Sei sulla pagina 1di 20

Drugs (2015) 75:777–796

DOI 10.1007/s40265-015-0385-y

ADIS DRUG EVALUATION

Alogliptin: A Review of Its Use in Patients with Type 2 Diabetes


Mellitus
Gillian M. Keating1

Published online: 9 April 2015


Ó Springer International Publishing Switzerland 2015

Abstract The dipeptidyl peptidase-4 inhibitor alogliptin


(NesinaÒ, VipidiaÒ) is approved in numerous countries Alogliptin in type 2 diabetes mellitus: a summary
worldwide for the treatment of type 2 diabetes mellitus.
Fixed-dose combinations of alogliptin/metformin (KazanoÒ, Oral dipeptidyl peptidase-4 inhibitor with a glucose-
VipdometÒ) and alogliptin/pioglitazone (OseniÒ, In- dependent mechanism of action
cresyncÒ) are also available. This article reviews the clinical
efficacy and tolerability of oral alogliptin in the treatment of Improves glycaemic control as monotherapy; as dual
type 2 diabetes. Results of randomized controlled trials therapy in combination with metformin,
demonstrated that oral alogliptin improved glycaemic control pioglitazone, a sulfonylurea, voglibose or insulin; or
when administered as monotherapy, as dual therapy in as triple therapy in combination with metformin and
combination with metformin, pioglitazone, a sulfonylurea, pioglitazone
voglibose or insulin, or as triple therapy in combination with Generally well tolerated and weight neutral, with a
metformin plus pioglitazone. Alogliptin was generally well low risk of hypoglycaemia
tolerated in patients with type 2 diabetes and was weight
No increase in the risk of major cardiovascular
neutral, with a low risk of hypoglycaemia. Results of the
events
large, well designed EXAMINE trial revealed that alogliptin
was not associated with an increased risk of major cardio-
vascular events in patients with type 2 diabetes and recent
acute coronary syndrome. In conclusion, alogliptin is a useful
option for the treatment of patients with type 2 diabetes. 1 Introduction

Type 2 diabetes mellitus places a substantial and growing


The manuscript was reviewed by: I. Bourdel-Marchasson, burden on healthcare systems and society. In 2010, the
Department of Gerontology-Centre Hospitalier Universitaire de global prevalence of type 2 diabetes was estimated to be
Bordeaux, Bordeaux, France; K. Kaku, Division of Diabetes,
Endocrinology and Metabolism, Kawasaki Medical School,
6.4 % (i.e. affecting 285 million adults globally); this is
Okayama, Japan; P. Marchetti, Department of Clinical and predicted to increase to 7.7 % by 2030 (i.e. affecting 439
Experimental Medicine, University of Pisa, Cisanello AOUP million adults globally) [1].
University Hospital, Pisa, Italy; J. Neumiller, College of Pharmacy, The incretin hormones glucagon-like peptide-1 (GLP-1)
Washington State University, Spokane, WA, USA; M. Rendell,
School of Medicine, Creighton University, and Association of
and glucose-dependent insulinotropic peptide (GIP) are
Diabetes Investigators, Creighton Diabetes Center, Omaha, NB, USA. released from the gut in response to food intake [2, 3]. Both
incretin hormones enhance insulin secretion in a glucose-
& Gillian M. Keating dependent manner, and GLP-1 also inhibits glucagon se-
demail@springer.com
cretion [2, 3]. Following release, both GLP-1 and GIP are
1
Springer, Private Bag 65901, Mairangi Bay 0754, Auckland, rapidly inactivated by dipeptidyl peptidase (DPP)-4 [2, 3].
New Zealand Given that the incretin effect is impaired in patients with
778 G. M. Keating

type 2 diabetes [2, 3], incretin-based agents (i.e. DPP-4 23]. For example, in terms of fasting markers, improve-
inhibitors and GLP-1 receptor agonists) represent a rational ments in the homeostasis model assessment of b-cell
approach to treatment. DPP-4 inhibitors prevent the function (HOMA-b) [12, 17–19, 21–23] and the proin-
degradation of incretin hormones by DPP-4, and GLP-1 sulin:insulin ratio [17–22] were seen with alogliptin alone
receptor agonists are resistant to cleavage by DPP-4 [3]. [12, 23], alogliptin plus metformin [18–20], alogliptin plus
The DPP-4 inhibitor alogliptin (NesinaÒ, VipidiaÒ) is pioglitazone [21], alogliptin plus glimepiride [17], and
approved in numerous countries worldwide, including the alogliptin plus voglibose [22]. Compared with placebo, the
US [4] and the EU [5], for the treatment of type 2 diabetes. fasting insulin secretion rate at a fixed glucose level was
Fixed-dose combinations of alogliptin/metformin significantly (p \ 0.001) increased with alogliptin plus
(KazanoÒ, VipdometÒ) [6, 7] and alogliptin/pioglitazone pioglitazone, but not with alogliptin alone; the difference
(OseniÒ, IncresyncÒ) [8, 9] are also available. This article between alogliptin plus pioglitazone recipients and
reviews the clinical efficacy and tolerability of alogliptin in alogliptin recipients was also significant (p \ 0.05) [16].
the treatment of type 2 diabetes, as well as summarizing its In some studies, alogliptin also reduced postprandial
pharmacological properties. glucagon levels in patients with type 2 diabetes who re-
ceived alogliptin alone [24], alogliptin plus pioglitazone
[24], alogliptin plus glimepiride [17] or alogliptin plus
2 Pharmacodynamic Properties voglibose [22].
In patients with type 2 diabetes receiving alogliptin 25,
2.1 Mechanism of Action 100 or 400 mg once daily for 14 days, postprandial glucose
(PPG) levels 4 h after breakfast, lunch and dinner were
Alogliptin forms noncovalent bonds with the catalytic site reduced to a significantly (p \ 0.05) greater extent than
of DPP-4, and is a selective, reversible inhibitor of this with placebo [13]. After 16 weeks’ therapy, PPG levels
enzyme [2, 10]. In animal studies, the half maximal in- were reduced to a significantly (p \ 0.01) greater extent
hibitory concentration (IC50) of alogliptin was 7 nmol/L with once-daily alogliptin 25 mg or alogliptin 25 mg plus
for DPP-4, compared with IC50 values of[100,000 nmol/L pioglitazone 30 mg than with placebo; the reduction
for DPP-2, DPP-8 and DPP-9 (i.e. [10,000-fold greater achieved with alogliptin plus pioglitazone was significantly
selectivity for DPP-4) [11]. (p \ 0.001) greater than with alogliptin monotherapy [16].
Alogliptin inhibited DPP-4 in a dose-dependent manner The effects of alogliptin on PPG levels in large clinical
[12, 13]. For example, mean DPP-4 inhibition at week 12 trials primarily designed to assess glycaemic control are
in patients with type 2 diabetes receiving alogliptin 6.25, discussed in Sect. 4.
12.5, 25 and 50 mg once daily was 67.8, 73.9, 81.3 and Enhancement of b-cell proliferation may also contribute
87.8 %, respectively [12]. Administration of alogliptin to the beneficial effect of alogliptin on b-cell function. A
25 mg once daily to patients with type 2 diabetes resulted tenfold increase in b-cell proliferation was seen in diabetic
in maximum inhibition of plasma DPP-4 activity of 94.7 % immunodeficient mice engrafted with human pancreatic
on day 1 and 93.8 % on day 14 [13]. Mean inhibition of islets administered alogliptin versus controls (average
plasma DPP-4 activity 24, 72 and 168 h after the final dose proliferation of 0.58 vs. 0.056 %) [25]. In another study,
on day 14 was 81.8, 66.3 and 20.5 %, respectively [13]. alogliptin restored b-cell structure and function in a mouse
Exposure to active GLP-1 in plasma was 2- to 6-fold model of diabetes [26].
higher following administration of alogliptin versus
placebo to healthy volunteers [14, 15], and changes from 2.3 Other Effects
baseline in the mean area under the concentration-time
curve from time zero to 2 h for active GLP-1 were sig- In most studies, no clinically significant changes in fasting
nificantly greater with alogliptin 6.25–50 mg once daily serum lipid levels were reported in patients with type 2
than with placebo in patients with type 2 diabetes (?4.23 to diabetes receiving alogliptin [12, 17, 20, 21, 27–29],
?9.04 vs. -1.50 pmolh/L) [12]. although some studies did report changes in lipid levels
favouring alogliptin [22, 30]. For example, significantly
2.2 Effects on b-Cell Function, Glucagon Secretion (p \ 0.05) greater reductions in fasting serum total
and Post-Prandial Glucose cholesterol and low-density lipoprotein-cholesterol levels
were seen with alogliptin 12.5 or 25 mg once daily plus
The increase in GLP-1 levels associated with alogliptin voglibose than with voglibose alone [22] and a sig-
enhanced glucose-dependent insulin secretion. Fasting and nificantly (p \ 0.05) greater reduction in fasting total
postprandial markers of b-cell function improved in pa- cholesterol levels was seen with alogliptin plus gliben-
tients with type 2 diabetes who received alogliptin [12, 16– clamide (glyburide) than with glibenclamide alone [30].
Alogliptin: A Review 779

Postprandial endothelial dysfunction was significantly 3.2 Metabolism and Excretion


(p = 0.03 vs. controls) improved by alogliptin 25 mg once
daily in healthy volunteers [31], and coronary flow reserve Alogliptin was not extensively metabolized, with 60–70 %
increased from baseline to a significantly (p = 0.006) of the dose excreted as unchanged drug in the urine [13–
greater extent with alogliptin than with glimepiride in pa- 15]. The minor metabolites M-I (N-demethylated
tients with type 2 diabetes and known or suspected coro- alogliptin) and M-II (N-acetylated alogliptin) were detected
nary artery disease [32]. The corrected QT interval was not following administration of radiolabelled alogliptin; M-I
increased in subjects receiving supratherapeutic dosages of and M-II accounted for \1 % and \6 % of the parent
alogliptin 50 or 400 mg once daily for 7 days [4]. compound [4, 5]. The active metabolite M-I was a highly
In patients with type 2 diabetes (12 postmenopausal selective inhibitor of DPP-4, whereas M-II had no in-
women and 8 men) who received alogliptin 25 mg once hibitory activity [4, 5, 36]. The cytochrome P450 (CYP)
daily for 16 weeks, serum bone alkaline phosphatase levels enzymes CYP2D6 and CYP3A4 contributed to the meta-
significantly decreased from baseline and calcitonin levels bolism of alogliptin [5].
were significantly increased (p-values not stated) [33]. Following administration of radiolabelled alogliptin, 76
However, no significant changes from baseline were seen and 13 % of the total radioactivity was recovered in the
in levels of GIP or biomarkers of bone turnover (e.g. os- urine and faeces [4, 5]. In patients with type 2 diabetes
teocalcin and serum N-terminal telopeptide) or in bone receiving alogliptin 25 mg once daily, alogliptin had a
mineral density at the lumbar vertebrae or proximal femur mean renal clearance of 175 mL/min, suggesting some
[33]. active renal tubular excretion and a mean terminal
Coadministration of alogliptin and warfarin did not alter elimination half-life of 21.1 h [4, 5, 13].
the prothrombin time or the international normalized ratio
[34]. 3.3 Special Patient Populations

The exposure of alogliptin following administration of a


3 Pharmacokinetic Properties single 50 mg dose was &1.7- to 3.8-fold greater in patients
with mild, moderate or severe renal impairment or end-
3.1 Absorption and Distribution stage renal disease than in healthy subjects [37]. The
alogliptin dosage does not need to be adjusted in patients
Alogliptin has an absolute bioavailability of &100 % [4, with mild renal impairment [creatinine clearance (CLCR) of
5]. Alogliptin was rapidly absorbed following adminis- [50 to B80 mL/min [5] or C60 mL/min [4]], but should
tration of single oral doses of 25–800 mg to healthy be reduced to 12.5 mg once daily in patients with moderate
volunteers, with the maximum plasma concentration renal impairment (CLCR of C30 to B50 mL/min [5] or C30
(Cmax) reached in a median 1–2 h [15]. In patients with to \60 mL/min [4]) and to 6.25 mg once daily in patients
type 2 diabetes receiving alogliptin 25 mg once daily for with severe renal impairment (CLCR of \30 mL/min [5,
14 days, a mean Cmax of 153 ng/mL was reached in a 36] or CLCR of C15 to \30 mL/min [4]) or end-stage renal
median 1.1 h [13]. The mean area under the plasma disease.
concentration-time curve from time zero to 24 h was No clinically significant changes in alogliptin exposure
1474 ngh/mL [13]. were seen in patients with moderate hepatic impairment
No clinically relevant accumulation was seen with re- who received a single 25 mg dose [38]. The EU summary
peated administration of alogliptin to healthy volunteers or of product characteristics (SPC) states that the alogliptin
patients with type 2 diabetes [5]. The exposure of alogliptin dosage does not need to be adjusted in patients with mild to
was not altered to a clinically relevant extent when it was moderate hepatic impairment [5]. Alogliptin is not rec-
administered with a high-fat meal [35]; alogliptin can be ommended for use in patients with severe hepatic impair-
administered with or without food [4, 5]. ment because of limited data in this patient group [5]. The
The fixed-dose combinations of alogliptin/metformin [6, US prescribing information recommends caution when
7] and alogliptin/pioglitazone [8, 9] were shown to be administering alogliptin to patients with hepatic impair-
bioequivalent to the corresponding doses of alogliptin and ment [4].
metformin or pioglitazone coadministered as separate The pharmacokinetics of alogliptin were not altered to a
tablets. clinically significant extent in elderly patients aged
Alogliptin had a volume of distribution of 417 L during 65–85 years [39]. No dosage adjustment is needed in
the terminal phase following administration of a single elderly patients receiving alogliptin [4, 5], although dosing
intravenous dose of 12.5 mg to healthy volunteers [4, 5]. should be conservative given the potential for decreased
Plasma protein binding of alogliptin was 20–30 % [5]. renal function in the elderly [5]. The pharmacokinetics of
780 G. M. Keating

