You are on page 1of 10

diabetes research and clinical practice 176 (2021) 108848

Contents available at ScienceDirect

Diabetes Research
and Clinical Practice
journal homepage: www.elsevier.com/locat e/dia bre s

Comparison of insulin glargine 300 U/mL versus


glargine 100 U/mL on glycemic control and
hypoglycemic events in East Asian patients with
type 2 diabetes: A Patient-level meta-analysis of
phase 3 studies

Linong Ji a,*, Yan Bi b, Shandong Ye c, Yun Huang d, Xia Zhang d, Shuhua Shang d, Nan Cui d,
Huiqiu Yin d, Minlu Zhang d
a
Peking University People’s Hospital, China
b
Drum Tower Hospital Affiliated to Nanjing University Medical School, China
c
The First Affiliated Hospital of University of Science and Technology of China, China
d
Sanofi, Shanghai, China

A R T I C L E I N F O A B S T R A C T

Article history: Aims: To evaluate efficacy and safety of Gla-300 with Gla-100 in a patient-level meta-
Received 10 February 2021 analysis among large East Asian patients with type 2 diabetes mellitus (T2DM).
Received in revised form Methods: A patient level meta-analysis of three EDITION studies with similar design and
23 April 2021 endpoints were conducted over 6-months treatment period. The analysis included 547
Accepted 28 April 2021 patients treated with Gla-300 and 348 patients treated with Gla-100.
Available online 1 May 2021 Results: Over 6-month treatment period, mean change in HbA1c was similar for Gla-300
[Least square (LS) mean, (SE): 1.13 (0.05) % and Gla-100: 1.14 (0.05) %], showing non-
inferiority of Gla-300 to Gla-100 (LS mean difference: 0.02%, 95% CI: 0.08 to 0.11). Gla-
Keywords:
300 was associated with reduced risk of hypoglycemic event (confirmed  3.9 mmol/L or
Insulin glargine
severe) vs Gla-100 at any time of day or at night (00:00–05:59 h). The event rates of hypo-
EDITION
glycemia were consistently lower with Gla-300 than Gla-100. Severe hypoglycemia was rare
Asians
in both treatment groups. Weight gain was minimal in both treatment groups.
HbA1c
Conclusion: Gla-300 provides comparable glycemic control to Gla-100 in East Asian patients
Diabetes
with broad clinical spectrum of T2DM, with consistently less hypoglycemia at any time of
Pooled analysis
the day and night.
Ó 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY
license (http://creativecommons.org/licenses/by/4.0/).

* Corresponding author at: Department of Endocrinology and Metabolism, Peking University People’s Hospital, 11 Xizhimen South
Street, Xicheng District, Beijing 100044, China.
E-mail address: jiln@bjmu.edu.cn (L. Ji).
https://doi.org/10.1016/j.diabres.2021.108848
0168-8227/Ó 2021 The Authors. Published by Elsevier B.V.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
2 diabetes research and clinical practice 176 (2021) 108848

