include
very diverse compounds, ranging from the MEK inhibitors to
rapamycin derivatives.
The allosteric inhibitor of MEK binds to a pocket adjacent
to the ATP-binding site, which is referred to as the allosteric
back-pocket(Ohren et al., 2004). These type 3 allosteric
inhibitors can either bind in the presence or absence of ATP.
Those that bind in the presence of ATP (DFG-in conformation,
such as MEK inhibitors) can be referred to as allosteric back-
pocket DFG-in inhibitors.Alternatively,allosteric back-pocket
DFG-out inhibitorscan bind to the allosteric back-pocket in the
absence of ATP (DFG-out conformation) and include allosteric
inhibitors of insulin-like growth factor 1 receptor (IGF-1R)
(Heinrich et al., 2010), FAK (Tomita et al., 2013), or p38 (Over
et al., 2013). In the case of IGF-1R, the inhibitor binds in the
DFG-out pocket and extends over the catalytic loop rather than
pushing it out from underneath like the FAK inhibitor, requiring
additional conformational changes (Tomita et al., 2013). The
allosteric AKT inhibitors are a special case of the allosteric back-
pocket DFG-out inhibitors, as they only bind to this site when
the pleckstrin homology domain is present (Barnett et al., 2005;
Lindsley et al., 2005). Thus, the identification of this allosteric
inhibitor that binds at the interface between the kinase domain
and pleckstrin homology domain was only possible using the full-
length protein for AKT (Barnett et al., 2005; Lindsley et al.,
2005). While lack of competition with ATP has, in some cases,
proven to be a useful way to identify type 3 inhibitors, it should
be pointed out that the allosteric back-pocket DFG-out inhib-
itors will score as ATP competitive.
Type 3 inhibitors that are further away from the ATP site
are the myristate (myr) pocket binders located in the bottom
of the C-lobe of ABL (Adrian et al., 2006; Zhang et al., 2009;
Fabbro et al., 2010), CHK1 inhibitors that occupy, in part,
the substrate-binding site (Converso et al., 2009), and JNK1
inhibitors that occupy the mitogen-activated protein kinase
insert region and A-loop (Comess et al., 2011), to only cite
a few. A more comprehensive review on type 3 inhibitors has
Kinase Inhibitors 769
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
been recently assembled by Cowan-Jacob et al. (Cowan-Jacob
et al., 2014).
Rapamycins appear to be further removed from the kinase
domain, as they specifically target the mTOR kinase in the
context of the Raptor-containing mTORC1 complex (Wang
and Sun, 2009; Yang et al., 2013). Similarly, agents targeting
the extracellular domains of RTKs act outside the kinase
domain (Christopoulos et al., 2014). The extracellular do-
mains of RTKs can be targeted by peptide mimetics, peptoids,
or antibodies (Fleishman et al., 2002; Udugamasooriya et al.,
2008; Cazorla et al., 2010; Jura et al., 2011). The monoclonal
antibodies trastuzumab (Herceptin) and pertuzumab (2C4,
Perjeta) act at different domains, with trastuzumab binding
to domain IV and pertuzumab binding to subdomain II of the
extracellular segments of the HER2/neu receptor, respectively
(Cho et al., 2003; Hynes and Lane, 2005; Hsieh and Moasser,
2007). On the other hand, small molecules like SSR128129E
[benzoic acid, 2-amino-5-[(1-methoxy-2-methyl-3-indolizinyl)
carbonyl]-, sodium salt], which targets the extracellular D2D3
domains of the fibroblast growth factor receptor (FGFR), mod-
ulate signaling of the FGFR RTKs (Bono et al., 2013; Herbert
et al., 2013). Examples of approved type 3 inhibitors are
trametinib and the rapamycins.
Activators, Paradoxical Activation, and Priming.
Targeting the allosteric sites on protein kinases may also
lead to the identification of activators rather than inhibitors,
which could be useful for therapeutic intervention or as phar-
macological tools. In particular, compounds targeting the PIF
pocket (the hydrophobic motif present in the N-terminal lobe
of AGC kinases) of either PDK1 or PKCzcan either act as
activators (Hindie et al., 2009) or substrate-selective inhibitors
(Lopez-Garcia et al., 2011; Sadowsky et al., 2011; Busschots
et al., 2012). Similarly, the myr-pocket binders of ABL can be
converted into activators if they are designed not to bend helix I
of the ABL kinase domain (Jahnke et al., 2010; Yang et al.,
2011).
On the other hand, there are a few protein kinases that
require activation rather than inhibition to fulfill their
therapeutic task, like in the case of AMP-activated protein
kinase and the insulin receptor for which activators have been
identified (Li et al., 2001; Pender et al., 2002; Sanders et al.,
2007; Lee et al., 2011; Salt and Palmer, 2012). PKC activation
by exogenous compounds acting at the diaclyglycerol-binding
site can have tumor-promoting or tumor-suppressive effects
(Martiny-Baron and Fabbro, 2007). These include phorbol
esters, bryostatin, and other compounds acting as diaclyglycerol
mimetics (Martiny-Baron and Fabbro, 2007). Another example
of kinase activators includes a mimetic of the brain-derived
neurotrophic factor that activates TrkB (NTRK-2) (Massa et al.,
2010).
In some cases, kinase inhibitors can lead to unintended
paradoxical activation either directly or via modulation of
feedback loops. The most striking example is the paradoxical
activation of RAF inhibitors, which can activate the mitogen-
activated protein kinase pathway in certain genetic backgrounds
(Hall-Jackson et al., 1999). This phenomenon is linked to a
complex regulation of RAF due to crossactivation of the wild-
type RAF isoforms, which is just beginning to be understood,
almost a decade after the first so-called RAF inhibitor sorafenib
was approved (Hall-Jackson et al., 1999; Hatzivassiliou et al.,
2010; Poulikakos et al., 2010; Holderfield et al., 2013).
Thus, BRAF inhibitors can activate MEK and extracellular
signal-regulated kinase (ERK) signaling in normal cells, result-
ing not only in the absence of hypoproliferative toxicities but
actual promotion of hyperproliferative toxicities (Su et al., 2012).
Another phenomenon is priming, which can lead to ac-
tivation via kinase inhibitors, which has been observed for
several kinases like AKT, MEK, and JAK (Okuzumi et al.,
2009; Andraos et al., 2012; Hatzivassiliou et al., 2013). Prim-
ing describes the up-regulation of the phosphorylated form of
the targeted kinase upon inhibition, which can lead to an
activation of the pathway once the inhibitor is removed. Prim-
ing may also depend on the mode of action of the inhibitor.
Inhibitors binding to active AKT do cause priming, whereas
allosteric inhibitors targeting the inactive conformation of AKT
do not (Lin et al., 2012).
There is currently no general strategy for the identification
of allosteric kinase inhibitors or activators, as most of them
have been discovered serendipitously by diverse approaches,
ranging from phenotypic screenings to highly sophisticated
structure-based drug design.
Resistance to Kinase Inhibitors (in Oncology)
Introduction to Drug Resistance. Most tumor cells
express only a few of the 150 so-called driver genes, which,
when mutated, promote tumorigenesis by affecting a dozen
signaling pathways regulating cell fate, cell survival, and
genome maintenance (Vogelstein et al., 2013). Insights into
particular mutations and their consequences for signaling
have already led to the development of more effective drugs
(Siegel et al., 2012). The finding that deregulation of protein
kinase activity by the gain of function mutations is essential
to maintain the oncogenic state is also reflected by the fact that
30 kinase inhibitors have been approved for various oncological
indications (Janne et al., 2009; Luo et al., 2009; Sharma and
Settleman, 2010; Yarden and Pines, 2012) (Table 1). In many
instances, these mutations can be linked to specific cancers
(Greenman et al., 2007; Thomas et al., 2007). Although many
activating mutations in various kinases have been found in
a variety of cancers, it will take another effort to unambigu-
ously identify the dependence of tumor growth on a particular
kinase or its pathway(s). The major difficulty is the intrinsic
heterogeneity of the late-stage forms of cancer that harbor
multiple mutations, chromosomal aberrations, and display
genomic instability. Demonstrating the cancer dependence of
the kinase target will not only lead to the identification of
inhibitors of the kinase, but may also accelerate the proof
of concept in clinical trials, allowing a better selection of pa-
tients most likely to respond to targeted therapy.
Kinase inhibitors are being and have been designed to spe-
cifically target kinase alleles with a gain of functions (Blume-
Jensen and Hunter, 2001; Fabbro and García-Echeverría,
2002a). Despite these successes, it should be emphasized that
patients most likely to benefit from these kinase inhibitors
often relapse after an initial response. Thus, the emergence of
drug resistance is not limited to conventional chemotherapeu-
tic drugs but extends to drugs with targeted modes of action
(Engelman and Settleman, 2008a). The mechanisms of multi-
drug resistance to chemotherapeutic drugs have been studied
and are not limited to reduced drug accumulation, but also
involve changes in the level of target proteins, mutations that
diminish drug binding, trapping of drugs in acidic vesicles,
enhanced metabolism of drugs by cytochrome P450 mixed
770 Fabbro
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
function oxidases, increased tolerance of cellular DNA damage,
and diminished apoptotic signaling (Gottesman, 2002; Szakacs
et al., 2006; Hall et al., 2009). Apart from the usual mechanisms
of drug inactivation in cancer (Szakacs et al., 2006; Hall et al.,
2009) as well as the findings that quiescent tumor stem cells are
refractory to kinase inhibitors (Graham et al., 2002), there are
additional target-related mechanisms for resistance that are not
based on mutations of the target kinase.
