You are on page 1of 10

Bioorganic & Medicinal Chemistry Letters 26 (2016) 241–250

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry Letters


journal homepage: www.elsevier.com/locate/bmcl

Digest paper

Biased agonism: An emerging paradigm in GPCR drug discovery


Zoran Rankovic a,⇑, Tarsis F. Brust b, Laura M. Bohn b,⇑
a
Discovery Chemistry and Research Technologies, Eli Lilly and Company, 893 South Delaware Street, Indianapolis, IN 46285, USA
b
Department of Molecular Therapeutics, and Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA

a r t i c l e i n f o a b s t r a c t

Article history: G protein coupled receptors have historically been one of the most druggable classes of cellular proteins.
Received 6 October 2015 The members of this large receptor gene family couple to primary effectors, G proteins, that have built in
Revised 4 December 2015 mechanisms for regeneration and amplification of signaling with each engagement of receptor and
Accepted 8 December 2015
ligand, a kinetic event in itself. In recent years GPCRs, have been found to interact with arrestin proteins
Available online 9 December 2015
to initiate signal propagation in the absence of G protein interactions. This pinnacle observation has chan-
ged a previously held notion of the linear spectrum of GPCR efficacy and uncovered a new paradigm in
Keywords:
GPCR research and drug discovery that relies on multidimensionality of GPCR signaling. Ligands were
Biased ligand
Functional selectivity
found that selectively confer activity in one pathway over another, and this phenomenon has been
GPCR referred to as ‘biased agonism’ or ‘functional selectivity’. While great strides in the understanding of this
Drug discovery phenomenon have been made in recent years, two critical questions still dominate the field: How can we
rationally design biased GPCR ligands, and ultimately, which physiological responses are due to G protein
versus arrestin interactions? This review will discuss the current understanding of some of the key
aspects of biased signaling that are related to these questions, including mechanistic insights in the nat-
ure of biased signaling and methods for measuring ligand bias, as well as relevant examples of drug dis-
covery applications and medicinal chemistry strategies that highlight the challenges and opportunities in
this rapidly evolving field.
Ó 2015 Elsevier Ltd. All rights reserved.

G protein-coupled receptors (GPCRs) are seven-transmembrane (b2AR).6–8 More recently, two additional families of arrestin-
domain proteins that function to transmit signals into the intracel- related proteins have been found to be broadly expressed in
lular space. It is estimated that nearly 36% of all FDA-approved eukaryotes, namely, a group of vacuolar protein sort 26 (Vps26)-
drugs target at least one member of the GPCR gene family.1,2 Clas- related proteins and aarrestins. Their tertiary structure is similar
sically, the categorization of GPCR-ligands has been done based on to that of the visual and barrestins, although their physiological
their efficacies for activation of G proteins, as full agonists, partial role and potential involvement in regulating GPCR signaling are
agonists, antagonists, or inverse agonists, depending on their abil- still not fully understood.9,10
ities to elicit a receptor-mediated response. However, it has While arrestins were named based on their initially discovered
become evident that GPCRs can mediate multiple signaling out- ability to arrest (turn off) GPCR signaling, it is now evident that
comes. For instance, barrestins, which have been long associated barrestins regulate GPCR trafficking as well as G protein-indepen-
with receptor desensitization, can also lead to signaling events.3 dent signaling.11–13 A general scheme for the barrestin-mediated
Arrestins are cytosolic proteins originally discovered in the regulation of GPCR function is depicted in Figure 1. Agonist binding
visual system, which include arrestin-1 (also referred to as visual to GPCR stabilizes an active conformation(s) of the receptor that
arrestin), arrestin-2 (barrestin1), arrestin-3 (barrestin2) and promotes its interaction with heterotrimeric G proteins, Gabc
arrestin-4 (termed a cone arrestin).4,5 While arrestins-1 and -4 (Fig. 1a). This is followed by the GDP to GTP exchange at the Ga
are localized in retinal rods and cones, molecular cloning revealed subunit, and the subsequent dissociation of G proteins (Fig. 1b).
two ubiquitously expressed isoforms termed barrestin1 and bar- The dissociated G proteins interact with and modulate the activity
restin2 due to their high homology with the visual arrestin and of downstream effectors (e.g., adenylyl cyclase and phospholi-
their potent functional regulation of the b2 adrenergic receptor pases) that produce second messengers such as cAMP and inositol
phosphate (G protein-dependent signaling).14
Since one activated receptor can sequentially couple to multiple
⇑ Corresponding authors.
G proteins with signal amplification occurring through to the enzy-
E-mail addresses: rankovic_zoran@lilly.com (Z. Rankovic), lbohn@scripps.edu
(L.M. Bohn).
matic activity of receptor second messengers (e.g., cyclases and

http://dx.doi.org/10.1016/j.bmcl.2015.12.024
0960-894X/Ó 2015 Elsevier Ltd. All rights reserved.
242 Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250

Figure 1. Paradigms of GPCR-mediated signaling and multiple roles of barrestins: binding of an agonist (a) results in activation of signaling pathways by G proteins (b), as
well as barrestins (f), in addition to desensitization and internalization by barrestins (d and e).

phospholipases), desensitization mechanisms have evolved to turn recycled slowly or undergo degradation, for example, the angioten-
off the potentially deleterious effects of sustained signaling.15 sin II type 1A, neurotensin 1, vasopressin V2, thyrotropin-releasing
Desensitization is considered a two-step process, which starts with hormone, and substance P receptors.23
phosphorylation of the agonist-occupied GPCR by the second mes- Recent barrestin crystallographic,24 mutation,25 and biophysical
senger-dependent protein kinases, such as protein kinase A or C studies26 suggest that barrestins undergo extensive conformational
(PKA or PKC), or by a corresponding GPCR Kinase (GRK, Fig. 1c).4 changes upon binding to the phosphorylated GPCR. In their basal
This triggers the second desensitization step, which involves the cytosolic receptor-unbound state, arrestins are elongated mole-
recruitment of barrestin to active phosphorylated receptors and cules, which consist of two (N- and C-) domains (Fig. 2a) and the
the consequent steric hindrance of further G protein activation C-terminus anchored in a polar core between them, unavailable
(Fig. 1d).16 The initial discovery of the critical involvement of bar- for interaction with partner proteins (Fig. 2b).27
restins in GPCR desensitization was subsequently followed up by The recently reported crystal structure of barrestin1 in complex
studies that indicated their important role in receptor internaliza- with a fully phosphorylated 29-amino-acid carboxy-terminal
tion. It has been found that the receptor-bound barrestins interact (derived from the vasopressin receptor-2 (V2Rpp) and shown to
with several key components of the receptor endocytotic process functionally and conformationally activate barrestin1) revealed
(Fig. 1e), including the adaptor-protein2 (AP2) complex,17 the extensive conformational changes in the C-terminus, which is
clathrin heavy chain,18 and the E3 ubiquitin ligase Mdm2.19 The released and becomes available for interactions with clathrin and
complex formed upon the binding of clathrin/AP2 to the recep- AP2 (Fig. 2c).24 The V2Rpp–barrestin1 crystal structure also
tor-bound arrestin translocates to the clathrin-coated pit, which revealed a 20° twist between the N- and C-domains. A similar
is then sequestered off the plasma membrane and hindered from 20° rotation has been observed in the crystal structure of the
subsequent stimuli.20 mouse visual arrestin bound to a constitutively active form of
While the receptor–G protein interaction that takes place upon human rhodopsin.28 It has been suggested that the twisting move-
receptor activation is unstable and brief, the receptor–arrestin ment of the two domains is part of the general mechanism by
complex can be relatively stable and exist on a time scale of min- which arrestins, upon activation, may expose an additional inter-
utes to hours.21,22 Analysis of the agonist-mediated arrestin face for interacting with their numerous binding partners.
translocation to multiple GPCRs identified two major classes of Over the past decades, a growing list of binding partners has
receptor–arrestin complexes, based on their strength and longev- implicated barrestins in a number of important cellular functions
ity. Class A complexes are transient and receptor–barrestin interac- in addition to GPCR desensitization and trafficking. In particular,
tions rapidly dissociate upon receptor internalization, as arrestins have been found to link activated GPCRs to signaling
exemplified by the b2 adrenergic, l-opioid, endothelin type A, effectors such as the Src family tyrosine kinases,29 components of
dopamine D1, and a1 adrenergic receptors. Receptors in this class the extracellular signal-regulated kinase 1/2 (ERK 1/2) and the
are rapidly recycled back to the plasma membrane. In contrast, c-Jun N-terminal kinase 3 (JNK3),30 mitogen-activated protein
Class B complexes have more stable receptor–barrestin interac- kinase (MAPK) cascades,31 Akt,32,33 and effectively convey the
tions that persist even as the receptor undergoes endosomal sort- G protein-independent signaling (Fig. 1f). Because arrestin
ing. Receptors in this class are sequestered in endosomes and binding can uncouple G protein from the activated receptor, the
Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250 243