1. Introduction A comprehensive evaluation of the efficacy of insulin glargine


in East Asians is desirable to make better informed decisions
The prevalence of diabetes is increasing globally, and also in the management of the disease. In this study, we present
there is an alarming increase in Asia [1]. Based on the latest the results of a patient-level meta-analysis based on the three
International Diabetes Federation (IDF) report, the regional EDITION studies which compared the efficacy of Gla-300 over
prevalence was 9.6% in Western Pacific and 8.8% in South East Gla-100 during 6 months in East Asian population with T2DM.
Asia [1]. China and Japan are among the top five global coun-
tries with an increasing number of people with diabetes (116.4 2. Materials and Methods
million and 7.4 million respectively) [1]. The management of
type 2 diabetes (T2DM) involves various approaches such as 2.1. Study design and participants
changes in the lifestyle and use of pharmacological interven-
tions for maintaining glycemic control. However, as the dis- This patient level meta-analysis included participants from
ease progresses, exogenous insulin therapy is required in three studies [27–29] (NCT01689142, NCT02855684 and
many patients. NCT01676220). All these three clinical trials were multicenter,
American Diabetes Association (ADA) guidelines recom- randomized, open-label, two-arm, parallel-group, phase III
mends basal insulin alone as the most convenient initial study described previously [27–29]. The inclusion/exclusion
insulin regimen and can be added to metformin and other criteria have been described in the individual studies previ-
oral agents [2]. Early introduction of insulin should be consid- ously [27–29]. In brief, all patients were aged  18 years with
ered if there is evidence of ongoing catabolism, symptoms of a diagnosis of T2DM (according to WHO criteria) [30] EDITION
hyperglycemia or when HbA1c levels (>10%) or blood glucose JP2 was conducted in Japanese patients and on basal insulin
levels (300 mg/dL) are very high [2]. Similarly, the Chinese (BI) with OAD(s), while EDITION AP and EDITION 3 was con-
Diabetes Society (CDS) and Korean Diabetes Association ducted in insulin-naı̈ve patients with T2DM not adequately
guidelines for T2DM recommends initiation of insulin therapy controlled with non-insulin antihyperglycemic drugs. Fur-
within 3 months of failure to achieve blood glucose target ther, EDITION AP included patients from Taiwan, Mainland
(<7% or < 6.5%) and on treatments with OAD(s) and lifestyle China, Korea, and only Japanese patients were included for
interventions. Further, intensive insulin treatment may be the analysis from EDITION 3 though the study was conducted
implemented in newly diagnosed patients with T2DM with worldwide. The main exclusion criteria included glycated
HbA1c > 9.0% or FPG  11.1 mmol/L and symptomatic hyper- haemoglobin levels (HbA1c) < 7.0% at screening for all three
glycaemia with or without OADs [3,4]. studies, HbA1c > 11.0% for EDITION 3 and AP, and > 10.0%
Abundance of studies have demonstrated that timely for EDITION JP2. All the studies included a 2-week screening
addition of insulin therapy in patients with T2DM could pro- phase, a 6-month treatment period and a 6-month safety
vide pronounced clinical improvements in glycemic control extension period; however, data from the first 6-month treat-
and improve patient satisfaction and quality of life [5–9]. ment period are reported here.
However, despite the proven efficacy of insulin therapy, evi- The included patients were randomized (1:1) to receive
dence suggests that insulin treatment is suboptimal in the once-daily subcutaneous injections of Gla-300 or Gla-100 in
current clinical practice in Asia due to issues such as delay EDITION JP2 and 3, while in EDITION AP, patients were ran-
in insulin initiation, inadequate insulin titration and rela- domized at a ratio of 2:1. Both the injections were self-
tively poor glucose control [10,11]. The reasons are complex administered at the same time each evening throughout the
and include both physician- and patient-related factors, 6-month treatment period. All participants were titrated to
among which concerns of hypoglycemia remains one of the a fasting self-monitored plasma glucose (SMPG) target of
main barriers [12–14]. 4.4–5.6 mmol/l (80–100 mg/dl), with adjustments made at
Insulin glargine 300 U/mL (Gla-300), as a new generation of least once per week, but no more often than every 3–4 days,
basal insulin analogue, provides a constant and prolonged in the absence of hypoglycemia. A detailed titration algorithm
pharmacokinetic and pharmacodynamic (PK/PD) profile com- for individual studies is listed in the Table S1. All the studies
pared with insulin glargine 100 U/mL (Gla-100), with a longer were approved by the institutional review boards and ethics
duration of action and more stable glucose-lowering activities committees of the participating centers, and were conducted
[15,16]. in accordance with the Declaration of Helsinki and Good Clin-
Various EDITION studies have reported comparable glyce- ical Practice guidelines.
mic control of Gla-300 versus Gla-100 in broad Caucasian/
non-Asian populations, with consistently lower risk of hypo- 2.2. Endpoints and outcomes
glycemia at any time during the day and at night in patients
with type 2 diabetes [17–22]. Owing to physiological and The efficacy outcomes of this study were to assess change in
pathological differences, studies have reported some differ- HbA1c from baseline to month 6; change in average pre-
ences in treatment response including glycemic control and breakfast SMPG (recognized as fasting SMPG) from baseline
hypoglycemia in Asians and non-Asians previously [23–26]. to month 6; change in laboratory-measured fasting plasma
Thus, the efficacy of two insulin glargine Gla-300 and Gla- glucose (FPG) from baseline to month 6; percentage of partic-
100 in large East Asian population is worth exploration. ipants achieving HbA1c < 7.0%; percentage of participants
diabetes research and clinical practice 176 (2021) 108848 3

achieving HbA1c < 7.0% without experiencing hypoglycaemia; was found for endpoint of weight change; hence a random-
eight-point SMPG profile, change in mean 24-hour plasma effect meta-analysis was conducted for weight change with
glucose (PG), variability of mean 24-hour PG, change in BI dose study-specific estimations using inverse-variance weights in
from baseline to month 6 of treatment period. random-effect model.
The safety endpoints included percentages of participants Efficacy endpoints used the modified intention-to-treat
having at least one nocturnal (00:00–05:59 h) hypoglycaemic (mITT) population, which included all randomized patients
event or hypoglycaemic event at any time of day (24 h) and who received 1 dose of the open label study drugs and had
annualized rates (events per participant-year), by study per- both a baseline assessment and 1 post baseline assessment
iod, and the cumulative mean number of hypoglycaemic of any primary or secondary efficacy variables. The safety
events per participant. All possible hypoglycaemic events population included all participants randomized and exposed
were recorded and categorized according to the definitions to 1 dose of study drug.
recommended by the ADA 2005 Working Group on Hypogly-
caemia [31]. ‘Documented symptomatic hypoglycaemia’ was 3. Results
defined as an event during which typical symptoms of hypo-
glycaemia are confirmed by a measured plasma glucose con- 3.1. Baseline characteristics
centration of 3.9 mmol/l (70 mg/dl)], asymptomatic
hypoglycaemia confirmed by a measured plasma glucose con- Of the 895 patients included in the pooled analysis of all three
centration of 3.9 mmol/l (70 mg/dl) and ‘severe hypogly- EDITION studies [27–29], 547 were randomized to Gla-300 and
caemia’ as events requiring assistance by another person to 348 were randomized to Gla-100. The mITT and safety popu-
administer carbohydrate, glucagon or other therapy. For the lation comprised 542 patients treated with Gla-300 and 346
main analysis of hypoglycaemic outcomes, the confirmed patients treated with Gla-100 respectively. The baseline
(with or without symptoms) and severe categories were com- demographic characteristics from the individual study popu-
bined, and the hypoglycaemic events with a plasma glucose lations and the pooled analysis population (all three studies)
measurement of <3.0 mmol/l (<54 mg/dl) were also analyzed. are shown in Table 1.
Other safety endpoints included change in body weight and
adverse events. 3.2. Glycemic control

