Trelegy Ellipta (Page 7 of 10)
12 CLINICAL PHARMACOLOGY
12.1 Mechanism of Action
TRELEGY ELLIPTA
TRELEGY ELLIPTA contains fluticasone furoate, umeclidinium, and vilanterol. The mechanisms of action described below for the individual components apply to TRELEGY ELLIPTA. These drugs represent 3 different classes of medications (an ICS, an anticholinergic, and a LABA), each having different effects on clinical and physiological indices.
Fluticasone Furoate
Fluticasone furoate is a synthetic trifluorinated corticosteroid with anti-inflammatory activity. Fluticasone furoate has been shown in vitro to exhibit a binding affinity for the human glucocorticoid receptor that is approximately 29.9 times that of dexamethasone and 1.7 times that of fluticasone propionate. The clinical relevance of these findings is unknown.
The precise mechanism through which fluticasone furoate affects COPD and asthma symptoms is not known. Inflammation is an important component in the pathogenesis of COPD and asthma. Corticosteroids have been shown to have a wide range of actions on multiple cell types (e.g., mast cells, eosinophils, neutrophils, macrophages, lymphocytes) and mediators (e.g., histamine, eicosanoids, leukotrienes, cytokines) involved in inflammation. Specific effects of fluticasone furoate demonstrated in in vitro and in vivo models included activation of the glucocorticoid response element, inhibition of pro-inflammatory transcription factors such as NFkB, and inhibition of antigen-induced lung eosinophilia in sensitized rats. These anti-inflammatory actions of corticosteroids may contribute to their efficacy.
Umeclidinium
Umeclidinium is a long-acting muscarinic antagonist, which is often referred to as an anticholinergic. It has similar affinity to the subtypes of muscarinic receptors M1 to M5. In the airways, it exhibits pharmacological effects through inhibition of M3 receptor at the smooth muscle leading to bronchodilation. The competitive and reversible nature of antagonism was shown with human and animal origin receptors and isolated organ preparations. In preclinical in vitro as well as in vivo studies, prevention of methacholine- and acetylcholine-induced bronchoconstrictive effects was dose-dependent and lasted longer than 24 hours. The clinical relevance of these findings is unknown. The bronchodilation following inhalation of umeclidinium is predominantly a site-specific effect.
Vilanterol
Vilanterol is a LABA. In vitro tests have shown the functional selectivity of vilanterol was similar to salmeterol. The clinical relevance of this in vitro finding is unknown.
Although beta2 -receptors are the predominant adrenergic receptors in bronchial smooth muscle and beta1 -receptors are the predominant receptors in the heart, there are also beta2 -receptors in the human heart comprising 10% to 50% of the total beta-adrenergic receptors. The precise function of these receptors has not been established, but they raise the possibility that even highly selective beta2 -agonists may have cardiac effects.
The pharmacologic effects of beta2 -adrenergic agonist drugs, including vilanterol, are at least in part attributable to stimulation of intracellular adenyl cyclase, the enzyme that catalyzes the conversion of adenosine triphosphate (ATP) to cyclic-3ʹ,5ʹ-adenosine monophosphate (cyclic AMP). Increased cyclic AMP levels cause relaxation of bronchial smooth muscle and inhibition of release of mediators of immediate hypersensitivity from cells, especially from mast cells.
12.2 Pharmacodynamics
Cardiac Electrophysiology
The effect of umeclidinium/vilanterol on cardiac rhythm in subjects diagnosed with COPD was assessed using 24-hour Holter monitoring in 6- and 12-month trials: 53 subjects received umeclidinium/vilanterol 62.5/25 mcg, 281 subjects received umeclidinium/vilanterol 125/25 mcg, and 182 subjects received placebo. No clinically meaningful effects on cardiac rhythm were observed.
The cardiovascular effects from dual combinations of fluticasone furoate/vilanterol and umeclidinium/vilanterol in healthy subjects are presented below.
Fluticasone Furoate/Vilanterol Combination: Healthy Subjects: QTc interval prolongation was studied in a double-blind, multiple-dose, placebo- and positive-controlled crossover study in 85 healthy volunteers. The maximum mean (95% upper confidence bound) difference in QTcF from placebo after baseline correction was 4.9 (7.5) milliseconds and 9.6 (12.2) milliseconds seen 30 minutes after dosing for fluticasone furoate/vilanterol 200/25 mcg and fluticasone furoate/vilanterol 800/100 mcg, respectively.