Resistance to Kinase Inhibitors. Drug resistance to tar-
geted agents like kinase inhibitors can either occur by compen-
satory mechanism or by reducing the affinity of the kinase to its
inhibitors (Szakacs et al., 2006; Fabbro et al., 2011).
The most commonly found point mutation leading to
resistance concomitant with relapses affects the gatekeeper
residue, whose size and shape regulates the properties of the
hydrophobic pocket located at the back of the ATP-binding
site. These mutations include the Thr gatekeeper of BCR-
ABL1 (T315I) (Gorre et al., 2001; Sawyers, 2004; Fabbro et al.,
2005), Kit (T670I) (Heinrich et al., 2003; Fletcher and Rubin,
2007), PDGFRa(T674I) (Cools et al., 2003), PDGFRb(T681I)
(Daub et al., 2004), SRC T341M (Bishop, 2004), as well as
other types of gatekeepers like L1196M in ALK (Katayama
et al., 2012), G697R in FLT3 (Cools et al., 2004), and V561M in
FGFR1 (Blencke et al., 2004). This is either due to a steric
clash between the inhibitor and mutated gatekeeper (Blencke
et al., 2004; Fabbro et al., 2005; Fabbro et al., 2011) or by
significantly increasing the affinity for ATP and thereby
reducing the affinity for the kinase inhibitors (Kobayashi et al.,
2005; Pao et al., 2005). Mutations in the gatekeeper as well as
other kinase domain mutations confer resistance to a wide spec-
trum of kinase inhibitors without affecting the kinase activity
and may explain a fraction of cases of acquired resistance.
There are additional mechanisms to circumvent kinase
inhibition (Sawyers, 2004; Rubin and Duensing, 2006, Fabbro
et al., 2011; Serra et al., 2011; Logue and Morrison, 2012;
Trusolino and Berlotti, 2012; Workman et al., 2013). Com-
pensatory changes in the signaling pathways of treated
cancer cells that can bypass drug-mediated kinase inhibition
and so restore downstream signaling in the presence of drug
include
1. Amplification of the target, like in the case of BCR-
ABL1 in CML (le Coutre et al., 2000) or dimerization of
aberrantly spliced BRAF (V-600E) (Poulikakos et al.,
2011); and
2. Up-regulation of RTKs following inhibition of PI3K
(Serra et al., 2011; Rodon et al., 2013) or up-regulation
of alternative kinase pathways through the receptors
for hepatocyte growth factor (MET), IGF-1R, or AXL in
the acquisition of resistance to EGFR kinase inhibition
(Engelman et al., 2007; Turke et al., 2010; Logue and
Morrison, 2012). Activation downstream of RTKs, like
the RAS-RAF-ERK and/or PI3K/AKT pathways, by
several mechanisms can override the effects of any RTK
inhibitor. Examples include activation of the PI3K/AKT
pathway by activating point mutations in PI3K sub-
units, loss-of-function/deletions of the PTEN phospha-
tase in EGFR-mutated lung cancer, mutations in RAS
isoforms downstream of oncogenic RTKs, MEK activation
by COT1 bypassing inhibited BRAF-V600E, and activa-
tion or the EGFR-mediating resistance to vemurafenib in
BRAF-V600E (She et al., 2003; Johannessen et al., 2010;
Prahallad et al., 2012). Signaling redundancies, inter-
connections through pathway crosstalk, and negative
feedback loops have also been identified as contrib-
utors to drug resistance or weaken the efficacy of kinase
inhibitors (Janne et al., 2009; Rodrik-Outmezguine et al.,
2011; Chandarlapaty, 2012). Negative feedback loops,
crossinhibition, crossactivation, and pathway conver-
gence connect signaling pathways through activated
components and are crucial for the maintenance of
normal cell functions as well as the dynamic and
adaptive responses to extracellular signals (Mendoza
et al., 2011; Logue and Morrison, 2012). Even if a driver
kinase is suppressed by the inhibitor, the tumor cells
can exploit its interconnected signaling pathways to
evolve drug resistance (Trusolino and Berlotti, 2012).
The Ras-ERK and PI3K-mTOR signaling provide major
mechanisms for the regulation of cell survival, differen-
tiation, proliferation, metabolism, and motility in re-
sponse to extracellular ligands and are activated in over
80% of all tumors (Engelman, 2009; Wong et al., 2010).
Selective inhibition of mTORC1 leads to disruption of
a negative feedback loop, which enhances the activity of
PI3K and its effector AKT, counteracting the antiproli-
ferative effects of mTOR inhibition (Chandarlapaty,
2012). Combined inhibition of the mTOR kinase and
RTKs therefore seems a promising approach to abolish
AKT signaling and prevent resistance formation (Rodrik-
Outmezguine et al., 2011). Alternatively, inhibition of
PI3K/mTOR signaling resulted in the activation of the
Jak/STAT5 pathway, suggesting a combination strategy
targeting both the PI3K/mTOR and JAK/STAT5 signal-
ing (Britschgi et al., 2012). Negative feedback regulation
hasalsobeenobservedintheRAS-RAF-ERKcascade
(Mendoza et al., 2011). The binding of vemurafenib to
wild-type BRAF in cells can cause BRAF/CRAF hetero-
dimerization and paradoxical activation of ERK1/2,
which can result in the development of kerato-acanthomas
in patients (Chapman et al., 2011; Poulikakos et al.,
2011; Su et al., 2012; Holderfield et al., 2014). This is
further exacerbated in the case where oncogenic RAS
and BRAF mutants are present in the same cells that
support RAS-dependent dimerization and paradoxical
ERK activation.
3. Factors regulating the bioavailability and intracellular
concentration of inhibitors like poor intestinal absorption,
tight binding to blood plasma proteins, overexpression of
the multidrug resistance genes, and/or increased metab-
olism of the drug by liver cytochrome P450 proteins have
also been linked to primary resistance (Mahon et al.,
2003; Apperley, 2007).
All of these mechanisms demonstrate the plasticity of
cancer cells and the many ways by which a tumor can evade
targeted therapies. The only way to approach these problems is
to use a rational combination of drugs, which is a mainstay in
cancer therapy.
Breaking the Resistance to Kinase Inhibitors. To cir-
cumvent target-dependent drug resistance, second-generation
kinase inhibitors have been developed. Inhibitors that
bind covalently to the ATP-binding site of EGFR have been
developed for the emerging resistance to gefitinib and erlotinib
(Kwak et al., 2005; Heymach et al., 2006; Felip et al., 2007;
Kinase Inhibitors 771
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
Zhou et al., 2011). Several of these covalent inhibitors are in
late-stage clinical trials (Zhang et al., 2009; Liu et al., 2013).
Alternatively, these types of covalent inhibitors, as in the case
of ibrutinib, have been designed upfront to bind covalently to
Cys481 of BTK and recently approved for B-cell malignancies
(Byrd et al., 2013; Wiestner, 2013; Akinleye et al., 2014). Although
ibrutinib has shown impressive clinical results, patients that have
disease progression revealed a C481S mutation in their BTK,
which abrogates the covalent binding to ibrutinib (Furman et al.,
2014). Additional approaches have attempted to develop non-
covalent inhibitors that can tolerate the amino acid exchange at
the gatekeeper position. Several ATP-site directed inhibitors
that are active against the T315I ABL1 gatekeeper mutant
have been designed, but thus far only ponatinib has been
approved (OHare et al., 2009; Hoy, 2014). To target the gate-
keeper mutation, these types of compounds display a rather
low selectivity that can have deleterious side effects, which may
lead to a temporary pull off the market over safety issues (Force
et al., 2007; Cheng and Force, 2010; Dalzell, 2013).
A further approach is to target the kinase outside the ATP-
binding sites with the goal of combining the ATPsite directed
inhibitors (type 1 and 2) with type 3 (Fabbro et al., 2011;
Cowan-Jacob et al., 2014) or to combine different kinase
inhibitors targeting kinases in the same pathway, like in the
case of vemurafenib, where the emerging resistance is not due to
mutations in BRAF (V600E) but rather in the downstream
MEK1 (Wagle et al., 2011; Medina et al., 2013). Unfortunately,
only a very limited number of non-ATP competitive kinase
inhibitors have thus far been identified, which could also be
used to address the resistance caused by mutations in the ATP
binding site (Fabbro et al, 2011; Cowan-Jacob et al., 2014)
A remote binding site on the kinase domain is addressed
by the GNF-2 compound, which was found by a phenotypic
screen shown to target the myr pocketbinding site of ABL
(Adrian et al., 2006; Zhang et al., 2009; Fabbro et al., 2010).
Exploration of the combined efficacy between the myr pocket
and ATP-binding sites significantly increased the survival of
mice in bone-marrow transplantation CML models relative
to treatment with either agent alone (Zhang et al., 2010). In
addition, the improved potency of a second-generation myr
pocket binder against wt-ABL and T315-ABL also translated
into a high level of degree of synergy in BaF3 cells trans-
formed with BcrABL-T315I when combined ATPsite di-
rected inhibitors like nilotinib or dasatinib, as it has been
noted in previous studies (Fabbro et al., 2002b; Zhang et al.,
2010). Surprisingly, NMR and small-angle X-ray scattering
analysis revealed an open state of the ABL when bound to
ATPsite directed inhibitors, like imatinib, leading to the
detachment of the SH3-SH2 domains from the kinase domain
and the formation of an open inactive state, which is inhibited
in the ATP site, which can be reverted by the addition of the
myr pocket binder (Skora et al., 2013). Whether these data
explain the synergy between the myr pocket binder and ATP-
directed inhibitors that appears to overcome the T315I-ABL
mediated drug resistance remains to be seen. The findings on
the actions of the two classes of inhibitors on a single target
kinase may help to devise new strategies for drug development.
