OBJECTIVE

Evaluate the safety and efficacy of RVT-1502, a novel oral glucagon receptor antagonist, in subjects with type 2 diabetes inadequately controlled on metformin.

RESEARCH DESIGN AND METHODS

In a phase 2, double-blind, randomized, placebo-controlled study, subjects with type 2 diabetes (n = 166) on a stable dose of metformin were randomized (1:1:1:1) to placebo or RVT-1502 5, 10, or 15 mg once daily for 12 weeks. The primary end point was change from baseline in HbA1c for each dose of RVT-1502 compared with placebo. Secondary end points included change from baseline in fasting plasma glucose (FPG) and safety assessments.

RESULTS

Over 12 weeks, RVT-1502 significantly reduced HbA1c relative to placebo by 0.74%, 0.76%, and 1.05% in the 5-, 10-, and 15-mg groups (P < 0.001), respectively, and FPG decreased by 2.1, 2.2, and 2.6 mmol/L (P < 0.001). The proportions of subjects achieving an HbA1c <7.0% were 19.5%, 39.5%, 39.5%, and 45.0% with placebo and RVT-1502 5, 10, and 15 mg (P ≤ 0.02 vs. placebo). The frequency of hypoglycemia was low, and no episodes were severe. Mild increases in mean aminotransferase levels remaining below the upper limit of normal were observed with RVT-1502 but were reversible and did not appear to be dose related, with no other liver parameter changes. Weight and lipid changes were similar between RVT-1502 and placebo. RVT-1502–associated mild increases in blood pressure were not dose related or consistent across time.

CONCLUSIONS

Glucagon receptor antagonism with RVT-1502 significantly lowers HbA1c and FPG, with a safety profile that supports further clinical development with longer-duration studies (NCT02851849).

Glucagon stimulates gluconeogenesis and glycogenolysis through the glucagon receptor, thereby counteracting the role of insulin in the regulation of glucose homeostasis (1). In subjects with diabetes, the normal feedback system becomes imbalanced, and glucagon secretion is dysregulated, remaining elevated in both fasting and postprandial states, increasing hepatic glucose production, and exacerbating the existing hyperglycemic state (2,3).

Dipeptidyl peptidase 4 inhibitors and glucagon-like peptide 1 receptor agonists (GLP1-RAs) reduce glucagon secretion by enhancing the effects of GLP1 (4). However, neither class fully alters glucagon-induced effects on glucose metabolism because they decrease glucagon secretion by only <10% (58). Nevertheless, the actions of dipeptidyl peptidase 4 inhibitors and GLP1-RAs on hepatic glucose output suggest that targeting glucagon metabolism can improve glycemic control.

In animal models, glucagon inhibition reduces plasma glucose, increases GLP1 levels, and reduces plasma triglycerides (1,9). Glucagon receptor knockout mice do not develop hyperglycemia or the metabolic disturbances of diabetes, even in the presence of severe insulinopenia (10). Glucagon-inhibiting agents tested in various animal models include antibodies that interfere with glucagon receptor signaling, antisense oligonucleotides that decrease glucagon receptor expression, and peptide and small molecule glucagon receptor antagonists (GRAs). These agents reduced blood glucose and improved glucose tolerance in preclinical studies (1120). In subjects with type 2 diabetes, small molecule GRAs decrease fasting plasma glucose (FPG) and hemoglobin A1c (HbA1c) (5,6,2123). However, GRA treatment has been associated with dose-dependent adverse effects, including increases in LDL cholesterol, body weight, blood pressure (BP), hypoglycemia, hepatic fat fraction, and transaminase levels, which have impeded clinical development (2426).

RVT-1502 (formerly LGD-6972) is a novel, orally bioavailable small molecule GRA being developed to improve glycemic control in adults with diabetes. In vitro, RVT-1502 binds to the glucagon receptor with high affinity and selectivity and suppresses glucagon-stimulated cAMP and glucose production (27). It is structurally distinct from other small molecule GRAs, containing a sulfonic acid tail rather than a carboxylic acid tail (28). Pharmacological activity of RVT-1502 appears to be mediated primarily by glucagon receptor signaling, with minimal evidence of off-target pharmacological effects. Binding and inhibition of the closely related GLP1 receptor and gastric inhibitory polypeptide receptor appear to be minimal, with >3,800-fold selectivity for the glucagon receptor. Similarly, RVT-1502 has >100-fold selectivity for the glucagon receptor versus a broad panel of receptors, ion channels, and transporters (27). In vivo, RVT-1502 reduced acute glucagon-stimulated hyperglycemia as well as chronic hyperglycemia in diabetic mouse models (27), effects that appear to be mediated primarily through inhibition of glucagon receptor signaling. In phase 1 studies, RVT-1502 demonstrated favorable safety, tolerability, and pharmacokinetics in healthy volunteers and subjects with type 2 diabetes in whom dose-dependent FPG reductions of up to 3.2 mmol/L (57 mg/dL) were observed after 14 days without clinically significant changes in liver enzymes or lipids, and no subject experienced a hypoglycemic event (29). Here, we describe the results of a 12-week, phase 2 dose-ranging study of RVT-1502 5, 10, or 15 mg once daily versus placebo in subjects with type 2 diabetes on a stable dose of metformin.

Design Overview

This phase 2, randomized, double-blind, placebo-controlled, four-arm, parallel-group study evaluated the efficacy and safety of RVT-1502 5, 10, and 15 mg compared with placebo over 12 weeks at 30 sites in the U.S. between September 2016 and June 2017. The study consisted of a 1-week, single-blind placebo lead-in period, active treatment for 12 weeks, and a 4-week posttreatment follow-up period (Supplementary Fig. 1). Randomization was stratified on the basis of HbA1c at the placebo lead-in visit (≤8.5% or >8.5%). The study was conducted in accordance with Good Clinical Practice guidelines, and the institutional review board at each study center reviewed and approved the protocols before initiating the study. All study subjects provided written informed consent.

