Ivacaftor

Name: Ivacaftor

Side effects

The following adverse reaction is discussed in greater detail in other sections of the label:

  • Transaminase Elevations [see WARNINGS AND PRECAUTIONS]

Clinical Trials Experience

Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared to rates in the clinical trials of another drug and may not reflect the rates observed in clinical practice.

The overall safety profile of KALYDECO is based on pooled data from three placebo-controlled clinical trials conducted in 353 patients 6 years of age and older with CF who had a G551D mutation in the CFTR gene (Trials 1 and 2) or were homozygous for the F508del mutation (Trial 3). In addition, the following clinical trials have also been conducted [see CLINICAL PHARMACOLOGY and Clinical Studies]:

  • An 8-week, crossover design trial (Trial 4) involving 39 patients between the ages of 6 and 57 years with a G1244E, G1349D, G178R, G551S, G970R, S1251N, S1255P, S549N, or S549R mutation in the CFTR gene.
  • A 24-week, placebo-controlled trial (Trial 5) involving 69 patients between the ages of 6 and 68 years with an R117H mutation in the CFTR gene.
  • A 24-week, open-label trial (Trial 6) in 34 patients 2 to less than 6 years of age. Patients eligible for Trial 6 were those with the G551D, G1244E, G1349D, G178R, G551S, G970R, S1251N, S1255P, S549N, or S549R mutation in the CFTR gene. Of 34 patients enrolled, 32 had the G551D mutation and 2 had the S549N mutation.
  • An 8-week, crossover design trial (Trial 7) involving patients between the ages of 12 and 72 years who were heterozygous for the F508del mutation and a second CFTR mutation predicted to be responsive to ivacaftor. A total of 156 patients were randomized to and received KALYDECO.

Of the 353 patients included in the pooled analyses of patients with CF who had either a G551D mutation or were homozygous for the F508del mutation in the CFTR gene, 50% of patients were female and 97% were Caucasian; 221 received KALYDECO, and 132 received placebo from 16 to 48 weeks.

The proportion of patients who prematurely discontinued study drug due to adverse reactions was 2% for KALYDECO-treated patients and 5% for placebo-treated patients. Serious adverse reactions, whether considered drug-related or not by the investigators, that occurred more frequently in KALYDECO-treated patients included abdominal pain, increased hepatic enzymes, and hypoglycemia.

The most common adverse reactions in the 221 patients treated with KALYDECO were headache (17%), upper respiratory tract infection (16%), nasal congestion (16%), nausea (10%), rash (10%), rhinitis (6%), dizziness (5%), arthralgia (5%), and bacteria in sputum (5%).

The incidence of adverse reactions below is based upon two double-blind, placebo-controlled, 48-week clinical trials (Trials 1 and 2) in a total of 213 patients with CF ages 6 to 53 who have a G551D mutation in the CFTR gene and who were treated with KALYDECO 150 mg orally or placebo twice daily. Table 2 shows adverse reactions occurring in ≥8% of KALYDECO-treated patients with CF who have a G551D mutation in the CFTR gene that also occurred at a higher rate than in the placebo-treated patients in the two double-blind, placebo-controlled trials.

Table 2: Incidence of Adverse Drug Reactions in ≥8% of KALYDECO-Treated Patients with a G551D Mutation in the CFTR Gene and Greater than Placebo in 2 Placebo-Controlled Phase 3 Clinical Trials of 48 Weeks Duration

Adverse Reaction (Preferred Term) Incidence: Pooled 48-Week Trials
KALYDECO
N=109 n (%)
Placebo
N=104 n (%)
Headache 26 (24) 17 (16)
Oropharyngeal pain 24 (22) 19 (18)
Upper respiratory tract infection 24 (22) 14 (14)
Nasal congestion 22 (20) 16 (15)
Abdominal pain 17 (16) 13(13)
Nasopharyngitis 16 (15) 12 (12)
Diarrhea 14(13) 10 (10)
Rash 14(13) 7 (7)
Nausea 13 (12) 11 (11)
Dizziness 10 (9) 1 (1)

Adverse reactions in the 48-week clinical trials that occurred in the KALYDECO group at a frequency of 4 to 7% where rates exceeded that in the placebo group include:

Infections and infestations: rhinitis

Investigations: aspartate aminotransferase increased, bacteria in sputum, blood glucose increased, hepatic enzyme increased

Musculoskeletal and connective tissue disorders: arthralgia, musculoskeletal chest pain, myalgia

Nervous system disorders: sinus headache

Respiratory, thoracic and mediastinal disorders: pharyngeal erythema, pleuritic pain, sinus congestion, wheezing

Skin and subcutaneous tissue disorders: acne

The safety profile for the CF patients enrolled in the other clinical trials (Trials 3-7) was similar to that observed in the 48-week, placebo-controlled trials (Trials 1 and 2).

