You are on page 1of 32

Perspective

pubs.acs.org/jmc

Ion Channels as Therapeutic Targets: A Drug Discovery Perspective


Sharan K. Bagal,† Alan D. Brown,† Peter J. Cox,‡ Kiyoyuki Omoto,† Robert M. Owen,† David C. Pryde,*,†
Benjamin Sidders,‡ Sarah E. Skerratt,† Edward B. Stevens,‡ R. Ian Storer,† and Nigel A. Swain†

Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park, Great Abington, Cambridge, CB21 6GS,
U.K.

Pfizer Neusentis, The Portway Building, Granta Park, Great Abington, Cambridge, CB21 6GS, U.K.

ABSTRACT: Ion channels are membrane proteins expressed in almost all living
cells. The sequencing of the human genome has identified more than 400 putative
ion channels, but only a fraction of these have been cloned and functionally tested.
The widespread tissue distribution of ion channels, coupled with the plethora of
physiological consequences of their opening and closing, makes ion-channel-
targeted drug discovery highly compelling. However, despite some important
drugs in clinical use today, as a class, ion channels remain underexploited in drug
discovery and many existing drugs are poorly selective with significant toxicities or
suboptimal efficacy. This Perspective seeks to review the ion channel family, its
structural and functional features, and the diseases that are known to be
modulated by members of the family. In particular, we will explore the structure
and properties of known ligands and consider the future prospects for drug
discovery in this challenging but high potential area.

■ INTRODUCTION
Ion Channels as Therapeutic Targets. Ion channels are
benzodiazepine diazepam (5), which binds to the γ-aminobutyric
acid class A (GABAA) channel (Figure 1).
transmembrane proteins that create a gated, water-filled pore to
help establish and control voltage potential across cell
membranes through control of the active flow of ions between
the intracellular and the extracellular environments.1,2 The ion
channel family is intimately involved in almost all aspects of
physiology and plays a critical role in diverse processes such as
nerve and muscle relaxation, cognition, sensory transduction,
regulation of blood pressure, and cell proliferation. Their
modulation has been linked to a wide range of diseases that
include cardiac disorders, neurological indications, kidney failure,
the perception of pain, and blindness. More than 60
“channelopathies” have been identified as human diseases that
are brought about through mutations in ion channels. Given the
central functional role that the ion channel superfamily plays in Figure 1. Example ion channel ligands.
human physiology, its membrane localization, and the diverse
tissue distribution of different members of the family, it
represents an attractive potential target class for drug discovery. An examination of the ion channel-based research landscape
The human genome comprises several hundred genes that reveals many opportunities to exploit the family in a drug
encode for pore-forming ion channels within plasma membranes, discovery context, not least of all due to the surprisingly large
broadly classified as either voltage or ligand gated depending on number of family members for which there have been no ligands
the primary factors that lead to channel opening and closing. identified to date. For a similarly large number of other family
Members of both subfamilies are known to be modulated by a members, some ligands have been found that are weak and/or
range of small molecule agents. For example, the low molecular nonselective, thereby limiting their utility as pharmacological
weight local anesthetics lidocaine (1) and bupivacaine (2) act on tools or as drug discovery starting points.
voltage-gated sodium channels (Nav). The transient receptor This Perspective is intended to offer a broad overview of the
potential (TRP) family recognizes a diverse array of ligand ion channel drug discovery landscape and introduce the reader to
structural types from pungent natural substances like capsaicin the members of the family, their classification, and the range of
(3) (TRP vanilloid 1 channel, TRPV1) and menthol (4) (TRP
melastatin 8 channel, TRPM8). Several well-known pharma- Received: August 3, 2012
ceutical agents act at certain ligand-gated ion channels such as the Published: November 2, 2012

© 2012 American Chemical Society 593 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

diseases that are reported to originate in channel dysfunction. We stimuli. Further intracellular and extracellular modules provide
review the structural features that underpin ion channel function binding sites for ligands and accessory proteins, adding further
and how ion channels are modulated by ligands. Finally we complexity to the system. Ion channels freely and rapidly sample
discuss the recent advances in ligand identification, trends in a range of states from the open to the closed states on a rapid time
their molecular properties, and the prospects for the drug scale, and the modulation of channel function by a ligand or
discovery industry being able to translate interesting ion channel external trigger manifests in a stabilization of the channel
targets into the next generation of drug therapies. structure in one particular state.
The scope of this article is, for the most part, limited to the Ion flux through the pore when in the open state is an
approximately 200 major pore-forming human ion channels incredibly fast process driven by an electrochemical gradient.
according to the International Union of Basic and Clinical Flux through the channel can be as fast as some 108 ions per
Pharmacology (IUPHAR) classification3 and does not include, second,2 leaving in the order of nanoseconds for the channel to
for example, the anion channels or other transport proteins such interact with each ion, thereby approaching the theoretical limits
as the connexins or aquaporins. The interested reader is referred described by Ohm’s law and Fick’s law. Despite the brevity of this
to several online curated databases of the entire ion channel contact time, potassium channels allow potassium ions to pass
family, for example, the Guide to Pharmacology database,4 for with a fidelity of approximately 10000:1 versus the smaller
details on these other channels. In some cases, the modulation of sodium ion6 through a careful spatial arrangement of carbonyl
regulatory subunits of channel proteins is described where their oxygen atoms within the selectivity filter that can accommodate
inclusion offers the reader a useful insight into alternative means two potassium ions in the pore at any one time.7,8 These contacts
of altering channel function beyond direct targeting of the pore- compensate for the energy penalty of desolvating ions as they
forming domains. enter the filter by providing favorable compensatory interactions
How Ion Channels Function. In the most basic terms, ion with the ion. Depending on the identity of the ion, this energetic
channels are a collection of protein domains that together create compensation varies relative to the energy of desolvation,
a water-filled pore to allow the passage of ions through a cell resulting in an energy-driven selectivity.9 From the perspective of
membrane in response to chemical stimuli, temperature changes, a single channel recording, the trace shown in Figure 3 depicts
or mechanical forces.5 These pore-forming domains vary
enormously in sequence and topology but are typically
composed of four or five helices that fit together to form a
barrel-like structure. The helices are made up of repeat domains
of a single subunit or of several different subunits. In order to
fulfill the function of selectively filtering ions through a biological
membrane, most ion channels also possess a pore-loop region
called the ion or selectivity filter that regulates which ions are
permitted through the pore (Figure 2). The channel features a
gating mechanism that controls the opening and closure of the
pore through conformational changes to the channel protein,
combined with a sensor mechanism that detects and responds to
Figure 3. An example single channel recording of an ion channel that
cycles between the open and closed state in response to a current gating
trigger applied repeatedly.

the gating of a single ion channel that starts at a resting potential


in the closed state, and at the time points indicated by the arrows,
a change in membrane potential creates a series of conforma-
tional changes in the protein that open the channel for a time
before it closes again.
This simple gating underpins all of the physiological
consequences that are controlled by ion channels, but the
detailed elucidation of precisely how channels gate is only just
beginning to emerge. Ion channels have been discovered across
species, and in particular the prokaryotic bacterial channels have
been extremely useful to the structural biology community in
providing research tools for high resolution X-ray crystallography
and understanding of how channels function.10 More recently
there has been significant progress in the elucidation of
mammalian structures of ion channels, most notably in the
voltage-gated family of potassium channels by crystallogra-
phy11,12 and nuclear magnetic resonance13 methods. Further
discussion of our structural understanding of ion channels is
presented later.

Figure 2. Cartoon representation of the basic structural components of


■ PHARMACOLOGY/DISEASES
Ion channels are expressed in all tissues and cell types in the
an ion channel featuring a pore region, ion, or selectivity filter and gate. human body, so it is unsurprising that these proteins cause, and
594 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

are modulated by, many diseases. The availability of selective and Table 1. Selected Channelopathies Arising through Channel
nonselective pharmacological tools and the ability to genetically Function Impairmenta
manipulate channels in situ are increasing our knowledge of their
ion channel
function and provide indirect evidence of contributions of ion family channel G/L disease
channels to normal function and the pathophysiology of disease.
Kir Kir1.1 L Bartter’s syndrome
The most compelling evidence for a role in disease has come
Kir2.1 L Andersen’s Syndrome
from studies of the inheritance of relatively rare monogenic
Kir6.2 L congenital hyperinsulinism
diseases caused by mutations that affect the function of specific
G neonatal diabetes
ion channels.14 Loss and gain of function mutations in the voltage
SUR2 L dilated cardiomyopathy
gated sodium channel Nav1.7 cause profound pain insensitivity15
Kv Kv1.1 L episodic ataxia type 1
and sensitivity,16 respectively, in carriers of these mutations. A
KCNQ1 L long QT syndrome
number of Nav1.7 blockers have now entered clinical develop- G short QT syndrome
ment,17 and it will be fascinating to learn if small molecule KCNQ2 L benign neonatal febrile convulsions
exogenous channel blockers can mimic the insensitivity-to-pain KCNQ4 L nonsyndromic deafness
state of the individuals that carry the genetic loss of function hERG L long QT syndrome
mutations. A loss of function mutation in the gene that encodes G short QT syndrome
the twin pore potassium (K2P) channel TWIK-related spinal TRP TRPP2 polycystic kidney disease
cord potassium channel (TRESK) renders the channel nonfunc- TRPA1 G familial episodic pain syndrome
tional and has recently been linked to familial migraine.26 There TRPC6 G focal segmental glomerulosclerosis
is extensive evidence for mutations in voltage gated calcium CNG CNGA1 L retinitis pigmentosa
channels (Cav) affecting central functions that give rise to KCa BK G epilepsy
conditions such as ataxia and epilepsy. These are a few examples Nav Nav1.1 G epilepsy
of disease associations that exist in every ion channel subfamily.18 L severe myoclonic epilepsy
Just some of the myriad diseases and disorders that have been Nav1.5 G long QT syndrome
linked to ion channels include Cav channels and retinal disease,19 Nav1.6 L cerebellar ataxia
Nav channels and epilepsy, cardiac arrhythmias and pain,20 Kv Nav1.7 G erythromelalgia, paroxysmal extreme
channels and seizures,21 voltage gated potassium channel, pain disorder
subfamily Q (KCNQ) channels and deafness and epilepsy,22 L congenital indifference to pain
TRP polycystic (TRPP) channels and renal cysts,23 inward Nav2.1 G benign familial neonatal seizures
rectifier potassium (Kir) channels and kidney transport and Cav Cav1.2 G timothy syndrome
hypoglycaemia,24 TRP mucolipin (TRPML) channels and Cav2.1 L episodic ataxia type 2
Niemann−Pick disease,25 and K2P channels and migraine and glycine GLRA1 L stiff baby syndrome
cardiac arrhythmias.26 Table 1 presents a summary selection of receptors
just some of these “channelopathies”, human diseases that are GABA GABAA L juvenile myoclonic epilepsy
brought about through mutations to specific ion channels.27 AChR CHRNA4 L autosomal dominant nocturnal frontal
lobe epilepsy
Large scale population genetic studies, focusing on common a
variants of ion channels, are also revealing new and unexpected G/L: gain or loss of function. Adapted from Ashcroft.27b
functions28 that would not have been predicted based on current
knowledge of their function and distribution. For example, four
large scale genome wide association studies have linked a
common single nucleotide polymorphism (SNP) in the Nav1.8
gene with a small increase in human cardiac conduction velocity,
yet this channel was considered to be expressed specifically in
pain sensing neurones.29 A functional effect of this SNP on
Nav1.8 biophysical properties has not been demonstrated.
Intensive sequencing efforts are also revealing that the repertoire
of ion channel variants that an individual expresses may be an
important determinant of health.
Relatively nonspecific ion channel modulators are a potential
source of additional information on ion channel function;
however, it can be difficult to draw firm conclusions because of
poorly characterized polypharmacology. As more selective ion
channel drugs become available, their use in the general human
population is likely to provide clearer insight into the functional Figure 4. Relative size of the ion channel and other protein families.
roles that ion channels play, in addition to providing better drugs
for the treatment of diseases with ion channel etiology.


A phylogenetic tree of the ion channel superfamily arranged by
voltage or ligand gated families is shown in Figure 5. The largest
PHYLOGENY branch of the family covers the voltage gated Kv potassium
In the human genome, the pore-forming ion channel family is channels, and overall the potassium channels form the largest
larger than the nuclear hormone receptor family, approximately proportion of the whole ion channel proteome. There is also a
half the size of the kinase and protease families, and accounts for marked difference between members of the same subfamily.
approximately 1% of the estimated 20 000 human protein coding Members of the Kv branch of the family are quite distinct in
genes (Figure 4). sequence and topology to other potassium channels, for example,
595 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 5. Ion channel phylogenetic tree.

the K2P or the Kir channels. Between different branches of the additional targets with a compelling biology will stimulate
tree, proteins are even more dissimilar, for example comparing increased interest from the biomedical research community to
the Nav channels to the Cav or Kv subfamilies. Within the ligand- ultimately identify better ligands.
gated family of proteins, the structural and functional divergence The vast majority of mammalian ion channels appear to have
is even more acute and subfamilies cluster as almost distinct had their origins in prokaryotes30 but have undergone substantial
collections of channels. Perhaps not surprisingly, the most evolution to produce the eukaryotic channels apparent today.
conserved regions of ion channels tend to cluster around the pore This is perhaps driven by an increasing need for neuronal
helices or the selectivity filter regions. signaling or an evolved response to external environmental
The phylogenetic tree of Figure 5 also demonstrates the sheer factors such as temperature or chemosensing. Comparisons of
number of members of the family. By surveying all of the patent gene numbers, however, do not indicate an exact correlation with
and publication landscape for ion channel research, we estimate anatomical complexity, as the crude numbers of channel genes in
that of all of the channels depicted in Figure 5, approximately humans and nematodes is surprisingly similar.31
40% of the family has no known ligands reported against them.
Of the remaining 60%, many of these are nonselective, reactive,
peptidic or occupy very poor physicochemical space for oral drug
■ SCREENING
Ion channel screening assays can be broadly divided into ligand
discovery programs. There appear to be several reasons for this. binding, ion flux assays, fluorescence readouts, flash lumines-
First, biological reagents have not been made available (or cence assays, and automated electrophysiology.
developed) for many of the family members, making screening High-throughput ion channel binding assays have been
campaigns to identify ligands impossible. In some cases, reagents configured for filter binding assays, scintillation proximity assays,
are available, but screening efforts have merely identified poor and fluorescence polarization assays.32 The technology is high-
quality leads, which may be a consequence of many large throughput and low cost and has been adapted to a range of ion
compound collections having been built around more traditional channels, for example, N-methyl-D-aspartate (NMDA) subtype
target space, for example, G-protein-coupled receptors glutamate receptors and Kv11.1.33 However, the technology is
(GPCRs), or screening campaigns having been conducted limited by availability of a suitable high-affinity ligand, provides
using much smaller sets of low structural diversity and no functional information, and only explores a single binding site.
questionable druglikeness. There are parallels to the early stages Functional cell based assays using ion flux, fluorescent dye
of kinase drug discovery in which a small subset of targets assays, or flash luminescence assays report mechanistic effects of
received a large amount of biopharmaceutical industry attention, compounds, identifying activators, inhibitors, and allosteric
which eventually began to bear fruit after significant investment. modulators. Ion flux assays use a radioactive species in
Significant attention has been devoted to certain TRP, Nav, and combination with a scintillation counter (e.g., 86Rb for potassium
Cav channels which has yielded more potent, selective, and channels and 14C-guanidinium for sodium channels)34 or use a
druglike compounds, and it can be anticipated that those tracer ion measured by atomic absorption/emission spectrom-
596 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

etry (e.g., Rb+ for potassium channels and Li+ for sodium Table 2. Some Common Ion Channel-Targeting Drugsa
channels). Radiometric flux assays are hindered by safety and
target year of first clinical
disposal challenges, while nonradiometric flux assays are limited drug channel disease target usage
by lengthy read times of atomic absorption spectrometers. Cell
verapamil L-type Cav hypertension 1982
based assays using fluorometric dyes have been developed that diltiazem L-type Cav hypertension 1982
either respond directly to changes in concentration of the ions amlodipine L-type Cav hypertension 1990
(e.g., calcium, halide, and thallium dyes) or indirectly monitor nifedipine L-type Cav hypertension 1977
ion channel activity by measuring their contribution to cell gabapentin Cav (α2δ) pain 1994
membrane potential using voltage-sensitive dyes (e.g., sodium pregabalin Cav (α2δ) pain 2004
channels, potassium channels, and GABAA).35 Next generation sotalol hERG arrhythmia 1992
fluorescence plate readers with built-in field stimulation flecainide Nav1.5 arrhythmia 1982
capabilities, suitable for activating voltage-gated ion channels, ziconotide Cav2.2 severe pain 2004
avoid pharmacological methods to open ion channels (e.g., lidocaine Nav local anesthetic 1949
scorpion toxins and veratrine for activating sodium channels) bupivacaine Nav local anesthetic 1987
and potentially provide better correlation with gold-standard lamotrigine Nav epilepsy, bipolar 1994
manual patch clamp data.36 Luminescence assays use bio- riluzole Nav amyotrophic lateral 1995
luminescent calcium-sensitive photoprotein (e.g., aequorin or sclerosis
mitochondria-targeted MitoPhotina) as a reporter protein that is phenytoin Nav epilepsy 1953
co-transfected with the target ion channel. Flash luminescence lacosamide Nav seizures and pain 2008
technology can provide lower cost and higher assay performance carbamazepine Nav epilepsy 1963
compared to fluorescence methods and has been adapted to a varenicline nAChR smoking cessation 2006
range of channels permeating calcium ions (e.g., Cav channels flupirtine KCNQ2/3 epilepsy 1984
and purinoreceptor (P2X) channels).37 retigabine KCNQ2/3 epilepsy 2011
Automated electrophysiology provides the ideal platform for diazepam GABAA depression 1963
a
supporting ion channel programs because of the high data quality Adapted from Clare.40
achievable. Currently the technology is based around two core
technologies.38 Planar patch is where a hole is fabricated in a chip discovered in the 1950s and progresses to a modern drug
in the horizontal plane and requires compound addition through discovery example that was launched within the past few years.
a robot arm, while lateral patch recordings involve holes Carbamazepine (6) is a first generation anticonvulsant
fabricated in the side of a microchannel. Current planar patch developed by J. R. Geigy (now Novartis) in the 1950s for the
clamp technology such as QPatch, PatchXpress, and Patchliner/ treatment of epilepsy, trigeminal neuralgia, and mania.41 In the
Synchropatch which provide high-quality gigaseal resistance early era of drug discovery when carbamazepine was first
recordings are ideal platforms for developing structure−activity launched, drugs were often discovered using empirical means,
relationships or screening focused libraries. Higher throughput often in isolated tissue preparations or in vivo efficacy models
screening (10−50000 compound screening libraries) can be designed to mimic some component of the clinical condition
performed using a loose-seal recording configuration from being targeted. The definitive characterization of the molecular
multiple cells in parallel (population patch clamp) using target of carbamazepine as voltage-gated sodium channels came
Ionworks Quattro/Barracuda planar patch clamp technologies.39 much later.42 Carbamazepine inhibits sustained repetitive firing
Recent developments in lateral patch technology giving rise to by blocking use-dependent sodium channels. Pain relief is
machines with a small footprint and a lower cost chip believed to be a consequence of blockade of synaptic
consumable unit are likely to lead to a higher throughput transmission in the trigeminal nucleus, and seizure control is
technology with lower costs. associated with reduction of potentiation of synaptic trans-


mission in the spinal cord. Also somewhat characteristic of ion
ION CHANNEL DRUGS channel modulators identified in this early era of drug discovery
was a general lack of ion channel selectivity. Carbamazepine is
A cursory glance over approved drugs would suggest that the ion known to block sodium channels, calcium channels, and GABA
channel family has yielded a significant number of important receptors at high micromolar levels of potency. This pan-ion-
drugs across several subfamilies. Table 2 summarizes some channel inhibition profile is likely reflected in carbamazepines'
common ion channel drugs and the indications for which they broad pharmacological properties that include anticholinergic,
were approved. Upon closer inspection, the number of discrete antiarrhythmic, antidepressant, sedative, and neuromuscular-
channels that has been successfully drugged is relatively small, blocking effects (Figure 6).
and drugs for cardiac, neurophysiological, or local anesthetic Interestingly, the structurally related oxcarbazepine42 (7) was
application are most common. It is also notable how few drugs launched nearly 3 decades later in 1990 by Novartis for the
have gained approval in the past decade, a likely consequence of treatment of epilepsy and partial seizures. It is a prodrug that is
clinical failures due to efficacy and safety issues and the apparent activated to S-(+)-licarbazepine (8) in the liver along with equal
difficulty in identifying clinical candidate quality chemical matter amounts of the less active R-enantiomer. Eslicarbazepine
across the ion channel family. acetate43 (9) is a more recent prodrug that predominantly
Many of the compounds depicted above were discovered over generates the active enantiomer 8 and was launched in 2009 for
a decade ago, with most possessing low levels of selectivity. To epilepsy. In this example, a detailed understanding of molecular
illustrate how ion channel drug discovery has evolved through target and metabolite structure led to rationally designed
the decades, we have chosen three examples to illustrate an improvements in safety and tolerability via small structural
increasing level of sophistication in the ability to identify potent changes and/or prodrugs to identify two further drugs of clinical
and selective ligands. This starts with a very early example utility.
597 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 6. Drugs based on the first generation sodium channel blocker carbamazepine.