Figure 2. barrestin1 crystal structures. (a) Superimposed structures of barrestin1 in a basal state (inactive state: blue; PDB 1G4 M) and in complex with V2Rpp (Vasopressin
Receptor 2, partial protein, active state: green; PDB 4JQI) reveal marked conformational differences, including the release of the C-terminus which contains binding sites for
partner proteins such as clathrin; (b) inactive state: the polar core consisting of five interacting residues keeping the C-terminus locked in the place is thought to be a critical
stabilizer of the barrestin1 inactive state (Arg393–Asp290 salt bridge highlighted by the broken black circle); (c) active state: upon V2Rpp binding (omitted for clarity) the
C-terminal strand residue Arg393 is displaced, and its interacting partner Asp297 undergoes a large movement together with the rest of the lariat loop, resulting in significant
conformational rearrangement in the polar core of the activated barrestin1.

G protein-dependent and arrestin-dependent signaling can be seg- target primarily the short H8 helix adjacent to the TM7 region.40
regated. Furthermore, the two signaling pathways, proved to be In addition, a detailed mapping of the b2AR phosphorylation in this
spatially and temporally distinct events, are also pharmacologi- study demonstrated that the barrestin-biased ligand (carvedilol)
cally separable.34 In recent years, there has been a movement to induced a phosphorylation pattern that was distinct from that of
develop ligands that will preferentially segregate GPCR to one isoproterenol, an unbiased full agonist.40 This suggests that by
pathway or another. These functionally selective ligands, also inducing distinct receptor conformations, the biased ligands are
referred to as the ‘functionally selective’ or ‘biased’ agonists, have able to recruit distinct GRKs and, consequently, produce a distinct
demonstrated the potential to harness GPCR signaling and have phosphorylation pattern. In fact, the experimental evidence accu-
collectively opened new possibilities in GPCR drug discovery.35 mulated over the recent years indicates that the biased ligands
Although the exact molecular mechanism of biased signaling is can stabilize both the receptor and barrestin in conformations that
not well understood, the studies reported to date suggest that the are distinct from those associated with unbiased ligands. For
GPCR conformation, stabilized by a G protein-biased agonist, is dis- example, by employing an intramolecular bioluminescence reso-
tinct from the conformation stabilized by a barrestin-biased ago- nance energy transfer (BRET)-based biosensor of barrestin2 and a
nist. For example, the fluorescence-based study on the arginine combination of biased ligands and/or biased mutants of three dif-
vasopressin type 2 receptor (V2R) activation using biased and unbi- ferent GPCRs, Shukla and colleagues at Duke University provided
ased agonists provided experimental evidence suggesting that the evidence suggesting that barrestins can adopt multiple ‘active’ con-
transmembrane helix 6 (TM6) and third intracellular loop (icl3) formations.41 One can speculate that these multiple barrestin con-
movements are associated with selective G protein signaling, formers with distinct surface topologies complex with different
whereas the movements between its TM7 and helix 8 (H8) region binding partners, and thereby engage in different downstream sig-
are required for selective barrestin recruitment.36 Similar conclu- naling pathways.
sions resulted from the 19F NMR study of b2AR in complex with a Another important advance toward unraveling the molecular
range of ligands with different functional profiles.37 The NMR sig- basis of biased signaling was the recently reported crystal struc-
nal changes of the site-specific 19F-labels located in the cytoplas- ture of two 5-hydroxytryptamine serotonin receptor subtypes,
mic region revealed that binding of an unbiased agonist (e.g., namely 5-HT1B and 5-HT2B, both bound to ergotamine (ERG,
formoterol) shifts the receptor conformational equilibrium toward Fig. 3a).42,43 ERG is an alkaloid produced by the ergot fungus, which
the specific ‘G protein active’ state of TM6, while the barrestin- has been shown to exhibits a strong barrestin-biased agonism at
biased ligands (e.g., carvedilol) impact primarily the conforma- 5-HT2B, and a relatively weak bias at the 5-HT1B receptor.
tional state of the TM7 helix. These findings are consistent with A close inspection of the two data sets revealed a small shrink-
the crystallographic38 and electron microscopy39 studies of the age of the ligand binding pocket in the ERG-5-HT2B structure, with
b2AR–Gabc heterotrimer complex that showed that the Ga sub- ERG in slightly different binding conformation and hydrophobic
unit contacts primarily the intracellular regions of TM5 and TM6. contacts with several additional residues when compared with
A study of the patterns of b2AR phosphorylation by individual the 5-HT1B structure (Fig. 3b). Intriguingly, a similar network of
GRKs, a prerequisite for b-arrestin binding, showed that GRKs additional interactions has been found in the structure of
244 Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250

Figure 3. (a) Superimposed crystal structures of 5-HT1B (blue; PDB 4IAR) and 5HT2B (gold; PDB 4IB4) in complex with ergotamine (ERG); (b) prominent conformational
difference of ERG bound to 5-HT1B (gray) and 5HT2B (green) is highlighted by the broken black circle. Contacts with additional residues observed in ERG-5HT2b structure are
also highlighted (gold); (c) conserved DRY motif exist in an ‘active’ state (the Asp152–Arg153 salt bridge broken) in 5HT1B structure (blue), and ‘inactive’ state (the Asp146–
Arg147 salt bridge intact) in 5HT2B structure (gold).

carvedilol, a barrestin-biased agonist bound to the b1AR.44 The methods for identifying biased ligands.52 However, not always a
analysis of the two crystal structures also revealed that the ERG- consensus was reached about the most appropriate way of mea-
bound 5HT1B complex exists in a classical full agonist-induced suring functional selectivity.53,54 Both qualitative and quantitative
active-like state, whereas the ERG-bound 5HT2B structure displays methods have been described.52,55,56 These methods compare test
conformational characteristics that are attributed to both the ligands with reference agonists in order to determine if a com-
active and inactive states. For example, both structures showed pound is biased for one signaling pathway relative to another.52
the conserved NPxxY motif (TM7) in an active-like conformation In the case of the quantitative methods, a ‘bias factor’ defining
with the intracellular TM7 helix moved toward the receptor core. the degree of bias of the test ligand is generated.52
However, only the ERG-5HT1B structure displayed another highly One of the most simplistic methods, developed by Ehlert and
conserved GPCR feature, the DRY motif in TM3, in an active state colleagues,57,58 utilizes data from standard four-parameter Hill
with the salt bridge between the Asp146–Arg147 residues being bro- equations that display slopes close to unity. This method uses
ken. In the ERG-5HT2B structure, the DRY motif salt bridge between ratios of relative efficacies (Emax) by potencies (EC50) (relative
Asp152 and Arg153 was intact, which is consistent with all inactive- activities) to quantify ligand bias, as shown in the equation bellow:
state GPCR structures known to date (Fig. 3c). Consequently, it is !
relative activity12;lig
tempting to attribute this ‘intermediate’ state of the 5HT2B receptor Bias factor ¼ log
relative activity12;ref
structure to the barrestin-biased signaling conformation character-
0 ! ! 1
ized by a strong barrestin and an inefficient G protein coupling. Emaxpath1  EC50-path2 Emaxpath1  EC50-path2
However, it is important to note that the observed difference ¼ log @  A
EC50-path1  Emaxpath2 EC50-path1  Emaxpath2
between the two structures may also reflect distinct signal trans- lig ref

