Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events from 19 clinical trials Michael Lehrke, Lawrence A. Leiter, Uwe Hehnke, Sandra Thiemann, Amit Bhandari, Thomas Meinicke, Odd Erik Johansen PII: DOI: Reference:
S1056-8727(16)30216-1 doi: 10.1016/j.jdiacomp.2016.06.015 JDC 6773
To appear in:
Journal of Diabetes and Its Complications
Received date: Revised date: Accepted date:
28 April 2016 13 June 2016 15 June 2016
Please cite this article as: Lehrke, M., Leiter, L.A., Hehnke, U., Thiemann, S., Bhandari, A., Meinicke, T. & Johansen, O.E., Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events from 19 clinical trials, Journal of Diabetes and Its Complications (2016), doi: 10.1016/j.jdiacomp.2016.06.015
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events
RI P
T
from 19 clinical trials
MA NU
Thomas Meinicke e, Odd Erik Johansen f*
SC
Michael Lehrke a, Lawrence A. Leiter b, Uwe Hehnke c, Sandra Thiemann c, Amit Bhandari d,
a
University Hospital Aachen, Aachen, Germany
b
Keenan Research Centre in the Li Ka Shing Research Institute, St. Michael’s Hospital,
ED
University of Toronto, Toronto, Canada
Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
d
Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
e
Boehringer Ingelheim Corporation, Biberach, Germany
CE
Boehringer Ingelheim Norway KS, Asker, Norway
AC
f
PT
c
*Corresponding Author: Odd Erik Johansen, Boehringer Ingelheim Drengsrudbekken 8, 1373 Asker, Oslo, Norway
[email protected]
Keywords: Linagliptin, dipeptidyl peptidase-4 inhibitor, type 2 diabetes mellitus, coronary artery disease, drug safety, heart failure
1
ACCEPTED MANUSCRIPT Abstract Aims: To examine the safety and efficacy of linagliptin in patients with type 2 diabetes
T
mellitus (T2DM) and coronary artery disease (CAD) using pooled data from the global
RI P
clinical trials program.
Methods: Patient-level data were pooled from randomized, placebo-controlled clinical trials
SC
of linagliptin (5 mg, monotherapy or combination therapy). Safety/efficacy analyses were
MA NU
conducted for patients with CAD and ≥12 and ≥24 weeks of treatment, respectively. Results: The safety analysis included 19 trials (linagliptin, n=451; placebo, n=272) and the efficacy analysis, 12 trials (linagliptin, n=328; placebo, n=198); mean (± standard deviation) exposure to study treatment was 212 (144) days linagliptin and 245 (171) days placebo.
ED
Occurrence of cardiac adverse events (AEs) was similar for linagliptin- and placebo-treated patients (9.1% and 9.2%, respectively); exposure-adjusted incidence rates (per 100 patient-
PT
years) were 16.6 and 14.0, respectively. Overall incidence of AEs was numerically lower with linagliptin than placebo. After 24 weeks, mean adjusted change (standard error) from
CE
baseline glycosylated hemoglobin was –0.64% (0.04) with linagliptin vs. –0.08% (0.05) with
AC
placebo (P<.001).
Conclusions: This comprehensive pooled analysis showed that addition of linagliptin to treatment regimens of patients with T2DM and CAD was not associated with an increased incidence of cardiac AEs, was well tolerated, and effective.
3
ACCEPTED MANUSCRIPT 1. Introduction Coronary artery disease (CAD) is a prevalent condition in patients with type 2 diabetes
T
mellitus (T2DM) (Go et al., 2014; Huxley et al., 2006); compared with individuals without
RI P
diabetes, patients with T2DM have a two to fourfold increased risk of cardiovascular (CV) disease (Beckman et al., 2002; Haffner et al., 1998; Preis et al., 2009). For patients with
SC
T2DM, the presence of CAD results in poorer clinical outcomes compared with those without CAD (Beckman et al., 2002; Franco et al., 2007; Miettinen et al., 1998). The pathophysiology
MA NU
of diabetic vascular disease is complex, and patients with T2DM may also have additional risk factors which increase the risk of atherosclerosis, including hypertension, dyslipidemia, obesity, smoking and insulin resistance. The importance of controlling these risk factors through medication and lifestyle changes are well established (Beckman et al., 2002). In
ED
particular, the control of hyperglycemia has been shown to reduce the risk of microvascular endpoints. Data from the United Kingdom Prospective Diabetes Study (UKPDS)
PT
demonstrated, over a 10-year follow-up period, that intensive glycemic control was associated with a reduction of microvascular endpoints, as well as risk reduction for
CE
myocardial infarction (MI) and death from any cause (Holman et al., 2008). Similarly, in the
AC
Kumamoto study, intensive glycemic control was shown to delay the onset and progression of early diabetic microvascular complications after 8-year follow-up of Japanese patients with T2DM (Shichiri et al., 2000). In contrast, the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study was prematurely terminated following a recommendation by the data and safety monitoring committee due to a 22% relative increase in risk of mortality and a 38% relative increase in CV death among patients receiving an intensive strategy to reduce glycosylated hemoglobin (HbA1c) levels, compared with those with a less stringent HbA1c target, although the groups did not differ with respect to the composite outcome measure of non-fatal MI, non-fatal stroke or CV death (in fact, non-fatal MI was significantly reduced) (Gerstein et al., 2008). Subsequent reports from the ACCORD Study Group on long-term effects, following 3.7 years of intensive glucose lowering, includes reports on
4
ACCEPTED MANUSCRIPT outcomes after 5 years of follow up (Gerstein et al., 2011) as well as 9-year outcomes (in the ACCORD International Ongoing [ACCORDION] Study) (The ACCORD Study Group Writing
RI P
attenuation of the overall mortality risk, shown in ACCORDION.