Another ion channel drug wherein subsequent drug


generations have shown improvements through pharmacoki-
netic modification is gabapentin. Gabapentin (10) is a lipophilic
GABA analogue developed by Parke-Davis (now Pfizer) over 10
years ago as an adjunctive treatment of partial seizure associated
with epilepsy (Figure 7). It was designed to increase passage

Figure 8. Varenicline and the starting point for its discovery.

nicotinic receptor using a high throughput binding assay to assess


binding to the α4β2 nicotinic receptor along with subtype
selectivity. Functional electrophysiology measurements were
Figure 7. Gabapentin and analogues. then used to assess the desired partial agonism pharmacology
profile, which was confirmed through several in vivo models that
across the blood−brain barrier, which indeed it does, but via evaluated the ability of varenicline to alter the central
recognition by the system L amino acid transporter. During dopaminergic response to nicotine.
patient studies and clinical use, gabapentin was observed to show This approach was rather more typical of a modern drug
efficacy in the treatment of neuropathic pain and postherpetic discovery effort, in which high throughput targeted screening was
neuralgia and subsequently demonstrated efficacy in a number of used to advance structure−activity relationships and lower
in vivo pain models.44 Gabapentin has no effect on GABA throughput functional assays were then used to hone in on the
desired pharmacology profile.


receptors, and the actual mechanism of action involves binding to
a high affinity binding site on the α2δ subunit of voltage gated
calcium channels which reduces calcium currents by impairing ANESTHETICS
the trafficking function of the subunits.45 For over a century, local anesthetics such as novocaine have been
DepoMed is currently developing a once-daily, extended- used to locally block sensory perception and thereby allow
release tablet formulation of gabapentin using its gastric surgical procedures to be carried out on peripheral tissues, to
retention technology.46 Gabapentin enacarbil (11) was approved reverse acute pain, or to treat chronic pain.50 These local
in 2011 by the FDA for the once-daily oral treatment of anesthetics are weak affinity amphipathic molecules that are
moderate-to-severe restless legs syndrome.47 Pregabalin (12) is administered at relatively high doses to primarily block voltage
another GABA analogue, launched in 2004 by Pfizer for the gated sodium channels through binding of the inner pore region.
treatment of peripheral neuropathic pain, epilepsy, postherpetic Local anesthetics are also known to block some potassium and
neuralgia, diabetic peripheral neuropathy, generalized anxiety calcium channels.51 Later generations of anesthetics including
disorder, and fibromyalgia. All of these agents were designed to lidocaine (1) and bupivacaine (2) offered duration of sensory
improve the limited and variable absorption rates of gabapentin, block or safety advantages over the earlier agents. It is the safety
giving distinct pharmacokinetic advantages over the prototype aspects of these agents (central nervous system (CNS) effects
agent.48 such as tinnitus and faintness or cardiac effects secondary to
The α2δ example illustrates how a focused medicinal respiratory depression are among the more common), coupled
chemistry and screening effort, along with a carefully executed with their weak, nonselective action at ion channels that limit
their use.


clinical trial program, ultimately led to the discovery of an
important class of ion channel targeted drugs. Their develop-
ment was dependent on efficacy readouts in animal models, from VENOMS, TOXINS, AND BIOLOGICS
which the pharmacological target of the drug class was eventually The venoms of various snakes, scorpions, spiders, insects and
identified. sea-dwelling animals are largely mixtures of neurotoxic peptides
Varenicline tartrate (14, Chantix) was launched by Pfizer in that these animals have evolved for prey capture or as a defense
2006 for use in smoking cessation and represents the product of a against predators. They act, often with exquisite potency and
modern day drug discovery program.49 The drug is specifically high selectivity at Kv, Cav, and Nav ion channels and some ligand-
designed to partially activate the nicotinic receptor and reduce gated ion channels (e.g., nicotinic acetylcholine receptors), to
the severity of the smoker’s craving and the withdrawal take advantage of the neuronal and muscle contractility effects of
symptoms from nicotine. Varenicline was discovered by channel modulation.52 Optimized versions of some of the huge
modification of a natural product (−)-cytisine49 (13, Figure 8) number of venomous peptides and toxins are being pursued by
that was known to have partial agonist activity at the α4β2 researchers,53 while mixtures of fractionated venoms or
598 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry


Perspective

individual toxins are available commercially as research tools ION CHANNEL MODULATORS IN CLINICAL
from several sources. Some common toxins are shown in Figure 9 DEVELOPMENT
Analysis of drugs that modulate ion channels and have reached
clinical development reveals some interesting trends. Figure 10
shows the classes of drug targets that have been pursued
according to the decade of their first disclosure in the patent
literature. In particular, both families show large spikes in activity
over the past 5 decades for certain classes of channels. For ligand-
gated targets, this occurred between 1980 and 2000 and can be
largely attributed to a significant industry effort targeting the
glutamate, nicotinic, and serotonin receptor subtype 3 (5-HT3)
channels, as described in the sections below. For voltage gated
channels this increase in activity occurred between 1980 and
1990 and corresponds to increased activity in the areas of calcium
channels blockers such as the dihydropyridine class of
antagonists and α2δ modulators, as well as an increased interest
in potassium channel modulators. While there has been an
overall drop in clinical activity around ion channels, interest in
ion channel targets has remained strong with a clear increase in
the past decade (2000−2010) in the areas of the P2X, TRP, and
sodium channel research.
Of the 606 compounds contained in this analysis, 28% have
been successfully launched as drugs with roughly an equal
proportion of launches between the ligand and voltage-gated
families (26% and 29%, respectively). In the context of drug
approvals, the increases in activity for certain classes of targets
Figure 9. Some common toxins. For the structure of protoxin 1, see the mentioned previously did not necessarily lead to an increase in
Protein Data Bank, structure code 1KVD. approved drugs. For example, from the 1970s to the 1980s, the
number of calcium channel modulators increased from 19 to 69
compounds in clinical development (Figure 11). However
despite this increase, the percentage of approved drugs targeting
that can be both small molecule, for example, saxitoxin (15) and calcium channels fell significantly in the same period, from 65%
tetrodotoxin (16), or small peptides, ranging from 8 to 70 amino to 22%. In addition, this latter value is closer to the percentage of
acids such as protoxin 1 (17). These peptides are highly compact approved drugs that targeted calcium channels in the 1990s
and are mainly stabilized by disulfide bonds, although some are (13%), during which time only 16 compounds reached the clinic.
stabilized by hydrogen bonds made from post-translation Although a wide range of reasons could have contributed to this
modified amino acids. trend, it does point to the fact that increased clinical activity in the
An alternative modality of targeting ion channels has been the field does not necessarily translate to an increased success rate for
use of functional antibodies. Typically, ligand-gated ion channels drug launch.
have been more susceptible to this approach, perhaps because of These data also demonstrate an increase in focus toward more
smaller and less defined binding sites on the extracellular selective agents entering clinical trials (Figure 12). For both
domains of voltage-gated channels. While research into this ligand and voltage gated targets, the most recent decade has
approach is making progress and could provide important shown a large increase in clinical compounds that target single
alternatives to small molecule approaches,54 it will not be covered channel subtypes within a family (for example, selective Nav1.7
any further in this article. modulators). It is worth noting that for ligand gated channels the

Figure 10. Ion channels in clinical development from the ligand and voltage gated families, broken down by decade and ion channel subfamily.

599 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Figure 11. Translating clinical activity into launched products. Compounds marked as “Not Launched” were part of the clinical data set derived from
Prous but were not flagged with a highest phase of “Launched” in the database.

Figure 12. Pursuit of selective ion channel modulators over time.

decade of 1980−1990 constituted something of an anomaly. Table 3. Physicochemical Space Comparisons across the Ion
During this decade, the industry focused a great deal on 5-HT3 Channel Familiesa
selective modulators. While these receptors are ion channels, the
family (no.) clogP PSA MW HBD rotatable bonds ring count
chemical matter that modulates them shares a great deal of
similarity to compounds that modulate the other members of the voltage (441) 3.7 72 393 1 6 3
5-HT family, which are GPCRs. If those compounds are ligand (311) 2.5 62 333 1 3 3
removed from the analysis, only 2% of the compounds that target rotatable ring
subfamily (no.) clogP PSA MW HBD bonds count
ligand gated receptors can be classed as subtype selective (from
the 29% value shown). This trend appears to have begun in the nicotinic (32) 1.8 34 242 1 1 3
1990s and is consistent with a move away from in vivo/tissue glutamate (99) 2.0 70 309 2 4 3
GABA (112) 2.9 62 346 0 3 3.5
based drug discovery approach as highlighted above in the
5HT (41) 2.4 57 333 1 2 4
carbamazepine example toward programs driven by an increased
ASIC (6) 3.5 55 337 3 3 3.5
understanding of the existence and physiological role of ion
calcium (123) 4.5 92 461 1 9 3
channel subtypes in human disease (Nav1.5 vs Nav1.7, for
nucleotide (7) 3.9 52 334 1 6 3
example) facilitated by the advances in molecular biology and
potassium (129) 2.9 75 335 1 4 3
genetics at this time. The intriguing aspect of the analysis is
P2X (23) 2.9 95 419 3 6 4
whether this change in strategy will be successful.
sodium (118) 3.3 66 365 2 5 3
We also examined these data for trends in physicochemical
TRP (49) 4.5 66 410 1 6 3
properties. To bolster the analysis to include more recent
other (13) 3.7 54 392 0 5 3
compounds that have not yet entered clinical evaluation, we a
included a set of manually curated ion channel ligands drawn The numbers in parentheses denote the number of compounds used
from the literature. Analysis of the physicochemical properties of to generate the corresponding median values. The compounds used
for this analysis were drawn from compounds that target ion channels
both ligand and voltage gated channel ligands showed that the and had progressed to the clinic and from a manually curated set
compounds lay squarely in the middle of druglike chemical space. drawn from the literature and described in the section Methods. All
Interestingly, the ligand gated modulators tended to be smaller, properties were calculated using Pfizer internal software packages.
less lipophillic, more rigid and have reduced polar surface area
(Table 3). This difference in properties may be explained in part
by the fact that a larger number of ligand gated targets are located values of their family, compounds that modulated the nicotinic
in the CNS, which has driven the property space toward receptors tended to be smaller and of lower polar surface area.
compounds that are more freely brain penetrant.55 A more Similarly, calcium channel modulators stood out relative to their
detailed analysis of compound properties at the subfamily level other family members in that they were more lipophilic, larger,
also revealed some interesting trends. Relative to the median contained more rotatable bonds, and were of higher polar surface
600 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 13. A voltage gated Nav channel and the most common binding sites identified for ligands. The four subunits of the channel are represented in
different colors.

area. Although the reasons behind these differences are unclear, it transmembrane region of the α-subunit of Nav channels, while
does suggest that ion channels cannot be viewed as a several small molecules have been proposed to bind to the
homogeneous population that prefer compounds in a particular voltage sensor domain.57 Examples of ligands that bind to all of
physicochemical space. Instead, like aminergic and peptidic these sites are described in the sections below.
GPCRs,56 different classes within the ion channel family may be In the case of voltage gated channels, the large channel protein
biased toward compounds in different physiochemical space. comprises four subunits that create a central ion conducting pore
Analysis of compounds that were classified as more subtype with a constricted selectivity filter at the top, surrounded by four
selective did not show any definitive differences in their voltage sensors. The voltage-sensing domain (VSD) is made up
physicochemical properties relative to the non-subtype-selective of the S1−S4 segments, which are linked to the S5 and S6
compounds, which could suggest that obtaining subtype segments that form the pore, and a P-loop joins the segments
selectivity has been a direct result of improved screening together. The VSD responds to membrane depolarization and
methods. initiates opening of the central pore through pulling on the S4−

■ ION CHANNEL STRUCTURE AND MODULATION BY


LIGANDS
S5 linker and opening of the gate at the base of the pore through
concerted movements of all four subunits. In this way, the ion
channel moves from one state (for example, closed) to another
In an earlier section, we introduced the basic structural features (open) through concerted movements and then through to an
that allow ion channels to sense environmental factors, inactivated state as the channel ceases to conduct ions. Of course,
selectively filter ions, and gate. Figure 13 shows an example of ligands will possess a different affinity for each of these states, and
a voltage gated ion channel and highlights the most common an attractive strategy to achieve functional selectivity is to target a
regions that have been reported as supporting ligand binding. high affinity activated state of high frequency firing channels
Much of this evidence has been gathered through site directed characteristic of disease states but not the low frequency firing of
mutagenesis, and while there are now several apo crystal the normal somatosensory system. Several compounds have
structures of ion channels, to date there are no cocrystal been reported to possess such “state-dependent” properties.
structures of any ligand bound in a channel. It is also possible to modulate ion channel function through
TTX is known to bind to a site toward the top of the pore ligand binding to auxiliary subunits. For example, the mechanism
region. Other toxins have been described to bind to other regions of action of gabapentin has already been highlighted, which elicits
of Nav channels spanning almost all protein domains, including its analgesic effects through binding to the α2δ accessory subunit
the pore. The pore module tends to be the most conserved of Cav channels, thereby impairing channel trafficking to the cell
region of protein across subfamilies, with pore blockers tending surface.58 Ionotropic glutamate receptors (iGluRs) are known to
to support the least selectivity. This includes many of the interact with transmembrane bound regulatory proteins and
nonselective drugs shown in Table 2. A hydrophobic fenestration affect receptor trafficking and proper localization at the cell
site in the side of the pore module has also been proposed to surface. These regulatory proteins are also known to affect
support small molecule binding.10b Local anesthetics bind to a channel conductance, activation, and densensitization. Other
601 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

post-translational regulation pathways have also been inves-


tigated for iGluRs, for example, the multiple phosphorylation
sites on AMPA receptors mediated by protein kinase C and
protein kinase A.59

■ ION CHANNEL MODULATOR CHEMOTYPES


In the below sections, we survey the prevalent chemotypes that
have been described as targeting the various subfamilies within
the ion channel superfamily according to one of the modalities
described above. We have organized this survey into five sections
according to the topology of the subfamilies (Figure 14): 6-

Figure 14. Cartoon representation of the five topologies of ion channels.


Note that this representation depicts the situation where the subunits
are heteromeric for clarity to distinguish the subunits involved in
creating the channel protein, but in some cases they may be homomeric.

transmembrane channels based on four subunits and an Figure 15. Structure of Kv1.2, a 6TM ion channel (PDB code 3LUT60).
appended transmembrane helix voltage sensor domain, 4-TM
Cys-loop channels that form pores as pentamers, glutamate
receptors with four subunits, the ASIC/P2X channels that have proposed three binding sites: a pore binding site, a cleft in
contain 2-TM domains and function as trimers, and the K2P/Kir the voltage sensor, and a fenestration site.65
channels that have a 2TM subunit topology comprising either a Although no crystal structures are available for ligand-gated
functional tetramer or two pairs of subunits. TRP channels, electron microscopy and sequence analyses
We relate the cartoon representations for each topology to an suggest that the topology for these channels is similar to voltage-
exemplar protein structure in each sectioǹ to orient the reader to gated channels.66 Homotetramers of four subunits comprise two
the structure and relative complexity of each topology. For domains: a sensor region (S1−S4) and a pore domain (S5−S6).
further detailed description of the structural aspects of each The discovery of small molecule TRP channel modulators dates
individual subfamily member, the reader is referred in each back to the 1990s; however, a binding site was not identified until
section to literature papers and reviews.


2002, when Julius proposed a key role for two residues located at
the intracellular loop between S2 and S3 of the sensor region.67
6-TRANSMEMBRANE (6TM) TOPOLOGY ION In addition, an alternative pore domain binding site has also been
CHANNELS (Nav, Kv, Cav, KCa, TRP) suggested for recognition of cholesterol and menthol by
Medicinal chemistry programs aimed at developing modulators TRPA1.68 This binding site is close to the fenestration binding
of 6TM voltage-gated Nav, Kv, Cav and ligand-gated TRP site of the voltage-gated channels and is thought that cholesterol
channels have been widely reported.20c,61 Structural studies on stabilizes the channel’s inactive state to suppress ion conduction
the α1-subunit of 6TM topology ion channels date back to the in the pore.
late 1980s.62 These channels are composed of approximately Voltage-Gated Sodium Channels (Nav). Sodium channels
2000 amino acids across four structurally distinct subunits with comprise a family of nine members Nav1.1 to Nav1.9.69 They
each 6TM subunit contributing two helices to the pore domain have been identified as regulators of cellular excitability,
responsible for ion conduction and four helices to the voltage contributing to the generation and propagation of action
sensor domain (Figure 15). Recently, structural understanding of potentials in neurons, muscle, and heart tissues. Consequently,
6TM topology has been improved by the publication of several sodium channels are precedented drug targets as antidysrhyth-
crystal structures: a Kv1.2/2.1 chimera crystal structure by mics, anticonvulsants, and anesthetics.
Mackinnon et al.63 and the bacterial channel NavAb crystal A number of natural toxins such as TTX (16) bind to all states
structure from the Catterall group.10b In addition, computational of sodium channels. Toxins of this class bind to occlude the
molecular dynamics has been used to understand the opening/ extracellular channel pore, and the sensitivity of the nine sodium
closing mechanism of voltage-gated channels. A publication from channels to TTX blockade has been used to divide the family into
Jensen et al.,64 which describes microsecond order molecular two classes: (i) TTX-S (sensitive), IC50 < 30 nM against Nav1.1,
dynamics for the Kv1.2/2.1 crystal structure, suggests that the -1.2, -1.3, -1.4, -1.6, -1.7 and (ii) TTX-R (resistant), IC50 > 30 nM
channel closes via hydrophobic collapse of a pore residue, against Nav1.5 and -1.8). This affinity difference is proposed to be
followed by movement of the voltage-sensor domain toward the due to a key cysteine residue in the TTX-binding site.70
pore. In the opening process, the voltage sensor moves outward, A variety of approved drugs are modulators of sodium
which pulls the pore domain open. The exact nature of small channels for the treatment of clinical conditions associated with
molecule binding sites is less defined, as no cocrystal structures abnormal cell excitability (Figure 16).71 Several are used to treat
have been reported. However, site-directed mutagenesis studies CNS disorders such as anticonvulsants (lamotrigine (18) and
602 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

channels with Nav1.3 IC50 = 415 nM, Nav1.7 IC50 = 182 nM, and
Nav1.8 IC50 = 16 μM.75 Lacosamide (25) was approved in 2008
for the treatment of partial onset seizures and diabetic
neuropathic pain.
Nav1.7 has received considerable attention driven by the
genetic data described above. The resulting industry-wide effort
has led to a diverse array of chemotypes (Figure 18) with several
companies moving compounds with Nav1.7 activity into clinical
trials including the proline derivative 26 from GSK/Con-
vergence.76 A structurally very different neutral spiro-oxindole
template exemplified by XEN402 (27) has been advanced to the
clinic by Xenon, and this compound has been shown to be
effective at treating pain in patients with congenital erythrome-
lalgia.77 Benzazepinone 28 has been reported by Merck to be a
state-dependent inhibitor of Nav1.7 (IC50 = 90 nM) and Nav1.8
(IC50 = 680 nM) while displaying ∼10-fold use-dependent
selectivity over Nav1.5.78 Pfizer, in collaboration with Icagen, has
published several patents in recent years based on acidic and
zwitterionic series with numerous examples (e.g., 29) reported to
possess Nav1.7 potency in the single digit nanomolar range.79
Recently Pfizer also advanced a Nav1.7 compound into clinical
trials for the treatment of pain.80 AstraZeneca has published
several patents that cover a diversity of chemical series for sodium
channel inhibition such as the chromane 30 with Nav1.7 IC50 =
66 nM, Nav1.5 IC50 = 13 μM, Nav1.2 IC50 > 33 μM.81
Vertex has published several patents in the area of TTX-S
selective channel modulators based on a series containing weakly
Figure 16. Nav channel modulator chemotypes. acidic N-heterocylic sulfonamides (estimated pKa ≈ 7),
suggesting Nav1.1 and Nav1.3 to be preferred targets for this
epilepsy drugs phenytoin (19) and carbamazepine (6)) by acting series with compound 31 having Nav1.1 and Nav1.3 IC50 < 2 μM
on sodium channels expressed in brain neurones. Antidysrhyth- (Figure 19).82 Icagen and a collaboration between Pfizer and
mics (mexiletine (20), flecainide (21), and tocainide (22)) Icagen have also filed patents in which N-heterocylic
rectify cardiac rhythm by targeting Nav channels in the heart. sulfonamides have been reported to modulate TTX-S channels
Local anesthetics (lidocaine (1), benzocaine (23), and with examples such as compound 32 having Nav1.3 IC50 = 30 nM
bupivacaine (2)) have been used for many years to modulate and >1000-fold selectivity over the cardiac channel Nav1.5.83
peripheral nerves as injectable or topical agents at the site of Pfizer reported a 6,6-biaryl series optimized for Nav1.8
action. On the whole these compounds lack subtype selectivity exemplified by 33 which has a whole cell Nav1.8 IC50 of 260
across sodium channels, leading to side effects that preclude their nM and selectivity of >20-fold over Nav1.1, Nav1.5, and Nav1.7
chronic use. From a physicochemical and structural basis all of (Figure 20). Pfizer has since reported entering clinical trials with
these compounds are either weakly basic or neutral and have a Nav1.8 selective compound.20c,84 A collaboration between
been suggested to bind to a site on the intracellular side of the Abbott and Icagen has resulted in 6,6- and 6,5-biaryls with an
channel pore.72 As this region of the channel shows a high degree alternative amide geometry. Substituted pyrazine 34 showed
of sequence conservation across the subtypes, it seems Nav1.8 IC50 of 30 nM and has good selectivity over the TTX-S
reasonable that compounds that bind to this region do so channel Nav1.2 and TTX-R channel Nav1.5. Interestingly,
without imparting appreciable levels of subtype selectivity. alternative cores were tolerated and had similar profiles, for
More recently several newer agents with general Nav activity example, the furan compound A-803467 35.61c
have been taken into clinical development (Figure 17). These Although significant progress has been made recently to define
the first subtype selective sodium channel series, there is still a
need for more research in this area to deliver alternative selective
chemical substrate.
Voltage-Gated Potassium Channels (Kv). Voltage-gated
potassium channels are found in all excitable cells. They open on
membrane depolarization, allowing potassium ions to exit the
cell, and thereby repolarize the cell.85
Figure 17. Recent Nav channel modulators. Researchers at Wyeth disclosed compounds of structure 36
(Figure 21) that disrupt Kv1.1 N-type inactivation and may be
include ralfinamide (24), a state and use dependent Nav useful for reducing neuronal hyperexcitability in diseases such as
modulator from Newron that is reported to act through epilepsy and neuropathic pain.86 Kv1.3 is widely regarded as a
Nav1.3, Nav1.7, Nav1.8 and the voltage-gated calcium channel promising new target for immunosuppression. Psora4 (37) is
Cav2.2.73 Lacosamide (25) from UCB, also based around a reported to block Kv1.3 in a use-dependent manner and exhibit a
benzylamino headgroup, is proposed to bind to a novel Nav 17- to 70-fold selectivity for Kv1.3 over several closely related
binding site that enhances slow inactivation.74 This compound Kv1-family channels. It also selectively suppresses the prolifer-
also possesses some Nav subtype selectivity toward TTX-S ation of human and rat myelin-specific effector memory T cells.87
603 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 18. Nav1.7 targeted modulators.