2.3. Statistical analysis In the pooled dataset of the three studies, the mean change in
HbA1c from baseline to month-6 was similar in both treat-
Change from baseline in HbA1c and all other continuous vari- ment groups [Least square (LS) mean, standard error (SE):
ables were analyzed using analysis of covariance (ANCOVA) Gla-300 vs Gla-100: 1.13 (0.05) % vs. 1.14 (0.05) %] showing
model with treatment group (Gla-300 and Gla-100), screening non-inferiority of Gla-300 to Gla-100 [LS mean difference
HbA1c (<8.0, 8.0%), use of sulfonylurea or glinides (Yes and (LSMD), 95% confidence interval (CI): 0.02% (-0.08 to 0.11)
No) and studies (EDITION 3, EDITION JP2, EDITION AP) as fixed (Fig. 1). The proportion of patients achieving an HbA1c target
effects and baseline measure as a covariate. In case, a partic- of <7.0% at month 6 were 46.4% (248/535) for Gla-300 and
ipant discontinued treatment prematurely or had no efficacy 43.1% (147/341) for Gla-100, with 42.4% (227/535) and 37.0%
measurement at month 6, the last post-baseline on- (126/341) achieved this target without hypoglycemia (con-
treatment measurement was used as the efficacy value at firmed <3.0 mmol/l or severe). Similar trend of results was
month 6 (last observation carried forward). demonstrated in patients achieving HbA1c target of <6.5%
The percentage of participants reporting 1 hypogly- (Table S2).
caemic event and all other categorical variables were ana- The laboratory measured fasting plasma glucose (FPG)
lyzed using the Cochran–Mantel–Haenszel method stratified decreased in both groups at month 6 from baseline [LS mean
by randomization strata of screening HbA1c (<8.0 and (SE): Gla-300: 2.86 mmol/L (0.10) vs. Gla-100: 2.99 mmol/L
8.0%), use of sulfonylurea or glinides (Yes and No) and stud- (0.11)] with LSMD (95% CI) between treatment groups of
ies (EDITION 3; EDITION JP2; EDITION AP) and the relative risk 0.13 mmol/L (-0.10 to 0.35) (Fig. 2A).
with 95% CI was estimated. The annualized rates of hypogly- The mean change in pre-breakfast SMPG values also
caemia (events per participant-year) were analyzed using an showed comparable reduction in both treatment groups from
over dispersed Poisson regression model stratified by ran- baseline to month 6 [LS mean (SE): Gla-300: 2.26 mmol/l
domization strata of screening HbA1c (<8.0 and 8.0%), use (0.08) vs. Gla-100: 2.40(0.08)], LSMD (95% CI) between treat-
of sulfonylurea or glinides (yes; no) and studies (EDITION 3; ment groups: 0.14 (-0.03 to 0.30) (Fig. 2B). The eight-point
EDITION JP2; EDITION AP) and rate ratio with 95% CI was esti- SMPG profiles in both groups showed a marked decrease from
mated. Cumulative mean number of hypoglycaemic events by baseline to month 6 (Fig. 2C), with the LS mean change in 24-
participant was analysed using Nelson-Aelen estimates. hour average SMPG values being similar between groups
Adverse events were analysed descriptively. For each end- (Table S2). There was also no between-treatment difference
point, homogeneity of treatment effect across all three stud- in the variability of 24-hour SMPG at month 6 [LS mean (SE):
ies was assessed by testing the study-by-treatment Gla-300: 4.86% (0.58) vs. Gla-100: 4.16% (0.62); LSMD (95% CI)
interaction for each endpoint before pooling. Homogeneity between two groups: 0.69% (-0.58 to 1.96) (Table S2).
4
Table 1 – Baseline demographic characteristics for individual studies and pooled analysis of all three studies (randomized population).
Patient-level pooled analysis EDITION 3 EDITION AP EDITION JP2
Gla-300 Gla-100 Gla-300 Gla-100 Gla-300 Gla-100 Gla-300 Gla-100
N = 547 N = 348 N = 25 N = 25 N = 401 N = 203 N = 121 N = 120

diabetes research and clinical practice


Age, years 59.1 (9.9) 59.1 (10.8) 59.7 (9.6) 62.0 (7.6) 58.5 (9.6) 57.9 (10.2) 61.1 (10.8) 60.5 (12.0)
Gender: male, n (%) 325 (59.4) 195 (56.0) 14 (56.0) 17 (68.0) 234 (58.4) 108 (53.2) 77 (63.6) 70 (58.3)
Body weight, kg 68.0 (12.3) 67.8 (12.1) 67 (15) 68.6 (13.3) 68.3 (11.7) 68.8 (11.5) 67.4 (13.6) 65.9 (12.8)
BMI, kg/m2 25.3 (3.5) 25.2 (3.4) 25.6 (4.8) 26.0 (4.1) 25.2 (3.2) 25.3 (3.2) 25.7 (4.0) 24.8 (3.6)
eGFR, mL/min/1.73 m2 88.0 (20.3) 86.8 (19.6) 88 (15.6) 83.8 (18.1) 90.4 (20.5) 88.7 (20.7) 80.2 (18.8) 84.2 (17.7)
C-peptide, nmol/l 0.54 (0.28) 0.50 (0.27) 0.56 (0.34) 0.43 (0.17) 0.59 (0.23) 0.61 (0.26) 0.38 (0.28) 0.32 (0.20)
Region
Japan 146 (26.7) 145 (41.7) 25 (100.0) 25 (100.0) – – 121(100.0) 120 (100.0)
China 315 (57.6) 159 (45.7) – – 315 (78.6) 159 (78.3) – –
South Korea 77 (14.1) 34 (9.8) – – 77 (19.2) 34 (16.7) – –
Taiwan 9 (1.7) 10 (2.9) – – 9 (2.2) 10 (4.9) – –
Duration of diabetes, years 11.4 (7.0) 11.8 (7.2) 9.9 (7.6) 11.6 (7.5) 10.7 (6.4) 10.5 (5.8) 14.0 (8.0) 13.9 (8.7)
Duration of basal insulin treatment, years 2.39 (2.33)† 2.52 (2.44)† – – – – 2.39 (2.33) 2.52 (2.44)
Prior use of insulin glargine, n(%) 118 (98.3)*,† 110 (91.7)*,† – – – – 118 (98.3)* 110 (91.7)*
Prior non-insulin anti-hyperglycaemic treatment, n (%)