Conclusions and Perspectives
Several kinase inhibitors have become successful drugs. A
large number of kinase inhibitors are in clinical development
mainly for oncology indications, which is a testimony for the
greater tolerability in this indication with respect to potential
side effects. One of the issues that is perceived as a major
challenge in kinase drug discovery is the selectivity for the
target kinases, which defines the on- and off-target pharma-
cology (Cohen, 2002; Fabbro and García-Echeverría, 2002a,b;
Vieth et al., 2004, 2005; Cowan-Jacob, 2006; Liu and Gray,
2006; Zhang et al., 2009). While development of selective
kinase inhibitors is likely to be extremely important from the
standpoint of minimizing drug side effects, inhibitors with
exquisite selectivity are a must for chronic administration in
nonlife-threatening diseases like many immunologic dysfunc-
tions (Cohen, 2002; Fabbro and García-Echeverría, 2002a;
Fabbro et al., 2002b; Vieth et al., 2004, 2005; Liu and Gray,
2006; Zhang et al., 2009). While the lack of selectivity of kinase
inhibitors can be advantageous in cancer treatment, it can also
lead to side effects (Cheng and Force, 2010). On the other hand,
low selectivity complicates mechanism of action studies as well
as biomarker discovery. The information linking adverse side
effects of protein and lipid kinase inhibitors with a particular
protein kinase selectivity profile is only now beginning to
emerge (Force et al., 2007; Olaharski et al., 2009). Moreover, we
are still trying to understand the side effects that are generated
by inhibiting multiple kinases, including undesired kinases that
may generate, at least in part, the adverse side effects. Recent
data, for example, suggest that simultaneous inhibition of ATM,
ATR, DNA-PK, Aurora, and/or other kinases involved in mitosis
may be genotoxic (Force et al., 2007; Olaharski et al., 2009).
The development of kinase inhibitors for nonlife-threatening
indications where chronic regimens are being used will require
a better target selectivity profile to minimize side effects.
Therefore, only few attempts have been made to position kinase
inhibitors in chronic diseases, such as inflammation and
immune disorders. Most prominent is the p38 kinase, which
has been targeted for treatment of nononcology indications
like rheumatoid arthritis.
Addressing the noncatalytic functions of kinases will result
in novel types of kinase inhibitors with improved selectivity.
The design and synthesis of non-ATP competitive (allosteric)
kinase inhibitors with high selectivity will not only lead to the
expansion of the kinase target space with less studied mem-
bers of the family like the atypical and the pseudo-kinases.
Having highly selective kinase inhibitors (ideally against
most of the members of the whole kinome) at hand will propel
the pharmacology as well as understanding of their roles in
signaling and regulation. Thanks to the available kinase
inhibitors, we have learned a lot about drug-protein inter-
actions and cancer cell signaling, in some cases only after
their use in the clinic. We only now begin to appreciate the
complex rules determining the mechanism of action and spec-
ificity of these types of inhibitors. In addition to its clinical
use, the various new drugs and drug candidates will be
instrumental and indispensable reagents for many biologic
disciplines to help decipher kinase regulation, interrogate
signaling pathways, perturb cellular networks, and study
complex cell biologic processes.
One of the major challenges in future kinase drug discovery
is to better understand the cancer dependence of the target
kinase and anticipate the emerging resistance to kinase
inhibitor treatment. With a few exceptions, the development
of kinase inhibitor resistance in cancer has lowered the en-
thusiasm about the initially observed outstanding clinical
772 Fabbro
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
effects. Although, in some cases, we have seen long-lasting
clinical responses with these kinase inhibitors, we have
inevitably and not unexpectedly seen relapses under treat-
ment. Understanding resistance mechanisms has not only
lead to a deeper understanding of cancer cell biology, but also
instructed ways to overcome resistance using second-generation
kinase inhibitors with a better selectivity and appropriate
potency, which are being applied to a genetically better defined
patient population that is most likely to respond. In addition,
clever combinations and sequential treatment regimens are
being tested. It should be noted that clinical responses have
also been noted with many other kinase inhibitors without
a clear rationale, offering encouragement that single agents
can be active in genetically complex neoplasms, although it is
formally possible that the single-agent activity may be due to
its fortuitous ability to inhibit multiple kinases. The recent
introduction of the concept of individualized cancer therapy
along with the development of selective drugs targeting specific
alteration in tumors has provided hope for the development of
more effective treatment strategies.
In summary, the available complement of clinically used
kinase inhibitors do not cover more than 1015% of the whole
kinome. Ongoing efforts using genome-wide screening in con-
junction with the use of genetic organisms will unravel new
disease associations and pave the way for the discovery of
many more new protein kinase targets in the coming years.
Finally, there are many selective protein kinase inhibitors
that cannot be used as drugs for reasons of toxicity or solubility
but which are still extremely useful as research reagents (Robert
et al., 2005; Force et al., 2007). Extending the complement of
very selective (allosteric) protein kinase inhibitors in conjunction
with a system biology approach will advance our understanding
of kinase biology in cellular networking and diseases.
Acknowledgments
The author thanks Adam Pawson and Elena Faccenda from the
IUPHAR database in helping to compile Table 1. This mini-review
could only be written with the valuable help of many publications.
Due to space restrictions, not all of the relevant publications could be
cited.
Authorship Contributions:
Wrote or contributed to the writing of the manuscript: Fabbro.
References
Adrián FJ, Ding Q, Sim T, Velentza A, Sloan C, Liu Y, Zhang G, Hur W, Ding S,
and Manley P et al. (2006) Allosteric inhibitors of Bcr-abl-dependent cell pro-
liferation. Nat Chem Biol 2:95102.
Akinleye A, Furqan M, and Adekunle O (2014) Ibrutinib and indolent B-cell lym-
phomas. Clin Lymphoma Myeloma Leuk 14:253260.
Ali S and Ali S (2007) Role of c-kit/SCF in cause and treatment of gastrointestinal
stromal tumors (GIST). Gene 401:3845.
Andraos R, Qian Z, Bonenfant D, Rubert J, Vangrevelinghe E, Scheufler C, Marque
F, Régnier CH, De Pover A, and Ryckelynck H et al. (2012) Modulation of
activation-loop phosphorylation by JAK inhibitors is binding mode dependent.
Cancer Discov 2:512523.
Apperley JF (2007) Part I: mechanisms of resistance to imatinib in chronic myeloid
leukaemia. Lancet Oncol 8:10181029.
Bardelli A, Parsons DW, Silliman N, Ptak J, Szabo S, Saha S, Markowitz S, Willson
JK, Parmigiani G, and Kinzler KW et al. (2003) Mutational analysis of the tyrosine
kinome in colorectal cancers. Science 300:949.
Barnett SF, Defeo-Jones D, Fu S, Hancock PJ, Haskell KM, Jones RE, Kahana JA,
Kral AM, Leander K, and Lee LL et al. (2005) Identification and characterization of
pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors.
Biochem J 385:399408.
Bishop AC (2004) A hot spot for protein kinase inhibitor sensitivity. Chem Biol 11:
587589.
Blencke S, Zech B, Engkvist O, Greff Z, Orfi L, Horváth Z, Kéri G, Ullrich A,
and Daub H (2004) Characterization of a conserved structural determinant con-
trolling protein kinase sensitivity to selective inhibitors. Chem Biol 11:691701.
Blume-Jensen P and Hunter T (2001) Oncogenic kinase signalling. Nature 411:355365.
Bono F, De Smet F, Herbert C, De Bock K, Georgiadou M, Fons P, Tjwa M, Alcouffe
C, Ny A, and Bianciotto M et al. (2013) Inhibition of tumor angiogenesis and
growth by a small-molecule multi-FGF receptor blocker with allosteric properties.
Cancer Cell 23:477488.
Boudeau J, Miranda-Saavedra D, Barton GJ, and Alessi DR (2006) Emerging roles of
pseudokinases. Trends Cell Biol 16:443452.
Britschgi A, Andraos R, Brinkhaus H, Klebba I, Romanet V, Müller U, Murakami M,
Radimerski T, and Bentires-Alj M (2012) JAK2/STAT5 inhibition circumvents re-
sistance to PI3K/mTOR blockade: a rationale for cotargeting these pathways in
metastatic breast cancer. Cancer Cell 22:796811.
Busschots K, Lopez-Garcia LA, Lammi C, Stroba A, Zeuzem S, Piiper A, Alzari PM,
Neimanis S, Arencibia JM, and Engel M et al. (2012) Substrate-selective inhibition
of protein kinase PDK1 by small compounds that bind to the PIF-pocket allosteric
docking site. Chem Biol 19:11521163.
Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman
JP, Coleman M, and Wierda WG et al. (2013) Targeting BTK with ibrutinib in
relapsed chronic lymphocytic leukemia. N Engl J Med 369:3242.
Cazorla M, Jouvenceau A, Rose C, Guilloux JP, Pilon C, Dranovsky A, and Prémont J
(2010) Cyclotraxin-B, the first highly potent and selective TrkB inhibitor, has
anxiolytic properties in mice. PLoS ONE 5:e9777.
Chandarlapaty S (2012) Negative feedback and adaptive resistance to the targeted
therapy of cancer. Cancer Discov 2:311319.
Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R,
Garbe C, Testori A, and Maio M et al.; BRIM-3 Study Group (2011) Improved
survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J
Med 364:25072516.
Cheng H and Force T (2010) Why do kinase inhibitors cause cardiotoxicity and what
can be done about it? Prog Cardiovasc Dis 53:114120.
Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW, Jr, and Leahy
DJ (2003) Structure of the extracellular region of HER2 alone and in complex with
the Herceptin Fab. Nature 421:756760.