Study Population

Eligible subjects included males and females 21–70 years of age with type 2 diabetes taking metformin monotherapy at a stable dose (≥1,000 mg daily) for at least 12 weeks before placebo lead-in. Inclusion criteria were an HbA1c of ≥7.0% (53 mmol/mol) to ≤10.5% (91 mmol/mol), FPG ≤14.4 mmol/L (≤260 mg/dL), BMI 25–40 kg/m2, and weight >45 kg. Female subjects were required to be either surgically sterile or naturally postmenopausal. Male subjects were required to have had a vasectomy or to agree that they and any female partners would use two acceptable forms of contraception, including a condom and another medical or surgical form of contraception (see Supplementary Data). Qualified subjects who required adjustment or stabilization of their metformin dose or washout of other specified oral antidiabetic medications participated in a run-in period of up to 12 additional weeks before randomization (see Supplementary Data).

Key exclusion criteria were a history of diabetic ketoacidosis or hypoglycemia unawareness, renal impairment (glomerular filtration rate <45 mL/min/1.73 m2), cardiovascular event within the past 6 months, history of uncontrolled BP or a screening systolic BP (SBP) >160 mmHg and/or diastolic BP (DBP) >100 mmHg, alanine transaminase (ALT) or aspartate aminotransferase (AST) levels >150% the upper limit of normal (ULN), or serum triglyceride level >4.52 mmol/L (>400 mg/dL). The full list of inclusion and exclusion criteria appears in the Supplementary Data.

Interventions

Subjects were randomly assigned in a 1:1:1:1 ratio to treatment with placebo or the 5-, 10-, or 15-mg doses of RVT-1502 given as three blinded capsules once daily.

Assessments

The primary efficacy end point was the change from baseline in HbA1c during 12 weeks of treatment. Secondary efficacy end points were changes from baseline to weeks 2, 4, 8, and 12 for HbA1c; FPG; fasting glucagon; total and active GLP1; insulin; insulin resistance by HOMA (HOMA-IR); and β-cell function by HOMA (HOMA-β). The number and percentage of subjects achieving HbA1c <7% at each postbaseline visit was also determined.

As exploratory end points, assessments of least squares mean (LSM) changes in glucose, glucagon, insulin, total GLP1, active GLP1, C-peptide area under the curve between 0 and 4 h (LSM ∆AUC0–4 h), and percent changes in oral glucose tolerance test (OGTT) were performed in a subset of randomized subjects. The analytic methods used are described in the Supplementary Data.

Safety end points included changes in lipids, BP, body weight, and BMI from baseline to weeks 2, 4, 8, and 12. The frequency of treatment-emergent adverse events (AEs) and hypoglycemic events was also evaluated at each visit and after the 4-week follow-up period. Hypoglycemic events included symptomatic and asymptomatic events documented by BP values ≤70 mg/dL; probable symptomatic events in which typical hypoglycemia symptoms were not accompanied by a plasma glucose determination but which were presumed to be caused by a plasma glucose concentration ≤70 mg/dL; and severe hypoglycemia, which was defined as hypoglycemia with symptoms requiring assistance from another person. Additional safety assessments included clinical laboratory tests (chemistry, hematology, and urinalysis), 12-lead electrocardiograms (ECGs), physical examinations, and other vital signs (temperature and pulse rate) at each visit. Trough plasma concentrations of RVT-1502 were assessed in samples collected from subjects in the RVT-1502 dose groups during weeks 2, 4, 8, and 12.

Statistical Methods

Subject information, such as disposition and demographic information, was summarized descriptively. Primary and secondary efficacy analyses were conducted on the intention-to-treat population, which was defined as all randomized subjects who received at least one dose of study drug and had a baseline and at least one postbaseline HbA1c measurement. The safety population included all subjects who received at least one dose of study drug. The OGTT population included all subjects in the safety population who had a baseline OGTT assessment and at least one postrandomization OGTT assessment. The primary end point of change in HbA1c from baseline to week 12 was analyzed by ANCOVA, with treatment group as a factor and baseline HbA1c as a covariate. If the week 12 measurement was missing, the last observation carried forward (LOCF) algorithm was applied to impute the missing week 12 value. A sample size of 33 subjects who completed the study per treatment group was determined to be sufficient to provide >90% power to detect between-group differences in mean HbA1c changes from a baseline of 0.5%, assuming an SD of 0.6% using a two-sided significance level of 0.05. To accommodate a dropout rate of 10% from randomization to study completion, a sample size of 37 randomized subjects per treatment group (148 subjects in total) was planned.

The LSMs, SEs, and two-tailed 95% CI for each treatment group and for each comparison were calculated. Two-sided P values were used to test for significance of within–treatment group changes from baseline and to make comparisons between treatment groups. Before this analysis was performed, the data were inspected for normality and homogeneity of variance, and a nonparametric method was applied. The same ANCOVA model and descriptive statistics used for the primary efficacy end point were applied to HbA1c, FPG, GLP1 (total and active), insulin, HOMA-IR, and HOMA-β at each scheduled visit without LOCF and at week 12 with LOCF.

The change in fasting lipids, SBP, DBP, body weight, and BMI from baseline to weeks 2, 4, 8, and 12 without LOCF was analyzed in the safety population (defined as all randomized subjects who received at least one dose of study drug) using a mixed-model repeated-measures procedure implemented with SAS PROC MIXED. The factors in the model were stratification group, treatment group, baseline value, visit, and the treatment group-by-visit interactions. An unstructured covariance matrix was used (TYPE = UN). No imputation was performed. The LSMs for change from baseline at each visit were estimated and compared between treatment groups.

Subject Disposition and Baseline Characteristics

A total of 166 subjects were randomized; 134 (80.7%) completed the study, and 32 (19.3%) withdrew early (Supplementary Fig. 2). Overall, the most common reason for withdrawal was loss to follow-up (6.6%) or withdrawal by subject for personal reasons (6.6%). Only one subject was withdrawn from the study because of an AE, which was assessed by the investigator as not related to study drug.