alogliptin were not altered to a clinically significant extent multicentre trials of 12–26 weeks’ duration [12, 27, 46,
by gender or race [39], including in Japanese patients [14]. 47]. One study included additional treatment arms exam-
ining the addition of alogliptin to metformin (see
3.4 Potential Drug Interactions Sect. 4.2.1.1) or pioglitazone (see Sect. 4.2.2) [47]. Pa-
tients in these studies had inadequately controlled type 2
This section provides an overview of drug interactions diabetes and were not currently receiving antidiabetic
potentially associated with alogliptin. Data were obtained therapy [12, 27, 46, 47] or were receiving metformin and/or
from studies in healthy volunteers [34, 35, 40–45], sup- a sulfonylurea [27]. Patients not currently receiving an-
plemented by data from the US prescribing information [4] tidiabetic therapy were newly diagnosed [27] or were
and the EU SPC [5]. Where specified, alogliptin was ad- inadequately controlled by diet and exercise [12, 27, 46,
ministered at a dosage of 25 mg [34, 40–45] or 100 mg 47]. Where specified, the mean duration of diabetes was 4.1
[35] once daily. [47] or 7.9 years [12]. Efficacy was assessed in the full
Alogliptin has a low potential for drug interactions. analysis set (n = 480 [12] and 185 [47]) or intent-to-treat
In vitro, alogliptin did not induce CYP1A2, CYP2B6 or population (n = 265 [27] and 329 [46]). Two studies are
CYP2C9 and did not inhibit CYP1A2, CYP2B6, CYP2C8, available as abstracts [27, 47], supplemented by informa-
CYP2C9, CYP2C19, CYP2D6 or CYP3A4 [4, 5]. Mild tion from ClinicalTrials.gov (NCT00755846 and
induction of CYP3A4 was seen with alogliptin in vitro, but NCT01289119).
not in in vivo studies [5]. In vitro, alogliptin was not a Alogliptin monotherapy improved glycaemic control in
substrate for, nor an inhibitor of, organic anion transporter patients with type 2 diabetes. Compared with placebo,
(OAT)-1, OAT-2 or organic cation transporter-2 [5]. significantly greater reductions from baseline in HbA1c
Clinical data suggest no interaction between alogliptin and were seen at week 12 with alogliptin dosages of
P-glycoprotein inhibitors or substrates [5]. 6.25–50 mg once daily in a dose-ranging study in patients
Interactions between alogliptin and CYP inhibitors are inadequately controlled by diet and exercise (mean changes
not expected [5]. Indeed, gemfibrozil (a CYP2C8/CYP2C9 of -0.51 to -0.82 vs. ?0.06 %; p \ 0.0001) [12], and
inhibitor), fluconazole (a CYP2C9 inhibitor) and keto- with alogliptin dosages of 12.5–100 mg once daily in a
conazole (a CYP3A4 inhibitor) did not alter the pharma- dose-ranging study in patients who were newly diagnosed
cokinetics of alogliptin to a clinically relevant extent [40]. or inadequately controlled by diet and exercise or by
Pioglitazone [41], metformin [35], voglibose [5], ci- metformin and/or a sulfonylurea (least squares mean
closporin [44], digoxin [42], cimetidine [35] and atorva- changes of -0.44 to -0.56 vs. -0.01 %; p \ 0.05) [27].
statin [43] did not alter the pharmacokinetics of alogliptin Other studies also demonstrated significantly (p \ 0.001)
to a clinically relevant extent. Alogliptin did not have a greater mean reductions from baseline in HbA1c with
clinically relevant effect on the pharmacokinetics of pi- alogliptin 25 mg once daily than with placebo after 16
oglitazone [41], metformin [35], glibenclamide [41], caf- (-0.99 vs. -0.42 %) [47] or 26 (-0.59 vs. -0.02 %) [46]
feine [4, 5], (R)- or (S)-warfarin [34], tolbutamide [4, 5], weeks’ therapy. Significantly (p \ 0.01 where stated) more
dextromethorphan [4, 5], atorvastatin [43], midazolam [4, alogliptin than placebo recipients achieved an HbA1c target
5], ethinylestradiol/norethisterone [45], digoxin [42], fex- of B7.0 % (44.3 vs. 23.4 % [46] and 63.3 vs. 30.0 % [47])
ofenadine [4, 5] or cimetidine [35]. or \6.9 % (29.1–44.3 vs. 8.0 %) [12], with significantly
more alogliptin than placebo recipients achieving an HbA1c
target of B6.5 % in one study (36.7 vs. 12.2 %; p \ 0.001)
4 Therapeutic Efficacy [47], but not in another study (20.6 vs. 10.9 %) [46].
Mean fasting plasma glucose (FPG) levels were reduced
This section mainly discusses studies designed to examine to a significantly (p \ 0.05) greater extent with once-daily
the effects of alogliptin on glycaemic control. In most alogliptin 6.25–50 mg (-0.52 to -1.25 vs. ?0.31 mmol/
studies, the primary endpoint was the change from baseline L) [12], 25–100 mg (-0.89 to -1.50 vs. ?0.47 mmol/L)
in glycated haemoglobin (HbA1c) [12, 17–22, 27–30, 46– [27] or 25 mg (-0.91 vs. ?0.63 mmol/L) [46] than with
53]. Several studies were conducted solely in Asian placebo. In addition, hyperglycaemic rescue was required
populations (e.g. studies were conducted in Japan [12, 17, by significantly (p B 0.001) fewer alogliptin 12.5 or 25 mg
18, 21, 22, 53] or in China, Taiwan and Hong Kong [47]). once daily than placebo recipients (9.8 and 7.6 vs. 29.7 %
of patients) [46].
4.1 Monotherapy During the 40-week extension phase of one of the
12-week dose-ranging studies (during which placebo re-
The efficacy of monotherapy with alogliptin in type 2 cipients switched to alogliptin 6.25–50 mg once daily),
diabetes was examined in four randomized, double-blind, HbA1c and FPG were significantly reduced from baseline
Alogliptin: A Review 781

with alogliptin 6.25–50 mg once daily at all time-points up baseline with alogliptin 12.5 or 25 mg once daily plus
to 52 weeks (p-values not stated) [12]. metformin at all time-points up to 52 weeks (p-values not
One of the 12-week dose-ranging studies also demon- stated) [18]. With alogliptin 12.5 or 25 mg once daily plus
strated that mean HbA1c was reduced to a significantly metformin, mean reductions from baseline in HbA1c were
greater extent with alogliptin 6.25–50 mg once daily than 0.44 and 0.58 % and in FPG were 0.91 and 0.98 mmol/L
with voglibose 0.2 mg three times daily (-0.51 to -0.82 [18].
vs. -0.16 %; p-values not stated) [12]. In addition, sig- In drug-naive patients with type 2 diabetes inadequately
nificantly more recipients of alogliptin 12.5–50 mg once controlled with diet and exercise (HbA1c of 7.5–10 %),
daily than voglibose achieved an HbA1c target of \6.9 % significantly greater reductions from baseline in HbA1c and
(35.7–44.3 vs. 19.3 %) and mean FPG was reduced to a FPG were seen with alogliptin 12.5 mg twice daily plus
significantly greater extent with alogliptin 12.5–50 mg metformin 500 or 1000 mg twice daily than with alogliptin
once daily than with voglibose (-0.81 to -1.25 vs. 12.5 mg twice daily alone or metformin 500 or 1000 mg
-0.17 mmol/L) [p-values not stated] [12]. twice daily alone [19] (Table 1).
In addition, significantly (p \ 0.05) greater least
4.2 Combination Therapy squares mean reductions from in baseline in 2-h PPG
levels were seen with alogliptin 12.5 mg twice daily plus
4.2.1 In Combination with Metformin metformin 1000 mg twice daily than with alogliptin
12.5 mg twice daily alone or metformin 1000 mg twice
Five randomized, double-blind, multicentre trials com- daily alone (-4.75 vs. -2.38 and -2.99 mmol/L), with
pared the efficacy of alogliptin plus metformin with no other significant between-group differences seen [19].
alogliptin or metformin alone [18–20, 47] or with glipizide Hyperglycaemic rescue was required in significantly
plus metformin [52] in patients with type 2 diabetes (p \ 0.05) fewer patients receiving alogliptin 12.5 mg
inadequately controlled by metformin [18, 20, 47, 52] or by twice daily plus metformin 500 mg twice daily than
diet and exercise [19]. The mean duration of diabetes alogliptin 12.5 mg twice daily alone or metformin
ranged from 4.0 to 6.3 years [18–20, 47, 52]. One study is 500 mg twice daily alone (12.3 vs. 17.3 and 22.9 %) or
available as an abstract [47], supplemented by information alogliptin 12.5 mg twice daily plus metformin 1000 mg
from ClinicalTrials.gov (NCT01289119). twice daily than alogliptin 12.5 mg twice daily alone or
metformin 1000 mg twice daily alone (2.6 vs. 17.3 and
4.2.1.1 Comparisons with Alogliptin or Metformin 10.8 %). In general, recipients of alogliptin plus met-
Alone Combination therapy with alogliptin plus met- formin were significantly more likely than recipients of
formin improved glycaemic control to a significantly alogliptin or metformin alone to achieve an HbA1c target
greater extent than metformin alone in patients with type 2 of \7.0 or \6.5 % [19] (Table 1).
diabetes inadequately controlled with metformin [18, 20,
47]. After 12–26 weeks’ therapy, HbA1c was reduced from 4.2.1.2 Comparisons with Glipizide plus Metformin The
baseline to significantly greater extent with alogliptin efficacy of alogliptin plus metformin was maintained over
25 mg once daily plus metformin than with metformin 2 years in patients with inadequately controlled type 2
alone [18, 20, 47] (Table 1). Patients receiving alogliptin diabetes [52]. Alogliptin 25 mg once daily plus metformin
25 mg once daily plus metformin were also significantly was noninferior to glipizide 5–20 mg/day plus metformin
more likely than those receiving metformin alone to in terms of the reduction from baseline in HbA1c after
achieve an HbA1c target of B7.0 [20, 47], \6.9 [18] or 104 weeks’ therapy in patients with type 2 diabetes
B6.5 % [20, 47] (Table 1). inadequately controlled with metformin, with superiority
FPG was reduced to a significantly greater extent with subsequently shown (Table 1). Significantly more patients
alogliptin 25 mg once daily plus metformin than with receiving alogliptin 25 mg once daily plus metformin than
metformin alone [18, 20] (Table 1). In addition, patients glipizide plus metformin achieved an HbA1c target of
receiving alogliptin 12.5 or 25 mg once daily plus met- B6.5 % or B7.0 % after 104 weeks’ therapy (Table 1)
formin were significantly (p \ 0.01) less likely than pa- [52].
tients receiving metformin alone to experience marked FPG was reduced from baseline to a significantly greater
hyperglycaemia (29 and 31 vs. 51 % of patients) or to extent with alogliptin 25 mg once daily plus metformin
require hyperglycaemic rescue (9 and 8 vs. 24 %) [20]. than with glipizide plus metformin after 104 weeks’ ther-
During the 40-week extension phase of the 12-week apy (Table 1) [52]. Hyperglycaemic rescue was required in
study (during which recipients of metformin alone significantly fewer patients receiving alogliptin 25 mg
switched to alogliptin 12.5 or 25 mg once daily plus met- once daily plus metformin than glipizide plus metformin
formin), HbA1c and FPG were significantly reduced from (23.2 vs. 27.4 %; p = 0.03) [52].
782 G. M. Keating

Table 1 Efficacy of alogliptin plus metformin in patients with type 2 diabetes inadequately controlled with metformin alone [18, 20, 47, 52] or
diet and exercise [19]. Results of trials of 12 [18], 16 [47], 26 [19, 20] or 104 [52] weeks’ duration
Study Treatmenta No. of LSM change from LSM change from HbA1c target (% of pts)
ptsb baseline in HbA1cc baseline in FPG (mmol/L)
(%) [baseline [baseline value] B7.0 or \6.9 %d B6.5 %
value]