A dose-dependent increase in heart rate was also observed. The maximum mean (95% upper confidence bound) difference in heart rate from placebo after baseline correction was 7.8 (9.4) beats/min and 17.1 (18.7) beats/min seen 10 minutes after dosing for fluticasone furoate/vilanterol 200/25 mcg and fluticasone furoate/vilanterol 800/100 mcg (8/4 times the recommended dosage), respectively.
Umeclidinium/Vilanterol Combination: Healthy Subjects: QTc interval prolongation was studied in a double-blind, multiple-dose, placebo- and positive-controlled crossover trial in 86 healthy subjects. The maximum mean (95% upper confidence bound) difference in QTcF from placebo after baseline correction was 4.6 (7.1) milliseconds and 8.2 (10.7) milliseconds for umeclidinium/vilanterol 125/25 mcg and umeclidinium/vilanterol 500/100 mcg (8/4 times the recommended dosage), respectively.
A dose-dependent increase in heart rate was also observed. The maximum mean (95% upper confidence bound) difference in heart rate from placebo after baseline correction was 8.8 (10.5) beats/min and 20.5 (22.3) beats/min seen 10 minutes after dosing for umeclidinium/vilanterol 125/25 mcg and umeclidinium/vilanterol 500/100 mcg, respectively.
HPA Axis Effects
Healthy Subjects: Inhaled fluticasone furoate at repeat doses up to 400 mcg was not associated with statistically significant decreases in serum or urinary cortisol in healthy subjects. Decreases in serum and urine cortisol levels were observed at fluticasone furoate exposures several-fold higher than exposures observed at the therapeutic dose.
Subjects with Chronic Obstructive Pulmonary Disease: In a trial with subjects with COPD, treatment with fluticasone furoate (50, 100, or 200 mcg)/vilanterol 25 mcg, vilanterol 25 mcg, or fluticasone furoate (100 or 200 mcg) for 6 months did not affect 24-hour urinary cortisol excretion. A separate trial with subjects with COPD demonstrated no effects on serum cortisol after 28 days of treatment with fluticasone furoate (50, 100, or 200 mcg)/vilanterol 25 mcg.
Subjects with Asthma: A randomized, double-blind, parallel-group trial in 185 subjects with asthma showed no difference between once-daily treatment with fluticasone furoate/vilanterol 100/25 mcg or fluticasone furoate/vilanterol 200/25 mcg compared with placebo on serum cortisol weighted mean (0 to 24 hours), serum cortisol AUC(0-24) , and 24-hour urinary cortisol after 6 weeks of treatment, whereas prednisolone 10 mg given once daily for 7 days resulted in significant cortisol suppression.
12.3 Pharmacokinetics
Linear pharmacokinetics was observed for fluticasone furoate (200 to 800 mcg), umeclidinium (62.5 to 500 mcg), and vilanterol (25 to 100 mcg). The pharmacokinetics of fluticasone furoate, umeclidinium, and vilanterol from TRELEGY ELLIPTA are comparable to the pharmacokinetics of fluticasone furoate, umeclidinium, and vilanterol when administered as fluticasone furoate/vilanterol or umeclidinium/vilanterol.
Systemic drug levels [steady-state Cmax and AUC(0-24) ] of fluticasone furoate, umeclidinium, and vilanterol following administration of TRELEGY ELLIPTA 100/62.5/25 mcg based on a combined pharmacokinetic dataset from 3 studies in subjects with COPD (N = 821) were within the range of those observed following administration of fluticasone furoate/vilanterol plus umeclidinium administered via 2 inhalers, fluticasone furoate/vilanterol and umeclidinium/vilanterol as the dual combinations, and after administration of fluticasone furoate, umeclidinium, and vilanterol as monotherapy.
