You are on page 1of 14

ANALYSIS

Trends in GPCR drug discovery:


new agents, targets and indications
Alexander S. Hauser1, Misty M. Attwood2, Mathias Rask-Andersen3, Helgi B. Schiöth2
and David E. Gloriam1
Abstract | G protein-coupled receptors (GPCRs) are the most intensively studied drug targets,
mostly due to their substantial involvement in human pathophysiology and their pharmacological
tractability. Here, we report an up-to-date analysis of all GPCR drugs and agents in clinical trials,
which reveals current trends across molecule types, drug targets and therapeutic indications,
including showing that 475 drugs (~34% of all drugs approved by the US Food and Drug
Administration (FDA)) act at 108 unique GPCRs. Approximately 321 agents are currently in
clinical trials, of which ~20% target 66 potentially novel GPCR targets without an approved drug,
and the number of biological drugs, allosteric modulators and biased agonists has increased.
The major disease indications for GPCR modulators show a shift towards diabetes, obesity and
Alzheimer disease, although several central nervous system disorders are also highly represented.
The 224 (56%) non-olfactory GPCRs that have not yet been explored in clinical trials have broad
untapped therapeutic potential, particularly in genetic and immune system disorders. Finally,
we provide an interactive online resource to analyse and infer trends in GPCR drug discovery.

Most drug targets are members of one of the follow- increased with the concept of biased agonism7,8 — pref-
Allosteric sites
Sites for ligand binding to a ing five protein families: G protein-coupled receptors erentially activating the desired intracellular signalling
receptor that are remote from (GPCRs), ion channels, kinases, nuclear hormone recep- pathway while minimizing undesired side effects due to
the binding site of the tors or proteases1. GPCRs have been of long-standing the activation of other pathways. So far, drugs targeting
physiological agonist (known as interest as pharmacological targets, as they regulate GPCRs have generally been developed without the help
the orthosteric site).
numerous diverse physiological processes and have of high-resolution structural information, but the crys-
druggable sites that are accessible at the cell surface. tal structures of 44 unique receptors and 205 ligand–
Drugs that target GPCRs also account for ~27% of the receptor complexes now provide a plethora of templates
global market share of therapeutic drugs, with aggre- for structure-based drug discovery and design. Of the
gated sales for 2011–2015 of ~US$890 billion2. GPCRs non-olfactory GPCRs, 130 (33%) are peptide or pro-
form the largest human membrane protein family, with tein receptors that are potential targets for biological
~800 members overall (of which ~400 are olfactory recep- drugs, and many more receptors have become acces-
1
Department of Drug Design
and Pharmacology, Faculty of tors that are not included in this analysis). The GPCR sible through advances in techniques that exploit nat-
Health and Medical Sciences, superfamily has been sub-divided into classes3 based on ural protein–protein interactions or that help develop
University of Copenhagen, evolutionary homology and receptor families4 with com- specific antibodies and derivatives. This has led to new
2100 Copenhagen, Denmark. mon physiological ligands, which are strikingly diverse: opportunities for both novel and established GPCR targets.
2
Department of Neuroscience,
Functional Pharmacology,
spanning ions, small molecular signalling molecules, Recent reviews on GPCRs have described drug target
University of Uppsala, lipids, peptides and proteins5. families9–11, disease indications12–14, novel tools15,16 or drug
751 05 Uppsala, Sweden. New avenues for GPCR drug discovery have emerged discovery mechanisms17–20. Here, we report an analysis
3
Department of Immunology, owing to recent advances in receptor pharmacology, of all GPCR drugs and agents in clinical trials, which
Genetics and Pathology,
breakthroughs in structural biology and innovations in reveals the current trends across all molecule types, drug
Science for Life Laboratory,
University of Uppsala, biotechnology. Modulation of GPCRs via allosteric sites, targets and therapeutic indications. General drug dis-
751 05 Uppsala, Sweden. which are distinct from binding sites for endogenous covery trends, such as biologics, allosteric modulators,
Correspondence to D.E.G.  ligands, can alter the structure, dynamics and function repurposing, GPCR structure data and ligands with
david.gloriam@sund.ku.dk of the receptor to achieve a potential therapeutic advan- biased signalling, are analysed to investigate whether
doi:10.1038/nrd.2017.178 tage, such as increased spatial and temporal selectivity 6. associated agents have reached clinical trials. We also
Published online 27 Oct 2017 Furthermore, our knowledge of receptor activation has outline the novel targets that are closest to reaching the

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 829


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Biased agonism market and assess the ‘dark matter’ of pharmaceuti- phase IV trials worldwide or that have been recently dis-
A newly discovered mechanism cally unexplored GPCR targets and unexploited disease continued. However, none of these resources is compre-
for potentially reducing drug associations. Finally, we provide our data for extended hensive with respect to the targets of agents in trials, which
side effects by using a analyses by the scientific community through a compre- can be collected from a combination of public resources,
surrogate agonist to
preferentially activate a
hensive public resource on GPCR drug data and statis- such as Drugbank 22, Pharos 23 and Open Targets 24
different intracellular signalling tics, and target mapping, which are integrated with the (which also has a disease ontology), the literature25–27
pathway from that of the GPCR database (GPCRdb)21. and company press releases.
physiological agonist. By manually curating CenterWatch’s Drugs in Clinical
GPCR drugs and agents in clinical trials Trials database (data extracted in July 2017) and cross-­
Established GPCR targets
Herein defined as the targets of
Numbers of GPCR-targeted agents. All drugs approved referencing with public sources, we were able to identify
a drug approved by the US in the United States are listed in the Drugs@FDA data- 475 approved drugs that target GPCRs. This accounts for
Food and Drug Administration. base. The primary resource for clinical trial registries is approximately 34% of all FDA-approved drugs, which is
the US National Institutes of Health (ClinicalTrials.gov), similar to previously reported estimates (27–33%) on the
which currently holds information on more than 256,000 number of GPCR-targeted drugs1,25,27. GPCR drug dis-
clinical trials worldwide. The current status of agents in covery has advanced rapidly. In the past 5 years, 69 new
clinical trials is available in commercial databases such as GPCR-targeting drugs have been approved by the US
CenterWatch’s Drugs in Clinical Trials database, which Food and Drug Administration (FDA) (see TABLE 1 for
covers more than 4,000 such agents that are in phase I to recent new molecular entities (NMEs)). The most recently

Table 1 | New molecular entities acting via GPCRs approved by the FDA since 2014
Substance Brand name Indications Targets* Approval
year
Droxidopa Northera Orthostatic hypotension ADRB1–3, ADA2A/B/C and 2014
ADA1A/B/D
Dulaglutide Trulicity Type 2 diabetes GLP1R 2014
Hydrocodone Hycodan Narcotic cough OPRD and OPRM 2014
bitartrate
Naloxegol Movantik Opioid-induced constipation OPRM 2014
Netupitant Akynzeo Nausea and/or vomiting NK1R 2014
Olodaterol Striverdi COPD ADRB2 2014
respimat
Suvorexant Belsomra Insomnia OX2R and OX1R 2014
Tasimelteon Hetlioz Non‑24‑hour sleep-wake disorder MTR1A and MTR1B 2014
Vorapaxar Zontivity Cardiovascular risk reduction PAR1 2014
Aripiprazole Aristada Schizophrenia 5HT1A, 5HT2A and DRD2 2015
lauroxil
Brexpiprazole Rexulti Depression 5HT2A, 5HT1A, DRD2 and 2015
5HT7R
Cangrelor Kengreal Percutaneous coronary intervention P2Y12 2015
Cariprazine Vraylar Schizophrenia and bipolar disorder DRD3 and DRD2 2015
Eluxadoline Viberzi Irritable bowel syndrome OPRM and OPRD 2015
Flibanserin Addyi Hypoactive sexual desire disorder 5HT2A and 5HT1A 2015
Parathyroid Natpara Hypoparathyroidism PTH1R and PTH2R 2015
hormone
Rolapitant Varubi Nausea and/or vomiting NK1R 2015
Selexipag Uptravi Pulmonary hypertension PI2R 2015
Sonidegib Odomzo Basal cell carcinoma SMO 2015
Lixisenatide Adlyxin Type 2 diabetes GLP1R 2016
Pimavanserin Nuplazid Parkinson disease psychosis 5HT2A 2016
Abaloparatide Tymlos Osteoporosis PTHR1 2017
Etelcalcetide Parsabiv Hyperparathyroidism CASR 2017
Naldemedine Symproic Opioid-induced constipation OPRM 2017
COPD, chronic obstructive pulmonary disease; GPCR, G protein-coupled receptor. *Listed using the protein name in UniProt; for
details on receptor nomenclature, see the IUPHAR/BPS Guide to PHARMACOLOGY (see Further information)

