You are on page 1of 13

REVIEWS

Current pharmacotherapy for obesity


Gitanjali Srivastava and Caroline M. Apovian
Abstract | More than one-third of adults in the USA have obesity, which causes, exacerbates or
adversely impacts numerous medical comorbidities, including diabetes mellitus and
cardiovascular disease. Despite intensive lifestyle modifications, the disease severity warrants
further aggressive intervention, including pharmacotherapy, medical devices and bariatric
surgery. Noninvasive anti-obesity drugs have thus now resurfaced as targeted adjunctive
therapeutic approaches to intensive lifestyle intervention, bridging the gap between lifestyle and
bariatric surgery. In this Review, we discuss FDA-approved anti-obesity drugs in terms of safety
and efficacy. As most of these drugs have a mean percentage weight loss reported in clinical trials
but individual variations in response rates, a future direction of obesity pharmacotherapy
research might include the potential for personalized medicine to target early responders to
these anti-obesity drugs.

Obesity is a chronic, complex, heterogeneous disease We also now know that the epidemic stems from
with a disturbing economic burden, and it can cause changes within the modern microenvironment and
more than 200 other medical disorders that affect entire macro­environment, with epi­genetic, genetic, imprint­
organ systems1. Since 1980, rates of obesity have doubled ing and early-life events affecting the individual pre­
in 73 countries, with a high BMI (>25 kg/m2) account­ disposition to weight gain and accrual of fat mass.
ing for ~4 million deaths worldwide and a high cardio­ Environmental and lifestyle factors such as circadian
vascular disease burden2. In the USA, over one-third of rhythm, pala­tability and availability of foods, taste pref­
adults (approximately 35% of men and 40% of women) erences, social habits and resources also signal in a feed­
have obesity; a disproportionate rise in the severity of forward manner to the ‘emotional or cognitive brain’
obesity has also occurred3. The rapid increase in obesity and central pathways of the ‘metabolic brain’ (REF. 6).
prevalence coupled with its devastating health effects These central nervous system (CNS) pathways within
and associated comorbidities highlights the immediate the arcuate nucleus of the hypothalamus either potenti­
need for early recognition and treatment of this prob­ ate or enhance two major opposing pathways that affect
lem in the context of the existing available therapeutic food intake and energy expenditure (FIG. 1): activation
armature. of the orexigenic peptides neuropeptide Y (NPY) and
This Review is thus timely and focuses on current agouti-related peptide (AgRP) increases food intake
anti-obesity drugs, as understanding their pharmaco­ and decreases energy expenditure, whereas activation
logical safety and efficacy is crucial to the clinical care of the anorexigenic peptides pro-­opiomelanocortin
of patients with obesity who might not have responded (POMC) and cocaine- and amphetamine-regulated
to lifestyle interventions. We also highlight important transcript (CART) decreases food intake and increases
clinical insights with regards to the prescription of these energy expenditure7–9. These pathways further receive
drugs. As obesity is influenced by a myriad of genetic, humoral, metabolic and neuronal7 afferent and efferent
Section of Endocrinology,
developmental, biological, environmental, behavioural signals from the gastrointestinal, muscular and adipose
Diabetes and Nutrition, and iatrogenic factors (resulting in the diversification tissue systems, becoming critical integral components
Department of Medicine, of obesity phenotypes with individual variations in of energy homeostasis10–16.
Boston University School of weight-loss response rates) discerning the pattern of Currently, there are six major FDA-approved anti-­
Medicine, 720 Harrison
response to specific anti-obesity drugs will probably obesity medications: phentermine, orlistat, phentermine/
Avenue, 8 th Floor, Suite 801,
Boston, increase their efficacy, which would be an early step topiramate extended release (ER), lorcaserin, naltrexone
Massachusetts 02118, USA. towards personalized obesity medicine. sustained release (SR)/bupropion SR and liraglutide, which
Correspondence to C.M.A.  Previously, obesity was regarded solely as a life­ is the only injectable formulation (TABLE 1). Most of these
caroline.apovian@bmc.org style or behavioural disorder; however, our current drugs work through CNS pathways that either reduce
doi:10.1038/nrendo.2017.122 perspective highlights the complex physiology of the appetite or enhance satiety, with the exception of orlistat,
Published online 13 Oct 2017 disorder 4–6 and its devastating effect on quality of life. which decreases the absorption of fat.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 1


©
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
.
REVIEWS

Key points or uncontrolled hypertension. In 1999, owing to safety


concerns voiced in Belgium, the drug was allowed to
• Obesity is a chronic, debilitating disease with devastating overall health effects and stay on the market provided that periodic safety moni­
a global burden of disease; weight loss can improve these outcomes toring updates and the impact on cardiovascular risk
• Anti-obesity drugs should be initiated promptly when appropriate criteria are met if factors were provided. In 2002, sibutramine was sus­
a patient fails self-directed or professionally directed lifestyle treatment pended in Italy owing to an increase in reported cardio­
• Anti-obesity drugs are approved in patients with a BMI ≥27 kg/m2 with at least one vascular adverse events21. In 2005, the 5‑year Sibutramine
obesity-related comorbidity such as diabetes mellitus, hypertension, hyperlipidaemia Cardiovascular Outcomes (SCOUT; n = 9,804) trial was
or sleep apnoea or in patients with a BMI ≥30 kg/m2 initiated to assess the long-term safety of sibutramine by
• Currently, six major FDA-approved anti-obesity medications are available: comparing patients on sibutramine with those on pla­
phentermine, orlistat, phentermine/topiramate extended release (ER), lorcaserin, cebo30. The SCOUT trial specifically enrolled patients
naltrexone sustained release (SR)/bupropion SR and liraglutide (the only injectable
with cardiovascular disease and/or a high risk of cardio­
formulation)
vascular events (that is, those with type 2 diabetes melli­
• Most of these anti-obesity drugs have an efficacy of 3–7% (estimated net weight loss)
tus (T2DM) and cardio­vascular disease, T2DM alone or
• Identifying the type of obesity on clinical presentation coupled with an understanding cardiovascular disease alone), despite the drug’s contra­
of anti-obesity drug safety, contraindications and adverse effect profiles can
indication in patients with cardio­vascular disease. The
selectively increase weight loss through appropriate use of these drugs
results showed that after 5 years, the overall hazard ratio
(HR) for cardio­vascular events in the sibutramine-treated
group increased (HR 1.16 (95% CI 1.03–1.31; P = 0.02)),
Historical perspective although the HR was similar across individual groups:
The development and approval of new anti-obesity drugs T2DM alone, 1.01 (95% CI 0.74–1.38); cardio­vascular
has been particularly challenging owing to an inauspi­ disease alone, 1.28 (95% CI 0.92–1.78); and T2DM with
cious history of adverse effects dating back to the 1890s, cardiovascular disease, 1.18 (95% CI 1.02–1.37)30. Before
when the use of sheep thyroid extract (to induce weight the study was published, in 2010, following the suspen­
loss in euthyroid patients) resulted in reports of cardiac sion of sibutramine from the European market due to
arrhythmias and death17. Subsequently, in the 1930s, potential risk factors and ongoing safety concerns, the
2,4‑dinitrophenol used for weight loss caused fatal hyper­ FDA asked Abbott Laboratories to voluntarily withdraw
thermia, agranulocytosis and the development of cata­ sibutramine from the US market22. Despite the withdrawal
racts18–20. In the 1950s, amphetamine use became popular, of sibutramine, a 2012 re‑analysis of the SCOUT trial
especially in combination with other mixed regimens showed that maintained intentional weight loss improved
such as diuretics, laxatives and thyroid hormone, but cardiovascular mortality even in those with pre-existing
resulted in reports of serious myocardial infarction and cardiovascular disease31. Because of the chequered initial
sudden deaths21,22. In 1992, a weight-loss trial (n = 121) history of weight-loss medications, anti-obesity drugs
involving combination low-dose fenfluramine with have typically garnered negative publicity over time.
phentermine showed a mean weight loss of 15.9% from The FDA criteria for a drug to be approved for the
baseline over 34 weeks, with no reported major safety treatment of obesity have been stringent since standard
concerns in patients, which resulted in the popularity of guidelines were issued in the mid‑1990s32. The new drug
its usage as a combination drug for weight loss23,24. must induce statistically significant placebo-­adjusted
In 1996, dexfenfluramine, an isomer of fenfluramine, weight loss of >5% at 1 year or >35% of patients should
was approved by the FDA, although preclinical studies achieve >5% weight loss (which must be at least twice that
demonstrated neurotoxicity 25 and epidemiological stud­ induced by placebo). In addition, the FDA also requires
ies associated it with pulmonary hypertension26. The drug that the medication show evidence of improvement in
was to be discontinued if weight loss of more than 1.8 kg metabolic biomarkers, including blood pressure, lipid
did not occur within the first month. Shortly afterwards, levels and glycaemic control32,33. Current anti-­obesity
associated reports of cardiac valvulopathy and pulmo­ medications (FIG. 2; TABLES 1,2) are indicated to be used as
nary hypertension resulted in its immediate withdrawal adjuncts to a reduced-calorie diet and increased physical
from the market, with a published study subsequently activity for chronic weight management in adults with
linking dexfenfluramine and fenfluramine to cardiac a BMI ≥30 kg/m2 or those with a BMI ≥27 kg/m2 who
valvular disease27. Phentermine was not withdrawn at have at least one weight-related comorbid condition such
that time and has remained on the market since, as the as diabetes mellitus, hypertension, hyperlipidaemia or
valvulopathy was linked solely to the serotonin-­releasing sleep apnoea.
agents fenfluramine and dexfenfluramine.
Sibutramine, a serotonin and noradrenaline reup­ Phentermine
take inhibitor that promotes satiety 28, was the next Phentermine, which was approved in 1959 for those
agent approved in 1997 in the USA and Canada after aged >16 years, is still the most commonly prescribed
several clinical trials showed this agent promoted 4.2 kg anti-obesity medication in the USA and in other coun­
(range 3.6–4.7 kg) more weight loss than placebo over tries, with the exception of the European Union due to
12 months29. Safety concerns of the sympathomimetic its potential adverse effects34. Phentermine is a sympa­
drug included elevation of heart rate and blood pres­ thomimetic amine that is believed to suppress appetite
sure, and it thus came with a clear contraindication in and release insignificant quantities of dopamine com­
patients with cardiovascular disease, stroke, arrhythmia pared with amphetamines and thus has a comparatively