176 (2021) 108848


Biguanides 381 (69.7) 240 (66.7) 18 (72.0) 18 (72.0) 293 (73.1) 151 (74.4) 70 (57.9) 71 (59.2)
Sulphonylureas 314 (57.4) 216 (60.0) 18 (72.0) 20 (80.0) 232 (57.9) 130 (64.0) 64 (52.9) 66 (55.0)
Alpha-glucosidase inhibitors 195 (35.6) 94 (27.0) 6 (24.0) 9 (36.0) 146 (36.4) 59 (29.1) 43 (35.5) 26 (21.7)
DPP-4 inhibitors 133 (24.3) 116 (32.2) 17 (68.0) 20 (80.0) 65 (16.2) 43 (21.2) 51 (42.1) 53 (44.2)
Thiazolidinediones 51 (9.3) 39 (10.8) 1 (4.0) 7 (28.0) 38 (9.5) 23 (11.3) 12 (9.9) 9 (7.5)
SGLT-2 inhibitors 12 (2.2) 9 (2.5) 0 (0.0) 0 (0.0) 12 (3.0) 9 (4.4) 0 (0.0) 0 (0.0)
Glinides 50 (9.1) 22 (6.1) 1 (4.0) 0 (0.0) 38 (9.5) 10 (4.9) 11 (9.1) 12 (10.0)
Fixed-dose oral combination drugs 36 (6.6) 16 (4.4) 0 (0.0) 0 (0.0) 30 (7.5) 13 (6.4) 6 (5.0%) 3 (2.5%)
HbA1c, % 8.4 (0.9) 8.3 (0.9) 8.0 (0.7) 8.0 (0.7) 8.6 (0.9) 8.5 (1.0) 8.0 (0.7) 8.1 (0.8)
HbA1c, mmol/mol 68.6 (10.1) 67.5 (10.1) 64.2 (8.0) 63.7 (7.4) 70.5 (9.8) 69.4 (10.9) 63.8 (7.9) 64.6 (8.4)
*
N = 120.

Excludes participants from EDITION 3 and EDITION AP (insulin-naive).
diabetes research and clinical practice 176 (2021) 108848 5

3.3. Hypoglycaemia

Over the 6-month treatment period, the number of partici-


pants experiencing confirmed (3.9 mmol/L [70 mg/dL]) or
severe hypoglycaemia was lower on receiving Gla-300 than
Gla-100 at any time of the day (24 h) (66.6% vs. 74.0%; relative
risk: 0.90; 95% CI: 0.83 to 0.98). Similarly, the number of partic-
ipants with nocturnal confirmed [3.9 mmol/l (70 mg/dl)] or
severe hypoglycaemic events was lower with Gla-300 than
Gla-100 (34.5% vs. 44.5%; relative risk: 0.76; 95% CI: 0.64 to
0.90) (Fig. 3 and Table S3). When analyzed by study period,
the reduction in incidence with Gla-300 compared with Gla-
100 was even apparent during the first 12 weeks of treatment.
The annualized rate (events per participant-year) of con-
Fig. 1 – HbA1c (mean ± SE) over the 6-month treatment firmed [3.9 mmol/l (70 mg/dl)] or severe hypoglycaemia
period (mITT population). at any time of day over the 6-month study period was 8.74
with Gla-300 and 11.83 with Gla-100 (rate ratio: 0.79, 95% CI:
0.65 to 0.96), showing a relative difference in rate of 21% in
favour of Gla-300 (Fig. 3). The annualized rates of nocturnal

Fig. 2 – (A) Laboratory-measured fasting plasma glucose (FPG) (B) Pre-breakfast Self-monitored plasma glucose (SMPG) (C)
Eight-point SMPG profile and (D) Insulin dose during the 6-month treatment period for pooled analysis of all three studies
(mITT population).
6 diabetes research and clinical practice 176 (2021) 108848

Fig. 3 – Percentage of participants with  1 hypoglycaemic event and annualized event rate (events per participant-year) over
the 6-month treatment period (Safety population). Abbreviations: RR, relative risk for percentage of participants with 1
event, rate ratio for annualized event rates; Gla-100, insulin glargine 100 U/ml, Gla-300, insulin glargine 300 U/ml.