duction mechanisms between the two receptor subtypes used in


this study. Additionally, it is critical to take into account the fact Additional methods use the Black and Leff operational model to
that crystal structures represent only a static snapshot of a quantify functional selectivity. As shown in the equation bellow,
potentially complex ensemble of conformations. It is now widely the Black and Leff model defines the terms s (coupling efficiency)
recognized that GPCRs are highly dynamic systems that assume and KA (conditional affinity) as the ligands’ (A) intrinsic efficacy
many conformations associated with different signaling states, and dissociation constant of agonist–receptor–signal transducer
which are stabilized by different ligands.45,46 Further research complex, respectively (n = transducer slope).59
efforts, most likely involving crystallography in conjunction with Emax  sn  ½An
other biophysical and computational47 methods to study GPCRs Response ¼
s n  ½An þ ð½A þ K A Þn
as dynamic systems, will be required to reliably delineate the
biased signaling mechanism at the molecular level. The Black and Leff operational model is as a mathematical rear-
Functional selectivity (biased signaling): definition, assays, quan- rangement of two distinct Hill equations (concentration of agonist
tification: The definition and identification of biased ligands has by receptor occupancy and receptor occupancy by effect) with the
been challenging. It appears that initially, differences in relative introduction of s (s = receptor density/receptor occupancy needed
efficacy or potency were used as the determinants of ligand for producing 50% of the system’s maximal effect).59 By plotting
bias.48–51 The realization that more efficient approaches for deter- ligand’s functional data into the Black and Leff equation, ratios of
mining bias were needed, led to the development of different s/KA quantify pharmacological responses regardless of signal
Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250 245

amplification and receptor reserve.52,60 This feature is very desir- display antagonism or inverse agonism for one of the signaling
able for ligand bias analyses and has been developed by Kenakin pathways analyzed.55 Extremely biased compounds have been
and colleagues as ‘transduction coefficients’ according to the fol- described.49,73,74 However, a framework for quantitatively ranking
lowing equation:60 the degree of bias of these compounds has only just begun to
  emerge.61,75 As these quantitative, mechanistic methods become
s
Bias factor ¼ DD log more accepted and elaborative there is certain to be a broader
KA
interest in both the discovery and interpretation of the molecular
    !
s s and physiological implications of extremely biased ligands. Again,
¼ log  log
KA lig KA ref
it is important to emphasize that bias is context dependent and
path1
    ! bias observed in one condition (i.e., cell-based signaling assay)
s s may not translate to the receptor expressed in an endogenous set-
 log  log
KA lig KA ref
ting (i.e., synapse).
path2
Drug discovery and medicinal chemistry highlights: The realiza-
The transduction coefficients and the relative activity ratios tion that barrestins mediate cellular signaling, which is indepen-
become proportional (and, thus, result in the same bias factors) dent and pharmacologically separable from G protein signaling,
when slopes of 1 are obtained from the dose–response curves.52,55 has changed a previously held notion of the linear spectrum of
The transduction coefficients have been recently modified in order GPCR efficacy and uncovered a new paradigm in drug discovery
to more accurately accommodate data from weak partial ago- that relies on the multidimensionality of GPCR signaling. Over
nists.61 This new method entitled ‘competitive model’ utilizes the past couple of decades a number of GPCR ligands displaying
ligand curves in agonist and antagonist mode to more precisely biased profiles were reported in the literature, most discovered ret-
define s and KA values for weak partial agonists.61 rospectively. In this section we discuss the hypothesis-driven drug
The utility of the methods of measuring ligand bias has been discovery and medicinal chemistry efforts that were intentionally
studied. For instance, the transduction coefficients have been suc- focused from the outset on the design and development of biased
cessfully incorporated into SAR studies to aid in the development GPCR ligands toward safer and/or more efficacious medicines. For
of biased ligands for the dopamine D2 and the j-opioid recep- a more general coverage outside of the scope of this section readers
tors.62–64 It also appears that these analyses can provide mechanis- are referred to recently published reviews of this topic.76,77
tic insight on the activation of more complex signaling pathways One of the earliest identified opportunities for the biased GPCR
that are downstream of immediate GPCR signal transducers.55 agonist approach in drug discovery steamed from the studies
Moreover, bias analyses have been successfully implemented in reported in 1999, 2000 and 2005 by Bohn and colleagues. These
studies to investigate ligand-specific functional consequences of studies showed that morphine administered to barrestin2 KO mice
different point mutations in the muscarinic M1 receptor.65 It is produce enhanced and prolonged analgesia with reduced tolerance
important to note that bias is a result of how a ligand interacts and fewer adverse events, when compared to wild-type mice.78–80
with the receptor (molecular bias, which translates across assays More recently, a similar profile of improved morphine potency
and expression systems) plus how the receptor response is with reduced side effects has been observed in mice and rats with
observed (an experimental artifact that is system- and assay- barrestin ablated by siRNA.81,82 Opioids produce powerful analge-
dependent and may not translate to other expression systems or sia, by activating l-opioid receptors (MOR), in addition to many
assays).52 As shown in the methods above, the solution found to efficacy-limiting adverse effects, which include tolerance, nausea,
correct for this observational issue has been to use a reference vomiting, sedation, constipation, and respiratory depression.83–85
ligand for comparisons of relative activities within specific assay Numerous MOR agonists have been developed over the years as
systems.52 Therefore, bias is always a measure relative to the refer- part of a global effort to improve the opioid safety and tolerability
ence ligand and assay systems. A reference agonist is typically cho- profile. However, all of these structurally diverse opioids display
sen based on its ability to robustly activate the receptor in the morphine-like adverse effects, suggesting that the analgesic effi-
assays that are to be compared. In some cases, the endogenous cacy, as well as the side effects, is directly related to MOR activa-
ligand of a receptor is used as the reference compound. However, tion. Intriguingly, experiments in the barrestin2 KO mice imply
nature may also exploit biased signaling and, thus, endogenous not only that barrestins reduce morphine’s analgesic potency, but
ligands may also present functional selectivity.66–71 This allows it may also be associated with producing morphine-induced side
for a test ligand being biased compared to an endogenous agonist, effects.
but that does not preclude the endogenous ligand from displaying This hypothesis suggests that it would be beneficial to develop
bias compared to another reference ligand.52 Therefore, it is impor- agonists that could selectively activate the G protein signaling
tant to emphasize that bias is not an absolute property, but is pathway without engaging barrestins as a means to preserve anal-
highly dependent on context of the assay and performance of the gesic efficacy while avoiding side effects. The first ligand identified
reference compound. that displayed this separation between signaling pathways was a
Another type of bias that needs to be considered in functional neoclerodane diterpene derived from the kappa opioid receptor
selectivity studies is the case of ‘perfect bias’. Ligands that display natural product agonist, salvinorin A.86 This compound, herkinorin,
perfect bias are agonists for one signaling pathway and do not dis- is structurally intriguing as it has no basic nitrogen, a highly con-
play any detectable activity for another pathway.52 Caution should served property of GPCR ligands.87,88 In antinociception studies
be taken in the classification of those ligands, since assay systems assessing formalin-induced pain responses, herkinorin proved effi-
that display low amplification or have low receptor reserve might cacious without tolerance; supporting the general hypothesis that
not result in any detectable responses for partial agonists. There- the antinociceptive properties could be dissected from tolerance by
fore, it has been suggested that those ligands should be defined driving G protein signaling over barrestin2 recruitment.89 How-
as ‘extremely biased’.72 Different assay systems with higher signal ever, herkinorin was not shown to be acting at the same sites as
amplification and receptor reserve should be employed for the morphine and therefore, it is difficult to conclude whether its bias
analyses of ligands that display this type of bias. properties produced the differences in physiological responses.
The cases of perfect or extreme bias also expose a deficiency in Further motivated by this hypothesis, Chen and colleagues at
the current quantitative methods for measuring ligand bias. That Trevena Inc. set out to identify G protein-biased MOR agonists as
is, these methods cannot accommodate data from compounds that potential novel analgesics with improved efficacy and side effect
246 Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250