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

Determinants affecting the Y1R • Forebrain


cognitive or emotional brain • Pituitary gland
PVN • Adrenal gland
Second-order TRH
Biological influences: neurons • GI afferents
• Early-life events CRH
MC3R/
• Imprinting Orexigenic signalling MC4R
• Epigenetics ↑ Food intake Orexin
• Genetics ↓ Satiety LHA
MCH
α-MSH
Higher cortical
Y2R centres
Anorexigenic signalling
Modern macroenvironment: GHSR ↓ Food intake
• Food production, ↑ Satiety
consumption and Limbic system
availability NPY–AgRP
NST
• Social structure and cues POMC–CART
• Weather or outdoor First-order IR Recent
neurons advances:
temperatures influencing LR
physical activity • Brown fat
Third • Microbiome
• Television and technology ventricle
• Cultural norms Vagus • Muscle (irisin)
• Endocrine disruptors nerve
AC
Median eminence
and area postrema
of blood–brain
barrier
Modern microenvironment: PYY Ghrelin Insulin Leptin GLP1
• Nutrition
• Exercise
• Sleep
Endocrine Produced
• Stressors ‘L’ cells primarily
• Circadian rhythm in ileum– in fundus
cecum
Large Stomach Pancreatic Adipose tissue Small intestine
intestine β cells Produced Produced primarily
primarily in in neuroendocrine
white adipose cells; GLP1R on
tissue dorsal vagal complex
(not shown)
PYY

Figure 1 | Neuronal and hormonal pathways influencing food intake influences food intake and satiety. In addition, biological and modern
Nature Reviews | Endocrinology
and satiety in the brain. Complex neuro–hormonal pathways, gut microenvironmental and macroenvironmental determinants affect the
hormones and adiposity signals reciprocally interact between the cognitive or emotional brain with an impact on energy regulatory
hypothalamus, brainstem, higher cortical areas and limbic system to pathways that gives rise to clinical heterogeneity (variation) in individuals
control appetite regulation. Peripheral signals can also interact via neural with obesity. α‑MSH, α‑melanocyte-stimulating hormone; CRH,
pathways directly. Neuropeptide Y–agouti-related protein (NPY–AgRP; corticotropin-­releasing hormone; GHSR, growth hormone secretagogue
orexigenic) and pro-opiomelanocortin–cocaine- and amphetamine-­ receptor; GI, gastrointestinal; GLP1, glucagon-like peptide 1; GLP1R,
related transcript (POMC–CART; anorexigenic) neurons reside within the GLP1 receptor; IR, insulin receptor; LHA, lateral hypothalamic area; LR,
arcuate nucleus (AC) of the hypothalamus. The cumulative effect of either leptin receptor; MCH, melanin-concentrating hormone; MC3R,
inhibition or activation of these orexigenic and anorexigenic neurons melanocortin receptor 3; NST, nucleus of the solitary tract; PVN,
from various signals in the bloodstream through the incomplete blood– paraventricular nucleus; PYY, peptide YY; TRH, thyrotropin-releasing
brain barriers (median eminence and area postrema) or neural pathways hormone; Y1R, Y1 receptor; Y2R, Y2 receptor.

lower abuse potential35. Although the exact mechanism patients were randomly assigned to three groups37. The
of action is unknown, weight loss might be mediated first group was given phentermine continuously for
through the release of catecholamines in the hypo­ 36 weeks; the second group was given placebo continu­
thalamus, which leads to reduced appetite and decreased ously; and the third group was given alternating phen­
food consumption36. Phentermine monotherapy is termine and placebo every 4 weeks. Mean weight loss
approved for 3 months (90 days), as the 1959 labelling has was −12.2 kg and −13.0 kg in the first and third groups,
not been updated despite our understanding of obesity as respectively, compared with −4.8 kg in the placebo group.
a chronic disease. The attrition rate was 41%, with no reported statistically
significant differences between groups; the data repre­
Efficacy sented only those who completed the trial. In a more
In a 1968 double-blind, placebo-controlled study evaluat­ recent 2013 clinical trial, which evaluated differences
ing the efficacy of continuous versus intermittent phenter­ in phentermine monotherapy compared with combina­
mine in 108 women who were overweight or had obesity, tions, a weight loss of 5.1% was reported at 28 weeks38.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 3


©
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
.
REVIEWS

Table 1 | Currently available anti-obesity medications*,‡,§


Drug (dosage) Mechanism of action Administration Contraindications Warnings Common adverse
effects
Phentermine Sympathomimetic 15 mg or 37.5 mg orally Cardiovascular Rare cases of Increase in HR
(8 mg (short acting), amine once daily; 8 mg orally disease, uncontrolled primary pulmonary and/or BP, dizziness,
15 mg capsule, 2–3 times daily; can hypertension, hypertension or serious dry mouth,
37.5 mg tablet) start with a quarter or a agitated states, regurgitant cardiac constipation,
half of a 37.5 mg tablet history of drug use, valvular disease insomnia and
once daily and titrate hyperthyroidism, irritability
upwards to a maximum glaucoma or MAOI
dosage of 37.5 mg use within 14 days
Orlistat (60 mg OTC, Pancreatic and gastric 120 mg orally three Malabsorption Malabsorption of Flatulence, bloating
120 mg) lipase inhibitor times daily syndrome or fat-soluble vitamins A, and diarrhoea
cholestasis D, E and K
Phentermine/ Combination of Start with 3.75/23 mg Glaucoma, Teratogenicity (risk of Peripheral
topiramate ER|| sympathomimetic orally once daily for hyperthyroidism or cleft palate and/or cleft neuropathy
(3.75/23 mg, amine, anorectic and 14 days; increase to MAOI use within lip), suicidal ideation, (usually transient),
7.5/46 mg, ER antiepileptic drug 7/46 mg once daily 14 days changes in memory dyspepsia, insomnia,
11.25/69 mg and and monthly titration or concentration, constipation and dry
15/92 mg) upwards to achieve metabolic acidosis, mouth
weight loss; discontinue hypokalaemia
if <3% weight loss (especially in patients
on 11.25/69 mg or on potassium-wasting
<5% weight loss diuretics), rise in
on maximum dose creatinine levels,
of 15/92 mg after increase in HR,
12 weeks acute myopia or
acute angle-closure
glaucoma
Lorcaserin|| (10 mg, 5‑HT2c receptor agonist 10 mg orally twice daily Pregnancy Serotonin syndrome Headache, dizziness,
20 mg ER) or 20 mg ER once daily or neuroleptic fatigue, dry mouth,
malignant syndrome; constipation, upper
safety unknown with respiratory tract-like
coadministration symptoms and
of serotonin or hypoglycaemia
antidopaminergic in patients with
agents, cognitive diabetes mellitus
impairment, suicidal
ideation and valvular
heart disease (though
not statistically
significant in clinical
trials)
Naltrexone SR Combination Upwards titration over Uncontrolled Suicidal ideation, Nausea, constipation,
(8 mg)/bupropion opioid antagonist 4 weeks to maximum of hypertension, seizure decrease in seizure headache, vomiting,
SR|| (90 mg) and aminoketone two tablets twice daily disorders, anorexia threshold, acute dizziness, dry mouth
antidepressant nervosa or bulimia, angle glaucoma, and diarrhoea
chronic opioid use, hepatotoxicity to
MAOI use within naltrexone component
14 days, abrupt and increase in HR
discontinuation of and/or BP
alcohol or seizure
medications
Liraglutide|| (3.0 mg) GLP1 receptor agonist Start with 0.6 mg Family or personal Thyroid C‑cell tumours, Nausea,
subcutaneously once history of medullary acute pancreatitis, hypoglycaemia,
daily for 7 days; titrate thyroid cancer or acute gallbladder diarrhoea,
upwards weekly to multiple endocrine disease, increase in HR, constipation,
1.2 mg, 2.4 mg, and then neoplasia type 2 renal impairment and vomiting, headache,
maximum dosage of (MEN2) syndrome hypoglycaemia decreased appetite,
3.0 mg once daily dyspepsia, abdominal
pain, fatigue,
dizziness, increase
in lipase levels and
suicidal behaviour
or ideation
*Approved for BMI ≥30 kg/m2 or ≥27 kg/m2 with at least one comorbid condition such as diabetes mellitus, hypertension, hyperlipidaemia or sleep apnoea.

All contraindicated in pregnancy; medications should not be prescribed with known allergy or hypersensitivity to any drug components.
§
With co‑use of antidiuretic medications, weight loss can cause hypoglycaemia; serum glucose levels should be monitored before and during treatment.
||
Effect on cardiovascular morbidity and mortality has not been established. 5‑HT2c, 5‑hydroxytryptamine receptor 2C; BP, blood pressure; ER, extended release;
GLP1, glucagon-like peptide 1; HR, heart rate; MAOI, monoamine oxidase inhibitor; OTC, over the counter; SR, sustained release.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

abrupt cessation of antihypertensive agents. Medical


Phentermine/topiramate ER
7.5/46 mg for 1 year decision-making regarding off-label, long-term use of
(Gadde et al. 2011)
Phentermine 15 mg daily for 28 weeks
phentermine should be done in concordance with state
(Aronne et al. 2013) medical boards and local laws. Long-term prescription
Naltrexone SR/bupropion SR of phentermine should be limited to patients without
Maximum dose for 56 weeks
(Greenway et al. 2010) evidence of cardiovascular disease or a history of sub­
Liraglutide (Saxenda [liraglutide] stance abuse disorders who have clinical evidence of
3.0 mg for 56 weeks
package insert 2014) drug efficacy and stable baseline heart rate and blood
Lorcaserin pressure readings. Off-label prescribing should also
10 mg twice daily for 1 year
(Smith et al. 2010) include appropriate documentation in medical records
Orlistat 120 mg thrice daily along with a discussion with the patient 8. Case reports of
(Aronne et al. 2013) for 1 year phentermine precipitating atrial fibrillation in patients
0 2 4 6 8 with obesity have been reported in the literature41–43.
% estimated weight loss (drug minus placebo)
Orlistat
Figure 2 | Efficacy of anti-obesity drugs. The percentage estimated weight loss (drug Orlistat is indicated for weight loss in conjunction
minus placebo) for the six currently available anti-obesityNature
drugs isReviews
depicted. ER,
| Endocrinology
with a reduced-calorie diet. It inhibits the activity of
extended release; SR, sustained release. pancreatic and gastric lipases, thereby decreasing fat
absorption by 30%44.