830 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

approved GPCR-targeted drug is abaloparatide28, a para­ families and classes. This suggests that general new
thyroid hormone (PTH1) receptor agonist that is used approaches, such as increased tractability with biolog-
to treat postmenopausal women with osteoporosis. The ics, have contributed to the overall increased thera­peutic
number of agents currently in clinical trials is 321 (68% targeting.
of the total number of approved drugs). Interestingly, 60
(19%) of the agents in trials target potentially novel GPCR Orphan receptors enter clinical trials. Interestingly,
targets for which a drug is not currently approved. current clinical trials feature several orphan GPCRs, for
To study the distribution of established and currently which endogenous ligands have not yet been discovered.
investigated drug targets, we mapped approved drugs These orphan GPCRs serve as potentially novel targets
and clinical trial candidates onto the GPCR super­family for the treatment of a diverse set of indications, such as
tree (FIGS 1,2). The 475 approved drugs mediate their GPR119 for the treatment of diabetes, leucine-­rich repeat-­
effects via 108 GPCR targets, accounting for 27% of the containing GPCR 4 (LGR4) and LGR5 for the treatment
human non-olfactory GPCRs. Aminergic receptors, of gastrointestinal disease, GPR35 for the treatment of an
which are all established drug targets, are targeted by allergic inflammatory condition, GPR55 as an antispas-
314 of the approved drugs. Currently established GPCR modic target, the proto-oncogene Mas (MAS) for the
drug targets are used by an average of 10.3 (median = 4) treatment of thrombocytopenia and GPR84 for the treat-
distinct approved agents. This indicates a near satura- ment of ulcerative colitis. This indicates that the drug
tion of the current target space, and emphasizes the need discovery process can still take place, and even advance,
to identify new druggable receptors in order to develop despite limited knowledge about the endogenous ligand
novel medications. The identification and exploitation of or ligands and/or signalling pathways. The entry of an
new targets is especially warranted for diseases with large agent and target into clinical trials serves as an important
unmet medical needs and few current viable targets, such qualifier, as it shows that animal studies, other preclin-
as Alzheimer disease (AD). ical data and disease relevance are promising enough
to involve humans in subsequent investigations. It can
Success rates of trials for GPCR-targeted agents. The FDA be expected that public information on orphan recep-
recently reported 70%, 33% and 25–30% success rates for tors will increase as candidate drugs progress through
phase I, II and III clinical trials, respectively, for all tar- clinical trials and/or upon approval.
get families (see Further information). We estimated the
success rates of GPCR-targeted agents in 2013–2017 (up Trends in agent type and mode of action
to July 2017), by counting successes as agents that suc- The proportion of GPCR-targeted biologics is increasing
cessfully reached at least one subsequent phase, and we in early stage clinical trials. Biopharmaceuticals such as
counted failures as agents that were reported to be discon- monoclonal antibodies (mAbs) are now well-established
tinued, terminated or withdrawn, or completed before this therapeutic modalities, accounting for 27–33% of the new
period but never progressed. This yielded GPCR-targeted drugs approved by the FDA in the years 2014 to 2016
agent success rates of 78%, 39% and 29% for phases I, II (REF. 30). Biologics have mostly targeted proteins such as
and III, respectively — slightly higher than the FDA’s aver- cytokines and their receptors, with no GPCR-targeted
age for all investigated agents. This may reflect the high mAbs yet approved by the FDA, although some are close
level of experience in targeting GPCRs. Both our and the to approval31. With 16 agents currently in clinical trials,
FDA’s analyses show a clear trend of more failures in later targeting GPCRs with mAb-based therapeutics might
phases. A recent example is the small-molecule drug fasig- have great therapeutic potential in various diseases,
lifam, an agonist of the free fatty acid 1 (FFA1) receptor including cancer, inflammatory disorders, and neuro­
(also known as GPR40). Fasiglifam showed efficacy in a logical and metabolic diseases. For example, targeting the
phase III trial for diabetes29, but further development was CGRP receptor with mAbs provides a new approach for
terminated due to hepatotoxicity. the treatment of chronic migraine. The CGRP receptor-­
specific mAb erenumab prevents the ligand CGRP from
Popularity of peptide or protein receptors. We identified binding, and has now been submitted to the FDA for
66 potentially novel GPCR targets in clinical trials; that approval based on positive data in phase III trials (three
is, targets that are not yet modulated by approved drugs. other mAbs targeting CGRP rather than its receptor are
Of these, 37 are peptide or protein-activated GPCRs (a also in late-stage development).
selection of which are shown in TABLE 2), including the Several peptide drugs targeting the glucagon-like
calcitonin gene-related peptide (CGRP) receptor for peptide 1 (GLP1) receptor have been approved for type 2
the treatment of migraine, the GPR55 receptor for the treat- diabetes, including exenatide, liraglutide, lixisenatide,
ment of epilepsy and the apelin receptor for the treatment albiglutide and dulaglutide32, and peptide drugs targeting
of cardiovascular disorders. Furthermore, the chemo­kine other GPCRs are currently in development. For example,
receptors alone have 22 agents in clinical trials for the bremelanotide is a non-­selective peptide melano­cortin
treatment of cancer, asthma, rheumatoid arthritis, chronic receptor agonist that has successfully completed two
obstructive pulmonary disease (COPD) and HIV. The six phase III trials for female sexual dysfunction, and a new
members of the glucagon receptor family are also a focus drug application (NDA) is expected to be submitted
of new approaches for the treatment of type 2 diabetes. to the FDA for approval in 2018. Setmelanotide —
Notably, although all recognize natural peptides or pro- another melanocortin receptor agonist that is selective
tein ligands, these targets span a breadth of receptor for the MC4 receptor — has shown promising results in

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 831


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

T2R60
T2R50
T2R46
T2R45
T2R43

5HT1D
5HT1A
T2R42

5HT1B
T2R41

5HT1E
T2R40

5HT2A
5HT1F
T2R39

5HT2C
5HT2B
T2R38

5HT4R
T2R31

5HT5A
T2R30

5HT6R
T2R209

5HT7R
T2R1
T2R164
T2R1 3

ACM 1
T2R1

ACM 2
ACM 3
T2R109

ACM

ACM54
TA2R 8

ADAA1A
TA2RR7
TA2 R5

AD 1B
AD A1D
TA2 R4

ADAA2A
TA2 3
TA2RR1

AD 2B
AD A2C
TA2

AD

AD RB1
ADRRB2
ine
FZD 9

B3
SMO
FZDD8
FZ D7

DR D1
FZ D6

D D2
10
FZ D5

DRRD3
tam
FZ 4

D R D4
DR
FZDD3

D5
FZ D2
FZ D1

H RH1
FZ

HRRH2
GPPC5DC

RH 3
G PC5 B

4
H
G PC5 I3

5-Hydroxytryp
C6

H
(muscaholine
G RA 79

1
AR
A

s
rinic)

1
GP 158 6

AGGTR
ptor

2
TA
GP 15

TR
1
GP

A
T S S 1R 1

BR RPRBR
T S1R

AP
1R
T

noce
Acetylc

G M
Taste 2
3

S3
N
2
GR RM 6

RB 1
Cla

BK KRB
G M 5

G A CK 2
M 7

ine
GR RMM4

Friz

R R
G R M3

A
G R M2 1

Bo pelinoten ine
ss

Adre

Anrace ne
G R M

pam

S
C
sin
G R

CO

AR R
G

zled
G

1
A gi m
2

C5 3A
i
GA AB AR

t am

C
BR R2 C5

a
A
NR RB

Do
rph
CA 1

en inin
ED DN

Co hole ykin n
M glu

His
SR E

C ad esi
GP R1
et ta

m cy in
an
FP PR2 3

em k
R9

Ta

T
A

pl sto
ab m
8
Ca

Br mb
A GR F PR

s
1

t
F

ste
A GR G7 LR 2
ot at
AGGRGG6 lci GAALR R3
ro e
um

ic
A R 5 G AL

Sensory
lin tide
1
AGGRGG4 G
pi
SR
he

nerg
AG RG 3
-se GA GH HR
c
RG 2
ot lpep
1 R
AD nsin BA
A
AGGRB GN
d

Pro
SR
RB 3
BA 2
GR g En rm n y KIS

Ami
1
I1
in HR 2
MCCHR
Fo lani n roph
AG

Or
AD V

tein
AG RF M
AG RF45 HR
GR Se i t
Ga hrel adoptin
ph
AG RF3 MS THR
AG RF2
G C
Am

ns A C3R
RF1
AD G onspe
an
M C4R
AG o M C5R
GR G is
in

AG RD2 ry M
K CH rtin
RD
B
o

1 LR
MT
oco
CE
AD
ac

CELLR3
Io M UR1
CEL R2 GR lan
NM UR2
id

R1
F n e NM

Taste 2
AG AD M U F1
NPFFF2
GR tilin din
AG RA3
AF R1

Cla
AGRRA2 NP
A1 ADG D Mo urome ide FF/ NPS
RC t
ss
CD9 e
N rope ide S p 1
AGRE 7
Adh
Cl

NPBW
4 ADG u p t B NPBW
2

F
AGRE3
esio e
N urop e /
as

AGRE2
RA id e W
Ne ropept
AGRE 1R
n PY
sC

1 N
NPY2R
AGRL
AGRL34
ADGR
E Neu eY
NPY4RR
NPY5 R
ide peptid NPY6
Cla Pept
AGRL2
AGRL1
ADGRL ss B Neuro NTR1
NTR2
VIPR2
2 nsin
Neurote
VIPR1
PACR VIP and PA OPRD
CAP OPRK
OPRM
Opioid
PTH2R
OPRX
PTH1R Parathyroid OX1R
SCTR
GLR Orex in OX2R
GLP2R
GLP1R Glucagon Peptide Class B1 Peptide P518 QRFPR
GIPR
GHRHR
family Prolactin PRLHR

CRFR2 Corticotropin Proteinase- PAR1


PAR2
CRFR1 activated
Calcitonin
PAR3
PAR4
CALRL Relaxin fa
CALCR mily RXFP1
RXFP2
TAAR9
TAAR8 Cl Somato
RL3R1
RL3R2
TAAR65 as statin
TAAR 3 sA SSR1
SSR2
TAAR 2
TAAR 0 Tach SSR3
Thyr ykinin
P2Y1 8 SSR4
P2RYX4 SSR5
MRG X3 Ur o tro
Va otensi pin
NK1R
MRG X2 NK
MRGGX1
oxysopres n
NK32R
MR RG
MRGGRF Pr t
R
ot Ch ocin sin/ TRFR
MR GRE
MR GRD an ein em
ph
UR2
MR AS1L erin R

Or
M MAS V1
6 V1 AR
LGRR5 V2 BR
LG R4 OX R
LG 83 YR
GP1182 Ch CM
L1
P
G 176
GPP174
G 173
em C
C CR1
ok C CR2
GPP171
ns CCCR3
Lip