T
Committee, 2016), which demonstrated persistence of the original findings, in addition to an
SC
However, the use of intensive glucose control showed no benefit on macrovascular events for patients with T2DM in the Action in Diabetes and Vascular Disease: Preterax and
MA NU
Diamicron Modified-Release Controlled Evaluation (ADVANCE) study. Although intensive glucose control was associated with a significant 10% relative reduction in the combined outcome of major macrovascular and microvascular events during a median 5-year follow-up period, this was mainly as a result of a 21% relative reduction in the incidence of
ED
nephropathy (P=.006), with no risk reduction in the incidence of major macrovascular events or death from CV causes (Patel et al., 2008). In the extended follow-up of this trial (median,
PT
5.4 years post-trial follow-up), intensive glucose control during the trial did not lead to long-
CE
term benefits with respect to mortality or macrovascular events (Zoungas et al., 2014). Similarly, a study of American veterans with T2DM, the Veterans Affairs Diabetes Trial
AC
(VADT), showed no significant effect of intensive glycemic control on the incidence of major CV events or on the risk of death from any cause, after a median follow-up period of 5.6 years (Duckworth et al., 2009). However, after extended follow-up of the VADT population to 9.8 years after the start of the study, patients who received intensive glucose control for the first 5.6 years were found to have a significant 17% reduction of the primary endpoint of macrovascular disease but no improvement in cardiovascular- or overall survival (Hayward et al., 2015).
In addition to uncertainties about the effectiveness of tight glycemic control in reducing the risk of macrovascular events, this approach can be associated with an
5
ACCEPTED MANUSCRIPT increased risk of severe hypoglycemic episodes, which can offset some of the CV benefits of therapy (Boussageon et al., 2011; Mannucci et al., 2009). Subgroup analyses of UKPDS, ACCORD, ADVANCE and VADT suggest that patients with T2DM of shorter duration and
RI P
T
without established CV disease (CVD) are more likely to benefit from intensive glycemic control, compared with those having more established disease for whom the risks might
SC
outweigh the potential CV benefits of intensive therapy (Skyler et al., 2009).
MA NU
Developing optimal treatment strategies in patients with T2DM and CAD remains a challenge in this high-risk population, where individuals often present with multiple risk factors which substantially increase the risk for morbidity and mortality (Turner et al., 1998). In view of the need for polypharmacy, frequent hospitalization and the risk of potential side
ED
effects or contraindications to some drugs, selecting the appropriate glucose-lowering treatment for these patients is uniquely challenging. The latest Diabetes Management
PT
Guidelines from the American Association of Clinical Endocrinologists (AACE) (Garber et al.,
CE
2016) and the joint Position Statement from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) (Inzucchi et al., 2015)
AC
recommend a patient-centered approach to management, with treatment individualized to take account of risk factors, including comorbid conditions such as CAD. Current uncertainty about the long-term CV safety of specific glucose-lowering drugs (Selvin et al., 2008), has led to the recommendation by the Food and Drug Administration (FDA) that the CV risk is evaluated for all compounds, except insulins, that are being developed as therapies for T2DM (U.S. FDA, 2008). As a result, many trials have been designed to assess the longterm CV outcomes of recently developed drugs for the management of T2DM, and there remains a clinical need for approaches to glycemic control that do not further increase CV risk (Cavender et al., 2015).
6
ACCEPTED MANUSCRIPT Linagliptin is a dipeptidyl-peptidase (DPP)-4 inhibitor approved for the treatment of T2DM. The overall safety and tolerability of linagliptin has been established in a large clinical trial program and further demonstrated in a pooled analysis of 22 placebo-controlled trials of
RI P
T
linagliptin (Lehrke et al., 2014). The findings of this analysis supported previous findings of a low incidence of adverse events and good overall tolerability of linagliptin in a broad range of patients with T2DM. The CV safety of linagliptin has been evaluated in a comprehensive
SC
patient-level pooled analysis of prospectively adjudicated CV events from the clinical trial
MA NU
program (Rosenstock et al., 2015). This analysis of 19 trials of at least 12 weeks’ duration, in which linagliptin was evaluated in comparison with placebo or one or more active comparators, showed that linagliptin was not associated with increased CV risk in patients with T2DM. A post-hoc pooled analysis of linagliptin as add-on to insulin therapy in T2DM also demonstrated a neutral effect of linagliptin on the occurrence of major adverse CV
ED
events (Zinman et al., 2016). Two CV outcome trials are underway with the aim of further
PT
evaluating the CV safety of linagliptin. The CARdiovascular Outcome Study of LINAgliptin versus Glimepiride in Patients with Type 2 Diabetes (CAROLINA®) (NCT01243424) has
CE
randomized 6041 patients with early T2DM who are at high CV risk to receive therapy with linagliptin or the sulfonylurea (SU) glimepiride (Marx et al., 2015; Rosenstock et al., 2013).
AC
The CARdiovascular Safety & Clinical outcoME with LINAgliptin (CARMELINA®) trial will compare the CV and renal safety of linagliptin vs. placebo, added to standard care in patients with T2DM who are at high risk of vascular complications. While the outcomes of dedicated CV safety trials of linagliptin are awaited, analysis of available data from completed clinical trials can provide some insights into the role of this agent in patients with CAD or at high risk of CV events. The aim of this analysis was to examine the safety and efficacy of linagliptin in patients with CAD using pooled data from a global clinical trials program.
2. Materials and methods 7
ACCEPTED MANUSCRIPT Patient-level data were pooled for this analysis from randomized, placebo-controlled clinical trials of linagliptin 5 mg, of at least 12 weeks’ duration. The trials included linagliptin administered either as monotherapy or in combination with other glucose-lowering drugs. All
RI P
T
trials were conducted in accordance with Good Clinical Practice guidelines and the principles of the Declaration of Helsinki. Eligibility criteria across the included trials were similar and included a diagnosis of T2DM, age ≥18 years, HbA1c 7–10% entrance criterion in most
SC
studies, and a body mass index (BMI) of 20–45 kg/m2. In all studies, rescue therapy could
MA NU
be initiated in response to a deterioration in glycemic control. In general, rescue therapy was initiated if glucose levels exceeded 240, 200 or 180 mg/dl (after an overnight fast) on two separate days during the first 12, 12–24 or >24 weeks, respectively. Data collected after
ED
initiation of rescue therapy were included in the analysis.
Patients with CAD were identified using relevant cardiac terms from the standardized
PT
Medical Dictionary for Regulatory Activities (MedDRA, v16.0) query “embolic and thrombotic
CE
events”. Included terms were: acute coronary syndrome; acute myocardial infarction; angina pectoris; angina unstable; arteriosclerosis coronary artery; arteriospasm coronary; coronary
AC
angioplasty; coronary arterial stent insertion; coronary artery bypass; coronary artery dilatation; coronary artery disease; coronary artery insufficiency; coronary artery occlusion; coronary artery stenosis; coronary revascularization; microvascular coronary artery disease; myocardial infarction; myocardial ischemia; percutaneous coronary intervention; post procedural myocardial infarction; postinfarction angina; Prinzmetal angina; silent myocardial infarction.