Comparisons with ALO or MET alone


Ji et al. [47]e ALO 25 od ? MET 98 -0.91*** -1.24f 55.1*** 21.4***
MET 97 -0.22 -0.51f 25.8 4.1
Nauck et al. [20] ALO 12.5 od ? MET 213 -0.6*** [7.9] -1.0*** [9.3] 52*** 20*
ALO 25 od ? MET 210 -0.6*** [7.9] -1.0*** [9.5] 44*** 17*
MET 104 -0.1 [8.0] 0 [10.0] 18 4
Pratley et al. [19] ALO 12.5 bid ? MET 500 bid 111 -1.22***   -1.76*  47.1**   20.6**
ALO 12.5 bid ? MET 1000 bid 114 -1.55***   -2.55*  59.5***   32.4*  
ALO 12.5 bid 113 -0.56 -0.54 20.2 13.5
ALO 25 od 112 -0.52 -0.3 20.2 6.7
MET 500 bid 114 -0.65 -0.64 27.2 6.8
MET 1000 bid 111 -1.11 -1.77 34.3 18.5
PL 109 ?0.15 ?0.7 3.9 1.0
Seino et al. [18] ALO 12.5 od ? MET 92 -0.55**** [7.89] -1.05g 28.3g
ALO 25 od ? MET 96 -0.64**** [8.02] -1.28g 27.1g
MET 100 ?0.22 [8.00] -0.04 2.0
Comparison with GPZ 1 MET
Del Prato et al. [52] ALO 12.5 od ? MET 371 -0.68h [7.6] -0.05ààà 45.6 23.5à
ààà àà
ALO 25 od ? MET 382 h
-0.72 [7.6] -0.18 48.5 24.1àà
GPZ 5–20 daily ? MET 336 -0.59 [7.6] ?0.30 42.8 19.0
ALO alogliptin, bid twice daily, FPG fasting plasma glucose, GPZ glipizide, HbA1c glycated haemoglobin, LSM least squares mean, MET
metformin, od once daily, PL placebo, pts patients
     à àà
* p \ 0.05, ** p \ 0.01, *** p \ 0.001, **** p \ 0.0001 vs. MET; p \ 0.05, p \ 0.001 vs. ALO 12.5 bid; p \ 0.05, p \ 0.01,
ààà
p \ 0.001 vs. GPZ ? MET
a
Pts received a stable MET dosage [18, 20, 47] (500–750 mg/day [18], C1000 mg/day [47] or C1500 mg/day [20]) or MET C1500 mg od or
the maximum tolerated dosage [52]
b
No. of patients in the full analysis set [18–20, 47] or the per-protocol population [52]
c
Primary endpoint
d
HbA1c target of B7.0 % [19, 20, 47, 52] or \6.9 % [18]
e
Available as an abstract, supplemented by data from ClinicalTrials.gov (NCT01289119). This study also included treatment arms in which pts
received ALO vs. PL (see Sect. 4.1 for results) or ALO ? PIO vs. PIO (see Table 2 for results)
f
Statistical analysis not reported
g
Significantly different vs. MET alone (p-value not stated)
h
Noninferiority was shown between ALO 12.5 od ? MET vs. GPZ ? MET and between ALO 25 od ? MET vs. GPZ ? MET, with supe-
riority also shown for ALO 25 od ? MET vs. GPZ ? MET

4.2.2 In Combination with Pioglitazone The mean duration of diabetes ranged from &3 to
7.6 years [21, 28, 47, 48].
Four randomized, double-blind, multicentre trials exam- Combination therapy with alogliptin plus pioglitazone
ined the efficacy of alogliptin in combination with piogli- significantly improved glycaemic control in patients with
tazone in patients with inadequately controlled type 2 type 2 diabetes inadequately controlled by a thiazolidine-
diabetes [21, 28, 47, 48]. Patients were inadequately con- dione with or without other oral antidiabetic drugs (OADs)
trolled despite diet and exercise (drug-naive patients) [48] [21, 28, 47]. After 12–26 weeks’ therapy, HbA1c was re-
or despite treatment with pioglitazone alone [21], piogli- duced from baseline to a significantly greater extent with
tazone with or without metformin [47] or a thiazolidine- alogliptin 25 mg once daily plus pioglitazone than with
dione with or without a sulfonylurea or metformin [28]. pioglitazone [21, 28, 47] (Table 2). HbA1c targets of
Alogliptin: A Review 783

Table 2 Efficacy of alogliptin plus pioglitazone in patients with type 2 diabetes inadequately controlled with pioglitazone (with or without other
oral antidiabetic drugs) [21, 28, 47] or diet and exercise [48]. Results of trials of 12 [21], 16 [47] or 26 [28, 48] weeks’ duration
Study Treatment (mg) No. of ptsa LSM change from Changec from baseline HbA1c target
baseline in HbA1cb in FPG (mmol/L) (% of pts)
(%) [baseline value] [baseline value]
B7.0 or B6.5 or
\6.9 %d \6.2 %e

Ji et al. [47]f ALO 25 od ? PIOg 60 -0.76*** -1.07h 61.7*** 30.0***


g
PIO 63 -0.25 -0.11h 31.7 9.5
Kaku et al. [21] ALO 12.5 od ? PIO 15 or 30 daily 111 -0.91**** [7.91] -0.83* 49.5* 6.3*
ALO 25 od ? PIO 15 or 30 daily 113 -0.97**** [7.89] -1.05* 49.6* 4.4*
PIO 15 or 30 daily 115 -0.19 [7.92] -0.13 20.0 0
i
Pratley et al. [28] ALO 12.5 od ? PIO 197 -0.66*** [8.1] -1.09** 44.2*
ALO 25 od ? PIOi 199 -0.80*** [8.0] -1.10** 49.2*
PIOi 97 -0.19 [8.0] -0.32 34.0
Rosenstock et al. [48] ALO 12.5 od ? PIO 30 od 163 -1.56* [8.85] -2.7* [11.0] 53.4* 26.4*
ALO 25 od ? PIO 30 od 164 -1.71*  [8.80] -2.8*  [10.2] 62.8*  27.4 
ALO 25 od 164 -0.96 [8.80] -1.4 [10.5] 24.4 11.6
PIO 30 od 163 -1.15 [8.76] -2.1 [10.5] 33.7 16.6
ALO alogliptin, FPG fasting plasma glucose, HbA1c glycated haemoglobin, LSM least squares mean, MET metformin, od once daily, PIO
pioglitazone, PL placebo, pts patients
 
* p \ 0.05, ** p \ 0.01, *** p \ 0.001, **** p \ 0.0001 vs. PIO; p \ 0.05 vs ALO
a
No. of patients in the full analysis set [21, 47] or intent-to-treat population [28, 48]
b
Primary endpoint
c
Mean [21] or least squares mean [28, 47, 48] changes
d
HbA1c target of B7.0 % [28, 47, 48] or \6.9 % [21]
e
HbA1c target of B6.5 % [47, 48] or \6.2 % [21]
f
Available as an abstract and supplemented by data from ClinicalTrials.gov (NCT01289119). This study also included treatment arms in which
pts received ALO vs. PL (see Sect. 4.1 for results) or ALO ? MET vs. MET (see Table 1 for results)
g
Pts continued to receive their stable dosage of PIO with or without MET
h
Statistical analysis not reported
i
Pts continued with their stable dosage of PIO or switched from rosiglitazone to PIO 30 or 45 mg. At baseline, 56.2 and 21.2 % of pts were also
receiving MET or a sulfonylurea, respectively; a stable dosage of these agents was administered throughout the trial

B7.0 % [28, 47],\6.9 % [21], B6.5 % [47] or\6.2 % [21] In drug-naive patients with type 2 diabetes inadequately
were achieved by significantly more alogliptin 25 mg once controlled with diet and exercise, HbA1c was reduced from
daily plus pioglitazone recipients than pioglitazone re- baseline to a significantly greater extent with alogliptin
cipients (Table 2). 25 mg once daily plus pioglitazone 30 mg once daily than
Significantly greater reductions in FPG were seen with with pioglitazone 30 mg once daily or alogliptin 25 mg
alogliptin 25 mg once daily plus pioglitazone than with once daily after 26 weeks’ therapy [48] (Table 2). In ad-
pioglitazone [21, 28] (Table 2). In addition, changes from dition, HbA1c targets of B7.0 and B6.5 % were achieved
baseline in PPG levels 2 h after a meal tolerance test were by significantly more recipients of alogliptin 25 mg once
significantly (p \ 0.05) greater with alogliptin 12.5 or daily plus pioglitazone 30 mg once daily than with pi-
25 mg once daily plus pioglitazone than with pioglitazone oglitazone 30 mg once daily or alogliptin 25 mg once daily
after 12 weeks’ therapy (-1.76 and -2.30 vs. [48] (Table 2).
-0.25 mmol/L) [21]. Significantly greater reductions in FPG were also seen
Results of the 40-week extension phase of the 12-week with alogliptin 25 mg once daily plus pioglitazone 30 mg
study (during which recipients of pioglitazone alone once daily than with pioglitazone 30 mg once daily or
switched to alogliptin 12.5 or 25 mg once daily plus pi- alogliptin 25 mg once daily [48] (Table 2). Hypergly-
oglitazone) showed that HbA1c, FPG and 2-h PPG levels caemic rescue was required by significantly fewer re-
were significantly reduced from baseline at all time-points cipients of alogliptin 25 mg once daily plus pioglitazone
during the 52-week follow-up period with alogliptin plus 30 mg once daily than alogliptin 25 mg once daily (2.5 vs.
pioglitazone (p-values not stated) [21]. 11.3 %; p \ 0.05) [48].
784 G. M. Keating

4.2.3 In Combination with a Sulfonylurea 12 weeks’ therapy, HbA1c was reduced from baseline to a
significantly greater extent with alogliptin 25 mg once
Two randomized, double-blind, multicentre trials of 12 daily plus voglibose 0.2 mg three times daily than with
[17] or 26 [30] weeks’ duration compared the efficacy of voglibose 0.2 mg three times daily alone (Table 3). In
alogliptin plus glibenclamide [30] or glimepiride [17] with addition, significantly more patients receiving alogliptin
glibenclamide [30] or glimepiride [17] alone in patients 12.5 or 25 mg once daily plus voglibose than voglibose
with type 2 diabetes inadequately controlled with a sul- alone achieved HbA1c targets of \6.9 % (Table 3) or
fonylurea [17, 30]. The mean duration of diabetes was 7.7 \6.2 % (5.3 and 10.1 vs. 0 %; p \ 0.05). FPG was re-
[30] or 9.8 [17] years. duced to a significantly greater extent with alogliptin
Alogliptin plus a sulfonylurea improved glycaemic 25 mg once daily plus voglibose than with voglibose alone
control in patients inadequately controlled with a sulfon- (Table 3). In addition, changes from baseline in PPG levels
ylurea alone. After 12 [17] or 26 [30] weeks’ therapy, 2 h after a meal tolerance test were significantly (p \ 0.05)
HbA1c was reduced from baseline to a significantly greater greater with alogliptin 12.5 or 25 mg once daily plus
extent with alogliptin 25 mg once daily plus glibenclamide voglibose than with voglibose after 12 weeks’ therapy
or glimepiride than with glibenclamide or glimepiride (-2.62 and -3.00 vs. -0.01 mmol/L) [22].
alone [17, 30] (Table 3). Patients receiving alogliptin Results of the 40-week extension phase of the 12-week
25 mg once daily plus glibenclamide or glimepiride were study (during which recipients of voglibose alone switched
significantly more likely than those receiving gliben- to alogliptin 12.5 or 25 mg once daily plus voglibose)
clamide or glimepiride alone to achieve an HbA1c target of showed that HbA1c and FPG were significantly reduced
B7.0 % [30] or \6.9 % [17] (Table 3). from baseline at all time-points during the 52-week follow-
In one trial, FPG was reduced to a significantly greater up period with alogliptin plus voglibose (p-values not
extent in patients receiving alogliptin 25 mg once daily stated) [22].
plus glimepiride than in those receiving glimepiride alone
[17] (Table 3). However, in the other trial, changes from 4.2.5 In Combination with Insulin
baseline in FPG did not significantly differ between
alogliptin 25 mg once daily plus glibenclamide recipients Two randomized, double-blind, multicentre trials exam-
and glibenclamide alone recipients [30] (Table 3). Changes ined the efficacy of alogliptin in combination with insulin
from baseline in PPG 2 h after a meal tolerance test were in patients with type 2 diabetes inadequately controlled by
significantly (p \ 0.05) greater with alogliptin 12.5 or insulin alone [29, 53] or insulin plus metformin [29]. Pa-
25 mg once daily plus glimepiride than with glimepiride tients received alogliptin plus insulin or insulin (with or
after 12 weeks’ therapy (-2.46 and -1.85 vs. ?0.45 without metformin [29]) for 12 [53] or 26 [29] weeks. Most
mmol/L) [17]. Patients receiving alogliptin 25 mg once patients were receiving premixed insulins or insulin com-
daily plus glibenclamide were significantly less likely than binations at baseline (see Table 3 for further details). Pa-
those receiving glibenclamide alone to require hypergly- tients had a mean duration of diabetes of 12.6 [29] or 14.9
caemic rescue [30] (Table 3). [53] years.
Results of the 40-week extension phase of the 12-week Combination therapy with alogliptin plus insulin im-
study (during which recipients of glimepiride alone proved glycaemic control in patients inadequately con-
switched to alogliptin 12.5 or 25 mg once daily plus trolled by insulin [29, 53]. After 12 [53] or 26 [29] weeks’
glimepiride) showed that HbA1c, FPG and 2-h PPG levels therapy, HbA1c was reduced from baseline to a sig-
were significantly reduced from baseline at all time-points nificantly greater extent with alogliptin 25 mg once daily
during the 52-week follow-up period with alogliptin plus plus insulin than with insulin (Table 3). FPG was also re-
glimepiride (p-values not stated) [17]. duced to a significantly greater extent with alogliptin
25 mg once daily plus insulin than with insulin in the
4.2.4 In Combination with Voglibose 26-week trial [29], but not in the 12-week trial [53], and
significantly fewer recipients of alogliptin 25 mg once
A randomized, double-blind, multicentre, 12 week trial daily plus insulin than insulin required hyperglycaemic
compared the efficacy of alogliptin plus voglibose with rescue [29] (Table 3). Consistent with the study protocol,
voglibose alone in patients with type 2 diabetes the mean insulin dosage at week 26 was essentially un-
inadequately controlled by an a-glucosidase inhibitor [22]. changed from baseline [29]. Similarly, minimal changes in
The mean duration of diabetes was 7.7 years [22]. insulin dosages were seen in the 12-week trial [53].
Combination therapy with alogliptin plus voglibose Results of the 40-week extension phase of the 12-week
improved glycaemic control in patients inadequately con- study (during which recipients of insulin alone switched to
trolled with an a-glucosidase inhibitor [22]. After alogliptin plus insulin) revealed that the efficacy of add-on
Alogliptin: A Review 785