G 162
GPP1610 a ine C R4
ph C CR5
ory
cid

G P16 3 C CR6
or
id
roid

Melatonin
Nucleotide

G P15 2 C CR
G P15 1 C CR 7
sA CCCR9 8
Sens

G P15 0
xylic a

as
Gl okin tty a

G P15 9 C R
C XC 10
Cl
G P14 8
Ste

yc e
Pr e fa

G P14 6 C XC R1
G P14 2 C XC R2
op tic

G P14 41 C XC R3
Fr

CX XCR R4
ro n

G P1 39
e

G P1 35 C C 5
acid xycarboxylic Alicarbo

X X3 R6
te

G P1 32
Le

G P1 01 A CR C1
in

G P1 88 A CK 1
A C R
uk

G PR 87 AC CK KR2 1
Lys

G PR 85 4 CC K R3
otr

G PR 8 3 R
G PR R8 2 FS RL2 4
op
Lys

cid

G P R8 HR
ien
Can 8/55/11

G P
LS H
ins
O PR7 78

GPR t-activat

ho

G
TS R1
HR R
G PS R 5

Plate
G PR

oph
Oxoglu ate
GP PR YR 1

e
tarate

Prosta
G

PK KR2
G

Estrogen
Ops

sph

P
Bile acid

nab
G R 63

P2Y
G PR 62

FF R 3
Succin
M PR 61

Adenosine

osp
Melatonin

FF FAR R4
AR 2
G TR 52

F FA 42
oli
le
G PR 1L

A
1
F PR
i
ET PR3 45

G
n
G BR 9

hol

pid
noid
G PR 2

LT T4R2 1
G PR 37

L R
G PR3 35

4R
id

CL LTR2R1
G PR3 4

C XE

1
GPPR3 3

ipid S1 NR1

T
O
G R3 2
G PR2 1

LPA R2
G PR2 7

LPAAR34
G PR2 6

LP AR
9
GPPR225

LP AR5

R1
LP AR6
Hydro
G R21

LP
GPPR20
GP R19

S1PPR2
ing
GP R17

S1 PR3
GP R15

S1 PR4

Number of drugs
R12

S1 PR5
GP R6

R1
GP R4
GPRR3

C 2
1
GP

CNR
GPRR55
OP SR
OP SG

GP 119
GP
OPNSB
OPN 5
OP 3

PTAFR
OP

OPSND4

> 75
PD2R 2
PD2R 1

18
PE2R
1

PE2R23
PE2R 4
OXGR1

PE2R
PF2R
SUCR

PI2R
HCAR 3

TA2R
HCAR12

MTR1A
MTR1B
HCAR

AA1R
AA2AR
AA2BR
AA3R
GPER1

P2Y14
GPBAR

P2Y13
P2Y11
P2Y12

P2RY1

50–75
P2RY2
P2RY4
P2RY6

40–50
30–40
Target established 20–30
Target in trials III II I 5–10
<5

Figure 1 | GPCR drug targets. Established targets have approved drugs as 117 approved agents and 41 agents in trials, and so the red circle is larger,
defined in the Drugs@FDA database, and targets of agents in clinical trials whereas for chemokine receptors, there are 2 approved agents and 37
were collected from manual annotation of CenterWatch’s Drugs in Clinical agents in trials and so the green circle is Nature
larger. At the family
Reviews level,Discovery
| Drug agents that
Trials database, OpenTargets, Drugbank, Pharos and company press releases. modulate multiple receptors in the family are only counted once to
Established (red) and phase I–III (green) targets across the G protein-coupled determine the circle size. For individual receptors (but not families), different
receptor (GPCR) classes, ligand types and receptor families (from the centre shades of green are used for each trial phase. An interactive tree that
to the outer ring) are shown. The sizes of the circles represent the number of includes the number of agents for each target is available at http://www.
agents. For receptor families, two concentric circles are superimposed: a red gpcrdb.org/drugs/drugmapping. MCH, melanin-concentrating hormone;
circle indicating the number of approved agents (that is, which have an PACAP, pituitary adenylate cyclase-activating peptide; VIP, vasoactive
established target in that family) and a green circle indicating the number of intestinal peptide. GPCRs are listed using the protein name in UniProt; for
agents in trials for the targets in that family. The area over which the two details on receptor nomenclature, see the IUPHAR/BPS Guide to
circles overlap is shown in brown. For example, for adrenoceptors, there are PHARMACOLOGY (see Further information).

832 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Established target families In-trial target families

Other Neuropeptide Y (phase II)


Opioid Dopamine Non-targeted (phase III) Class A orphans (phase III)
Other Acetylcholine Ghrelin (phase III)
4%
7% 5%
23% 11% 5% Calcitonin
56%
32% 6% (phase III)
12% GPR18, GPR55
7% and GPR119
20% (phase II)
6%
12% 27% 17% Metabotropic
14% 20% 8% glutamate
(phase III)
In trials
Adrenoceptors Histamine 5-Hydroxtryptamine Established Chemokine Melanocortin
(phase III) (phase III)

Figure 2 | Major GPCR families that are established drug targets and targets of agents in clinical trials. There are
398 non-olfactory G protein-coupled receptors (GPCRs), of which 108 have drugs approved (red),
Nature 66 have
Reviews agents
| Drug that
Discovery
have reached clinical trials but that have not yet been approved (green) and 224 are yet to be targeted by agents in
clinical trials (grey). The 108 established GPCR drug targets (left-hand side) are primarily aminergic and opioid receptors,
whereas most new targets of agents that have reached clinical trials but not yet been approved are types of peptide
receptor (right-hand side).

phase II trials for the treatment of rare genetic disorders Increasing focus on target selectivity rather than poly­
of obesity, and is now in phase III development. Positive pharmacology. Many drugs are known to exert their
phase III data for a synthetic human angiotensin II pep- therapeutic effect through multiple targets; that is, poly-
tide LJPC‑501 in patients with catecholamine-­resistant pharmacology. To complement the previous studies by
vasodilatory shock has recently been reported33, and an Santos et al.27 and Overington et al.25, which assigned
NDA is expected to be submitted by the end of 2017. 1:1 relationships between approved drugs and their
Overall, there are early indications that modalities targets, our annotation covers all primary and second-
other than small molecules are becoming more popu- ary targets, including those for agents in clinical trials.
lar for agents targeting GPCRs, as the share of peptides, Interestingly, a trend was found for increasing selectivity
mAbs and other recombinant proteins31 being studied in (decreasing polypharmacology) for earlier trials (FIG. 3c).
phase I trials is higher than that in later phases (FIG. 3a). Furthermore, the currently investigated phase I agents
have fewer targets than the discontinued agents, further
More allosteric modulators in early stage clinical trials. suggesting an increasing focus on target selectivity rather
Allosteric modulators offer an attractive novel mecha- than on polypharmacology.
nism: remotely modulating the activity that is induced Polypharmacology could still lead to future oppor-
at the binding site of the physiological ligand. This often tunities with established targets, as specifically dis-
has advantages, such as higher selectivity at the target cussed for the dopaminergic system38,39. Drugs that
(perhaps by binding to a less well-conserved site across a exhibit polypharmacology at dopamine and serotonin
receptor family with the same endogenous ligand), tem- receptors include, for example, haloperidol, amoxa-
poral selectivity (action linked to the release of the endo­ pine and asenapine39. Amitriptyline, as an example
genous ligand) and sometimes also functional selectivity of a tricyclic antidepressant, also has high affinity at
through biased signalling 19, as shown for the FFA1 recep- muscarinic and histamine H1 receptors (which can be
tor34,35. Approved allosteric modulators include cinacalcet, useful therapeutically). Furthermore, antimuscarinics
a positive allosteric modulator of the calcium-­sensing used in bladder dysfunction are non-selective across
(CaS) receptor for the treatment of hyperparathyroidism, the range of five human receptors, although the pri-
and maraviroc, a negative allosteric modulator of the mary targets seem to be the muscarinic acetylcholine
chemokine receptor CCR5 for the prevention of cellular M2 and M3 receptors.
entry of HIV‑1. Allosteric modulation has also been sug-
gested as a viable option for the peptide hormone class B Trends in disease indications
receptors, which have not been tractable when targeting Our data show that the indications for GPCR-targeted
the orthosteric site18. The allosteric database currently agents are expanding from historically popular areas,
lists 27,769 GPCR-targeted allosteric modulators36, and such as hypertension, allergy, analgesics, schizo­
several positive and negative allosteric modulators are in phrenia and depression, into novel areas such as AD
phase I and phase II trials, respectively (FIG. 3b). There is and obesity (FIG. 4a). Furthermore, in the past 5 years,
also an indication that agonists may be more prevalent GPCRs have also been targeted for new indications,
Polypharmacology and that antagonists may be less prevalent in phase I including multiple sclerosis (MS), smoking cessation,
Ligand binding to multiple
targets, all of which contribute
trials. Interestingly, several of the allosteric modulators short bowel syndrome and hypocalcaemia. Major
to the pharmacological currently in clinical trials are combination therapies with trends in the indications of GPCR-targeted agents are
response. orthosteric ligands for the treatment of CNS disorders37. highlighted below.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 833


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Table 2 | New GPCR target families and late-stage targets for agents currently in clinical trials
Receptor family Targets* Indications Agents
Angiotensin AGTR2 Catecholamine resistant hypotension LJPC‑501
Apelin APJ Cardiovascular disorders and insulin Apelin
sensitivity
Bile acid GPBAR Liver fibrosis INT‑767
Calcitonin CALCRL Migraine Erenumab and ubrogepant
Chemerin CML1 Dry eye RX‑10045
Chemokine CCR2, CCR4 and HIV infection, cancer and type 1 Cenicriviroc, mogamulizumab and
CXCR1 diabetes reparixin
Class A orphans GPR84, GPR35, Ulcerative colitis, irritable bowel GLPG1205, PA101B, TXA127
MAS and LGR5 syndrome, autoimmune diseases, (angiotensin 1–7) and BNC101
multiple myeloma and colorectal
cancer
Complement peptide C5AR1 Autoimmune diseases CCX168
Free fatty acid FFAR2 Neutrophil-driven inflammation and GLPG0974
ulcerative colitis
GPR18, GPR55 and GPR55 Spasticity related to multiple VSN16R, cannabidiol (GWP42003)
GPR119 sclerosis and epilepsy and cannabidivarin (GWP42006)
Ghrelin GHSR Appetite stimulant, antidiabetic, Unacylated ghrelin (AZP‑531),
cancer cachexia, gastroparesis and anamorelin, macimorelin and
digestive system disease ulimorelin
LPA LPAR1 Pulmonary fibrosis AM‑152
Melanocortin MC1R, MC3R and Sexual dysfunction, anti-obesity and Bremelanotide, RM‑493 and
MC4R dermatological afamelanotide
Motilin MTLR Gastroparesis Camicinal
Prostanoid PD2R2 (GPR44) Asthma and allergic rhinitis Fevipiprant and setipiprant
Relaxin RXFP1 and RXFP2 Heart failure Serelaxin
Tachykinin NK3R Polycystic ovarian syndrome MLE-4901 and AZD4901
VIP and PACAP VIPR1 and VIPR2 Sexual dysfunction and hypertension Vasomera (PB1046) and vasoactive
intestinal peptide
GPCR, G protein-coupled receptor; LPA, lysophosphatidic acid; PACAP, pituitary adenylate cyclase-activating peptide;
VIP, vasoactive intestinal peptide. *Listed using the protein name in UniProt; for details on receptor nomenclature, see the
IUPHAR/BPS Guide to PHARMACOLOGY (see Further information).