Christopoulos A, Changeux JP, Catterall WA, Fabbro D, Burris TP, Cidlowski JA,
Olsen RW, Peters JA, Neubig RR, and Pin JP et al. (2014) International union of
basic and clinical pharmacology. XC. Multisite pharmacology: recommendations
for the nomenclature of receptor allosterism and allosteric ligands. Pharmacol Rev
66:918947.
Cohen P (2002) Protein kinasesthe major drug targets of the twenty-first century?
Nat Rev Drug Discov 1:309315.
Comess KM, Sun C, Abad-Zapatero C, Goedken ER, Gum RJ, Borhani DW, Argiriadi
M, Groebe DR, Jia Y, and Clampit JE et al. (2011) Discovery and characterization
of non-ATP site inhibitors of the mitogen activated protein (MAP) kinases. ACS
Chem Biol 6:234244.
Converso A, Hartingh T, Garbaccio RM, Tasber E, Rickert K, Fraley ME, Yan Y,
Kreatsoulas C, Stirdivant S, and Drakas B et al . (2009) De velopment of thio-
quinazolinones, allosteric Chk1 kinase inhibitors. Bioorg Med Chem Lett 19:
12401244.
Cools J, Mentens N, Furet P, Fabbro D, Clark JJ, Griffin JD, Marynen P,
and Gilliland DG (2004) Prediction of resistance to small molecule FLT3 inhibitors:
implications for molecularly targeted therapy of acute leukemia. Cancer Res 64:
63856389.
Cools J, Stover EH, Boulton CL, Gotlib J, Legare RD, Amaral SM, Curley DP, Duclos
N, Rowan R, and Kutok JL et al. (2003) PKC412 overcomes resistance to imatinib
in a murine model of FIP1L1-PDGFRa-induced myeloproliferative disease. Cancer
Cell 3:459469.
Courtney KD, Corcoran RB, and Engelman JA (2010) The PI3K pathway as drug
target in human cancer. J Clin Oncol 28:10751083.
Cowan-Jacob SW (2006) Structural biology of protein tyrosine kinases. Cell Mol Life
Sci 63:26082625.
Cowan-Jacob SW, Jahnke W, and Knapp S (2014) Novel approaches for targeting
kinases: allosteric inhibition, allosteric activation and pseudokinases. Future Med
Chem 6:541561.
Cowan-Jacob SW, Möbitz H, and Fabbro D (2009) Structural biology contributions to
tyrosine kinase drug discovery. Curr Opin Cell Biol 21:280287.
Dalzell MD (2013) Ponatinib pulled off market over safety issues. Manag Care 22:
4243.
Daub H, Specht K, and Ullrich A (2004) Strategies to overcome resistance to targeted
protein kinase inhibitors. Nat Rev Drug Discov 3:10011010.
Engelman JA (2009) Targeting PI3K signalling in cancer: opportunities, challenges
and limitations. Nat Rev Cancer 9:550562.
Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, Maira M,
McNamara K, Perera SA, and Song Y et al. (2008b) Effective use of PI3K and MEK
inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers.
Nat Med 14:13511356.
Engelman JA and Settleman J (2008a) Acquired resistance to tyrosine kinase
inhibitors during cancer therapy. Curr Opin Genet Dev 18:7379.
Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N,
Gale CM, Zhao X, and Christensen J et al. (2007) MET amplification leads to
gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316:
10391043.
Fabbro D, Cowan-Jacob SW, Möbitz H, and Martiny-Baron G (2011) Targeting cancer
with small-molecular-weight kinase inhibitors. Methods Mol Biol 795:134.
Fabbro D, Fendrich G, Guez V, Meyer T, Furet P, Mestan P, Griffin JD, Manley PW,
and Cowan-Jacob SW (2005). Targeted therapy with imatinib: an exception or
a rule?, in Inhibitors of Protein Kinases and Protein Phosphates (Pinna LA and
Cohen PTW eds) vol 167, 361389, Springer, New York.
Fabbro D and García-Echeverría C (2002a) Targeting protein kinases in cancer
therapy. Curr Opin Drug Discov Devel 5:701712.
Fabbro D, Manley PW, Jahnke W, Liebetanz J, Szyttenholm A, Fendrich G, Strauss A,
Zhang J, Gray NS, and Adrian F et al. (2010) Inhibitors of the Abl kinase directed at
either the ATP- or myristate-binding site. Biochim Biophys Acta 1804:454462.
Kinase Inhibitors 773
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
Fabbro D, Ruetz S, Buchdunger E, Cowan-Jacob SW, Fendrich G, Liebetanz J, Mestan
J, OReilly T,Traxler P, and ChaudhuriB et al. (2002b) Protein kinases as targets for
anticancer agents: from inhibitors to useful drugs. Pharmacol Ther 93:7998.
Faivre S, Demetri G, Sargent W, and Raymond E (2007) Molecular basis for sunitinib
efficacy and future clinical development. Nat Rev Drug Discov 6:734745.
Fedorov O, Müller S, and Knapp S (2010) The (un)targeted cancer kinome. Nat Chem
Biol 6:166169.
Felip E, Santarpia M, and Rosell R (2007) Emerging drugs for non-small-cell lung
cancer. Expert Opin Emerg Drugs 12:449460.
Fleishman SJ, Schlessinger J, and Ben-Tal N (2002) A putative molecular-activation
switch in the transmembrane domain of erbB2. Proc Natl Acad Sci USA 99:
1593715940.
Fletcher JA and Rubin BP (2007) KIT mutations in GIST. Curr Opin Genet Dev 17:
37.
Force T, Krause DS, and Van Etten RA (2007) Molecular mechanisms of cardiotoxicity
of tyrosine kinase inhibition. Nat Rev Cancer 7:332344.
Furman RR, Cheng S, Lu P, Setty M, Perez AR, Guo A, Racchumi J, Xu G, Wu H,
and Ma J et al. (2014) Ibrutinib resistance in chronic lymphocytic leukemia. N Engl
JMed370:23522354.
Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, and Sawyers CL
(2001) Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene
mutation or amplification. Science 293:876880.
Gottesman MM (2002) Mechanisms of cancer drug resistance. Annu Rev Med 53:
615627.
Graham SM, JørgensenHG, Allan E, Pearson C, Alcorn MJ, Richmond L, and Holyoake
TL (2002) Primitive, quiescent, Philadelphia-positive stem cells from patients with
chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 99:319325.
Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H,
Teague J, Butler A, and Stevens C et al. (2007) Patterns of somatic mutation in
human cancer genomes. Nature 446:153158.
Hahn WC and Weinberg RA (2002) Modelling the molecular circuitry of cancer. Nat
Rev Cancer 2:331341.
Hall MD, Handley MD, and Gottesman MM (2009) Is resistance useless? Multidrug
resistance and collateral sensitivity. Trends Pharmacol Sci 30:546556.
Hall-Jackson CA, Goedert M, Hedge P, and Cohen P (1999) Effect of SB 203580 on
the activity of c-Raf in vitro and in vivo. Oncogene 18:20472054.
Hanahan D and Weinberg RA (2000) The hallmarks of cancer. Cell 100:5770.
Hanks SK and Hunter T (1995) Protein kinases 6. The eukaryotic protein kinase
superfamily: kinase (catalytic) domain structure and classification. FASEB J 9:
576596.
Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R, Hewitt JF, Zak M,
Peck A, and Orr C et al. (2013) Mechanism of MEK inhibition determines efficacy
in mutant KRAS- versus BRAF-driven cancers. Nature 501:232236.
Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R,
Ludlam MJ, Stokoe D, Glo or SL, and Vigers G et al. ( 2010) RAF inhibitors prime
wild-type RAF to activate the MAPK pathway and enhance growth. Nature 464:
431435.
Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H,
McGreevey LS, Chen CJ, Van den Abbeele AD, and Druker BJ et al. (2003) Kinase
mutations and imatinib response in patients with metastatic gastrointestinal
stromal tumor. J Clin Oncol 21:43424349.
Heinrich T, Grädler U, Böttcher H, Blaukat A, and Shutes A (2010) Allosteric IGF-1R
Inhibitors. ACS Med Chem Lett 1:199203.
Herbert C, Schieborr U, Saxena K, Juraszek J, De Smet F, Alcouffe C, Bianciotto M,
Saladino G, Sibrac D, and Kudlinzki D et al. (2013) Molecular mechanism of
SSR128129E, an extracellularly acting, small-molecule, allosteric inhibitor of FGF
receptor signaling. Cancer Cell 23:489501.
Heymach JV, Nilsson M, Blumenschein G, Papadimitrakopoulou V, and Herbst R
(2006) Epidermal growth factor receptor inhibitors in development for the treat-
ment of non-small cell lung cancer. Clin Cancer Res 12:4441s4445s.
Hill ZB, Perera BG, Andrews SS, and Maly DJ (2012) Targeting diverse signaling
interaction sites allows the rapid generation of bivalent kinase inhibitors. ACS
Chem Biol 7:487495.
Hindie V, Stroba A, Zhang H, Lopez-Garcia LA, Idrissova L, Zeuzem S, Hirschberg D,
Schaeffer F, Jørgensen TJ, and Engel M et al. (2009) Structure and allosteric
effects of low-molecular-weight activators on the protein kinase PDK1. Nat Chem
Biol 5:758764.
Holderfield M, Merritt H, Chan J, Wallroth M, Tandeske L, Zhai H, Tellew J, Hardy S,
Hekmat-Nejad M, and Stuart DD et al. (2013) RAF inhibitors activate the MAPK
pathway by relieving inhibitory autophosphorylation. Cancer Cell 23:594602.
Holderfield M, Nagel TE, and Stuart DD (2014) Mechanism and consequences of RAF
kinase activation by small-molecule inhibitors. Br J Cancer 111:640645.