Safety analyses were conducted on patients who had at least 12 weeks of treatment and who had received at least one dose of study drug (treated set). Data on adverse events (AEs) were collected by the study investigators using electronic case report forms. The
8
ACCEPTED MANUSCRIPT incidence and intensity of AEs, including cardiac and hypoglycemic events, were coded using MedDRA v16.0 and analyzed using descriptive statistical methods. Data on investigator-reported cardiac AEs were defined by the MedDRA System Organ Class of
RI P
T
“cardiac disorders” based on clinical interpretation. Exposure-adjusted incidence rates were calculated using the number of patients with the respective events per treatment divided by the time at risk, expressed as 100 patient-years. Heart failure data were calculated as a total
SC
of the narrow standardized MedDRA queries (SMQ), and consisted of the following terms:
MA NU
acute left ventricular failure; acute pulmonary edema; acute right ventricular failure; cardiac asthma; cardiac failure; cardiac failure, acute; cardiac failure, chronic; cardiac failure, congestive; cardiac failure, high output; cardiogenic shock; cardiopulmonary failure cardiorenal syndrome; chronic left ventricular failure; chronic right ventricular failure; cor pulmonale; cor pulmonale, acute; cor pulmonale, chronic; ejection fraction decreased;
ED
hepatic congestion; hepatojugular reflux; left ventricular failure; low cardiac output syndrome;
PT
neonatal cardiac failure; pulmonary edema; pulmonary edema, neonatal; right ventricular failure; ventricular failure. Reporting of AEs was also analyzed according to concomitant use
AC
CE
of insulin and/or SU therapy.
Overall hypoglycemic events included both asymptomatic and symptomatic hypoglycemic events. In general, asymptomatic hypoglycemia was defined as an event that was not accompanied by typical symptoms of hypoglycemia but with a measured plasma glucose concentration ≤70 mg/dl (3.9 mmol/l). Symptomatic hypoglycemia was defined as a measured plasma glucose concentration ≥54 mg/dl and ≤70 mg/dl (≥3.0 mmol/l and ≤3.9 mmol/l), accompanied by typical symptoms of hypoglycemia, or symptomatic hypoglycemia with a measured plasma glucose concentration <54 mg/dl (<3.0 mmol/l): event accompanied by typical symptoms of hypoglycemia but no need for external assistance. A severe hypoglycemic episode was defined as an event requiring the assistance of another person to actively administer carbohydrate, glucagon or other resuscitative actions. 9
ACCEPTED MANUSCRIPT
Efficacy analyses were conducted using data from patients who had received at least 24
T
weeks of treatment and at least one dose of study drug at the 5-mg dosage, and who also
RI P
had an HbA1c value at baseline and at least one on-treatment value (full analysis set). The change from baseline glycemic parameters for linagliptin and placebo groups were
SC
compared using an analysis of covariance (ANCOVA). The model included treatment, washout, study, and continuous baseline HbA1c (HbA1c analysis) and baseline HbA1c and
MA NU
baseline fasting plasma glucose (FPG) (FPG analysis). The analysis used the last observation carried forward (LOCF) method to assign values to missing data. ANCOVA and logistic regression techniques were applied to assess all continuous and categorical secondary and safety endpoints, respectively.
ED
3. Results
PT
The safety analysis included patients from 19 trials (linagliptin, n=451; placebo, n=272) (Supplementary Table 1). The mean [± standard deviation (SD)] exposure to study treatment
CE
was 212 (144) days in the linagliptin group and 245 (171) days for patients allocated to placebo. Baseline characteristics, including the presence of CV risk factors and use of non-
AC
study drugs for CV indications, were well matched between the treatment groups (Table 1). Similar proportions of patients were receiving none, one or more than two glucose-lowering drugs. However, there were modest differences in the composition of classes of drugs used, i.e., use of concomitant insulin (± other glucose-lowering drugs), SU (± other glucoselowering drugs) or insulin + SU (Table 1).
Cardiac AEs were reported by 9.1% of patients in the linagliptin group and 9.2% in the placebo group (Fig. 1A; Table 2). Exposure adjusted incidence rates (per 100 patient-years) of cardiac AEs were 16.6 for linagliptin and 14.0 for placebo. The proportion of patients reporting cardiac AEs was higher among those receiving concomitant SU and/or insulin 10
ACCEPTED MANUSCRIPT (linagliptin, 11.9%; placebo, 11.3%) compared with patients not receiving these drugs
T
(linagliptin, 2.8%; placebo, 2.9%) (Fig. 1B).
RI P
The overall incidence of AEs was numerically lower with linagliptin than placebo (Supplementary Table 2). In the linagliptin group, the proportion of patients with any AE was
SC
higher among patients receiving concomitant SU and/or insulin compared with patients who were not receiving these drugs (73.2% vs. 47.5%); corresponding values in the placebo
MA NU
group were 77.0% vs. 70.6% (Table 3). The overall occurrence of investigator-reported events that were suggestive of congestive heart failure (CHF) was low for linagliptin- and placebo-treated patients, but higher among patients on SU and/or insulin background therapy (Table 3). Similarly, among linagliptin-treated patients, the proportion of patients with
ED
investigator-reported hypoglycemia was greater among those receiving concomitant SU and/or insulin compared with patients who were not taking these treatments (29.4% vs.
PT
2.1%); corresponding values were 32.8% and 0% in the placebo group (Supplementary
CE
Table 3). Overall, the rates of exposure-adjusted investigator-reported hypoglycemia were
AC
slightly lower with linagliptin than with placebo (Fig. 2).
The efficacy analysis included patients from 12 trials (linagliptin, n=328; placebo, n=198). The analysis demonstrated a significantly greater reduction in HbA1c among patients receiving linagliptin vs. placebo: after 24 weeks of treatment the mean adjusted change (± standard error) from baseline HbA1c was –0.65% (± 0.04) for linagliptin-treated patients compared with –0.08% (± 0.05) for the placebo group (P<.001) (Supplementary Table 4). Patients were more likely to achieve HbA1c lowering to <7% with linagliptin vs. placebo (odds ratio 3.70; P<.0001).
4. Discussion 11
ACCEPTED MANUSCRIPT The results from this comprehensive pooled analysis show that the addition of linagliptin to the treatment regimens of patients with a previous history of CAD was well tolerated, with no changes to the known safety profile listed in the current prescribing information
RI P
T
(Boehringer Ingelheim Pharmaceuticals, 2015). Thus, the findings, in patients with a preexisting diagnosis of CAD and at high risk for further CV events, are consistent with those of previous pooled analyses of double-blind, randomized, controlled trials of lower risk
SC
populations that have evaluated safety data from the linagliptin global clinical trials program
MA NU
(Lehrke et al., 2014; Rosenstock et al., 2015; Schernthaner et al., 2014).