Figure 19. Nav1.3 targeted modulators.

the Kv2 family, Kv2.2 (IC50 100−200 nM) and are >10-fold
selective over Nav and other Kv channels. At clinically relevant
concentrations, the selective serotonin reuptake inhibitor
fluoxetine (39) and its major metabolite norfluoxetine (40)
inhibit Kv3.1, with IC50 of 13.1 and 0.80 μM, respectively.89
KCNQ channels play a central role in regulating several critical
functions, such as regulation of the heart beat or modulation of
neuronal activity.61b Potent and selective KCNQ1 inhibitors
have been reported by Merck90 that function as antiarrhythmic
agents both in vitro and in vivo (41, Figure 22). A number of
KCNQ2/3 channel openers have been reported. An example of
this class is ICA-2724357 42. Researchers at Icagen investigated
the location and nature of the binding site for this selective
KCNQ2/3 selective activator, and unlike nonselective KCNQ
Figure 20. Nav1.8 selective compounds. activators including retigabine91 (43) and flupirtine92 (44), 42
reportedly binds to a novel voltage sensor domain site.57
The human ether-a-go-go-related gene (hERG, KV11.1)
potassium channel plays a significant role in cardiac action
potential repolarization. Inhibitors of this channel can induce
long QT syndrome. hERG is a promiscuous channel and binds to
a structurally diverse set of small molecules and has resulted in
many drugs being removed from the market or terminated
during clinical development because of cardiac side effects. These
include terfenadine (45), astemizole (46), and cisapride (47)
(Figure 23). It is now common practice to assess the hERG
blocking liability of compounds before they are taken to the
clinic.93 hERG channel blockers that are administered as
treatments for cardiac arrhythmias include sotalol (48). hERG
channel activators are considered potential therapeutics for
antiarrhythmia including PD-118057 49,89 an acid-containing
Figure 21. Kv channel modulators. compound that contacts the pore domain of hERG channels to
attenuate inactivation and enhance K+ conductance. Another
Merck scientists have recently disclosed compounds such as example is NS3623 50 which incorporates an acidic tetrazole
RY79688 (38) that potently inhibit Kv2.1 and another member of group and also induces hERG inactivation.89
604 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 22. Kv7 channel modulators.

Figure 23. Modulators of the hERG channel.

Figure 24. L-type Cav channel blockers.

Voltage-Gated Calcium Channels (Ca v). In many dyhydropyridines such as amlodipine (53) and nifedipine (54).
physiological systems, Cav channels are the molecular link Each drug class has been established to bind to different sites on
between membrane potential and intracellular calcium signaling the α1 subunit.94
pathways. As a result, they have become drug targets for a range Selective Cav2.1 and Cav2.3 inhibitors have not been disclosed
of neurological and cardiovascular diseases.61c The principal to date. Small molecule N-type Cav2.2 selective compounds,
protein that forms Cav channels is the α1-subunit which shares however, have been identified, driven largely by the established
similar topology with the equivalent Nav and Kv channel clinical link of this subtype to neuropathic pain.95 Ziconotide
subunits. However, there are three other Cav auxiliary subunits (Figure 25) is an approved analgesic peptide but requires
that also modulate the function of the α1 subunit: a cytoplasmic intrathecal administration and is associated with some significant
β-subunit, a membrane anchored extracellular α2δ subunit, and side effects.96
an integral membrane γ subunit. The 10 α1 subunits are divided Efforts have been made to develop systemically efficacious
into 3 subfamilies: (1) Cav1.1−Cav1.4 (L-type); (2) Cav2.1 (P/ small molecule N-type inhibitors to overcome these limitations.
Q-type), Cav2.2 (N-type), Cav2.3 (R-type); (3) Cav3.1−Cav3.3 Several groups have disclosed a variety of series including Purdue
(T-type), with all three classes involved in neuronal signaling. To Pharma 55, Merck 56, which has Cav2.2 IC50 = 150 nM and
date the L-type (Cav1) channels have been the most frequently Cav1.2 IC50 = 910 nM, Ionix 57 which has Cav2.2 IC50 = 730 nM
targeted by inhibitors for the treatment of angina and and Cav1 IC50 = 23 μM, and Ajinomoto Pharma 58 which has
hypertension, although compounds reported to date have not Cav2.2 IC50 = 720 nM and Cav1 IC50 = 44 μM (Figure 26).97
been able to readily achieve subtype selectivity (Figure 24). The Notably compounds patented by GSK have been progressed by
three main classes of drugs are phenylalkylamines such as Convergence Pharmaceuticals, e.g., CNV2197944 (structure not
verapamil (51), benzothiazepines such as diltiazem (52), and disclosed), into phase II clinical studies in 2012 for neuropathic
605 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

and voltage-gated. All are tetrameric channels with α1 subunit


topology similar to that of voltage-gated channels.
Several launched drugs are known to impart some of their
efficacy via opening of the BK (KCa1.1) channel (Figure 27).

Figure 27. KCa1.1 channel modulators.

These include the thiazides hydroflumethiazide saluron100 (61)


and chlorothiazide diuril101 (62), which work as diuretics
Figure 25. Structure of the ω-conotoxin ziconotide. For a detailed through inhibition of the NaCl co-transporter in the kidney but
structure of ziconotide, see the Protein Data Bank, structure code also have additional antihypertensive properties suggested to be
1DW5. due to BK channel activity. Flindokalner102 (63) is a positive
modulator of KCa1.1 (EC50 = 352 nM) but also a positive
pain. On the basis of the patents assigned to Convergence, the modulator of Kv7.2−7.5 (EC50 = 2.4 μM) and a negative
clinical compound appears to be related to the piperazinesulfo- modulator of the Kv7.1 channel. Flindokalner was developed as a
namide 59.98 Also, compound 60,99 originally from Neuromed, first in class compound for the treatment of stroke but did not
has been reformulated and progressed into phase I by Zalicus in achieve clinical efficacy over placebo.103
2012. A variety of unselective IK/SK channel openers are known,
Currently, selective T-type Cav3 inhibitors have not been such as NS309104 64 (Figure 28). Some SK channel selective
identified, although several nonselective compounds have been compounds are also known, such as the Neurosearch compound
proposed, at least in part, to elicit their antihypertensive efficacy CyPPA (65) (SK2 EC50 = 14 μM, SK3 EC50 = 5.6 μM, no activity
through Cav3 inhibition.61c at SK1 or IK channels).105 GSK also published GSK542573X 66
The gabapentanoid drugs gabapentin and pregabalin are used as the first selective KCa2.1 subtype selective opener via binding
to treat epilepsy and neuropathic pain through Cav channel to a novel site on the channel.106 Several recent patents have
modulation via binding to the α2δ-1 and α2δ-2 subunits, as disclosed SK channel openers with data for KCa2.3 provided but
described above. no further selectivity data. Neurosearch disclosed the imidazo-
Calcium Activated Potassium Channels. This family pyrimidine 67 with an EC50 of 20 nM.107 As these channels are
contains eight members: big potassium (BK) channel (KCa1.1), expressed in the CNS, there is a suggestion that modulators
small conductance (SK) channels (KCa2.1, KCa2.2, KCa2.3), could be beneficial in a number of neurological diseases such as
intermediate conductance (IK) channel (KCa3.1), voltage gated epilepsy or for enhancing learning and memory. However, more
potassium channel, subfamily T (KCNT) (KCa4.1, KCa4.2), and selective modulators will likely be required in the future.108
high conductance (Slo) (KCa5.1). Most of these channels are Senicapoc/ICA-17043 (68) currently owned by Pfizer is a
activated by intracellular calcium except for the KCa4 and KCa5 selective dual inhibitor of KCa3.1 (IC50 = 6 nM) and the Gardos
channels which are activated by intracellular sodium and channel KCCN4 (IC50 = 11 nM). It has been reported that this
chloride, respectively, and the KCa1 channels are both calcium- compound has IC50 > 1 μM for Kv1.5, hERG, TTX-S Nav's, IK,

Figure 26. N-type Cav channel blockers.

606 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Figure 28. IK/SK channel modulators.

Kv7.1, and Nav1.5. Senicapoc has been progressed to phase II nate, and the endogenous ligand 4-hydroxynonenal (4-
clinical trials for sickle cell anemia and for asthma.109 HNE).111,113b Electrophilic ligands have been shown to activate
TRP Channels. TRP channels constitute a family of 28 TRPA1 through covalent binding to cysteine residues present in
mammalian cation channels with biological functions ranging the cytoplasmic N-terminus of TRPA1 in in vitro systems.118
from pain perception and thermosensation to calcium and Nonelectrophilic compounds that activate TRPA1 include
magnesium absorption. Consequently, TRP channels are flufenamic acid (FFA), thymol, and 2-tert-butyl-5-methylphenol.
believed to play a role in the pathogenesis of several diseases.110 Recombinant TRPA1 is activated by noxious cold (<17 °C).
TRP channels are classified into six subfamilies: TRPA (ankyrin), Hydra Biosciences has disclosed several TRPA1 inhibitors such
TRPV (vanilloid), TRPM (melastatin), TRPC (canonical), as HC-030031119 74 (IC50 = 6.2 μM) and the related TRPA1
TRPP (polycystin), and TRPML (mucolipin) and are activated ligand HC-068559120 75 that have demonstrated efficacy in
by a range of chemical and thermal stimuli.111 TRPV1, TRPV3, preclinical pain models (Figure 30). Other known TRPA1
TRPV4, TRPA1, and TRPM8 are the most advanced in terms of selective inhibitors include AP18 76 and AMG9090 77.111,113b
TRPV3 is activated by natural products such as camphor (78)
availability of potent and selective chemical modulators.112
and carvacrol (79) and by 2-APB (80) and warm temperatures
TRPV1 is activated by numerous stimuli including heat (>42
(>32 °C).121 Several classes of TRPV3 antagonists have been
°C), vanilloids, lipids, and protons/cations. Capsaicin 3 and
disclosed over recent years (Figure 31). These include
resiniferatoxin RTX (69) are potent small molecule TRPV1 benzothiazole 81121a and quinazolin-4-one 82.121b GRC15300
agonists. Several selective TRPV1 antagonists have advanced (structure not disclosed), a Glenmark TRPV3 modulator for the
into clinical development for the potential treatment of pain and potential treatment of osteoarthritis and neuropathic pain, has
have been covered in recent reviews.113 These include recently entered phase I clinical trials in partnership with
AMG517114 70, SB-705498115 71, ABT-102116 72, and MK- Sanofi.122
2295117 73 (Figure 29), which cover a range of chemical classes. TRPV4 is activated by heat (27−34 °C) and various small
Crucially, reports from these clinical studies have shown TRPV1 molecules (Figure 32) including anandamide (ANA, 83), 5,6-
blockade to have effects on core body temperature, which has epoxyeicosatrienoic acid (5,6-EET, 84), and GSK1016790A
halted much activity on this channel. 85.123 In recent years, a number of apparently selective TRPV4
TRPA1 is activated by a variety of ligands including exogenous antagonists have been disclosed such as RN-1734 (86, IC50 = 2.3
electrophiles such as cinnamaldehyde, acrolein, allyl isothiocya- μM)124 from Renovis and pyrrole HC-067047 87 from Hydra
which has been reported to be efficacious in preclinical models of
bladder cystitis.125 Interestingly, Renovis also reported on a
structural analogue RN-1747 (88, EC50 = 770 nM) that is an
agonist of the TRPV4 channel.124
TRPM8 is activated in vitro by cool temperatures (10−23 °C)
and cooling agents such as icilin (89) and menthol (4).121a,126
The Dendreon TRPM8 agonist D-3263127 90 advanced to
clinical trials in 2009 (Figure 33). A number of TRPM8
antagonists such as the GlaxoSmithKline compound AMTB (91)
and the Janssen carboxylic acid 92 have been disclosed, with the
latter compound (TRPM8 IC50 = 4 nM) showing efficacy in
preclinical models of pain.128
Although there is much potent and selective substrate for the
modulation of TRP channels, to date no compounds have
successfully progressed to launched drug status.

■ Cys-LOOP CHANNELS (GABAA, nAChR, 5-HT3,


GlyR, GluR)
Cys-loop channels are ligand-gated channels that control fast
synaptic transmission. The Cys-loop family comprises GABA-A
receptors, nAChR (nicotinic acetylcholine receptor), 5-HT3
Figure 29. TRPV1 channel modulators. receptors, glycine receptors, and glutamate receptors.
607 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 30. TRPA1 channel modulators.

Figure 31. TRPV3 channel modulators.

Figure 32. TRPV4 channel modulators.


Figure 33. TRPM8 channel modulators.

Cys-loop channels adopt pentameric structures (Figure 34) site, including muscimol (94), THIP (95), and 4-PIOL (96)
with C5 symmetric assembly of five subunits, each of which (Figure 35).
consists of two domains: an extracellular N-terminal domain and Benzodiazepines were introduced into clinical practice in the
a transmembrane domain. The transmembrane domain is 1960s and proved successful in the treatment of anxiety and
composed of four helices, which are connected to each other insomnia. They bind to a site that is separate from the GABA
by extracellular and intracellular loops.130 A number of cocrystal binding site, and a diverse number of ligands have been reported
structures of nicotinoids and AChBP (a homologous protein of (Figure 36) that bind to this benzodiazepine site including
nAChR) have been disclosed.131 According to these structures, diazepam (5) (GABAA α5β3γ2 EC50 = 17 nM),133 flumazenil
the binding site lies in a cleft at the interface between the (97) (GABAA α3β3γ2 Kd = 0.45 nM),134 flavonoids such as
subunits, which is located in the middle of the N-terminal genistein (98), and β-carbolines such as CCP (99).135
domain. The effects of benzodiazepines on GABAA receptors are
GABA channels. GABAA is a ligand-gated ion channel whose dependent on subunit composition. The α1 subtype is associated
endogenous ligand is γ-aminobutyric acid (GABA, 93), a major with sedation and addiction, while α2 and α3 subtypes are
neurotransmitter in the CNS. The subunits are grouped into associated with anxiolysis and muscle relaxation effects. A
eight families (α1−6, β1−3, γ1−3, δ, ε, π, θ, and ρ).132 A limited number of clinical compounds with higher affinity to α1
number of other ligands are known to bind to the GABA binding containing GABAA receptors, such as zolpidem (100) and
608 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 37. α1 selective GABAA ligands.

Figure 34. Structure of an anion-selective Cys-loop receptor (GluCl,


PDB code 3RHW129).
Figure 38. α2/3 selective GABAA ligands.

The α5 subunit has reported links to cognition.141 Merck


developed L-655708 105 (α1, Ki = 49 nM; α2, Ki = 27 nM; α3, Ki
= 25 nM; α5, Ki = 0.45 nM) and MRK-016 106 (α1, Ki = 0.8 nM,
23% enhancement over baseline; α2, Ki = 0.8 nM, 4%
enhancement; α3, Ki = 0.8 nM, −15% enhancement; α5, Ki =
Figure 35. GABAA ligands that are known to bind to the GABA binding 1.4 nM, −96% enhancement) (Figure 39), both partial inverse
site. α5 agonists selective over α2, α3, α1, and α6142 which enhance
cognition in animals without anxiogenic or convulsive side
effects.
zaleplon (101) (subtype selectivity GABAA α5β3γ3 EC50 > 1
μM, GABAA α1β2γ2 EC50 = 0.29 nM, GABAA α3β3γ2 EC50 > 1
μM, GABAA α2β3γ2 EC50 = 1.63 μM),136 are used for the
treatment of insomnia (Figure 37).
More recently research has focused on the delivery of α2 and/
or α3 functionally selective ligands to deliver nonsedating
anxiolytics.137 SL651498 102 (Figure 38) is a full positive
allosteric modulator (PAM) of α2 but a partial PAM of α1
subtypes.138 L-838417 103 is a Merck partial PAM of α2-, α3-,
and α5-containing receptors (α2, Ki = 0.7 nM, 34% enhancement
over baseline; α3, Ki = 0.7 nM, 39% enhancement; α5, Ki = 2.3
nM, 36% enhancement) that does not enhance the GABA Figure 39. α5 selective GABAA ligands.
response at α1 receptors139 and exhibits a nonsedating anxiolytic
effect in non-human primates. MRK-529 104 exhibits functional AChR Channels. Acetylcholine (ACh) 107 (Figure 40) is a
selectivity for α2 and α3 over α1 (α1, Ki = 1.3 nM, 0% neurotransmitter and the endogenous ligand for the nicotinic
enhancement; α2, Ki = 0.6 nM, 37% enhancement over baseline; acetylcholine receptor (nAChR). Molecular biology of nAChR
α3, Ki = 0.5 nM, 45% enhancement) and is reported to be has shown this functional receptor to consist of five subunits.143
efficacious in animal models of anxiety with no sedation or A limited number of subunit combinations have so far shown
ethanol potentiation effects.140 therapeutic promise, including α7 and α4β2, which are expressed

Figure 36. Benzodiazepine binding site ligands of the GABAA channel.

609 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Figure 40. α7 and α4β2 AChR ligands.

in the brain and known to be involved in several biological


processes.144 Nicotine 108 is an agonist of these receptors and
■ IONOTROPIC GLUTAMATE RECEPTOR CHANNELS
Ionotropic glutamate receptors (iGluRs) are ligand gated ion
has been reported to improve cognition and attention in rodents,
channels that mediate excitatory neurotransmission and play a
primates, and humans.145 This, coupled with the observation that
nAChRs have reduced expression in the brain of schizophrenia key role in the development and function of the mammalian
and Alzheimer's disease (AD) patients, has driven a search for α7 central nervous system. Many human neurological disorders
and α4β2 receptor agonists for the treatment of cognitive such as epilepsy, stroke, and Alzheimer’s disease can be linked to
impairment disorders. A number of α7 and α4β2 receptor overstimulation or underactivity of iGluRs.157 Because of a
agonists and partial agonists have been reported, including the wealth of structural biology, they also represent one of the best
pyridyl ethers A85380146 109 (α4β2 agonist, EC50 = 700 nM, Ki understood classes of ion channel.158 These findings have aided
= 50 pM) and AZD-3480147 110 (α4β2 agonist, EC50 = 106 nM), development of iGluR agonists, antagonists, and modulators as
the piperidine derivative GTS-21148 111 (α7 weak agonist EC50 research tools and potential therapeutic agents.159
= 81 μM), and the piperidine derivative PNU-282987149 112 (α7 The iGluRs function as a complex of four individual subunits
agonist EC50 = 150 nM). Preclinical studies have confirmed their (Figure 42). The AMPA and KA receptors can exist as homo-
efficacy in a wide range of indications, including the use of the and heterotetramers that bind glutamate, whereas NMDA are
partial agonist varenicline (14) for smoking cessation which is heterotetramers with subunits that are activated by glycine (N1
described above (binding Ki = 60 pM at rat cortex α4β2, with and N3) and glutamate (N2). The iGluRs are subdivided into
∼30−60% the in vivo efficacy of nicotine with block of the in vivo three categories based on selective activation by small molecule
nicotine response).150 agonists (Figure 43); AMPA 119 (α-amino-3-hydroxyl-5-
5-HT3 Channel. 5-HT3 is expressed in the CNS, the
peripheral nervous system, and the gut and is associated with
excitatory synaptic transmission. Its endogenous ligand is
serotonin (113), a monoamine neurotransmitter (Figure 41).