Systemic drug levels [steady-state Cmax and AUC(0-24) ] of fluticasone furoate, umeclidinium, and vilanterol following administration of TRELEGY ELLIPTA (100/62.5/25 or 200/62.5/25 mcg) based on a population pharmacokinetic analysis from subjects with asthma (1,265 subjects for fluticasone furoate; 634 subjects for umeclidinium; 1,263 subjects for vilanterol) were within the range of those observed following administration of fluticasone furoate/vilanterol as the dual combination when compared with fluticasone furoate 100 and 200 mcg, respectively; the systemic exposure of umeclidinium 62.5 mcg following TRELEGY ELLIPTA (100/62.5/25 or 200/62.5/25 mcg) was within the range of those observed following administration of umeclidinium 62.5 mcg as monotherapy.
The pharmacokinetics of the individual components of TRELEGY ELLIPTA are presented as follows. Plasma levels of fluticasone furoate, umeclidinium, and vilanterol may not predict therapeutic effect.
Absorption
Fluticasone Furoate: Following inhaled administration of fluticasone furoate, Cmax occurred within 0.5 to 1 hour. Absolute bioavailability of fluticasone furoate when administered by inhalation was 15.2%, primarily due to absorption of the inhaled portion of the dose delivered to the lung. Oral bioavailability from the swallowed portion of the dose is low (approximately 1.3%) due to extensive first-pass metabolism. Following repeat dosing of inhaled fluticasone furoate, steady state was achieved within 6 days with up to 2.6-fold accumulation.
Umeclidinium: Following inhaled administration of umeclidinium in healthy subjects, Cmax occurred at 5 to 15 minutes. Umeclidinium is mostly absorbed from the lung after inhaled doses with minimum contribution from oral absorption. Following repeat dosing of inhaled umeclidinium, steady state was achieved within 14 days with up to 1.8-fold accumulation.
Vilanterol: Following inhaled administration of vilanterol in healthy subjects, Cmax occurred at 5 to 15 minutes. Vilanterol is mostly absorbed from the lung after inhaled doses with negligible contribution from oral absorption. Following repeat dosing of inhaled vilanterol, steady state was achieved within 14 days with up to 1.7-fold accumulation.
Distribution
Fluticasone Furoate: Following intravenous administration to healthy subjects, the mean volume of distribution at steady state was 661 L. Binding of fluticasone furoate to human plasma proteins was high (>99%).
Umeclidinium: Following intravenous administration to healthy subjects, the mean volume of distribution was 86 L. In vitro plasma protein binding in human plasma was on average 89%.
Vilanterol: Following intravenous administration to healthy subjects, the mean volume of distribution at steady state was 165 L. In vitro plasma protein binding in human plasma was on average 94%.
Elimination
Metabolism: Fluticasone Furoate: Fluticasone furoate is cleared from systemic circulation principally by hepatic metabolism via CYP3A4 to metabolites with significantly reduced corticosteroid activity. There was no in vivo evidence for cleavage of the furoate moiety resulting in the formation of fluticasone.
Umeclidinium: In vitro data showed that umeclidinium is primarily metabolized by the enzyme cytochrome P450 2D6 (CYP2D6) and is a substrate for the P-glycoprotein (P-gp) transporter. The primary metabolic routes for umeclidinium are oxidative (hydroxylation, O-dealkylation) followed by conjugation (e.g., glucuronidation), resulting in a range of metabolites with either reduced pharmacological activity or for which the pharmacological activity has not been established. Systemic exposure to the metabolites is low.
Vilanterol: In vitro data showed that vilanterol is metabolized principally by CYP3A4 and is a substrate for the P-gp transporter. Vilanterol is metabolized to a range of metabolites with significantly reduced β1 — and β2 -agonist activity.
Excretion: Fluticasone Furoate: The plasma elimination half-life following repeat-dose inhaled administration averaged 24 hours. Following intravenous dosing with radiolabeled fluticasone furoate, mass balance showed 90% of the radiolabel in the feces and 2% in the urine. Following oral dosing, radiolabel recovered in feces was 101% of the total dose and that in urine was approximately 1% of the total dose.