A key finding relative to the high-risk CAD patients that were the focus of this analysis was that the overall incidence of cardiac AEs was similar for patients receiving linagliptin and
ED
placebo (9.1% vs. 9.2%, respectively). The exposure-adjusted incidence rate (per 100 patient-years), which allows for possible differences in follow-up duration between treatment
PT
groups, also indicated that the incidence of cardiac AEs was similar among linagliptin- vs.
AC
CE
placebo-treated patients (16.6% vs. 14.0%, respectively).
Given the potential influence of background medication on AE rates, the occurrence of AEs was specifically investigated with regard to patients receiving SU and insulin. We found that the proportion of patients who reported cardiac AEs with linagliptin was numerically greater among patients treated with concomitant SU and/or insulin compared with patients who were not receiving these drugs. As with cardiac AEs, the overall rate of AEs with linagliptin was higher with concomitant SU and/or insulin therapy than without. One obvious explanation for this difference might be the presence of more advanced T2DM among patients who required additional SU and/or insulin to achieve glycemic control, and hence the presence of a greater number of comorbidities in these patients than in those who do not require SU and/or insulin. Previous studies have shown linagliptin to be generally well
12
ACCEPTED MANUSCRIPT tolerated when used as add-on therapy to SU and/or insulin (Barnett et al., 2013; McGill et
T
al., 2014; McGill et al., 2012; Yki-Järvinen et al., 2013).
RI P
Randomized trials have demonstrated a low incidence of hypoglycemia with linagliptin therapy, both alone and when used in combination with other glucose-lowering agents (Del
SC
Prato et al., 2011; Inagaki et al., 2013; Owens et al., 2011; Taskinen et al., 2011). In this study, rates of hypoglycemia with linagliptin were low and comparable with prior pooled
MA NU
analyses, with slightly fewer hypoglycemic episodes reported in the linagliptin group vs. placebo. Of course, this finding could be a consequence of differences in background therapies – there was more insulin use in the placebo group, although conversely there was
ED
also greater SU use in the linagliptin group.
PT
The improvements in glucose control in this vulnerable population were consistent with previously reported data for the general T2DM population (Del Prato et al., 2011; Owens et
AC
CE
al., 2011; Taskinen et al., 2011).
Several longer term studies designed to evaluate the CV safety of other DPP-4 inhibitors have reported results: SAVOR-TIMI 53 (Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus – Thrombolysis in Myocardial Infarction 53 trial) (Scirica et al., 2013), EXAMINE (Examination of Cardiovascular Outcomes With Alogliptin Versus Standard of Care) (White et al., 2013) and TECOS (Trial Evaluating Cardiovascular Outcomes with Sitagliptin) (Green et al., 2015). All of these placebo-controlled trials have demonstrated a neutral effect of these agents (saxagliptin, alogliptin and sitagliptin, respectively) on composite CV outcomes [CV death, stroke and myocardial infarction (MI) in SAVOR and EXAMINE, and CV death, stroke, MI and hospitalization for unstable angina in TECOS] in patients with high risk for CV events (Scirica et al., 2013; White et al., 2013). 13
ACCEPTED MANUSCRIPT However, in SAVOR-TIMI 53, a statistically significant increased risk of hospitalization for CHF was associated with saxagliptin therapy compared with the placebo group [3.5% vs. 2.8%, respectively; HR, 1.27 (95% CI, 1.07 to 1.51; P=.007)] (Scirica et al., 2013). Further
RI P
T
evaluation of the EXAMINE data has shown no increase in the risk of heart failure outcomes associated with alogliptin therapy (Zannad et al., 2015). More recently, results have been published from the TECOS study, which evaluated sitagliptin in patients with T2DM and
SC
established CVD (Green et al., 2015). In this large study, sitagliptin was found to be
MA NU
associated with no increase in the risk of major adverse CV events [sitagliptin was noninferior to placebo for the composite outcome of CV death, non-fatal MI, non-fatal stroke or hospitalization for unstable angina; HR, 0.98 (95% CI, 0.88 to 1.09; P<.001)] or hospitalization for heart failure [HR, 1.00 (95% CI, 0.83 to 1.20; P=.98)] (Green et al., 2015). A recent retrospective, observational study, using information from a US insurance claims
ED
database, found no association between hospitalization for heart failure and treatment with a
PT
DPP-4 inhibitor relative to SU therapy, or treatment with saxagliptin relative to sitagliptin (Fu et al., 2016). At present, therefore, it remains unclear whether the observation of an increase
CE
in hospitalization for CHF with saxagliptin (Scirica et al., 2013) was an adverse effect of this
AC
specific therapy or a chance finding.
For linagliptin, where there are currently no published CV outcomes data, the aforementioned pooled analyses of linagliptin trial data provide the best indication of CV safety. This pooled analysis also showed that the occurrence of investigator-reported events that were suggestive of CHF was low for linagliptin- and placebo-treated patients [0.5% (26 events), 0.2% (8 events)] (Rosenstock et al., 2015). In the current analysis, which evaluated linagliptin in patients with a pre-existing diagnosis of CAD and, therefore, at high risk for further CV events, the incidence of reported cardiac AEs (per 100 patient-years) was comparable (16.6 for linagliptin and 14.0 for placebo) with those of the previous pooled analyses of linagliptin (Lehrke et al., 2014; Rosenstock et al., 2015). Further understanding 14
ACCEPTED MANUSCRIPT of the CV safety of linagliptin will be provided by the results of the ongoing CAROLINA® and
T
CARMELINA® trials.
RI P
The present analysis has several limitations because of its pooled design and the small number of patients with T2DM and CAD available for inclusion in the analysis. Further
SC
limitations are the relatively short and differing durations of the included studies, the fact that none of the individual studies included in this analysis was powered or designed to assess
MA NU
CV risk or events, and that there was no randomization procedure in this analysis for the patients that were identified with CAD. In addition, the mean individual patient exposure to linagliptin and placebo was less than 1 year (212 days and 245 days, respectively) and,
ED
therefore, no conclusions on the long-term CV safety of linagliptin can be drawn.