Hoy SM (2014) Ponatinib: a review of its use in adults with chronic myeloid leu-
kaemia or Philadelphia chromosome-positive acute lymphoblastic leukaemia.
Drugs 74:793806.
Hsieh AC and Moasser MM (2007) Targeting HER proteins in cancer therapy and the
role of the non-target HER3. Br J Cancer 97:453457.
Hunter T (2000) Signaling2000 and beyond. Cell 100:113127.
Hynes NE and Lane HA (2005) ERBB receptors and cancer: the complexity of tar-
geted inhibitors. Nat Rev Cancer 5:341354.
Ihle NT, Williams R, Chow S, Chew W, Berggren MI, Paine-Murrieta G, Minion DJ,
Halter RJ, Wipf P, and Abraham R et al. (2004) Molecular pharmacology and
antitumor activity of PX-866, a novel inhibitor of phosphoinositide-3-kinase sig-
naling. Mol Cancer Ther 3:763772.
Jahnke W, Grotzfeld RM, Pellé X, Strauss A, Fendrich G, Cowan-Jacob SW, Cotesta
S, Fabbro D, Furet P, and Mestan J et al. (2010) Binding or bending: distinction of
allosteric Abl kinase agonists from antagonists by an NMR-based conformational
assay. J Am Chem Soc 132:70437048.
Jänne PA, Gray N, and Settleman J (2009) Factors underlying sensitivity of cancers
to small-molecule kinase inhibitors. Nat Rev Drug Discov 8:709723.
Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, John son LA,
Emery CM, Stransky N, Cog dill AP, and Barretina J et al. (2010) COT drives
resistance to RAF inhi bition thr ough MAP kinase pathway reactivation. Nature
468:968972.
Jura N, Zhang X, Endres NF, Seeliger MA, Schindler T, and Kuriyan J (2011) Cat-
alytic control in the EGF receptor and its connection to general kinase regulatory
mechanisms. Mol Cell 42:922.
Kannan N and Taylor SS (2008) Rethinking pseudokinases. Cell 133:204205.
Katayama R, Shaw AT, Khan TM, Mino-Kenudson M, Solomon BJ, and Halmos B
et al. (2012). Mechanisms of acquired crizotinib resistance in ALK-rearranged lung
cancers. Sci Transl Med 4:120117.
Knapp S, Arruda P, Blagg J, Burley S, Drewry DH, Edwards A, Fabbro D, Gillespie
P, Gray NS, and Kuster B et al. (2013) A public-private partnership to unlock the
untargeted kinome. Nat Chem Biol 9:36.
Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, Johnson BE,
Eck MJ, Tenen DG, and Halmos B (2005) EGFR mutation and resistance of non-
small-cell lung cancer to gefitinib. N Engl J Med 352:786792.
Kornev AP, Haste NM, Taylor SS, and Eyck LF (2006) Surface comparison of active
and inactive protein kinases identifies a conserved activation mechanism. Proc
Natl Acad Sci USA 103:1778317788.
Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW,
Harris PL, Driscoll DR, Fidias P, and Lynch TJ et al. (2005) Irreversible inhibitors
of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl
Acad Sci USA 102:76657670.
Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A,
Amzallag A, Ramaswamy S, and Tesar B et al. (2014) Targeting transcription
regulation in cancer with a covalent CDK7 inhibitor. Nature 511:616620.
le Coutre P, Tassi E, Varella-Garcia M, Barni R, Mologni L, Cabrita G, Marchesi E,
Supino R, and Gambacorti-Passerini C (2000) Induction of resistance to the
Abelson inhibitor STI571 in human leukemic cells through gene amplification.
Blood 95:17581766.
Lee KH, Hsu EC, Guh JH, Yang HC, Wang D, Kulp SK, Shapiro CL, and Chen CS
(2011) Targeting energy metabolic and oncogenic signaling pathways in triple-
negative breast cancer by a novel adenosine monophosphate-activated protein ki-
nase (AMPK) activator. J Biol Chem 286:3924739258.
Levitzki A (2003) Protein kinase inhibitors as a therapeutic modality. Acc Chem Res
36:462469.
Li M, Youngren JF, Manchem VP, Kozlowski M, Zhang BB, Maddux BA,
and Goldfine ID (2001) Small molecule insulin receptor activators potentiate in-
sulin action in insulin-resistant cells. Diabetes 50:23232328.
Lin K, Lin J, Wu W-I, Ballard J, Lee BB, Gloor SL, Vigers GP, Morales TH, Friedman
LS, and Skelton N et al. (2012) An ATP-site on-off switch that restricts phospha-
tase accessibility of Akt. Sci Signal 5:ra37.
Lindsley CW, Zhao Z, Leister WH, Robinson RG, Barnett SF, Defeo-Jones D, Jones
RE, Hartman GD, Huff JR, and Huber HE et al. (2005) Allosteric Akt (PKB)
inhibitors: discovery and SAR of isozyme selective inhibitors. Bioorg Med Chem
Lett 15:761764.
Liu Q, Sabnis Y, Zhao Z, Zhang T, Buhrlage SJ, Jones LH, and Gray NS (2013)
Developing irreversible inhibitors of the protein kinase cysteinome. Chem Biol 20:
146159.
Liu Y and Gray NS (2006) Rational design of inhibitors that bind to inactive kinase
conformations. Nat Chem Biol 2:358364.
Logue JS and Morrison DK (2012) Complexity in the signaling network: insights from
the use of targeted inhibitors in cancer therapy. Genes Dev 26:641650.
Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cor-
nelius LA, Das J, and Doweyko AM et al. (2004) Discovery of N-(2-chloro-6-methyl-
phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)
thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent
antitumor activity in preclinical assays. J Med Chem 47:66586661.
Lopez-Garcia LA, Schulze JO, Fröhner W, Zhang H, Süss E, Weber N, Navratil J,
Amon S, Hindie V, and Zeuzem S et al. (2011) Allosteric regulation of protein
kinase PKCzby the N-terminal C1 domain and small compounds to the PIF-pocket.
Chem Biol 18:14631473.
Luo J, Solimini NL, and Elledge SJ (2009) Principles of cancer therapy: oncogene and
non-oncogene addiction. Cell 136:823837.
Mahon FX, Belloc F, Lagarde V, Chollet C, Moreau-Gaudry F, Reiffers J, Goldman
JM, and Melo JV (2003) MDR1 gene overexpression confers resistance to imatinib
mesylate in leukemia cell line models. Blood 101:23682373.
Manning G, Whyte DB, Martinez R, Hunter T, and Sudarsanam S (2002) The protein
kinase complement of the human genome. Science 298:19121934.
Martiny-Baron G and Fabbro D (2007). Classical PKC isoforms in cancer. Pharmacol
Res 55:477486.
Massa SM, Yang T, Xie Y, Shi J, Bilgen M, Joyce JN, Nehama D, Rajadas J,
and Longo FM (2010) Small molecule BDNF mimetics activate TrkB signaling and
prevent neuronal degeneration in rodents. J Clin Invest 120:17741785.
Medina T, Amaria MN, and Jimeno A (2013) Dabrafenib in the treatment of ad-
vanced melanoma. Drugs Today (Barc) 49:377385.
Mendoza MC, Er EE, and Blenis J (2011) The Ras-ERK and PI3K-mTOR pathways:
cross-talk and compensation. Trends Biochem Sci 36:320328.
Moebitz H and Fabbro D (2012) Conformational bias: a key concept for protein kinase
inhibition. Eur Pharm Rev 17:4151.
Motzer RJ, Hoosen S, Bello CL, and Christensen JG (2006) Sunitinib malate for the
treatment of solid tumours: a review of current clinical data. Expert Opin Investig
Drugs 15:553561.
Noble ME, Endicott JA, and Johnson LN (2004) Protein kinase inhibitors: insights
into drug design from structure. Science 303:18001805.
Nolen B, Taylor S, and Ghosh G (2004) Regulation of protein kinases; controlling
activity through activation segment conformation. Mol Cell 15:661675.
OHare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T,
Huang WS, and Xu Q et al. (2009) AP24534, a pan-BCR-ABL inhibitor for chronic
774 Fabbro
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-
based resistance. Cancer Cell 16:401412.
Ohren JF, Chen H, Pavlovsky A, Whitehead C, Zhang E, Kuffa P, Yan C, McConnell
P, Spessard C, and Banotai C et al. (2004) Structures of human MAP kinase kinase
1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition. Nat Struct
Mol Biol 11:11921197.
Okuzumi T, Fiedler D, Zhang C, Gray DC, Aizenstein B, Hoffman R, and Shokat KM
(2009) Inhibitor hijacking of Akt activation. Nat Chem Biol 5:484493.
Olaharski AJ, Gonzaludo N, Bitter H, Goldstein D, Kirchner S, Uppal H, and Kolaja
K (2009) Identification of a kinase profile that predicts chromosome damage in-
duced by small molecule kinase inhibitors. PLOS Comput Biol 5:e1000446.
Over B, Wetzel S, Grütter C, Nakai Y, Renner S, Rauh D, and Waldmann H (2013)
Natural-product-derived fragments for fragment-based ligand discovery. Nat Chem
5:2128.
Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, Kris MG,
and Varmus H (2005) Acquired resistance of lung adenocarcinomas to gefitinib or
erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS
Med 2:e73.
Pender C, Goldfine ID, Manchem VP, Evans JL, Spevak WR, Shi S, Rao S, Bajjalieh
S, Maddux BA, and Youngren JF (2002) Regulation of insulin receptor function by
a small molecule insulin receptor activator. J Biol Chem 277:4356543571.
Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C, Moriceau G, Shi H, Atefi
M, Titz B, and Gabay MT et al. (2011) RAF inhibitor resistance is mediated by
dimerization of aberrantly spliced BRAF(V600E). Nature 480:387390.
Poulikakos PI, Zhang C, Bollag G, Shokat KM, and Rosen N (2010) RAF inhibitors
transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature
464:427430.
Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen
RL, Bardelli A, and Bernards R (2012) Unresponsiveness of colon cancer to BRAF
(V600E) inhibition through feedback activation of EGFR. Nature 483:100103.
Quintás-Cardama A, Kantarjian H, and Cortes J (2007) Flying under the radar: the
new wave of BCR-ABL inhibitors. Nat Rev Drug Discov 6:834848.
Rauch J, Volinsky N, Romano D, and Kolch W (2011) The secret life of kinases:
functions beyond catalysis. Cell Commun Signal 9:23.
Robert C, Soria JC, Spatz A, Le Cesne A, Malka D, Pautier P, Wechsler J, Lhomme C,
Escudier B, and Boige V et al. (2005) Cutaneous side-effects of kinase inhibitors
and blocking antibodies. Lancet Oncol 6:491500.
Rodon J, Dienstmann R, Serra V, and Tabernero J (2013) Development of PI3K
inhibitors: lessons learned from early clinical trials. Nat Rev Cli n Oncol 10:
143153.
Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI, Scaltriti M,
Moskatel E, Baselga J, Guichard S, and Rosen N (2011) mTOR kinase inhibition
causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov 1:
248259.
Rubin BP and Duensing A (2006) Mechanisms of resistance to small molecule kinase
inhibition in the treatment of solid tumors. Lab Invest 86:981986.
Sadowsky JD, Burlingame MA, Wolan DW, McClendon CL, Jacobson MP, and Wells
JA (2011) Turning a protein kinase on or off from a single allosteric site via
disulfide trapping. Proc Natl Acad Sci USA 108:60566061.
Salt IP and Palmer TM (2012) Exploiting the anti-inflammatory effects of AMP-
activated protein kinase activation. Expert Opin Investig Drugs 21:11551167.
Sanders MJ, Ali ZS, Hegarty BD, Heath R, Snowden MA, and Carling D (2007)
Defining the mechanism of activation of AMP-activated protein kinase by the small
molecule A-769662, a member of the thienopyridone family. J Biol Chem 282:
3253932548.
Sawyers C (2004) Targeted cancer therapy. Nature 432:294297.
Serra V, Scaltriti M, Prudkin L, Eichhorn PJA, Ibrahim YH, Chandarlapaty S,
Markman B, Rodriguez O, Guzman M, and Rodriguez S et al. (2011) PI3K in-
hibition results in enhanced HER signaling and acquired ERK dependency in
HER2-overexpressing breast cancer. Oncogene 30:25472557.
Sharma SV and Settleman J (2010) Exploiting the balance between life and death:
targeted cancer therapy and oncogenic shock.Biochem Pharmacol 80:666673.
She QB, Solit D, Basso A, and Moasser MM (2003) Resistance to gefitinib in PTEN-
null HER-overexpressing tumor cells can be overcome through restoration of
PTEN function or pharmacologic modulation of constitutive phosphatidylinositol
39-kinase/Akt pathway signaling. Clin Cancer Res 9:43404346.
Siegel R, Naishadham D, and Jemal A (2012) Cancer statistics for Hispanics/Latinos,
2012. CA Cancer J Clin 62:283298.
Skora L, Mestan J, Fabbro D, Jahnke W, and Grzesiek S (2013) NMR reveals the
allosteric opening and closing of Abelson tyrosine kinase by ATP-site and myristoyl
pocket inhibitors. Proc Natl Acad Sci USA 110:E4437E4445.
Su F, Viros A, Milagre C, Trunzer K, Bollag G, Spleiss O, Reis-Filho JS, Kong X,
Koya RC, and Flaherty KT et al. (2012) RAS mutations in cutaneous squamous-cell
carcinomas in patients treated with BRAF inhibitors. NEnglJMed366:207215.
Szakács G, Paterson JK, Ludwig JA, Booth-Genthe C, and Gottesman MM (2006)
Targeting multidrug resistance in cancer. Nat Rev Drug Discov 5:219234.
Takano T, Ohe Y, Sakamoto H, Tsuta K, Matsuno Y, Tateishi U, Yamamoto S,
Nokihara H, Yamamoto N, and Sekine I et al. (2005) Epidermal growth factor
receptor gene mutations and increased copy numbers predict gefitinib sensitivity
in patients with recurrent non-small-cell lung cancer. J Clin Oncol 23:68296837.
Tao ZF, Wang L, Stewart KD, Chen Z, Gu W, Bui MH, Merta P, Zhang H, Kovar P,
and Johnson E et al. (2007) Structure-based design, synthesis, and biological
evaluation of potent and selective macrocyclic checkpoint kinase 1 inhibitors.
JMedChem50:15141527.
Taylor SS and Kornev AP (2011) Protein kinases: evolution of dynamic regulatory
proteins. Trends Biochem Sci 36:6577.
Thomas RK, Baker AC, Debiasi RM, Winckler W, Laframboise T, Lin WM, Wang M,
Feng W, Zander T, and MacConaill L et al. (2007) High-throughput oncogene
mutation profiling in human cancer. Nat Genet 39:347351.
Tomita N, Hayashi Y, Suzuki S, Oomori Y, Aramaki Y, Matsushita Y, Iwatani M,
Iwata H, Okabe A, and Awazu Y et al. (2013) Structure-based discovery of cellular-
active allosteric inhibitors of FAK. Bioorg Med Chem Lett 23:17791785.
Trusolino L and Bertotti A (2012) Compensatory pathways in oncogenic kinase sig-
naling and resistance to targeted therapies: six degrees of separation. Cancer
Discov 2:876880.
Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L,
Rogers A, Mok T, and Sequist L et al. (2010) Preexistence and clonal selection of
MET amplification in EGFR mutant NSCLC. Cancer Cell 17:7788.
Udugamasooriya DG, Dineen SP, Brekken RA, and Kodadek T (2008) A peptoid
antibody surrogatethat antagonizes VEGF receptor 2 activity. J Am Chem Soc
130:57445752.
Ventura JJ and Nebreda AR (2006) Protein kinases and phosphatases as therapeutic
targets in cancer. Clin Transl Oncol 8:153160.
Vieth M, Higgs RE, Robertson DH, Shapiro M, Gragg EA, and Hemmerle H (2004)
Kinomics-structural biology and chemogenomics of kinase inhibitors and targets.
Biochim Biophys Acta 1697:243257.
Vieth M, Sutherland JJ, Robertson DH, and Campbell RM (2005) Kinomics: charac-
terizing the therapeutically validated kinase space. Drug Discov Today 10:839846.
Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Jr, and Kinzler KW
(2013) Cancer genome landscapes. Science 339:15461558.
Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, Kehoe SM,
Johannessen CM, Macconaill LE, and Hahn WC et al. (2011) Dissecting therapeutic
resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol 29:
30853096.
Wang X and Sun SY (2009) Enhancing mTOR-targeted cancer therapy. Expert Opin
Ther Targets 13:11931203.
Weinstein IB (2002) Cancer. Addiction to oncogenesthe Achilles heal of cancer.
Science 297:6364.
Weisberg E, Manley PW, Breitenstein W, Brüggen J, Cowan-Jacob SW, Ray A,
Huntly B, Fabbro D, Fendrich G, and Hall-Meyers E et al. (2005) Characterization
of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 7:
129141.
Wiestner A (2013) Targeting B-cell receptor signaling for anticancer therapy: the
Brutons tyrosine kinase inhibitor ibrutinib induces impressive responses in B-cell
malignancies. J Clin Oncol 31:128130.
Wolf-Yadlin A, Kumar N, Zhang Y, Hautaniemi S, Zaman M, Kim HD, Grantcharova
V, Lauffenburger DA, and White FM (2006) Effects of HER2 overexpression on cell
signaling networks governing proliferation and migration. Mol Syst Biol 2:54.
Wong KK, Engelman JA, and Cantley LC (2010) Targeting the PI3K signaling
pathway in cancer. Curr Opin Genet Dev 20:8790.
Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, Ellis B,
Pennisi C, Horne E, and Lackey K et al. (2004) A unique structure for epidermal
growth factor receptor bound to GW572016 (Lapatinib): relationships among pro-
tein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer
Res 64:66526659.
Workman P, Al-Lazikani B, and Clarke PA (2013) Genome-based cancer therapeu-
tics: targets, kinase drug resistance and future strategies for precision oncology.
Curr Opin Pharmacol 13:486496.
Wymann MP, Bulgarelli-Leva G, Zvelebil MJ, Pirola L, Vanhaesebroeck B, Waterfield
MD, and Panayotou G (1996) Wortmannin inactivates phosphoinositide 3-kinase by
covalent modification of Lys-802, a residue involved in the phosphate transfer re-
action. Mol Cell Biol 16:17221733.
Yang H, Rudge DG, Koos JD, Vaidialingam B, Yang HJ, and Pavletich NP (2013)
mTOR kinase structure, mechanism and regulation. Nature 497:217223.
Yang J, Campobasso N, Biju MP, Fisher K, Pan XQ, Cottom J, Galbraith S, Ho T,
Zhang H, and Hong X et al. (2011) Discovery and characterization of a cell-permeable,
small-molecule c-Abl kinase activator that binds to the myristoyl binding site. Chem
Biol 18:177186.
Yarden Y and Pines G (2012) The ERBB network: at last, cancer therapy meets
systems biology. Nat Rev Cancer 12:553563.