Table 1 shows baseline characteristics of the study population. Overall, most subjects were white and male, and 65% were Hispanic. A large majority (91.6%) of subjects did not require metformin stabilization or a washout period. At baseline, mean HbA1c was 8.2–8.3% (66–67 mmol/mol), and mean FPG ranged from 9.0 mmol/L (162 mg/dL) to 9.9 mmol/L (178 mg/dL). A total of 108 (65.1%) subjects were stratified to the HbA1c ≤8.5% group, and 58 (34.9%) were stratified to the HbA1c >8.5% group. No meaningful differences in other demographic or baseline characteristics were noted across treatment groups.

Table 1

Baseline demographics and characteristics

CharacteristicPlacebo (n = 41)RVT-1502 5 mg (n = 43)RVT-1502 10 mg (n = 40)RVT-1502 15 mg (n = 42)Total (N = 166)
Age (years), mean (SD) 56.0 (8.0) 56.9 (8.5) 54.4 (9.0) 58.1 (8.1) 56.4 (8.4) 
Female, n (%) 17 (41.5) 25 (58.1) 15 (37.5) 18 (42.9) 75 (45.2) 
Race and ethnicity, n (%)      
 White 35 (85.4) 32 (74.4) 28 (70.0) 36 (85.7) 131 (78.9) 
 Black 6 (14.6) 10 (23.3) 9 (22.5) 5 (11.9) 30 (18.1) 
 Asian 0 (0.0) 1 (2.3) 2 (5.0) 1 (2.4) 4 (2.4) 
 Other 0 (0.0) 0 (0.0) 1 (2.5) 0 (0.0) 1 (0.6) 
 Hispanic or Latino 29 (70.7) 28 (65.1) 25 (62.5) 26 (61.9) 108 (65.1) 
HbA1c, mean (SD)      
 % 8.2 (1.0) 8.2 (1.1) 8.3 (0.9) 8.2 (0.9) 8.2 (1.0) 
 mmol/mol 66 (11.0) 66 (11.7) 67 (10.2) 66 (9.8) 66 (10.6) 
FPG, mean (SD)      
 mmol/L 9.0 (2.5) 9.7 (2.6) 9.9 (2.6) 9.5 (2.2) 9.5 (2.5) 
 mg/dL 161.6 (45.5) 175.0 (46.5) 177.5 (46.7) 171.8 (38.7) 171.5 (44.5) 
Weight (kg), mean (SD) 88.4 (18.2) 86.1 (13.8) 84.4 (16.8) 85.4 (16.2) 86.1 (16.2) 
BMI (kg/m2), mean (SD) 31.8 (5.1) 31.9 (4.2) 30.5 (4.3) 31.2 (4.0) 31.4 (4.4) 
Stratification group, n (%)      
 HbA1c ≤8.5% 27 (65.9) 28 (65.1) 26 (65.0) 27 (64.3) 108 (65.1) 
 HbA1c >8.5% 14 (34.1) 15 (34.9) 14 (35.0) 15 (35.7) 58 (34.9) 
Required metformin stabilization or washout period, n (%) 1 (2.4) 4 (9.3) 5 (12.5) 4 (9.5) 14 (8.4) 
CharacteristicPlacebo (n = 41)RVT-1502 5 mg (n = 43)RVT-1502 10 mg (n = 40)RVT-1502 15 mg (n = 42)Total (N = 166)
Age (years), mean (SD) 56.0 (8.0) 56.9 (8.5) 54.4 (9.0) 58.1 (8.1) 56.4 (8.4) 
Female, n (%) 17 (41.5) 25 (58.1) 15 (37.5) 18 (42.9) 75 (45.2) 
Race and ethnicity, n (%)      
 White 35 (85.4) 32 (74.4) 28 (70.0) 36 (85.7) 131 (78.9) 
 Black 6 (14.6) 10 (23.3) 9 (22.5) 5 (11.9) 30 (18.1) 
 Asian 0 (0.0) 1 (2.3) 2 (5.0) 1 (2.4) 4 (2.4) 
 Other 0 (0.0) 0 (0.0) 1 (2.5) 0 (0.0) 1 (0.6) 
 Hispanic or Latino 29 (70.7) 28 (65.1) 25 (62.5) 26 (61.9) 108 (65.1) 
HbA1c, mean (SD)      
 % 8.2 (1.0) 8.2 (1.1) 8.3 (0.9) 8.2 (0.9) 8.2 (1.0) 
 mmol/mol 66 (11.0) 66 (11.7) 67 (10.2) 66 (9.8) 66 (10.6) 
FPG, mean (SD)      
 mmol/L 9.0 (2.5) 9.7 (2.6) 9.9 (2.6) 9.5 (2.2) 9.5 (2.5) 
 mg/dL 161.6 (45.5) 175.0 (46.5) 177.5 (46.7) 171.8 (38.7) 171.5 (44.5) 
Weight (kg), mean (SD) 88.4 (18.2) 86.1 (13.8) 84.4 (16.8) 85.4 (16.2) 86.1 (16.2) 
BMI (kg/m2), mean (SD) 31.8 (5.1) 31.9 (4.2) 30.5 (4.3) 31.2 (4.0) 31.4 (4.4) 
Stratification group, n (%)      
 HbA1c ≤8.5% 27 (65.9) 28 (65.1) 26 (65.0) 27 (64.3) 108 (65.1) 
 HbA1c >8.5% 14 (34.1) 15 (34.9) 14 (35.0) 15 (35.7) 58 (34.9) 
Required metformin stabilization or washout period, n (%) 1 (2.4) 4 (9.3) 5 (12.5) 4 (9.5) 14 (8.4) 

Baseline was defined as the measurement at week 1, day 1. If missing, the last valid measurement on or before the first date administration of study drug was used as baseline.