Efficacy
An 8 mg tablet formulation of phentermine was also In a 4‑year double-blind, prospective study in which
approved as a citizen petition under a new abbreviated 3,305 patients with obesity were randomly assigned to
drug application, which was based on earlier studies39. lifestyle changes plus either 120 mg orlistat or placebo
No current studies have evaluated the efficacy of the three times daily, mean weight loss was statistically
8 mg formulation of phentermine compared with stand­ significant with orlistat (5.8 kg versus 3.0 kg with pla­
ard drug dosages of phentermine. Phentermine, like all cebo; P < 0.001)45. Orlistat has been shown to reduce the
sympathomimetic amines, is a Schedule IV controlled incidence of T2DM (6.2% with orlistat compared with
substance. 9.0% with placebo), corresponding to a risk reduction of
37.3% (P < 0.0032)45 and to reduced levels of total cho­
Safety lesterol and LDL independent of weight loss46. Pooled
All sympathomimetics can cause stimulant-like adverse data from randomized, double-blind, placebo-controlled
effects, which include increases in heart rate and blood multicentre clinical trials show a statistically significant
pressure and the provocation of nervousness and/or improvement in glucose tolerance, progression to T2DM
insomnia40. The most common treatment-emergent in adults with obesity and impaired fasting glucose levels
adverse events (TEAEs) associated with phentermine (3.0% with orlistat compared with 7.5% with placebo)47.
include dizziness, dry mouth, difficulty sleeping, irrita­
bility, nausea and/or vomiting, diarrhoea and constipa­ Safety
tion33. Phentermine is contraindicated in patients with The most common TEAEs with orlistat (incidence of
cardiovascular disease, hyperthyroidism, glaucoma 5% and at least twice that of placebo) include flatulence,
or a history of drug abuse and in pregnant women. oily spotting, faecal urgency, fatty/oily stool, oily defeca­
Phentermine should not be used while taking, and for tion, increased defecation and faecal incontinence and
14 days after stopping, a monoamine oxidase (MAO) other adverse effects such as nephrotoxicity, hepato­
inhibitor because of the risk of hypertensive crisis. The toxicity, nephrolithiasis and pancreatitis48,49. Due to fat
recommended dosage of phentermine is 15–37.5 mg mal­absorption, the risk of fat-soluble vitamin deficien­
orally once daily, taken 1–2 h before breakfast; phen­ cies is considerably increased, and patients should be
termine should not be prescribed later in the evening advised to take a multivitamin with the formulation in
owing to risk of insomnia. Dosage adjustment should conjunction with a diet in which ~30% or less of the
be individualized to obtain an adequate treatment calories come from fat. Orlistat is also available as a non-­
response with the lowest effective dose. The 8 mg for­ prescription, over-the-counter lower dose of 60 mg com­
mulation of phentermine is given orally twice or thrice pared with the prescription-strength 120 mg formulation;
daily at least 30 minutes before meals owing to its both formulations are taken orally three times a day.
short-acting effect.
Clinical insight
Clinical insight Undesirable adverse effects coupled with only a mod­
Although phentermine is FDA-approved for weight loss, est average weight loss have decreased the utility and
it is not approved for long-term use. This caveat presents popularity of this particular medication, although
challenges for providers, as weight regain will probably orlistat might have some benefits in patients with
occur following prompt discontinuation of the drug due impaired fasting glucose levels45 and hyperlipidaemia50
to the loss of drug-mediated physiologic effects, simi­ and in those who cannot tolerate the centrally acting
lar to patients with rebound hypertension following the appetite-suppressant drugs.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 5


©
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
.
REVIEWS

Table 2 | Major clinical trials of anti-obesity pharmacotherapy


Drug (trial) Study design n Mean Mean % of patients Comments Refs
weight loss weight loss losing ≥5% of
(kg or %)* (kg or %)‡ baseline weight§
Phentermine 36‑week 108 women with overweight –12.2 kg –4.8 kg 56% (all phentermine Data presented for 37
double-blind, or obesity (continuous groups) versus completers; statistically
placebo- dose phen- 20% with ‘dummy’ significant differences
controlled termine); capsules; although were not reported; attrition
study; conti- −13.0 kg participants lost rate 41%
nuous versus (intermittent weight, the study did
intermittent dose phen- not report whether
phentermine termine) ≥5% weight loss
every other from baseline weight
4 weeks was achieved
Orlistat 4‑year 3,305 patients randomly –5.8 kg –3.0 kg 52.8% versus Cumulative diabetes mellitus 45
double-blind assigned to lifestyle changes 37.3% at 4 years for incidence decreased in the
prospective or lifestyle plus orlistat completers orlistat group versus the
study 120 mg three times a day placebo group (6.2% versus
9.0%, with risk reduction
of 37.3%; P < 0.0032)
Phentermine/ Double-blind, 2,487 patients (BMI –9.8 kg –1.2 kg 70% (15/92 mg) and Patients on 15/92 mg 53
topiramate ER placebo- 27–45 kg/m2 with two (15/92 mg 62% (7.5/46 mg) had greater changes in
(CONQUER) controlled or more risk factors) dose); versus 21% (placebo) blood pressure, waist
trial over were randomly assigned −7.8 kg circumference and levels of
1 year (4‑week to placebo (n = 979), (7.5/46 mg lipids, fasting glucose and
titration plus phentermine/topiramate dose) insulin than those on placebo
52 weeks of ER 7.5/46 mg (n = 488) or
treatment) phentermine/topiramate
ER 15/92 mg (n = 981)
once daily
Phentermine/ 2‑year study 227 patients who completed –10.9 kg –2.1 kg 79.3% (15/92 mg) Patients on 15/92 mg had 54
topiramate overall; 1‑year the original blinded (15/92 mg and 75.2% greater changes in levels of
ER (SEQUEL) extension of treatment dose); (7.5/46 mg) versus lipids, triglycerides, fasting
CONQUER −9.6 kg 30.0% (placebo) glucose and insulin and in
(additional (7.5/46 mg waist circumference than
52 weeks of dose) those on placebo
treatment)
Lorcaserin 1‑year 4,008 patients, aged –4.7 kg –2.9 kg 40.2% (once daily) Key exclusion criteria 67
(BLOSSOM) randomized, 18–65 years, BMI 30–45 kg/ (daily dose); and 47.2% (twice included recent cardio-
double-blind, m2 or 27–29.9 kg/m2 with an −5.8 kg daily) versus 25.0% vascular events, diabetes
placebo- obesity-related condition, (twice daily (placebo) mellitus, systolic blood
controlled randomly assigned in a 2:1:2 dose) pressure ≥150 mmHg or
trial ratio to receive lorcaserin diastolic blood pressure
10 mg twice daily, 10 mg ≥95 mmHg; patients on
once daily or placebo lorcaserin lost more weight
than those on placebo
Lorcaserin 2‑year 3,182 adults with overweight –5.8 kg –2.2 kg 47.5% versus 20.3% Weight loss was maintained 68
(BLOOM) randomized, or obesity (mean BMI in a greater proportion of
double-blind, 36.2 kg/m2) randomly patients who continued to
placebo- assigned to receive receive lorcaserin during
controlled lorcaserin 10 mg twice year 2 than those who were
trial daily or placebo; after switched over to placebo
52 weeks, the placebo group
continued placebo, while
those in lorcaserin arm were
reassigned to either placebo
or an additional 52 weeks of
lorcaserin
Lorcaserin 1‑year 604 patients with baseline –5.0 kg –1.6 kg 44.7% (daily) and Significant decreases in heart 69
(BLOOM‑DM) randomized, HbA1c 7–10%, BMI 27–45 kg/ (daily dose); 37.5% (twice daily) rate with both dosages of
double-blind, m2 and treatment with −4.7 kg versus 16.1% lorcaserin compared with
placebo- metformin, sulfonylurea (twice-daily (placebo) placebo (−2.0 bpm twice-daily
controlled or both were randomly dose) dosing and −2.9 bpm daily
trial assigned to treatment dosing versus −0.4 bpm
(lorcaserin 10 mg twice placebo) and in HDL levels
daily or 10 mg once daily) or and waist circumference.
placebo FDA-defined valvulopathy
was not statistically
significant (P = 0.70; 2.3%
placebo versus 2.7%
lorcaserin group at year 1;
2.7% placebo versus 2.6%
lorcaserin at year 2)