Central nervous system disorders remain highly repre- drug has been approved: fingolimod, a sphingosine
sented among the indications of GPCR-targeted agents. 1‑­phosphate receptor 1 (S1P1) modulator, which reduces
Grouping the indications of approved GPCR-targeted relapse rates and the risk of disability progression43. Several
drugs into higher-level disease terms using the Open other S1P1 receptor modulators — ozanimod, ponesimod
Target 24 ontology shows that central nervous system and siponimod — are currently in phase II and phase III
(CNS) diseases are the most abundant, accounting trials. These drugs are expected to have advantages over
for 124 (26%) of all approved GPCR-targeted drugs. fingolimod, such as higher selectivity for the S1P1 recep-
Furthermore, at least 79 GPCR-targeted agents are cur- tor, faster clearance and improved tissue penetration44.
rently in clinical trials for CNS indications, demonstrat- Other MS targets include the cannabinoid receptors.
ing a continued strong interest. An analysis of receptor A tetrahydrocannabinol and cannabidiol (THC-CBD)
baseline expression from the human protein atlas40, and oromucosal spray has been tested in phase II and phase
studies on the mouse brain41, show that more than 50% III studies and has been shown to reduce spasticity 45. A
of all non-olfactory GPCRs are expressed in the cerebral GPR55‑selective compound currently in phase II trials
cortex. Malfunctions in GPCR-mediated neurotransmis- may also potentially be better tolerated than comparable
sion can lead to multiple neurological and psychiatric antispasmodics. Taken together, the multitude of agents
disorders, making GPCRs promising targets42. MS, AD, and targets in late-stage clinical trials indicates that addi-
Huntington disease (HD) and fragile X syndrome (FXS) tional MS therapies acting through GPCRs are likely to
are highlighted below. emerge in the near future.
MS, the most common chronic autoimmune disorder GPCRs are also involved in several neurotransmitter
that affects the CNS, is caused by damage to the insulat- systems that are associated with AD; glutamatergic, sero-
ing myelin covers of axons. Data from clinical studies and tonergic, adrenergic and peptidergic pathways in particu-
animal models have uncovered several GPCRs involved lar are deregulated in this neurodegenerative disorder 46.
in the pathogenesis of MS12, and one GPCR-targeted Targeting these systems might protect against disease

834 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

a Molecule type In trial Approved Discontinued trials progression by modulating the formation of amyloid-­β
I II III I II III plaques (one of the cardinal disease features) or aber-
n = 75 n = 155 n = 104 n = 475 n = 42 n = 127 n = 62 rant signalling following plaque formation46. There is
100
a huge unmet medical need for new therapies for AD,
80 particularly those that might modify disease progres-
Percentage of agents

sion, as the small number of existing therapies (most


60 of which act by increasing levels of acetylcholine by
inhibiting its breakdown by acetylcholinesterase) only
40 have limited effectiveness at improving disease symp-
toms. Leuprolide, a gonadotropin-­releasing hormone
20
receptor agonist approved for treating prostate cancer,
has been tested in a phase III trial for AD, but it failed
0
to meet the primary or secondary end points, although
Small molecule Peptide Protein Antibody Other
there were signs of an effect on disease progression in
b Mode of action In trials Approved Discontinued trials
patients taking an acetylcholinesterase inhibitor 47. An
I II III I II III additional nine GPCR-targeted agents are currently in
n = 72 n = 157 n = 105 n = 481 n = 37 n = 127 n = 66 clinical trials for the treatment of AD (FIG. 4). Serotonin
100 (5-HT) receptor modulators are of particular interest,
including 5‑HT6 receptor antagonists to improve disease
80
Percentage of agents

symptoms (rather than to modify the disease course)


60
by promoting the release of acetylcholine48. However,
Lundbeck recently terminated the development of their
40 5‑HT6 receptor antagonist idalopirdine owing to insuf-
ficient efficacy in phase III trials. Pfizer also terminated
20 a 5‑HT6 receptor antagonist PF‑05212377 due to lack
of efficacy after a phase II trial in 2016. A phase III trial of
0
another 5‑HT6 receptor antagonist, intepirdine, licensed
Antagonist Inverse Agonist Partial PAM NAM by Axovant Sciences from GlaxoSmithKline, failed very
agonist agonist
recently, but intepirdine is still being investigated in
patients with dementia with Lewy bodies.
c Target selectivity or polypharmacology
GPCRs are also potential targets for HD, another
In trials Approved Discontinued trials
neuro­degenerative disorder 49, for which current ther-
I II III I II III
apies can also only improve some symptoms. HD is
n = 75 n = 154 n = 103 n = 475 n = 42 n = 127 n = 62 caused by numerous repetitions of CAG-triplet repeats
Mean = 1.3 Mean = 1.5 Mean = 1.7 Mean = 2.2 Mean = 1.6 Mean = 1.6 Mean= 1.9
100 within the Huntingtin gene (HTT), leading to the
expression of an abnormal pathogenic huntingtin pro-
80 tein and subsequent cell damage; however, the under­
Percentage of agents

lying mechanisms are not fully elucidated. Several GPCR


60
pathways are downregulated in patients with HD, and
40 two GPCR-targeted agents are currently in clinical trials
for HD: the adenosine A1 receptor antagonist pbf‑999
20 (which is in phase I trials) and the dopamine D2 receptor
antagonist pridopidine (which is in phase III trials).
0 Finally, GPCRs have attracted considerable invest-
Number of targets 1 2 3 4 5 ≥6 ment as targets for FXS — the most common inher-
ited form of intellectual disability and autism — which
Figure 3 | Trends in agent molecule types and modes of action for GPCR-targeted is caused by alterations in FMR1, the gene coding for
agents. a | Most G protein-coupled receptor (GPCR)-targeted agents in clinical trials FMRP. Studies in Fmr1‑knockout mice, which have been
are still small molecules, but the earlier phases have increasing proportions of
peptides, monoclonal antibodies, other recombinant proteins and other agent types.
widely used as an animal model for FXS, showed that
Discontinued trials have the same trend, and similar proportions of small molecules the inhibition of the metabotropic glutamate receptor 5
versus biologics, suggesting that their overall attrition rates are also similar. b | Looking (mGlu5) improved synaptic function in these animals and
at their modes of action, GPCR-targeted agents that have been approved or that are in highlighted the importance of mGlu5 in FXS37. However,
Nature Reviews | Drugallosteric
Discovery
clinical trials are predominantly agonists and antagonists, but more positive negative allosteric modulators of mGlu5, such as basim-
modulators (PAMs) and negative allosteric modulators (NAMs) have entered phases I glurant, mavoglurant and STX107, failed in phase II
and II, respectively. There are similar proportions of agonists and antagonists among the trials, as no improvement over placebo could be demon-
agents for which trials have been discontinued, suggesting similar rates of attrition. strated. Agents targeting the GABAB receptor, which
Modes of action had not been reported for 19 agents, which were excluded. c | Target
improved function in Fmr1‑knockout mice, have also
selectivity is increasing (that is, polypharmacology is decreasing) in ongoing clinical
trials of GPCR-targeted agents. The mean number of targets is shown for each group. been clinically investigated for FXS. However, despite
Data from ClinicalTrials.gov were aggregated for agents in trial (completed: 168; some indications of efficacy in a phase II trial with the
recruiting: 91; ongoing: 55; not yet open: 19; and suspended: 3) or discontinued GABAB agonist arbaclofen50, subsequent phase III trials
(discontinued: 205; terminated: 14; and withdrawn: 4). failed due to lack of efficacy compared with placebo.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 835


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

a b Number of indications
In trials Approved Discontinued trials
I II III I II III
30
n = 75 n = 156 n = 103 n = 475 n = 42 n = 127 n = 62
Diabetes Mean = 1.3 Mean = 1.4 Mean= 1.5 Mean = 1.5 Mean = 1.4 Mean= 1.3 Mean = 1.5
100
25 Neoplasms

Percentage of agents
80

60
20
Number of agents in trial

40
Analgesics
Parkinson disease 20

15 0
Schizophrenia
Asthma Number of indications 1 2 3 4 ≥5
Hypertension
Alzheimer
10 disease Depression c Repurposed in current trials
Migraine Allergy 16 15%
Obesity 14% 14%

Percentage of agents
14
5 12
10
8 8%
7% 7%
6 5%
0 4
0 10 20 30 40 50 2
Number of agents approved 0

Figure 4 | Trends in the indications of approved GPCR-targeted drugs Data were manually annotated from the CenterWatch
Nature Reviews database and public
| Drug Discovery
and agents in clinical trials. a | The largest numbers (>40) of approved resources. b | The numbers of indications are similar for approved drugs and
agents (x axis) are seen for analgesics, allergy and hypertension, whereas currently investigated agents. c | There are 23 approved G protein-coupled
among agents in trials (y axis) the highest numbers of listings (>20) are for receptor (GPCR) drugs currently being repurposed for other indications (5%
agents for diabetes and neoplasms. The colour gradient from red to green of the total number of approved drugs), accounting for 8%, 7% and 14% of
highlights the most established and novel indications, calculated as the agents in phases I, II and III, respectively. For parts b and c, the data from
ratio of approved and in‑trial agents, respectively. Alzheimer disease and ClinicalTrials.gov were aggregated for agents in trial (completed: 168;
obesity are the areas with the highest ratio of in‑trial agents to approved recruiting: 91; ongoing: 55; not yet open: 19; and suspended: 3) or
agents, followed by asthma, diabetes, Parkinson disease and neoplasms. discontinued (discontinued: 205; terminated: 14; and withdrawn: 4).

Diabetes is highly represented among the indications new non-peptide agonists of the GLP1 receptor, includ-
for GPCR-targeted agents currently in clinical trials. ing the orally bioavailable TTP273, which is currently in
The growing market share of metabolic disease medica- phase II trials.
tions51 is reflected in the high number of GPCR-targeted The challenges of treating type 2 diabetes and associ-
agents in clinical trials for diabetes and obesity, with ated diseases such as diabetic neuropathy and foot ulcers
27 agents targeting GPCRs for diabetes and seven for have catalysed investment in further GPCR-targeted
obesity (together constituting ~9% of the total number agents32,55. Currently, 11 GPCRs mediate the therapeutic
of agents in clinical trials). There are an estimated 415 effects of approved treatments for these conditions, and
million people worldwide with diabetes mellitus52, 90% agents targeting a further 25 GPCRs are under investiga-
of whom have type 2 diabetes. In contrast to type 1 dia- tion in clinical trials. Among them is MBX‑2982, which
betes, which requires regular insulin injections due to the is a small-molecule GPR119 agonist currently in phase II
failure of the pancreas to produce enough insulin, type 2 trials. MBX‑2982 increases both insulin secretion and
diabetes can be treated with medications that stimulate GLP1 release56,57. Another novel target for which ago-
insulin secretion or that increase insulin sensitivity53. The nists stimulate insulin secretion is the FFA1 receptor34.
first GPCR-targeted drug for type 2 diabetes — the GLP1 Despite the recently discontinued development of the
receptor agonist (or incretin mimetic) exenatide — was small-­molecule agonist fasiglifam (TAK‑875) due to
approved in 2005. As noted above, there are now several hepatotoxicity (mentioned above)29, the FFA1 receptor
other approved peptidic GLP1 receptor agonists, includ- remains a viable target, albeit with a need for further
ing liraglutide, lixisenatide, dulaglutide and albiglutide. characterization of its signalling spectrum, and at least
Modes of action They differ in their durations of action, but all are formu- one FFA1 receptor modulator, LY2881835, is in clinical
Receptor activity defined as lated as injectable drugs. However, semaglutide, a once- trials. Another novel target for the treatment of diabe-
ligand stimulation (agonism), weekly GLP1 analogue that improved glycaemic control in tes is the dopamine D2 receptor. The first dopaminer-
blocking (antagonism),
inhibition (inverse agonism),
a phase II trial54 is now also being tested for oral dosing gic agent, bromocriptine, was recently approved for
or negative or positive in phase III trials. Advances in small-molecule screening, improved glycaemic control and glucose tolerance in
allosteric modulation. including structure-based techniques9, have identified type 2 diabetes58.