Umeclidinium: The effective half-life after once-daily oral dosing is 11 hours. Following intravenous dosing with radiolabeled umeclidinium, mass balance showed 58% of the radiolabel in the feces and 22% in the urine. The excretion of the drug-related material in the feces following intravenous dosing indicated elimination in the bile. Following oral dosing to healthy male subjects, radiolabel recovered in feces was 92% of the total dose and that in urine was <1% of the total dose, suggesting negligible oral absorption.
Vilanterol: The effective half-life for vilanterol, as determined from inhalation administration of multiple doses, is 11 hours. Following oral administration of radiolabeled vilanterol, mass balance showed 70% of the radiolabel in the urine and 30% in the feces.
Specific Populations
The effects of intrinsic and extrinsic factors on the pharmacokinetics of fluticasone furoate, umeclidinium, and vilanterol are shown in Figures 1, 2, 3, and 4. Based on population pharmacokinetic analyses in COPD and asthma, none of the covariates assessed (i.e., age, race, gender) had a clinically relevant effect on fluticasone furoate, umeclidinium, or vilanterol pharmacokinetics when administered as TRELEGY ELLIPTA.
Figure 1. Impact of Intrinsic Factors on the Pharmacokinetics (PK) of Fluticasone Furoate (FF), Umeclidinium (UMEC), and Vilanterol (VI) following Coadministration in COPD
Figure 2. Impact of Intrinsic Factors on the Pharmacokinetics (PK) of Fluticasone Furoate (FF), Umeclidinium (UMEC), and Vilanterol (VI) following Coadministration in Asthmaa
a Age, ethnicity, and gender comparison for TRELEGY ELLIPTA 200/62.5/25 mcg in subjects with ashtma
Figure 3. Impact of Intrinsic Factorsa and Coadministered Drugsb on the Pharmacokinetics (PK) of Fluticasone Furoate (FF) and Vilanterol (VI) following Administration as Fluticasone Furoate/Vilanterol Combination or following Vilanterol Coadministered with Umeclidinium
a Renal groups (fluticasone furoate/vilanterol 200/25 mcg) and hepatic groups (fluticasone furoate/vilanterol 200/25 mcg or fluticasone furoate/vilanterol 100/12.5 mcg).
b Compared with placebo group.
Figure 4. Impact of Intrinsic Factors and Coadministered Drugs on the Systemic Exposure of Umeclidinium
Racial or Ethnic Groups: Fluticasone Furoate: Systemic exposure [AUC(0-24) ] to inhaled fluticasone furoate was approximately 30% higher in subjects with COPD of East Asian heritage (n = 113) compared with White subjects (Figure 1). However, this higher exposure to fluticasone furoate is not expected to have clinically relevant effects on serum or urine cortisol or on efficacy in these racial groups. In East Asian subjects with asthma (Japanese, East Asian, and Southeast Asian heritage) (n = 92), there was no effect of race on pharmacokinetics of fluticasone furoate (Figure 2).
Umeclidinium: There was no effect of race on the pharmacokinetics of umeclidinium in subjects with COPD or asthma (Figures 1 and 2).
Vilanterol: There was no effect of race on the pharmacokinetics of vilanterol in subjects with COPD (Figure 1). In East Asian subjects with asthma (Japanese, East Asian, and Southeast Asian heritage) (n = 92), estimates of vilanterol Cmax at steady state were approximately 3-fold higher than non-East Asian subjects (Figure 2). However, the higher systemic exposure is not expected to have a clinically relevant effect on heart rate.
Patients with Hepatic Impairment: Fluticasone Furoate: Following repeat dosing of fluticasone furoate/vilanterol 200/25 mcg (100/12.5 mcg in the severe impairment group) for 7 days, there was an increase of 34%, 83%, and 75% in fluticasone furoate systemic exposure (AUC) in subjects with mild, moderate, and severe hepatic impairment, respectively, compared with healthy subjects (Figure 3).
In subjects with moderate hepatic impairment receiving fluticasone furoate/vilanterol 200/25 mcg, mean serum cortisol (0 to 24 hours) was reduced by 34% (90% CI: 11%, 51%) compared with healthy subjects. In subjects with severe hepatic impairment receiving fluticasone furoate/vilanterol 100/12.5 mcg, mean serum cortisol (0 to 24 hours) was increased by 14% (90% CI: -16%, 55%) compared with healthy subjects. Patients with moderate to severe hepatic disease should be closely monitored.