PT
In summary, and despite the limitations of this pooled analysis, the finding that linagliptin is not associated with an increase in CV risk in patients with T2DM and CAD will be
CE
reassuring to clinicians. The results of CAROLINA® and CARMELINA® and other ongoing
AC
trials of the CV safety of glucose-lowering therapies will further assist clinicians in their efforts to optimize treatment strategies for patients with or at risk of CVD and T2DM.
15
ACCEPTED MANUSCRIPT Acknowledgments This study was supported by Boehringer Ingelheim, and Eli Lilly and Company. The authors
T
were fully responsible for all content and editorial decisions, were involved at all stages of
RI P
manuscript development, and have approved the final version. Medical writing assistance, supported financially by Boehringer Ingelheim, was provided by Paul MacCallum, PhD and
SC
Jennifer Edwards, MB BS of Envision Scientific Solutions, during the preparation of this
MA NU
manuscript.
Conflict of Interest
ML has acted as an advisor to BMS, Astra-Zeneca, Roche, GSK, MSD, Amgen, Sanofi, and
ED
a speaker for BMS, Astra-Zeneca, Roche, GSK, Boehringer Ingelheim, Merck Sharp & Dohme, Novo Nordisk, Sanofi, Amgen.
PT
LAL has received research funding from, has provided CME on behalf of, and/or has acted
CE
as an advisor to: AstraZeneca, Boehringer Ingelhiem, Eli Lilly, GSK, Janssen, Merck, Pfizer, Sanofi, and Sevier.
AC
UH, ST, OEJ, AB and TM are all employees of Boehringer Ingelheim.
Prior Publication The data in this manuscript have been previously presented in posters at the American Diabetes Association 75th Scientific Sessions, Boston, MA, USA, June 5–9, 2015 (Poster 1246-P), and at the 51st Annual Meeting of the European Association for the Study of Diabetes, Stockholm, Sweden, 14–18 September, 2015 (Poster 814).
16
ACCEPTED MANUSCRIPT References Bajaj, M., Gilman, R., Patel, S., Kempthorne-Rawson, J., Lewis-D'Agostino, D., & Woerle, H.
T
J. (2014). Linagliptin improved glycaemic control without weight gain or
RI P
hypoglycaemia in patients with type 2 diabetes inadequately controlled by a combination of metformin and pioglitazone: a 24-week randomized, double-blind
SC
study. Diabetic Medicine, 31(12), 1505–1514.
Barnett, A. H., Huisman, H., Jones, R., von Eynatten, M., Patel, S., & Woerle, H. J. (2013).
MA NU
Linagliptin for patients aged 70 years or older with type 2 diabetes inadequately controlled with common antidiabetes treatments: a randomised, double-blind, placebo-controlled trial. Lancet, 382(9902), 1413–1423. Barnett, A. H., Patel, S., Harper, R., et al. (2012). Linagliptin monotherapy in type 2 diabetes
ED
patients for whom metformin is inappropriate: an 18-week randomized, double-blind, placebo-controlled phase III trial with a 34-week active-controlled extension.
PT
Diabetes, Obesity and Metabolism, 14(12), 1145–1154. Beckman, J. A., Creager, M. A., & Libby, P. (2002). Diabetes and atherosclerosis:
CE
epidemiology, pathophysiology, and management. Journal of the American Medical
AC
Association, 287(19), 2570–2581. Boehringer Ingelheim Pharmaceuticals. Highlights of prescribing information. Tradjenta (linagliptin) tablets. 2015. Accessed: 20 August 2015. Available at: http://bidocs.boehringeringelheim.com/BIWebAccess/ViewServlet.ser?docBase=renetnt&folderPath=/Prescri bing+Information/PIs/Tradjenta/Tradjenta.pdf. Boussageon, R., Bejan-Angoulvant, T., Saadatian-Elahi, M., et al. (2011). Effect of intensive glucose lowering treatment on all cause mortality, cardiovascular death, and microvascular events in type 2 diabetes: meta-analysis of randomised controlled trials. British Medical Journal, 343, d4169.
17
ACCEPTED MANUSCRIPT Cavender, M. A., Steg, P. G., Smith, S. C., Jr., et al. (2015). Impact of diabetes mellitus on hospitalization for heart failure, cardiovascular events, and death: outcomes at 4 years from the Reduction of Atherothrombosis for Continued Health (REACH)
RI P
T
Registry. Circulation, 132(10), 923–931.
Chen, Y., Ning, G., Wang, C., et al. (2015). Efficacy and safety of linagliptin monotherapy in Asian patients with inadequately controlled type 2 diabetes mellitus: a multinational,
SC
24-week, randomized, clinical trial. Journal of Diabetes Investigation, 6(6), 692–698.
MA NU
Del Prato, S., Barnett, A. H., Huisman, H., Neubacher, D., Woerle, H. J., & Dugi, K. A. (2011). Effect of linagliptin monotherapy on glycaemic control and markers of β-cell function in patients with inadequately controlled type 2 diabetes: a randomized controlled trial. Diabetes, Obesity and Metabolism, 13(3), 258–267. Duckworth, W., Abraira, C., Moritz, T., et al. (2009). Glucose control and vascular
PT
360(2), 129–139.
ED
complications in veterans with type 2 diabetes. New England Journal of Medicine,
Franco, O. H., Steyerberg, E. W., Hu, F. B., Mackenbach, J., & Nusselder, W. (2007).
CE
Associations of diabetes mellitus with total life expectancy and life expectancy with and without cardiovascular disease. Archives of Internal Medicine, 167(11), 1145–
AC
1151.
Fu, A. Z., Johnston, S. S., Ghannam, A., et al. (2016). Association between hospitalization for heart failure and dipeptidyl peptidase-4 inhibitors in patients with type 2 diabetes: an observational study. Diabetes Care, [Epub ahead of print]. Garber, A. J., Abrahamson, M. J., Barzilay, J. I., et al. (2016). Consensus statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the comprehensive type 2 diabetes management algorithm – 2016 executive summary. Endocrine Practice, 22(1), 84–113. Gerstein, H. C., Miller, M. E., Byington, R. P., et al. (2008). Effects of intensive glucose lowering in type 2 diabetes. New England Journal of Medicine, 358(24), 2545–2559.
18
ACCEPTED MANUSCRIPT Gerstein, H. C., Miller, M. E., Genuth, S., et al. (2011). Long-term effects of intensive glucose lowering on cardiovascular outcomes. New England Journal of Medicine, 364(9), 818-828.