Figure 41. 5-HT3 channel modulators.

Drug discovery approaches toward this channel have been very


successful, with several launched drugs having been identified,
many based on the structure of 113. Antagonists of the 5-HT3
channel have proved clinically effective in the treatment of
chemotherapy-induced nausea and vomiting and irritable bowel
syndrome. Some of the approved 5-HT3 antagonists include
indisetron151 (114), ramosetron152 (115), ondansetron153 Figure 42. Structure of an AMPA-subtype glutamate receptor (GluA2,
(116), palonosetron154 (117), and azasetron155 (118).156 PDB code 3KG2160).

610 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Figure 43. Agonists of glutamate receptors.

methyl-4-isoxazolepropionic acid), NMDA 120 (N-methyl-D-


aspartate), and KA 121 (kainic acid). Molecular cloning
identified four receptor subunits for AMPA (GluA1−4), five
for KA (GluK1−5), and seven for NMDA (GluN1, GluN2A−
GluN2D, GluN3A, GluN3B).
Agonists of the iGluRs are typically derived from glutamate,
AMPA, NMDA, or KA and feature or mimic the α-amino acid
moiety of the neurotransmitter. However, small structural
changes alter selectivity across subtypes and receptor families.
For instance AMPA has negligible effects on KA receptors but
replacement of the isoxazole oxygen with a sulfur atom and
increasing steric bulk at the 5-position provide analogues such as
thio-ATPA 122 which invert potency and selectivity for GluK1
over the AMPA receptors (GluA1 EC50 = 5.2 μM, GluK1 EC50 =
0.1 μM).161 Agonist activity for a series of substituted willardiines
has been compared between kainate preferring receptors in
dorsal root ganglion (DRG) neurons and AMPA preferring
receptors in the hippocampal (HPC) neurons.162 (S)-5-
Fluorowillardiine 123 shows high selectivity for AMPA receptors Figure 44. Competitive antagonists of glutamate receptors.
(HPC EC50 = 1.5 μM, 46-fold selective over DRG neurons). (S)-
5-Chlorowillardiine 124 demonstrates similar potencies across pharmacologically between AMPA and KA receptor families
both neurons (EC50 of 2−7 μM), whereas (S)-5-iodowillardiine with similar affinities at both. 5,7-Dichlorokynurenic acid166 133
125 provides greater agonist activity and selectivity for the KA and AP-V167 (134) represent antagonists targeting the glutamate
receptors (DRG EC50 = 0.14 μM, 137-fold selective over HPC (GluN2) or glycine (GluN1, GluN3) site of NMDA receptors,
neurons). This preference for KA receptor agonist activity was respectively.
rationalized on steric arguments analogous to the AMPA/thio- Clinically, agonists and competitive antagonists of iGluRs
ATPA observations. A large, lipophilic C4 substituent on typically exhibit many severe CNS-mediated side effects. It is
glutamate derived LY-339434 126 also provides agonist widely anticipated that allosteric inhibition of iGluRs will display
selectivity for GluK1 over AMPA receptors and other KA a smaller number of side effects at therapeutic doses. However,
receptor subtypes (GluK1 EC50 = 2.5 μM, GluK2 EC50 > 100 they are less well studied than competitive inhibitors and the
μM, GluA EC50 > 300 μM).163 Agonists of NMDA receptors that limited structural information to guide design has impeded
bind to the glutamate site are typically derivatives of glutamic acid development of such therapeutic agents.
or NMDA such as homoquinolinic acid 127. Similarly, agonists Not surprisingly, noncompetitive antagonists and allosteric
that bind to the glycine site are usually amino acid derivatives modulators of iGluRs show a much wider structural variety than
such as cyclopropyl analogue 128. agonists and competitive antagonists. Benzodiazepine GYKI
Most of the competitive antagonists of glutamate receptors are 52466 135 is the prototypical noncompetitive antagonist at the
expanded versions of agonists often incorporating additional AMPA receptor and has been used as a lead compound to
acidic moieties or with negatively charged non amino acid ends. develop analogues with improved potency and biopharmaceutics
AMPA and KA receptor antagonists ATPO (129) and willardiine (Figure 45).168 Benzothiadiazine derivatives, such as cyclo-
derivative 130 are typical exemplifications of this pharmacophore thiazide (136), and sulfonamide analogues, such as LY-404187
(Figure 44).164 Similarly, tezampanel (131), which represents a 137169 from Lilly, are positive allosteric modulators of AMPA
ring expanded and homologated version of KA, inhibits both receptors. Modulators generally bind to the ligand-binding
AMPA and GluK1 receptors with similar affinity (IC50 of 0.5−2.5 domain and stabilize the agonist-bound conformation, slowing
μM).165 Quinoxalinediones, such as CNQX164 (132), represent receptor desensitization and/or deactivation. An increased
an alternative structural class of AMAP/KA receptors. These structural understanding of glutamate receptors has helped
classical antagonists are typically unable to differentiate lead research into design of subtype selective blockers in an
611 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 45. Noncompetitive antagonists and allosteric modulators of glutamate receptors.

attempt to further separate efficacy from adverse side effects ASIC Channels. There are several known subunits of the
associated with pan-receptor blockade. In particular, subtype acid-sensing ion channels (ASICs)174 (ASIC1a/b, ASIC2a/b,
selective NMDA-GluN2B receptor antagonists have attracted ASIC3 and ASIC4) that are susceptible to activation by changes
considerable attention in the pharmaceutical industry for the in pH, implying a role in conveying pain from tissue acidosis.175
targeting of CNS disorders including stroke and Parkinson’s Channels containing the ASIC3 subunit are highly expressed in
disease.170 Ifenprodil171 (138) is the prototype GluN2B nociceptors and have received significant interest from the
antagonist that served as a template for the design of more pharmaceutical industry as targets for blocking chronic
selective derivatives, such as traxoprodil (139),172 which has inflammatory pain.176 Diuretic agent amiloride (140) is the
progressed into clinical trials for a number of indications prototype weak nonselective blocker of ASIC channels (IC50 of
including stroke and neuropathic pain. 10−50 μM) (Figure 47).177 Amidine A-317567 141 was the first

■ ASIC AND P2X CHANNELS


The acid-sensing ion channels (ASICs) and purinergic P2X
receptors are trimeric ion channels whereby each subunit
contains two transmembrane domains (Figure 46), a large
extracellular loop, and relatively short intracellular amino and
carboxyl termini.

Figure 47. Examples of ASIC inhibitors.

non-amiloride small molecule blocker of ASIC demonstrating


increased potency over amiloride in vitro (ASIC3 IC50 = 9.5 μM)
and in vivo.178 The potency of amidine containing molecules has
been optimized further by several groups including an indole
series from Merck represented by compound 142 (ASIC3 IC50 =
133 nM).179 The acylguanidine or arylamidine is seen as the key
pharmacophore for ASIC activity. This motif is implicated in
potential off-target effects and poor bioavailability, hampering
development of either tools or potential drug candidates.
Attempts to explore non-amidine containing inhibitors at the
ASIC3 channel led to identification of amine 143.180 However,
this compound also demonstrated polypharmacology across
numerous targets and CNS related side effects through increased
brain penetration. Further research is required to identify potent
selective ASIC inhibitors to fully elucidate their role in pain and
neurological disease states.
P2X Channels. P2X receptors are purinergic cell surface ion
channels gated by extracellular ATP.181 Seven receptor subtypes
(P2X1−7) have been characterized in mammalian cells which
Figure 46. Structure of a P2X receptor (P2X4, PDB code 4DW1173). assemble as homotrimeric and/or heterotrimeric functional
612 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Figure 48. Examples of P2X3 and P2X2/3 receptor antagonists.

Figure 49. Examples of P2X7 receptor antagonists.

channels (depending on subtype). P2X3, P2X2/3, P2X4, and P2X7 149 is potent at the hP2X3 receptor (IC50 = 24 nM) and selective
receptors have received the most attention from the over rP2X2/3 (IC50 = 8580 nM), but incorporation of a methyl
pharmaceutical industry as potential targets to treat a variety of substituent (150) at the benzylic carbon resulted in an almost
conditions including chronic pain, arthritis, and bladder equipotent inhibitor at the hP2X3 (IC50 = 21 nM) and rP2X2/3
malfunction.182 (IC50 = 74 nM) receptors.187
The discovery of A-317491 144 provided the first potent and Several groups have disclosed P2X7 receptor antagonists based
selective, non-nucleotide, small molecule dual P2X3/P2X2/3 on lipophilic benzamides (Figure 49).188 AstraZeneca described
antagonist (Figure 48).183 While this compound delivered a series of adamantane containing benzamides whereby the
significant in vivo activity in pain and overactive bladder models, physicochemical properties could be significantly manipulated
the presence of the three carboxylic acids severely limited its oral through variation of the 5-phenyl substituent.189 Incorporation
bioavailability. Since this discovery, many companies have of basic groups was well tolerated (e.g., compound 151, hP2X7
explored alternative lead matter to identify P2X 3/P2X2/3 pIC50 > 5) providing improved solubility and metabolic stability.
antagonists in more clinically viable oral drug space.182c Pfizer researchers described multiparameter optimization of a
Following extensive SAR investigation on a diaminopyrimidine high throughput screening hit to optimize potency and
series derived from an HTS hit, researchers at Roche identified physicochemical and pharmacokinetic properties within an
RO-4 (145, rP2X3 IC50 = 13 nM, hP2X2/3 IC50 = 25 nM) azauracil series, culminating in clinical candidate CE-224,535
followed by the more potent analogue RO-51 (146, rP2X3 IC50 = (152, IL-1β IC 50 = 1 nM). 190 The ortho-substituted
2 nM, hP2X2/3 IC50 = 5 nM) for the treatment of pain.184 The arylbenzamide remained a key structural feature of numerous
incorporation of the diol moiety in 146 could potentially be a P2X7 receptor antagonists described by Evotec (formerly
strategy to impart some CNS restriction, although this is not Renovis). Of particular interest are the oxoisoquinoline
discussed by the authors and no data relating to this are disclosed. benzamides which demonstrated exquisite potency in an IL-1β
Biarylbenzamide derivatives are also described as potent P2X3 inhibition assay (e.g., compound 153, IL-1β IC50 = 0.01 nM).191
and P2X2/3 receptor antagonists by a number of companies A small number of series described in the literature do not
including Roche (compound 147, hP2X2/3 IC50 = 8 nM)185 and possess the benzamide pharmacophore typified by the majority
Merck (compound 148, hP2X2/3 IC50 = 8 nM).186 An interesting of disclosed P2X7 receptor antagonists. For example, workers at
example from AstraZeneca’s pyrrolopyrimidinone series dem- GlaxoSmithKline have recently reported a series of pyrazolea-
onstrated that subtype selectivity can be achieved across cetamides with impressive ligand and ligand lipophilic
homotrimeric and heterotrimeric channels. Quinoline derivative efficiencies (e.g., compound 154, hP2X7 pIC50 = 7.7).192
613 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

Evotec, Pfizer, and AstraZeneca have all progressed P2X7 It has been reported that caffeic acid derived esters such as
receptor antagonists to clinical trials, although the lack of efficacy CDC (155) and CAPE (156) act as openers of the native TREK-
in treatment of rheumatoid arthritis for CE-224535 and 1 (K2P2.1) channel at micromolar concentrations (Figure 51).196
AZD9056 is likely to focus future industry efforts on potential Acidic tetrazole compound BL-1249 157 has been shown to be
alternative indications such as chronic obstructive pulmonary an opener of TREK-1 with micromolar activity in both binding
disease and pain.193 and electrophysiology assays.197 Riluzole (158), an established

■ TWIN-PORE AND INWARDLY RECTIFYING


POTASSIUM CHANNELS (K2P AND Kir)
neuroprotective agent with anticonvulsant, sedative, and anti-
ischemic properties, has also been reported to be a micromolar
affinity activator of TREK-1 and TRAAK (K2P4.1). Interestingly,
K2P Channels. Twin-pore potassium channels contain two while activation of TRAAK was sustained, the activation of
pore forming P-loop domains and four transmembrane segments TREK-1 was transient and followed by inhibition thought to be
(M1−M4) that organize to form a single transmembrane pore mediated by a protein kinase A dependent inhibition of TREK-1.
(Figure 50) selective for potassium.195 The class contains 15 TRAAK was permanently activated by riluzole, as this channel
lacks the negative regulation pathway.198
In addition to openers of the K2P channels, several
compounds have been reported as blockers. Interestingly,
dihydropyridine calcium channel antagonists have been shown
to have activity for the inhibition of TREK-1. Amlodipine (53)
has pronounced submicromolar activity at TREK-1 IC50 = 400
nM.199 The local anesthetic lidocaine (1) has also been shown to
inhibit TREK-1, albeit with weak activity (IC50 = 180 μM).200
Similarly, lamotrigine 18 inhibits TRESK (K2P18.1) but not
TREK-1.201 Sanofi-Aventis patented a series of compounds that
inhibit TASK-1/2 with sulfonamide 159 having TASK-1 IC50 of
100 nM in Xenopus oocytes and a patch clamp TASK-3 IC50 of 1
μM.202 Merck has reported an alternative TASK-1/3 inhibiting
series, compound 160, which has TASK-1 IC50 of 300 nM and
TASK-3 IC50 of 35 nM and has also been shown to be active in
preclinical rodent sleep pattern pharmacology models.203
The inward rectifier family of potassium (Kir) channels
comprises at least 16 members that have broad tissue distribution
and are implicated in a variety of functional roles. There is a
Figure 50. Structure of a Kir potassium channel (KirBac3.1, PDB code paucity of potent and selective small-molecule modulators that
2WLK194).
target specific family members. However, considerable work has
been undertaken to deliver selective Kir1.1 (ROMK) inhibitors
members that are widely distributed, voltage-insensitive, non- (Figure 52), as Kir1.1 is thought to play a central role in the
inactivating background channels expressed in neurons, muscle regulation of salt and potassium homeostasis.204 A class of
cells, and endocrine cells. They are commonly named according selective small molecule Kir1.1 inhibitors has been described by
to their historic origins. These channels act to dampen electrical Merck and exemplified by piperazine 161, which shows a narrow
activity and thereby regulate cell functions including secretion of window over the hERG channel (ROMK IC50 = 50 nM, hERG
neurotransmitters and hormones. Activators (openers) are IC50 = 170 nM).205 Additionally, a Kir2 selective inhibitor has
anticipated to have potential as therapeutics to treat neurological been disclosed, ML133 (162) (Kir2.1 IC50 = 1.8 μM).206
and cardiac diseases.196 Currently there are relatively few Benzimidazoleimine analogues such as R70 163 were reported
compound classes that have been shown to modulate a subset by Vanderbilt University, TN, to selectively inhibit Kir2.3 (IC50 =
of K2P channels. 24 μM) over other Kir channels.207

Figure 51. K2P channel modulators.

614 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Figure 52. Kir channel modulators.

■ RECENT TRENDS
In a survey of the range of ion channel targeted chemotypes, it is
It is also striking how similar templates/fragments appear quite
often as ligands for different channels. Small amide substituted
heterocycles have appeared as Kv7 channel openers, Kv2 channel
apparent that the breadth of ion channels being targeted in drug
blockers, and Nav channel blockers, and templates such as these
discovery programs has greatly expanded in the past decade. This
may well offer a very useful basis for building homology models
is especially true of those channels for which a clear link to a
for specific channels and increase our understanding of the
specific disease phenotype has been established, for example, the
structural features that relate to a particular selectivity profile.
voltage-gated sodium channels. As the drug discovery Small fragment-like compounds such as riluzole have also been
community has increased their activity in these areas, the reported as ligands for several different channels.
number of clinical agents across the ion channel cohort has also Finally, as the field provides more human clinical data, this will
increased, and the field has begun to see the disclosure of more critically inform pharmacokinetic−pharmacodynamic relation-
clinical data that in some cases has raised warning flags about the ships and thereby the relationship between in vitro measures of
target. For example, core body temperature changes with TRPV1 potency, compound ADME properties, and even selectivity.
blockers have limited their further progression, while for other While the drug discovery community has moved toward more
channels the full clinical utility of the target has not been fully potent and selective ion channel agents, it remains to be seen if
elucidated, as with the P2X ligands currently undergoing clinical this is matched by improved human safety and efficacy.


evaluation.
It is interesting that the majority of the currently launched ion FUTURE PERSPECTIVE
channel modulators in clinical use today are relatively unselective
and in many cases state-independent modulators of ion channel Over the past few years, there have been significant develop-
function. This may imply that many of these compounds bind to ments in the field of ion channel drug discovery. Key highlights
somewhat conserved regions of the channel pore domains. To include major advances in ion channel cloning and screening
address some of the efficacy and/or safety shortcomings of some capabilities and an increased awareness of the various ion
channels and their subtypes. This, coupled with clinical data from
of these agents, there has been a definite trend toward targeting
non selective ion channel modulators, has increased under-
more selective agents. Non pore domain binding compounds
standing of the role that ion channels play in disease and
have been implicated in providing greater selectivity, for example,
underpinned research interest in ion channels as therapeutic
through binding to the subunits of voltage gated calcium
targets.
channels, and even compounds that achieve their selectivity More recently, the ability to screen ligands against a wide range
profile through selectively targeting a particular channel state are of high quality functional ion channel assays has become possible,
all being pursued. However, the structural knowledge of ion including the capability of screening against a number of those
channels is still at such an early stage that we have not noted any known to elicit CNS and cardiovascular side effects. This,
truly predictive methods that can be applied to any chemotype to coupled with the emergence of strong genetic linkages of specific
increase selectivity against a particular channel. Rather, the ion channels to certain disease states, has focused delivery on
realization of more selective agents appears to have been driven highly selective ligands. Furthermore, several ground breaking
by a greater availability of quality high throughput assays that publications have begun to elucidate ion channel structure and its
companies and organizations have been able to screen their relationship to function.
proprietary compound collections against. This is consistent with In the future, we anticipate continued advances in the
a move away from recognizable derivatives of natural products development of higher throughput, physiologically relevant ion
such as menthol, camphor, capsaicin, and GABA to more channel screens. We also anticipate significant advances in the
traditional small molecule chemotypes. Commensurately, the field of structural biology with the ability to generate cocrystal
diversity of chemotypes reported as active against members of structures of bound ligands in specific channel states coupled
the ion channel superfamily has greatly increased in the past with a capability to characterize ion channel−ligand interactions
decade. We note that several companies are now describing ion in biophysical assays. This will increase our understanding of
channel targeted screening subsets based at least in some cases ligand-binding events and their functional relevance which,
on known ion channel chemotypes. It is also notable that in many coupled with improved screening, should enhance our ability to
cases the definition of selective appears to be based on a relatively design safe and selective ligands. The availability of selective
small number of channel homologues from the same subfamily, ligands, associated chemical biology probes, and clinical
and there are very few reports of broad spectrum ion channel candidates, along with additional genetic data, will further aid
screening to bring greater definition to just how selective understanding of the role of specific ion channels in disease.
compounds may be, much in the same way that kinase selectivity Currently, similarities exist between chemotypes across numer-
screens are reported now. ous ion channels. This may be a function of cross-ion channel
615 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624
Journal of Medicinal Chemistry Perspective

pharmacophores or indicative of similar screening collections. Oxford, U.K. (2001), and Ph.D. from the University of Oxford under the
These chemotypes sometimes offer little in the way of ion supervision of Professor Jack E. Baldwin and Dr. Robert M. Adlington in
channel selectivity, and therefore, a challenge to the drug the field of biomimetic total synthesis (2004). This was followed by
discovery community may be to generate additional ion channel postdoctoral research with Professor Samir Z. Zard at É cole
chemical space for renewed opportunity in this area. Other areas Polytechnique, France, on xanthate based radical chemistry method
that may be of benefit are an increased understanding of ion development before joining Pfizer in 2006.
channel trafficking and how ion channels interact with accessory Alan D. Brown is Head of Chemistry at Pfizer Neusentis. He earned his
proteins. Ph.D. under the supervision of Dr. Ernie Colvin at the University of
This is an exciting time for ion channel research. In a similar Glasgow, U.K., in the field of asymmetric synthesis before joining Pfizer
way compared to the kinase field more than a decade ago, we as a medicinal chemist in 1990.
anticipate that the next decade will bring a significant growth in
the ability to design and deliver safe and selective ion channel Peter J. Cox is a biologist at Pfizer Neusentis in the U.K. He has 14 years
modulators to the clinic by harnessing advances in functional of experience in drug discovery aimed at identifying treatments for
screening, biophysical characterization, and structure-based chronic pain conditions. Prior to joining Pfizer, Peter completed
design techniques. This step−change in our ability to design postdoctoral studies in Neurosciences at Cambridge University, U.K.,
selective ligands should rapidly broaden our understanding of the and INSERM, Paris, France. He has a Ph.D. in Molecular Virology from
role of ion channels and increase the impact of ion channel the University of Glasgow, U.K., and a B.Sc. (Hons) degree in
modulators in the treatment of human disease. Microbiology from the National University of Ireland.