profile.90 They initiated a hit finding effort by screening the inter- Interestingly, the in vitro MOR biased profile of compound TRV-
nal compound collection in the cAMP accumulation assay (G pro- 130 was found to be species dependent. It showed higher barrestin
tein signaling) and the barrestin2-based b-galactosidase fragment potency and efficacy at the mouse-(EC50 = 12.6 nM; efficacy = 74%)
compartmentalization assay (barrestin signaling). Both assays and dog-(EC50 = 9.4 nM; efficacy = 24%) receptors, compared to the
were developed in the HEK-293 cell line expressing human MOR. rat MOR (inactive) and human MOR (EC50 = 40 nM; efficacy = 14%).
This effort led to the discovery of tetrahydropyran 1 (Table 1) This phenomenon, potentially resulting from differences in phos-
with a submicromolar G protein MOR potency and a low MOR bar- phorylation sites and/or receptor expression, is important to
restin recruitment activity (32% vs 100% in comparison to mor- remember when designing or interpreting translational in vivo
phine), which was deemed a suitable starting point for an data for biased ligands.
optimization program. The optimization efforts around compound Gratifyingly, the observed pre-clinical profile of TRV-130
1, which primarily focused on improving the MOR G protein proved translatable in the clinic, where in healthy volunteers it
potency while diminishing barrestin recruitment, resulted in the produced analgesia with less reduction in a respiratory drive and
discovery of thiophenyl analogue 2. It is worth noting that some a less severe nausea than morphine.92 However, in a recently
closely related analogues displayed an unbiased full agonist profile, reported phase 2, randomized, placebo- and active-controlled
such as compound 3, which showed high efficacy in both the cAMP study in acute pain following bunionectomy, intravenously admin-
and barrestin assays (104% and 197%). Displaying the desired istered TRV130 produced greater categorical pain relief compared
in vitro profile, compound 2 was progressed to the in vivo evalua- with morphine (P <0.005), with tolerability similar to morphine
tion and benchmarking against morphine’s efficacy and potential and no serious adverse effects; the respiratory effects in this study
for adverse effects. These studies showed that at equianalgesic did not provide a statistically significant benefit over morphine but
doses, established in the mouse hot-plate assay, morphine (6 mg/ it is hoped that the improved analgesic benefit will offset this dif-
kg) exhibited a more severe constipation effect than compound 2 ference.93 These results provide an early clinical translation of
(3 mg/kg) in a mouse glass bead retention model of colonic motil- ligand bias as an important new paradigm in GPCR-targeted phar-
ity (158 min vs 65 min retention times, respectively). However, macotherapy. However, considering a profound effect seen in bar-
despite the favorable in vivo pharmacology profile, the potent restin2 KO mice, one may be disappointed with the relatively
hERG channel inhibition (IC50 = 2.3 lM) and the cardiac abnormal- limited improvements brought about by TRV-130 in both the ani-
ities observed in the rabbit ventricular wedge assay prevented fur- mal models and clinical trials. For example, in comparison to mor-
ther progression of compound 2. Encouraged by the initial in vivo phine, TRV-130 showed only approximately a two-fold improved
data that reaffirmed the original hypothesis, the authors continued therapeutic index (analgesia vs respiratory depression).92 This
their optimization efforts around 2, which led to the discovery of 3- raises the question of whether this result is the maximum one
methoxy thiophenyl derivative 4 with not only an improved biased could expect from this approach, or if a more biased G protein
MOR profile (Table 1), but also reduced cardiovascular liabilities MOR agonist would show a safer profile with an even greater ther-
with hERG IC50 >200-fold over MOR cAMP EC50 value of 8 nM. apeutic index than TRV-130.
The authors calculated a bias factor of 3 based on the equiactive Similarly to the MOR for pain, the dopamine D2 receptor has
comparison method based on the intrinsic relative activity model been for a long time a focus of research and drug discovery for psy-
outlined by Griffin et al.58 and Rajagopal et al.56,91 chiatric disorders. With the exception of aripiprazole 5, reportedly
On the basis of its in vitro profile, compound 4 (TRV-130) was a D2 partial agonist, all clinically used antipsychotics are unbiased
progressed for in vivo evaluation, where it displayed a 5–10 times antagonists of the D2 receptor.94 The D2 antagonism is thought to
higher potency than morphine in the mouse (ED50 values 0.9 mg/ underlie the therapeutic benefit in the treatment of psychotic
kg vs 4.9 mg/kg) and rat (ED50 0.32 mg/kg vs 3.2 mg/kg) hot-plate symptoms, as well as serious extrapyramidal side effects including
assays, as well as an improved safety profile in the mouse glass catalepsy and other dyskinesias.95 Investigating a possibility of
bead colonic motility assay (Emax = 53% for TRV-130 compared to separating the D2 antipsychotic efficacy from the target-related
100% for morphine at equianalgesic doses). Despite showing at side effects, Chen and colleagues were interested in developing a
higher doses an effect on blood gases, TRV-130 therapeutic index barrestin signaling biased D2 receptor agonist.96 They selected
proved superior to morphine in respect of the analgesia versus res- the FDA approved antipsychotic aripiprazole as a starting point
piratory suppression in the rat, with no effect on blood CO2 and O2 because of its unique D2 profile and hoping that its closely related
levels at doses of up to eight-fold over the equianalgesic dose.91 analogues may share its favorable pharmacokinetic properties,

Table 1
G protein-biased MOR agonists90

Compound R1, R2, R3 MOR cAMP pEC50 MOR cAMP Emax (%) MOR barr2 pEC50 MOR barr2 Emax (%)
Morphine NA 7.4 100 6.3 100
1 NA 6.3 74 5.7 32
2 H, H, H 7.8 95 6.6 15
3 H, Me, Me 8.3 104 6.3 197
4 (TRV-130) OMe, H, H 8.1 84 7.3 15
Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250 247

Table 2
From aripiprazole to the barrestin-biased D2 agonists96

Cl
Cl 6 O
O N
N N H
N O
H
N
Cl Cl O

N N Cl Cl O S

N N
5, aripiprazole 7

Compound D2 cAMP EC50 (nM) D2 cAMP Emax (%) D2 barr2 EC50 (nM) D2 barr2 Emax (%)
5 1.0 51 4.0 62
6 Inactive <20 1.6 47
7 Inactive <20 50 97

including CNS exposure. A range of aripiprazole analogues were compounds were profiled in the D2 cAMP accumulation assay,
synthesized using diversity-oriented high throughput synthetic whereas barrestin signaling was evaluated by measuring phospho-
methods. The compounds were profiled in the cAMP accumulation rylation of extracellular signal-regulated kinase 1/2 (ERK 1/2). A
(G protein signaling) and barrestin recruitment assays, in which particularly interesting aspect of this work is the application of
aripiprazole itself displayed an unbiased partial agonist profile the transduction coefficients, as shown in Table 3.
(Table 2). In this testing paradigm, cariprazine 8 displayed a 230-fold bias
This effort resulted in the discovery of potent and barrestin- toward the cAMP pathway compared with dopamine, whereas a
biased D2 agonists 6 and 7 (Table 2). Compound 6 displayed a closely related carbamate 9 showed a 42-fold bias decrease. Inter-
potent partial agonist profile in the D2 barrestin assay, similar to estingly, while all cariprazine derivatives disclosed in this Letter
the parent compound 5, while in the same assay 7 was found to showed a bias toward the G protein signaling pathway, subtle
be a slightly less potent but full agonist. Importantly, both com- changes of the D2 unbiased partial agonist 10, a structurally related
pounds displayed a potent antipsychotic-like efficacy in the aryl piperazine reported by Tschammer et al.,99 led to analogues
amphetamine-induced hyperlocomotion studies in mouse, with- that displayed a strong bias toward barrestin signaling, for exam-
out inducing motor side effects that are characteristic for unbiased ple, 11; DD log (s/KA) = 1.2 (Fig. 4).
D2 antagonists such as haloperidol. At the same doses both com- Cardiovascular disease is another therapeutic area where biased
pounds produced a haloperidol-like catalepsy in barrestin2 KO agonists are extensively investigated as an approach to improve
mice.97 Taken together, these results suggest that the D2 receptor the current standards of care. Angiotensin II is a powerful endoge-
barrestin2 signaling blocking is not required for antipsychotic effi- nous vasoconstrictor that exerts its physiological effects through
cacy, while the barrestin2 agonist activity may protect from the activation of angiotensin II receptors (AT1R), a Gaq-coupled
motor side effects triggered by D2-mediated G protein antagonism. GPCR.100 AT1R antagonism proved to be an effective approach for
It remains to be seen if these encouraging preclinical data for the the treatment of hypertension since the 1985 launch of losartan,
barrestin-biased D2 agonists can be translated to the clinic. the first of the AT1 receptor blockers (ARBs) currently used in the
A similar literature-inspired approach in the design of clinic.101 However, in addition to blocking adverse effects of angio-
barrestin-biased D2 receptor ligands has been taken by Shonberg tensin II, such as hypertension and cardiac remodeling, ARBs also
and colleagues at Monash University.62 The focus of their struc- prevent beneficial effects of angiotensin II, including the AT1R-
ture–functional selectivity exploration was on cariprazine 8, mediated inotropy which supports cardiac function. Consequently,
another D2 partial agonist currently in clinical development for reduced cardiac output was long thought to be an inseparable side
schizophrenia.98 Again, to assess G protein-related signaling, the effect associated with AT1R antagonism and ARBs in general. This