Table 3 Efficacy of alogliptin plus a sulfonylurea, voglibose or insulin in patients with inadequately controlled type 2 diabetes. Results of trials
of 12 [17, 22, 53] or 26 [29, 30] weeks’ duration
Study Treatment (mg) No. of LSM change from Changec from HbA1c target Hyperglycaemic
ptsa baseline in HbA1cb baseline in of B7.0 or rescue
(%) [baseline value] FPG (mmol/L) \6.9 %d (% of pts)
[baseline value] (% of pts)

In combination with a sulfonylurea


Pratley et al. [30] ALO 12.5 od ? GBCe 203 -0.39*** -0.26 29.6 14.9*
ALO 25 od ? GBCe 198 -0.53*** -0.47 34.8** 15.7*
GBCe 99 ?0.01 ?0.12 18.2 28.3
Seino et al. [17] ALO 12.5 od ? GMP 1–4 daily 104 -0.59**** [8.54] -1.24* 9.6*
ALO 25 od ? GMP 1–4 daily 104 -0.65**** [8.54] -0.88* 7.7*
GMP 1–4 daily 103 ?0.35 [8.62] ?0.33 0
In combination with VOG
Seino et al. [22] ALO 12.5 od ? VOG 0.2 tid 76 -0.96   [8.02] -1.06  46.1 
ALO 25 od ? VOG 0.2 tid 79 -0.93   [7.91] -1.03  50.6 
VOG 0.2 tid 75 ?0.06 [8.12] -0.31 12.2
In combination with INS
Rosenstock et al. [29] ALO 12.5 od ? INSf 131 -0.63àà [9.3] ?0.1 [10.5] 21àà
àà à
ALO 25 od ? INS f
129 -0.71 [9.3] -0.6 [10.3] 20àà
INSf 130 -0.13 [9.3] ?0.3 [10.9] 40
Kaku et al. [53] ALO 25 od ? INSg 90 -0.96ààà [8.4] -1.1 [8.6] 23.3
INSg 89 -0.29 [8.4] -0.5 [8.6] 5.7
ALO alogliptin, FPG fasting plasma glucose, GBC glibenclamide, GMP glimepiride, HbA1c glycated haemoglobin, INS insulin, LSM least
squares mean, od once daily, pts patients, tid three times daily, VOG voglibose
     à àà
* p \ 0.05, ** p \ 0.01, *** p \ 0.001, **** p \ 0.0001 vs. sulfonylurea; p \ 0.05, p \ 0.0001 vs. VOG; p \ 0.05, p \ 0.001,
ààà
p \ 0.0001 vs. INS
a
No. of patients in the full analysis set [17, 22, 29, 53] or intent-to-treat population [30]
b
Primary endpoint
c
Mean [17, 22, 29, 53] or least squares mean [30] changes
d
HbA1c target of B7.0 [30, 53] or \6.9 % [17, 22]
e
Pts continued a stable GBC dosage (median 10 mg/day at study entry)
f
Pts continued their existing INS regimen, with premixed INS or INS combinations, long-acting INS and short-acting INS administered to 64,
34 and 2 % of pts, respectively. The mean INS dosage at baseline was 57 IU/day, and 59 % of pts were also receiving metformin
g
Pts continued their existing INS regimen, with premixed INS, long-acting INS and intermediate-acting INS administered to 80, 15 and 5 % of
pts, respectively. The mean INS dosage at baseline was 23 IU/day

therapy with alogliptin was maintained in the longer term plus metformin in patients with type 2 diabetes
[53]. At week 52, the mean reduction from baseline in inadequately controlled by metformin plus pioglitazone or
HbA1c was 1.0 % in patients initially randomized to another OAD [49]. In terms of the reduction from baseline
alogliptin plus insulin or insulin alone [53]. in HbA1c, alogliptin 25 mg once daily plus pioglitazone
30 mg once daily and metformin was deemed to be non-
4.2.6 In Combination with Pioglitazone Plus Metformin inferior to pioglitazone 45 mg once daily and metformin at
weeks 26 and 52, with superiority also shown at week 52
The efficacy of triple therapy with alogliptin plus piogli- (Table 4). In addition, significantly more patients receiving
tazone and metformin was examined in two randomized, alogliptin plus pioglitazone and metformin than uptitrated
double-blind, multicentre in patients with type 2 diabetes pioglitazone plus metformin achieved an HbA1c target of
inadequately controlled by metformin alone [50] or met- B7.0 or B6.5 % (Table 4). FPG was reduced to a sig-
formin plus pioglitazone or another OAD [49]. The trials nificantly greater extent with alogliptin plus pioglitazone
were of 26 [50] or 52 [49] weeks’ duration and patients had and metformin than with uptitrated pioglitazone plus
a mean diabetes duration of 6.2 [50] or 7.2 [49] years. metformin (Table 4) [49].
Triple therapy with alogliptin plus pioglitazone and Triple therapy with alogliptin plus pioglitazone and
metformin was more effective than uptitrated pioglitazone metformin also improved glycaemic control in patients
786 G. M. Keating

Table 4 Efficacy of triple therapy with alogliptin, pioglitazone and metformin in patients with inadequately controlled type 2 diabetes
Study Treatmenta No. of Time- LSM change from LSM change from HbA1c target (%
ptsb point baseline in HbA1cc baseline in FPG of pts)
(weeks) (%) [baseline value] (mmol/L)
[baseline value] B7.0 % B6.5 %

Bosi et al. [49] ALO 25 od ? PIO 30 od ? MET 303 26 -0.89d [8.3] -0.9* [9.0]
d
52 -0.70 -0.8* 33.2** 8.7**
PIO 45 od ? MET 306 26 -0.42 [8.1] -0.3 [9.0]
52 -0.29 -0.2 21.3 4.3
DeFronzo et al. [50]e ALO 12.5 od ? PIO 15 od ? MET 130 26 -1.34  [8.5] -2.33  [10.2] 49.2f 21.5f
ALO 12.5 od ? PIO 30 od ? MET 128 26 -1.39  [8.5] -2.34  [10.0] 53.1f 30.0f
ALO 12.5 od ? PIO 45 od ? MET 127 26 -1.55  [8.5] -2.85  [9.7] 61.5f 32.3f
    f
ALO 25 od ? PIO 15 od ? MET 127 26 -1.27 [8.5] -2.11 [10.0] 54.6 24.6f
    f
ALO 25 od ? PIO 30 od ? MET 124 26 -1.39 [8.5] -2.31 [9.9] 53.1 30.0f
    f
ALO 25 od ? PIO 45 od ? MET 126 26 -1.60 [8.6] -2.92 [9.9] 60.0 33.1f
f
ALO 12.5 od ? MET 122 26 -0.64 [8.6] -0.73 [10.2] 22.7 8.6f
ALO 25 od ? MET 123 26 -0.90 [8.6] -1.03 [10.2] 27.1f 12.4f
f
PIO 15 od ? MET 127 26 -0.75 [8.5] -1.31 [9.8] 25.6 6.2f
f
PIO 30 od ? MET 123 26 -0.92 [8.5] -1.60 [9.7] 29.5 11.6f
PIO 45 od ? MET 126 26 -1.00 [8.5] -1.80 [10.0] 36.4f 19.4f
PL ? MET 126 26 -0.13 [8.5] ?0.36 [9.8] 6.2f 0.8f
ALO alogliptin, FPG fasting plasma glucose, HbA1c glycated haemoglobin, LSM least squares mean, MET metformin, od once daily, PIO
pioglitazone, PL placebo, pts patients
 
* p \ 0.01, ** p \ 0.001 vs. PIO ? MET; p B 0.001 vs. corresponding dosages of ALO ? MET or PIO ? MET
a
Pts received a stable MET dosage of 1500 mg/day or the maximum tolerated dosage [49] or C1500 mg/day [50]
b
No. of patients in the full analysis set [50] or per-protocol population [49]
c
Primary endpoint
d
Noninferiority was shown between ALO ? PIO ? MET and PIO ? MET at weeks 26 and 52, with superiority also shown at week 52
e
Supplemented by data from ClinicalTrials.gov (NCT00328627)
f
Statistical analysis not reported

with type 2 diabetes inadequately controlled by metformin for the individual treatment arms are shown in Table 4. In
alone [50]. When the pioglitazone dosage groups were addition, significantly (p \ 0.01) fewer patients receiving
pooled, the least squares mean reduction from baseline in alogliptin 12.5 or 25 mg once daily plus pioglitazone 15, 30
HbA1c at week 26 was significantly (p \ 0.001) greater or 45 mg once daily plus metformin than pioglitazone 15, 30
with both alogliptin 12.5 and 25 mg once daily plus pi- or 45 mg once daily plus metformin required hypergly-
oglitazone 15, 30 or 45 mg once daily plus metformin than caemic rescue (3.9 and 3.4 vs. 11.4 %) [50].
with pioglitazone 15, 30 or 45 mg once daily plus met-
formin (-1.4 % for both alogliptin dosages vs. -0.9 %). 4.3 Special Patient Populations
The proportion of patients achieving an HbA1c target of
B7.0 % was significantly (p \ 0.001) greater with 4.3.1 Elderly Patients
alogliptin 12.5 or 25 mg once daily plus pioglitazone 15,
30 or 45 mg once daily plus metformin than with piogli- The efficacy of alogliptin monotherapy was compared with
tazone 15, 30 or 45 mg once daily plus metformin (54.6 that of glipizide monotherapy in elderly patients aged
and 55.9 vs. 30.5 %). Results for the individual treatment 65–90 years with type 2 diabetes in a randomized, double-
arms are shown in Table 4 [50]. blind, multicentre, 52-week study [51]. Patients had mild
The least squares mean reduction from baseline in FPG at hyperglycaemia despite diet and exercise with or without
week 26 was also significantly (p \ 0.001) greater with both OAD monotherapy. The mean duration of type 2 diabetes
alogliptin 12.5 and 25 mg once daily plus pioglitazone 15, 30 was 6.1 years and the mean baseline HbA1c was 7.50 % in
or 45 mg once daily plus metformin than with pioglitazone alogliptin recipients and 7.45 % in glipizide recipients. The
15, 30 or 45 mg once daily plus metformin (-2.5 mmol/L primary efficacy analysis was conducted in the per-protocol
for both alogliptin dosages vs. -1.6 mmol/L) [50]. Results population (n = 342) [51].
Alogliptin: A Review 787

After 52 weeks’ therapy, alogliptin 25 mg once daily 5 Tolerability and Safety


was noninferior to glipizide 5–10 mg once daily in terms of
the change from baseline in HbA1c [51]. The least squares In addition to the trials discussed in Sect. 4, tolerability
mean reduction from baseline in HbA1c was 0.14 % with data were obtained from a pooled analysis of tolerability
alogliptin and 0.09 % with glipizide (mean baseline values data reported in the US prescribing information [4]. The
of 7.54 and 7.46 %). An HbA1c target of B7.0 was pooled analysis included 14 randomized, double-blind tri-
achieved in 49 % of alogliptin recipients and 45 % of als in &8500 alogliptin recipients (as monotherapy or in
glipizide recipients and an HbA1c target of B6.5 % was combination with other OADs or insulin), &2200 active
achieved in 22 and 18 % of patients in the corresponding comparator recipients and &2900 placebo recipients [4].
treatment groups [51]. The least squares mean reduction Data regarding cardiovascular outcomes were obtained
from baseline in FPG was 0.11 mmol/L with alogliptin and from the randomized, double-blind, multinational EXAM-
0.22 mmol/L with glipizide (mean baseline values of 8.21 INE trial [56]. In EXAMINE, patients with type 2 diabetes
and 7.99 mmol/L). In addition, the mean change in 2-h and a recent acute coronary syndrome [i.e. acute myocar-
PPG was -0.33 mmol/L in alogliptin recipients (from a dial infarction (MI) or unstable angina pectoris requiring
baseline of 10.93 mmol/L) versus ?0.33 in glipizide re- hospitalization within the previous 15–90 days] received
cipients (from a baseline of 10.21 mmol/L). Hypergly- alogliptin 6.25–25 mg once daily (dosage dependent on
caemic rescue was required in 25 % of alogliptin recipients renal function) [n = 2701] or placebo (n = 2679), in ad-
and in 22 % of glipizide recipients [51]. ditional to standard of care treatment for type 2 diabetes.
The primary endpoint in EXAMINE was a composite of
4.3.2 Patients Undergoing Haemodialysis death from cardiovascular causes, nonfatal MI or nonfatal
stroke. Patients were followed for up to 40 months (median
Two small (n = 16 [54] and 30 [55]) studies of 48 weeks duration of alogliptin exposure of 533 days) [56]. An ad-
[55] or 2 years [54] duration examined the efficacy of ditional analysis of EXAMINE is available, reporting heart
alogliptin in patients with type 2 diabetes who were un- failure and mortality outcomes [57]. In EXAMINE, 1533
dergoing haemodialysis [54, 55]. The trials were of non- patients (28 %) had a history of heart failure, although
comparative, single-centre [54] or open-label, multicentre patients with New York Heart Association (NYHA) class
[55] design. Patients had inadequate glycaemic control IV heart failure were excluded from the trial [56].
despite receiving diet and exercise therapy [54] or treat-
ment with dietary modification alone (n = 15) or mit- 5.1 General Tolerability Profile
iglinide and/or voglibose (n = 15) [55]. At baseline, mean
HbA1c was 7.1 % [54, 55], and mean haemoglobin levels Oral alogliptin was generally well tolerated in patients with
were 10.8 g/dL [54] and 10.9 g/dL [55]. Given that anae- type 2 diabetes when administered as monotherapy [27, 46]
mia is common in patients undergoing haemodialysis and as dual therapy in combination with metformin [18–20],
may falsely lower HbA1c levels [55], glycated albumin pioglitazone [21, 28], voglibose [22], a sulfonylurea [17,
levels were also assessed in these trials. 30] or insulin [29, 53] or as triple therapy in combination
Alogliptin 6.25 mg once daily improved glycaemic with pioglitazone plus metformin [49, 50]. Most adverse
control in patients undergoing haemodialysis who had type events were of mild to moderate severity [12, 17–20, 28–
2 diabetes inadequately controlled by diet and exercise 30, 46, 49] and were not considered treatment related [17–
alone [54]. After 24 months’ therapy, mean HbA1c was 21, 29, 30, 49, 53].
significantly (p \ 0.05) reduced from 7.1 % at baseline to In the pooled analysis, adverse events were reported in
5.8 %. Mean glycated albumin levels were also sig- 66 % of alogliptin 25 mg once daily recipients, in 70 % of
nificantly (p \ 0.05) reduced from 22.5 % at baseline to active comparator recipients and in 62 % of placebo re-
19.6 % at 24 months [54]. cipients, with discontinuation because of adverse events
Alogliptin 6.25 mg once daily improved glycaemic occurring in 4.7, 6.2 and 4.5 % of patients in the corre-
control in patients undergoing haemodialysis who had type sponding treatment groups [4]. Adverse reactions reported
2 diabetes inadequately controlled by diet or by mitiglinide in C4 % of the 5902 alogliptin 25 mg once daily recipients
and/or voglibose [55]. After 48 weeks’ therapy, mean and in numerically more alogliptin than placebo recipients
HbA1c was significantly (p \ 0.0001) reduced from 7.1 % included nasopharyngitis (4.4 % of alogliptin 25 mg once
at baseline to 6.3 %. Significant (p \ 0.0001) reductions daily recipients vs. 5.0 % of active comparator recipients
from baseline were also seen in glycated albumin levels and 3.0 % of placebo recipients), headache (4.2 vs. 5.4 and
(from 25.6 to 20.7 %) and in PPG (from 11.77 to 2.5 %) and upper respiratory tract infection (4.2 vs. 5.0 and
8.66 mmol/L) [55]. 2.1 %) [4].
788 G. M. Keating