RI P
T
Go, A. S., Mozaffarian, D., Roger, V. L., et al. (2014). Heart disease and stroke statistics-2014 update: a report from the American Heart Association. Circulation, 129(3), e28– e292.
SC
Gomis, R., Espadero, R. M., Jones, R., Woerle, H. J., & Dugi, K. A. (2011). Efficacy and
MA NU
safety of initial combination therapy with linagliptin and pioglitazone in patients with inadequately controlled type 2 diabetes: a randomized, double-blind, placebocontrolled study. Diabetes, Obesity and Metabolism, 13(7), 653–661. Green, J. B., Bethel, M. A., Armstrong, P. W., et al. (2015). Effect of sitagliptin on
373(3), 232–242.
ED
cardiovascular outcomes in type 2 diabetes. New England Journal of Medicine,
PT
Haak, T., Meinicke, T., Jones, R., Weber, S., von Eynatten, M., & Woerle, H. J. (2012). Initial combination of linagliptin and metformin improves glycaemic control in type 2
CE
diabetes: a randomized, double-blind, placebo-controlled study. Diabetes, Obesity and Metabolism, 14(6), 565–574.
AC
Haffner, S. M., Lehto, S., Rönnemaa, T., Pyörälä, K., & Laakso, M. (1998). Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. New England Journal of Medicine, 339(4), 229–234. Hayward, R. A., Reaven, P. D., Wiitala, W. L., et al. (2015). Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. New England Journal of Medicine, 372(23), 2197–2206. Holman, R. R., Paul, S. K., Bethel, M. A., Matthews, D. R., & Neil, H. A. (2008). 10-Year follow-up of intensive glucose control in type 2 diabetes. New England Journal of Medicine, 359(15), 1577–1589.
19
ACCEPTED MANUSCRIPT Huxley, R., Barzi, F., & Woodward, M. (2006). Excess risk of fatal coronary heart disease associated with diabetes in men and women: meta-analysis of 37 prospective cohort studies. British Medical Journal, 332(7533), 73–78.
RI P
T
Inagaki, N., Watada, H., Murai, M., et al. (2013). Linagliptin provides effective, well-tolerated add-on therapy to pre-existing oral antidiabetic therapy over 1 year in Japanese patients with type 2 diabetes. Diabetes, Obesity and Metabolism, 15(9), 833–843.
SC
Inzucchi, S. E., Bergenstal, R. M., Buse, J. B., et al. (2015). Management of hyperglycemia
MA NU
in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care, 38(1), 140–149.
Kawamori, R., Inagaki, N., Araki, E., et al. (2012). Linagliptin monotherapy provides superior glycaemic control versus placebo or voglibose with comparable safety in Japanese
ED
patients with type 2 diabetes: a randomized, placebo and active comparator-
PT
controlled, double-blind study. Diabetes, Obesity and Metabolism, 14(4), 348–357. Laakso, M., Rosenstock, J., Groop, P. H., et al. (2015). Treatment with the dipeptidyl
CE
peptidase-4 inhibitor linagliptin or placebo followed by glimepiride in patients with type 2 diabetes with moderate to severe renal impairment: a 52-week, randomized,
AC
double-blind clinical trial. Diabetes Care, 38(2), e15–17. Lehrke, M., Marx, N., Patel, S., et al. (2014). Safety and tolerability of linagliptin in patients with type 2 diabetes: a comprehensive pooled analysis of 22 placebo-controlled studies. Clinical Therapeutics, 36(8), 1130–1146. Lewin, A. J., Arvay, L., Liu, D., Patel, S., von Eynatten, M., & Woerle, H. J. (2012). Efficacy and tolerability of linagliptin added to a sulfonylurea regimen in patients with inadequately controlled type 2 diabetes mellitus: an 18-week, multicenter, randomized, double-blind, placebo-controlled trial. Clinical Therapeutics, 34(9), 1909–1919.e1915. Mannucci, E., Monami, M., Lamanna, C., Gori, F., & Marchionni, N. (2009). Prevention of cardiovascular disease through glycemic control in type 2 diabetes: a meta-analysis 20
ACCEPTED MANUSCRIPT of randomized clinical trials. Nutrition, Metabolism and Cardiovascular Diseases, 19(9), 604–612. Marx, N., Rosenstock, J., Kahn, S. E., et al. (2015). Design and baseline characteristics of
RI P
T
the CARdiovascular Outcome Trial of LINAgliptin Versus Glimepiride in Type 2 Diabetes (CAROLINA(R)). Diabetes and Vascular Disease Research, 12(3), 164– 174.
SC
McGill, J. B., Barnett, A. H., Lewin, A. J., et al. (2014). Linagliptin added to sulphonylurea in
MA NU
uncontrolled type 2 diabetes patients with moderate-to-severe renal impairment. Diabetes and Vascular Disease Research, 11(1), 34–40. McGill, J. B., Sloan, L., Newman, J., et al. (2012). Long-term efficacy and safety of linagliptin in patients with type 2 diabetes and severe renal impairment: a 1-year, randomized, double-blind, placebo-controlled study. Diabetes Care, 36(2), 237-244.
ED
Miettinen, H., Lehto, S., Salomaa, V., et al. (1998). Impact of diabetes on mortality after the
PT
first myocardial infarction. The FINMONICA Myocardial Infarction Register Study Group. Diabetes Care, 21(1), 69–75.
CE
Owens, D. R., Swallow, R., Dugi, K. A., & Woerle, H. J. (2011). Efficacy and safety of linagliptin in persons with type 2 diabetes inadequately controlled by a combination of
AC
metformin and sulphonylurea: a 24-week randomized study. Diabetic Medicine, 28(11), 1352–1361. Patel, A., MacMahon, S., Chalmers, J., et al. (2008). Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. New England Journal of Medicine, 358(24), 2560–2572. Preis, S. R., Hwang, S. J., Coady, S., et al. (2009). Trends in all-cause and cardiovascular disease mortality among women and men with and without diabetes mellitus in the Framingham Heart Study, 1950 to 2005. Circulation, 119(13), 1728–1735. Rosenstock, J., Marx, N., Kahn, S. E., et al. (2013). Cardiovascular outcome trials in type 2 diabetes and the sulphonylurea controversy: rationale for the active-comparator CAROLINA trial. Diabetes and Vascular Disease Research, 10(4), 289–301. 21
ACCEPTED MANUSCRIPT Rosenstock, J., Marx, N., Neubacher, D., et al. (2015). Cardiovascular safety of linagliptin in type 2 diabetes: a comprehensive patient-level pooled analysis of prospectively adjudicated cardiovascular events. Cardiovascular Diabetology, 14, 57.