836 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Opportunities are emerging for GPCR-targeted agents do not necessarily succeed more often than new agents.
in oncology. Currently, 21 approved drugs with an anti- This suggests that efficacy is typically the limiting factor,
neoplastic indication mediate their effects via 14 distinct rather than safety.
GPCRs. Among these are degarelix, a gonadotropin-­
releasing hormone (GnRH) receptor antagonist that is Emerging trends and opportunities
approved for patients with advanced prostate cancer, and GPCR knowledge in the literature is disproportionately
vismodegib, a smoothened (SMO) receptor inhibitor for focused. More than 50% of the human genome-encoded
the treatment of basal cell carcinoma. The most recent non-olfactory GPCRs (n = 224) remain therapeutically
FDA approval for a GPCR-targeted agent in oncology unexploited (FIG. 2). Drug targets are often first identified
was in 2015 for another SMO receptor inhibitor, sonide- and explored in an academic environment. However,
gib, which was also approved for the treatment of basal due to the long time span of drug development, there is
cell carcinoma. usually a lag time of many years or even decades before
An additional 23 GPCR-targeted agents for treating such discoveries are translated into marketed drugs.
cancer — seven of which have potentially novel targets For this reason, we investigated how research results
— are currently in clinical trials. Chemokine receptors in the form of publication output relate to drug discov-
and proteins in the WNT pathway are among the novel ery efforts, and whether this output could indicate new
GPCR targets being pursued13, often with peptide or trends in GPCR-targeted drug discovery.
mAb therapeutics. For example, CCR2 is the target of
the mAb plozalizumab, which is in phase I trials for GPCR research is an area of immense exploration.
melanoma, and the small-molecule CCR2 inhibitor Gene and protein name searches in PubMed abstracts
CCX872 is in phase I trials for advanced pancreatic can- show that the chemokine receptor type 4 (CXCR4) and
cer. Vantictumab (also known as OMP‑18R5) — a mAb the putative adhesion G protein-coupled receptor E4P
specific for the Frizzled‑7 receptor (FZD7) that has been (ADGRE4P; also known as AGRE4) are mentioned
tested in clinical trials for breast and pancreatic cancer in the abstracts of the most publications (11,123) and
— targets the WNT signalling pathway, which is dys- fewest publications (0; although this may be partly due
regulated in many cancers, leading to cancer stem cell to new nomenclature for adhesion receptors), respec-
activity and tumour growth59. Other biologics in clinical tively (FIG. 5a; note logarithmic scale). GPCRs with
development with targets in the WNT signalling path- approved drugs or agents in clinical trials are men-
way include ipafricept (a fusion protein targeting FZD8), tioned in the abstracts of an average of ~400 publica-
OTSA‑101‑DTPA-90Y (a radiolabelled mAb targeting tions, whereas this figure is only 20 for non-targeted
FZD10) and BNC‑101 (a mAb targeting LGR5). receptors. Receptors with a crystal structure (FIG. 5b)
Furthermore, several other GPCRs have been sug- are mentioned in the abstracts of many publications
gested as potential cancer targets based on mRNA (this probably actually reflects the fact that crystalli-
expression analyses of tumours60. In addition to directly zation efforts in the past decade or so initially focused
targeting GPCR-mediated cancer pathways, over­ on therapeutically important and well-characterized
expressed receptor homodimers and heterodimers that GPCRs). Orphan receptors ( FIG. 5a; shown in light
are overexpressed in certain cancers might function as red) with unknown physiological agonists and func-
selective markers for cancer treatment 61. tions often have very few mentions in publication
Drugs for established GPCR targets in oncology also abstracts. Nonetheless, some orphan receptors, such
continue to be investigated, including GnRH antago- as GPR143 and GPR15, are mentioned in more than
nists such as relugolix for prostate cancer. GnRH 100 article abstracts. The disproportionate data foun-
antagonists such as relugolix and elagolix are also being dation underlines the fact that the research efforts on
investigated for other hormone-related indications such many GPCRs are still in their infancy, and that fur-
as endometriosis. ther characterization is needed to assess their role in
physiology and pathophysiology. This is in accordance
Repurposing of existing GPCR-targeted drugs for new with a general knowledge deficit and lack of funding
indications. The repurposing of existing drugs for for many understudied human proteins, as has been
new indications can reduce the time and cost of bring- recently highlighted by a US National Institutes of
ing a therapeutic to market 62. Of the approved GPCR- Health (NIH) programme called Illuminating the
targeted drugs, 156 (33%) have more than one indication Druggable Genome2.
and the overall average is 1.5 indications (FIG. 4b), demon-
strating that many such drugs are already used for Emerging targets based on disease associations, tool
several indications. Agents in clinical trials have a sim- compounds and patents. Approximately 63% of human
ilar average number of indications to that of currently GPCRs have at least one ligand reported in ChEMBL
approved drugs. (FIG. 5c), including various promising emerging targets
Ongoing clinical trials are evaluating the potential for which agents have not yet been explored in clinical
of repurposing 23 (5%) of the approved GPCR-targeted trials. For example, the neuropeptide S system, which
drugs, which account for ~8%, ~7% and ~14% of the was first described only a few years ago, has been associ-
agent–indication pairs in phase I, II and III trials, respec- ated with several mental illnesses63, and the relaxin fam-
tively (FIG. 4c). Repurposed agents occur as frequently in ily receptors have recently been examined as potential
ongoing and discontinued trials, indicating that they targets for the treatment of addiction, anxiety, obesity

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 837


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

a b c d e

50

100

Number of GPCR targets

200

300

Orphan GPCR Approved


Non-orphan GPCR In trials
400
101 102 103 104 0 5 10 15 100 500 1,000 0 10 20 30 40 0 10 25 50
Citations in PubMed (log scale) Number of available Number of ChEMBL Number of WIPO Number of agents
crystal structures compounds per target patents after 2014 approved or in trials

Figure 5 | GPCR targets from publication to drugs. For each non-olfactory G protein-coupled receptor| (GPCR;
Nature Reviews y axis),
Drug Discovery
the parts show the number of: publications in PubMed in which the GPCR is mentioned in the abstract (part a); crystal
structures in the Protein Data Bank (part b; cut-off at 20 structures); ligands in ChEMBL (part c; cut-off at 1,200
compounds); patents filed through the World Intellectual Property Organization (WIPO) after 2014 (part d; cut-off at
40 patents); and drugs on the market and in clinical trials (part e). GPCRs are sorted by the number of publications.
This comparison reveals a highly disproportionate knowledge landscape for GPCR research and drug discovery,
indicating an unutilized expansion potential.

and anorexia64. With a range of specific antagonists Untapped GPCRs as a source for new drug targets in
at hand, the modulation of formyl peptide receptors a variety of disease areas. The diverse roles of GPCRs
has been suggested to inhibit tumour angiogenesis in are reflected by our analysis of target disease associations
glioblastoma and other cancers65. Bombesin 3 recep- from Open Targets (FIG. 6), which shows that GPCRs are
tor-knockout mice develop metabolic disturbances, associated with nearly every aspect of human patho­
obesity and hypertension66,67. The disruption of gala- physiology. Strikingly, untapped or emerging targets
nin receptor signalling in several preclinical studies — that is, GPCRs that have not yet been targeted or
has indicated that this system is involved in a range of those for which agents are in trials but none have yet
pathologies, including AD, epilepsy, depression and been approved — are found in 17 and 19 of the 20 dis-
cancer 68,69. The aforementioned target families are played categories, respectively. Most emerging targets for
expected to be among the first to enter clinical trials which agents are in trials have already reached phase III,
in the coming years due to their promising preclinical indicating that they may soon transition to established
findings. targets. The largest share of GPCRs with a disease associ-
To further evaluate potentially active pharmaceutical ation that has not yet been investigated at all clinically are
investigations into promising new targets, we analysed found for genetic disorders, followed by eye diseases and
patents filed between 2014 and October 2016 in med- the immune system.The exploitation of the emerging tar-
ical sciences through the World Intellectual Property gets will be facilitated by new avenues in GPCR drug dis-
Organization (WIPO) (FIG. 5d). On the basis of these covery, such as the application of structural data, biased
filings, chemokine receptors, with more than 100 filed signalling, allosteric modulation and de‑­orphanization
patents, are currently the most vigorously pursued target (described below).
family, followed by CaS receptors (n = 11), glycoprotein
hormone receptors (n = 9) and Frizzled receptors (n = 9). Impact of GPCR structures on drug discovery. Structure-
In addition, 35 patents on orphan receptors, including based drug design has long had a valuable role in drug
GPR84, GPR1, GPR17 and LGR5, have also been filed discovery, particularly for drugs with enzyme targets,
in this timeframe. Finally, targets for which there are such as the HIV protease inhibitor indinavir 70, the tyros-
approved drugs or drugs in clinical trials tend to have a ine kinase inhibitor imatinib71 and the influenza neur­
high number of associated publications and compounds a­minidase inhibitor zanamivir 72. However, until recently,
in ChEMBL (FIG. 5e). major challenges in applying X‑ray crystallography to

838 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Genetic disorder 70 for crystallization based on their high therapeutic rele-


Eye 45 vance, such as many and early validated disease asso-
Immune system 46 ciations. However, these targets also have a significant
Skeletal system 31
proportion of agents in phase I and phase II trials, in
Neoplasm 48
which the structural templates could have been available
Disease or organ association