Serious adverse events considered to be possibly or parameters were generally reported as being of no clinical
probably related to treatment occurred infrequently in relevance or of mild severity [12, 17–22, 29, 30, 46, 47,
alogliptin recipients. For example, no serious adverse 49–51].
events considered to be related to treatment occurred in
patients receiving monotherapy with alogliptin 25 mg once 5.2 Adverse Cardiovascular Events
daily [46]. In addition, serious adverse events considered to
be possibly or probably related to treatment occurred in The risk of major cardiovascular events was not increased
0–1 % of alogliptin 25 mg once daily plus metformin re- with alogliptin in patients with type 2 diabetes and a recent
cipients versus 0 % of metformin monotherapy recipients acute coronary syndrome, according to the results of the
[18, 20], in 0–1.5 % of alogliptin 25 mg once daily plus EXAMINE trial [56]. The primary composite endpoint
pioglitazone recipients versus 1 % of pioglitazone mono- (death from cardiovascular causes, nonfatal MI or nonfatal
therapy recipients [21, 28], in 0 % of alogliptin 25 mg once stroke) occurred in 11.3 % of alogliptin recipients and in
daily plus glibenclamide recipients versus 1 % of gliben- 11.8 % of placebo recipients [hazard ratio (HR) 0.96, with
clamide monotherapy recipients [30], in 0 % of alogliptin an upper limit for the one-sided repeated CI of 1.16;
25 mg once daily plus insulin recipients versus 0 % of p \ 0.001 for noninferiority; p = 0.32 for superiority].
insulin recipients [29], and in 0.3 % of patients receiving When components of the primary composite endpoint were
alogliptin 25 mg once daily plus pioglitazone and met- considered separately, there was no significant difference
formin versus 0.3 % of patients receiving pioglitazone plus between alogliptin and placebo recipients in the rate of
metformin [50]. cardiovascular mortality (3.3 vs. 4.1 %) [HR 0.79; 95 % CI
The good tolerability profile of alogliptin was main- 0.60–1.04], nonfatal MI (6.9 vs. 6.5 %) [HR 1.08; 95 % CI
tained in the longer-term, according to extension studies 0.88–1.33] or nonfatal stroke (1.1 vs. 1.2 %) [HR 0.91;
examining the use of up to 52 weeks’ alogliptin therapy 95 % CI 0.55–1.50]. In addition, there was no significant
[12, 17, 18, 21, 22, 53] and results of a 2-year study [52]. difference between alogliptin and placebo recipients in the
Alogliptin 25 mg once daily was generally well toler- incidences of the key secondary endpoint comprising the
ated in elderly patients with type 2 diabetes [51]. Adverse primary composite endpoint plus urgent revascularization
events were reported in 73.4 % of alogliptin recipients and because of unstable angina within 24 h of hospital admis-
in 68.9 % of glipizide recipients, with drug-related adverse sion (12.7 vs. 13.4 %) [HR 0.95; with an upper limit for the
events reported in 16.2 and 21.5 % of patients, respec- one-sided repeated CI of 1.14], all-cause mortality (5.7 vs.
tively, and discontinuation because of adverse events oc- 6.5 %) [HR 0.88; 95 % CI 0.71–1.09] or cardiovascular
curring in 8.6 and 12.3 % of patients, respectively. The mortality when both primary endpoint events and death
most commonly reported adverse events included urinary occurring after a nonfatal primary endpoint were included
tract infection, dizziness and headache [51]. (4.1 vs. 4.9 %) [HR 0.85; 95 % CI 0.66–1.10] [56].
Alogliptin was also generally well tolerated in patients The risk of the prespecified composite endpoint of the first
with type 2 diabetes who were undergoing haemodialysis, occurrence of all-cause mortality, nonfatal MI, nonfatal
with no significant increase in adverse events such as stroke, urgent revascularization because of unstable angina
symptomatic hypoglycaemia, liver impairment, anaemia or and hospitalized heart failure did not significantly differ
skin rash [55]. Moreover, bodyweight gain between dialysis between alogliptin and placebo recipients in EXAMINE
sessions was significantly (p = 0.04) reduced from baseline (16.0 vs. 16.5 %) [HR 0.98; 95 % CI 0.86–1.12] [57]. Within
with alogliptin [55]. In another study in patients with type 2 this composite endpoint, first hospitalized heart failure oc-
diabetes who were undergoing haemodialysis, none of the curred in 3.1 % of alogliptin recipients and in 2.9 % of
alogliptin recipients experienced significant adverse events placebo recipients (HR 1.07; 95 % CI 0.79–1.46). Moreover,
(e.g. hypoglycaemia), although drug-related rash occurred the risk of the post hoc composite endpoint of cardiovascular
in one patient [54]. death and hospitalized heart failure did not significantly
Death was reported infrequently among patients re- differ between alogliptin and placebo recipients in the
ceiving alogliptin or comparator agents in trials of 26 [19, overall population (7.4 vs. 7.5 %) [HR 1.00; 95 % CI
20, 29, 46, 50] or 52 [17, 18, 21, 22, 49] weeks’ duration. 0.82–1.21] or in the subgroups of patients with (13.9 vs.
In a 2-year study, death was reported in 0.3 % of alogliptin 15.7 %) [HR 0.90; 95 % CI 0.70–1.17] or without (4.9 vs.
12.5 mg once daily plus metformin recipients, 0.3 % of 4.2 %) [HR 1.14; 95 % CI 0.85–1.54] a history of heart
alogliptin 25 mg once daily plus metformin recipients and failure. In terms of the hospitalized heart failure component,
0.6 % of glipizide plus metformin recipients [52]. there was no significant difference between alogliptin and
No clinically relevant changes in vital signs and/or ECG placebo recipients in the overall population (3.9 vs. 3.3 %)
recordings were reported in alogliptin recipients [12, 17– [HR 1.19; 95 % CI 0.90–1.58] or in the subgroup of patients
22, 29, 30, 46, 49–51, 53]. Changes in laboratory with a history of heart failure (8.2 vs. 8.5 %) [HR 1.00; 95 %
Alogliptin: A Review 789

CI 0.71–1.42]. However, in the subgroup of patients without comparator [4]; the analysis included interim data from the
a history of heart failure, alogliptin recipients were sig- EXAMINE trial [58]. In EXAMINE, there was no sig-
nificantly more likely than placebo recipients to experience nificant difference between alogliptin and placebo re-
hospitalized heart failure (2.2 vs. 1.3 %) [HR 1.76; 95 % CI cipients in the incidence of acute pancreatitis (0.4 vs.
1.07–2.90; p = 0.026]. Analysis of the post hoc composite 0.3 %) or chronic pancreatitis (0.2 vs. 0.1 %), and there
endpoint by baseline levels of N-terminal prohormone of were no reports of pancreatic cancer [56]. The proportion
brain natriuretic peptide revealed no significant difference of patients with serum ALT or AST levels [3 times the
between alogliptin and placebo in all quartiles [57]. upper limit of normal did not significantly differ between
alogliptin and placebo recipients [56].
5.3 Hypoglycaemia
5.5 Other Adverse Events of Interest
Alogliptin was generally associated with a low risk of
hypoglycaemia in patients with type 2 diabetes [12, 17–22, Alogliptin monotherapy is generally considered weight
28, 46, 47, 49, 50, 52], and most hypoglycaemic events neutral [12, 19, 27]. For example, a bodyweight change of
were of mild to moderate severity [12, 17–21, 28, 46, 47, ?0.15 kg was seen in patients who received alogliptin
49, 50, 52]. The incidence of hypoglycaemia was reported 25 mg once daily for 52 weeks [12]. In addition, only
to be similar in recipients of alogliptin 25 mg once daily small changes from baseline in bodyweight were reported
versus placebo (0 vs. 1.3 % [12]), alogliptin 25 mg once in patients receiving alogliptin in combination with met-
daily plus metformin versus metformin alone (0 vs. 3 % formin [19, 20], pioglitazone [21, 28], a sulfonylurea [30]
[20] and 2 vs. 0 % [18]), alogliptin 25 mg once daily plus or insulin [29, 53]. Moreover, changes in bodyweight sig-
pioglitazone versus pioglitazone alone (7.0 vs. 5.2 % [28]), nificantly (p \ 0.001) favoured recipients of alogliptin
alogliptin 25 mg once daily plus voglibose versus vogli- 25 mg once daily plus metformin versus recipients of
bose alone (1.3 vs. 1.3 % [22]), and alogliptin 25 mg once glipizide plus metformin after 2 years’ therapy (-0.89 vs.
daily plus a sulfonylurea versus a sulfonylurea alone (1.9 ?0.95 kg) [52].
vs. 1.0 % [17] and 9.6 vs. 11.1 % [30]). Malignancy was reported rarely among patients receiv-
The incidence of hypoglycaemia was numerically higher ing alogliptin in the trials discussed in Sect. 4 [12, 17, 20].
in patients receiving glipizide plus metformin than in those In the EXAMINE trial, the incidence of malignancy did not
receiving alogliptin 25 mg once daily plus metformin (23.2 significantly differ between alogliptin and placebo re-
vs. 1.4 %) [52]. Among elderly patients, the incidence of cipients (2.0 vs. 1.9 %) [56].
hypoglycaemia was almost fivefold higher with glipizide The incidence of skin reactions (e.g. pruritus, rash) or
than with alogliptin (26.0 vs. 5.4 %) [51]. skin and subcutaneous disorders in alogliptin recipients
The incidence of hypoglycaemia was threefold higher in was generally low [18–20, 30, 46, 49, 50], although it was
patients receiving triple therapy with alogliptin 25 mg once numerically higher with alogliptin than with comparator
daily plus pioglitazone and metformin than in patients re- agents in some trials [18, 20, 49]. For example, drug-
ceiving uptitrated pioglitazone plus metformin (4.5 vs. related skin and subcutaneous disorders occurred in 5.2 %
1.5 %) in one trial [49]. However, in a second trial, the of patients receiving alogliptin 25 mg once daily plus pi-
incidence of hypoglycaemia was 1.5 % in patients receiv- oglitazone and metformin and in 2.3 % of patients re-
ing alogliptin 25 mg once daily plus pioglitazone and ceiving uptitrated pioglitazone plus metformin [49].
metformin and 2.1 % in patients receiving pioglitazone Facial oedema and peripheral oedema occurred in 0.9
plus metformin [50]. and 2.7 % of patients, respectively, receiving alogliptin
As expected, a higher rate of hypoglycaemia was seen in 25 mg once daily plus pioglitazone; oedema-related events
patients receiving background insulin therapy [29, 53]. The were of mild severity and were generally considered to be
incidence of hypoglycaemia was 22.2 % [53] and 27.1 % related to treatment [21]. In patients receiving triple ther-
[29] in patients receiving alogliptin 25 mg once daily plus apy, peripheral oedema was reported in 2.6 % of patients
insulin and 22.5 % [53] and 24.0 % [29] in patients re- receiving alogliptin 25 mg once daily plus pioglitazone and
ceiving insulin. metformin versus 3.6 % of patients receiving pioglitazone
plus metformin [50], and drug-related peripheral oedema
5.4 Pancreatitis and Other Gastrointestinal Events was reported in 2.0 % of patients receiving alogliptin
25 mg once daily plus pioglitazone and metformin and in
In the pooled analysis reported in the US prescribing in- 3.0 % of patients receiving uptitrated pioglitazone plus
formation, pancreatitis was reported in 11 of 5902 (0.19 %) metformin [49]. The incidence of angioedema did not
alogliptin 25 mg once daily recipients and in 5 of 5183 significantly differ between alogliptin and placebo re-
(0.096 %) patients receiving placebo or an active cipients in the EXAMINE trial (0.6 vs. 0.5 %) [56].
790 G. M. Keating