events over the 6-month study period were lower with Gla- and to 0.30 (0.14) U/kg [20.7 (11.1) U] with Gla-100. The major-
300 than Gla-100 (2.26 vs. 3.27; rate ratio: 0.74; 95% CI: 0.53 ity of increase in insulin dose occurred during the first
to 1.04), with a relative difference of 26% in favour of Gla- 12 weeks of treatment (Fig. 2D).
300 (Fig. 3). The cumulative number of confirmed
(3.9 mmol/l) or severe hypoglycaemic events per participant 3.5. Adverse events
was lower with Gla-300 compared with Gla-100 both at any
time and at night (Fig. 4). The percentages of participants experiencing treatment
Similar trends of reduction in hypoglycaemic events were emergent adverse events (TEAEs) were comparable for Gla-
observed across other hypoglycaemia definitions, including 300 and Gla-100 [52.2% (283/542) vs 54.3% (188/346)]. Serious
documented symptomatic hypoglycaemia (3.9 mmol/l), adverse events were infrequent, reported in 28 participants
and confirmed or severe hypoglycaemia at the lower thresh- (5.2%) in Gla-300 group and 18 participants (5.2%) in Gla-100
old of < 3.0 mmol/L (<54 mg/dL) (Table S3). The distribution group.
of annualized rates of confirmed (3.9 mmol/l) or severe Injection site reactions were rare and observed among 10
hypoglycaemic events over 24 h is reported in Fig. 5. A lower (1.8%) participants in Gla-300 group and 2 (0.6%) in the Gla-
rate of hypoglycaemia was shown during night and beyond 100 group. Overall, 11 patients discontinued treatment due
the predefined nocturnal period (00:00–05:59 h) with Gla-300 to adverse events, 8 (1.5%) in Gla-300 and 3 (0.9%) in Gla-
compared with Gla-100. Hypoglycemic events were most fre- 100, respectively, while two patients (0.4%) in Gla-300 and 1
quently reported between 04:00 and 08:00 h. patient (0.3%) in Gla-100 had TEAEs leading to death
Severe hypoglycemia was rare in both the treatment (Table S4).
groups. The number of participants experiencing severe
hypoglycaemia at any time of day was 4 (0.7%) with Gla-300 4. Discussion
and 3 (0.9%) with Gla-100 (relative risk: 1.15 95% CI: 0.28 to
4.76) (Table S3). Studies in several Western populations with type 2 diabetes in
the EDITION programs have shown that treatment with Gla-
3.4. Weight gain and insulin dose 300 was associated with comparable glycemic control but
reduced incidence of hypoglycaemia than Gla-100 [17,18,29].
There was a slight weight gain with Gla-300 and Gla-100 (LS In this present pooled analysis, treatment with Gla-300 was
mean (SE): 0.40 (0.86) kg vs 0.65 (0.44) kg, respectively). LS shown to be as effective as Gla-100 in improving glycaemic
mean difference in weight change for Gla-300 versus Gla- control over 6 months, and was accompanied by a lower risk
100 was 0.26 kg; (95% CI: 1.19 to 0.68). Mean daily basal of confirmed (3.9 mmol/l) or severe hypoglycemia at any
insulin dose was comparable at baseline between Gla-300 time during the day and at night. Weight gain was observed
[mean (SD) 0.19 (0.08) U/kg [13.09 (6.05) U] and Gla-100 [0.20 low in both groups (<1kg). The present meta-analysis also
(0.08) U/kg [13.56 (5.96) U]. The daily basal insulin dose showed that the percentage of patients achieving HbA1c < 7%
increased in both treatment groups at month 6 from baseline, without confirmed or severe hypoglycaemia was higher with
to a mean (SD) of 0.35 (0.18) U/kg [24.4 (13.7) U] with Gla-300 Gla-300 vs Gla-100. No new adverse events were identified
diabetes research and clinical practice 176 (2021) 108848 7

Fig. 4 – Cumulative mean number of confirmed [3.9 mmol/l (70 mg/dl)] or severe hypoglycaemic events (A) at any time of
day (24 h) and (B) during the night (00.00–05.59 h) for pooled analysis of all three studies (safety population). Gla-100, insulin
glargine 100 U/ml; Gla-300, insulin glargine 300 U/ml.

during this 6-month analysis and were comparable between Gla-300 were observed (26% and 24% respectively during the
the two treatment groups. entire 6 months of treatment). Nocturnal hypoglycemia is
In the overall pooled analysis, the relative difference in worrisome and need to be drawn attention, as it is often
rate of confirmed [3.9 mmol/l (70 mg/dl)] or severe hypo- undetected and may lead to unconsciousness and even death
glycaemia at any time of day was 21% in favour of Gla-300 in severe cases [32]. This important finding reveals the advan-
over 6 months of treatment period. The corresponding differ- tage of using Gla-300 for better management of nocturnal
ence in the individual studies were 36% for EDITION JP2, 12% hypoglycemia. Our hypoglycemic findings are in accordance
and 25% for EDITION AP and EDITION 3 respectively [27–29]. with the real-world evidence studies which report lower
These findings suggest that the pooled analysis results were hypoglycemic events with Gla-300 over other insulin ana-
mainly driven by the size of the reductions observed in EDI- logues [33,34].
TION JP2 study [28]. One of the possible explanations is that Over the course of 24-hours, it was noted that confirmed
those insulin pre-treated patients who were included in EDI- [3.9 mmol/l (70 mg/dl)] or severe hypoglycemic events were
TION JP2 had rather higher hypoglycaemia risk than insulin reported the most during 04:00 and 08:00 h, a lesser number
naı̈ve patients included in EDITION 3 and AP, allowing more of events with Gla-300 than Gla-100. This lower frequency of
pronounced safety advantages of Gla-300 to be demonstrated. hypoglycemia extended from the early morning till the after-
A greater difference in the annualized rate and incidence of noon (15:59 h) thus favouring treatment to Gla-300. This can
nocturnal confirmed or severe hypoglycaemia in patients on be attributed with the more stable and prolonged action of
8 diabetes research and clinical practice 176 (2021) 108848

Asians. Our study results confirm the consistent glycemic


control effect and reduced hypoglycaemia risk of Gla-300 in
East Asian patients with T2DM, who usually have lower BMI
and demand lower insulin dose compared with Western pop-
ulation. However, this study is not without limitations. The
individual clinical trials were open-label in design that
reflects difficulties in blinding the two insulins, which has
the potential to introduce bias along with a shorter duration
of study period. Besides, the efficacy of Gla-300 in broader
East Asian population with other ethnic, or with various reg-
imens (e.g., basal bolus or switching from premix insulin)
could be further investigated. However, taking into considera-
tion the successful results found in the earlier EDITION stud-
ies on Asian populations and worldwide, initiating insulin
therapy with Gla-300 will be beneficial in the management
Fig. 5 – Number of confirmed (3.9 mmol/L [70 mg/dL]) or of diabetes control.
severe hypoglycaemic events per participant-year by time of
day; safety population. 5. Conclusion