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

Table 2 (cont.) | Major clinical trials of anti-obesity pharmacotherapy


Drug (trial) Study design n Mean Mean % of patients Comments Refs
weight loss weight loss losing ≥5% of
(kg or %)* (kg or %)‡ baseline weight§
Naltrexone 1‑year 1,742 patients randomly –5.0% –1.3% 39% (16/360 mg) and Decreases in body weight 74
SR/bupropion randomized, assigned in a 1:1:1 ratio (16/360 mg 48% (32/360 mg) were greater with NB than
SR (COR‑I) double-blind, to receive placebo, NB dose); −6.1% versus 16% (placebo) with placebo
placebo- 16/360 mg or NB 32/360 mg (32/360 mg
controlled trial dose)
Naltrexone 1‑year 1,496 patients randomly –6.4% –1.2% 50.5% versus 17.1% Weight loss was statistically 75
SR/bupropion randomized, assigned in a 2:1 ratio to greater in the NB group versus
SR (COR‑II) double-blind, NB 32/360 mg or placebo; placebo and was maintained
placebo- patients on NB with <5% with continued NB through
controlled trial weight loss between week 56; randomly reassigning
28–44 weeks were randomly those who did not lose
reassigned to continue NB weight to the higher dose (NB
32/360 mg or escalate to NB 48/360 mg) did not change
48/360 mg outcomes
Naltrexone 1‑year 793 patients with obesity –11.5% –7.3% 66.4% versus 42.5% Weight loss was greater in the 76
SR/ randomized, randomly assigned in a 1:3 NB + BMOD group
bupropion SR double-blind, ratio to placebo + BMOD or compared with placebo + 
(COR-BMOD) placebo- BMOD + NB 32/350 mg BMOD; the efficacy of the
controlled trial drug is fixed, and an intensive
lifestyle modification can add
to weight loss
Naltrexone 1‑year 505 patients with overweight –5.0% –1.8% 44.5% versus 18.9% The NB group lost more 77
SR/bupropion randomized, or obesity and T2DM ± oral weight than the placebo
SR (COR- double-blind, anti-hypoglycaemic group, with improved HbA1c
Diabetes) placebo- medication randomly reduction and improvements
controlled trial assigned in a 2:1 ratio to NB in select cardiovascular risk
32/360 mg or placebo factors. NB was associated
with a higher incidence of
nausea, constipation and
vomiting than placebo. There
were no differences in rates of
depression or suicidal ideation
between the groups
Liraglutide 1‑year 3,731 patients with –8.4 kg –2.8 kg 63.2% versus 27.1% Treatment with liraglutide 85,
(3.0 mg; randomized, overweight or obesity 3.0 mg improved weight 88
SCALE- double-blind, without evidence of T2DM loss, glycaemic index,
Obesity and placebo- were randomly assigned blood pressure and waist
Prediabetes) controlled trial to liraglutide 3.0 mg circumference, while
subcutaneously once daily lowering prevalence of
versus placebo; 61.2% had prediabetes. In the 3‑year
prediabetes follow‑up assessment,
patients with obesity and
prediabetes on liraglutide had
reduced risk of progression to
T2DM by 160 weeks
Liraglutide 1‑year 846 adults with T2DM –6.0% –2.0% 54.2% versus 21.4% Among patients with T2DM 86
(3.0 mg; randomized, and overweight or obesity who had overweight or
SCALE- double-blind, were randomly assigned to obesity, weight loss was
Diabetes) placebo- liraglutide versus placebo greater in the liraglutide
controlled trial group. More gastrointestinal
disorders were reported
with liraglutide 3.0 mg
versus liraglutide 1.8 mg and
placebo. No pancreatitis was
reported
Liraglutide 1‑year 422 adults with overweight –6.2% –0.2% 81.4% versus 48.9% Liraglutide, with diet and 87
(3.0 mg; randomized, or obesity who had lost ≥5% exercise, maintained weight
SCALE- double-blind, of initial body weight during loss achieved by calorie
Maintenance) placebo- a calorie-restriction period restriction and induced
controlled trial were randomly assigned to further weight loss over 56
liraglutide versus placebo weeks. Improvements in some
cardiovascular disease risk
factors were also observed
*Treatment from baseline reported. ‡Placebo from baseline reported. §Treatment versus placebo. BLOOM, Behavioural Modification and Lorcaserin for Overweight
and Obesity Management; BLOSSOM, Behavioural Modification and Lorcaserin Second Study Group for Obesity Management; BMOD, behavioural modification;
bpm, beats per minute; CONQUER, Controlled-Release Phentermine plus Topiramate Combination in Overweight and Obese Adults; COR, Contrave Obesity
Research; ER, extended release; NB, naltrexone SR/bupropion SR; SCALE, Satiety and Clinical Adiposity — Liraglutide Evidence in Nondiabetic and Diabetic
Individuals; SEQUEL, 2‑year Sustained Weight Loss and Metabolic Benefits with Controlled-release Phentermine/Topiramate in Obese and Overweight Adults;
SR, sustained release; T2DM, type 2 diabetes mellitus.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 7


©
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
.
REVIEWS

Phentermine/topiramate ER (n = 295)) were entered into an additional 52‑week


Phentermine/topiramate is an extended-release combina­ (total 108 weeks, approximately 2 years) SEQUEL trial54.
tion that was approved by the FDA in 2012. Topiramate Overall, 84% of the patients completed the SEQUEL study.
is also an antiepileptic drug and is used to treat migraine Reported mean percentage changes in body weight were
headaches. The exact anorexigenic mechanism of significantly greater on the combination drug (−9.3%
topiramate is not well understood, although it is postu­ in the 7.5/46 mg group, −10.5% in the 15/92 mg group)
lated that the effects are mediated through modulation compared with the placebo group (−1.8%; P < 0.0001).
of various neurotransmitters, including the inhibition of In addition, compared with 56 weeks (CONQUER), at
voltage-dependent sodium channels, glutamate recep­ 108 weeks (SEQUEL), a greater percentage of patients lost
tors and carbonic anhydrase and the potentiation of >5% of baseline weight (placebo group: 30.0%; 7.5/46 mg
γ-aminobutyrate activity 51. The combination drug has group: 75.2%; and 15/92 mg group: 79.3%)54. In the
shown greater potential weight-loss effects than mono­ secondary end point analysis of all three studies, patients
therapy alone for each while also reducing adverse effects. on the combination drug had statistically significant
improvements in their lipid profile, glycaemic control
Efficacy and waist circumference52–54.
Two large randomized, double-blind, placebo-­
controlled trials over 52 weeks with a 4‑week titration Safety
period (Controlled-Release Phentermine/Topiramate Concerns expressed by the FDA included teratogenicity
in Severely Obese Adults (EQUIP)52 and Controlled- (specific to both components of the combination drug)
Release Phentermine plus Topiramate Combination and cardiovascular concerns (specific to the phentermine
in Overweight and Obese Adults (CONQUER)53), fol­ component) as well as cognitive, psychiatric and metabolic
lowed by a 2‑year extension trial with an additional acidosis (the last three being specific to the topiramate
52 weeks of treatment (2‑year Sustained Weight Loss and component)55. In the EQUIP and CONQUER trials, the
Metabolic Benefits with Controlled-release Phentermine/ most common adverse events with an incidence of >1%
Topiramate in Obese and Overweight Adults (SEQUEL)54 leading to treatment discontinuation included blurred
for completers from the CONQUER study) assessed the vision, headache, irritability, dizziness, paraesthesia,
efficacy of the combination drug in conjunction with insomnia, depression and anxiety 52,53. In addition, in the
intensive lifestyle modification and a well-balanced, SEQUEL trial, upper respiratory tract infection, constipa­
reduced-­calorie diet. The EQUIP trial included 1,267 tion, paraesthesia and dry mouth were noted54. Rates of
adults with severe obesity (BMI >35 kg/m2) who were discontinuation at 1 year due to adverse effects were 11.6%
randomly assigned to either phentermine/topiramate in the 3.75/23 mg group and 7.5/46 mg group, 17.4% in the
ER 3.75 mg/23  mg or phentermine/topiramate ER 15/92 mg group and 8.4% in the placebo group52,53.
15 mg/92 mg once daily. No statistically significant differ­ In unrelated studies, an increased risk of cleft palate
ences were observed between baseline groups, and 59.9% and/or cleft lip in infants born to mothers on topiramate
of the participants completed the study regardless of their therapy during the first trimester (prevalence ratio 5.4
assigned treatment. for women on topiramate versus those not exposed to
In the primary efficacy end point analysis, patients topiramate (95% CI 2.0–14.5)) was observed56. In addi­
had a statistically significant greater weight loss on the tion, topiramate might decrease the efficacy of oral con­
combination overall than on placebo, with an average traceptives, although this is unlikely at doses <200 mg per
weight loss of −5.1 kg from baseline in the patients who day 57. Although topiramate is not a controlled Schedule
received 3.75/23 mg compared with −10.9 kg in those IV drug, owing to the phentermine component, the drug
who received 15/92 mg. Approximately 17% of those on falls under the Schedule IV Controlled Substances Act.
placebo, 45% of those in the 3.75/23 mg group and 67% of Phentermine/topiramate ER should be taken once daily
those in the 15/92 mg group lost >5% of baseline weight52. in the morning with or without food. Gradual titration
In CONQUER, a total of 2,487 patients (BMI 27–45 kg/m2 is required, with an initial starting dose of 3.75/23 mg
with two or more risk factors) were randomly assigned to once daily for 14 days and then an increase to 7.5/46 mg
placebo (n = 979), phentermine/topiramate ER 7.5/46 mg daily. Further dose escalation to 11.25/69 mg and then
(n = 488) or phentermine/topiramate ER 15/92  mg to 15/92 mg is recommended in nonresponders if <3%
(n = 981) once daily with no statistically significant dif­ weight loss has occurred by 12 weeks on the middle
ferences across groups. The combination drug overall dose (7.5/46 mg) or <5% weight loss by 12 weeks on
resulted in significantly greater weight loss than placebo, the maximum dose (15/92 mg). Due to the increased
with the higher dosage resulting in greater weight loss risk of seizures associated with the abrupt withdrawal of
than the lower dosage of the medication (−7.8 kg and topiramate, graduated downward titration over 3–5 days
−9.8 kg from baseline, respectively, versus −1.2 kg for is recommended52–54,58.
placebo). Approximately 21% of patients in the pla­
cebo group, 62% of those in the 7.5/46 mg group and Clinical insight
70% of those in the 15/92 mg group lost >5% of baseline Although expensive, the combination drug has robust
body weight53. weight-loss potential. The individual components of the
Participants who completed CONQUER and drug can be prescribed separately in available mono­therapy
who continued their randomly assigned treatments dosages (phentermine 8 mg, 15 mg, 30 mg and 37.5 mg;
(placebo (n = 227), 7.5/46 mg (n = 153) and 15/92 mg topiramate 15 mg, 25 mg, 50 mg, 100 mg and 200 mg).