RI P
T
Ross, S. A., Rafeiro, E., Meinicke, T., Toorawa, R., Weber-Born, S., & Woerle, H. J. (2012). Efficacy and safety of linagliptin 2.5 mg twice daily versus 5 mg once daily in patients with type 2 diabetes inadequately controlled on metformin: a randomised, double-
SC
blind, placebo-controlled trial. Current Medical Research and Opinion, 28(9), 1465–
MA NU
1474.
Schernthaner, G., Barnett, A. H., Patel, S., Hehnke, U., von Eynatten, M., & Woerle, H. J. (2014). Safety and efficacy of the dipeptidyl peptidase-4 inhibitor linagliptin in elderly patients with type 2 diabetes: a comprehensive analysis of data from 1331 individuals aged ≥ 65 years. Diabetes, Obesity and Metabolism, 16(11), 1078–1086.
ED
Scirica, B. M., Bhatt, D. L., Braunwald, E., et al. (2013). Saxagliptin and cardiovascular
PT
outcomes in patients with type 2 diabetes mellitus. New England Journal of Medicine, 369(14), 1317–1326.
CE
Selvin, E., Bolen, S., Yeh, H. C., et al. (2008). Cardiovascular outcomes in trials of oral diabetes medications: a systematic review. Archives of Internal Medicine, 168(19),
AC
2070–2080.
Shichiri, M., Kishikawa, H., Ohkubo, Y., & Wake, N. (2000). Long-term results of the Kumamoto Study on optimal diabetes control in type 2 diabetic patients. Diabetes Care, 23(Suppl 2), B21–29. Skyler, J. S., Bergenstal, R., Bonow, R. O., et al. (2009). Intensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA diabetes trials: a position statement of the American Diabetes Association and a scientific statement of the American College of Cardiology Foundation and the American Heart Association. Circulation, 119(2), 351–357. Taskinen, M. R., Rosenstock, J., Tamminen, I., et al. (2011). Safety and efficacy of linagliptin as add-on therapy to metformin in patients with type 2 diabetes: a randomized, 22
ACCEPTED MANUSCRIPT double-blind, placebo-controlled study. Diabetes, Obesity and Metabolism, 13(1), 65– 74. The ACCORD Study Group Writing Committee. (2016). 9-Year Effects of 3.7 Years of
RI P
T
Intensive Glycemic Control on Cardiovascular Outcomes. Diabetes Care. Thrasher, J., Daniels, K., Patel, S., Whetteckey, J., & Woerle, H. J. (2014). Efficacy and safety of linagliptin in black/African American patients with type 2 diabetes: a 6-
SC
month, randomized, double-blind, placebo-controlled study. Endocrine Practice,
MA NU
20(5), 412–420.
Turner, R. C., Millns, H., Neil, H. A., et al. (1998). Risk factors for coronary artery disease in non-insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). British Medical Journal, 316(7134), 823–828. U.S. FDA. U.S. Department of Health and Human Services, Food and Drug Administration,
ED
Center for Drug Evaluation and Research (CDER). Guidance for industry diabetes
PT
mellitus – evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. 2008. Accessed: 15 September, 2015. Available at:
CE
http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Gui dances/ucm071627.pdf.
AC
Wang, W., Yang, J., Yang, G., et al. (2016). Efficacy and safety of linagliptin in Asian patients with type 2 diabetes mellitus inadequately controlled by metformin: a multinational 24-week, randomized clinical trial. Journal of Diabetes, 8(2), 229–237. White, W. B., Cannon, C. P., Heller, S. R., et al. (2013). Alogliptin after acute coronary syndrome in patients with type 2 diabetes. New England Journal of Medicine, 369(14), 1327–1335. Yki-Järvinen, H., Rosenstock, J., Durán-Garcia, S., et al. (2013). Effects of adding linagliptin to basal insulin regimen for inadequately controlled type 2 diabetes: a ≥52-week randomized, double-blind study. Diabetes Care, 36(12), 3875–3881. Zannad, F., Cannon, C. P., Cushman, W. C., et al. (2015). Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in 23
ACCEPTED MANUSCRIPT EXAMINE: a multicentre, randomised, double-blind trial. Lancet, 385(9982), 2067– 2076. Zinman, B., Ahrén, B., Neubacher, D., Patel, S., Woerle, H. J., & Johansen, O. E. (2016).
RI P
T
Efficacy and cardiovascular safety of linagliptin as an add-on to insulin in type 2 diabetes: a pooled comprehensive post hoc analysis. Canadian Journal of Diabetes, 40(1), 50–57.
SC
Zoungas, S., Chalmers, J., Neal, B., et al. (2014). Follow-up of blood-pressure lowering and
MA NU
glucose control in type 2 diabetes. New England Journal of Medicine, 371(15), 1392–
AC
CE
PT
ED
1406.
24
ACCEPTED MANUSCRIPT Figure captions Fig. 1. Investigator-reported cardiac adverse events a for the treated set (A), and by use of
T
concomitant SU/insulin (B). Data are unadjudicated; defined by the MedDRA System Organ Class “cardiac disorders”.
b
Exposure-adjusted incidence rates are calculated using the number of patients with the
RI P
a
MA NU
MedDRA version 16.0 used for reporting.
SC
respective events per treatment divided by the time at risk expressed as 100 patient-years.
Fig. 2. Rate of exposure-adjusted investigator-reported hypoglycemia (treated set). Asymptomatic or symptomatic hypoglycemia reported as adverse event.
b
Severe hypoglycemia is a hypoglycemic event requiring the assistance of another person
ED
a
c
PT
to actively administer carbohydrate, glucagon or other resuscitative actions. Symptomatic hypoglycemia is an adverse event reported as a hypoglycemic event with
CE
typical symptoms of hypoglycemia.
AC
PG, plasma glucose.