Table 3
From cariprazine to the G protein-biased D2 agonists62

O
Cl Cl R
N
N N H

8, R= NMe2, cariprazine
9, R= OtBu

Compound cAMP pERK1/2 cAMP–pERK1/2


pKA Log s Log (s/KA) D log (s/KA) pKA Log s Log (s/KA) D log(s/KA) DD log (s/KA)
8 8.71 0.85 9.59 1.04 7.61 0.37 7.25 1.3 2.34
9 7.11 0.76 7.92 0.63 7.51 0.31 7.21 1.34 0.71
248 Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250

Figure 4. Aryl piperazine analogues of cariprazine and aripiprazole with different D2 receptor functional selectivity profiles.99

notion has been recently challenged by studies showing that, in a where a systematic SAR exploration around a known ligand,
stark contrast to angiotensin II (a balanced AT1R agonist) a bar- regardless of its functional selectivity, led to identification of a suit-
restin-biased analogue Sar1Ile4Ile8-angiotensin (SII) robustly pro- able starting compound with a desired biased profile. However,
motes contractility of isolated cardiac myocytes.102 This finding these steep and often unpredictable ‘SAR cliffs’ present a signifi-
suggested that the AT1R G protein signaling pathway is the pri- cant challenge at the hit/lead optimization stage, when SAR-driven
mary mediator of the angiotensin II vasoconstrictive effect, medicinal chemistry efforts are focused on preserving the desired
whereas barrestin signaling is solely responsible for the ionotropic biased profile while optimizing other properties that are required
effect of AT1R stimulation. In line with this hypothesis, another in a successful clinical candidate. Great strides have been made
biased AT1R ligand that blocks the G proteins but activates the bar- in unravelling the biostructural mechanisms of the biased signal-
restin signaling pathway, Sar-Arg-Val-Tyr-Ile-His-Pro-D-Ala-OH ing.117 However, to better understand the SAR and enable a
(TRV027), has been shown to reduce blood pressure and improve rational design of biased ligands, more atomic-level biostructural
cardiac performance in rats, whereas the unbiased ARB losartan information about the nature of ligand–receptor–effector interac-
decreased both blood pressure and cardiac performance.103 It has tions will be required. The most useful information in this context
been shown that the in vivo effect of a selective barrestin-biased will likely come from research that effectively combines the crys-
AT1R agonist on cardiac contractility is lost in barrestin2 KO ani- tallographic and other biophysical and computational methods to
mals, which further supports the hypothesis that positive ionotro- study GPCRs as dynamic systems.
pic effects of AT1R stimulation are specific to barrestin-mediated Availability of high quality biased ligands is a critical prerequi-
signaling.104 Since administration of a barrestin-biased AT1R ago- site to addressing the ultimate challenge of deconvoluting in vivo
nist does not lead to increased intracellular levels of secondary physiological effects that are associated with different GPCR signal-
messengers such as calcium, IP1 or DAG, it is hypothesized that ing pathways. This is proving particularly difficult, as the manner
the observed barrestin-dependent inotropic effect is likely related in which a receptor signals is highly context dependent and the
to enhanced sensitivity of the myofilament to calcium. In addition diversity of expression of GPCRs in vivo is contextually rich. When
to the positive effect on cardiac contractility, barrestin-biased AT1R translating in vitro bias to in vivo pharmacology one should be
agonists were also found to promote cell survival during cardiac aware of possible complications resulting from potential species
injury. Together, these findings present intriguing opportunities differences, which may affect ligand potency or functional selectiv-
for cardiac therapy in congestive heart failure in general and ity in vivo as indicated earlier. One should also consider potential
ischemic injury in particular, which is often characterized clinically effects of in vivo pharmacokinetics, which may result in subopti-
by peripheral arterial vasoconstriction and diminished cardiac con- mal ligand exposure at the site of action, or formation of active
tractility. Indeed, the preclinical efficacy and safety profile of metabolites with unbiased agonist profiles. For example buprenor-
TRV027 has been translated into the clinic where it lowered blood phine, a clinically used opioid analgesic, is reportedly a partial and
pressure in healthy volunteers on a salt restricted diet, as well as in biased agonist of the MOR, whereas its main metabolite in humans,
patients with acute heart failure (AHF), without significant adverse norbuprenorphine, is a potent, full and balanced MOR agonist.118
safety signals. On the basis of these data TRV027 has recently pro- Interestingly, since norbuprenorphine is a P-gp substrate actively
gressed to a larger Phase 2b study in AHF patients.105 effluxed at the blood brain barrier, it does not affect buprenor-
Concluding remarks and future perspectives: The classical under- phine’s centrally-mediated pharmacology.119
standing of GPCR signaling106,107 has undergone major revisions in Moreover, many receptors were found to couple to different
recent years with the introduction of the biased agonism para- types of Ga subunits of G proteins,120,121 as well as the multiple
digm.108–111 The realization that barrestin has a role in GPCR sig- isoforms and combinations of Gbc subunits, which could result
naling and the discovery of ‘biased’ or ‘functionally selective’ in distinct signaling events and further increase the overall com-
ligands that can produce specific receptor activation profiles have plexity of GPCR signaling.122–124 The concept of biased signaling
stimulated significant interest in the physiological, pharmacologi- will inevitably broaden to encompass additional complexities
cal and biostructural mechanisms that underline this para- and continue to evolve as an important approach toward safer
digm.33,64,78–80,112–116 Based on experimental evidence suggesting and more effective medications. Indeed, the sheer extent of the
that the physiological effects of GPCR activation are pathway current scientific literature and ongoing clinical developments for
dependent, functional selectivity has been proposed as a strategy a range of indications assure increasingly prominent role of the
for improving current drug therapies that target GPCRs.66 biased agonism paradigm in GPCR drug discovery.
Emerging reports of drug discovery programs aimed to evaluate
potential therapeutic benefits of the functional selectivity para- References and notes
digm provide an insight into the current challenges and opportuni-
ties in this rapidly evolving field. For example, understanding SAR 1. Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. Nat. Rev. Drug Disc. 2006, 5, 993.
2. Rask-Andersen, M.; Almen, M. S.; Schioth, H. B. Nat. Rev. Drug Disc. 2011, 10,
data is one of the commonly reported challenges for medicinal che- 579.
mists working on biased ligand programs, since even small struc- 3. Lefkowitz, R.; Shenoy, S. Science 2005, 308, 512.
tural modifications are often found to result in large shifts in the 4. Shukla, A. K.; Xiao, K.; Lefkowitz, R. J. Trends Biochem. Sci. 2011, 36, 457.
5. Lefkowitz, R. J. Prog. Mol. Biol. Transl. Sci. 2013, 118, 3.
functional selectivity. Interestingly, this phenomenon has been 6. Lohse, M. J.; Benovic, J. L.; Codina, J.; Caron, M. G.; Lefkowitz, R. J. Science 1990,
successfully leveraged as an effective hit generation strategy, 248, 1547.
Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250 249