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

Although topiramate monotherapy is not currently The BLOOM trial (n = 3,182 patients with an average
FDA-approved for the treatment of obesity, it has shown BMI of 36.2 kg/m2) evaluated primary outcomes on lor­
benefit in patients with emotional eating and binge-­ caserin (10 mg twice daily versus placebo) at 1 year and
eating disorder 59 and against weight regain following maintenance of weight loss at 2 years. After 1 year,
bariatric surgery 60. Topiramate is usually prescribed patients either continued placebo, or those on lorcaserin
starting at 25 mg per day for the first 1–2 weeks and is were randomly assigned to either placebo or continuation
then increased in increments of 25 mg monthly (to a of the drug for an additional 52 weeks. Mean body weight
maximum of 100–150 mg); dose and titration should be was lower in patients treated with lorcaserin for 2 years
guided by clinical outcome61. Close follow-up is recom­ compared with the other groups. Mean weight loss in
mended once monthly for the initial 3 months and then the lorcaserin group was greater at year 1 (−5.8 kg versus
every 2–3 months thereafter. Transient adverse effects that −2.2 kg for placebo; P < 0.001) and continued into year 2
resolve quickly (for example, tingling or numbness) are (−8.1 kg versus −3.3 kg for placebo; P < 0.001), at which
not usually concerning; however, persistent and worsen­ time the weight loss was maintained in a larger portion of
ing peripheral neuropathy or cognitive impairment such patients who had >5% weight loss at year 1 on lorcaserin
as personality changes, lethargy, language impairment, and who continued to receive the drug than in those who
word-finding difficulty and forgetfulness warrant prompt were switched over to placebo in year 2 (67.9% versus
attention with immediate lowering of the drug dosage or 50.5%, respectively; P < 0.001)68. The 10 mg twice-daily
discontinuation. These adverse effects are reversible62. dosing was also associated with greater improvements
in the secondary end points of lipid profile and waist
Lorcaserin circumference than placebo.
Lorcaserin, a 5‑hydroxytryptamine receptor 2C (5‑HT2c) The BLOOM‑DM study enrolled patients with
agonist that acts on anorexigenic POMC neurons in the T2DM (n = 604) with a baseline HbA1c of 7–10% on met­
hypothalamus, was also approved by the FDA in 2012 as formin ± sulfonylurea in patients who were overweight
an adjunct to a reduced-calorie diet and increased physi­ or had obesity with at least one obesity-related medical
cal activity for chronic weight management in adults with comorbidity. These patients were assigned to lorcaserin
a BMI ≥30 kg/m2 or those with a BMI ≥27 kg/m2 who have 10 mg twice daily (n = 256; 66.6% completion rate), lor­
at least one weight-related comorbid condition such as caserin 10 mg once daily (n = 95; 78.9% completion rate) or
diabetes mellitus, hypertension, hyperlipidaemia or sleep placebo (n = 253; 61.2% completion rate). The lorcaserin
apnoea63. Lorcaserin has specific selectivity towards the groups had statistically significant reductions in mean
5‑HT2c receptor, which alleviates risk associated with percentage body weight change from baseline (−4.5%
prior agents of this class, such as fenfluramine, which and −5.0% for 10 mg twice daily and 10 mg once daily,
was found to have affinity for both 5‑HT2A (causing hal­ respectively) compared with placebo (−1.5%) and greater
lucinations) and 5‑HT2B receptors (causing cardiac valve reductions in baseline HbA1c levels (>0.5%, P < 0.001)69.
insufficiency and pulmonary hypertension)64. Serotonin
has been known to modulate food intake and appetite65,66. Safety
The most common adverse effects reported by >2% of
Efficacy patients on lorcaserin without diabetes mellitus and more
The efficacy of lorcaserin was addressed in three major commonly than in those on placebo include headache, diz­
randomized, double-blind, placebo-controlled trials of ziness, fatigue, nausea, dry mouth and constipation. The
52‑week or 104‑week duration that enrolled patients most common adverse effects reported in patients with
with overweight or obesity: Behavioural Modification and diabetes mellitus on lorcaserin include hypo­glycaemia
Lorcaserin Second Study Group for Obesity Management (blood glucose levels <65 mg/dl), headache, back pain,
(BLOSSOM)67; Behavioural Modification and Lorcaserin nasopharyngitis, cough and fatigue. Patients in all three
for Overweight and Obesity Management (BLOOM)68; trials had serial echocardiograms due to past cardiac
and BLOOM-DM69, which evaluated the drug specifically valvulo­pathy concerns with the older serotonin analogues.
in adults with T2DM. The incidence of new cardiac valvulopathy in lorcaserin
The 1-year BLOSSOM trial (n = 4,008 patients with a versus placebo groups was not statistically significant
BMI ≥27 kg/m2 and more than one obesity-related con­ across all three trials; the incidence of worsening valvulo­
dition or a BMI of 30–45 kg/m2, aged 18–65 years without pathy in patients who already had existing mitral or aortic
cardiovascular disease or diabetes mellitus, all of whom regurgitation was also not statistically significant (12.1%
received diet and exercise counselling) randomly assigned with lorcaserin twice daily; 11.1% with lorcaserin once
patients to three treatment arms: 10 mg lorcaserin twice daily; and 30.6% with placebo in BLOSSOM; P = 0.056)67.
daily, 10 mg lorcaserin once daily or placebo; 55.5% of the In a pooled analysis of all phase III trials, lorcaserin did
participants completed the trial. Both lorcaserin dosage not adversely increase FDA-defined cardiac valvulopathy
arms lost significantly more weight than the placebo arm, in patients with pre-existing disease69. Lorcaserin should
with the twice-daily dosing associated with more weight be administered at a dose of 10 mg twice daily orally or
loss. The mean percentage weight change from baseline 20 mg extended-release once daily with or without food.
was −2.8%, −5.8% and −4.7% in the placebo, 10 mg twice Lorcaserin should be discontinued if <5% weight loss is
daily and 10 mg once daily groups, with 25.0%, 47.2% achieved at 12 weeks and should not be coadministered
and 40.2% of patients losing >5% of baseline weight, with another drug of the serotonin class, which might lead
respectively 67. to neuroleptic malignant-like syndrome63.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 9


©
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
.
REVIEWS

Clinical insight of initial body weight on the NB + BMOD regimen


Although lorcaserin is recommended as a 10 mg twice- versus BMOD alone (P < 0.001)76. In COR-Diabetes,
daily dose, in clinical trials, the 10 mg once-daily participants had diagnosed T2DM ± antidiabetic med­
dose produced similar weight-loss outcomes as the ication. The NB 32/360 mg group lost on average −5.1%
twice-daily dosing 67,69. In addition, a 20 mg once-daily of baseline weight compared with the placebo group
extended-­release formulation of lorcaserin is now avail­ (−1.8%; P < 0.001), with 44.5% of the patients losing
able. In patients with cardiovascular disease, where sym­ >5% of baseline weight compared with those on pla­
pathomimetics should be avoided, lorcaserin might be a cebo (18.9%; P < 0.001), and had significantly greater
good option. Lorcaserin has a lower potential for increas­ reductions in HbA1c and improved cardiovascular risk
ing heart rate and blood pressure than phentermine and factors, including lipid profile77.
phentermine/topiramate ER68.
Safety
Naltrexone SR/bupropion SR The combination, NB, did not increase rates of depres­
The naltrexone SR/bupropion SR (NB) combination sion and suicidal ideation more than placebo in the
was demonstrated in preclinical animal models to func­ clinical trials74–77. Transient nausea, most frequently
tion in a synergistic manner, augmenting POMC activ­ occurring during the 3‑week dose-escalation period, was
ity to cumulatively reduce food intake and satiety 70,71. the most common adverse effect in approximately one-
Naltrexone blocks opioid receptor-mediated POMC third of patients. Other more common adverse events
auto-inhibition, and bupropion selectively inhibits reup­ reported with an incidence of at least 2% among patients
take of dopamine and noradrenaline. Monotherapy of treated with NB compared with placebo included nau­
these two drugs has also been used to treat addiction to sea, constipation, headaches, vomiting, dizziness and dry
nicotine (bupropion) and alcohol (naltrexone), and thus mouth78. The current NB formulation is available in 8 mg
the combination has an effect on CNS reward pathways, naltrexone SR and 90 mg bupropion combined dosing,
food intake and satiety through antagonistic feedback which is upwards titrated over a 4‑week period for a total
inhibition33,72. Bupropion monotherapy (300 mg per day) dosage of two tablets twice daily: one tablet once daily in
in patients with obesity and depressive symptoms has the morning during week 1, one tablet twice daily during
also been shown to facilitate weight loss (−4.6% versus week 2, two tablets in the morning and one tablet in the
−1.8% with placebo (n = 191); P < 0.001)73. evening during week 3, and finally two tablets twice daily
during week 4. The drug should be discontinued if <5%
Efficacy weight loss is achieved over 12 weeks and should not be
Four major 56‑week phase III randomized, placebo-­ used in patients with a history of seizures, drug addic­
controlled trials have evaluated the efficacy of combina­ tion and bulimia and/or anorexia nervosa. Patients
tion NB (variable dosages) versus placebo — Contrave should be advised to avoid the medication before a
Obesity Research  I (COR-I; n = 1,742) 74, COR-II procedure such as dental work, in which opiates might
(n = 1,496)75, COR-Behavioural Modification (COR- be administered.
BMOD; n = 793)76 and COR-Diabetes (n = 505)77 — in
patients with either a BMI of 30–45 kg/m2 or a BMI Clinical insight
>27–29.9 kg/m2 with evidence of dyslipidaemia and/or Adverse effects such as nausea are particularly com­
hypertension. mon in patients prescribed NB. During the initial drug-­
In COR‑I, the percentage weight loss from base­ dosage titration stage, if a patient experiences significant
line on NB 16/360 mg, NB 32/360 mg or placebo was nausea or other adverse effects, the dose should not be
−5.0%, −6.1% and −1.3%, respectively (P < 0.000), further escalated until the adverse effects have subsided
with 39% (16/360 mg) and 48% (32/360 mg) of the or the patient has demonstrated better tolerability.
patients on NB losing >5% of initial body weight Persistent or lingering adverse effects might warrant dis­
compared with those on placebo (16%; P < 0.0001; continuation of the drug. If no more than 5% of initial
P < 0.0099 for NB 16/360 mg versus NB 32/360 mg)74. body weight is lost within 12 weeks, NB should be dis­
COR‑II evaluated patients on NB 32/360 mg versus continued. In patients with food addiction and obesity,
placebo; the 16/360 mg dosing was not included in NB is advantageous74,78. A particular efficacy in patients
this trial. Patients on NB 32/360 mg lost significantly with binge-eating disorder and concomitant alcohol
more weight from baseline (−6.4% versus −1.2% with abuse has been anecdotally observed with NB, but this
placebo; P < 0.001), with 50.5% of the patients losing observation requires further study 79–82.
>5% of initial body weight (compared with 17.1% of
those on placebo; P < 0.001)75. COR-BMOD compared Liraglutide
the obesity drug NB 32/360 mg added to an intensive Liraglutide is a glucagon-like peptide 1 (GLP1) receptor
behavioural modification intervention (NB + BMOD) agonist of an incretin-derived hormone that acts through
versus the intervention alone (BMOD) in conjunction both peripheral and central receptor pathways affecting
with a reduced-calorie diet. At week 56, the average glucose homeostasis, food intake and satiety. Because of
changes in body weight were significantly greater for its incretin properties, this compound is an attractive
the NB + BMOD group versus BMOD alone (−11.5% drug for patients with T2DM and obesity. Currently, lira­
versus −7.3%; P < 0.001), with more patients losing >5% glutide is the only injectable anti-obesity drug; all others
(66.4% versus 42.5%) and >10% (41.5% versus 20.2%) are taken orally.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