Respiratory system 38

Number of GPCR targets


Haematological system 14 long enough to contribute to the lead discovery and/or
Digestive system 48 optimization processes.
Nervous system 70 The real impact of structural data may be under­
Reproductive system 36 estimated due to unpublished proprietary structures. For
Cardiovascular 40
Endocrine system
example, Receptos announced in January 2011 that they
47
Skin 26
were targeting the S1P1 receptor with a proprietary struc-
Kidney 27 ture, which was not published until the following year.
Liver 27 This S1P1 agonist, ozanimod (which is now being devel-
Other 69 oped by Celgene following their acquisition of Receptos),
Metabolic 51 is expected to be submitted for FDA approval in 2017.
Bladder 14
Head
Furthermore, Heptares Therapeutics’ pipeline (see Further
25
Infectious 22 information) currently lists ten specified, and an even
larger number of undisclosed, targets for which agents are
0 20 40 60 80 100
in preclinical and clinical trials. Known structure-based
Percentage of targets
candidates that target GPCRs include agents that target
Non-targeted Phase I Phase II Phase III Approved the μ-opioid receptor for pain77, M1 and M4 muscarinic
receptors for AD, the mGlu5 receptor for psychiatric dis-
Figure 6 | Disease associations for all 398 non-olfactory GPCRs. Disease associations orders, orexin receptor 2 (OX2; also known as HCRTR2)
for receptors not yet targeted (grey), with an agent in a clinical
Nature trial (green)
Reviews | Drug and with an
Discovery for narcolepsy, proteinase-activated receptor 2 (PAR2) for
approved (red) drug. The 224 non-targeted G protein-coupled receptors (GPCRs) are
inflammatory disorders (see Heptares pipeline in Further
associated with a wide range (17/20) of disease types, systems or organs, demonstrating
broad untapped therapeutic potential. The numbers of GPCR targets associated with
information) and the adenosine A2A receptor for cancer79.
each disease type, system or organ are listed on the right-hand side. It is important to
note that the association of a GPCR target with an approved drug with a disease type, Biased signalling as a novel mechanism for functional
system or organ does not mean that a drug has been approved for that disease type, selectivity. Activating the appropriate cellular response
system or organ (for example, many genetic disorders for which an association exists through one of the four major G protein families and
may not actually have an approved GPCR drug). Disease associations have been their intracellular effectors (such as adenylyl cyclase and
agglomerated from the Open Targets platform by combining association scores above phospholipase C) or through β‑arrestin-dependent acti-
a value of 0.5 (at least one-half of the highest association confidence). The association vation of kinases and others, is crucial for a favourable
score summarizes the strength of evidence from each data source (genome-wide physiological response80–82. Recent discoveries of mole-
association studies, genetic variants, expression data and animal models) (see Further
cules that preferentially trigger one of these pathways —
information). Three ontology terms, ‘measurement’, ‘phenotype’ and ‘biological process’,
were excluded as they cannot be interpreted as a distinct category. It should be noted
referred to as biased agonists — offer a new mechanism
that receptors can be associated with multiple systems. for reducing side effects8,20,83,84. This has led to the idea of
distinct receptor conformational states that are stabilized
by different ligands, and can lead to activation of specific
GPCRs limited the potential for structure-based drug signalling and regulatory proteins85.
design for such targets. However, thanks to recent break- Several promising agents with bias through β‑­ar-
throughs in GPCR crystallography 73,74, 44 distinct GPCR restin, a G protein or allosteric modulation are being
structures and 205 ligand–receptor complexes are now investigated in preclinical studies, as well as clinical tri-
available across all human GPCR classes A–C and F21 als8. Most notable is the μ-opioid receptor ligand oliceri-
(FIG. 7). This has paved the way for novel lead discovery dine (TRV130), currently in phase III trials, which was
through virtual screening and better off-target ration- granted ‘Breakthrough Therapy’ designation by the FDA
alization75,76. For example, recent docking experiments owing to its improved analgesic profile86. Oliceridine,
against the μ-opioid receptor structure identified PZM21, and its current phase I follow‑on agent TRV734, do
a Gi protein-biased agonist with potency and efficacy not engage the β‑arrestin pathway, which is associated
similar to morphine, but with reduced adverse effects with opioid-induced respiratory depression and con-
in mice77. In another exciting example, two chemokine stipation87. Conversely, no G protein engagement was
receptor structures — for CCR9 and CCR2 — revealed a observed for the β‑arrestin-biased ligand TRV027, which
previously unknown intracellular binding pocket, which targets the angiotensin II type 1 receptor and which is a
might provide a new strategy for drug design78. drug candidate for the treatment of acute heart failure88.
We investigated whether the availability of crystal However, in May 2016, Trevena announced that TRV027
structures has had a measurable impact on the number had failed to meet the primary or secondary end points
of agents in clinical trials. FIGURE 7a shows that most of in a phase IIb trial.
the receptors with many approved drugs (red bars) have Therapeutic implications for biased agonists have only
an early crystal structure published in 2007–2012, with begun to be elucidated for a number of GPCR systems,
the exception of the muscarinic M1 receptor (ACM1) including adrenergic, angiotensin, opioid, dopamine,
from 2016. It is likely that these receptors were selected serotonin and chemokine receptors7. More knowledge

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 839


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

a b

5HT1B
5HT2B
Phase I Phase II Phase III Approved

ACM 2
Crystallised

SMO
AC 4
AC 3
Year receptor

M
AC

GR RM5
M

1
M
2000 OPSD

AD

M
AD

G
RB
2007 ADRB2 RB

2
1 R
2008 ADRB1 DR GL P1R
AA2AR D3 GL

ein
Am
2010 FR1

id
CXCR4 HR CR

Prot
ine

ac
H1
DRD3

rg

no
CR

ic

i
2011 CAL

Am
HRH1 AGTR t ide
2012 ACM2
AGTR2
1 C
F Pep
S1PR1 B1
ACM3 Class Other US28
OPRM APJ Sensory
OPRK A Nu OPSD
OPRX EDNRB tid
e cle
oti
OPRD Pep de
P2Y
D 12
NTR1 OPR P2R
RK Y1

Lip
PAR1 OP X

Protein
R

id
5HT2B OP M AA
R AA 2AR
2013 5HT1B OP 1R
R
SMO X1
O

CN
X2
CRFR1

R1
O

PA 1

S1
GLR

R
R2
PA

PR
LPA
1
CCR5

1
FFAR
CCR2
NTR

CXCR4
CCR5
CCR9

R1
2014 GRM1

1
P2Y12
GRM5
FFAR1
c Receptor ligand types
2015 OX2R
45
US28 Other Nucleotide Peptide
P2RY1
Number of GPCRs with a crystal structure

40 Amino acid Lipid Aminergic


AGTR1
LPAR1 Sensory Protein
35
2016 ACM1
OX1R 30
ACM4
EDNRB 25
CNR1
CCR9 20
CCR2
2017 AA1R 15
AGTR2
10
CALCR
PAR2
5
GLP1R
APJ
0
0 20 40 60 80 2000 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017
Number of agents in clinical phases Year

Figure 7 | Crystallized GPCRs. a | Number of agents in each clinical collected from http://gpcrdb.org/structure/statistics. GPCRs are listed
phase for each crystallized G protein-coupled receptor (GPCR) sorted by Nature Reviews
using the protein name in UniProt; for definitions Drug details
and |other Discovery
on
year. b | Classification tree of all crystallized GPCRs. c | Proportion of each receptor nomenclature, see the IUPHAR/BPS Guide to PHARMACOLOGY
receptor ligand type among all crystallized GPCRs. Parts b and c were (see Further information).

of the signalling repertoire of GPCRs will be required to to be expected that many of the GPCR-targeted agents
fully exploit the potential of biased signalling and currently in clinical trials will not ultimately gain reg-
to fine-tune intracellular responses to therapy 89. New ulatory approval, the demonstrated druggability of the
ways to validate biased signalling in animal models will GPCR protein family and the important role of GPCRs
help the development of the tools needed to transition in diseases such as diabetes, obesity, AD and psychiatric
biased ligands towards preclinical development as drug disorders, provide a strong driving force for continued
candidates and beyond90. drug discovery and development efforts in this field.
As the physiology and pathophysiology of GPCRs
Outlook for targeting GPCRs becomes better characterized, certain groups of recep-
GPCR drug discovery has gained new momentum, as tors may prove intractable, whereas others may expand
demonstrated by the large number of new drug tar- the druggable ‘GPCRome’. There is emerging evidence of
gets and the scientific impetus in GPCR structural excreted gut microbiota metabolites that serve as ligands
biology, pharmacology and modelling. Although it is for GPCRs and thereby influence hormone release91,92.

840 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

Additionally, many GPCRs have been identified as targeted by drugs with a specific mechanism of action,
nutrient sensors, which could serve as potential targets such as allosteric modulation, inhibitors interfering with
to treat metabolic dysfunction and inflammatory dis- natural protein–protein interactions and antibodies,
eases93. Furthermore, several orphan receptors may have which are currently used to target, for example, adhesion
evolved to recognize pathogens and to invoke appropri- receptors for antineoplastic treatment 96.
ate immune responses94. The characterization of the Moving forwards in GPCR drug discovery depends
remaining orphan receptors could reveal new targets on solving several crucial issues. Suitable tool com-
for a multitude of indications. Strikingly, the number pounds are required to establish target biological func-
of orphan receptors could increase considerably, as tions and disease relevance97. This could be aided by
many of the ~400 olfactory GPCRs are now known to novel high-throughput ligand identification methods
be widely expressed throughout the body and to have able to probe a larger chemical space, such as DNA-
important functions beyond the detection of odor- encoded libraries98. Furthermore, improved disease
ants95. Olfactory receptors have also been found to be models and genetically engineered systems are needed
overexpressed in tumour cells95. for unambiguous target validation, which could be aided
However, some orphan receptors do not follow by gene-editing technologies such as CRISPR99. The
the classical paradigm of endogenous activation by an huge and heterogeneous published and patented chem-
external ligand. GPR50 heterodimerizes constitutively ical and biological data need to be made available and
and specifically with melatonin receptors MT1 and MT2. structured in long-term committing databases such as
The class C orphan receptors GPR156 and GPRC5A–D ChEMBL, Guide to PHARMACOLOGY and GPCRdb.
lack the amino terminus, which contains the ortho- These endeavours are too large for any single entity to pur-
steric binding site for the liganded members of this sue, and will need to engage the wider interdisciplinary
class. Furthermore, the non-orphan protease-activated basic and applied GPCR field, calling for continued invest-
and adhesion receptors are activated by their own N ter- ment in public–private pre-competitive partnerships and
minus upon truncation. These receptors could still be consortia to accelerate progress.