7. Sterne-Marr, R.; Gurevich, V. V.; Goldsmith, P.; Bodine, R. C.; Sanders, C.; 45. Kahsai, A. W.; Xiao, K.; Rajagopal, S.; Ahn, S.; Shukla, A. K.; Sun, J.; Oas, T. G.;
Donoso, L. A.; Benovic, J. L. J. Biol. Chem. 1993, 268, 15640. Lefkowitz, R. J. Nat. Chem. Biol. 2011, 7, 692.
8. Benovic, J. L.; Kuhn, H.; Weyand, I.; Codina, J.; Caron, M. G.; Lefkowitz, R. J. 46. Nygaard, R.; Zou, Y.; Dror, R. O.; Mildorf, T. J.; Arlow, D. H.; Manglik, A.; Pan, A.
Proc. Natl. Acad. Sci. U.S.A. 1987, 84, 8879. C.; Liu, C. W.; Fung, J. J.; Bokoch, M. P.; Thian, F. S.; Kobilka, T. S.; Shaw, D. E.;
9. Aubry, L.; Klein, G. Prog. Mol. Biol. Transl. Sci. 2013, 118, 21. Mueller, L.; Prosser, R. S.; Kobilka, B. K. Cell 2013, 152, 532.
10. Kang, D. S.; Tian, X.; Benovic, J. L. Curr. Opin. Cell Biol. 2014, 27, 63. 47. Costanzi, S. Trends Pharmacol. Sci. 2014, 35, 277.
11. Gurevich, V. V.; Gurevich, E. V.; Cleghorn, W. M. Handb. Exp. Pharmacol. 2008, 48. Urban, J.; Clarke, W.; von Zastrow, M.; Nichols, D.; Kobilka, B.; Weinstein, H.;
15. Javitch, J.; Roth, B.; Christopoulos, A.; Sexton, P.; Miller, K.; Spedding, M.;
12. Lefkowitz, R. J.; Rajagopal, K.; Whalen, E. J. Mol. Cell 2006, 24, 643. Mailman, R. J. Pharmacol. Exp. Ther. 2007, 320, 1.
13. Tian, X.; Kang, D. S.; Benovic, J. L. Handb. Exp. Pharmacol. 2014, 219, 173. 49. Gay, E. A.; Urban, J. D.; Nichols, D. E.; Oxford, G. S.; Mailman, R. B. Mol.
14. Birnbaumer, L. Biochim. Biophys. Acta 2007, 1768, 756. Pharmacol. 2004, 66, 97.
15. Benovic, J. L.; DeBlasi, A.; Stone, W. C.; Caron, M. G.; Lefkowitz, R. J. Science 50. Holloway, A. C.; Qian, H.; Pipolo, L.; Ziogas, J.; Miura, S.; Karnik, S.; Southwell,
1989, 246, 235. B. R.; Lew, M. J.; Thomas, W. G. Mol. Pharmacol. 2002, 61, 768.
16. Zhang, J.; Ferguson, S. S.; Barak, L. S.; Aber, M. J.; Giros, B.; Lefkowitz, R. J.; 51. Sneddon, W. B.; Magyar, C. E.; Willick, G. E.; Syme, C. A.; Galbiati, F.; Bisello,
Caron, M. G. Receptors Channels 1997, 5, 193. A.; Friedman, P. A. Endocrinology 2004, 145, 2815.
17. Laporte, S. A.; Oakley, R. H.; Zhang, J.; Holt, J. A.; Ferguson, S. S.; Caron, M. G.; 52. Kenakin, T.; Christopoulos, A. Nat. Rev. Drug Disc. 2013, 12, 205.
Barak, L. S. Proc. Natl. Acad. Sci. U.S.A. 1999, 96, 3712. 53. Kenakin, T.; Christopoulos, A. Nat. Rev. Drug Disc. 2013, 12, 483.
18. Goodman, O. B., Jr.; Krupnick, J. G.; Santini, F.; Gurevich, V. V.; Penn, R. B.; 54. Rajagopal, S. Nat. Rev. Drug Disc. 2013, 12, 483.
Gagnon, A. W.; Keen, J. H.; Benovic, J. L. Nature 1996, 383, 447. 55. Brust, T. F.; Hayes, M. P.; Roman, D. L.; Burris, K. D.; Watts, V. J. J. Pharmacol.
19. Shenoy, S. K.; Barak, L. S.; Xiao, K.; Ahn, S.; Berthouze, M.; Shukla, A. K.; Exp. Ther. 2015, 352, 480.
Luttrell, L. M.; Lefkowitz, R. J. J. Biol. Chem. 2007, 282, 29549. 56. Rajagopal, S.; Ahn, S.; Rominger, D.; Gowen-MacDonald, W.; Lam, C.; Dewire,
20. Praefcke, G. J.; McMahon, H. T. Nat. Rev. Mol. Cell Biol. 2004, 5, 133. S.; Violin, J.; Lefkowitz, R. Mol. Pharmacol. 2011, 80, 367.
21. Charest, P. G.; Terrillon, S.; Bouvier, M. EMBO Rep. 2005, 6, 334. 57. Ehlert, F. J. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2008, 377, 549.
22. Pfleger, K. D.; Dromey, J. R.; Dalrymple, M. B.; Lim, E. M.; Thomas, W. G.; Eidne, 58. Griffin, M. T.; Figueroa, K. W.; Liller, S.; Ehlert, F. J. J. Pharmacol. Exp. Ther. 2007,
K. A. Cell. Signal. 2006, 18, 1664. 321, 1193.
23. Luttrell, L. M.; Gesty-Palmer, D. Pharmacol. Rev. 2010, 62, 305. 59. Black, J. W.; Leff, P. Proc. R. Soc. London, Ser. B 1983, 220, 141.
24. Shukla, A. K.; Manglik, A.; Kruse, A. C.; Xiao, K.; Reis, R. I.; Tseng, W. C.; Staus, 60. Kenakin, T.; Watson, C.; Muniz-Medina, V.; Christopoulos, A.; Novick, S. ACS
D. P.; Hilger, D.; Uysal, S.; Huang, L. Y.; Paduch, M.; Tripathi-Shukla, P.; Koide, Chem. Neurosci. 2012, 3, 193.
A.; Koide, S.; Weis, W. I.; Kossiakoff, A. A.; Kobilka, B. K.; Lefkowitz, R. J. Nature 61. Stahl, E. L.; Zhou, L.; Ehlert, F. J.; Bohn, L. M. Mol. Pharmacol. 2015, 87, 866.
2013, 497, 137. 62. Shonberg, J.; Klein Herenbrink, C.; Lopez Munoz, L.; Christopoulos, A.;
25. Gurevich, V. V.; Gurevich, E. V. Pharmacol. Ther. 2006, 110, 465. Scammells, P. J.; Capuano, B.; Lane, J. R. J. Med. Chem. 2013.
26. Nobles, K. N.; Guan, Z.; Xiao, K.; Oas, T. G.; Lefkowitz, R. J. J. Biol. Chem. 2007, 63. Lovell, K. M.; Frankowski, K. J.; Stahl, E. L.; Slauson, S. R.; Yoo, E.; Prisinzano, T.
282, 21370. E.; Aube, J.; Bohn, L. M. ACS Chem. Neurosci. 2015, 6, 1411.
27. Han, M.; Gurevich, V. V.; Vishnivetskiy, S. A.; Sigler, P. B.; Schubert, C. Structure 64. Zhou, L.; Lovell, K. M.; Frankowski, K. J.; Slauson, S. R.; Phillips, A. M.;
2001, 9, 869. Streicher, J. M.; Stahl, E.; Schmid, C. L.; Hodder, P.; Madoux, F.; Cameron, M.
28. Kang, Y.; Zhou, X. E.; Gao, X.; He, Y.; Liu, W.; Ishchenko, A.; Barty, A.; White, T. D.; Prisinzano, T. E.; Aube, J.; Bohn, L. M. J. Biol. Chem. 2013, 288, 36703.
A.; Yefanov, O.; Han, G. W.; Xu, Q.; de Waal, P. W.; Ke, J.; Tan, M. H.; Zhang, C.; 65. Keov, P.; Lopez, L.; Devine, S. M.; Valant, C.; Lane, J. R.; Scammells, P. J.; Sexton,
Moeller, A.; West, G. M.; Pascal, B. D.; Van Eps, N.; Caro, L. N.; Vishnivetskiy, S. P. M.; Christopoulos, A. J. Biol. Chem. 2014, 289, 23817.
A.; Lee, R. J.; Suino-Powell, K. M.; Gu, X.; Pal, K.; Ma, J.; Zhi, X.; Boutet, S.; 66. Luttrell, L. M.; Maudsley, S.; Bohn, L. M. Mol. Pharmacol. 2015, 88, 579.
Williams, G. J.; Messerschmidt, M.; Gati, C.; Zatsepin, N. A.; Wang, D.; James, 67. Schmid, C. L.; Raehal, K. M.; Bohn, L. M. Proc. Natl. Acad. Sci. U.S.A. 2008, 1079,
D.; Basu, S.; Roy-Chowdhury, S.; Conrad, C. E.; Coe, J.; Liu, H.; Lisova, S.; Kupitz, 105.
C.; Grotjohann, I.; Fromme, R.; Jiang, Y.; Tan, M.; Yang, H.; Li, J.; Wang, M.; 68. Groer, C. E.; Schmid, C. L.; Jaeger, A. M.; Bohn, L. M. J. Biol. Chem. 2011, 286,