Efficacy unknown in humans and no incidence of medullary thy­


In dose–response studies, liraglutide at all dosages was roid cancer on liraglutide has been reported to date92,93.
found to cause a greater mean weight loss than placebo; Liraglutide is administered subcutaneously once daily
the weight-loss response was also dose-dependent. in a 5‑week upwards dose titration to 3.0 mg. Moreover,
Liraglutide 3.0 mg caused significantly greater weight loss liraglutide is of particular benefit in patients with T2DM
than placebo or orlistat83,84. The efficacy of 3.0 mg liraglu­ or those with severe insulin resistance.
tide subcutaneously once weekly versus placebo in combi­
nation with a reduced-calorie diet and increased physical Clinical insight
activity was further assessed in four 56‑week studies: Liraglutide is initiated at 0.6 mg subcutaneously once
Satiety and Clinical Adiposity — Liraglutide Evidence in daily for the first week; the dose is escalated by 0.6 mg
Nondiabetic and diabetic individuals (SCALE)-Obesity increments every week, up to a maximum of 3.0 mg. If a
and Prediabetes85 (n = 3,731 individuals without diabetes patient experiences adverse effects such as nausea dur­
with a BMI >30 kg/m2 and overweight individuals with­ ing dose escalation, dose increases should be aborted
out diabetes with a BMI >27 kg/m2 plus more than one until better tolerability or cessation of adverse effects is
comorbidity); SCALE-Diabetes86 (n = 846 adults with achieved84. In patients with T2DM and overweight or
T2DM); SCALE-Maintenance87 (n = 422 patients who obesity, efforts to tailor pharmacological therapy for
had already lost >5% of initial body weight by calorie diabetes mellitus to promote weight loss, such as the
restriction); and follow-up 3‑year assessment 88 of the addition of GLP1 receptor agonists to the first-line agent
SCALE-Obesity and Prediabetes trial (n = 2,254 patients metformin, are recommended8.
with prediabetes who completed SCALE-Obesity and
Prediabetes re‑randomized 2:1 to either liraglutide Future directions — personalized therapy
3.0 mg subcutaneously once daily for 2 years versus pla­ The field of obesity pharmacotherapy has evolved over
cebo with a 12‑week, off-treatment follow‑up period with time. We now know that this complex and regulated
the primary outcome defined as time to diabetes onset by human energy system yields biological heterogeneity
160 weeks). In all three primary trials, patients on lira­ that translates into clinical heterogeneity and, in turn,
glutide lost significantly more weight from baseline than innumerable different obesity phenotypes and varia­
those on placebo (8% versus 2.6%, respectively, in SCALE- tions in treatment response. We have only just begun
Obesity and Prediabetes; 6.0% versus 2.0%, respectively, to recognize clinical obesity phenotypes and have not
in SCALE-Diabetes; additional 6.2% versus 0.2%, yet progressed to pharmacometabonomics research
respectively, in SCALE-Maintenance; P < 0.0001)85–87. that could identify therapeutic gene clusters involved in
A higher percentage of patients lost statistically more distinguishing early responders from nonresponders to
weight (>5% from baseline weight) on liraglutide than weight-loss medications94.
those in the placebo arm, with significant improvements Delineation, elucidation and translation of these
in glycaemic control, lipid profile and cardio­vascular clinical phenotypes into identifiable genotypes for
markers, including waist circumference 85–87. In the therapeutic purposes is not new to personalized med­
3‑year follow‑up to SCALE-Obesity and Prediabetes, icine; in fact, metabolic and genetic profiling in order
66% of patients in the liraglutide group (970/1,472) had to target responders is a novel approach to personalized
regressed from prediabetes to normoglycaemia by week drug therapy and can positively affect patient care and
160, compared with 36% of patients in the placebo group outcomes95. Psychiatric medicine, where only a subset
(268/738; odds ratio 3.6, 95% CI 3.0–4.4, P < 0.001). of patients respond to pharmacotherapy and ineffec­
Liraglutide induced greater weight loss than placebo at tive treatment results in unnecessary adverse effects of
week 160 (−6.1% (s.d. 7.3) for liraglutide versus −1.9% drug exposure, has utilized pharmacogenetic genom­
for placebo, P < 0.0001)88. ics to predict underlying treatment failure or success
response rates96. For example, a recent psychiatric study
Safety suggested that genetic variation in ERBB4 might dif­
The most common TEAEs across all four trials were ferentially affect schizophrenia treatment response to a
mostly gastrointestinal (nausea, diarrhoea, constipa­ specific drug 97. Similarly, tamoxifen therapy in oncologic
tion, vomiting, dyspepsia and abdominal pain), tran­ treatment of patients with breast cancer is influenced
sient and of mild to moderate intensity 85–88. In past years, by genetic variants and drug interaction of cytochrome
lira­glutide, an incretin-based agent, has been reported P450 2D6 inhibitors, which influence plasma concentra­
to cause pancreatitis on the basis of spontaneous post-­ tions of active drug metabolites and hence outcomes of
marketing reports, although the association has not been patients treated with tamoxifen98–101.
found to be statistically significant or clearly evident for Recognizing and targeting phenotypes early in the
pancreatitis or pancreatic cancer 89,90. If pancreatitis is treatment algorithm can result in successful clinical
suspected, the medication should be discontinued with response rates. Over the past decade, metformin has
appropriate management. Liraglutide is contraindicated demonstrated efficacy in the prevention of weight gain
in patients with a family or personal history of medul­ and, in some cases, in the promotion of weight loss in
lary thyroid carcinoma or patients with multiple endo­ both adults and children on antipsychotic medications
crine neoplasia type 2 (MEN2) syndrome91. Malignant (a distinct phenotype)102–104. Our paradigm approach
thyroid C‑cell carcinomas were detected in rats and for patients meeting criteria for anti-obesity pharmaco­
mice exposed to liraglutide, although this correlation is therapy (that is, a BMI >27 kg/m2 with comorbidity or

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 11


©
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
.
REVIEWS

a BMI >30 kg/m2) most commonly recommends the most commonly reported in clinical trials through linear
addition of FDA-approved medications to lifestyle mod­ regression models. This point is particularly important
ification, provided the patient does not have a specific to note when evaluating the efficacy of an anti-obesity
contraindication or adverse effect on the medication8. drug in an individual patient. Although current obesity
However, there are currently no recommendations for pharmaco­therapy research has not yet progressed to pre­
which type or class of patients the drugs will be supra-­ dictive biological markers of successful clinical response,
therapeutic for, and this art is left to the skilled obesity the future holds promise given the recent availability of
specialist with expertise and experience in recognizing newly FDA-approved anti-obesity drugs.
and appreciating these phenotypes.
Again, variation in individual treatment response is Conclusions
evident, and thus successful patient phenotyping, as a Weight-loss intervention requires a multimodal com­
first step, would provide more accurate therapy selection prehensive approach that utilizes both lifestyle and
and an improved therapeutic algorithm. We also know behavioural interventions. In addition, pharmacological
individual variation in weight occurs in response to therapy as an adjunct therapy can now be used for those
diet 105 despite dietary adherence, obesity drugs67,68,75 and patients who fail to respond to lifestyle modification
even bariatric surgery 106. Whereas some patients lose a alone. However, pharmacological application of these
significant portion of weight as expected (>5% of initial anti-obesity drugs continues to be an art, given varia­
body weight), others actually gain weight, and a subset tion between individuals in their response to the drugs
might have treatment failures (losing <5% of initial body despite a statistically significant >5% average weight loss
weight), which results in an average weight loss being reported in clinical trials.