■ METHODS
The clinical compound analysis was conducted as follows. All ion
Kiyoyuki Omoto is a computational scientist based at Pfizer Neusentis
in the U.K. He gained his Ph.D. from Kyoto University, Japan, on
molecular orbital theory of organic reactions. He joined Pfizer Nagoya
channel targeted compounds (voltage and ligand gated) from the Prous Labs in 2002 before moving to the U.K. in 2007. His research interests
Integrity database that had a highest phase above preclinical were include lead discovery through HTS triage, 2D and 3D virtual screening
selected. All combination drug products that contained more than one
methods, and structure-based drug design.
active ingredient were removed. Compounds with a molecular
mechanism that was not strongly linked to an ion channel were Robert M. Owen is a medicinal chemist based at Pfizer Neusentis in the
removed. Dates were established from patent priority date of compound U.K. He earned his B.A. in Chemistry from St. Olaf College, MN (1997),
determined from PatBase and manual searching of compound structure and Ph.D. from the University of Wisconsin, Madison, under the
in SciFinder. For those compounds without a structure in Prous, the supervision of Professor Laura Kiessling, developing methods to
date was set to be the earliest milestone date recorded in Prous.
understand and explore multivalency in biological systems (2003),
Selectivity designations were made as follows: multiple families
(molecular mechanism designation in Prous that covered at least two and carried out postdoctoral research with Professor William Roush at
target families, e.g., Na and Ca channels), single family (molecular the University of Michigan on the total synthesis of peloruside A before
mechanism designation in Prous that covered only one target family, joining Pfizer in 2006.
e.g., AMPA), subtype selective (molecular mechanism designation in David C. Pryde is a medicinal chemist based at Pfizer Neusentis in the
Prous that covered a single member of a given family, e.g., Nav1.8). All
U.K. He earned his B.Sc. in Pure and Applied Chemistry from the
molecular properties were calculated using standard Pfizer in-house
software. PSA values were calculated using the method of Ertl et al.208 University of Strathclyde, U.K. (1991), and Ph.D. from the University of
and clogP values using methodology from BioByte (www.biobyte.com). Nottingham, U.K., under the supervision of Professor Gerry Pattenden
Compounds with MW > 700 were removed from the analysis. Analysis in the field of biomimetic radical cyclizations (1994) and carried out
was focused on all available data up to 2010. Family designation was postdoctoral research with Professor Albert Meyers at Colorado State
made based on the first molecular mechanism designation or the first ion University on stereoselective metalation chemistry before joining Pfizer
channel designation if the first was not an ion channel. For the manually in 1997.
curated set of compounds used in the analysis, reviews were conducted
of the patent and journal literature using PatBase and Scifinder, and Benjamin Sidders is a bioinformatician at Pfizer Neusentis in
small molecule compounds were selected that were the most potent Cambridge, U.K. He has a B.Sc. in Molecular Microbiology from the
and/or selective from the hits identified. University of Surrey, U.K., and a Ph.D. in Bioinformatics from the
The phylogenetic tree was constructed as follows. The primary University of London, U.K., where he undertook transcriptional analysis
sequence of proteins classified as ion channels in the IUPHAR database3 of Mycobacterium tuberculosis with Professor Neil Stoker in order to
were retrieved from Uniprot and aligned using clustalw2. Protdist and define diagnostic peptides. He joined Pfizer in 2008 and has worked in
Kitsch from the Phylip suite of programs were used to calculate distances both the antiviral and pain research units.
and build a tree from the alignments. Voltage and ligand gated channels
were analyzed separately. The final representation is artistic and has Sarah E. Skerratt is a medicinal chemist based at Pfizer Neusentis in the
been produced from alignments of single families and larger subgroups U.K. She earned her Masters in Chemistry from the University of
of proteins to aid separation and visualization. The tree may be Sheffield, U.K.(1998), and Ph.D. from the University of Nottingham,
downloaded from http://www.neusentis.com/IonChannels.php.


U.K., under the supervision of Professor Jim Anderson on the
preparation of unnatural α,α-disubstituted α-amino acid building blocks
AUTHOR INFORMATION (2002) and carried out postdoctoral research with Professor Paul
Corresponding Author Wender at Stanford University, CA, on the design and synthesis of
*Phone: +44 (0) 1304 643687. E-mail: David.Pryde@pfizer. (−)-laulimalide analogues for biological evaluation (2003) before
com. joining Pfizer in 2004.
Notes Edward B. Stevens is a biologist at Pfizer Neusentis with over 20 years
The authors declare no competing financial interest. experience in electrophysiology and ion channel biology. He obtained a
Biographies B.Sc. (Hons) degree and Ph.D. from Imperial College London, U.K. He
Sharan K. Bagal is a medicinal chemist based at Pfizer Neusentis in the held postdoctoral positions at the University of Cambridge, U.K., and
U.K. She earned her MChem in Chemistry from the University of Parke-Davis Neuroscience Research Centre, Cambridge, U.K. He then

616 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

worked as a Section Head at BioFocus and then as Chief Operating (10) (a) Doyle, D. A.; Morais-Cabral, J.; Pfuetzner, R. A.; Kuo, A.;
Officer of NeuroSolutions Ltd., Warwick, U.K., before joining Pfizer. Gulbis, J. M.; Cohen, S. L.; Chait, B. T.; MacKinnon, R. The structure of
the potassium channel: molecular basis of K+ conduction and selectivity.
R. Ian Storer earned his MSci MA in Natural Sciences from the Science (Washington, DC, U. S.) 1998, 280 (5360), 69−77. (b) Payandeh,
University of Cambridge, U.K. (1999), where he remained to undertake J.; Scheuer, T.; Zheng, N.; Catterall, W. A. The crystal structure of a
Ph.D. research on the application of solid-supported reagents to the total voltage-gated sodium channel. Nature (London, U. K.) 2011, 475
synthesis of natural products with Professor Steven V. Ley (2003). This (7356), 353−358. (c) Shaya, D.; Kreir, M.; Robbins, R. A.; Wong, S.;
was followed by a postdoctoral research fellowship at California Institute Hammon, J.; Brüggemann, A.; Minor, D. L. Voltage-gated sodium
of Technology, working with Professor David W. C. MacMillan on the channel (NaV) protein dissection creates a set of functional pore-only
development of new methods for enantioselective organocatalysis proteins. Proc. Natl. Acad. Sci. U.S.A. 2011, 108 (30), 12313−12318.
before joining Pfizer as a medicinal chemist in 2006. (11) Long, S. B.; Campbell, E. B.; MacKinnon, R. Crystal structure of a
mammalian voltage-dependent Shaker family K+ channel. Science
Nigel A. Swain is a medicinal chemist based at Pfizer Neusentis in the (Washington, DC, U. S.) 2005, 309 (5736), 897−903.
U.K. He earned his MChem from the University of Southampton, U.K. (12) (a) Miller, A. N.; Long, S. B. Crystal structure of the human two-
(1999), where he remained to undertake Ph.D. research under the pore domain potassium channel K2P1. Science (Washington, DC, U. S.)
supervision of Professor Richard C. D. Brown, working in the field of C− 2012, 335 (6067), 432−436. (b) Brohawn, S. G.; del Mármol, J.;
H insertion chemistry in natural product synthesis (2002) before joining MacKinnon, R. Crystal structure of the human K2P TRAAK, a lipid- and
Pfizer in 2003. mechano-sensitive K+ ion channel. Science (Washington, DC, U. S.) 2012,


335 (6067), 436−441.
(13) Butterwick, J. A.; MacKinnon, R. Solution structure and
ABBREVIATIONS USED phospholipid interactions of the isolated voltage-sensor domain from
TRESK, TWIK-related spinal cord potassium channel; TWIK, KvAP. J. Mol. Biol. 2012, 403 (4), 591−606.
tandem of P-domains in a weakly inward rectifying potassium (14) Hübner, C. A.; Jentsch, T. J. Ion channel diseases. Hum. Mol.
channel; CNS, central nervous system; SNP, single nucleotide Genet. 2002, 11 (20), 2435−2445.
polymorphism; PAM, positive allosteric modulator; TRP, (15) (a) Cox, J. J.; Reimann, F.; Nicholas, A. K.; Thornton, G.; Roberts,
transient receptor potential; KCNQ, voltage gated potassium E.; Springell, K.; Karbani, G.; Jafri, H.; Mannan, J.; Raashid, Y.; Al-Gazali,
L.; Hamamy, H.; Valente, E. M.; Gorman, S.; Williams, R.; McHale, D.
channel, subfamily Q; Kir, inward rectifier potassium channel; P.; Wood, J. N.; Gribble, F. M.; Woods, C. G. An SCN9A channelopathy
K2P, twin-pore potassium channel; SUR, sulfonylurea receptor; causes congenital inability to experience pain. Nature (London, U. K.)
hERG, human ether-a-go-go-related gene; CNG, cyclic nucleo- 2006, 444 (7121), 894−898. (b) Goldberg, Y. P.; MacFarlane, J.;
tide gated; BK, big potassium channel; GLR, glycine receptor; MacDonald, M. L.; Thompson, J.; Dube, M. P.; Mattice, M.; Fraser, R.;
GABA, γ-aminobutyric acid; CHRNA, cholinergic receptor, Young, C.; Hossain, S.; Pape, T.; Payne, B.; Radomski, C.; Donaldson,
nicotinic, α; AChR, acetylcholine receptor; NMDA, N-methyl-D- G.; Ives, E.; Cox, J.; Younghusband, H. B.; Green, R.; Duff, A.;
aspartate; P2X, ATP-activated purinoreceptor; 5-HT, serotonin Boltshauser, E.; Grinspan, G. A.; Dimon, J. H.; Sibley, B. G.; Andria, G.;
receptor; ASIC, acid-sensing ion channel; SK, small conductance Toscano, E.; Kerdraon, J.; Bowsher, D.; Pimstone, S. N.; Samuels, M. E.;
calcium-activated potassium channel; IK, intermediate con- Sherrington, R.; Hayden, M. R. Loss-of-function mutations in the
ductance calcium-activated potassium channel; KCNT, voltage Nav1.7 gene underlie congenital indifference to pain in multiple human
gated potassium channel, subfamily T; Slo, high conductance populations. Clin. Genet. 2007, 71 (4), 311−319.
(16) Dib-Hajj, S. D.; Cummins, T. R.; Black, J. A.; Waxman, S. G. From
potassium channel; iGluR, ionotropic glutamate receptor; Kv,
genes to pain: Nav1.7 and human pain disorders. Trends Neurosci. 2007,
voltage-gated potassium channel; Nav, voltage-gated sodium 30 (11), 555−563.
channel; Cav, voltage-gated calcium channel


(17) England, S. Voltage-gated sodium channels: the search for
subtype-selective analgesics. Expert Opin. Invest. Drugs 2008, 17 (12),
REFERENCES 1849−1864.
(1) Minor, D. L. An Overview of Ion Channel Structure, Functioning of (18) Catterall, W. A.; Dib-Hajj, S.; Meisler, M. H.; Pietrobon, D.
Transmembrane Receptors in Cell Signalling; Elsevier Inc.: San Diego, CA, Inherited neuronal ion channelopathies: new windows on complex
2011. neurological diseases. J. Neurosci. 2008, 28 (46), 11768−11777.
(2) Hille, B. Ion Channels of Excitable Membranes, 3rd ed.; Sinauer (19) Bech-Hansen, N. T.; Naylor, M. J.; Maybaum, T. A.; Pearce, W.
Associates Inc.; Sunderland, MA, 2001. G.; Koop, B.; Fishman, G. A.; Mets, M.; Musarella, M. A.; Boycott, K. M.
(3) International Union of Basic and Clinical Pharmacology. IUPHAR Loss-of-function mutations in a calcium-channel alpha1-subunit gene in
Database. http://www.iuphar-db.org/ (accessed July 14, 2012). Xp11.23 cause incomplete X-linked congenital stationary night
(4) British Pharmacological Society Guide to Receptors and Channels. blindness. Nat. Genet. 1998, 19 (3), 264−267.
http://www.guidetopharmacology.org/GRAC/ (accessed August 4, (20) (a) Escayg, A.; MacDonald, B. T.; Meisler, M. H.; Baulac, S.;
2012). Huberfeld, G.; An-Gourfinkel, I.; Brice, A.; LeGuern, E.; Moulard, B.;
(5) Hebert, S. C. General principles of the structure of ion channels. Chaigne, D.; Buresi, C.; Malafosse, A. Mutations of SCN1A, encoding a
Am. J. Med. 1998, 104 (1), 87−98. neuronal sodium channel, in two families with GEFS+2. Nat. Genet.
(6) French, R. J.; Wells, J. B. Sodium ions as blocking agents and charge 2000, 24 (4), 343−345. (b) Wang, Q.; Shen, J.; Splawski, I.; Atkinson,
carriers in the potassium channel of the squid giant axon. J. Gen. Physiol. D.; Li, Z.; Robinson, J. L.; Moss, A. J.; Towbin, J. A.; Keating, M. T.
1977, 70 (6), 707−724. SCN5A mutations associated with an inherited cardiac arrhythmia, long
(7) Zhou, Y.; Morais-Cabral, J. H.; Kaufman, A.; MacKinnon, R. QT syndrome. Cell (Cambridge, MA, U. S.) 1995, 80 (5), 805−811.
Chemistry of ion coordination and hydration revealed by a K+ channel- (c) Kemp, M. I. Structural trends among second-generation voltage-
Fab complex at 2.0Å resolution. Nature (London, U.K.) 2001, 414 gated sodium channel blockers. Prog. Med.Chem. 2010, 49, 81−111.
(6859), 43−48. (21) Browne, D. L.; Gancher, S. T.; Nutt, J. G.; Brunt, E. R. P.; Smith, E.
(8) Morais-Cabral, J. H.; Zhou, Y.; MacKinnon, R. Energetic A.; Kramer, P.; Litt, M. Episodic ataxia/myokymia syndrome is
optimization of ion conduction rate by the K+ selectivity filter. Nature associated with point mutations in the human potassium channel
(London, U. K.) 2001, 414 (6859), 37−42. gene, KCNA1. Nat. Genet. 1994, 8 (2), 136−140.
(9) Gouaux, E.; MacKinnon, R. Principles of selective ion transport in (22) (a) Kubisch, C.; Schroeder, B. C.; Friedrich, T.; Lütjohann, B.; El-
channels and pumps. Science (Washington, DC, U. S.) 2005, 310 (5753), Amraoui, A.; Marlin, S.; Petit, C.; Jentsch, T. J. KCNQ4, a novel
1461−1465. potassium channel expressed in sensory outer hair cells, is mutated in

617 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

dominant deafness. Cell (Cambridge, MA, U. S.) 1999, 96 (3), 437−446. stimulation and fluorescence detection of membrane potential. Nat.
(b) Jentsch, T. J. Neuronal KCNQ potassium channels: physislogy and Biotechnol. 2006, 24 (4), 439−446.
role in disease. Nat. Rev. Neurosci. 2000, 1 (1), 21−30. (37) Dupriez, V. J. Aequorin-based functional assays for G-protein
(23) Peters, D. J. M.; Spruit, L.; Saris, J. J.; Ravine, D.; Sandkuijl, L. A.; coupled receptors, ion channels and tyrosine kinase receptors. Recept.
Fossdal, R.; Boersma, J.; van Eijk, R.; Norby, S.; Constantinou-Deltas, C. Channels 2002, 8 (5/6), 319−330.
D.; Pierides, A.; Briessenden, J. E.; Frants, R. R.; van Ommen, G. J. B.; (38) Dunlop, J.; Bowlby, M.; Peri, R.; Vasilyev, D.; Arias, R. High-
Breuning, M. H. Chromosome 4 localization of a second gene for throughput electrophysiology: an emerging paradigm for ion-channel
autosomal dominant polycystic kidney disease. Nat. Genet. 1993, 5 (4), screening and physiology. Nat. Rev. Drug Discovery 2008, 7 (4), 358−
359−362. 368.
(24) Simon, D. B.; Karet, F. E.; Rodriguez-Soriano, J.; Hamdan, J. H.; (39) John, V. H.; Dale, T. J.; Hollands, E. C.; Chen, M. X.; Partington,
DiPietro, A.; Trachtman, H.; Sanjad, S. A.; Lifton, R. P. Genetic L.; Downie, D. L.; Meadows, H. J.; Trezise, D. J. Novel 384-well
heterogeneity of Bartter’s syndrome revealed by mutations in the K+ population patch clamp electrophysiology assays for Ca2+ activated K+
channel, ROMK. Nat. Genet. 1996, 14 (2), 152−156. channels. J.Biomol. Screening 2007, 12 (1), 50−60.
(25) Shen, D.; Wang, X.; Li, X.; Zhang, X.; Yao, Z.; Dibble, S.; Dong, X. (40) Clare, J. J. Targeting ion channels for drug discovery. Discovery
P.; Yu, T.; Lieberman, A. P.; Showalter, H. D.; Xu, H. Lipid storage Med. 2010, 9 (46), 253−260.
disorders block lysosomal trafficking by inhibiting a TRP channel and (41) Schindler, W.; Häfliger, F. Ü ber derivate des iminodibenzyls. Helv.
lysosomal calcium release. Nat. Commun. 2012, 3, 731/1−731/11. Chim. Acta 1954, 37 (2), 472−483.
(26) (a) Curran, M. E.; Splawski, I.; Timothy, K. W.; Vincen, G. M.; (42) Grant, S. M.; Faulds, D. Oxcarbazepine: a review of its
Green, E. D.; Keating, M. T. A molecular basis for cardiac arrhythmia: pharmacology and therapeutic potential in epilepsy, trigeminal neuralgia
HERG mutations cause long QT syndrome. Cell (Cambridge, MA, U. S.) and affective disorders. Drugs 1992, 43 (6), 873−888.
1995, 80 (5), 795−803. (b) Lafreniere, R. G.; Cader, M. Z.; Poulin, J. F.; (43) Dulsat, C.; Mealy, N.; Castaner, R.; Bolos, J. Eslicarbazepine
Andres-Enguix, I.; Simoneau, M.; Gupta, N.; Boisvert, K.; Lafreniere, F.; acetate. Drugs Future 2009, 34 (3), 189−195.
McLaughlan, S.; Dube, M. P.; Marcinkiewicz, M. M.; Ramagopalan, S.; (44) Backonja, M.; Glanzman, R. L. Gabapentin dosing for neuropathic
Ansorge, O.; Brais, B.; Sequeiros, J.; Pereira-Monteiro, J. M.; Griffiths, L. pain: evidence from randomized, placebo-controlled clinical trials. Clin.
R.; Tucker, S. J.; Ebers, G.; Rouleau, G. A. A dominant-negative Ther. 2003, 25 (1), 81−104.
mutation in the TRESK potassium channel is linked to familial migraine (45) Hendrich, J.; Van Minh, A. T.; Heblich, F.; Nieto-Rostro, M.;
with aura. Nat. Med. 2010, 16 (10), 1157−1160. Watschinger, K.; Striessnig, J.; Wratten, J.; Davies, A.; Dolphin, A. C.
(27) (a) Ashcroft, F. M. Ion Channels and Disease: Channelopathies; Pharmacological disruption of calcium channel trafficking by the α2δ
Academic Press: San Diego, CA, 2000; (b) Ashcroft, F. M. From ligand gabapentin. Proc. Natl. Acad. Sci. U.S.A. 2008, 105 (9), 3628−
molecule to malady. Nature (London, U. K.) 2006, 440 (7083), 440− 3633.
447. (46) Abramovicz, M. Once daily gabapentin (Gralise) for postherpetic
(28) Holm, H.; Gudbjartsson, D. F.; Arnar, D. O.; Thorleifsson, G.; neuralgia. Med. Lett. 2011, 53 (1378), 94−95.
Thorgeirsson, G.; Stefansdottir, H.; Gudjonsson, S. A.; Jonasdottir, A.; (47) Cundy, K. C.; Sastry, S.; Luo, W.; Zou, J.; Moors, T. L.; Canafax,
Mathiesen, E. B.; Njolstad, I.; Nyrnes, A.; Wilsgaard, T.; Hald, E. M.; D. M. Clinical pharmacokinetics of XP13512, a novel transported
Hveem, K.; Stoltenberg, C.; Lochen, M.; Kong, A.; Thorsteinsdottir, U.;
prodrug of gabapentin. J. Clin. Pharmacol. 2008, 48 (12), 1378−1388.
Stefansson, K. Several common variants modulate heart rate, PR interval (48) Bockbrader, H. N.; Wesche, D.; Miller, R.; Chapel, S.; Janiczek,
and QRS duration. Nat. Genet. 2010, 42 (2), 117−122.
N.; Burger, P. A comparison of the pharmacokinetics and pharmacody-
(29) Chambers, J. C.; Zhao, J.; Terracciano, C. M. N.; Bezzina, C. R.;
namics of pregabalin and gabapentin. Clin. Pharmacokinet. 2010, 49
Zhang, W.; Kaba, R.; Navaratnarajah, M.; Lotlikar, A.; Sehmi, J. S.;
(10), 661−669.
Kooner, M. K.; Deng, G.; Siedlecka, U.; Parasramka, S.; El-Hamamsy, I.;
(49) Coe, J. W.; Brooks, P. R.; Vetelino, M. G.; Wirtz, M. C.; Arnold, E.
Wass, M. N.; Dekker, L. R. C.; de Jong, J. S. S. G.; Sternberg, M. J. E.;
P.; Huang, J.; Sands, S. B.; Davis, T. I.; Lebel, L. A.; Fox, C. B.;
McKenna, W.; Severs, N. J.; de Silva, R.; Wilde, A. A. M.; Anand, P.;
Shrikhande, A.; Heym, J. H.; Schaeffer, E.; Rollema, H.; Lu, Y.;
Yacoub, M.; Scott, J.; Elliott, P.; Wood, J. N.; Kooner, J. S. Genetic
variation in SCN10A influences cardiac conduction. Nat. Genet. 2010, Mansbach, R. S.; Chambers, L. K.; Rovetti, C. C.; Schulz, D. W.; Tingley,
42 (2), 149−152. F. D.; O’Neill, B. T. Varenicline: α4β2 nicotinic receptor partial agonist
(30) Kuo, M. M. C.; Haynes, W. J.; Loukin, S. H.; Kung, C.; Saimi, Y. for smoking cessation. J. Med. Chem. 2005, 48 (10), 3474−3477.
Prokaryotic K+ channels: from crystal structures to diversity. FEMS (50) Yanagidate, F.; Strichartz, G. R. Local anaesthetics. Handb. Exp.
Microbiol. Rev. 2005, 29 (5), 961−985. Pharmacol. 2007, 177, 95−127.
(31) Jegla, T. J.; Zmasek, C. M.; Batalov, S.; Nayak, S. K. Evolution of (51) Xiong, Z.; Strichartz, G. R. Inhibition by local anesthetics of Ca2+
the human ion channel set. Comb. Chem. High Throughput Screening channels in rat anterior pituitary cells. Eur. J. Pharmacol. 1998, 363 (1),
2009, 12 (1), 2−23. 81−90.
(32) Zhang, S. P.; Kauffman, J.; Yagel, S. K.; Codd, E. E. High- (52) Dutertre, S.; Lewis, R. J. Use of venom peptides to probe ion
throughput screening for N-type calcium channel blockers using a channel structure and function. J. Biol. Chem. 2010, 285 (18), 13315−
scintillation proximity assay. J. Biomol. Screening 2006, 11 (6), 672−677. 13320.
(33) Deacon, M.; Singleton, D.; Szalkai, N.; Pasieczny, R.; Peacock, C.; (53) Clark, R. J.; Jensen, J.; Nevin, S. T.; Callaghan, B. P.; Adams, D. J.;
Price, D.; Boyd, J.; Boyd, H.; Steidl-Nichols, J. V.; Williams, C. Early Craik, D. J. The engineering of an orally active conotoxin for the
evaluation of compound QT prolongation effects: a predictive 384-well treatment of neuropathic pain. Angew. Chem., Int. Ed. 2010, 49 (37),
fluorescence polarization binding assay for measuring hERG blockade. 6545−6548.
J.Pharmacol. Toxicol. Methods 2007, 55 (3), 255−264. (54) (a) Buell, G.; Chessell, I. P.; Michel, A. D.; Collo, G.; Salazzo, M.;
(34) Ford, J. W. Potassium channels: gene family, therapeutic Herren, S.; Gretener, D.; Grahames, C.; Kaur, R.; Kosco-Vilbois, M. H.;
relevance, high-throughput screening technologies and drug discovery. Humphrey, P. P. A. Blockade of human P2X7 receptor function with a
Prog. Drug Res. 2002, 58, 133−168. monoclonal antibody. Blood 1998, 92 (10), 3521−3528. (b) Zhou, B. Y.;
(35) Baxter, D. F.; Kirk, M.; Garcia, A. F.; Raimondi, A.; Holmqvist, M. Ma, W.; Huang, X. Y. Specific antibodies to the external vestibule of
H.; Flint, K. K.; Bojanic, D.; Distefano, P. S.; Curtis, R.; Xie, Y. A novel voltage-gated potassium channels block current. J. Gen. Physiol. 1998,
membrane potential-sensitive fluorescent dye improves cell-based 111 (4), 555−563.
assays for ion channels. J. Biomol. Screening 2002, 7 (1), 79−85. (55) Wager, T. T.; Hou, X.; Verhoest, P. R.; Villalobos, A. Moving
(36) Huang, C. J.; Harootunian, A.; Maher, M. P.; Quan, C.; Raj, C. D.; beyond rules: the development of a central nervous system multi-
McCormack, K.; Numann, R.; Negulescu, P. A.; Gonzalez, J. E. parameter optimization (CNS MPO) approach to enable alignment of
Characterization of voltage-gated sodium-channel blockers by electrical druglike properties. ACS Chem. Neurosci. 2012, 1 (6), 435−449.