Glycemic Efficacy

After 12 weeks of treatment, mean HbA1c changes relative to placebo were −0.7% (95% CI −1.1 to −0.4%; P < 0.001), −0.8% (−1.1 to −0.4%; P < 0.001), and −1.1% (−1.4 to −0.7%; P < 0.001) in the RVT-1502 5-, 10-, and 15-mg groups, respectively (Fig. 1A and Supplementary Table 1). The change from baseline was significantly different from placebo (P ≤ 0.0003) at weeks 2, 4, 8, and 12 for all RVT-1502 dose groups. As shown in Fig. 2, 8 of 41 (19.5%) subjects receiving placebo and 17 of 43 (39.5%), 15 of 38 (39.5%), and 18 of 40 (45.0%) of those receiving RVT-1502 5, 10, and 15 mg, respectively, achieved an HbA1c <7% (odds ratio [OR] vs. placebo 3.7–4.4; P ≤ 0.022). An HbA1c <6.5% was achieved by 7 of 43 (16.3%), 7 of 38 (18.4%), and 10 of 40 (25.0%) of subjects in the RVT-1502 5-, 10-, and 15-mg groups, respectively, compared with 2 of 41 (4.9%) of the placebo group (OR vs. placebo 4.4–7.9; P < 0.05 with RVT-1502 10 and 15 mg) (Fig. 2). FPG changes relative to placebo were −2.1 mmol/L (−38 mg/dL) (95% CI −2.9 to −1.3 mmol/L [−53 to −23 mg/dL]; P < 0.001), −2.2 mmol/L (−40 mg/dL) (−3.1 to −1.3 mmol/L [−55 to −24 mg/dL]; P < 0.001), and −2.6 mmol/L (−47 mg/dL) (−3.5 to −1.8 mmol/L [−63 to −32 mg/dL]; P < 0.001) after 12 weeks (Fig. 1B).

Figure 1

Glycemic control over 12 weeks in the intention-to-treat population (data are from patients with nonmissing values from baseline and the specified visit). A: LSM change from baseline in HbA1c. Placebo-adjusted differences at week 12 were −0.7% (95% CI −1.1 to −0.4%), −0.8% (−1.1 to −0.4%), and −1.1% (−1.4 to −0.7%) for RVT-1502 5, 10, and 15 mg, respectively (P < 0.001 vs. placebo for all doses). B: LSM change from baseline in FPG. Placebo-adjusted differences at week 12 were −2.1 mmol/L (95% CI −2.9 to −1.3 mmol/L), −2.2 mmol/L (−3.1 to −1.3 mmol/L), and −2.6 mmol/L (−3.5 to −1.8 mmol/L) for RVT-1502 5, 10, and 15 mg, respectively (P < 0.001 vs. placebo for all doses). Error bars represent SE.

Figure 1

Glycemic control over 12 weeks in the intention-to-treat population (data are from patients with nonmissing values from baseline and the specified visit). A: LSM change from baseline in HbA1c. Placebo-adjusted differences at week 12 were −0.7% (95% CI −1.1 to −0.4%), −0.8% (−1.1 to −0.4%), and −1.1% (−1.4 to −0.7%) for RVT-1502 5, 10, and 15 mg, respectively (P < 0.001 vs. placebo for all doses). B: LSM change from baseline in FPG. Placebo-adjusted differences at week 12 were −2.1 mmol/L (95% CI −2.9 to −1.3 mmol/L), −2.2 mmol/L (−3.1 to −1.3 mmol/L), and −2.6 mmol/L (−3.5 to −1.8 mmol/L) for RVT-1502 5, 10, and 15 mg, respectively (P < 0.001 vs. placebo for all doses). Error bars represent SE.

Close modal
Figure 2

HbA1c target achievement in each group after 12 weeks of therapy. OR, 95% CI, and P values are based on a logistic regression model with treatment group as a factor and baseline HbA1c as a covariate. Data are week 12 LOCF.

Figure 2

HbA1c target achievement in each group after 12 weeks of therapy. OR, 95% CI, and P values are based on a logistic regression model with treatment group as a factor and baseline HbA1c as a covariate. Data are week 12 LOCF.

Close modal

Nonglycemic End Points

Mean fasting glucagon increased relative to placebo by 118 ng/L (95% CI 43–192 ng/L; P = 0.0021), 268 ng/L (191–345 ng/L; P < 0.0001), and 335 ng/L (260–410 ng/L; P < 0.0001) after 12 weeks with RVT-1502 5, 10, and 15 mg, respectively (Supplementary Table 1). Mean fasting active GLP1 increased significantly from baseline with RVT-1502 5 and 10 mg, although differences from placebo were not statistically significant (Supplementary Table 1). Fasting total GLP1 increased significantly from baseline with all three RVT-1502 doses, and the difference between the 10-mg dose and placebo was significant (LSM difference from placebo 1.88 pmol/L [95% CI 0.38–3.38]; P = 0.0145) (Supplementary Table 1). The LSM difference in HOMA-IR between RVT-1502 5 mg and placebo was −2.56 (−5.04 to −0.07; P = 0.0436). No other statistically significant differences from placebo were observed for fasting insulin, HOMA-IR, or HOMA-β. Mean HOMA-β increased from baseline in the 15-mg group (P = 0.0120), and there was a trend toward an increase with all RVT-1502 treatment groups and a decrease with placebo (Supplementary Table 1).

Postprandial End Points

During the exploratory OGTT evaluation, the mean fasting glucose before oral glucose administration (preglucose load) was reduced at week 12 compared with week 1 in subjects administered RVT-1502, and the incremental change (LSM ∆AUC0–4 h) in glucose was decreased (Supplementary Fig. 3A). For example, in the 15-mg group, there was a 31.7% decrease in the glucose LSM ∆AUC0–4 h. Although the mean preglucose load glucagon level was increased with RVT-1502 treatment at week 12 compared with week 1, the incremental change in glucagon generally decreased compared with week 1 (Supplementary Fig. 3B). In contrast, while the preglucose load insulin levels were unchanged at week 12, insulin incrementally increased with RVT-1502 treatment compared with week 1 (Supplementary Fig. 3C). The incremental change in C-peptide mirrored the increases seen in insulin (data not shown). Subjects receiving placebo exhibited no incremental changes between weeks 1 and 12 for glucose, glucagon, or insulin (Supplementary Fig. 3). The LSM ∆AUC0–4 h for total and active GLP1 did not change significantly between weeks 1 and 12 in any treatment group (Supplementary Fig. 3).