Bone fractures were reported in 1.5 % of patients re- alogliptin/pioglitazone 25/15, 25/30 or 25/45 mg (as ap-
ceiving alogliptin 25 mg once daily plus pioglitazone and propriate based on current therapy) are recommended for
metformin and in 1.0 % of patients receiving uptitrated patients receiving pioglitazone who require additional
pioglitazone plus metformin [49], whereas no treatment- glycaemic control and alogliptin/pioglitazone 25/15 mg is
related bone fractures were reported in patients receiving recommended in patients with NYHA class I or II con-
triple therapy with alogliptin plus pioglitazone and met- gestive heart failure [8]. In patients switching from
formin in another trial [50], or in patients receiving alogliptin coadministered with pioglitazone, alogliptin/pi-
alogliptin 25 mg once daily alone or in combination with oglitazone may be initiated at the same total daily
pioglitazone [48]. alogliptin and pioglitazone dosage [8].
There have been postmarketing reports of serious hy- In the EU, alogliptin/metformin is available as 12.5/850
persensitivity reactions, including anaphylaxis, angioede- and 12.5/1000 mg fixed-dose tablets; the maximum rec-
ma and Stevens-Johnson syndrome, in alogliptin recipients ommended daily dose of alogliptin/metformin is
[4]. In the pooled analysis of clinical trial data reported in 25/2000 mg [7]. Alogliptin/metformin is approved as an
the US prescribing information, hypersensitivity reactions adjunct to diet and exercise to improve glycaemic control
were reported in 0.6 % of alogliptin 25 mg once daily re- in patients inadequately controlled on a maximal tolerated
cipients and in 0.8 % patients receiving placebo or an ac- dose of metformin alone (recommended dosage of
tive comparator [4]. alogliptin/metformin 12.5/850 or 12.5/1000 mg twice dai-
ly), inadequately controlled on maximal tolerated doses of
metformin and pioglitazone (alogliptin/metformin 12.5/850
6 Dosage and Administration or 12.5/1000 mg twice daily and the same dosage of pi-
oglitazone) or inadequately controlled on metformin and a
Alogliptin is approved in the US for use as monotherapy or stable insulin dosage (alogliptin 12.5 mg twice daily and a
combination therapy as an adjunct to diet and exercise to metformin dosage similar to that already being taken).
improve glycaemic control in adults with type 2 diabetes Alogliptin/metformin is also approved for use in patients
[4], and in the EU for use in combination with other an- already receiving combination therapy with alogliptin and
tidiabetic agents (including insulin) as an adjunct to diet metformin administered separately (maintain the same total
and exercise to improve glycaemic control in adults aged daily dosage of alogliptin and metformin) [7].
C18 years with inadequately controlled type 2 diabetes [5]. In the EU, alogliptin/pioglitazone is available as 25/30,
In the US, the recommended alogliptin dosage is 25 mg 25/45, 12.5/30 and 12.5/45 mg fixed-dose tablets; the max-
once daily [4]. In the EU, the recommended alogliptin imum recommended daily dosage of alogliptin and piogli-
dosage is 25 mg once daily as add-on therapy to met- tazone is 25 and 45 mg, respectively [9].
formin, a thiazolidinedione, a sulfonylurea or insulin, and Alogliptin/pioglitazone is indicated as second- or third-line
as triple therapy in combination with metformin and a treatment as an adjunct to diet and exercise to improve gly-
thiazolidinedione or insulin [5]. caemic control in patients inadequately controlled with pi-
In the US, the fixed-dose combinations of alogliptin/ oglitazone alone and in whom metformin is inappropriate
metformin and alogliptin/pioglitazone are approved as (alogliptin/pioglitazone 25/30 or 25/45 mg once daily) or
adjuncts to diet and exercise to improve glycaemic control inadequately controlled on pioglitazone and a maximal tol-
when treatment with both alogliptin and metformin [6] or erated dose of metformin (alogliptin/pioglitazone 25/30 or
alogliptin and pioglitazone [8] is appropriate. Alogliptin/ 25/45 mg once daily and the same dosage of metformin) [9].
metformin is available as 12.5/500 and 12.5/1000 mg Alogliptin/pioglitazone is also approved for use in patients
fixed-dose tablets and should be taken twice daily with already receiving combination therapy with alogliptin and
food; the dose should be gradually escalated and subse- pioglitazone administered separately (maintain the same
quently adjusted based on efficacy and tolerability [6]. The total daily dosage of alogliptin and pioglitazone) [9].
maximum recommended daily dosage of alogliptin/met- The US prescribing information for alogliptin/met-
formin is 25/2000 mg [6]. Alogliptin/pioglitazone is formin contains a black-box warning relating to the risk of
available as 25/15, 25/30, 25/45, 12.5/15, 12.5/30 and 12.5/ lactic acidosis in patients receiving metformin [6], and the
45 mg fixed-dose tablets; the alogliptin/pioglitazone US prescribing information for alogliptin/pioglitazone
dosage can be titrated to a maximum of 25/45 mg once contains a black-box warning relating to the risk of con-
daily [8]. The recommended starting dosage of aloglipt- gestive heart failure in patients receiving thiazolidine-
in/pioglitazone is 25/15 or 25/30 mg for patients diones, including pioglitazone [8].
inadequately controlled on diet or exercise or metformin Local prescribing information should be consulted for
alone or for patients receiving alogliptin who require ad- contraindications, warnings and precautions relating to
ditional glycaemic control [8]. Starting dosages of alogliptin, alogliptin/metformin and alogliptin/pioglitazone.
Alogliptin: A Review 791

7 Place of Alogliptin in the Management of Type 2 glycaemic control to a significantly greater extent than
Diabetes Mellitus pioglitazone alone in patients with type 2 diabetes
inadequately controlled with pioglitazone (with or without
Initial treatment in type 2 diabetes usually comprises life- other OADs) (Sect. 4.2.2). Initial combination therapy with
style changes, with metformin added at or soon after di- alogliptin plus pioglitazone was also more effective than
agnosis [59]. If the HbA1c target (7.0 % for most patients pioglitazone alone in drug-naive patients (Sect. 4.2.2) [48].
[60]) is not reached after &3 months of treatment, add-on Alogliptin is currently the only DPP-4 inhibitor available in
therapy with a sulfonylurea, a thiazolidinedione, a DPP-4 a fixed-dose combination with a thiazolidinedione
inhibitor, a GLP-1 receptor agonist or insulin should be (alogliptin/pioglitazone); this fixed-dose combination has
considered [59]. Both the treatment regimen and the HbA1c been shown to be bioequivalent to alogliptin and pioglita-
target should be tailored to the individual patient [59]. zone coadministered separately (Sect. 3.1). In terms of
DPP-4 inhibitors are currently recommended as a first-line other dual OAD regimens, studies have demonstrated the
option if metformin cannot be used, as add-on therapy in efficacy of alogliptin plus a sulfonylurea (Sect. 4.2.3) or
patients who do not achieve HbA1c targets with metformin voglibose (Sect. 4.2.4).
alone and as a component of triple therapy in patients not Given the weight neutrality and low risk of hypogly-
achieving HbA1c targets with metformin plus a sulfonyl- caemia associated with DPP-4 inhibitors, there is a strong
urea, a thiazolidinedione or insulin [59]. rationale for combining these agents with insulin [62].
Combining antidiabetic agents with complementary Indeed, the addition of alogliptin to insulin improved gly-
mechanisms of action represents a rational approach to the caemic control in patients with type 2 diabetes
treatment of type 2 diabetes. For example, DPP-4 inhibitors (Sect. 4.2.5), without exacerbating hypoglycaemia
such as alogliptin improve insulin secretion and suppress (Sect. 5.3). However, it should be noted that insulin
postprandial glucagon in a glucose-dependent manner, dosages were not optimized during these studies, which is
whereas metformin improves insulin sensitivity and re- not reflective of standard clinical practice [29].
duces hepatic glucose production [20]. Indeed, adding Triple therapy with three OADs is an option in patients
alogliptin to metformin improved glycaemic control in who have not responded to dual therapy [59]. Alogliptin in
patients inadequately controlled with metformin alone combination with metformin and pioglitazone was effec-
(Sect. 4.2.1), whilst preserving the advantages of met- tive in patients with inadequately controlled type 2 dia-
formin (absence of weight gain and low risk of hypogly- betes, and was more effective than uptitrating the
caemia) (Sect. 5) [20]. Moreover, a recent diabetes pioglitazone dosage in patients receiving dual therapy
management algorithm recommends the use of initial (Sect. 4.2.6). Trials comparing alogliptin plus metformin
combination therapy with metformin and another agent, and pioglitazone with an alternative triple therapy regimen
such as a DPP-4 inhibitor, in patients with an initial HbA1c (e.g. metformin plus pioglitazone in combination with in-
of C7.5 % [61]. Initial combination therapy with alogliptin sulin or a sulfonylurea) would be of interest [49].
plus metformin was shown to be more effective than either Alogliptin is also approved for use as monotherapy in
agent alone in drug-naive patients with type 2 diabetes the US (Sect. 6). Trials demonstrated that alogliptin
inadequately controlled by diet and exercise (HbA1c of monotherapy improved glycaemic control in patients with
7.5–10 % at study entry) (Sect. 4.2.1) [19]. inadequately controlled type 2 diabetes (Sect. 4.1).
In studies published to date, patients assigned to com- Results of a meta-analysis suggested that the magnitude
bination therapy received alogliptin and metformin ad- of the HbA1c reduction decreases as the duration of
ministered as separate tablets. However, alogliptin/ alogliptin therapy increases [63]. However, results of long-
metformin is available as a fixed-dose combination tablet term extension studies demonstrated that significant im-
that has been shown to be bioequivalent to alogliptin and provements from baseline in glycaemic control were
metformin coadministered separately (Sect. 3.1). A study maintained with up to 52 weeks of alogliptin therapy [12,
currently underway in China, Malaysia and South Korea is 17, 18, 21, 22], and alogliptin demonstrated sustained ef-
comparing the efficacy of the alogliptin/metformin fixed- ficacy in a 104-week trial [52] (Sect. 4).
dose combination with that of alogliptin and metformin Alogliptin demonstrated efficacy in both Caucasian and
alone in patients with type 2 diabetes (NCT01890122; this Asian patients (Sect. 4). In terms of differences between
study is being conducted to support a regulatory require- these populations, Asian patients with type 2 diabetes tend to
ment in China) [58]. be leaner and insulin deficiency, rather than increased insulin
The thiazolidinedione pioglitazone is an insulin sensi- resistance, underlies their loss of glycaemic control [18]. In
tizer that increases peripheral glucose uptake [48]. Com- addition, Asian patients have a lower insulin secretory ca-
bination therapy with alogliptin plus pioglitazone improved pacity, particularly in the early phase when impaired insulin
792 G. M. Keating