Zheng, Z.; Li, D.; Howe, N.; Zhao, Y.; Standfuss, J.; Diederichs, K.; Dong, Y.; 31731.
Potter, C. S.; Carragher, B.; Caffrey, M.; Jiang, H.; Chapman, H. N.; Spence, J. C.; 69. Rajagopal, S.; Kim, J.; Ahn, S.; Craig, S.; Lam, C. M.; Gerard, N. P.; Gerard, C.;
Fromme, P.; Weierstall, U.; Ernst, O. P.; Katritch, V.; Gurevich, V. V.; Griffin, P. Lefkowitz, R. J. Proc. Natl. Acad. Sci. U.S.A. 2010, 107, 628.
R.; Hubbell, W. L.; Stevens, R. C.; Cherezov, V.; Melcher, K.; Xu, H. E. Nature 70. Kohout, T. A.; Nicholas, S. L.; Perry, S. J.; Reinhart, G.; Junger, S.; Struthers, R. S.
2015, 523, 561. J. Biol. Chem. 2004, 279, 23214.
29. Luttrell, L. M.; Ferguson, S. S.; Daaka, Y.; Miller, W. E.; Maudsley, S.; Della 71. Zidar, D. A.; Violin, J. D.; Whalen, E. J.; Lefkowitz, R. J. Proc. Natl. Acad. Sci. U.S.A.
Rocca, G. J.; Lin, F.; Kawakatsu, H.; Owada, K.; Luttrell, D. K.; Caron, M. G.; 2009, 106, 9649.
Lefkowitz, R. J. Science 1999, 283, 655. 72. Kenakin, T. J. Pharmacol. Exp. Ther. 2011, 336, 296.
30. Seo, J.; Tsakem, E. L.; Breitman, M.; Gurevich, V. V. J. Biol. Chem. 2011, 286, 73. Brust, T. F.; Hayes, M. P.; Roman, D. L.; Watts, V. J. Biochem. Pharmacol. 2015,
27894. 93, 85.
31. Chavkin, C.; Schattauer, S. S.; Levin, J. R. Handb. Exp. Pharmacol. 2014, 219, 281. 74. Wei, H.; Ahn, S.; Shenoy, S. K.; Karnik, S. S.; Hunyady, L.; Luttrell, L. M.;
32. Schmid, C. L.; Bohn, L. M. J. Neurosci. 2010, 30, 13513. Lefkowitz, R. J. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 10782.
33. Schmid, C. L.; Streicher, J. M.; Groer, C. E.; Munro, T. A.; Zhou, L.; Bohn, L. M. J. 75. Kenakin, T. Mol. Pharmacol. 2015.
Biol. Chem. 2013, 288, 22387. 76. Correll, C. C.; McKittrick, B. A. J. Med. Chem. 2014, 57, 6887.
34. Ahn, S.; Shenoy, S. K.; Wei, H.; Lefkowitz, R. J. J. Biol. Chem. 2004, 279, 35518. 77. Shonberg, J.; Lopez, L.; Scammells, P. J.; Christopoulos, A.; Capuano, B.; Lane, J.
35. Rajagopal, S.; Rajagopal, K.; Lefkowitz, R. J. Nat. Rev. Drug Disc. 2010, 9, 373. R. Med. Res. Rev. 2014, 34, 1286.
36. Rahmeh, R.; Damian, M.; Cottet, M.; Orcel, H.; Mendre, C.; Durroux, T.; 78. Bohn, L. M.; Gainetdinov, R. R.; Lin, F. T.; Lefkowitz, R. J.; Caron, M. G. Nature
Sharma, K. S.; Durand, G.; Pucci, B.; Trinquet, E.; Zwier, J. M.; Deupi, X.; Bron, 2000, 408, 720.
P.; Baneres, J. L.; Mouillac, B.; Granier, S. Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 79. Bohn, L. M.; Lefkowitz, R. J.; Gainetdinov, R. R.; Peppel, K.; Caron, M. G.; Lin, F.
6733. T. Science 1999, 286, 2495.
37. Liu, J. J.; Horst, R.; Katritch, V.; Stevens, R. C.; Wuthrich, K. Science 2012, 335, 80. Raehal, K. M.; Walker, J. K.; Bohn, L. M. J. Pharmacol. Exp. Ther. 2005, 314, 1195.
1106. 81. Li, Y.; Liu, X.; Liu, C.; Kang, J.; Yang, J.; Pei, G.; Wu, C. Int. J. Mol. Sci. 2009, 10,
38. Rasmussen, S. G.; DeVree, B. T.; Zou, Y.; Kruse, A. C.; Chung, K. Y.; Kobilka, T. S.; 954.
Thian, F. S.; Chae, P. S.; Pardon, E.; Calinski, D.; Mathiesen, J. M.; Shah, S. T.; 82. Yang, C. H.; Huang, H. W.; Chen, K. H.; Chen, Y. S.; Sheen-Chen, S. M.; Lin, C. R.
Lyons, J. A.; Caffrey, M.; Gellman, S. H.; Steyaert, J.; Skiniotis, G.; Weis, W. I.; Br. J. Anaesth. 2011, 107, 774.
Sunahara, R. K.; Kobilka, B. K. Nature 2011, 477, 549. 83. Pasternak, G. W. J. Clin. Oncol. 2014, 32, 1655.
39. Westfield, G. H.; Rasmussen, S. G.; Su, M.; Dutta, S.; DeVree, B. T.; Chung, K. Y.; 84. Massotte, D.; Kieffer, B. L. Essays Biochem. 1998, 33, 65.
Calinski, D.; Velez-Ruiz, G.; Oleskie, A. N.; Pardon, E.; Chae, P. S.; Liu, T.; Li, S.; 85. Gaveriaux-Ruff, C.; Kieffer, B. L. Neuropeptides 2002, 36, 62.
Woods, V. L., Jr.; Steyaert, J.; Kobilka, B. K.; Sunahara, R. K.; Skiniotis, G. Proc. 86. Roth, B. L.; Baner, K.; Westkaemper, R.; Siebert, D.; Rice, K. C.; Steinberg, S.;
Natl. Acad. Sci. U.S.A. 2011, 108, 16086. Ernsberger, P.; Rothman, R. B. Proc. Natl. Acad. Sci. U.S.A. 2002, 99, 11934.
40. Nobles, K. N.; Xiao, K.; Ahn, S.; Shukla, A. K.; Lam, C. M.; Rajagopal, S.; 87. Groer, C. E.; Tidgewell, K.; Moyer, R. A.; Harding, W. W.; Rothman, R. B.;
Strachan, R. T.; Huang, T. Y.; Bressler, E. A.; Hara, M. R.; Shenoy, S. K.; Gygi, S. Prisinzano, T. E.; Bohn, L. M. Mol. Pharmacol. 2007, 71, 549.
P.; Lefkowitz, R. J. Sci. Signal. 2011, 4, 1. 88. Tidgewell, K.; Harding, W. W.; Lozama, A.; Cobb, H.; Shah, K.; Kannan, P.;
41. Shukla, A. K.; Violin, J. D.; Whalen, E. J.; Gesty-Palmer, D.; Shenoy, S. K.; Dersch, C. M.; Parrish, D.; Deschamps, J. R.; Rothman, R. B.; Prisinzano, T. E. J.
Lefkowitz, R. J. Proc. Natl. Acad. Sci. U.S.A. 2008, 105, 9988. Nat. Prod. 2006, 69, 914.
42. Wacker, D.; Wang, C.; Katritch, V.; Han, G. W.; Huang, X. P.; Vardy, E.; 89. Lamb, K.; Tidgewell, K.; Simpson, D. S.; Bohn, L. M.; Prisinzano, T. E. Drug
McCorvy, J. D.; Jiang, Y.; Chu, M.; Siu, F. Y.; Liu, W.; Xu, H. E.; Cherezov, V.; Alcohol Depend. 2012, 121, 181.
Roth, B. L.; Stevens, R. C. Science 2013, 340, 615. 90. Chen, X. T.; Pitis, P.; Liu, G.; Yuan, C.; Gotchev, D.; Cowan, C. L.; Rominger, D.
43. Wang, C.; Jiang, Y.; Ma, J.; Wu, H.; Wacker, D.; Katritch, V.; Han, G. W.; Liu, W.; H.; Koblish, M.; Dewire, S. M.; Crombie, A. L.; Violin, J. D.; Yamashita, D. S. J.
Huang, X. P.; Vardy, E.; McCorvy, J. D.; Gao, X.; Zhou, X. E.; Melcher, K.; Zhang, Med. Chem. 2013, 56, 8019.
C.; Bai, F.; Yang, H.; Yang, L.; Jiang, H.; Roth, B. L.; Cherezov, V.; Stevens, R. C.; 91. DeWire, S. M.; Yamashita, D. S.; Rominger, D. H.; Liu, G.; Cowan, C. L.; Graczyk,
Xu, H. E. Science 2013, 340, 610. T. M.; Chen, X. T.; Pitis, P. M.; Gotchev, D.; Yuan, C.; Koblish, M.; Lark, M. W.;
44. Warne, T.; Edwards, P. C.; Leslie, A. G.; Tate, C. G. Structure 2012, 20, 841. Violin, J. D. J. Pharmacol. Exp. Ther. 2013, 344, 708.
250 Z. Rankovic et al. / Bioorg. Med. Chem. Lett. 26 (2016) 241–250