Patients receiving Gla-300 showed comparable glycemic con-


Gla-300 previously described in various PK/PD studies [15,35]
trol compared to patients on Gla-100 in East Asian patients
and is similar to hypoglycaemia risk reduction beyond
with a broad clinical spectrum of T2DM, with consistently
06:00 h already reported in Western type 2 diabetes popula-
less hypoglycemia at any time of the day and at night. These
tions [17,29]. These results highlight that prescribing Gla-300
results show similar efficacy and safety profile of Gla-300 as
over Gla-100 may provide greater stability in patients suffer-
observed in the earlier global EDITION programs. In consider-
ing from hypoglycaemia during daytime in East Asian popula-
ation of these similarities, the present findings may have
tion and thereby help to lessen the impact of diabetes on daily
meaningful clinical implications in successfully starting and
activities.
maintaining treatment with Gla-300.
Further, reduction in the incidences of hypoglycemia as
well as the annualized rates of hypoglycaemia even occurred
during first 12 weeks of treatment, where most of the basal Declaration of Competing Interest
insulin dose titration takes place. Hypoglycaemia occurs fre-
quently during the titration period and hypoglycaemic events The authors declare that they have no known competing
during this period can lead to a higher risk in the longer term. financial interests or personal relationships that could have
Therefore, it is important to minimize the risk of hypogly- appeared to influence the work reported in this paper.
caemia within the initial weeks of starting basal insulin ther- L.J. reports receiving consulting and lecture fees from Eli
apy, which may facilitate to improve treatment adherence Lilly, Bristol-Myers Squibb, Novartis, Novo Nordisk, Merck,
and moreover the long-term clinical outcomes [36,37]. These Bayer, Takeda, Sanofi, Roche and Boehringer Ingelheim and
results are consistent with the BRIGHT treat-to-target trial research grants from Roche and Sanofi. Y.B. reports receiving
randomized controlled trial comparing efficacy of Gla-300 lecture fees from Eli Lilly, Bristol-Myers Squibb, Novartis,
and IDeg-100 which showed a reduced risk of hypoglycaemia Novo Nordisk, Merck, Bayer, Sanofi, Roche and Boehringer
with Gla-300 during 12-week active titration period, along Ingelheim. S.Y. has no conflicts of interests. Y.H., X.Z., S.S,
with majority of dose increase and HbA1c, FPG reductions N.C., H.Y. and M.Z. are employees of Sanofi.
[38].
Increase in basal insulin dose from baseline to month 6 Acknowledgments
was observed in both treatment groups especially during
the first 12 weeks of treatment and quite slight change there- The authors thank the study participants, trial staff and
after. There was higher dose with Gla-300 which was in line investigators for their participation. Editorial and writing
with the observations from the individual EDITION studies. assistance was provided by Anwesha Mandal of Indegene
This may be due to the subcutaneous administration of Gla- Pvt Ltd. Bangalore, India and was funded by Sanofi.
300 which leads to increased residence time, thereby leading
to longer exposure to tissue peptidases and decreasing its Funding
bioavailability [15,39]. Despite the higher dose of Gla-300,
comparable glycemic control with reduced incidence of hypo- This study was funded by Sanofi China.
glycemia were observed.
The strength of this study lies in that three head-to-head Appendix A. Supplementary material
randomized trials with similar study designs were pooled
together, allowing to evaluate the efficacy and safety of Gla- Supplementary data to this article can be found online at
300 compared with Gla-100 in a large population of East https://doi.org/10.1016/j.diabres.2021.108848.
diabetes research and clinical practice 176 (2021) 108848 9