secretion is seen after the ingestion of nutrients, which may patients with NYHA class III or IV heart failure, given that
partly reflect lower intact GLP-1 levels [18, 22]. data are limited in this patient group [5].
Given that DPP-4 inhibitors act by enhancing b-cell In the SAVOR-TIMI 53 trial, saxagliptin had no effect
function, there has been concern that the progressive loss of on the risk of cardiovascular death, MI or stroke (primary
b-cell function seen in type 2 diabetes may mean these agents composite endpoint) in patients with type 2 diabetes and a
are less beneficial in patients with more advanced disease history of established cardiovascular disease or multiple
[62]. However, beneficial effects on glycaemic control have risk factors for vascular disease [66]. However, the risk of
been seen with alogliptin in patients with a relatively long hospitalization for heart failure was significantly
disease duration (e.g. mean disease duration of[7 years [12, (p = 0.007) increased with saxagliptin versus placebo [66].
17, 22, 28–30, 53]), including patients receiving long-term The reason for the discrepancy between EXAMINE and
insulin therapy [29] (Sect. 4). The effect of alogliptin on a- SAVOR-TIMI 53 in terms of the risk of hospitalized heart
cell function (i.e. suppressing postprandial glucagon) may failure is not clear [57]. Results of the VIVIDD trial
play a greater role in later stages of the disease [62]. demonstrated that vildagliptin was noninferior to placebo
Elderly patients with type 2 diabetes present a particular in terms of the increase in left ventricular ejection fraction
challenge, given that they are more susceptible to hypo- seen in patients with type 2 diabetes and NYHA class I–III
glycaemia and more likely to experience disease-related heart failure [67]. More data are needed concerning the risk
morbidity and comorbidities, including renal impairment, of hospitalized heart failure in patients receiving DPP-4
frailty, physical disability and cognitive impairment [64]. inhibitors. Two other large cardiovascular outcomes trials
Thus, DPP-4 inhibitors, with their low risk of hypogly- examining the use of sitagliptin (TECOS) [68] and li-
caemia and good tolerability profile, are a good choice for nagliptin (CAROLINA) [69] are still underway. In addi-
use in the elderly [65]. Indeed, alogliptin was noninferior to tion, the SPEAD-A (Study of Preventive Effects of
glipizide in terms of the improvement in glycaemic control Alogliptin on Diabetic Atherosclerosis) study is examining
(Sect. 4.3.1) and associated with a fivefold lower incidence the ability of alogliptin to prevent the progression of
of hypoglycaemia (Sect. 5.3) in elderly patients with type 2 atherosclerosis (assessed using the carotid intima-media
diabetes. The low potential for drug interactions associated thickness) in patients with type 2 diabetes [70].
with alogliptin (Sect. 3.4) also makes it an appropriate There have been concerns that incretin-based therapies
choice for use in the elderly, given that polypharmacy is may be associated with an increased risk of pancreatitis and
often an issue in this patient population [64, 65]. pancreatic cancer [71–74]. However, type 2 diabetes itself
Treatment options are limited in patients with renal seems to be associated with an increased risk of pancre-
disease, which is commonly seen in patients with type 2 atitis [75] and pancreatic cancer [76], and no increased risk
diabetes [62]. Similar to several other DPP-4 inhibitors, of pancreatitis or cancer was seen in other analyses of DPP-
alogliptin is excreted renally, meaning that reduced dosa- 4 inhibitors [77–81]. In addition, no increase in the risk of
ges are required in patients with moderate or severe renal pancreatitis, pancreatic cancer and/or cancer was seen with
impairment (Sect. 3.3). Alogliptin appeared to improve alogliptin [56] or saxagliptin [66] in the EXAMINE [56]
glycaemic control in patients with type 2 diabetes who and SAVOR-TIMI 53 [66] trials. The US FDA and Euro-
were undergoing haemodialysis, according to the results of pean Medicines Agency (EMA) recently concluded that
two small studies (Sect. 4.3.2). Larger, double-blind trials assertions regarding a causal association between these
examining the use of alogliptin in patients with renal im- agents and pancreatitis or pancreatic cancer are inconsis-
pairment would be of interest. tent with the current data [82]. The FDA and the EMA state
The EXAMINE trial demonstrated that the risk of major that they have not yet reached a final conclusion regarding
cardiovascular events was not increased with alogliptin in a causal relationship between incretin-based agents and
patients with type 2 diabetes and a recent acute coronary pancreatitis or pancreatic cancer, and that while the totality
syndrome [56] (Sect. 5.2). Importantly, the risk of cardio- of data provides reassurance, pancreatitis will continue to
vascular outcomes that included the endpoint of hospitalized be considered a risk associated with these agents until more
heart failure was not increased with alogliptin, and alogliptin data are available [82].
was not associated with an increased risk of first hospitalized Patients with type 2 diabetes are at increased risk of
heart failure overall. In addition, alogliptin was not associ- bone fracture and results of a meta-analysis suggest that
ated with worsening of heart failure outcomes in patients DPP-4 inhibitors may reduce the risk of bone fracture [83].
with a history of heart failure. The higher rate of hospitalized Although it appears that alogliptin may have beneficial
heart failure seen in alogliptin versus placebo recipients in effects on bone quality via the suppression of deleterious
the lower-risk subgroup of patients without a history of heart bone metabolism (Sect. 2.3), more data are needed. Bone
failure may be due to chance [57]. Currently, the EU SPC fracture rates were low in patients with type 2 diabetes
recommends that alogliptin should be used with caution in receiving alogliptin in clinical trials (Sect. 5.5).
Alogliptin: A Review 793

Several DPP-4 inhibitors besides alogliptin are currently Springer. During the peer review process, the manufacturer of the
available for the treatment of type 2 diabetes, including agent under review was offered an opportunity to comment on this
article. Changes resulting from comments received were made by the
vildagliptin, sitagliptin, saxagliptin, linagliptin, gemigliptin author on the basis of scientific and editorial merit.
(available in South Korea) and teneligliptin (available in
Japan). Although there are pharmacodynamic and phar-
macokinetic differences between the various DPP-4 in- References
hibitors (e.g. differences in the potency of DPP-4
inhibition) [3], it is not clear if these differences translate 1. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the
into differences in efficacy. The once-daily administration prevalence of diabetes for 2010 and 2030. Diabetes Res Clin
Pract. 2010;87(1):4–14.
regimen for alogliptin is supported by the finding of DPP-4 2. Deacon CF, Holst JJ. Dipeptidyl peptidase-4 inhibitors for the
inhibition of [80 % 24 h after administration of alogliptin treatment of type 2 diabetes: comparison, efficacy and safety.
25 mg once daily (Sect. 2.1) [13]. Expert Opin Pharmacother. 2013;14(15):2047–58.
DPP-4 inhibitors are more expensive than older treat- 3. Capuano A, Sportiello L, Maiorino MI, et al. Dipeptidyl pepti-
dase-4 inhibitors in type 2 diabetes therapy: focus on alogliptin.
ment options such as metformin and sulfonylureas [84]. Drug Des Dev Ther. 2013;7:989–1001.
Data concerning the cost effectiveness of alogliptin are 4. Takeda Pharmaceuticals America Inc. Nesina (alogliptin) tablets:
limited. A US budget impact analysis, available as an ab- US prescribing information. 2013. http://www.takeda.us/
stract, in patients with type 2 diabetes inadequately con- products/default.aspx. Accessed 3 Mar 2015.
5. European Medicines Agency. Vipidia (alogliptin): EU summary
trolled with a pioglitazone- or DPP-4 inhibitor-containing of product characteristics. 2014. http://www.ema.europa.eu/.
regimen concluded that inclusion of the fixed-dose com- Accessed 3 Mar 2015.
bination of alogliptin/pioglitazone on a managed care 6. Takeda Pharmaceuticals America Inc. Kazano (alogliptin and
health plan formulary had negligible impact on the annual metformin HCl) tablets for oral administration: US prescribing
information. 2013. http://www.takeda.us/products/default.aspx.
pharmacy budget [85]. Accessed 3 Mar 2015.
In conclusion, oral alogliptin is effective and well tol- 7. European Medicines Agency. Vipdomet (alogliptin/metformin):
erated in the treatment of type 2 diabetes. Results of ran- EU summary of product characteristics. 2015. http://www.ema.
domized controlled trials demonstrated that oral alogliptin europa.eu/. Accessed 3 Mar 2015.
8. Takeda Pharmaceuticals America Inc. Oseni (alogliptin and pi-
improved glycaemic control when administered as mono- oglitazone) tablets: US prescribing information. 2013. http://
therapy, as dual therapy in combination with metformin, www.takeda.us/products/default.aspx. Accessed 3 Mar 2015.
pioglitazone, a sulfonylurea, voglibose or insulin, or as 9. European Medicines Agency. Incresync (alogliptin/pioglitazone):
triple therapy in combination with metformin plus piogli- EU summary of product characteristics. 2015. http://www.ema.
europa.eu/. Accessed 3 Mar 2015.
tazone. Alogliptin was generally well tolerated in patients 10. Feng J, Zhang Z, Wallace MB, et al. Discovery of alogliptin: a
with type 2 diabetes and was weight neutral, with a low potent, selective, bioavailable, and efficacious inhibitor of
risk of hypoglycaemia. Results of the large, well designed dipeptidyl peptidase IV. J Med Chem. 2007;50(10):2297–300.
EXAMINE trial revealed that alogliptin was not associated 11. Christopher RJ, Davenport JM, Gwaltney S, et al. Pharmacoki-
netic and pharmacodynamic profiles of SYR-322, a novel in-
with an increased risk of major cardiovascular events in hibitor of dipeptidyl peptidase-IV, in rats, dogs, and monkeys
patients with type 2 diabetes and recent acute coronary [abstract no. 452-P]. In: 66th Annual Scientific Sessions of the
syndrome. Thus, alogliptin is a useful option for the American Diabetes Association. 2006.
treatment of patients with type 2 diabetes. 12. Seino Y, Fujita T, Hiroi S, et al. Efficacy and safety of alogliptin
in Japanese patients with type 2 diabetes mellitus: a randomized,
double-blind, dose-ranging comparison with placebo, followed
Data selection sources: Relevant medical literature (including by a long-term extension study. Curr Med Res Opin.
published and unpublished data) on alogliptin was identified by 2011;27(9):1781–92.
searching databases including MEDLINE (from 1946) and 13. Covington P, Christopher R, Davenport M, et al. Pharmacoki-
EMBASE (from 1996) [searches last updated 16 March 2015], netic, pharmacodynamic, and tolerability profiles of the dipep-
bibliographies from published literature, clinical trial reg- tidyl peptidase-4 inhibitor alogliptin: a randomized, double-blind,
istries/databases and websites. Additional information was also placebo-controlled, multiple-dose study in adult patients with
requested from the company developing the drug. type 2 diabetes. Clin Ther. 2008;30(3):499–512.
Search terms: Alogliptin, type-2 diabetes mellitus, diabetes 14. Hirayama M, Matsuno K, Fujita H, et al. Pharmacokinetics,
mellitus, type 2, non-insulin dependent diabetes mellitus. pharmacodynamics, and tolerability of single and multiple doses
Study selection: Studies in patients with type 2 diabetes mellitus of the dipeptidyl peptidase-4 inhibitor alogliptin in Japanese
who received alogliptin. When available, large, well designed, healthy male subjects [abstract no. 521-P]. In: 68th Scientific
comparative trials with appropriate statistical methodology were Sessions of the American Diabetes Association. 2008.
preferred. Relevant pharmacodynamic and pharmacokinetic data 15. Christopher R, Covington P, Davenport M, et al. Pharmacoki-
are also included. netics, pharmacodynamics, and tolerability of single increasing
doses of the dipeptidyl peptidase-4 inhibitor alogliptin in healthy
male subjects. Clin Ther. 2008;30(3):513–27.
Disclosure The preparation of this review was not supported by any 16. Van Raalte DH, van Genugten RE, Eliasson B, et al. The effect of
external funding. Gillian Keating is a salaried employee of Adis/ alogliptin and pioglitazone combination therapy on various
794 G. M. Keating