Safety and Tolerability

All three doses of RVT-1502 were safe and well tolerated after 12 weeks on the basis of evaluation of AEs, clinical laboratory variables, ECG observations, and vital signs. Overall, 59 (35.5%) subjects experienced an AE during or after treatment with RVT-1502: 17 (39.5%), 11 (27.5%), and 16 (38.1%) subjects receiving RVT-1502 5, 10, and 15 mg, respectively, and 15 (36.6%) receiving placebo (Supplementary Table 2). The majority of AEs were considered mild or moderate in severity.

No deaths occurred during the study. Three patients experienced serious AEs: one receiving placebo (pyelonephritis, acute kidney injury, leukocytosis, and liver function test elevation), one receiving RVT-1502 10 mg (transient ischemic attack, which occurred 30 days after the last dose), and one receiving RVT-1502 15 mg (angina pectoris). None of these events were considered related to study drug, and no subjects were withdrawn because of these events.

AEs experienced by five (11.6%), three (7.5%), and four (9.5%) subjects receiving RVT-1502 5, 10, and 15 mg, respectively, and one (2.4%) receiving placebo were determined to be treatment related (Supplementary Table 2). No dose-related trends were observed. The most frequently reported study drug–related AEs were diarrhea, increased AST, proteinuria, and urinary tract infection (Supplementary Table 2).

Hypoglycemia

Overall, 10 (8.0%) subjects randomized to RVT-1502 had 19 mild hypoglycemic events during the study as defined by blood glucose ≤70 mg/dL (Supplementary Table 3). Eight subjects had events categorized as asymptomatic (three, one, and four subjects in the RVT-1502 5-, 10-, and 15-mg groups, respectively). One subject (5-mg group) had a documented symptomatic event, and one (5-mg group) had a probable symptomatic event. One subject in the RVT1502 5-mg group and one in the 10-mg group had a blood glucose <54 mg/dL, and no subjects had a severe hypoglycemic event.

Liver Function

Mild increases in mean ALT and AST levels were observed with RVT-1502 treatment. Mean values remained within normal limits, were reversible, and did not appear to be dose related (Fig. 3). No concomitant increases were observed in mean γ-glutamyl transferase, alkaline phosphatase, or total bilirubin levels. No occurrences of Hy’s Law were observed. One subject in the RVT-1502 10-mg group experienced a single incidence of elevated ALT >3× ULN at week 2, and one subject in the RVT-1502 15-mg group experienced a single incidence of elevated AST >3× ULN at week 4. In both cases, transaminase elevations were not present when measured at subsequent visits. Neither subject had an associated increase in bilirubin. No notable changes from baseline in mean values were observed for any other chemistry laboratory parameters.

Figure 3

Mean ALT (A) and AST (B) levels over the 12-week treatment and 4-week follow-up periods. Error bars represent SEM.

Figure 3

Mean ALT (A) and AST (B) levels over the 12-week treatment and 4-week follow-up periods. Error bars represent SEM.

Close modal

Cardiovascular and Other Safety End Points

Transient increases in lipid parameters occurred during the treatment period, but no significant differences between RVT-1502 and placebo were observed at week 12 (Supplementary Fig. 4). From baseline to week 12, SBP increased by 5.0 mmHg (95% CI 1.4–8.7 mmHg), 2.1 mmHg (−1.8 to 6.0), and 1.7 mmHg (−2.1 to 5.5) with RVT-1502 5, 10, and 15 mg, respectively, and decreased by 1.8 mmHg (−5.5 to 2.0) with placebo. LSM changes in DBP from baseline to week 12 were 2.6 mmHg (0.4–4.9), 3.2 mmHg (0.8–5.7), 1.6 mmHg (−0.8 to 4.0), and −1.1 mmHg (−3.4 to 1.3) with RVT-1502 5, 10, and 15 mg and placebo, respectively. The increases in SBP and DBP compared with placebo were neither dose related nor consistent across time (Supplementary Fig. 5). No notable changes from baseline in body weight (Supplementary Fig. 6), BMI, ECGs, heart rate, or other vital signs were observed.

In this phase 2 study of RVT-1502, a novel GRA, all doses of the drug were well tolerated and significantly reduced HbA1c and FPG over 12 weeks relative to placebo. Placebo-adjusted changes in both HbA1c and FPG were statistically significant at week 2 and remained so throughout the treatment period, with 12-week differences from placebo of 1.1% and 2.6 mmol/L (47 mg/dL), respectively. The incidence of hypoglycemia was low, and no severe events occurred during the study. These results support this GRA as a potential novel treatment for type 2 diabetes.

Glucagon receptor antagonism to treat hyperglycemia in type 2 diabetes by reducing hepatic glucose production (5,6) has been pursued for several decades, but no drugs with this mechanism of action are currently available for clinical use. Despite promising efficacy results with previous compounds, adverse effects on lipids, BP, weight, and liver enzymes (particularly isolated ALT and AST) and increases in hypoglycemia have so far hampered clinical development of GRAs (5,6,2426,30). RVT-1502 shows similar reductions in HbA1c at lower doses than other GRAs tested to date, which could potentially reduce the risk of off-target adverse effects during chronic therapy. In addition, the distinct chemical structure of RVT-1502 compared with other small molecule GRAs may lead to a unique receptor pharmacology that plays a role in the clinical profile observed in phase 2 studies (28). In this study, the glycemic reductions observed with RVT-1502 were not accompanied by clinically significant, dose-related changes in lipids (including total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides) and body weight. Although hypoglycemia was observed, the frequency was low relative to the associated glucose reduction. Mild elevations in BP were inconsistent and not dose responsive and will require further evaluation in longer studies with more-precise BP evaluations.

Elevations in ALT, and to a lesser extent AST, have been observed in the clinical trials of other GRAs, including small molecules (2123,31,32), a monoclonal antibody (33), and an antisense oligonucleotide (34), suggesting that these changes may be a pharmacological effect of interfering with glucagon receptor signaling. Although the mechanism is not known, it is possible that the changes in liver enzymes may be due to increased glycogen content or an alteration in the handling of amino acids that would otherwise be used for gluconeogenesis (3537). There was an increase in hepatic fat in the study of GRA LY2409021, which suggests other pathophysiologic mechanisms may be at work (24,35). In the current study, mild increases in mean ALT and AST were observed but were not dose related or associated with increases in bilirubin or other symptoms of liver toxicity, and levels returned to baseline during the follow-up period. Two subjects experienced transient transaminase increases >3× ULN, but these had resolved by subsequent visits with continued dosing. Larger clinical trials of longer duration will be required to fully examine the effects of RVT-1502 on liver metabolism.