1. Rask-Andersen, M., Masuram, S. & Schiöth, H. B. The 12. Du, C. & Xie, X. G protein-coupled receptors as 28. Miller, P. D. et al. Effect of abaloparatide versus placebo
druggable genome: evaluation of drug targets in therapeutic targets for multiple sclerosis. Cell Res. 22, on new vertebral fractures in postmenopausal women
clinical trials suggests major shifts in molecular class 1108–1128 (2012). with osteoporosis. JAMA 316, 722–733 (2016).
and indication. Annu. Rev. Pharmacol. Toxicol. 54, 13. Bar-Shavit, R. et al. G protein-coupled receptors in 29. Kaku, K., Enya, K., Nakaya, R., Ohira, T. &
9–26 (2014). cancer. Int. J. Mol. Sci. 17, 1320 (2016). Matsuno, R. Efficacy and safety of fasiglifam
2. The IDG Knowledge Management Center. Unexplored 14. Lappano, R. & Maggiolini, M. G protein-coupled (TAK‑875), a G protein-coupled receptor 40 agonist,
opportunities in the druggable human genome. Nat. receptors: novel targets for drug discovery in cancer. in Japanese patients with type 2 diabetes
Rev. Drug Disc. http://www.nature.com/nrd/posters/ Nat. Rev. Drug Discov. 10, 47–60 (2011). inadequately controlled by diet and exercise:
druggablegenome/nrd_druggablegenome.pdf (2016). 15. Cromie, K. D., Van Heeke, G. & Boutton, C. a randomized, double-blind, placebo-controlled,
Peer-reviewed poster outlining a major NIH Nanobodies and their use in GPCR drug discovery. phase III trial. Diabetes Obes. Metab. 17, 675–681
programme to characterize the ‘dark space’ of Curr. Top. Med. Chem. 15, 2543–2557 (2015). (2015).
major drug target families. 16. Zhang, R. & Xie, X. Tools for GPCR drug discovery. 30. Mullard, A. 2016 FDA drug approvals. Nat. Rev. Drug
3. Kolakowski, L. F. Jr. GPCRDb: a G‑protein-coupled Acta Pharmacol. Sin. 33, 372–384 (2012). Discov. 16, 73–76 (2017).
receptor database. Recept. Channels 2, 1–7 (1994). 17. Jacobson, K. A. New paradigms in GPCR drug 31. Hutchings, C. J., Koglin, M., Olson, W. C. &
4. Schiöth, H. B. & Fredriksson, R. The GRAFS discovery. Biochem. Pharmacol. 98, 541–555 (2015). Marshall, F. H. Opportunities for therapeutic
classification system of G‑protein coupled receptors in 18. Wootten, D., Miller, L. J., Koole, C., Christopoulos, A. antibodies directed at G‑protein-coupled receptors.
comparative perspective. Gen. Comp. Endocrinol. & Sexton, P. M. Allostery and biased agonism at class Nat. Rev. Drug Discov. 16, 1–24 (2017).
142, 94–101 (2005). B G protein-coupled receptors. Chem. Rev. 117, A review on therapeutic antibodies targeting
5. Southan, C. et al. The IUPHAR/BPS Guide to 111–138 (2017). GPCRs and the current pipeline of agents in clinical
PHARMACOLOGY in 2016: Towards curated 19. Wootten, D., Christopoulos, A. & Sexton, P. M. trials.
quantitative interactions between 1300 protein Emerging paradigms in GPCR allostery: implications 32. Oh, D. Y. & Olefsky, J. M. G protein-coupled receptors
targets and 6000 ligands. Nucleic Acids Res. 44, for drug discovery. Nat. Rev. Drug Discov. 12, as targets for anti-diabetic therapeutics. Nat. Rev.
D1054–D1068 (2016). 630–644 (2013). Drug Discov. 15, 161–172 (2016).
6. Christopoulos, A. et al. International Union of Basic A key review article on the therapeutic potential of 33. Khanna, A. et al. Angiotensin II for the treatment of
and Clinical Pharmacology. XC. Multisite allosteric ligands. vasodilatory Shock. N. Engl. J. Med. 377, 419–430
pharmacology: recommendations for the 20. Pupo, A. S. et al. Recent updates on GPCR biased (2017).
nomenclature of receptor allosterism and allosteric agonism. Pharmacol. Res. 112, 49–57 (2016). 34. Mancini, A. D. & Poitout, V. GPR40 agonists for the
ligands. Pharmacol. Rev. 66, 918–947 (2014). 21. Isberg, V. et al. GPCRdb: an information system for G treatment of type 2 diabetes: life after ‘TAKing’ a hit.
7. Rajagopal, S., Rajagopal, K. & Lefkowitz, R. J. protein-coupled receptors. Nucleic Acids Res. 44, Diabetes. Obes. Metab. 17, 622–629 (2015).
Teaching old receptors new tricks: biasing seven- D356–D364 (2016). 35. Gao, Z. G. & Jacobson, K. A. Allosteric modulation
transmembrane receptors. Nat. Rev. Drug Discov. 9, A specialized GPCR database and analysis tool and functional selectivity of G protein-coupled
373–386 (2010). spanning structures, mutants, crystallization receptors. Drug Discov. Today Technol. 10,
8. Violin, J. D., Crombie, A. L., Soergel, D. G. & construct design, and drugs and indications. e237–e243 (2013).
Lark, M. W. Biased ligands at G‑protein-coupled 22. Law, V. et al. DrugBank 4.0: shedding new light on 36. Shen, Q. et al. ASD v3.0: unraveling allosteric
receptors: promise and progress. Trends Pharmacol. drug metabolism. Nucleic Acids Res. 42, regulation with structural mechanisms and biological
Sci. 35, 308–316 (2014). D1091–D1097 (2014). networks. Nucleic Acids Res. 44, D527–D535
A review discussing how biased ligands may deliver 23. Ahlgren, N. A., Ren, J., Lu, Y. Y., Fuhrman, J. A. & (2016).
safer, better tolerated and more efficacious drugs, Sun, F. Alignment-free d2* oligonucleotide frequency 37. Nickols, H. H. & Conn, J. P. Development of allosteric
which highlights several biased ligands that are in dissimilarity measure improves prediction of hosts modulators of GPCRs for treatment of CNS disorders.
clinical development. from metagenomically-derived viral sequences. Neurobiol. Dis. 61, 55–71 (2014).
9. de Graaf, C. et al. Glucagon-like peptide‑1 and its class Nucleic Acids Res. 45, 39–53 (2017). 38. Anighoro, A., Bajorath, J. & Rastelli, G.
B G protein-coupled receptors: a long march to 24. Koscielny, G. et al. Open Targets: a platform for Polypharmacology: challenges and opportunities in
therapeutic successes. Pharmacol. Rev. 68, therapeutic target identification and validation. drug discovery. J. Med. Chem. 57, 7874–7887
954–1013 (2016). Nucleic Acids Res. 45, D985–D994 (2016). (2014).
10. Solari, R., Pease, J. E. & Begg, M. Chemokine 25. Overington, J. P., Al‑Lazikani, B. & Hopkins, A. L. How 39. Butini, S. et al. Polypharmacology of dopamine
receptors as therapeutic targets: why aren’t there many drug targets are there? Nat. Rev. Drug Discov. receptor ligands. Prog. Neurobiol. 142, 68–103
more drugs? Eur. J. Pharmacol. 746, 363–367 5, 993–996 (2006). (2016).
(2015). 26. Rask-Andersen, M., Almén, M. S. & Schiöth, H. B. 40. Uhlen, M. et al. Tissue-based map of the human
11. Nicoletti, F., Bruno, V., Ngomba, R. T., Gradini, R. & Trends in the exploitation of novel drug targets. proteome. Science 347, 1260419 (2015).
Battaglia, G. Metabotropic glutamate receptors as Nat. Rev. Drug Discov. 10, 579–590 (2011). 41. Regard, J. B., Sato, I. T. & Coughlin, S. R. Anatomical
drug targets: what’s new? Curr. Opin. Pharmacol. 20, 27. Santos, R. et al. A comprehensive map of molecular profiling of G protein-coupled receptor expression.
89–94 (2015). drug targets. Nat. Rev. Drug Disc. 16, 19–34 (2017). Cell 135, 561–571 (2008).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 16 | DECEMBER 2017 | 841