618 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

(56) Vieth, M.; Sutherland, J. J. Dependence of molecular properties (71) Ragsdale, D. S.; McPhee, J. C.; Scheuer, T.; Catterall, W. A.
on proteomic family for marketed oral drugs. J. Med. Chem. 2006, 49 Common molecular determinants of local anesthetic, antiarrhythmic,
(12), 3451−3453. and anticonvulsant block of voltage-gated Na+ channels. Proc. Natl.
(57) Padilla, K.; Wickenden, A. D.; Gerlach, A. C.; McCormack, K. The Acad. Sci. U.S.A. 1996, 93 (17), 9270−9275.
KCNQ2/3 selective channel opener ICA-27243 binds to a novel (72) England, S.; de Groot, M. J. Subtype-selective targeting of voltage-
voltage-sensor domain site. Neurosci. Lett. 2009, 465 (2), 138−142. gated sodium channels. Br. J. Pharmacol. 2009, 158 (6), 1413−1425.
(58) Bauer, C. S.; Tran-Van-Minh, A.; Kadurin, I.; Dolphin, A. C. A (73) Cattabeni, F. Ralfinamide, Newron Pharmaceuticals. IDrugs 2004,
new look at calcium channel α2δ subunits. Curr. Opin. Neurobiol. 2010, 7 (10), 935−939.
20 (5), 563−571. (74) Errington, A. C.; Stohr, T.; Heers, C.; Lees, G. The investigational
(59) Traynelis, S. F.; Wollmuth, L. P.; McBain, C. J.; Menniti, F. S.; anticonvulsant lacosamide selectively enhances slow inactivation of
Vance, K. M.; Ogden, K. K.; Hansen, K. B.; Yuan, H.; Myers, S. J.; voltage-gated sodium channels. Mol. Pharmacol. 2008, 73 (1), 157−169.
Dingledine, R. Glutamate receptor ion channels: structure, regulation, (75) Sheets, P. L.; Heers, C.; Stoehr, T.; Cummins, T. R. Differential
and function. Pharmacol. Rev. 2010, 62 (3), 405−496. block of sensory neuronal voltage-gated sodium channels by lacosamide
(60) Chen, X.; Wang, Q.; Ni, F.; Ma, J. Structure of the full-length [(2R)-2-(acetylamino)-N-benzyl-3-methoxypropanamide], lidocaine,
Shaker potassium channel Kv1.2 by normal-mode-based X-ray crystallo- and carbamazepine. J. Pharmacol. Exp. Ther. 2008, 326 (1), 89−99.
graphic refinement. Proc. Natl. Acad. Sci. U.S.A. 2010, 107 (25), 11352− (76) Zajac, M. A. Process for Preparing α-Carboxamide Pyrrolidine
11357. Derivatives. WO2011029762, March 17, 2011.
(61) (a) Cortright, D. N.; Szallasi, A. TRP channels and pain. Curr. (77) Goldberg, Y. P.; Price, N.; Namdari, R.; Cohen, C. J.; Lamers, M.
Pharm. Des. 2009, 15 (15), 1736−1749. (b) Dorange, I.; Swahn, B. M. H.; Winters, C.; Price, J.; Young, C. E.; Verschoof, H.; Sherrington, R.;
Recent progress in the discovery of Kv7 modulators. Annu. Rep. Med. Pimstone, S. N.; Hayden, M. R. Treatment of Nav1.7-mediated pain in
Chem. 2011, 46, 53−65. (c) Triggle, D. J. Calcium channel antagonists: inherited erythromelalgia using a novel sodium channel blocker. Pain
clinical usespast, present and future. Biochem. Pharmacol. 2007, 74 2012, 153 (1), 80−85.
(1), 1−9. (78) Williams, B. S.; Felix, J. P.; Priest, B. T.; Brochu, R. M.; Dai, K.;
(62) Noda, M.; Ikeda, T.; Suzuki, H.; Takeshima, H.; Takahashi, T.; Hoyt, S. B.; London, C.; Tang, Y. S.; Duffy, J. L.; Parsons, W. H.;
Kuno, M.; Numa, S. Expression of functional sodium channels from Kaczorowski, G. J.; Garcia, M. L. Characterization of a new class of
cloned cDNA. Nature (London, U. K.) 1986, 322 (6082), 826−828. potent inhibitors of the voltage-gated sodium channel Nav1.7.
(63) Long, S. B.; Tao, X.; Campbell, E. B.; MacKinnon, R. Atomic Biochemistry 2007, 46 (50), 14693−14703.
structure of a voltage-dependent K+ channel in a lipid membrane-like (79) Beaudoin, S.; Laufersweiler, M. C.; Markworth, C. J.; Marron, B.
environment. Nature (London, U. K.) 2007, 450 (7168), 376−382. E.; Millan, D. S.; Rawson, D. J.; Reister, S. M.; Sasaki, K.; Storer, R. I.;
(64) Jensen, M. O.; Jogini, V.; Borhani, D. W.; Leffler, A. E.; Dror, R. Stupple, P. A.; Swain, N. A.; West, C. W.; Zhou, S. Preparation of
O.; Shaw, D. E. Mechanism of voltage gating in potassium channels. Heteroarylsulfonamide Derivatives for Use as Analgesics.
Science (Washington, DC, U. S.) 2012, 336 (6078), 229−233. WO2010079443, July 15, 2010.
(65) (a) Tikhonov, D. B.; Zhorov, B. S. Architecture and pore block of (80) Nantermet, P. G.; Henze, D. A. Recent advances toward pain
eukaryotic voltage-gated sodium channels in view of NavAb bacterial therapeutics. Annu. Rep. Med. Chem. 2011, 46, 19−32.
sodium channel structure. Mol. Pharmacol. 2012, 82 (1), 97−104. (81) Aurell, C. J.; Besidski, Y.; Kers, I.; Nyloef, M. Preparation of
(b) Peretz, A.; Pell, L.; Gofman, Y.; Haitin, Y.; Shamgar, L.; Patrich, E.; Chromane Compounds for the Treatment of Pain. WO2012039657,
Kornilov, P.; Gourgy-Hacohen, O.; Ben-Tal, N.; Attali, B. Targeting the March 29, 2012.
voltage sensor of Kv7.2 voltage-gated K+ channels with a new gating- (82) Zimmermann, N.; Grootenhuis, P. D. J.; Numa, M. M. D.;
modifier. Proc. Natl. Acad. Sci. U.S.A. 2010, 107 (35), 15637−15642. Stamos, D.; Anderson, C. D.; Whitney, T. Preparation of Thiazolyl
(c) Casis, O.; Olesen, S. P.; Sanguinetti, M. C. Mechanism of action of a Benzenesulfonamides as Modulators of Ion Channels for Treating Pain,
novel human ether-a-go-go-related gene channel activator. Mol. Nervous System Disorders, and Other Diseases. WO2010002956,
Pharmacol. 2006, 69 (2), 658−665. (d) Lange, W.; Geissendorfer, J.; January 7, 2010.
Schenzer, A.; Grotzinger, J.; Seebohm, G.; Friedrich, T.; Schwake, M. (83) (a) Beaudoin, S.; Johnson, M. S.; Marron, B. E.; Suto, M. J.
Refinement of the binding site and mode of action of the anticonvulsant Sodium Channel Inhibitors. WO2009012241, January 22, 2009.
retigabine on KCNQ K+ channels. Mol. Pharmacol. 2009, 75 (2), 272− (b) Marron, B. E.; Fritch, P. C.; Markworth, C. J.; Maynard, A. T.;
280. Swain, N. A. N-Azolyl Aminocarbonylarylsulfonamide Derivatives as
(66) Moiseenkova-Bell, V. Y.; Stanciu, L. A.; Serysheva, I. I.; Tobe, B. J.; Inhibitors of Ion Channels and Their Preparation, Pharmaceutical
Wensel, T. G. Structure of TRPV1 channel revealed by electron Compositions and Use in the Treatment of Diseases. WO2008118758,
cryomicroscopy. Proc. Natl. Acad. Sci. U.S.A. 2008, 105 (21), 7451− October 2, 2008.
7455. (84) Bagal, S. K.; Gibson, K. R.; Kemp, M. I.; Poinsard, C.; Stammen, B.
(67) Jordt, S. E.; Julius, D. Molecular basis for species-specific L.; Denton, S. M.; Glossop, M. S. Pyridine Derivatives as Voltage-Gated
sensitivity to “hot” chilli peppers. Cell (Cambridge, MA, U. S.) 2002, 108 Sodium Channel Modulators and Their Preparation, Pharmaceutical
(3), 421−430. Compositions and Use in the Treatment of Pain. WO 2008135830,
(68) (a) Picazo-Juarez, G.; Romero-Suarez, S.; Nieto-Posadas, A.; November 13, 2008.
Llorente, I.; Jara-Oseguera, A.; Briggs, M.; McIntosh, T. J.; Simon, S. A.; (85) (a) McGivern Joseph, G. Advantages of voltage-gated ion
Ladron-de-Guevara, E.; Islas, L. D.; Rosenbaum, T. Identification of a channels as drug targets. Expert Opin. Ther. Targets 2007, 11 (3), 265−
binding motif in the S5 helix that confers cholesterol sensitivity to the 271. (b) Castle, N. A. Pharmacological modulation of voltage-gated
TRPV1 ion channel. J. Biol. Chem. 2011, 286 (28), 24966−24976. potassium channels as a therapeutic strategy. Expert Opin. Ther. Pat.
(b) Xiao, B.; Dubin, A. E.; Bursulaya, B.; Viswanath, V.; Jegla, T. J.; 2010, 20 (11), 1471−1503.
Patapoutian, A. Identification of transmembrane domain 5 as a critical (86) Lu, Q.; Peevey, J.; Jow, F.; Monaghan Michael, M.; Mendoza, G.;
molecular determinant of menthol sensitivity in mammalian TRPA1 Zhang, H.; Wu, J.; Kim Callain, Y.; Bicksler, J.; Greenblatt, L.; Lin
channels. J. Neurosci. 2008, 28 (39), 9640−9651. Stephen, S.; Childers, W.; Bowlby Mark, R. Disruption of Kv1.1 N-type
(69) Ogata, N.; Yoshida, S. New perspectives on the structure and inactivation by novel small molecule inhibitors (disinactivators). Bioorg.
function of the Na+ channel multigene family. Curr. Med. Chem.: Cent. Med. Chem. 2008, 16 (6), 3067−3075.
Nerv. Syst. Agents 2002, 2 (1), 59−81. (87) Vennekamp, J.; Wulff, H.; Beeton, C.; Calabresi, P. A.; Grissmer,
(70) Lipkind, G. M.; Fozzard, H. A. A structural model of the S.; Haensel, W.; Chandy, K. G. Kv1.3-blocking 5-phenylalkoxypsor-
tetrodotoxin and saxitoxin binding site of the Na+ channel. Biophys. J. alens: a new class of immunomodulators. Mol. Pharmacol. 2004, 65 (6),
1994, 66 (1), 1−13. 1364−1374.

619 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

(88) Herrington, J.; Solly, K.; Ratliff, K. S.; Li, N.; Zhou, Y.-P.; Howard, (103) Jensen, B. S. BMS-204352: a potassium channel opener
A.; Kiss, L.; Garcia, M. L.; McManus, O. B.; Deng, Q.; Desai, R.; Xiong, developed for the treatment of stroke. CNS Drug Rev. 2002, 8 (4),
Y.; Kaczorowski, G. J. Identification of novel and selective Kv2 channel 353−360.
inhibitors. Mol. Pharmacol. 2011, 80 (6), 959−964. (104) Jensen, B. S.; Jorgensen, T. D.; Ahring, P. K.; Christophersen, P.;
(89) Choi, B. H.; Choi, J. S.; Yoon, S. H.; Rhie, D. J.; Min, D. S.; Jo, Y. Strobaek, D.; Teuber, L.; Olesen, S. P. Use of Isatin Oxime Derivatives as
H.; Kim, M. S.; Hahn, S. J. Effects of norfluoxetine, the major metabolite Ion Channel Activating Agents. WO2000033834, June 15, 2000.
of fluoxetine, on the cloned neuronal potassium channel Kv3.1. (105) Sorensen, U. S.; Strobaek, D.; Christophersen, P.; Hougaard, C.;
Neuropharmacology 2001, 41 (4), 443−453. Jensen, M. L.; Nielsen, E. O.; Peters, D.; Teuber, L. Synthesis and
(90) Selnick, H. G.; Liverton, N. J.; Baldwin, J. J.; Butcher, J. W.; structure−activity relationship studies of 2-(N-substituted)-amino-
Claremon, D. A.; Elliott, J. M.; Freidinger, R. M.; King, S. A.; Libby, B. E.; benzimidazoles as potent negative gating modulators of small
McIntyre, C. J.; Pribush, D. A.; Remy, D. C.; Smith, G. R.; Tebben, A. J.; conductance Ca2+ activated K+ channels. J. Med. Chem. 2008, 51 (23),
Jurkiewicz, N. K.; Lynch, J. J.; Salata, J. J.; Sanguinetti, M. C.; Siegl, P. K. 7625−7634.
S.; Slaughter, D. E.; Vyas, K. Class III antiarrhythmic activity in vivo by (106) Hougaard, C.; Jensen, M. L.; Dale, T. J.; Miller, D. D.; Davies, D.
selective blockade of the slowly activating cardiac delayed rectifier J.; Eriksen, B. L.; Stroebaek, D.; Trezise, D. J.; Christophersen, P.
potassium current IKs by (R)-2-(2,4-trifluoromethyl)-N-[2-oxo-5- Selective activation of the SK1 subtype of human small-conductance
phenyl-1-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-benzo[e][1,4]diazepin- Ca2+ activated K+ channels by 4-(2-methoxy-phenylcarbamoyloxymeth-
3-yl]acetamide. J. Med. Chem. 1997, 40 (24), 3865−3868. yl)-piperidine-1-carboxylic acid tert-butyl ester (GW542573X) is
(91) Rostock, A.; Tober, C.; Rundfeldt, C.; Bartsch, R.; Engel, J.; dependent on serine 293 in the S5 segment. Mol. Pharmacol. 2009, 76
Polymeropoulos, E. E.; Kutscher, B.; Loscher, W.; Honack, D.; White, (3), 569−578.
H. S.; Wolf, H. H. D-23129: a new anticonvulsant with a broad spectrum (107) Eriksen, B. L.; Soerensen, U. S.; Hougaard, C.; Peters, D.;
activity in animal models of epileptic seizures. Epilepsy Res. 1996, 23 (3), Johansen, T. H.; Christophersen, P. Preparation of Purine Derivatives
211−223. and Their Use as Potassium Channel Modulators. WO2008116910,
(92) Jakovlev, V.; Sofia, R. D.; Achterrath-Tuckermann, U.; von October 2, 2008.
Schlichtegroll, A.; Thiemer, K. The pharmacologic effect of flupirtine, a (108) Liegeois, J. F.; Mercier, F.; Graulich, A.; Graulich-Lorge, F.;
structurally new analgesic. Arzneim.-Forsch. 1985, 35 (1), 30−43. Scuvee-Moreau, J.; Seutin, V. Modulation of small conductance calcium-
(93) Zhou, P.; Babcock, J.; Liu, L.-q.; Li, M.; Gao, Z. Activation of activated potassium (SK) channels: a new challenge in medicinal
human ether-a-go-go related gene (hERG) potassium channels by small chemistry. Curr. Med. Chem. 2003, 10 (8), 625−647.
molecules. Acta Pharmacol. Sin. 2011, 32 (6), 781−788. (109) (a) Ataga, K. I.; Smith, W. R.; De Castro, L. M.; Swerdlow, P.;
(94) Pexton, T.; Moeller-Bertram, T.; Schilling, J. M.; Wallace, M. S. Saunthararajah, Y.; Castro, O.; Vichinsky, E.; Kutlar, A.; Orringer, E. P.;
Targeting voltage-gated calcium channels for the treatment of Rigdon, G. C.; Stocker, J. W. Efficacy and safety of the Gardos channel
neuropathic pain: a review of drug development. Expert Opin. Invest.
blocker, senicapoc (ICA-17043), in patients with sickle cell anemia.
Drugs 2011, 20 (9), 1277−1284.
Blood 2008, 111 (8), 3991−3997. (b) Nardi, A.; Demnitz, J.; Garcia, M.
(95) Yamamoto, T.; Takahara, A. Recent updates of N-type calcium
L.; Polosa, R. Potassium channels as drug targets for therapeutic
channel blockers with therapeutic potential for neuropathic pain and
intervention in respiratory diseases. Expert Opin. Ther. Pat. 2008, 18
stroke. Curr. Top. Med. Chem. 2009, 9 (4), 377−395.
(12), 1361−1384.
(96) McGivern, J. G. Ziconotide: a review of its pharmacology and use
(110) Nilius, B. TRP channels in disease. Biochim. Biophys. Acta 2007,
in the treatment of pain. Neuropsychiatric Dis. Treat. 2007, 3 (1), 69−85.
(97) (a) Yamamoto, T.; Niwa, S.; Tokumasu, M.; Onishi, T.; Ohno, S.; 1772 (8), 805−812.
Hagihara, M.; Koganei, H.; Fujita, S.; Takeda, T.; Saitou, Y.; Iwayama, S.; (111) Bakthavatchalam, R.; Kimball, S. D. Modulators of transient
Takahara, A.; Iwata, S.; Shoji, M. Discovery and evaluation of selective receptor potential ion channels. Annu. Rep. Med. Chem. 2010, 45, 37−53.
N-type calcium channel blockers: 6-unsubstituted-1,4-dihydropyridine- (112) Preti, D.; Szallasi, A.; Patacchini, R. TRP channels as therapeutic
5-carboxylic acid derivatives. Bioorg. Med. Chem. Lett. 2012, 22 (11), targets in airway disorders: a patent review. Expert Opin. Ther. Pat. 2012,
3639−3642. (b) Knutsen, L. J. S.; Hobbs, C. J.; Earnshaw, C. G.; 22 (6), 663−695.
Fiumana, A.; Gilbert, J.; Mellor, S. L.; Radford, F.; Smith, N. J.; Birch, P. (113) (a) Gavva, N. R. Setbacks in the clinical development of TRPV1
J.; Russell Burley, J.; Ward, S. D. C.; James, I. F. Synthesis and SAR of antagonists: what next? Open Drug Discovery J. 2009, 1, 1−35. (b) Viana,
novel 2-arylthiazolidinones as selective analgesic N-type calcium F.; Ferrer-Montiel, A. TRPA1 modulators in preclinical development.
channel blockers. Bioorg. Med. Chem. Lett. 2007, 17 (3), 662−667. Expert Opin. Ther. Pat. 2009, 19 (12), 1787−1799.
(c) Tyagarajan, S.; Chakravarty, P. K.; Park, M.; Zhou, B.; Herrington, J. (114) Gavva, N. R.; Bannon, A. W.; Hovland, D. N., Jr.; Lehto, S. G.;
B.; Ratliff, K.; Bugianesi, R. M.; Williams, B.; Haedo, R. J.; Swensen, A. Klionsky, L.; Surapaneni, S.; Immke, D. C.; Henley, C.; Arik, L.; Bak, A.;
M.; Warren, V. A.; Smith, M.; Garcia, M.; Kaczorowski, G. J.; McManus, Davis, J.; Ernst, N.; Hever, G.; Kuang, R.; Shi, L.; Tamir, R.; Wang, J.;
O. B.; Lyons, K. A.; Li, X.; Madeira, M.; Karanam, B.; Green, M.; Forrest, Wang, W.; Zajic, G.; Zhu, D.; Norman, M. H.; Louis, J.-C.; Magal, E.;
M. J.; Abbadie, C.; McGowan, E.; Mistry, S.; Jochnowitz, N.; Duffy, J. L. Treanor, J. J. S. Repeated administration of vanilloid receptor TRPV1
A potent and selective indole N-type calcium channel (Cav2.2) blocker antagonists attenuates hyperthermia elicited by TRPV1 blockade. J.
for the treatment of pain. Bioorg. Med. Chem. Lett. 2011, 21 (2), 869− Pharmacol. Exp. Ther. 2007, 323 (1), 128−137.
873. (115) Rami, H. K.; Thompson, M.; Stemp, G.; Fell, S.; Jerman, J. C.;
(98) Heer, J. P.; Norton, D.; Ward, S. E. Preparation of Pyrazolo[1,5- Stevens, A. J.; Smart, D.; Sargent, B.; Sanderson, D.; Randall, A. D.;
a]pyrimidine Derivatives for Blocking Cav2.2 Calcium Channels. Gunthorpe, M. J.; Davis, J. B. Discovery of SB-705498: a potent,
WO2010007074, January 21, 2010. selective and orally bioavailable TRPV1 antagonist suitable for clinical
(99) Snutch, T. P. Synthesis and Activity of Substituted Piperazine and development. Bioorg. Med. Chem. Lett. 2006, 16 (12), 3287−3291.
Piperidine Calcium Channel Blockers. WO2001045709, June 28, 2001. (116) Gomtsyan, A.; Bayburt, E. K.; Schmidt, R. G.; Surowy, C. S.;
(100) Ford, R. V.; Nickell, J. The clinical pharmacological investigation Honore, P.; Marsh, K. C.; Hannick, S. M.; McDonald, H. A.; Wetter, J.
of hydroflumethiazide (Saluron). Antibiot. Med. Clin. Ther. 1959, 6, M.; Sullivan, J. P.; Jarvis, M. F.; Faltynek, C. R.; Lee, C.-H. Identification
461−469. of (R)-1-(5-tert-butyl-2,3-dihydro-1H-inden-1-yl)-3-(1H-indazol-4-yl)-
(101) Young, D. S.; Forrester, T. M.; Morgan, T. N. A comparison of urea (ABT-102) as a potent TRPV1 antagonist for pain management. J.
the diuretic action of chlorthiazide analogs. Lancet 1959, 1959-II, 765− Med. Chem. 2008, 51 (3), 392−395.
768. (117) Bakthavatchatam, R.; Blum, C. A.; Brielmann, H. L.; Caldwell, T.
(102) Hewawasam, P.; Meanwell, N. A.; Gribkoff, V. K. 3-Substituted M.; De Lombaert, S. Preparation of Substituted Quinazolin-4-ylamine
Oxindole Derivatives ss Potassium Channel Modulators, and Prepara- Analogs as VR1 Capsaicin Receptor Antagonists for Relieving Pain.
tion and Therapeutic Use Thereof. US5602169, February 11, 1997. WO2003062209, July 31, 2003.