aspects of beta-cell function in patients with recent-onset type 2 32. Kato S, Kirigaya H, Gyotoku D, et al. DPP-4 inhibition by
diabetes. Eur J Endocrinol. 2014;170(4):565–74. alogliptin improves the coronary flow reserve evaluated by phase
17. Seino Y, Hiroi S, Hirayama M, et al. Efficacy and safety of contrast cine magnetic resonance imaging in type 2 diabetic pa-
alogliptin added to sulfonylurea in Japanese patients with type 2 tients with coronary artery disease [abstract no. 16542]. In: 86th
diabetes: a randomized, double-blind, placebo-controlled trial Annual Scientific Sessions of the American Heart Association.
with an open-label, long-term extension study. J Diabetes In- 2013.
vestig. 2012;3(6):517–25. 33. Yamauchi Y, Tunekawa S, Seino Y, et al. The effect of DPP-4
18. Seino Y, Miyata Y, Hiroi S, et al. Efficacy and safety of alogliptin inhibitor, alogliptin, on bone metabolism in patients with type 2
added to metformin in Japanese patients with type 2 diabetes: a diabetes mellitus [abstract no. 1071-P]. In: 73rd Annual Scientific
randomized, double-blind, placebo-controlled trial with an open- Sessions of the American Diabetes Association. 2013.
label, long-term extension study. Diabetes Obes Metab. 34. Karim A, Harris S, Fleck P, et al. Assessment of drug interaction
2012;14(10):927–36. between alogliptin benzoate (SYR-322), a highly selective
19. Pratley RE, Fleck P, Wilson C. Efficacy and safety of initial dipeptidyl peptidase-4 inhibitor, and warfarin at steady state
combination therapy with alogliptin plus metformin versus either [abstract no. 106]. J Clin Pharmacol. 2007;47(9):1207.
as monotherapy in drug-naive patients with type 2 diabetes: a 35. Karim A, Covington P, Christopher R, et al. Pharmacokinetics of
randomized, double-blind, 6-month study. Diabetes Obes Metab. alogliptin when administered with food, metformin, or cime-
2014;16(7):613–21. tidine: a two-phase, crossover study in healthy subjects. Int J Clin
20. Nauck MA, Ellis GC, Fleck PR, et al. Efficacy and safety of Pharmacol Ther. 2010;48(1):46–58.
adding the dipeptidyl peptidase-4 inhibitor alogliptin to met- 36. European Medicines Agency. Vipidia (alogliptin): EU public
formin therapy in patients with type 2 diabetes inadequately assessment report. 2013. http://www.ema.europa.eu/. Accessed 3
controlled with metformin monotherapy: a multicentre, ran- Mar 2015.
domised, double-blind, placebo-controlled study. Int J Clin Pract. 37. Karim A, Fleck P, Hetman L, et al. Single-dose pharmacokinetics
2009;63(1):46–55. of the dipeptidyl peptidase-4 inhibitor alogliptin in subjects with
21. Kaku K, Itayasu T, Hiroi S, et al. Efficacy and safety of alogliptin renal impairment [abstract no. 538-P]. In: 68th Annual Scientific
added to pioglitazone in Japanese patients with type 2 diabetes: a Sessions of the American Diabetes Association. 2008.
randomized, double-blind, placebo-controlled trial with an open- 38. Karim A, Fleck P, Dorsey D, et al. Single-dose pharmacokinetics
label long-term extension study. Diabetes Obes Metab. of alogliptin benzoate (SYR-322), a highly selective dipeptidyl
2011;13(11):1028–35. peptidase-4 inhibitor, in subjects with moderate hepatic impair-
22. Seino Y, Fujita T, Hiroi S, et al. Alogliptin plus voglibose in ment [abstract no. 107]. J Clin Pharmacol. 2007;47(9):1207.
Japanese patients with type 2 diabetes: a randomized, double- 39. Karim A, Fleck P, Harrell RE, et al. Effects of age, race, and
blind, placebo-controlled trial with an open-label, long-term ex- gender on the pharmacokinetics and pharmacodynamics of
tension. Curr Med Res Opin. 2011;27(Suppl 3):21–9. alogliptin, a novel and highly selective dipeptidyl peptidase-4
23. Kutoh E, Ukai Y. Alogliptin as an initial therapy in patients with inhibitor in healthy subjects [abstract no. PI-14]. Clin Pharmacol
newly diagnosed, drug naive type 2 diabetes: a randomized, Ther. 2008;83(Suppl 1):S13.
control trial. Endocrine. 2012;41(3):435–41. 40. Karim A, Fleck P, Harris S, et al. Effect of fluconazole, keto-
24. Eliasson B, Möller-Goede D, Eeg-Olofsson K, et al. Lowering of conazole, and gemfibrozil on the pharmacokinetics of alogliptin
postprandial lipids in individuals with type 2 diabetes treated with benzoate (SYR-322) in healthy subjects [abstract no. 105]. J Clin
alogliptin and/or pioglitazone: a randomised double-blind place- Pharmacol. 2007;47(9):1207.
bo-controlled study. Diabetologia. 2012;55(4):915–25. 41. Karim A, Laurent A, Munsaka M, et al. Coadministration of
25. Jurczyk A, Dilorio P, Brostowin D, et al. Improved function and pioglitazone or glyburide and alogliptin: pharmacokinetic drug
proliferation of adult human beta cells engrafted in diabetic im- interaction assessment in healthy participants. J Clin Pharmacol.
munodeficient NOD-scid IL2rcnull mice treated with alogliptin. 2009;49(10):1210–9.
Diabetes Metab Syndr Obes. 2013;6:493–9. 42. Karim A, Fleck P, Harris S, et al. Lack of pharmacokinetic in-
26. Zhang X, Wang Z, Huang Y, et al. Effects of chronic adminis- teraction between multiple doses of the dipeptidyl peptidase-4
tration of alogliptin on the development of diabetes and b-cell inhibitor alogliptin and digoxin in healthy subjects [abstract no.
function in high fat diet/streptozotocin diabetic mice. Diabetes PI-13]. Clin Pharmacol Ther. 2008;83(Suppl 1):S12–3.
Obes Metab. 2011;13(4):337–47. 43. Karim A, Fleck P, Harris S, et al. Assessment of drug interaction
27. Fleck P, Christopher R, Covington P. Efficacy and safety of between alogliptin, a highly selective dipeptidyl peptidase-4 in-
alogliptin monotherapy over 12 weeks in patients with type 2 hibitor, and atorvastatin in healthy subjects [abstract no. PI-17].
diabetes [abstract no. 479-P]. Diabetes. 2008;57(Suppl 1):143. Clin Pharmacol Ther. 2008;83(Suppl 1):S14.
28. Pratley RE, Reusch JE-B, Fleck PR, et al. Efficacy and safety of the 44. Karim A, Chiselko P, Fleck P, et al. Lack of effect of cy-
dipeptidyl peptidase-4 inhibitor alogliptin added to pioglitazone in closporine on the single-dose pharmacokinetics of alogliptin, a
patients with type 2 diabetes: a randomized, double-blind, placebo- novel dipeptidyl peptidase-4 inhibitor, in healthy subjects [ab-
controlled study. Curr Med Res Opin. 2009;25(10):2361–71. stract no. PI-15]. Clin Pharmacol Ther. 2008;83(Suppl 1):S13.
29. Rosenstock J, Rendell MS, Gross JL, et al. Alogliptin added to 45. Karim A, Copa A, Fleck P, et al. Effects of alogliptin on the
insulin therapy in patients with type 2 diabetes reduces HbA1C pharmacokinetics and pharmacodynamics of norethindrone and
without causing weight gain or increased hypoglycaemia. Dia- ethinyl estradiol (Ortho-NovumÒ 1/35) in healthy adult female
betes Obes Metab. 2009;11(12):1145–52. subjects [abstract no. PI-16]. Clin Pharmacol Ther.
30. Pratley RE, Kipnes MS, Fleck PR, et al. Efficacy and safety of the 2008;83(Suppl 1):S14.
dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 46. DeFronzo RA, Fleck PR, Wilson CA, et al. Efficacy and safety of
diabetes inadequately controlled by glyburide monotherapy. the dipeptidyl peptidase-4 inhibitor alogliptin in patients with
Diabetes Obes Metab. 2009;11(2):167–76. type 2 diabetes and inadequate glycemic control: a randomized,
31. Noda Y, Miyoshi T, Oe H, et al. Alogliptin ameliorates post- double-blind, placebo-controlled study. Diabetes Care.
prandial lipemia and postprandial endothelial dysfunction in non- 2008;31(12):2315–7.
diabetic subjects: a preliminary report. Cardiovasc Diabetol. 47. Ji Q, Han P, Li C, et al. Efficacy and safety of alogliptin in
2013;12:8. subjects with type 2 diabetes: a randomised, double-blind,
Alogliptin: A Review 795

placebo-controlled, phase 3 study in mainland China, Taiwan and 65. Pratley RE. Alogliptin: a new, highly selective dipeptidyl pepti-
Hong Kong [abstract no. 915]. In: 49th Annual Meeting of the dase-4 inhibitor for the treatment of type 2 diabetes. Expert Opin
European Association for the Study of Diabetes. 2013. Pharmacother. 2009;10(3):503–12.
48. Rosenstock J, Inzucchi SE, Seufert J, et al. Initial combination 66. Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and car-
therapy with alogliptin and pioglitazone in drug-naive patients diovascular outcomes in patients with type 2 diabetes mellitus.
with type 2 diabetes. Diabetes Care. 2010;33(11):2406–8. N Engl J Med. 2013;369(14):1317–26.
49. Bosi E, Ellis GC, Wilson CA, et al. Alogliptin as a third oral 67. McMurray JJV, Ponikowski P, Bolli GB, et al. The Vildagliptin
antidiabetic drug in patients with type 2 diabetes and inadequate in Ventricular Dysfunction Diabetes trial (VIVIDD) [late-break-
glycaemic control on metformin and pioglitazone: a 52-week, ing abstract]. In: Heart Failure 2013. 2013.
randomized, double-blind, active-controlled, parallel-group 68. Green JB, Bethel MA, Paul SK, et al. Rationale, design, and
study. Diabetes Obes Metab. 2011;13(12):1088–96. organization of a randomized, controlled Trial Evaluating Car-
50. DeFronzo RA, Burant CF, Fleck P, et al. Efficacy and tolerability diovascular Outcomes with Sitagliptin (TECOS) in patients with
of the DPP-4 inhibitor alogliptin combined with pioglitazone, in type 2 diabetes and established cardiovascular disease. Am Heart
metformin-treated patients with type 2 diabetes. J Clin En- J. 2013;166(6):983–9.
docrinol Metab. 2012;97(5):1615–22. 69. Rosenstock J, Marx N, Kahn SE, et al. Cardiovascular outcome
51. Rosenstock J, Wilson C, Fleck P. Alogliptin versus glipizide trials in type 2 diabetes and the sulphonylurea controversy: ra-
monotherapy in elderly type 2 diabetes mellitus patients with tionale for the active-comparator CAROLINA trial. Diabetes
mild hyperglycaemia: a prospective, double-blind, randomized, Vasc Dis Res. 2013;10(4):289–301.
1-year study. Diabetes Obes Metab. 2013;15(10):906–14. 70. Katakami N, Mita T, Yoshii H, et al. Rationale, design, and
52. Del Prato S, Camisasca R, Wilson C, et al. Durability of the baseline characteristics of a trial for the prevention of diabetic
efficacy and safety of alogliptin compared with glipizide in type 2 atherosclerosis using a DPP-4 inhibitor: the Study of Preventive
diabetes mellitus: a 2-year study. Diabetes Obes Metab. Effects of Alogliptin on Diabetic Atherosclerosis (SPEAD-A).
2014;16(12):1239–46. J Atheroscler Thromb. 2013;20(12):893–902.
53. Kaku K, Mori M, Kanoo T, et al. Efficacy and safety of alogliptin 71. Singh S, Chang H-Y, Richards TM, et al. Glucagonlike peptide
added to insulin in Japanese patients with type 2 diabetes: a 1-based therapies and risk of hospitalization for acute pancreatitis
randomized, double-blind, 12-week, placebo-controlled trial fol- in type 2 diabetes mellitus: a population-based matched case-
lowed by an open-label, long-term extension phase. Expert Opin control study. JAMA Intern Med. 2013;173(7):534–9.
Pharmacother. 2014;15(15):2121–30. 72. Faillie J-L, Babai S, Crépin S, et al. Pancreatitis associated with
54. Nakamura Y, Inagaki M, Shimizu T, et al. Long-term effects of the use of GLP-1 analogs and DPP-4 inhibitors: a case/non-case
alogliptin benzoate in hemodialysis patients with diabetes: a study from the French Pharmacovigilance Database. Acta Dia-
2-year study. Nephron Clin Pract. 2013;123(1–2):46–51. betol. 2014;51(3):491–7.
55. Fujii Y, Abe M, Higuchi T, et al. The dipeptidyl peptidase-4 73. Scheen A. Gliptins (dipeptidyl peptidase-4 inhibitors) and risk of
inhibitor alogliptin improves glycemic control in type 2 diabetic acute pancreatitis. Expert Opin Drug Saf. 2013;12(4):545–57.
patients undergoing hemodialysis. Expert Opin Pharmacother. 74. Butler AE, Campbell-Thompson M, Gurlo T, et al. Marked ex-
2013;14(3):259–67. pansion of exocrine and endocrine pancreas with incretin therapy
56. White WB, Cannon CP, Heller SR, et al. Alogliptin after acute in humans with increased exocrine pancreas dysplasia and the
coronary syndrome in patients with type 2 diabetes. N Engl J potential for glucagon-producing neuroendocrine tumors. Dia-
Med. 2013;369(14):1327–35. betes. 2013;62(7):2595–604.
57. Zannad F, Cannon CP, Cushman WC, et al. Heart failure and 75. Yang L, He Z, Tang X, et al. Type 2 diabetes mellitus and the risk
mortality outcomes in patients with type 2 diabetes taking of acute pancreatitis: a meta-analysis. Eur J Gastroenterol
alogliptin versus placebo in EXAMINE: a multicentre, ran- Hepatol. 2013;25(2):225–31.
domised, double-blind trial. Lancet. 2015. doi:10.1016/S0140- 76. Brodovicz KG, Kou TD, Alexander CM, et al. Impact of diabetes
6736(14)62225-X. duration and chronic pancreatitis on the association between type
58. Data on file, Takeda, 2014. 2 diabetes and pancreatic cancer risk. Diabetes Obes Metab.
59. Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of 2012;14(12):1123–8.
hyperglycemia in type 2 diabetes: a patient-centered approach. 77. Monami M, Dicembrini I, Martelli D, et al. Safety of dipeptidyl
Position statement of the American Diabetes Association (ADA) peptidase-4 inhibitors: a meta-analysis of randomized clinical
and the European Association for the Study of Diabetes (EASD). trials. Curr Med Res Opin. 2011;27(Suppl 3):57–64.
Diabetes Care. 2012;35(6):1364–79. 78. Gokhale M, Sturmer T, Gray C, et al. Dipeptidyl peptidase 4
60. Standards of medical care in diabetes: 2013. Diabetes Care. inhibitors and comparative pancreatic cancer risk [abstract no.
2013;36 Suppl 1:S11–66. 111-LB]. In: 73rd Scientific Sessions of the American Diabetes
61. Garber AJ, Abrahamson MJ, Barzilay JI, et al. AACE compre- Association. 2013.
hensive diabetes management algorithm 2013. Endocr Pract. 79. Richard KR, Shelburne JS, Kirk JK. Tolerability of dipeptidyl
2013;19(2):327–36. peptidase-4 inhibitors: a review. Clin Ther.
62. Charbonnel B, Schweizer A, Dejager S. Combination therapy 2011;33(11):1609–29.
with DPP-4 inhibitors and insulin in patients with type 2 diabetes 80. Monami M, Dicembrini I, Mannucci E. Dipeptidyl peptidase-4
mellitus: what is the evidence? Hosp Pract (Minneap). inhibitors and pancreatitis risk: a meta-analysis of randomized
2013;41(2):93–107. clinical trials. Diabetes Obes Metab. 2014;16(1):48–56.
63. Berhan A, Berhan Y. Efficacy of alogliptin in type 2 diabetes 81. Faillie JL, Azoulay L, Patenaude V, et al. Incretin based drugs
treatment: a meta-analysis of randomized double-blind controlled and risk of acute pancreatitis in patients with type 2 diabetes:
studies. BMC Endocrine Disord. 2013;13:9. cohort study. BMJ. 2014;348:g2780.
64. Bourdel-Marchasson I, Schweizer A, Dejager S. Incretin thera- 82. Egan AG, Blind E, Dunder K, et al. Pancreatic safety of incretin-
pies in the management of elderly patients with type 2 diabetes based drugs: FDA and EMA assessment. N Engl J Med.
mellitus. Hosp Pract. 2011;39(1):7–21. 2014;370(9):794–7.
796 G. M. Keating

83. Monami M, Dicembrini I, Antenore A, et al. Dipeptidyl pepti- 85. Bron M, Ferrufino CP, Samyshkin Y, et al. Evaluation of the
dase-4 inhibitors and bone fractures: a meta-analysis of ran- pharmacy budget impact of alogliptin plus pioglitazone fixed
domized clinical trials. Diabetes Care. 2011;34(11):2474–6. dose combination in the treatment of type-2 diabetes mellitus
84. Marino AB, Cole SW. Alogliptin: safety, efficacy, and clinical [abstract no. PDB25]. Value Health. 2013;16(3):A160.
implications. J Pharm Pract. 2015;28(1):99–106.

Potrebbero piacerti anche