Zhang J, Adrián FJ, Jahnke W, Cowan-Jacob SW, Li AG, Iacob RE, Sim T, Powers J,
Dierks C, and Sun F et al. (2010) Targeting Bcr-Abl by combining allosteric with
ATP-binding-site inhibitors. Nature 463:501506.
Zhang J, Yang PL, and Gray NS (2009) Targeting cancer with small molecule kinase
inhibitors. Nat Rev Cancer 9:2839.
Zhou W, Ercan D, Chen L, Yun CH, Li D, Capelletti M, Cortot AB, Chirieac L, Iacob
RE, and Padera R et al. (2009) Novel mutant-selective EGFR kinase inhibitors
against EGFR T790M. Nature 462:10701074.
Zhou W, Ercan D, Jänne PA, and Gray NS (2011) Discovery of selective irreversible
inhibitors for EGFR-T790M. Bioorg Med Chem Lett 21:638643.
Zuccotto F, Ardini E, Casale E, and Angiolini M (2010) Through the gatekeeper
door: exploiting the active kinase conformation. J Med Chem 53:26812694.
Address correspondence to: Doriano Fabbro, Ph.D., PIQUR Therapeutics
AG, Hochbergerstrasse 60C, CH-4057 Basel, Switzerland. E-mail: doriano.
fabbro@piqur.com
Kinase Inhibitors 775
at ASPET Journals on April 17, 2015molpharm.aspetjournals.orgDownloaded from
... Les PKI déjà sur le marché sont principalement dérivés de pyridine, pyrimidine, quinolone, quinazoline ou carboxamide 35,251 . Ils sont, le plus souvent, compétitifs de l'ATP (types I, I 1/2 et II) et moins efficaces lors de l'apparition de mutations amenant des résistances à ces traitements 261 . C'est par exemple le cas de la protéine kinase de fusion ciblée par l'imatinib (Breakpoint Cluster Region-Abelson, BCR-ABL1) pouvant muter sur son acide aminé gatekeeper. ...
... The starting point to define the human kinome was the original paper of manning (Manning et al., 2002) updated by the pkinfam UniProt -Swiss-Prot Protein Knowledgebase at https://www.uniprot.org/docs/pkinfam.txt. Additional annotations about motif specific to kinase family (DFG, hinge, G-Loop ect …) were extracted from literature (Fabbro et al., 2015) and converted to GFF format using a multiple sequence alignment of 490 kinase domain from möbitz paper (Möbitz, 2015). Then each data (e.g. ...
... Finally, after identification, biological knowledge is transferred in PyMOL 3D structure visualizer. We mapped mutations specific kinase family motifs (DFG, hinge, G-Loop etc …) (Fabbro et al., 2015) as well as natural variants from Uniprot. Then, each kinase chain is superposed on the same coordinate space using one of the first PDB entry of kinase as reference: PRKCA (PDB code 1ATP). ...
Thesis
Les protéines kinases sont une famille de protéines ayant des rôles clés dans la régulation de fonctions cellulaires. Ce sont des cibles thérapeutiques majeures étudiées principalement en oncologie et plus récemment dans d’autres pathologies. Cette famille de protéines est composée de plus de 500 membres qui présentent une grande similarité tridimensionnelle entre eux, ce qui complique leur inhibition spécifique. Les macrocycles sont une classe de molécules qui par leur rigidité peuvent apporter cette sélectivité.Frags2Drugs est un outil in silico qui a été récemment développé afin de concevoir des inhibiteurs de protéines kinases à partir de fragments moléculaires, composés organiques de faible poids moléculaire. Au cours de cette thèse, cet outil a été analysé et amélioré pour l’appliquer dans quatre projets de recherche d’inhibiteurs de protéines kinases. Ainsi, en collaboration avec des équipes de chimistes et biologistes, de nouveaux inhibiteurs et macrocycles ont été conçus sur des cibles impliquées en oncologie. Pour cela, Frags2drugs a été combiné à d’autres méthodes pour la sélection des meilleurs inhibiteurs. Plusieurs molécules et macrocycles obtenus sont en cours de synthèse et seront ensuite évalués biologiquement. Un site internet (http://frags2drugs.icoa.fr) a été développé pour rendre accessible l’outil Frags2drugs à la communauté scientifique. Les travaux de thèse ont aussi consisté à mettre en place deux autres outils, MolDesc (http://moldesc.icoa.fr) pour le calcul de descripteurs moléculaires et KinoMine (http://kinomine.icoa.fr) pour l’exploration du kinome humain.
... The majority of these small molecules are quinazoline-based substances, and numerous attempts have been undertaken to make changes in their structure in order to create more effective anticancer drugs, thus, the search for novel scaffolds that can function as tyrosine kinase inhibitors is still ongoing today [13][14][15][16]. ...
... The evaluated synthesized compounds showed a variable degree of inhibition, with an IC 50 range of 0.1165 to 1.8364 µM. The most active derivatives that shown good inhibition against VEGFR-2 were found to be derivatives 10d, 15 an IC 50 of 3.07 µM. ...
... The positions of these motifs (termed IN or OUT) correspond to active and inactive kinase conformations, respectively. ATPcompetitive RAF inhibitors can be classified based on their preferential binding to different (IN or OUT) conformations of the αC-helix and the DFG motif [38][39][40]. At present, three types of RAF inhibitors, which preferably bind to (1) [41], yet with much larger K d 's. ...
Article
Full-text available
Citation: Imoto, H.; Rauch, N.; Neve, A.J.; Khorsand, F.; Kreileder, M.; Alexopoulos, L.G.; Rauch, J.; Okada, M.; Kholodenko, B.N.; Rukhlenko, O.S. A Combination of Conformation-Specific RAF Inhibitors Overcome Drug Resistance Brought about by RAF Overexpression. Biomolecules 2023, 13, 1212. https://doi. Abstract: Cancer cells often adapt to targeted therapies, yet the molecular mechanisms underlying adaptive resistance remain only partially understood. Here, we explore a mechanism of RAS/RAF/MEK/ERK (MAPK) pathway reactivation through the upregulation of RAF isoform (RAFs) abundance. Using computational modeling and in vitro experiments, we show that the upreg-ulation of RAFs changes the concentration range of paradoxical pathway activation upon treatment with conformation-specific RAF inhibitors. Additionally, our data indicate that the signaling output upon loss or downregulation of one RAF isoform can be compensated by overexpression of other RAF isoforms. We furthermore demonstrate that, while single RAF inhibitors cannot efficiently inhibit ERK reactivation caused by RAF overexpression, a combination of two structurally distinct RAF inhibitors synergizes to robustly suppress pathway reactivation.
... [16][17][18] One of the ongoing challenges nowadays would be the quest for new scaffolds, able to act as tyrosine kinase inhibitors. [19][20][21][22] Interestingly, there are many FDA-approved agents that target VEGFRs (Sorafenib, Sunitinib, Vandetanib, Axitinib, Regorafenib, etc.). However, drug resistance is a major concern associated with the monotherapy of these agents. ...
Article
Full-text available
Since VEGFR-2 plays a crucial role in tumor growth, angiogenesis, and metastasis, it is a prospective target for cancer treatment. In this work, a series of 3-phenyl-4-(2-substituted phenylhydrazono)-1H-pyrazol-5(4H)-ones (3a-l) were synthesized and investigated for their cytotoxicity against the PC-3 human cancer cell line compared to Doxorubicin and Sorafenib as reference drugs. Two compounds 3a and 3i showed comparable cytotoxic activity with IC 50 values of 1.22 and 1.24 mM compared to the reference drugs (IC 50 = 0.932, 1.13 mM). Compound 3i was found to be the most effective VEGFR-2 inhibitor using in vitro testing of the synthesized compounds, with nearly 3-fold higher activity than Sorafenib (30 nM), with IC 50 8.93 nM. Compound 3i significantly stimulated total apoptotic prostate cancer cell death 55.2-fold (34.26% compared to 0.62% for the control) arresting the cell cycle at the S-phase. The genes involved in apoptosis were also impacted, with proapoptotic genes being upregulated and antiapoptotic Bcl-2 being downregulated. These results were supported by docking studies of these two compounds within the active site of the VEGFR2 enzyme. Finally, in vivo, the study revealed the potentiality of compound 3i to inhibit tumor proliferation by 49.8% reducing the tumor weight from 234.6 mg in untreated mice to 83.2 mg. Therefore, 3i could be a promising anti-prostate cancer agent.
... This principle has been successfully exploited in the discovery of small molecule kinase modulators. [14][15][16] Conformational transitions between kinase states are orchestrated by three conserved structural motifs in the catalytic domain: the αC-helix, the DFG-Asp motif (DFG-Asp in, active; DFG-Asp out, inactive), and the activation loop (A-loop open, active; A-loop closed, inactive). The conserved His-Arg-Asp (HRD) motif in the catalytic loop and the DFG motif are coupled with the αC-helix to form conserved intramolecular networks termed regulatory spine (R-spine) and catalytic spine (C-spine). ...
Article
Full-text available
In the current study, multiscale simulation approaches and dynamic network methods are employed to examine the dynamic and energetic details of conformational landscapes and allosteric interactions in the ABL kinase domain that determine the kinase functions. Using a plethora of synergistic computational approaches, we elucidate how conformational transitions between the active and inactive ABL states can employ allosteric regulatory switches to modulate the intramolecular communication networks between the ATP site, the substrate binding region, and the allosteric binding pocket. A perturbation-based network approach that implements mutational profiling of allosteric residue propensities and communications in the ABL states is proposed. Consistent with the biophysical experiments, the results reveal functionally significant shifts of the allosteric interaction networks in which preferential communication paths between the ATP binding site and substrate regions in the active ABL state become suppressed in the closed inactive ABL form, which in turn features favorable allosteric couplings between the ATP site and the allosteric binding pocket. By integrating the results of atomistic simulations with dimensionality reduction methods and Markov state models we analyze the mechanistic role of the macrostates and characterize kinetic transitions between the ABL conformational states. Using network-based mutational scanning of allosteric residue propensities, this study provides a comprehensive computational analysis of the long-range communications in the ABL kinase domain and identifies conserved regulatory hotspots that modulate kinase activity and allosteric cross-talk between the allosteric pocket, ATP binding site and substrate binding regions.