620 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

(118) (a) Hinman, A.; Chuang, H.; Bautista, D. M.; Julius, D. TRP Wells, K. M.; Youells, S. C.; Youngman, M. A. N-Benzothienyl
channel activation by reversible covalent modification. Proc. Natl. Acad. Sulfonamides as TRPM8 Modulators and Their Preparation,
Sci. U.S.A. 2006, 103 (51), 19564−19568. (b) Macpherson, L. J.; Dubin, Pharmaceutical Compositions and Use in the Treatment of Diseases.
A. E.; Evans, M. J.; Marr, F.; Schultz, P. G.; Cravatt, B. F.; Patapoutian, A. WO2009012430, January 22, 2009.
Noxious compounds activate TRPA1 ion channels through covalent (129) Hibbs, R. E.; Gouaux, E. Principles of activation and permeation
modification of cysteines. Nature (London, U. K.) 2007, 445 (7127), in an anion-selective Cys-loop receptor. Nature (London, U. K.) 2011,
541−545. 474 (7349), 54−60.
(119) Eid, S. R.; Crown, E. D.; Moore, E. L.; Liang, H. A.; Choong, K.- (130) Unwin, N. Refined structure of the nicotinic acetylcholine
C.; Dima, S.; Henze, D. A.; Kane, S. A.; Urban, M. O. HC-030031, a receptor at 4Å resolution. J. Mol. Biol. 2005, 346 (4), 967−989.
TRPA1 selective antagonist, attenuates inflammatory- and neuropathy- (131) (a) Bourne, Y.; Talley, T. T.; Hansen, S. B.; Taylor, P.; Marchot,
induced mechanical hypersensitivity. Mol. Pain 2008, 4 (1), 48/1−48/ P. Crystal structure of a Cbtx-AChBP complex reveals essential
10. interactions between snake alpha-neurotoxins and nicotinic receptors.
(120) Ng, H.; Weigele, M.; Moran, M.; Chong, J.; Fanger, C.; Larsen, EMBO J. 2005, 24 (8), 1512−1522. (b) Brejc, K.; van Dijk, W. J.;
G. R.; Del Camino, D.; Hayward, N.; Adams, S.; Ripka, A. Preparation of Klaassen, R. V.; Schuurmans, M.; van Der Oost, J.; Smit, A. B.; Sixma, T.
Purine Nucleobases via Coupling Reaction for Treating Disorders K. Crystal structure of an ACh-binding protein reveals the ligand-
Related to TRPA1. WO2009002933, December 31, 2008. binding domain of nicotinic receptors. Nature (London, U. K.) 2001, 411
(121) (a) Chong, J. A.; Fanger, C.; Moran, M. M.; Underwood, D. J.; (6835), 269−276.
Zhen, X.; Ripka, A.; Weigele, M.; Lumma, W. C.; Larsen, G. R. (132) Sieghart, W. Structure, pharmacology, and function of GABAA
Benzothiazole Derivatives and Related Compounds for Modulating receptor subtypes. Adv. Pharmacol. 2006, 54, 231−263.
TRPV3 Function and Their Preparation, Pharmaceutical Compositions (133) Dawson, G. R.; Maubach, K. A.; Collinson, N.; Cobain, M.;
and Their Use for Treatment of Pain. WO2006122156, November 16, Everitt, B. J.; MacLeod, A. M.; Choudhury, H. I.; McDonald, L. M.;
2006. (b) Chong, J. A.; Fanger, C.; Larsen, G. R.; Lumma, W. C.; Moran, Pillai, G.; Rycroft, W.; Smith, A. J.; Sternfeld, F.; Tattersall, F. D.;
M. M.; Ripka, A.; Underwood, D. J.; Weigele, M.; Zhen, X. Wafford, K. A.; Reynolds, D. S.; Seabrook, G. R.; Atack, J. R. An inverse
Dihydroquinazolinone Compounds for Modulating TRPV3 Function agonist selective for α5 subunit-containing GABAA receptors enhances
and Their Preparation, Pharmaceutical Compositions and Use in the cognition. J. Pharmacol. Exp. Ther. 2006, 316 (3), 1335−1345.
Treatment of Pain and Related Disorders. WO2007056124, May 18, (134) Atack, J. R.; Wafford, K. A.; Tye, S. J.; Cook, S. M.; Sohal, B.;
2007. (c) Lingam, P.; Thomas, A.; Gharat, L. A.; Ukirde, D. V.; Mindhe, Pike, A.; Sur, C.; Melillo, D.; Bristow, L.; Bromidge, F.; Ragan, I.; Kerby,
A. S.; Khairatkar Joshi, N.; Kattige, V. G.; Phatangare, S. K. Preparation J.; Street, L.; Carling, R.; Castro, J. L.; Whiting, P.; Dawson, G. R.;
of Chromane Derivatives as Vanilloid TRPV3 Modulators. McKernan, R. M. TPA023 [7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-
WO2009084034, July 9, 2009. (d) Lingam, P.; Thomas, A.; More, D. triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]-
A.; Khatik, J. Y.; Khairatkar, J. N.; Kattige, V. G. Preparation of Fused pyridazine], an agonist selective for α2- and α3-containing GABAA
Pyrimidine Derivatives as TRPV3 Modulators. WO2009109987, receptors, is a nonsedating anxiolytic in rodents and primates. J.
September 11, 2009. Pharmacol. Exp. Ther. 2006, 316 (1), 410−422.
(122) Glenmark Pharmaceuticals. Research Pipeline. http://www. (135) Atack, J. R. The benzodiazepine binding site of GABA(A)
glenmarkpharma.com/UITemplate/HtmlContainer.aspx?res=P_ receptors as a target for the development of novel anxiolytics. Expert
GLN_GDY_FLIN. (accessed August 4, 2012). Opin. Invest. Drugs 2005, 14 (5), 601−618.
(123) Thorneloe, K. S.; Sulpizio, A. C.; Lin, Z.; Figueroa, D. J.; Clouse, (136) Sanna, E.; Busonero, F.; Talani, G.; Carta, M.; Massa, F.; Peis,
A. J.; McCafferty, G. P.; Chendrimada, T. P.; Lashinger, E. S. R.; M.; Maciocco, E.; Biggio, G. Comparison of the effects of zaleplon,
Gordon, E. A.; Evans, L.; Misajet, B. A.; DeMarini, D. J.; Nation, J. H.; zolpidem, and triazolam at various GABAA receptor subtypes. Eur. J.
Casillas, L. N.; Marquis, R. W.; Votta, B. J.; Sheardown, S. A.; Xu, X.; Pharmacol. 2002, 451 (2), 103−110.
Brooks, D. P.; Laping, N. J.; Westfall, T. D. GSK1016790A, a Novel and (137) Atack, J. R. GABAA receptor subtype-selective modulators. I.
Potent TRPV4 Channel Agonist Induces Urinary Bladder Contraction α2α3-Selective agonists as non-sedating anxiolytics. Curr. Top. Med.
and Hyperactivity. J. Pharmacol. Exp. Ther. 2008, 326 (2), 432−442. Chem. 2011, 11 (9), 1176−1202.
(124) Vincent, F.; Acevedo, A.; Nguyen, M. T.; Dourado, M.; DeFalco, (138) Griebel, G.; Perrault, G.; Simiand, J.; Cohen, C.; Granger, P.;
J.; Gustafson, A.; Spiro, P.; Emerling, D. E.; Kelly, M. G.; Duncton, M. A. Decobert, M.; Francon, D.; Avenet, P.; Depoortere, H.; Tan, S.; Oblin,
Identification and characterization of novel TRPV4 modulators. A.; Schoemaker, H.; Evanno, Y.; Sevrin, M.; George, P.; Scatton, B.
Biochem. Biophys. Res. Commun. 2009, 389 (3), 490−494. SL651498: an anxioselective compound with functional selectivity for
(125) Everaerts, W.; Zhen, X.; Ghosh, D.; Vriens, J.; Gevaert, T.; alpha2- and alpha3-containing gamma-aminobutyric acid(A) (GABA-
Gilbert, J. P.; Hayward, N. J.; McNamara, C. R.; Xue, F.; Moran, M. M.; (A)) receptors. J. Pharmacol. Exp. Ther. 2001, 298 (2), 753−768.
Strassmaier, T.; Uykal, E.; Owsianik, G.; Vennekens, R.; De Ridder, D.; (139) McKernan, R. M.; Rosahl, T. W.; Reynolds, D. S.; Sur, C.;
Nilius, B.; Fanger, C. M.; Voets, T. Inhibition of the cation channel Wafford, K. A.; Atack, J. R.; Farrar, S.; Myers, J.; Cook, G.; Ferris, P.;
TRPV4 improves bladder function in mice and rats with cyclo- Garrett, L.; Bristow, L.; Marshall, G.; Macaulay, A.; Brown, N.; Howell,
phosphamide-induced cystitis. Proc. Natl. Acad. Sci. U.S.A. 2010, 107 O.; Moore, K. W.; Carling, R. W.; Street, L. J.; Castro, J. L.; Ragan, C. I.;
(44), 19084−19089. Dawson, G. R.; Whiting, P. J. Sedative but not anxiolytic properties of
(126) (a) Dhaka, A.; Murray, A. N.; Mathur, J.; Earley, T. J.; Petrus, M. benzodiazepines are mediated by the GABAA receptor α1 subtype. Nat.
J.; Patapoutian, A. TRPM8 is required for cold sensation in mice. Neuron Neurosci. 2000, 3 (6), 587−592.
2007, 54 (3), 371−378. (b) Bautista, D. M.; Siemens, J.; Glazer, J. M.; (140) Crawforth, J. M.; Gibson, K. R.; Rowley, M. Preparation of
Tsuruda, P. R.; Basbaum, A. I.; Stucky, C. L.; Jordt, S.; Julius, D. The Tricyclic Pyridin-2-one Analogue as a GABA Receptor Ligand.
menthol receptor TRPM8 is the principal detector of environmental WO20010053776, July 26, 2001.
cold. Nature (London, U. K.) 2007, 448 (7150), 204−208. (141) McNamara, R. K.; dePape, G. E.; Skelton, R. W. Differential
(127) Dendreon. Research and Development. http://www.dendreon. effects of benzodiazepine receptor agonists on hippocampal long-term
com/research-development/trpm8 (accessed August 12 2012). potentiation and spatial learning in the Morris water maze. Brain Res.
(128) (a) Lashinger, E. S. R.; Steiginga, M. S.; Hieble, J. P.; Leon, L. A.; 1993, 626 (1−2), 63−70.
Gardner, S. D.; Nagilla, R.; Davenport, E. A.; Hoffman, B. E.; Laping, N. (142) (a) Chambers, M. S.; Atack, J. R.; Broughton, H. B.; Collinson,
J.; Su, X. AMTB, a TRPM8 channel blocker: evidence in rats for activity N.; Cook, S.; Dawson, G. R.; Hobbs, S. C.; Marshall, G.; Maubach, K. A.;
in overactive bladder and painful bladder syndrome. Am. J. Physiol. 2008, Pillai, G. V.; Reeve, A. J.; MacLeod, A. M. Identification of a novel,
295 (3), F803−F810. (b) Branum, S. T.; Colburn, R. W.; Dax, S. L.; selective GABAA α5 receptor inverse agonist which enhances cognition.
Flores, C. M.; Jetter, M. C.; Liu, Y.; Ludovici, D.; Macielag, M. J.; J. Med. Chem. 2003, 46 (11), 2227−2240. (b) Sternfeld, F.; Carling, R.
Matthews, J. M.; McNally, J. J.; Reaney, L. M.; Russell, R. K.; Qin, N.; W.; Jelley, R. A.; Ladduwahetty, T.; Merchant, K. J.; Moore, K. W.;