1. Tremmel, M., Gerdtham, U. G., Nilsson, P. M. & 17. Bhasin, S., Wallace, W., Lawrence, J. B. & Lesch, M. 33. Apovian, C. M., Aronne, L. & Powell, A. Clinical
Saha, S. Economic burden of obesity: a systematic Sudden death associated with thyroid hormone abuse. Management of Obesity 1 st edn (Professional
literature review. Int. J. Environ. Res. Public Health Am. J. Med. 71, 887–890 (1981). Communications, 2015).
14, E435 (2017). 18. Grundlingh, J., Dargan, P. I., El‑Zanfaly, M. & 34. Hampp, C., Kang, E. M. & Borders-Hemphill, V. Use of
2. GBD 2015 Obesity Collaborators. Health effects of Wood, D. M. 2,4‑Dinitrophenol (DNP): a weight loss prescription antiobesity drugs in the United States.
overweight and obesity in 195 countries over agent with significant acute toxicity and risk of death. Pharmacotherapy 33, 1299–1307 (2013).
25 years. N. Engl. J. Med. 377, 13–27 (2017). J. Med. Toxicol. 7, 205–212 (2011). 35. Hendricks, E. J. et al. Addiction potential of
3. Flegal, K. M., Kruszon-Moran, D., Carroll, M. D., 19. Kamour, A. et al. Increasing frequency of severe phentermine prescribed during long-term treatment
Fryar, C. D. & Ogden, C. L. Trends in obesity among clinical toxicity after use of 2,4‑dinitrophenol in the of obesity. Int. J. Obes. 38, 292–298 (2014).
adults in the United States, 2005 to 2014. JAMA UK: a report from the National Poisons Information 36. Samanin, R. & Garattini, S. Neurochemical mechanism
315, 2284–2291 (2016). Service. Emerg. Med. J. 32, 383–386 (2015). of action of anorectic drugs. Pharmacol. Toxicol. 73,
4. Zheng, H., Lenard, N. R., Shin, A. C. & Berthoud, H. R. 20. Gehring, P. J. & Buerge, J. F. The cataractogenic 63–68 (1993).
Appetite control and energy balance regulation in the activity of 2,4‑dinitrophenol in ducks and rabbits. 37. Munro, J. F., MacCuish, A. C., Wilson, E. M. &
modern world: reward-driven brain overrides repletion Toxicol. Appl. Pharmacol. 14, 475–486 (1969). Duncan, L. J. Comparison of continuous and
signals. Int. J. Obes. 33 (Suppl. 2), S8–S13 (2009). 21. Haslam, D. Weight management in obesity — past intermittent anorectic therapy in obesity. Br. Med. J.
5. Lenard, N. R. & Berthoud, H. R. Central and and present. Int. J. Clin. Pract. 70, 206–217 (2016). 1, 352–354 (1968).
peripheral regulation of food intake and physical 22. Bray, G. A. Medical treatment of obesity: the past, the 38. Aronne, L. J. et al. Evaluation of phentermine and
activity: pathways and genes. Obesity (Silver Spring) present and the future. Best Pract. Res. Clin. topiramate versus phentermine/topiramate extended-
16 (Suppl. 3), S11–S22 (2008). Gastroenterol. 28, 665–684 (2014). release in obese adults. Obesity (Silver Spring) 21,
6. Berthoud, H. R. Interactions between the “cognitive” 23. Weintraub, M. et al. Long-term weight control study. I 2163–2171 (2013).
and “metabolic” brain in the control of food intake. (weeks 0 to 34). The enhancement of behavior 39. [No authors listed.] In brief: phentermine (Lomaira) for
Physiol. Behav. 91, 486–498 (2007). modification, caloric restriction, and exercise by weight loss. Med. Lett. Drugs Ther. 58, 158 (2016).
7. Vetter, M. L., Faulconbridge, L. F., Webb, V. L. & fenfluramine plus phentermine versus placebo. 40. Yanovski, S. Z. & Yanovski, J. A. Long-term drug
Wadden, T. A. Behavioral and pharmacologic therapies Clin. Pharmacol. Ther. 51, 586–594 (1992). treatment for obesity: a systematic and clinical review.
for obesity. Nat. Rev. Endocrinol. 6, 578–588 (2010). 24. Weintraub, M. Long-term weight control study: JAMA 311, 74–86 (2014).
8. Apovian, C. M. et al. Pharmacological management of conclusions. Clin. Pharmacol. Ther. 51, 642–646 (1992). 41. Magnani, J. W., Hylek, E. M. & Apovian, C. M. Obesity
obesity: an endocrine Society clinical practice 25. McCann, U. et al. Dexfenfluramine and serotonin begets atrial fibrillation: a contemporary summary.
guideline. J. Clin. Endocrinol. Metab. 100, 342–362 neurotoxicity: further preclinical evidence that clinical Circulation 128, 401–405 (2013).
(2015). caution is indicated. J. Pharmacol. Exp. Ther. 269, 42. Apovian, C. M. & Aronne, L. J. The 2013 American
9. Suzuki, K., Simpson, K. A., Minnion, J. S., Shillito, J. C. 792–798 (1994). Heart Association/American College of Cardiology/The
& Bloom, S. R. The role of gut hormones and the 26. Kramer, M. S. & Lane, D. A. Aminorex, Obesity Society Guideline for the management of
hypothalamus in appetite regulation. Endocr. J. 57, dexfenfluramine, and primary pulmonary overweight and obesity in adults: what is new about
359–372 (2010). hypertension. J. Clin. Epidemiol. 51, 361–364 (1998). diet, drugs, and surgery for obesity? Circulation 132,
10. Clemmensen, C. et al. Gut–brain cross-talk in 27. Connolly, H. M. et al. Valvular heart disease 1586–1591 (2015).
metabolic control. Cell 168, 758–774 (2017). associated with fenfluramine–phentermine. N. Engl. 43. Apovian, C. M. & Gokce, N. Obesity and
11. Yeo, G. S. & Heisler, L. K. Unraveling the brain J. Med. 337, 581–588 (1997). cardiovascular disease. Circulation 125, 1178–1182
regulation of appetite: lessons from genetics. 28. Araujo, J. R. & Martel, F. Sibutramine effects on (2012).
Nat. Neurosci. 15, 1343–1349 (2012). central mechanisms regulating energy homeostasis. 44. Hvizdos, K. M. & Markham, A. Orlistat: a review of
12. Rodriguez, A., Ezquerro, S., Mendez-Gimenez, L., Curr. Neuropharmacol. 10, 49–52 (2012). its use in the management of obesity. Drugs 58,
Becerril, S. & Fruhbeck, G. Revisiting the adipocyte: a 29. Rucker, D., Padwal, R., Li, S. K., Curioni, C. & 743–760 (1999).
model for integration of cytokine signaling in the Lau, D. C. Long term pharmacotherapy for obesity 45. Torgerson, J. S., Hauptman, J., Boldrin, M. N. &
regulation of energy metabolism. Am. J. Physiol. and overweight: updated meta-analysis. BMJ 335, Sjostrom, L. XENical in the prevention of diabetes in
Endocrinol. Metab. 309, E691–E714 (2015). 1194–1199 (2007). obese subjects (XENDOS) study: a randomized study
13. Qian, S., Huang, H. & Tang, Q. Brown and beige fat: 30. James, W. P. et al. Effect of sibutramine on of orlistat as an adjunct to lifestyle changes for the
the metabolic function, induction, and therapeutic cardiovascular outcomes in overweight and obese prevention of type 2 diabetes in obese patients.
potential. Front. Med. 9, 162–172 (2015). subjects. N. Engl. J. Med. 363, 905–917 (2010). Diabetes Care 27, 155–161 (2004).
14. Contreras, C., Nogueiras, R., Dieguez, C., 31. Caterson, I. D. et al. Maintained intentional weight 46. Muls, E., Kolanowski, J., Scheen, A. & Van Gaal, L. The
Rahmouni, K. & Lopez, M. Traveling from the loss reduces cardiovascular outcomes: results from effects of orlistat on weight and on serum lipids in
hypothalamus to the adipose tissue: the thermogenic the Sibutramine Cardiovascular OUTcomes (SCOUT) obese patients with hypercholesterolemia: a
pathway. Redox Biol. 12, 854–863 (2017). trial. Diabetes Obes. Metab. 14, 523–530 (2012). randomized, double-blind, placebo-controlled,
15. Mathur, R. & Barlow, G. M. Obesity and the 32. Department of Health and Human Services, Food and multicentre study. Int. J. Obes. Relat. Metabol.
microbiome. Exp. Rev. Gastroenterol. Hepatol. 9, Drug Administration & Center for Drug Evaluation and Disord. 25, 1713–1721 (2001).
1087–1099 (2015). Research (CDER). Guidance for Industry: developing 47. Heymsfield, S. B. et al. Effects of weight loss with
16. Bradlow, H. L. Obesity and the gut microbiome: products for weight management. FDA https://www. orlistat on glucose tolerance and progression to type 2
pathophysiological aspects. Horm. Mol. Biol. Clin. fda.gov/downloads/Drugs/%E2%80%A6/Guidances/ diabetes in obese adults. Arch. Intern. Med. 160,
Investig. 17, 53–61 (2014). ucm071612.pdf (2007). 1321–1326 (2000).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo


©
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
.
REVIEWS

48. Weir, M. A. et al. Orlistat and acute kidney injury: an 69. O’Neil, P. M. et al. Randomized placebo-controlled 91. Saxenda [package insert]. Liraglutide (rDNA) injection
analysis of 953 patients. Arch. Intern. Med. 171, clinical trial of lorcaserin for weight loss in type 2 https://www.accessdata.fda.gov/drugsatfda_docs/
703–704 (2011). diabetes mellitus: the BLOOM‑DM study. Obesity label/2014/206321Orig1s000lbl.pdf (Novo Nordisk,
49. Xenical (Orlistat) package insert https://www.gene. (Silver Spring) 20, 1426–1436 (2012). 2014).
com/download/pdf/xenical_prescribing.pdf 70. Kelly, M. J., Loose, M. D. & Ronnekleiv, O. K. 92. Gallo, M. Thyroid safety in patients treated with
(Genentech, 2016). Opioids hyperpolarize beta-endorphin neurons via liraglutide. J. Endocrinol. Investig. 36, 140–145
50. Derosa, G., Mugellini, A., Ciccarelli, L. & Fogari, R. mu‑receptor activation of a potassium (2013).
Randomized, double-blind, placebo-controlled conductance. Neuroendocrinology 52, 268–275 93. Bjerre Knudsen, L. et al. Glucagon-like peptide‑1
comparison of the action of orlistat, fluvastatin, or (1990). receptor agonists activate rodent thyroid C‑cells
both an anthropometric measurements, blood 71. Greenway, F. L. et al. Rational design of a combination causing calcitonin release and C‑cell proliferation.
pressure, and lipid profile in obese patients with medication for the treatment of obesity. Obesity Endocrinology 151, 1473–1486 (2010).
hypercholesterolemia prescribed a standardized diet. (Silver Spring) 17, 30–39 (2009). 94. Tanaka, T. [Therapeutic gene clusters as drug action
Clin. Ther. 25, 1107–1122 (2003). 72. Apovian, C. M. Naltrexone/bupropion for the mechanisms] [Japanese]. Nihon Yakurigaku Zasshi
51. Antel, J. & Hebebrand, J. Weight-reducing side effects treatment of obesity and obesity with type 2 122 (Suppl.) 5P–7P (2003).
of the antiepileptic agents topiramate and diabetes. Future Cardiol. 12, 129–138 (2016). 95. Maslov, D. L., Balashova, E. E., Lokhov, P. G. &
zonisamide. Handb. Exp. Pharmacol. 209, 433–466 73. Jain, A. K. et al. Bupropion SR versus placebo for Archakov, A. I. [Pharmacometabonomics — the novel
(2012). weight loss in obese patients with depressive way to personalized drug therapy] [Russian]. Biomed.
52. Allison, D. B. et al. Controlled-release phentermine/ symptoms. Obes. Res. 10, 1049–1056 (2002). Khim. 63, 115–123 (2017).
topiramate in severely obese adults: a randomized 74. Greenway, F. L. et al. Effect of naltrexone plus 96. Adkins, D. E. et al. Genotype-based ancestral
controlled trial (EQUIP). Obesity (Silver Spring) 20, bupropion on weight loss in overweight and obese background consistently predicts efficacy and side
330–342 (2012). adults (COR‑I): a multicentre, randomised, double- effects across treatments in CATIE and STAR*D. PLoS
53. Gadde, K. M. et al. Effects of low-dose, controlled- blind, placebo-controlled, phase 3 trial. Lancet 376, ONE 8, e55239 (2013).
release, phentermine plus topiramate combination 595–605 (2010). 97. Wang, D. et al. Large-scale candidate gene study to
on weight and associated comorbidities in overweight 75. Apovian, C. M. et al. A randomized, phase 3 trial of identify genetic risk factors predictive of
and obese adults (CONQUER): a randomised, naltrexone SR/bupropion SR on weight and obesity- paliperidone treatment response in patients with
placebo-controlled, phase 3 trial. Lancet 377, related risk factors (COR‑II). Obesity (Silver Spring) schizophrenia. Pharmacogenet. Genomics 25,
1341–1352 (2011). 21, 935–943 (2013). 173–185 (2015).
54. Garvey, W. T. et al. Two-year sustained weight loss 76. Wadden, T. A. et al. Weight loss with naltrexone SR/ 98. Brauch, H., Murdter, T. E., Eichelbaum, M. &
and metabolic benefits with controlled-release bupropion SR combination therapy as an adjunct to Schwab, M. Pharmacogenomics of tamoxifen therapy.
phentermine/topiramate in obese and overweight behavior modification: the COR-BMOD trial. Obesity Clin. Chem. 55, 1770–1782 (2009).
adults (SEQUEL): a randomized, placebo-controlled, (Silver Spring) 19, 110–120 (2011). 99. Blue Cross Blue Shield Association, Kaiser Foundation
phase 3 extension study. Am. J. Clin. Nutr. 95, 77. Hollander, P. et al. Effects of naltrexone sustained- Health Plan & Southern California Permanente
297–308 (2012). release/bupropion sustained-release combination Medical Group. CYP2D6 pharmacogenomics of
55. Roberts, M. D. Clinical briefing document: therapy on body weight and glycemic parameters in tamoxifen treatment. Technol. Eval. Cent. Assess.
Endocrinologic and Metabolic Drugs Advisory overweight and obese patients with type 2 diabetes. Program Exec. Summ. 28, 1–4 (2014).
Committee Meeting (February 22, 2012). New Drug Diabetes Care 36, 4022–4029 (2013). 100. Zembutsu, H. Pharmacogenomics toward personalized
Application 22580: VI‑0521 Qnexa (phentermine/ 78. Contrave package insert https://www.accessdata. tamoxifen therapy for breast cancer.
topiramate). Sponsor: Vivus. FDA fda.gov/drugsatfda_docs/ Pharmacogenomics 16, 287–296 (2015).
https://wayback.archive-it. label/2014/200063s000lbl.pdf (Takeda 101. Kiyotani, K., Mushiroda, T., Nakamura, Y. &
org/7993/20170405220351/https://www.fda.gov/ Pharmaceuticals, 2014). Zembutsu, H. Pharmacogenomics of tamoxifen: roles
downloads/AdvisoryCommittees/ 79. McElroy, S. L., Guerdjikova, A. I., Mori, N., of drug metabolizing enzymes and transporters. Drug
CommitteesMeetingMaterials/Drugs/ Munoz, M. R. & Keck, P. E. Overview of the treatment Metab. Pharmacokinet. 27, 122–131 (2012).
EndocrinologicandMetabolicDrugsAdvisoryCommittee/ of binge eating disorder. CNS Spectr. 20, 546–556 102. Hendrick, V., Dasher, R., Gitlin, M. & Parsi, M.
UCM292315.pdf (2017). (2015). Minimizing weight gain for patients taking
56. Mines, D. et al. Topiramate use in pregnancy and the 80. White, M. A. & Grilo, C. M. Bupropion for overweight antipsychotic medications: the potential role for early
birth prevalence of oral clefts. Pharmacoepidemiol. women with binge-eating disorder: a randomized, use of metformin. Ann. Clin. Psychiatry 29, 120–124
Drug Saf. 23, 1017–1025 (2014). double-blind, placebo-controlled trial. J. Clin. (2017).
57. Bialer, M. et al. Pharmacokinetic interactions of Psychiatry 74, 400–406 (2013). 103. Wu, R. R. et al. Lifestyle intervention and metformin
topiramate. Clin. Pharmacokinet. 43, 763–780 81. Yeomans, M. R. & Gray, R. W. Opioid peptides and the for treatment of antipsychotic-induced weight gain: a
(2004). control of human ingestive behaviour. Neurosci. randomized controlled trial. JAMA 299, 185–193
58. Qsymia (phentermine and topiramate extended- Biobehav. Rev. 26, 713–728 (2002). (2008).
release) package insert https://www.qsymia.com/pdf/ 82. Calderone, A. et al. Psychopathological behaviour 104. Anagnostou, E. et al. Metformin for treatment of
prescribing-information.pdf (Vivus, 2014). and cognition in morbid obesity. Recent Pat. overweight induced by atypical antipsychotic
59. Grilo, C. M., Reas, D. L. & Mitchell, J. E. Combining Endocr. Metab. Immune Drug Discov. 10, 112–118 medication in young people with autism spectrum
pharmacological and psychological treatments for (2016). disorder: a randomized clinical trial. JAMA Psychiatry
binge eating disorder: current status, limitations, and 83. Astrup, A. et al. Effects of liraglutide in the 73, 928–937 (2016).
future directions. Curr. Psychiatry Rep. 18, 55 treatment of obesity: a randomised, double-blind, 105. Dansinger, M. L., Gleason, J. A., Griffith, J. L.,
(2016). placebo-controlled study. Lancet 374, 1606–1616 Selker, H. P. & Schaefer, E. J. Comparison of the
60. Stanford, F. C. et al. The utility of weight loss (2009). Atkins, Ornish, Weight Watchers, and Zone diets for
medications after bariatric surgery for weight regain 84. Astrup, A. et al. Safety, tolerability and sustained weight loss and heart disease risk reduction: a
or inadequate weight loss: a multi-center study. weight loss over 2 years with the once-daily human randomized trial. JAMA 293, 43–53 (2005).
Surg. Obes. Relat. Dis. 13, 491–500 (2017). GLP‑1 analog, liraglutide. Int. J. Obes. 36, 843–854 106. Hatoum, I. J. & Kaplan, L. M. Advantages of percent
61. Topamax (topiramate) package insert https://www. (2012). weight loss as a method of reporting weight loss after
accessdata.fda.gov/drugsatfda_docs/ 85. Pi‑Sunyer, X. et al. A randomized, controlled trial of Roux‑en‑Y gastric bypass. Obesity (Silver Spring) 21,
label/2012/020844s041lbl.pdf (Janssen 3.0 mg of Liraglutide in weight management. N. Engl. 1519–1525 (2013).
Pharmaceuticals, 2009). J. Med. 373, 11–22 (2015).
62. Ojemann, L. M. et al. Language disturbances as side 86. Davies, M. J. et al. Efficacy of liraglutide for weight Acknowledgements
effects of topiramate and zonisamide therapy. loss among patients with type 2 diabetes: the SCALE The authors thank A. McCarthy for editorial and administra-
Epilepsy Behav. 2, 579–584 (2001). Diabetes randomized clinical trial. JAMA 314, tive assistance.
63. Belviq (lorcaserin HCl) package insert https://www. 687–699 (2015).
accessdata.fda.gov/drugsatfda_docs/ 87. Wadden, T. A. et al. Weight maintenance and Author contributions
label/2012/022529lbl.pdf (Arena Pharmaceuticals, additional weight loss with liraglutide after low-calorie- G.S. and C.M.A. researched data for the article, made sub-
2012). diet-induced weight loss: the SCALE Maintenance stantial contributions to discussions of the content, wrote the
64. Thomsen, W. J. et al. Lorcaserin, a novel selective randomized study. Int. J. Obes. 37, 1443–1451 article and reviewed and/or edited the article before
human 5‑hydroxytryptamine2C agonist: in vitro and (2013). submission.
in vivo pharmacological characterization. 88. le Roux, C. W. et al. 3 years of liraglutide versus
J. Pharmacol. Exp. Ther. 325, 577–587 (2008). placebo for type 2 diabetes risk reduction and Competing interests statement
65. Lam, D. D., Garfield, A. S., Marston, O. J., Shaw, J. & weight management in individuals with prediabetes: G.S. declares no competing interests. C.M.A. has received
Heisler, L. K. Brain serotonin system in the a randomised, double-blind trial. Lancet 389, personal fees from Gelesis, GI Dynamics, Johnson and
coordination of food intake and body weight. 1399–1409 (2017). Johnson, Merck, NovoNordisk, Nutrisystem, Orexigen,
Pharmacol. Biochem. Behav. 97, 84–91 (2010). 89. Chalmer, T., Almdal, T. P., Vilsboll, T. & Knop, F. K. Sanofi-Aventis, Scientific Intake, Takeda and Zafgen; grants
66. Blundell, J. E., Lawton, C. L. & Halford, J. C. Adverse drug reactions associated with the use of from Aspire Bariatrics, the Robert C. and Veronica Atkins
Serotonin, eating behavior, and fat intake. Obes. Res. liraglutide in patients with type 2 diabetes — focus on Foundation, Coherence Lab, Energesis, Gelesis, GI Dynamics,
3 (Suppl. 4), 471S–476S (1995). pancreatitis and pancreas cancer. Exp. Opin. Drug Myos, Orexigen, PCORI, Takeda and Vela Foundation; and
67. Fidler, M. C. et al. A one-year randomized trial of Saf. 14, 171–180 (2015). other forms of support from Science-Smart. No grant support
lorcaserin for weight loss in obese and overweight 90. Funch, D., Gydesen, H., Tornoe, K., Major- or external funding was provided to G.S. or C.M.A. for the
adults: the BLOSSOM trial. J. Clin. Endocrinol. Metab. Pedersen, A. & Chan, K. A. A prospective, claims- drafting of this manuscript.
96, 3067–3077 (2011). based assessment of the risk of pancreatitis and
68. Smith, S. R. et al. Multicenter, placebo-controlled trial pancreatic cancer with liraglutide compared to other Publisher’s note
of lorcaserin for weight management. N. Engl. J. Med. antidiabetic drugs. Diabetes Obes. Metab. 16, Springer Nature remains neutral with regard to jurisdictional
363, 245–256 (2010). 273–275 (2014). claims in published maps and institutional affiliations.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 13


©
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