In the current study, mean fasting glucagon increased relative to placebo with all three RVT-1502 doses, as has been reported with other GRAs (21,22,3032). In addition, there were increases in total and active GLP1 relative to baseline with some doses of RVT-1502, and a significant increase in total GLP1 with RVT-1502 10 mg compared with placebo. Preclinical models have demonstrated that GLP1 contributes to the improved glucose tolerance in the setting of reduced glucagon receptor signaling (38,39). Taken together with our results, these findings suggest a mechanism that merits further investigation.

Mean fasting insulin concentrations exhibited no significant change from baseline during 12 weeks of treatment with all dose groups of RVT-1502. Considered in the context of the reduction in HbA1c and FPG, this result may reflect increased hepatic insulin sensitivity in the fasting state. The trends observed in HOMA-IR and HOMA-β tend to support this possibility. Although fasting insulin did not change from baseline in response to RVT-1502, insulin levels increased during the OGTT. As might be expected, the increased insulin levels in subjects who received RVT-1502 during the OGTT were followed by a greater decrement in subsequent glucose levels compared with placebo. This confirms a previous observation made during an OGTT conducted in the phase 1 multiple-ascending dose study (29). Exogenous administration of GLP1-RAs has been shown to increase insulin levels during an OGTT (40,41). Thus, the apparent improvement in insulin sensitivity and insulin secretion in response to glucose load in subjects who received RVT-1502 may be related to the observed increases in fasting total and active GLP1 in this study. The increases in fasting total and active GLP1, as well as the higher postprandial insulin levels, may be responsible for the decrease in glucagon levels after glucose load because glucagon secretion is normally highly sensitive to insulin and GLP1 levels. The clinical significance of these findings suggests that inhibition of the glucagon response by RVT-1502 in the fasted state may abrogate the increased overnight gluconeogenesis found in early type 2 diabetes, as reflected by the decrease in FPG, while the potential concomitant improvements in fasting insulin sensitivity and insulin secretion in response to an oral glucose load may address the impaired insulin action in the fed state in clinically manifest type 2 diabetes.

RVT-1502 was generally well tolerated and demonstrated a low frequency and severity of AEs during the study similar to placebo. There were no drug-related serious AEs, and no subjects were withdrawn from the study because of a drug-related AE. Mild increases in mean transaminase levels were observed, but overall values remained below the ULN. There were no other clinically meaningful or dose-dependent changes in hematology, clinical chemistry, urinalysis, ECG, or vital signs. The overall frequency of study drug–related AEs was higher with RVT-1502 versus placebo, but no AE occurred in more than one subject in any treatment group, and no dose-related trends were observed. The GRA mechanism of action is potentially associated with hypoglycemia. However, in this study, as in the recent phase 2 trials of LY2409021 and PF-06291874 (22,30), improvements in HbA1c were associated with a relatively low incidence of hypoglycemia, with no severe events and very few symptomatic events. Weight increased modestly but significantly in the PF-06291874 study (30). In this trial, weight remained stable in all treatment groups.

Mild increases in average SBP and DBP were observed at week 12 in the RVT-1502 dose groups, but no consistent time- or dose-related increases in SBP and DBP compared with placebo were observed during 12 weeks of treatment. Elevations in BP have been observed in clinical trials with other GRAs, including in a 6-week study of LY2409021 using 24-h ambulatory BP monitoring in subjects with type 2 diabetes (23,30,33,34). The mechanism by which GRAs could increase BP remains unexplained.

In conclusion, glucagon receptor antagonism with RVT-1502 significantly lowers HbA1c and FPG levels, with a low incidence of hypoglycemia. Transient changes in liver enzymes, mild increases in BP, and the incidence of hypoglycemia observed in this study warrant further investigation. Possible end points to be considered for future clinical trials would include measurement of liver fat, 24-h BP monitoring, and continuous glucose monitoring. The results of the current study support continued clinical development of RVT-1502 in subjects with type 2 diabetes.

Acknowledgments. The authors thank Robert Henry (University of California San Diego) for advice and consultation, the participating clinical site staff and principal investigators for study conduct, and the study team members from Medpace for contributions to the study design, implementation, and management. The authors also thank Amanda Justice (independent consultant, Brooklyn, NY) for medical writing and editorial support, which was funded by Metavant Pharmaceuticals, Inc.

Funding and Duality of Interest. This study was supported and conducted by Ligand Pharmaceuticals, Inc. J.H.P. has received consulting fees from Diasome, Eli Lilly, Mannkind, Novo Nordisk, and Sanofi. D.D. has received consulting fees from Eli Lilly and Intarcia and research support from Merck and Eli Lilly. J.P.F. has received research support from AbbVie, Akcea, Allergan, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Cirius, Cymabay, Eli Lilly, Genentech, IONIS, Janssen, Lexicon, Ligand Pharmaceuticals, Madrigal, Merck, Mylan, NGM Biopharmaceuticals, Novartis, Novo Nordisk, Pfizer, Sanofi, Second Genome, and Theracos. He has served on advisory boards or received fees for consulting from Boehringer Ingelheim, Eli Lilly, Gilead, LifeScan, Merck, Novo Nordisk, and Sanofi. E.G.V., J.D.P., L.Z., and K.B.M. are employees of and own stock in Ligand Pharmaceuticals Incorporated. J.R. reports receiving grants from Applied Therapeutics, AstraZeneca, Boehringer Ingelheim, Eli Lilly, Genentech, Intarcia, Janssen, Lexicon, Merck, Novo Nordisk, Oramed, PegBio, Pfizer, and Sanofi. He has received honoraria or consulting fees from Applied Therapeutics, Boehringer Ingelheim, Eli Lilly, Intarcia, Janssen, MannKind, Novo Nordisk, and Sanofi. No other potential conflicts of interest relevant to this article were reported.