Umeclidinium: The impact of hepatic impairment on the pharmacokinetics of umeclidinium has been evaluated in subjects with moderate hepatic impairment (Child-Pugh score of 7-9). There was no evidence of an increase in systemic exposure to umeclidinium (Cmax and AUC) (Figure 4). There was no evidence of altered protein binding in subjects with moderate hepatic impairment compared with healthy subjects. TRELEGY ELLIPTA has not been evaluated in subjects with severe hepatic impairment.
Vilanterol: Hepatic impairment had no effect on vilanterol systemic exposure (Cmax and AUCss on Day 7) following repeat-dose administration of fluticasone furoate/vilanterol 200/25 mcg (100/12.5 mcg in the severe impairment group) for 7 days (Figure 3).
There were no additional clinically relevant effects of the fluticasone furoate/vilanterol combinations on heart rate or serum potassium in subjects with mild or moderate hepatic impairment (vilanterol 25 mcg combination) or with severe hepatic impairment (vilanterol 12.5 mcg combination) compared with healthy subjects.
Patients with Renal Impairment: Fluticasone Furoate: Systemic exposure was not increased in subjects with severe renal impairment compared with healthy subjects (Figure 3). There was no evidence of greater corticosteroid class-related systemic effects (assessed by serum cortisol) in subjects with severe renal impairment compared with healthy subjects.
Umeclidinium: The pharmacokinetics of umeclidinium has been evaluated in subjects with severe renal impairment (CrCl <30 mL/min). There was no evidence of an increase in systemic exposure to umeclidinium (Cmax and AUC) (Figure 4). There was no evidence of altered protein binding in subjects with severe renal impairment compared with healthy subjects.
Vilanterol: Systemic exposure (AUCss ) was 56% higher in subjects with severe renal impairment compared with healthy subjects (Figure 3). There was no evidence of greater beta‑agonist class-related systemic effects (assessed by heart rate and serum potassium) in subjects with severe renal impairment compared with healthy subjects.
Drug Interaction Studies
No drug-drug interaction studies have been conducted with TRELEGY ELLIPTA. The information below is from drug-drug interaction studies conducted with umeclidinium, fluticasone furoate/vilanterol, or umeclidinium/vilanterol. The potential for fluticasone furoate, umeclidinium, and vilanterol to inhibit or induce metabolic enzymes and transporter systems is negligible at low inhalation doses.
Inhibitors of Cytochrome P450 3A4: The exposure (AUC) of fluticasone furoate and vilanterol were 36% and 65% higher, respectively, when coadministered with ketoconazole 400 mg compared with placebo (Figure 3). The increase in fluticasone furoate exposure was associated with a 27% reduction in weighted mean serum cortisol (0 to 24 hours). The increase in vilanterol exposure was not associated with an increase in beta-agonist–related systemic effects on heart rate or blood potassium.
Cytochrome P450 2D6: In vitro metabolism of umeclidinium is mediated primarily by CYP2D6. However, no clinically meaningful difference in systemic exposure to umeclidinium (500 mcg) (8 times the approved dose) was observed following repeat daily inhaled dosing to normal (ultra‑rapid, extensive, and intermediate metabolizers) and CYP2D6 poor metabolizer subjects.
Inhibitors of P-glycoprotein: Fluticasone furoate, umeclidinium, and vilanterol are substrates of P‑gp. Coadministration of repeat-dose (240 mg once daily) verapamil (a moderate CYP3A4 inhibitor and a P-gp inhibitor) did not affect the vilanterol Cmax or AUC in healthy subjects (Figure 3). Drug interaction trials with a specific P-gp inhibitor and fluticasone furoate have not been conducted. The effect of the moderate P-gp transporter inhibitor verapamil (240 mg once daily) on the steady-state pharmacokinetics of umeclidinium was assessed in healthy subjects. No effect on umeclidinium Cmax was observed; however, an approximately 1.4-fold increase in umeclidinium AUC was observed (Figure 4).
All MedLibrary.org resources are included in as near-original form as possible, meaning that the information from the original provider has been rendered here with only typographical or stylistic modifications and not with any substantive alterations of content, meaning or intent.