... As a result, they have been suggested as powerful targets for the treatment of MetS. 7,9,10 DYRK1B participates in differentiation and undergoes different splicing during fat formation. 11 Moreover, DYRK1B is overexpressed and mutated in cancers. ...
Article
Full-text available
Background: Metabolic syndrome (MetS) is a group of signs and symptoms that are associated with a higher risk of type 2 diabetes mellitus and cardiovascular diseases. The major risk factor for developing MetS is abdominal obesity, which is caused by an increase in adipocyte size or quantity. Increased adipocyte quantity is a result of differentiation of stem cells into adipose tissue. Numerous studies have investigated the expression of key transcription factors, including PPARG and CEBPB during adipocyte differentiation in murine cells such as 3T3-L1 cell lines. To better understand the expression changes during the process of fat accumulation in adipose-derived stem cells (ASCs), we compared the expression of DYRK1B, PPARG, and ẟB in ASCs between the patient (harboring DYRK1B R102C) and control (healthy individuals) groups. Methods: Gene expression was evaluated on the eighth day before induction and days 1, 5, and 15 postinduction. The pluripotent capacity of ASCs and the potential for differentiation into adipocytes were confirmed by flow cytometry analysis of surface markers (CD34, CD44, CD105, and CD90), and Oil Red O staining, respectively. The Expression of DYRK1B, PPARG, and CEBPB were assessed by real-time-polymerase chain reaction in patients and normal individuals. The effects of AZ191, a potent small molecule inhibitor on DYRK1B and CEBPB expression in patients' samples were studied. Result: The expression of DYRK1B kinase and transcription factors (CEBPB and PPARG) are higher in ASCs harboring DYRK1B R102C compared with noncarriers on days 5 and 15 during adipocyte differentiation. These proteins may be helpful to elucidate the mechanisms underlying obesity and obesity-related disorders like MetS. Furthermore, the new compound AZ191 exhibited inhibitory activity toward DYRK1B and CEBPB. We suggest that AZ191 may be helpful in defining the potential roles of DYRK1B and CEBPB in adipogenesis.
... These drugs are absorbed in the stomach and small intestine and metabolised mainly in the liver by the cytochrome P450 system [2]. Jakinibs have been approved for the treatment of several immune-mediated inflammatory diseases (IMIDs), including RA, psoriasis (and hence psoriatic arthritis, PsA), and inflammatory bowel disease (IBD), with variations across countries [4,5]. Four jakinibs are currently approved in Spain: tofacitinib, baricitinib, upadacitinib and filgotinib [6]. ...
Article
Full-text available
Nurses’s support of patients needs an evidence base as much as that of specialists management. However, some more practical aspects need specific questions that are not addressed in medical societies’ recommendations. Our objective was to investigate the effect of Janus kinase inhibitors (jakinibs) on efficacy, safety, infections, cardiovascular risk, vaccination, pregnancy and lactation, interactions, surgery, and switch in adult patients with rheumatic diseases. We used the methodology for rapid reviews. Medline was searched for systematic reviews of randomised clinical trials and longitudinal observational studies reporting on the target aspects, without limits, yielding 540 titles, of which 70 articles were selected for detailed reading after the screening of title and abstract. In the case of no systematic review being published on a specific question, we resorted to the information provided by primary studies. The efficacy and safety profiles are similar to that of TNF-inhibitors to which they are compared in most studies; however, there is an increased risk of herpes zoster infections with jakinibs. The evidence on pregnancy, surgery and switches between jakinibs is very limited, although, so far, there are no major issues to inform patients about or to implement specific measures. In general, evidence to support nursing management in patients being treated with jakinibs is of moderate quality and scarce, ought to the recent incursion of jakinibs as a treatment.
Article
The development of peptide drugs has made tremendous progress in the past few decades because of the advancements in modification chemistry and analytical technologies. The novel‐designed peptide drugs have been modified through various biochemical methods with improved diagnostic, therapeutic, and drug‐delivery strategies. Researchers found it a helping hand to overcome the inherent limitations of peptides and bring continued advancements in their applications. Furthermore, the emergence of peptide‐drug conjugates (PDCs)—utilizes target‐oriented peptide moieties as a vehicle for cytotoxic payloads via conjugation with cleavable chemical agents, resulting in the key foundation of the new era of targeted peptide drugs. This review summarizes the various classifications of peptide drugs, suitable chemical modification strategies to improve the ADME (adsorption, distribution, metabolism, and excretion) features of peptide drugs, and recent (2015–early 2024) progress/achievements in peptide‐based drug delivery systems as well as their fruitful implication in preclinical and clinical studies. Furthermore, we also summarized the brief description of other types of PDCs, including peptide‐MOF conjugates and peptide‐UCNP conjugates. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development and progress toward a bright future of novel peptide drugs.
Article
There is a need for non-hormonal contraceptives. One area that needs further investigation is the development of male contraceptives. Comparatively little is understood about potential drug targets in men to achieve a reversible contraceptive effect. In this article, we review the need for male contraceptives and some thoughts around the characteristics of a male contraceptive and the potential development pathway. We then discuss different potential approaches to discovering male contraceptives and then highlight potential targets that have been discussed in the literature.
Article
Full-text available
Tumour oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacological inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymatic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here we present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukaemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide analysis in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumour cells. Pharmacological modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumour types that are dependent on transcription for maintenance of the oncogenic state.
Article
Full-text available
Allosteric interactions play vital roles in metabolic processes and signal transduction and, more recently, have become the focus of numerous pharmacological studies because of the potential for discovering more target-selective chemical probes and therapeutic agents. In addition to classic early studies on enzymes, there are now examples of small molecule allosteric modulators for all superfamilies of receptors encoded by the genome, including ligand- and voltage-gated ion channels, G protein-coupled receptors, nuclear hormone receptors, and receptor tyrosine kinases. As a consequence, a vast array of pharmacologic behaviors has been ascribed to allosteric ligands that can vary in a target-, ligand-, and cell-/tissue-dependent manner. The current article presents an overview of allostery as applied to receptor families and approaches for detecting and validating allosteric interactions and gives recommendations for the nomenclature of allosteric ligands and their properties.
Article
Full-text available
To the Editor: Ibrutinib, an inhibitor that binds covalently to C481 of Bruton's tyrosine kinase (BTK), has produced remarkable responses in patients with relapsed and refractory chronic lymphocytic leukemia (CLL).(1)-(4) However, 5.3% of patients have disease progression, and the mechanism of resistance is largely unknown. Herein we describe the mechanism of resistance in such a case. A 49-year-old woman had a diagnosis of CLL established in 2000. After the failure of multiple treatments, she began receiving ibrutinib at a dose of 560 mg daily in 2010 as part of a phase 1, dose-escalation study of ibrutinib in B-cell cancers. . . .
Article
ALK fusion oncogenes represent a novel molecular target in a small subset of non-small cell lung cancers (NSCLC). The vast majority of patients with ALK-positive NSCLC are highly responsive to ALK tyrosine kinase inhibitor (TKI) therapy. However, these patients invariably relapse, typically within one year, due to the development of resistance. Herein, we report findings from the first series of patients with acquired resistance to the ALK TKI crizotinib. In approximately one-third of patients, we identified a diverse array of secondary mutations distributed throughout the ALK TK domain. We also identified aberrant activation of other kinases as alternative mechanisms of crizotinib resistance. Additionally, we generated laboratory models of acquired crizotinib resistance which closely recapitulate the diversity of resistance mechanisms observed in patients. Our results highlight the marked heterogeneity of TKI resistance mechanisms in ALK-positive NSCLC, and provide the rationale for pursuing combinatorial therapeutic strategies in patients who relapse on crizotinib. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5593. doi:1538-7445.AM2012-5593
Article
In clinical trials, the tyrosine kinase inhibitor STI571 has proven highly effective in reducing leukemic cell burden in chronic myeloid leukemia (CML). The overall sensitivity of CML CD34+ progenitor cells to STI571 and the degree to which cell death was dependent on cell cycle status were determined. Stem cells (Lin−CD34+) from the peripheral blood of patients with CML in chronic phase and from granulocyte–colony-stimulating factor–mobilized healthy donors were labeled with carboxy-fluorescein diacetate succinimidyl diester dye to enable high-resolution tracking of cell division. Then they were cultured for 3 days with and without growth factors ± STI571. After culture, the cells were separated by fluorescence-activated cell sorting into populations of viable quiescent versus cycling cells for genotyping. For healthy controls, in the presence of growth factors, STI571 affected neither cell cycle kinetics nor recovery of viable cells. In the absence of growth factors, normal cells were unable to divide. For CML samples, in the presence or absence of growth factors, the response to STI571 was variable. In the most sensitive cases, STI571 killed almost all dividing cells; however, a significant population of viable CD34+ cells was recovered in the undivided peak and confirmed to be part of the leukemic clone. STI571 also appeared to exhibit antiproliferative activity on the quiescent population. These studies confirm that CML stem cells remain viable in a quiescent state even in the presence of growth factors and STI571. Despite dramatic short-term responses in vivo, such in vitro insensitivity to STI571, in combination with its demonstrated antiproliferative activity, could translate into disease relapse after prolonged therapy.