As of March 2018, Metavant Sciences GmbH holds certain rights to develop and commercialize RVT-1502.

Author Contributions. J.H.P., D.D., J.P.F., E.G.V., J.D.P., J.R., G.W., M.A.Z., L.Z., and K.B.M. participated in the drafting and critical revision of the manuscript. J.H.P., D.D., J.P.F., J.D.P., J.R., G.W., and M.A.Z. conducted the study, including data acquisition, analysis, and interpretation. J.H.P., E.G.V., J.D.P., G.W., and K.B.M. contributed to the study design. D.D., J.P.F., and M.A.Z. performed the statistical analysis. E.G.V. and K.B.M. conceived the study and oversaw interpretation of data. G.W. and M.A.Z. contributed to the statistical design. J.H.P. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 78th Scientific Sessions of the American Diabetes Association, Orlando, FL, 22–26 June 2018.

Clinical trial reg. no. NCT02851849, clinicaltrials.gov.

This article is featured in a podcast available at http://www.diabetesjournals.org/content/diabetes-core-update-podcasts.

1.
Ali
S
,
Drucker
DJ
.
Benefits and limitations of reducing glucagon action for the treatment of type 2 diabetes
.
Am J Physiol Endocrinol Metab
2009
;
296
:
E415
E421
2.
Edgerton
DS
,
Cherrington
AD
.
Glucagon as a critical factor in the pathology of diabetes
.
Diabetes
2011
;
60
:
377
380
3.
Dunning
BE
,
Gerich
JE
.
The role of alpha-cell dysregulation in fasting and postprandial hyperglycemia in type 2 diabetes and therapeutic implications
.
Endocr Rev
2007
;
28
:
253
283
4.
Drucker
DJ
,
Nauck
MA
.
The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes
.
Lancet
2006
;
368
:
1696
1705
5.
Lefèbvre
PJ
,
Paquot
N
,
Scheen
AJ
.
Inhibiting or antagonizing glucagon: making progress in diabetes care
.
Diabetes Obes Metab
2015
;
17
:
720
725
6.
Bagger
JI
,
Knop
FK
,
Holst
JJ
,
Vilsbøll
T
.
Glucagon antagonism as a potential therapeutic target in type 2 diabetes
.
Diabetes Obes Metab
2011
;
13
:
965
971
7.
Degn
KB
,
Juhl
CB
,
Sturis
J
, et al
.
One week’s treatment with the long-acting glucagon-like peptide 1 derivative liraglutide (NN2211) markedly improves 24-h glycemia and alpha- and beta-cell function and reduces endogenous glucose release in patients with type 2 diabetes
.
Diabetes
2004
;
53
:
1187
1194
8.
Farngren
J
,
Persson
M
,
Schweizer
A
,
Foley
JE
,
Ahrén
B
.
Vildagliptin reduces glucagon during hyperglycemia and sustains glucagon counterregulation during hypoglycemia in type 1 diabetes
.
J Clin Endocrinol Metab
2012
;
97
:
3799
3806
9.
Christensen
M
,
Bagger
JI
,
Vilsbøll
T
,
Knop
FK
.
The alpha-cell as target for type 2 diabetes therapy
.
Rev Diabet Stud
2011
;
8
:
369
381
10.
Conarello
SL
,
Jiang
G
,
Mu
J
, et al
.
Glucagon receptor knockout mice are resistant to diet-induced obesity and streptozotocin-mediated beta cell loss and hyperglycaemia
.
Diabetologia
2007
;
50
:
142
150
11.
Xiong
Y
,
Guo
J
,
Candelore
MR
, et al
.
Discovery of a novel glucagon receptor antagonist N-[(4-{(1S)-1-[3-(3, 5-dichlorophenyl)-5-(6-methoxynaphthalen-2-yl)-1H-pyrazol-1-yl]ethyl}phenyl)carbonyl]-β-alanine (MK-0893) for the treatment of type II diabetes
.
J Med Chem
2012
;
55
:
6137
6148
12.
Madsen
P
,
Kodra
JT
,
Behrens
C
, et al
.
Human glucagon receptor antagonists with thiazole cores. A novel series with superior pharmacokinetic properties
.
J Med Chem
2009
;
52
:
2989
3000
13.
Winzell
MS
,
Brand
CL
,
Wierup
N
, et al
.
Glucagon receptor antagonism improves islet function in mice with insulin resistance induced by a high-fat diet
.
Diabetologia
2007
;
50
:
1453
1462
14.
Rivera
N
,
Everett-Grueter
CA
,
Edgerton
DS
, et al
.
A novel glucagon receptor antagonist, NNC 25-0926, blunts hepatic glucose production in the conscious dog
.
J Pharmacol Exp Ther
2007
;
321
:
743
752
15.
Shen
DM
,
Zhang
F
,
Brady
EJ
, et al
.
Discovery of novel, potent, and orally active spiro-urea human glucagon receptor antagonists
.
Bioorg Med Chem Lett
2005
;
15
:
4564
4569
16.
Qureshi
SA
,
Rios Candelore
M
,
Xie
D
, et al
.
A novel glucagon receptor antagonist inhibits glucagon-mediated biological effects
.
Diabetes
2004
;
53
:
3267
3273
17.
Sloop
KW
,
Cao
JX
,
Siesky
AM
, et al
.
Hepatic and glucagon-like peptide-1-mediated reversal of diabetes by glucagon receptor antisense oligonucleotide inhibitors
.
J Clin Invest
2004
;
113
:
1571
1581
18.
Liang
Y
,
Osborne
MC
,
Monia
BP
, et al
.
Reduction in glucagon receptor expression by an antisense oligonucleotide ameliorates diabetic syndrome in db/db mice
.
Diabetes
2004
;
53
:
410
417
19.
Gu
W
,
Yan
H
,
Winters
KA
, et al
.
Long-term inhibition of the glucagon receptor with a monoclonal antibody in mice causes sustained improvement in glycemic control, with reversible alpha-cell hyperplasia and hyperglucagonemia