25
ACCEPTED MANUSCRIPT Tables Table 1
RI P
T
Patient demographics and clinical characteristics at baseline (treated set) Placebo
(n=451)
(n=272)
SC
Linagliptin 5 mg
Age (years)
64.8 (9.1)
64.6 (9.3)
62.7
65.8
84.4 (17.4)
87.4 (16.2)
30.4 (4.9)
31.0 (4.8)
77.8
78.3
16.0
10.7
Black African/American
2.7
4.4
Other
0.4
1.1
3.1
5.5
Never smoked
51.7
47.8
Ex-smoker
35.0
38.2
Current smoker
13.3
14.0
37.9
36.8
MA NU
Sex (% male) Body weight (kg) Body mass index (kg/m2)
ED
Race (%)
PT
White
AC
Missing
CE
Asian
Smoking status (%)
Alcohol status (%) Drinker
26
ACCEPTED MANUSCRIPT 8.1 (0.9)
8.1 (0.9)
<7.0%
6.2
6.3
7.0% to <8.0%
39.0
44.5
RI P
T
HbA1c a (%)
8.0% to <9.0%
30.5
19.3
18.8
SC
≥9.0%
35.3
Fasting plasma glucose a, mg/dl
159.6 (46.7)
71.2
66.5
28.8
33.5
Systolic blood pressure (mmHg)
135.2 (15.8)
135.9 (16.8)
Pulse rate (bpm)
70.2 (10.5)
69.9 (10.5)
Total cholesterol
178.9 (49.6)
173.1 (41.2)
Triglyceride
178.5 (108.7)
173.4 (120.5)
High-density lipoprotein
44.0 (11.7)
43.1 (11.0)
Low-density lipoprotein
100.2 (41.6)
97.2 (34.2)
≤1 year
5.8
4.8
>1 to 5 years
23.3
17.6
MA NU
162.7 (46.7)
Renal function (eGFR) according to MDRD (%)
Normal or mild impairment (60 to ≥90 ml/min/1.73 m2)
ED
Moderate or severe impairment, or ESRD (<30 to
CE
AC
Lipids a (mg/dl)
PT
<60 ml/min/1.73 m2)
Time since diagnosis of T2DM (%)
27
ACCEPTED MANUSCRIPT >5 years
71.0
77.6
0
10.2
6.6
1
38.4 51.4
49.6
RI P
T
Glucose-lowering drugs (%)
43.8
31.0
52.6
36.8
21.7
1.1
0.7
74.9
77.6
92.9
87.1
62.3
67.6
Peripheral artery disease (%)
11.3
13.6
Cerebrovascular disease (%)
11.1
12.5
Myocardial infarction (%)
30.4
32.4
Insulin ± other glucose-lowering drugs, excluding
MA NU
SU
SC
≥2
SU ± other glucose-lowering drugs, excluding insulin
ED
Insulin + SU ± other glucose-lowering drugs
Aspirin
AC
Statins
CE
Antihypertensives
PT
Cardiovascular drugs (non-study) (%)
Data are mean (SD) unless stated otherwise. a
The number of patients with data available for these parameters is as follows. HbA1c:
linagliptin n=450, placebo n=272; fasting plasma glucose: linagliptin n=445, placebo n=269; total cholesterol: linagliptin n=372, placebo n=242; triglyceride: linagliptin n=371, placebo n=242; high-density lipoprotein: linagliptin n=370, placebo n=242; low-density lipoprotein: linagliptin n=371, placebo n=239.
28
ACCEPTED MANUSCRIPT eGFR, estimated glomerular filtration rate; MDRD, modification of diet in renal disease; ESRD,
AC
CE
PT
ED
MA NU
SC
RI P
T
end-stage renal disease.
29
ACCEPTED MANUSCRIPT Table 2 Investigator-reported cardiac adverse events by preferred term a (treated set)
SC
Incidence
RI P
(n=451)
T
Linagliptin 5 mg
rate/100 patient-
(n=272) Incidence rate/100 patient-
%
years b
16.6
9.2
14.0
Acute myocardial infarction
0.7
1.1
0.4
0.5
Angina pectoris
2.0
3.4
2.6
3.8
Atrial fibrillation
1.3
2.3
0
0
0.4
0.8
0.7
1.1
Congestive cardiac failure c
0.7
1.1
0
0
Coronary artery disease
0.9
1.5
0.7
1.1
Left ventricular failure
0.7
1.1
0
0
Myocardial ischemia
0.9
1.5
0.7
1.1
Palpitations
0.4
0.8
0.7
1.1
Tachycardia
0.7
1.1
0
0
Narrow SMQ cardiac failure§
2.0
3.4
1.1
1.6
ED
9.1
CE
Cardiac AEs
AC
PT
Cardiac failure c
a
years b
MA NU
%
Placebo
Data are unadjudicated; defined by the MedDRA System Organ Class “cardiac disorders”;
preferred term frequency ≥0.5%. b
Exposure-adjusted incidence rates are calculated using the number of patients with the
respective events per treatment divided by the time at risk expressed as 100 patient-years. MedDRA version 16.0 used for reporting. 30
ACCEPTED MANUSCRIPT c
Cardiac failure and congestive cardiac failure AEs are separate terms derived from the
specific investigator reporting of these events. § Based on reported preferred terms.
RI P
T
MedDRA version 16.0 used for reporting.
AC
CE
PT
ED
MA NU
SC
SMQ, standardized MedDRA query.
31
ACCEPTED MANUSCRIPT Table 3 Any adverse events and heart failure adverse events by concomitant use of SU and/or
Linagliptin 5 mg
Placebo
(n=451)
(n=272)
310
204
73.2
77.0
141
68
47.5
70.6
310
204
SU/insulin: yes (%)
2.6
1.5
Patients analyzed, n
141
68
SU/insulin: no (%)
0.7
0
SC
RI P
T
insulin during screening
Any adverse event
MA NU
Concomitant SU/insulin Patients analysed, n SU/insulin: yes (%)
ED
Patients analysed, n
PT
SU/insulin: no (%) Narrow SMQ heart failure
CE
Concomitant SU/insulin
AC
Patients analyzed, n
32
ACCEPTED MANUSCRIPT Figures
AC
CE
PT
ED
MA NU
SC
RI P
T
Fig. 1.
33
ACCEPTED MANUSCRIPT
AC
CE
PT
ED
MA NU
SC
RI P
T
Fig. 2.
34
ACCEPTED MANUSCRIPT Safety and efficacy of linagliptin in patients with type 2 diabetes mellitus and coronary artery disease: analysis of pooled events
RI P
T
from 19 clinical trials
SC
Highlights
This analysis evaluated the cardiovascular (CV) safety of linagliptin
Linagliptin clinical trial data were pooled for patients at high CV risk
In patients at high CV risk, linagliptin demonstrated safety and tolerability
Efficacy/safety profile was consistent with that in populations of lower CV risk
The results of long-term dedicated linagliptin CV outcome trials are awaited
AC
CE
PT
ED
MA NU
35