R E F E R E N C E S Diabetes Obes Metab 2018;20:488–96. https://doi.org/


10.1111/dom.13132.
[15] Becker RHA, Dahmen R, Bergmann K, Lehmann A, Jax T,
Heise T. New insulin glargine 300 Units  mL-1 provides a
[1] International Diabetes Federation - Facts & figures n.d.
more even activity profile and prolonged glycemic control at
https://www.idf.org/aboutdiabetes/what-is-diabetes/facts-
steady state compared with insulin glargine 100 Units  mL-1.
figures.html (accessed November 15, 2019).
Diabetes Care 2015;38:637–43. https://doi.org/10.2337/dc14-
[2] Pharmacologic Approaches to Glycemic Treatment:
0006.
Standards of Medical Care in Diabetes 2019 n.d.
[16] Bergenstal RM, Bailey TS, Rodbard D, Ziemen M, Guo H,
[3] Jia W, Weng J, Zhu D, Ji L, Lu J, Zhou Z, et al. Standards of
Muehlen-Bartmer I, et al. Comparison of Insulin Glargine 300
medical care for type 2 diabetes in China 2019. Diabetes
Units/mL and 100 Units/mL in Adults With Type 1 Diabetes:
Metab Res Rev 2019;35. https://doi.org/10.1002/dmrr.3158
Continuous Glucose Monitoring Profiles and Variability Using
e3158.
Morning or Evening Injections. Diabetes Care 2017;40:554–60.
[4] Kim MK, Ko S-H, Kim B-Y, Kang ES, Noh J, Kim S-K, et al. 2019
https://doi.org/10.2337/dc16-0684.
Clinical Practice Guidelines for Type 2 Diabetes Mellitus in
[17] Yki-Järvinen H, Bergenstal R, Ziemen M, Wardecki M,
Korea. Diabetes Metab J 2019;43:398–406. https://doi.org/
Muehlen-Bartmer I, Boelle E, et al. New insulin glargine 300
10.4093/dmj.2019.0137.
units/mL versus glargine 100 units/mL in people with type 2
[5] Lovre D, Fonseca V. Benefits of timely basal insulin control in
diabetes using oral agents and basal insulin: glucose control
patients with type 2 diabetes. J Diabetes Complications
and hypoglycemia in a 6-month randomized controlled trial
2015;29:295–301. https://doi.org/10.1016/j.
(EDITION 2). Diabetes Care 2014;37:3235–43. https://doi.org/
jdiacomp.2014.11.018.
10.2337/dc14-0990.
[6] Blicklé J-F, Hancu N, Piletic M, Profozic V, Shestakova M, Dain
[18] Riddle MC, Bolli GB, Ziemen M, Muehlen-Bartmer I, Bizet F,
M-P, et al. Insulin glargine provides greater improvements in
Home PD, et al. New insulin glargine 300 units/mL versus
glycaemic control vs. intensifying lifestyle management for
glargine 100 units/mL in people with type 2 diabetes using
people with type 2 diabetes treated with OADs and 7–8% A1c
basal and mealtime insulin: glucose control and
levels. The TULIP study. Diabetes Obes Metab 2009;11:379–86.
hypoglycemia in a 6-month randomized controlled trial
https://doi.org/10.1111/j.1463-1326.2008.00980.x.
(EDITION 1). Diabetes Care 2014;37:2755–62. https://doi.org/
[7] Owens DR, Traylor L, Dain M-P, Landgraf W. Efficacy and
10.2337/dc14-0991.
safety of basal insulin glargine 12 and 24 weeks after
[19] Ritzel R, Roussel R, Bolli GB, Vinet L, Brulle-Wohlhueter C,
initiation in persons with type 2 diabetes: a pooled analysis of
Glezer S, et al. Patient-level meta-analysis of the EDITION 1, 2
data from treatment arms of 15 treat-to-target randomised
and 3 studies: glycaemic control and hypoglycaemia with
controlled trials. Diabetes Res Clin Pract 2014;106:264–74.
new insulin glargine 300 U/ml versus glargine 100 U/ml in
https://doi.org/10.1016/j.diabres.2014.08.003.
people with type 2 diabetes. Diabetes Obes Metab
[8] Gerstein HC, Yale J-F, Harris SB, Issa M, Stewart JA, Dempsey
2015;17:859–67. https://doi.org/10.1111/dom.12485.
E. A randomized trial of adding insulin glargine vs. avoidance
[20] Riddle MC, Yki-Järvinen H, Bolli GB, Ziemen M, Muehlen-
of insulin in people with Type 2 diabetes on either no oral
Bartmer I, Cissokho S, et al. One-year sustained glycaemic
glucose-lowering agents or submaximal doses of metformin
control and less hypoglycaemia with new insulin glargine
and/or sulphonylureas. The Canadian INSIGHT
300 U/ml compared with 100 U/ml in people with type 2
(Implementing New Strategies with Insulin Glargine for
diabetes using basal plus meal-time insulin: the EDITION 1
Hyperglycaemia Treatment) Study. Diabet Med
12-month randomized trial, including 6-month extension.
2006;23:736–42. https://doi.org/10.1111/j.1464-
Diabetes Obes Metab 2015;17:835–42. https://doi.org/
5491.2006.01881.x.
10.1111/dom.12472.
[9] Houlden R, Ross S, Harris S, Yale J-F, Sauriol L, Gerstein HC.
[21] Yki-Järvinen H, Bergenstal RM, Bolli GB, Ziemen M, Wardecki
Treatment satisfaction and quality of life using an early
M, Muehlen-Bartmer I, et al. Glycaemic control and
insulinization strategy with insulin glargine compared to an
hypoglycaemia with new insulin glargine 300 U/ml versus
adjusted oral therapy in the management of Type 2 diabetes:
insulin glargine 100 U/ml in people with type 2 diabetes using
the Canadian INSIGHT Study. Diabetes Res Clin Pract
basal insulin and oral antihyperglycaemic drugs: the
2007;78:254–8. https://doi.org/10.1016/j.diabres.2007.03.021.
EDITION 2 randomized 12-month trial including 6-month
[10] Ji L, Zhang P, Zhu D, Li X, Ji J, Lu J, et al. Observational Registry
extension. Diabetes Obes Metab 2015;17:1142–9. https://doi.
of Basal Insulin Treatment (ORBIT) in patients with type 2
org/10.1111/dom.12532.
diabetes uncontrolled with oral antihyperglycaemic drugs:
[22] Ritzel R, Roussel R, Giaccari A, Vora J, Brulle-Wohlhueter C,
Real-life use of basal insulin in China. Diabetes Obes Metab
Yki-Järvinen H. Better glycaemic control and less
2017;19:822–30. https://doi.org/10.1111/dom.12886.
hypoglycaemia with insulin glargine 300 U/mL vs glargine
[11] Tsai S-T, Pathan F, Ji L, Yeung VTF, Chadha M, Suastika K,
100 U/mL: 1-year patient-level meta-analysis of the EDITION
et al. First insulinization with basal insulin in patients with
clinical studies in people with type 2 diabetes. Diabetes Obes
Type 2 diabetes in a real-world setting in Asia. J Diabetes
Metab 2018;20:541–8. https://doi.org/10.1111/dom.13105.
2011;3:208–16. https://doi.org/10.1111/j.1753-0407.2011.00137.
[23] Venn BJ, Williams SM, Mann JI. Comparison of postprandial
x.
glycaemia in Asians and Caucasians: Glycaemia in Asians
[12] Chan WB, Chen JF, Goh S-Y, Vu TTH, Isip-Tan IT, Mudjanarko
and Caucasians. Diabet Med 2010;27:1205–8. https://doi.org/
SW, et al. Challenges and unmet needs in basal insulin
10.1111/j.1464-5491.2010.03069.x.
therapy: lessons from the Asian experience. Diabetes Metab
[24] Ma RC, Chan JC. Type 2 diabetes in East Asians: similarities
Syndr Obes 2017;10:521–32. https://doi.org/10.2147/DMSO.
and differences with populations in Europe and the United
S143046.
States. Ann N Y Acad Sci 2013;1281:64. https://doi.org/
[13] Berard L, Bonnemaire M, Mical M, Edelman S. Insights into
10.1111/nyas.12098.
optimal basal insulin titration in type 2 diabetes: Results of a
[25] Chan JCN, Bunnag P, Chan SP, Tan ITI, Tsai S-T, Gao L, et al.
quantitative survey. Diabetes Obes Metab 2018;20:301–8.
Glycaemic responses in Asian and non-Asian people with
https://doi.org/10.1111/dom.13064.
type 2 diabetes initiating insulin glargine 100 units/mL: A
[14] Russell-Jones D, Pouwer F, Khunti K. Identification of barriers
patient-level pooled analysis of 16 randomised controlled
to insulin therapy and approaches to overcoming them.
10 diabetes research and clinical practice 176 (2021) 108848