92. Soergel, D. G.; Subach, R. A.; Sadler, B.; Connell, J.; Marion, A. S.; Cowan, C. L.; 107. Weiss, J. M.; Morgan, P. H.; Lutz, M. W.; Kenakin, T. P. J. Ther. Biol. 1996, 181,
Violin, J. D.; Lark, M. W. J. Clin. Pharmacol. 2014, 54, 351. 381.
93. Viscusi, E. R.; Webster, L.; Kuss, M.; Daniels, S.; Bolognese, J. A.; Zuckerman, S.; 108. Deupi, X.; Kobilka, B. K. Physiology (Bethesda) 2010, 25, 293.
Soergel, D. G.; Subach, R. A.; Cook, E.; Skobieranda, F. Pain 2015. 109. Katritch, V.; Cherezov, V.; Stevens, R. C. Annu. Rev. Pharmacol. Toxicol. 2013, 53,
94. Masri, B.; Salahpour, A.; Didriksen, M.; Ghisi, V.; Beaulieu, J.; Gainetdinov, R.; 531.
Caron, M. Proc. Natl. Acad. Sci. U.S.A. 2008, 105, 13656. 110. Kenakin, T. BMC Pharmacol. Toxicol. 2012, 13, 3.
95. Kapur, S.; Mamo, D. Prog. Neuropsychopharmacol. Biol. Psychiatry 2003, 1081, 111. Venkatakrishnan, A. J.; Deupi, X.; Lebon, G.; Tate, C. G.; Schertler, G. F.; Babu,
27. M. M. Nature 2013, 494, 185.
96. Chen, X.; Sassano, M. F.; Zheng, L.; Setola, V.; Chen, M.; Bai, X.; Frye, S. V.; 112. Bohn, L. M.; Dykstra, L. A.; Lefkowitz, R. J.; Caron, M. G.; Barak, L. S. Mol.
Wetsel, W. C.; Roth, B. L.; Jin, J. J. Med. Chem. 2012, 55, 7141. Pharmacol. 2004, 66, 106.
97. Allen, J.; Yost, J.; Setola, V.; Chen, X.; Sassano, M.; Chen, M.; Peterson, S.; 113. Bruchas, M. R.; Land, B. B.; Aita, M.; Xu, M.; Barot, S. K.; Li, S.; Chavkin, C. J.
Yadav, P.; Huang, X.; Feng, B.; Jensen, N.; Che, X.; Bai, X.; Frye, S.; Wetsel, W.; Neurosci. 2007, 27, 11614.
Caron, M.; Javitch, J.; Roth, B.; Jin, J. Proc. Natl. Acad. Sci. U.S.A. 2011, 108, 114. Bruchas, M. R.; Macey, T. A.; Lowe, J. D.; Chavkin, C. J. Biol. Chem. 2006, 281,
18488. 18081.
98. Agai-Csongor, E.; Domany, G.; Nogradi, K.; Galambos, J.; Vago, I.; Keseru, G. 115. Raehal, K. M.; Schmid, C. L.; Groer, C. E.; Bohn, L. M. Pharmacol. Rev. 2011,
M.; Greiner, I.; Laszlovszky, I.; Gere, A.; Schmidt, E.; Kiss, B.; Vastag, M.; 1001, 63.
Tihanyi, K.; Saghy, K.; Laszy, J.; Gyertyan, I.; Zajer-Balazs, M.; Gemesi, L.; 116. Whalen, E.; Rajagopal, S.; Lefkowitz, R. Trends Mol. Med. 2011, 17, 126.
Kapas, M.; Szombathelyi, Z. Bioorg. Med. Chem. Lett. 2012, 22, 3437. 117. Shukla, A. K.; Singh, G.; Ghosh, E. Trends Biochem. Sci. 2014, 39, 594.
99. Tschammer, N.; Bollinger, S.; Kenakin, T.; Gmeiner, P. Mol. Pharmacol. 2011, 118. McPherson, J.; Rivero, G.; Baptist, M.; Llorente, J.; Al-Sabah, S.; Krasel, C.;
79, 575. Dewey, W. L.; Bailey, C. P.; Rosethorne, E. M.; Charlton, S. J.; Henderson, G.;
100. Skeggs, L. T.; Dorer, F. E.; Kahn, J. R.; Lentz, K. E.; Levine, M. Am. J. Med. 1976, Kelly, E. Mol. Pharmacol. 2010, 78, 756.
60, 737. 119. Alhaddad, H.; Cisternino, S.; Decleves, X.; Tournier, N.; Schlatter, J.; Chiadmi,
101. Burnier, M. Circulation 2001, 103, 904. F.; Risede, P.; Smirnova, M.; Besengez, C.; Scherrmann, J. M.; Baud, F. J.;
102. Rajagopal, K.; Whalen, E. J.; Violin, J. D.; Stiber, J. A.; Rosenberg, P. B.; Premont, Megarbane, B. Crit. Care Med. 2012, 40, 3215.
R. T.; Coffman, T. M.; Rockman, H. A.; Lefkowitz, R. J. Proc. Natl. Acad. Sci. U.S.A. 120. Laugwitz, K. L.; Allgeier, A.; Offermanns, S.; Spicher, K.; Van Sande, J.; Dumont,
2006, 103, 16284. J. E.; Schultz, G. Proc. Natl. Acad. Sci. U.S.A. 1996, 93, 116.
103. Violin, J. D.; DeWire, S. M.; Yamashita, D.; Rominger, D. H.; Nguyen, L.; 121. Offermanns, S.; Wieland, T.; Homann, D.; Sandmann, J.; Bombien, E.; Spicher,
Schiller, K.; Whalen, E. J.; Gowen, M.; Lark, M. W. J. Pharmacol. Exp. Ther. 2010, K.; Schultz, G.; Jakobs, K. H. Mol. Pharmacol. 1994, 45, 890.
335, 572. 122. Dupre, D. J.; Robitaille, M.; Rebois, R. V.; Hebert, T. E. Annu. Rev. Pharmacol.
104. Kim, K. S.; Abraham, D.; Williams, B.; Violin, J. D.; Mao, L.; Rockman, H. A. Am. Toxicol. 2009, 49, 31.
J. Physiol. Heart Circ. Physiol. 2012, 303, H1001. 123. Khan, S. M.; Sleno, R.; Gora, S.; Zylbergold, P.; Laverdure, J. P.; Labbe, J. C.;
105. Violin, J. D.; Crombie, A. L.; Soergel, D. G.; Lark, M. W. Trends Pharmacol. Sci. Miller, G. J.; Hebert, T. E. Pharmacol. Rev. 2013, 65, 545.
2014, 35, 308. 124. Lin, Y.; Smrcka, A. V. Mol. Pharmacol. 2011, 80, 551.
106. Stephenson, R. P. Br. J. Pharmacol. Chemother. 1956, 11, 379.

You might also like