.
J Pharmacol Exp Ther
2009
;
331
:
871
881
20.
Yan
H
,
Gu
W
,
Yang
J
, et al
.
Fully human monoclonal antibodies antagonizing the glucagon receptor improve glucose homeostasis in mice and monkeys
.
J Pharmacol Exp Ther
2009
;
329
:
102
111
21.
Kazierad
DJ
,
Bergman
A
,
Tan
B
, et al
.
Effects of multiple ascending doses of the glucagon receptor antagonist PF-06291874 in patients with type 2 diabetes mellitus
.
Diabetes Obes Metab
2016
;
18
:
795
802
22.
Kazda
CM
,
Ding
Y
,
Kelly
RP
, et al
.
Evaluation of efficacy and safety of the glucagon receptor antagonist LY2409021 in patients with type 2 diabetes: 12- and 24-week phase 2 studies
.
Diabetes Care
2016
;
39
:
1241
1249
23.
Kelly
RP
,
Garhyan
P
,
Raddad
E
, et al
.
Short-term administration of the glucagon receptor antagonist LY2409021 lowers blood glucose in healthy people and in those with type 2 diabetes
.
Diabetes Obes Metab
2015
;
17
:
414
422
24.
Guzman
CB
,
Zhang
XM
,
Liu
R
, et al
.
Treatment with LY2409021, a glucagon receptor antagonist, increases liver fat in patients with type 2 diabetes
.
Diabetes Obes Metab
2017
;
19
:
1521
1528
25.
Kazda
CM
,
Frias
J
,
Foga
I
, et al
.
Treatment with the glucagon receptor antagonist LY2409021 increases ambulatory blood pressure in patients with type 2 diabetes
.
Diabetes Obes Metab
2017
;
19
:
1071
1077
26.
Scheen
AJ
,
Paquot
N
,
Lefèbvre
PJ
.
Investigational glucagon receptor antagonists in Phase I and II clinical trials for diabetes
.
Expert Opin Investig Drugs
2017
;
26
:
1373
1389
27.
Vajda
EG
,
Potter
SC
,
Fujitaki
JM
, et al
.
LGD-6972, a potent, orally-bioavailable, small molecule glucagon receptor antagonist for the treatment of type 2 diabetes
(Abstract).
Diabetes
2012
;
61
:
A252
28.
Vajda
EG
,
Zhi
L
,
Marschke
K
.
An allosteric glucagon receptor antagonist, LGD-6972, displays biased receptor signaling
(Abstract).
Diabetes
2018
;
67
:
A298
29.
Vajda
EG
,
Logan
D
,
Lasseter
K
, et al
.
Pharmacokinetics and pharmacodynamics of single and multiple doses of the glucagon receptor antagonist LGD-6972 in healthy subjects and subjects with type 2 diabetes mellitus
.
Diabetes Obes Metab
2017
;
19
:
24
32
30.
Kazierad
DJ
,
Chidsey
K
,
Somayaji
VR
,
Bergman
AJ
,
Calle
RA
.
Efficacy and safety of the glucagon receptor antagonist PF-06291874: a 12-week, randomized, dose-response study in patients with type 2 diabetes mellitus on background metformin therapy
.
Diabetes Obes Metab
2018
;
20
:
2608
2616
31.
Engel
SS
,
Xu
L
,
Andryuk
PJ
, et al
.
Efficacy and tolerability of MK-0893, a glucagon receptor antagonist (GRA), in patients with type 2 diabetes (T2DM)
(Abstract).
Diabetes
2011
;
60
:
A85
32.
Engel
SS
,
Reitman
M
,
Xu
L
, et al
.
Glycemic and lipid effects of the short-acting glucagon receptor antagonist MK-3577 in patients with type 2 diabetes
(Abstract).
Diabetes
2012
;
61
:
A266
33.
Kelly
RP
,
Garhyan
P
,
Reynolds
VL
, et al
.
Glucagon receptor antibody LY2786890 reduced glucose levels in type 2 diabetes mellitus patients
(Abstract).
Diabetes
2015
;
64
:
ALB27
34.
Morgan
ES
,
Tai
LJ
,
Pham
NC
, et al
.
Antisense inhibition of glucagon receptor by IONIS-GCGRRx improves type 2 diabetes without increase in hepatic glycogen content in patients with type 2 diabetes on stable metformin therapy
.
Diabetes Care
2019
;
42
:
585
593
35.
Rui
L
.
Energy metabolism in the liver
.
Compr Physiol
2014
;
4
:
177
197
36.
Dean
ED
,
Li
M
,
Prasad
N
, et al
.
Interrupted glucagon signaling reveals hepatic α cell axis and role for L-glutamine in α cell proliferation
.
Cell Metab
2017
;
25
:
1362
1373.e5
37.
Galsgaard
KD
,
Winther-Sørensen
M
,
Ørskov
C
, et al
.
Disruption of glucagon receptor signaling causes hyperaminoacidemia exposing a possible liver-alpha-cell axis
.
Am J Physiol Endocrinol Metab
2018
;
314
:
E93
E103
38.
Ali
S
,
Lamont
BJ
,
Charron
MJ
,
Drucker
DJ
.
Dual elimination of the glucagon and GLP-1 receptors in mice reveals plasticity in the incretin axis
.
J Clin Invest
2011
;
121
:
1917
1929
39.
Jun
LS
,
Millican
RL
,
Hawkins
ED
, et al
.
Absence of glucagon and insulin action reveals a role for the GLP-1 receptor in endogenous glucose production
.
Diabetes
2015
;
64
:
819
827
40.
Retnakaran
R
,
Kramer
CK
,
Choi
H
,
Swaminathan
B
,
Zinman
B
.
Liraglutide and the preservation of pancreatic β-cell function in early type 2 diabetes: the LIBRA trial
.
Diabetes Care
2014
;
37
:
3270
3278
41.
Thorkildsen
C
,
Neve
S
,
Larsen
BD
,
Meier
E
,
Petersen
JS
.
Glucagon-like peptide 1 receptor agonist ZP10A increases insulin mRNA expression and prevents diabetic progression in db/db mice
.
J Pharmacol Exp Ther
2003
;
307
:
490
496
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data