trials. Diabetes Res Clin Pract 2018;135:199–205. https://doi. with Type 2 Diabetes on Insulin Glargine 300 U/ml Versus
org/10.1016/j.diabres.2017.11.025. First- and Second-Generation Basal Insulin Analogs: The
[26] Cai X-L, Ji L-N. Treatment response between Asian and non- Real-World LIGHTNING Study. Diabetes Ther 2019;10:617–33.
Asian patients with type 2 diabetes: is there any similarity or https://doi.org/10.1007/s13300-019-0568-8.
difference?. Chin Med J (Engl) 2019;132:1–3. https://doi.org/ [34] Zhou FL, Ye F, Berhanu P, Gupta VE, Gupta RA, Sung J, et al.
10.1097/CM9.0000000000000012. Real-world evidence concerning clinical and economic
[27] Ji L, Kang ES, Dong X, Li L, Yuan G, Shang S, et al. Efficacy and outcomes of switching to insulin glargine 300 units/mL vs
safety of insulin glargine 300 U/mL versus insulin glargine other basal insulins in patients with type 2 diabetes using
100 U/mL in Asia Pacific insulin-naı̈ve people with type 2 basal insulin. Diabetes Obes Metab 2018;20:1293–7. https://
diabetes: The EDITION AP randomized controlled trial. doi.org/10.1111/dom.13199.
Diabetes Obes Metab 2020;22:612–21. https://doi.org/ [35] Shiramoto M, Eto T, Irie S, Fukuzaki A, Teichert L, Tillner J,
10.1111/dom.13936. et al. Single-dose new insulin glargine 300 U/ml provides
[28] Terauchi Y, Koyama M, Cheng X, Takahashi Y, Riddle MC, prolonged, stable glycaemic control in Japanese and
Bolli GB, et al. New insulin glargine 300 U/ml versus glargine European people with type 1 diabetes. Diabetes Obes Metab
100 U/ml in Japanese people with type 2 diabetes using basal 2015;17:254–60. https://doi.org/10.1111/dom.12415.
insulin and oral antihyperglycaemic drugs: glucose control [36] Mauricio D, Meneghini L, Seufert J, Liao L, Wang H, Tong L,
and hypoglycaemia in a randomized controlled trial et al. Glycaemic control and hypoglycaemia burden in
(EDITION JP 2). Diabetes Obes Metab 2016;18:366–74. https:// patients with type 2 diabetes initiating basal insulin in
doi.org/10.1111/dom.12618. Europe and the USA. Diabetes Obes Metab 2017;19:1155–64.
[29] Bolli GB, Riddle MC, Bergenstal RM, Ziemen M, Sestakauskas https://doi.org/10.1111/dom.12927.
K, Goyeau H, et al. New insulin glargine 300 U/ml compared [37] Dalal MR, Kazemi M, Ye F, Xie L. Hypoglycemia After
with glargine 100 U/ml in insulin-naı̈ve people with type 2 Initiation of Basal Insulin in Patients with Type 2 Diabetes in
diabetes on oral glucose-lowering drugs: a randomized the United States: Implications for Treatment
controlled trial (EDITION 3). Diabetes Obes Metab Discontinuation and Healthcare Costs and Utilization. Adv
2015;17:386–94. https://doi.org/10.1111/dom.12438. Ther 2017;34:2083–92. https://doi.org/10.1007/s12325-017-
[30] World Health Organization, International Diabetes 0592-x.
Federation. Definition and diagnosis of diabetes mellitus and [38] Rosenstock J, Cheng A, Ritzel R, Bosnyak Z, Devisme C, Cali
intermediate hyperglycaemia: report of a WHO/IDF AMG, et al. More Similarities Than Differences Testing
consultation. 2006. Insulin Glargine 300 Units/mL Versus Insulin Degludec 100
[31] Association AD. Defining and Reporting Hypoglycemia in Units/mL in Insulin-Naive Type 2 Diabetes: The Randomized
Diabetes: A report from the American Diabetes Association Head-to-Head BRIGHT Trial. Diabetes Care 2018;41:2147–54.
Workgroup on Hypoglycemia. Diabetes Care 2005;28:1245–9. https://doi.org/10.2337/dc18-0559.
https://doi.org/10.2337/diacare.28.5.1245. [39] Heise T, Mathieu C. Impact of the mode of protraction of
[32] Allen KV, Frier BM. Nocturnal hypoglycemia: clinical basal insulin therapies on their pharmacokinetic and
manifestations and therapeutic strategies toward pharmacodynamic properties and resulting clinical
prevention. Endocr Pract 2003;9:530–43. https://doi.org/ outcomes. Diabetes Obes Metab 2017;19:3–12. https://doi.org/
10.4158/EP.9.6.530. 10.1111/dom.12782.
[33] Pettus J, Roussel R, Liz Zhou F, Bosnyak Z, Westerbacka J,
Berria R, et al. Rates of Hypoglycemia Predicted in Patients

You might also like