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A N A LY S I S

42. Komatsu, H. Novel therapeutic GPCRs for psychiatric 65. Prevete, N., Liotti, F., Marone, G., Melillo, R. M. & 89. Khoury, E., Clément, S. & Laporte, S. A. Allosteric
disorders. Int. J. Mol. Sci. 16, 14109–14121 (2015). De Paulis, A. Formyl peptide receptors at the interface and biased G protein-coupled receptor signaling
43. Brinkmann, V. et al. Fingolimod (FTY720): discovery of inflammation, angiogenesis and tumor growth. regulation: potentials for new therapeutics.
and development of an oral drug to treat multiple Pharmacol. Res. 102, 184–191 (2015). Front. Endocrinol. 5, 68 (2014).
sclerosis. Nat. Rev. Drug Discov. 9, 883–897 (2010). 66. Ramos-Álvarez, I. et al. Insights into bombesin 90. Beaulieu, J. M. In vivo veritas, the next frontier for
44. Radick, L. & Mehr, S. R. The latest innovations in the receptors and ligands: highlighting recent advances. functionally selective GPCR ligands. Methods 92,
drug pipeline for multiple sclerosis. Am. Health Drug Peptides 72, 128–144 (2015). 64–71 (2016).
Benefits 8, 448–453 (2015). 67. Yamada, K., Wada, E. & Wada, K. Bombesin-like 91. Psichas, A., Reimann, F. & Gribble, F. M. Gut
45. Zettl, U. K., Rommer, P., Hipp, P. & Patejdl, R. peptides studies on food intake and social behaviour chemosensing mechanisms. J. Clin. Invest. 125,
Evidence for the efficacy and effectiveness of THC-CBD with receptor knock-out mice. Ann. Med. 32, 908–917 (2015).
oromucosal spray in symptom management of 519–529 (2000). 92. Cohen, L. J., Esterhazy, D., Kim, S. H., Lemetre, C.,
patients with spasticity due to multiple sclerosis. 68. Lang, R. et al. Physiology, signaling, and Aguilar, R. R. et al. Commensal bacteria make GPCR
Ther. Adv. Neurol. Disord. 9, 9–30 (2016). pharmacology of galanin peptides and receptors: ligands that mimic human signalling molecules.
46. Thathiah, A. & De Strooper, B. The role of G protein- three decades of emerging diversity. Pharmacol. Rev. Nature 549 48–53 (2017).
coupled receptors in the pathology of Alzheimer’s 67, 118–175 (2015). 93. Richards, J. L., Yap, Y. A., McLeod, K. H., Mackay, C. R.
disease. Nat. Rev. Neurosci. 12, 73–87 (2011). 69. Freimann, K., Kurrikoff, K. & Langel, Ü. Galanin & Marino, E. Dietary metabolites and the gut
47. Bowen, R. L., Perry, G., Xiong, C., Smith, M. A. & receptors as a potential target for neurological disease. microbiota: an alternative approach to control
Atwood, C. S. A clinical study of lupron depot in the Expert Opin. Ther. Targets 19, 1665–1676 (2015). inflammatory and autoimmune diseases. Clin. Transl
treatment of women with Alzheimer’s disease: 70. Wlodawer, A. & Vondrasek, J. Inhibitors of HIV‑1 Immunol. 5, e82 (2016).
preservation of cognitive function in patients taking an protease: a major success of structure-assisted drug 94. Bufe, B. et al. Recognition of bacterial signal peptides
acetylcholinesterase inhibitor and treated with high design. Annu. Rev. Biophys. Biomol. Struct. 27, by mammalian formyl peptide receptors. J. Biol.
dose lupron over 48 weeks. J. Alzheimers Dis. 44, 249–284 (1998). Chem. 290, 7369–7387 (2015).
549–560 (2015). 71. Capdeville, R., Buchdunger, E., Zimmermann, J. & 95. Foster, S. R., Roura, E. & Thomas, W. G. Extrasensory
48. Ferrero, H., Solas, M., Francis, P. T., & Ramirez, M. J. Matter, A. Glivec (STI571, imatinib), a rationally perception: odorant and taste receptors beyond the
Serotonin 5‑HT6 receptor antagonists in Alzheimer’s developed, targeted anticancer drug. Nat. Rev. Drug nose and mouth. Pharmacol. Ther. 142, 41–61 (2014).
disease: therapeutic rationale and current Discov. 1, 493–502 (2002). A review summarizing the evidence for expression
development status. CNS Drugs 31, 19–32 (2017). 72. Varghese, J. N. Development of neuraminidase and function of odorant and taste receptors in
49. Dowie, M. J., Scotter, E. L., Molinari, E. & Glass, M. inhibitors as anti-influenza virus drugs. Drug Dev. Res. tissues beyond the nose and mouth and
The therapeutic potential of G‑protein coupled 46, 176–196 (1999). highlighting their broad potential in physiology and
receptors in Huntington’s disease. Pharmacol. Ther. 73. Piscitelli, C. L., Kean, J., de Graaf, C. & Deupi, X. A. pathophysiology.
128, 305–323 (2010). Molecular pharmacologist’s guide to G protein- 96. Hamann, J. et al. International Union of Basic and
50. Veenstra-VanderWeele, J. et al. Arbaclofen in children coupled receptor crystallography. Mol. Pharmacol. Clinical Pharmacology. XCIV. Adhesion G Protein-
and adolescents with autism spectrum disorder: 88, 536–551 (2015). Coupled Receptors. Pharmacol. Rev. 67, 338–367
a randomized, controlled, phase 2 trial. 74. Jazayeri, A., Dias, J. M. & Marshall, F. H. From G (2015).
Neuropsychopharmacology 42, 1390–1398 protein-coupled receptor structure resolution to 97. Wacker, D. et al. How ligands illuminate GPCR
(2017). rational drug design. J. Biol. Chem. 290, molecular pharmacology. Cell 170, 414–427 (2017).
51. Hampp, C., Borders-Hemphill, V., Moeny, D. G. & 19489–19495 (2015). A review highlighting the many understudied
Wysowski, D. K. Use of antidiabetic drugs in the U. 75. Cooke, R. M., Brown, A. J. H., Marshall, F. H. & GPCRs and new methods for the identification of
S., 2003–2012. Diabetes Care 37, 1367–1374 Mason, J. S. Structures of G protein-coupled receptors tool compounds to elucidate their pharmacology.
(2014). reveal new opportunities for drug discovery. 98. Ahn, S. et al. Allosteric ‘beta-blocker’ isolated from a
52. International Diabetes Federation. IDF Diabetes Atlas Drug Discov. Today 20, 1355–1364 (2015). DNA-encoded small molecule library. Proc. Natl Acad.
Sixth Ed. IDF https://www.idf.org/e-library/ 76. Tautermann, C. S. & Gloriam, D. E. Editorial overview: Sci. USA 114, 1708–1713 (2017).
epidemiology-research/diabetes-atlas/19-atlas-6th- new technologies: GPCR drug design and function — 99. Fellmann, C., Gowen, B. G., Lin, P.‑C., Doudna, J. A. &
edition.html (2013). exploiting the current (of) structures. Curr. Opin. Corn, J. E. Cornerstones of CRISPR-Cas in drug
53. Olokoba, A. B., Obateru, O. A. & Olokoba, L. B. Pharmacol. 30, vii–x (2016). discovery and therapy. Nat. Rev. Drug Discov. 16,
Type 2 diabetes mellitus: a review of current trends. Special issue with leading academic and industrial 89–100 (2017).
Oman Med. J. 27, 269–273 (2012). groups describing developments in technologies for
54. Nauck, M. A. et al. A phase 2, randomized, dose- structure-based drug design. Acknowledgements
finding study of the novel once-weekly human GLP‑1 77. Manglik, A. et al. Structure-based discovery of opioid The authors thank A. Hersey, S. M. Soisson, V. Chelliah, L.
analog, semaglutide, compared with placebo and analgesics with reduced side effects. Nature 537, Jahn, K. Harpsøe and M. Shehata for their valuable com-
open-label liraglutide in patients with type 2 diabetes. 185–190 (2016). ments on this work. D.E.G. has received financial support
Diabetes Care 39, 231–241 (2016). Computational drug design study to develop a from the European Research Council (DE‑ORPHAN 639125)
55. Kolar, G. R., Grote, S. M. & Yosten, G. L. C. Targeting drug that mimics the pain-relieving activity of and the Lundbeck Foundation (R163‑2013‑16327).
orphan G protein-coupled receptors for the treatment opioid compounds but that has fewer side effects.
of diabetes and its complications: C‑peptide and 78. Sakmar, T. P. & Huber, T. Pharmacology: inside-out Competing interests statement
GPR146. J. Intern. Med. 281, 25–40 (2017). receptor inhibition. Nature 540, 344–345 (2016). The authors declare no competing interests.
56. Lauffer, L., Iakoubov, R. & Brubaker, P. L. GPR119: 79. Jazayeri, A., Andrews, S. P. & Marshall, F. H.
‘double-dipping’ for better glycemic control. Structurally enabled discovery of adenosine A2A Publisher’s note
Endocrinology 149, 2035–2037 (2008). receptor antagonists. Chem. Rev. 117, 21–37 Springer Nature remains neutral with regard to jurisdictional
57. Ritter, K., Buning, C., Halland, N., Pöverlein, C. & (2017). claims in published maps and institutional affiliations.
Schwink, L. G. Protein-coupled receptor 119 (GPR119) 80. Marinissen, M. J. & Gutkind, J. S. G‑Protein-coupled
agonists for the treatment of diabetes: recent receptors and signaling networks: emerging
progress and prevailing challenges. J. Med. Chem. 59, paradigms. Trends Pharmacol. Sci. 22, 368–376 FURTHER INFORMATION
3579–3592 (2016). (2001). CenterWatch: http://www.centerwatch.com
58. Lopez Vicchi, F. et al. Dopaminergic drugs in type 2 81. Neves, S. R., Ram, P. T. & Iyengar, R. G protein ChEMBL: https://www.ebi.ac.uk/chembl
diabetes and glucose homeostasis. Pharmacol. Res. pathways. Science 296, 1636–1639 (2002). ClinicalTrials.gov: https://clinicaltrials.gov
109, 74–80 (2016). 82. Ludwig, A., Belfiore, N. M., Pitra, C., Svirsky, V. & Drugbank: https://www.drugbank.ca
59. Lynch, J. & Wang, J. G. Protein-coupled receptor Jenneckens, I. Genome duplication events and Drugs@FDA: https://www.accessdata.fda.gov/scripts/
signaling in stem cells and cancer. Int. J. Mol. Sci. 17, functional reduction of ploidy levels in sturgeon cder/daf
707 (2016). (Acipenser, Huso and Scaphirhynchus). Genetics 158, FDA information on attrition rates in clinical trials: https://
60. Li, S., Huang, S. & Peng, S. B. Overexpression of G 1203–1215 (2001). www.fda.gov/forpatients/approvals/drugs/ucm405622.htm
protein-coupled receptors in cancer cells: involvement 83. Masuho, I. et al. Distinct profiles of functional GPCRdb: http://www.gpcrdb.org
in tumor progression. Int. J. Oncol. 27, 1329–1339 discrimination among G proteins determine the GPCRdb statistics: http://gpcrdb.org/drugs/drugstatistics
(2005). actions of G protein-coupled receptors. Sci. Signal. 8, GPCRdb drug data: http://gpcrdb.org/drugs/drugbrowser
61. Moreno, E. et al. Targeting CB2‑GPR55 receptor ra123 (2015). GPCRdb target mapping: http://gpcrdb.org/drugs/
heteromers modulates cancer cell signaling. 84. Costa-Neto, C. M., Parreiras-e-Silva, L. T. & drugmapping
J. Biol. Chem. 289, 21960–21972 (2014). Bouvier, M. A pluridimensional view of biased Guide To PHARMACOLOGY:
62. Nosengo, N. Can you teach old drugs new tricks? agonism. Mol. Pharmacol. 90 587–595 (2016). http://www.guidetopharmacology.org
Nature 534, 314–316 (2016). 85. Nygaard, R. et al. The dynamic process of Heptares Therapeutics: https://www.heptares.com
A news feature on the repurposing of old drugs to ß2‑adrenergic receptor activation. Cell 152, 532–542 Open Targets: https://www.targetvalidation.org
overcome skyrocketing costs in drug development. (2013). Protein Data Bank: https://www.wwpdb.org/
63. Reinscheid, R. K. in Handbook of Biologically Active 86. Viscusi, E. R. et al. A randomized, phase 2 study Pharos: https://pharos.nih.gov/idg/index
Peptides (ed. Kastin, A.) 869–874 (Academic Press, investigating TRV130, a biased ligand of the μ‑opioid PubMed: https://www.ncbi.nlm.nih.gov/pubmed
2013). receptor, for the intravenous treatment of acute pain. Receptos ozanimod: http://www.msdiscovery.org/research-
64. Halls, M. L., Bathgate, R. A. D., Sutton, S. W., Pain 157, 264–272 (2016). resources/drug-pipeline/337-ozanimod
Dschietzig, T. B. & Summers, R. J. International Union 87. Bohn, L. M. Enhanced morphine analgesia in mice Reuters article on idalopirdine: http://www.reuters.com/
of Basic and Clinical Pharmacology. XCV. Recent lacking β-arrestin 2. Science 286, 2495–2498 article/us-health-alzheimers-lundbeck-idUSKBN15N16R
advances in the understanding of the pharmacology (1999). UniProt: http://uniprot.org
and biological roles of relaxin family peptide receptors 88. Ikeda, Y., Kumagai, H., Motozawa, Y., Suzuki, J.‑I. & WIPO: http://worldwide.espacenet.com
1–4, the receptors for relaxin family peptides. Komuro, I. Biased agonism of the angiotensin II type I ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Pharmacol. Rev. 67, 389–440 (2015). receptor. Int. Heart J. 56, 485–488 (2015).

842 | DECEMBER 2017 | VOLUME 16 www.nature.com/nrd


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like