621 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

Reeve, A. J.; Street, L. J.; O’Connor, D.; Sohal, B.; Atack, J. R.; Cook, S.; (157) Bowie, D. Ionotropic glutamate receptors & CNS disorders.
Seabrook, G.; Wafford, K.; Tattersall, F. D.; Collinson, N.; Dawson, G. CNS Neurol. Disord.: Drug Targets 2008, 7 (2), 129−143.
R.; Castro, J. L.; MacLeod, A. M. Selective, orally active α-aminobutyric (158) (a) Kaczor, A. A.; Matosiuk, D. Molecular structure of ionotropic
acid-A α5 receptor inverse agonists as cognition enhancers. J. Med. glutamate receptors. Curr. Med. Chem. 2010, 17 (24), 2608−2635.
Chem. 2004, 47 (9), 2176−2179. (b) Mayer, M. L. Structure and mechanism of glutamate receptor ion
(143) (a) Arias, H. R. Localization of agonist and competitive channel assembly, activation and modulation. Curr. Opin. Neurobiol.
antagonist binding sites on nicotinic acetylcholine receptors. Neurochem. 2011, 21 (2), 283−290.
Int. 2000, 36 (7), 595−645. (b) Arias, H. R.; Bhumireddy, P.; Bouzat, C. (159) Stawski, P.; Janovjak, H.; Trauner, D. Pharmacology of
Molecular mechanisms and binding site locations for noncompetitive ionotropic glutamate receptors: a structural perspective. Bioorg. Med.
antagonists of nicotinic acetylcholine receptors. Int. J. Biochem. Cell Biol. Chem. 2010, 18 (22), 7759−7772.
2006, 38 (8), 1254−1276. (160) Sobolevsky, A. I.; Rosconi, M. P.; Gouaux, E. X-ray structure,
(144) Gotti, C.; Riganti, L.; Vailati, S.; Clementi, F. Brain neuronal symmetry and mechanism of an AMPA-subtype glutamate receptor.
nicotinic receptors as new targets for drug discovery. Curr. Pharm. Des. Nature (London, U. K.) 2009, 462 (7274), 745−756.
2006, 12 (4), 407−428. (161) Stensbol, T. B.; Jensen, H. S.; Nielsen, B.; Johansen, T. N.;
(145) Spinelli, S.; Ballard, T.; Feldon, J.; Higgins, G. A.; Pryce, C. R. Egebjerg, J.; Frydenvang, K.; Krogsgaard-Larsen, P. Stereochemistry and
Enhancing effects of nicotine and impairing effects of scopolamine on molecular pharmacology of (S)-thio-ATPA, a new potent and selective
distinct aspects of performance in computerized attention and working GluR5 agonist. Eur. J. Pharmacol. 2001, 411 (3), 245−253.
memory tasks in marmoset monkeys. Neuropharmacology 2006, 51 (2), (162) Wang, L. A.; Mayer, M. L.; Jane, D. E.; Watkins, J. C. Willardiines
238−250. differentiate agonist binding sites for kainate- versus AMPA-preferring
(146) Koren, A. O.; Horti, A. G.; Mukhin, A. G.; Guendisch, D.; Kimes, glutamate receptors in DRG and hippocampal neurons. J. Neurosci.
A. S.; Dannals, R. F.; London, E. D. 2-, 5-, and 6-Halo-3-(2(S)- 1994, 14 (6), 3881−3897.
azetidinylmethoxy)pyridines: synthesis, affinity for nicotinic acetylcho- (163) Pedregal, C.; Collado, I.; Escribano, A.; Ezquerra, J.; Dominguez,
line receptors, and molecular modelling. J. Med. Chem. 1998, 41 (19), C.; Mateo, A. I.; Rubio, A.; Baker, S. R.; Goldsworthy, J.; Kamboj, R. K.;
3690−3698. Ballyk, B. A.; Hoo, K.; Bleakman, D. 4-Alkyl- and 4-cinnamylglutamic
(147) Caldwell, W. S.; Dull, G. M.; Bhatti, B. S.; Hadimani, S. B.; Park, acid analogues are potent GluR5 kainate receptor agonists. J. Med. Chem.
H.; Wagner, J. M.; Crooks, P. A.; Lippiello, P. M.; Bencherif, M. 2000, 43 (10), 1958−1968.
Preparation of Aryl Substituted Olefinic Amines and Their Use as (164) Shou, X.; Chamberlin, A. R. Ligands for kainate subtype
Cholinergic Receptors Agonists. WO9965876, December 23, 1999. glutamate receptors. Expert Opin. Ther. Pat. 2004, 14 (4), 471−486.
(148) Briggs, C. A.; Anderson, D. J.; Brioni, J. D.; Buccafusco, J. J.; (165) Gilron, I. Therapeutic potential of the AMPA/kainate receptor
Buckley, M. J.; Campbell, J. E.; Decker, M. W.; Donnelly-Roberts, D.; antagonist LY293558 in the management of pain and other central
nervous system disorders. Curr. Opin. Invest. Drugs 2001, 2 (9), 1273−
Elliott, R. L.; Gopalakrishnan, M.; Holladay, M. W.; Hui, Y. H.; Jackson,
1278.
W. J.; Kim, D. J.; Marsh, K. C.; O’Neill, A.; Prendergast, M. A.; Ryther, K.
(166) Baron, B. M.; Harrison, B. L.; Miller, F. P.; McDonald, I. A.;
B.; Sullivan, J. P.; Arneric, S. P. Functional characterization of the novel
Salituro, F. G.; Schmidt, C. J.; Sorensen, S. M.; White, H. S.; Palfreyman,
neuronal nicotinic acetylcholine receptor ligand GTS-21 in vitro and in
M. G. Activity of 5,7-dichlorokynurenic acid, a potent antagonist at the
vivo. Pharmacol., Biochem. Behav. 1997, 57 (1−2), 231−241.
N-methyl-D-aspartate receptor-associated glycine binding site. Mol.
(149) Bodnar, A. L.; Cortes-Burgos, L. A.; Cook, K. K.; Dinh, D. M.;
Pharmacol. 1990, 38 (4), 554−561.
Groppi, V. E.; Hajos, M.; Higdon, N. R.; Hoffmann, W. E.; Hurst, R. S.; (167) Danysz, W.; Fadda, E.; Wroblewski, J. T.; Costa, E. Kynurenate
Myers, J. K.; Rogers, B. N.; Wall, T. M.; Wolfe, M. L.; Wong, E. and 2-amino-5-phosphonovalerate interact with multiple binding sites
Discovery and structure−activity relationship of quinuclidine benza- of the N-methyl-D-aspartate-sensitive glutamate receptor domain.
mides as agonists of alpha7 nicotinic acetylcholine receptors. J. Med. Neurosci. Lett. 1989, 96 (3), 340−344.
Chem. 2005, 48 (4), 905−908. (168) (a) Tarnawa, I.; Berzsenyi, P.; Andrasi, F.; Botka, P.; Hamori, T.;
(150) Haydar, S. N.; Dunlop, J. Neuronal nicotinic acetylcholine Ling, I.; Korosi, J. Structure−activity relationships of 2,3-benzodiazepine
receptorstargets for the development of drugs to treat cognitive compounds with glutamate antagonistic action. Bioorg. Med. Chem. Lett.
impairment associated with schizophrenia and Alzheimer’s disease. Curr. 1993, 3 (1), 99−104. (b) Chimirri, A.; De Luca, L.; Ferro, S.; De Sarro,
Top. Med. Chem. 2010, 10 (2), 144−152. G.; Ciranna, L.; Gitto, R. Combined strategies for the discovery of
(151) Taniguchi, S.; Watanabe, F.; Hagihara, K. Pharmacological ionotropic glutamate receptor antagonists. ChemMedChem 2009, 4 (6),
characterization of indisetron hydrochloride, as a novel 5-HT3 receptor 917−922.
antagonist. Jpn. Pharmacol. Ther. 2004, 32 (11), 789−797. (169) Quirk, J. C.; Nisenbaum, E. S. LY404187: a novel positive
(152) Miyata, K.; Kamato, T.; Nishida, A.; Ito, H.; Katsuyama, Y.; Iwai, allosteric modulator of AMPA receptors. CNS Drug Rev. 2002, 8 (3),
A.; Yuki, H.; Yamano, M.; Tsutsumi, R.; Ohta, M.; Takeda, M.; Honda, 255−282.
K. Pharmacologic profile of (R)-5-[(1-methyl-3-indolyl)carbonyl]- (170) Gogas, K. R. Glutamate-based therapeutic approaches: NR2B
4,5,6,7-tetrahydro-1H-benzimidazole hydrochloride (YM060), a potent receptor antagonists. Curr. Opin. Pharmacol. 2006, 6 (1), 68−74.
and selective 5-hydroxytryptamine3 receptor antagonist, and its (171) Reynolds, I. J.; Miller, R. J. Ifenprodil is a novel type of N-methyl-
enantiomer in the isolated tissue. J. Pharmacol. Exp. Ther. 1991, 259 D-aspartate receptor antagonist: interaction with polyamines. Mol.
(1), 15−21. Pharmacol. 1989, 36 (5), 758−765.
(153) Jones, B. J.; Costall, B.; Domeney, A. M.; Kelly, M. E.; Naylor, R. (172) Chenard, B. L.; Bordner, J.; Butler, T. W.; Chambers, L. K.;
J.; Oakley, N. R.; Tyers, M. B. The potential anxiolytic activity of Collins, M. A.; De Costa, D. L.; Ducat, M. F.; Dumont, M. L.; Fox, C. B.;
GR38032F, a 5-HT3-receptor antagonist. Br. J. Pharmacol. 1988, 93 (4), Mena, E. E.; Menniti, F. S.; Nielsen, J.; Pagnozzi, M. J.; Richter, K. E. G.;
985−993. Ronau, R. T.; Shalaby, I. A.; Stemple, J. Z.; White, W. F. (1S,2S)-1-(4-
(154) Wong, E. H.; Clark, R.; Leung, E.; Loury, D.; Bonhaus, D. W.; Hydroxyphenyl)-2-(4-hydroxy-4-phenylpiperidino)-1-propanol: a po-
Jakeman, L.; Parnes, H.; Whiting, R. L.; Eglen, R. M. The interaction of tent new neuroprotectant which blocks N-methyl-D-aspartate responses.
RS 25259-197, a potent and selective antagonist, with 5-HT3 receptors, J. Med. Chem. 1995, 38 (16), 3138−3145.
in vitro. Br. J. Pharmacol. 1995, 114 (4), 851−859. (173) Hattori, M.; Gouaux, E. Molecular mechanism of ATP binding
(155) Sato, N.; Sakamori, M.; Haga, K.; Takehara, S.; Setoguchi, M. and ion channel activation in P2X receptors. Nature (London, U. K.)
Antagonistic activity of Y-25130 on 5-HT3 receptors. Jpn. J. Pharmacol. 2012, 485 (7397), 207−212.
1992, 59 (4), 443−448. (174) Wemmie, J. A.; Price, M. P.; Welsh, M. J. Acid-sensing ion
(156) Chong, Y.; Choo, H. 5-HT(3) antagonists under development. channels: advances, questions and therapeutic opportunities. Trends
Expert Opin. Invest. Drugs 2010, 19 (11), 1309−1319. Neurosci. 2006, 29 (10), 578−586.

622 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

(175) Lingueglia, E. Acid-sensing ion channels in sensory perception. J. (190) Duplantier, A. J.; Dombroski, M. A.; Subramanyam, C.; Beaulieu,
Biol. Chem. 2007, 282 (24), 17325−17329. A. M.; Chang, S.; Gabel, C. A.; Jordan, C.; Kalgutkar, A. S.; Kraus, K. G.;
(176) Wu, W.; Cheng, C.; Sun, W.; Wong, C.; Chen, C. Targeting Labasi, J. M.; Mussari, C.; Perregaux, D. G.; Shepard, R.; Taylor, T. J.;
ASIC3 for pain, anxiety, and insulin resistance. Pharmacol. Ther. 2012, Trevena, K. A.; Whitney-Pickett, C.; Yoon, K. Optimization of the
134 (2), 127−138. physicochemical and pharmacokinetic attributes in a 6-azauracil series of
(177) Xiong, Z.; Pignataro, G.; Li, M.; Chang, S.; Simon, R. P. Acid- P2X7 receptor antagonists leading to the discovery of the clinical
sensing ion channels (ASICs) as pharmacological targets for neuro- candidate CE-224,535. Bioorg. Med. Chem. Lett. 2011, 21 (12), 3708−
degenerative diseases. Curr. Opin. Pharmacol. 2008, 8 (1), 25−32. 3711.
(178) Dube, G. R.; Lehto, S. G.; Breese, N. M.; Baker, S. J.; Wang, X.; (191) Kelly, M. G.; Kincaid, J.; Fang, Y.; Cao, Y.; Kaub, C.; Gowlugari,
Matulenko, M. A.; Honore, P.; Stewart, A. O.; Moreland, R. B.; Brioni, J. S.; Wang, Z. Preparation of Isoquinolonecarboxamides as P2X7
D. Electrophysiological and in vivo characterization of A-317567, a Purinoceptor Modulators and Uses Thereof. WO2008112205,
novel blocker of acid sensing ion channels. Pain 2005, 117 (1−2), 88− September 18, 2008.
96. (192) Chambers, L. J.; Stevens, A. J.; Moses, A. P.; Michel, A. D.;
(179) Kuduk, S. D.; Chang, R. K.; Wai, J. M. C.; Di Marco, C. N.; Cofre, Walter, D. S.; Davies, D. J.; Livermore, D. G.; Fonfria, E.; Demont, E. H.;
V.; Di Pardo, R. M.; Cook, S. P.; Cato, M. J.; Jovanovska, A.; Urban, M.
Vimal, M.; Theobald, P. J.; Beswick, P. J.; Gleave, R. J.; Roman, S. A.;
O.; Leitl, M.; Spencer, R. H.; Kane, S. A.; Hartman, G. D.; Bilodeau, M.
Senger, S. Synthesis and structure−activity relationships of a series of
T. Amidine derived inhibitors of acid-sensing ion channel-3 (ASIC3).
(1H-pyrazol-4-yl)acetamide antagonists of the P2X7 receptor. Bioorg.
Bioorg. Med. Chem. Lett. 2009, 19 (15), 4059−4063.
(180) Kuduk, S. D.; Chang, R. K.; Di Marco, C. N.; Dipardo, R. M.; Med. Chem. Lett. 2010, 20 (10), 3161−3164.
Cook, S. P.; Cato, M. J.; Jovanovska, A.; Urban, M. O.; Leitl, M.; (193) (a) Keystone, E. C.; Wang, M. M.; Layton, M.; Hollis, S.;
Spencer, R. H.; Kane, S. A.; Hartman, G. D.; Bilodeau, M. T. McInnes, I. B. Clinical evaluation of the efficacy of the P2X7 purinergic
Identification of non-amidine inhibitors of acid-sensing ion channel-3 receptor antagonist AZD9056 on the signs and symptoms of
(ASIC3). Bioorg. Med. Chem. Lett. 2011, 21 (14), 4255−4258. rheumatoid arthritis in patients with active disease despite treatment
(181) Khakh, B. S.; North, R. A. P2X receptors as cell-surface ATP with methotrexate or sulphasalazine. Ann. Rheum. Dis. 2012, 71 (10),
sensors in health and disease. Nature (London, U. K.) 2006, 442 (7102), 1630−1635. (b) Stock, T. C.; Bloom, B. J.; Wei, N.; Ishaq, S.; Park, W.;
527−532. Wang, X.; Gupta, P.; Mebus, C. A. Efficacy and safety of CE-224,535, an
(182) (a) Guile, S. D.; Ince, F.; Ingall, A. H.; Kindon, N. D.; Meghani, antagonist of P2X7 receptor, in treatment of patients with rheumatoid
P.; Mortimore, M. P. The medicinal chemistry of the P2 receptor family. arthritis inadequately controlled by methotrexate. J. Rheumatol. 2012, 39
Prog. Med. Chem. 2001, 38, 115−187. (b) Jacobson, K. A.; Jarvis, M. F.; (4), 720−727.
Williams, M. Purine and pyrimidine (P2) receptors as drug targets. J. (194) Clarke, O. B.; Caputo, A. T.; Hill, A. P.; Vandenberg, J. I.; Smith,
Med. Chem. 2002, 45 (19), 4057−4093. (c) Gunosewoyo, H.; Kassiou, B. J.; Gulbis, J. M. Domain reorientation and rotation of an intracellular
M. P2X purinergic receptor ligands: recently patented compounds. assembly regulate conduction in Kir potassium channels. Cell (Cam-
Expert Opin. Ther. Pat. 2010, 20 (5), 625−646. bridge, MA, U. S.) 2010, 141 (6), 1018−1029.
(183) Jarvis, M. F.; Burgard, E. C.; McGaraughty, S.; Honore, P.; (195) Mathie, A.; Al-Moubarak, E.; Veale, E. L. Gating of two pore
Lynch, K.; Brennan, T. J.; Subieta, A.; Van Biesen, T.; Cartmell, J.; domain potassium channels. J. Physiol. (Oxford, U. K.) 2010, 588 (17),
Bianchi, B.; Niforatos, W.; Kage, K.; Yu, H.; Mikusa, J.; Wismer, C. T.; 3149−3156.
Zhu, C. Z.; Chu, K.; Lee, C.; Stewart, A. O.; Polakowski, J.; Cox, B. F.; (196) Danthi, S.; Enyeart, J. A.; Enyeart, J. J. Caffeic acid esters activate
Kowaluk, E.; Williams, M.; Sullivan, J.; Faltynek, C. A-317491, a novel TREK-1 potassium channels and inhibit depolarization-dependent
potent and selective non-nucleotide antagonist of P2X3 and P2X2/3 secretion. Mol. Pharmacol. 2004, 65 (3), 599−610.
receptors, reduces chronic inflammatory and neuropathic pain in the rat. (197) Tertyshnikova, S.; Knox, R. J.; Plym, M. J.; Thalody, G.; Griffin,
Proc. Natl. Acad. Sci. U.S.A. 2002, 99 (26), 17179−17184. C.; Neelands, T.; Harden, D. G.; Signor, L.; Weaver, D.; Myers, R. A.;
(184) (a) Carter, D. S.; Alam, M.; Cai, H.; Dillon, M. P.; Ford, A. P. D. Lodge, N. J. BL-1249 [(5,6,7,8-tetrahydronaphthalen-1-yl)-[2-(1H-
W.; Gever, J. R.; Jahangir, A.; Lin, C.; Moore, A. G.; Wagner, P. J.; Zhai, tetrazol-5-yl)phenyl]amine]: a putative potassium channel opener with
Y. Identification and SAR of novel diaminopyrimidines. Part 1: The bladder-relaxant properties. J. Pharmacol. Exp. Ther. 2005, 313 (1),
discovery of RO-4, a dual P2X3/P2X2/3 antagonist for the treatment of 250−259.
pain. Bioorg. Med. Chem. Lett. 2009, 19 (6), 1628−1631. (b) Jahangir, A.; (198) Duprat, F.; Lesage, F.; Patel, A. J.; Fink, M.; Romey, G.;
Alam, M.; Carter, D. S.; Dillon, M. P.; Du Bois, D. J.; Ford, A. P. D. W.; Lazdunski, M. The neuroprotective agent riluzole activates the two P
Gever, J. R.; Lin, C.; Wagner, P. J.; Zhai, Y.; Zira, J. Identification and domain K+ channels TREK-1 and TRAAK. Mol. Pharmacol. 2000, 57
SAR of novel diaminopyrimidines. Part 2: The discovery of RO-51, a
(5), 906−912.
potent and selective, dual P2X3/P2X2/3 antagonist for the treatment of
(199) Flockerzi, V. An introduction on TRP channels. Handb. Exp.
pain. Bioorg. Med. Chem. Lett. 2009, 19 (6), 1632−1635.
Pharmacol. 2007, 179, 1−19.
(185) Chen, L.; Dillon, M. P.; Feng, L.; Hawley, R. C.; Yang, M.
(200) Nayak, T. K.; Harinath, S.; Nama, S.; Somasundaram, K.; Sikdar,
Preparation of Tetrazole Substituted Arylamides as P2X Receptor
S. K. Inhibition of human two-pore domain K+ channel TREK1 by local
Antagonists. WO2009077371, June 25, 2009.
(186) Burgey, C. S.; Deng, Z. J.; Nguyen, D. N.; Paone, D. V.; anesthetic lidocaine: negative cooperativity and half-of-sites saturation
Potteiger, C. M.; Vacca, J. P., Biarylcarboxamides as P2X3 Receptor kinetics. Mol. Pharmacol. 2009, 76 (4), 903−917.
Antagonists for Treatment of Pain and Their Preparation. (201) Kang, D.; Kim, G.; Kim, E.; La, J.; Lee, J.; Lee, E.; Park, J.; Hong,
WO2009058298, May 7, 2009. S.; Han, J. Lamotrigine inhibits TRESK regulated by G-protein coupled
(187) Bayrakdarian, M.; Buon, C.; Cantin, L.; Hu, Y.; Luo, X.; receptor agonists. Biochem. Biophys. Res. Commun. 2008, 367 (3), 609−
Santhakumar, V.; Tomaszewski, M. Preparation of 5,6-Dihydro-7H- 615.
pyrrolo[3,4-d]pyrimidin-7-one Derivatives as P2X3 Receptor Antago- (202) Brendel, J.; Goegelein, H.; Wirth, K.; Kamm, W. Inhibitors of the
nists for Treating Especially Pain. WO2008136756, November 13, 2008. TASK-1 and TASK-3 Ion Channel, and Their Therapeutic Use.
(188) Romagnoli, R.; Baraldi, P. G.; Cruz-Lopez, O.; Lopez-Cara, C.; WO2007124849, November 8, 2007.
Preti, D.; Borea, P. A.; Gessi, S. The P2X7 receptor as a therapeutic (203) Coburn, C. A.; Luo, Y.; Cui, M.; Wang, J.; Soll, R.; Dong, J.; Hu,
target. Expert Opin. Ther. Targets 2008, 12 (5), 647−661. B.; Lyon, M. A.; Santarelli, V. P.; Kraus, R. L.; Gregan, Y.; Wang, Y.; Fox,
(189) Guile, S. D.; Alcaraz, L.; Birkinshaw, T. N.; Bowers, K. C.; Ebden, S. V.; Binns, J.; Doran, S. M.; Reiss, D. R.; Tannenbaum, P. L.; Gotter, A.
M. R.; Furber, M.; Stocks, M. J. Antagonists of the P2X7 receptor. From L.; Meinke, P. T.; Renger, J. J. Discovery of a Pharmacologically Active
lead identification to drug development. J. Med. Chem. 2009, 52 (10), Antagonist of the Two-Pore-Domain Potassium Channel K2P9.1
3123−3141. (TASK-3). ChemMedChem 2012, 7 (1), 123−133.

623 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624


Journal of Medicinal Chemistry Perspective

(204) Welling, P. A.; Ho, K. A comprehensive guide to the ROMK


potassium channel: form and function in health and disease. Am. J.
Physiol. 2009, 297, F849−F863.
(205) Tang, H.; Walsh, S. P.; Yan, Y.; de Jesus, R. K.; Shahripour, A.;
Teumelsan, N.; Zhu, Y.; Ha, S.; Owens, K. A.; Thomas-Fowlkes, B. S.;
Felix, J. P.; Liu, J.; Kohler, M.; Priest, B. T.; Bailey, T.; Brochu, R.;
Alonso-Galicia, M.; Kaczorowski, G. J.; Roy, S.; Yang, L.; Mills, S. G.;
Garcia, M. L.; Pasternak, A. Discovery of selective small molecule
ROMK inhibitors as potential new mechanism diuretics. ACS Med.
Chem. Lett. 2012, 3 (5), 367−372.
(206) Wang, H.; Wu, M.; Yu, H.; Long, S.; Stevens, A.; Engers, D. W.;
Sackin, H.; Daniels, J. S.; Dawson, E. S.; Hopkins, C. R.; Lindsley, C. W.;
Li, M.; McManus, O. B. Selective inhibition of the Kir2 family of inward
rectifier potassium channels by a small molecule probe: the discovery,
SAR, and pharmacological characterization of ML133. ACS Chem. Biol.
2011, 6 (8), 845−856.
(207) Raphemot, R.; Lonergan, D. F.; Nguyen, T. T.; Utley, T.; Lewis,
L. M.; Kadakia, R.; Weaver, C. D.; Gogliotti, R.; Hopkins, C.; Lindsley,
C. W.; Denton, J. S. Discovery, characterization, and structure−activity
relationships of an inhibitor of inward rectifier potassium (Kir) channels
with preference for Kir2.3, Kir3.X, and Kir7.1. Front. Pharmacol. 2011, 2,
75/1−75/18.
(208) Ertl, P.; Rohde, B.; Selzer, P. Fast calculation of molecular polar
surface area as a sum of fragment-based contributions and its application
to the prediction of drug transport properties. J. Med. Chem. 2000, 43
(20), 3714−3717.

■ NOTE ADDED AFTER ASAP PUBLICATION


After this paper was published online November 29, 2012,
changes were made to references 123 and 124. The corrected
version was reposted December 3, 2012.

624 dx.doi.org/10.1021/jm3011433 | J. Med. Chem. 2013, 56, 593−624

You might also like