Laboratory Abnormalities

Transaminase Elevations

In Trials 1, 2, and 3 the incidence of maximum transaminase (ALT or AST) >8, >5, or >3 x ULN was 2%, 2%, and 6% in KALYDECO-treated patients and 2%, 2%, and 8% in placebo-treated patients, respectively. Two patients (2%) on placebo and 1 patient (0.5%) on KALYDECO permanently discontinued treatment for elevated transaminases, all >8 x ULN. Two patients treated with KALYDECO were reported to have serious adverse reactions of elevated liver transaminases compared to none on placebo. Transaminase elevations were more common in patients with a history of transaminase elevations [see WARNINGS AND PRECAUTIONS].

During the 24-week, open-label, clinical trial in 34 patients ages 2 to less than 6 years (Trial 6), where patients received either 50 mg (less than 14 kg) or 75 mg (14 kg or greater) ivacaftor granules twice daily, the incidence of patients experiencing transaminase elevations (ALT or AST) >3 x ULN was 14.7% (5/34). All 5 patients had maximum ALT or AST levels >8 x ULN, which returned to baseline levels following interruption of KALYDECO dosing. Transaminase elevations were more common in patients who had abnormal transaminases at baseline. KALYDECO was permanently discontinued in one patient [see WARNINGS AND PRECAUTIONS].

Clinical pharmacology

Mechanism Of Action

Ivacaftor is a potentiator of the CFTR protein. The CFTR protein is a chloride channel present at the surface of epithelial cells in multiple organs. Ivacaftor facilitates increased chloride transport by potentiating the channel open probability (or gating) of CFTR protein located at the cell surface. The overall level of ivacaftor-mediated CFTR chloride transport is dependent on the amount of CFTR protein at the cell surface and how responsive a particular mutant CFTR protein is to ivacaftor potentiation.

CFTR Chloride Transport Assay In Fisher Rat Thyroid (FRT) Cells Expressing Mutant CFTR

In order to evaluate the response of mutant CFTR protein to ivacaftor, total chloride transport was determined in Ussing chamber electrophysiology studies using a panel of FRT cell lines transfected with individual CFTR mutations. Ivacaftor increased chloride transport in FRT cells expressing CFTR mutations that result in CFTR protein being delivered to the cell surface.

Data shown in Figure 1 are the mean (n=3-7) net change over baseline in CFTR mediated chloride transport following the addition of ivacaftor in FRT cells expressing mutant CFTR proteins. The in vitro CFTR chloride response threshold was designated as a net increase of at least 10% of normal over baseline (dotted line) because it is predictive or reasonably expected to predict clinical benefit. Mutations with an increase in chloride transport of 10% or greater are considered responsive. A patient must have at least one CFTR mutation responsive to ivacaftor to be indicated. Mutations including F508del that are not responsive to ivacaftor potentiation, based on the in vitro CFTR chloride response threshold, are listed in Figure 1 below the dotted line.

Figure 1: Net Change Over Baseline (% of Normal) in CFTR-Mediated Chloride Transport Following Addition of Ivacaftor in FRT Cells Expressing Mutant CFTR (Ussing Chamber Electrophysiology Data)

*Clinical data exist for these mutations [see Clinical Studies].

#A46D, G85E, E92K, P205S, R334W,R347P, T338I, S492F, I507del, V520F, A559T, R560S, R560T, A561E, L927P, H1054D, G1061R, L1065P, R1066C, R1066H, R1066M, L1077P, H1085R, M1101K, W1282X, N1303K mutations in the CFTR gene do not meet the threshold of change in CFTR mediated chloride transport of at least 10% of normal over baseline.

Note that splice mutations cannot be studied in this FRT assay and are not included in Figure 1. Evidence of clinical efficacy exists for non-canonical splice mutations 2789+5G→A, 3272-26A→G, 3849+10kbC→T, 711+3A→G and E831X and these are listed in Table 3 below [see also Clinical Studies]. The G970R mutation causes a splicing defect resulting in little-to-no CFTR protein at the cell surface that can be potentiated by ivacaftor [see Clinical Studies].

Ivacaftor also increased chloride transport in cultured human bronchial epithelial (HBE) cells derived from CF patients who carried F508del on one CFTR allele and either G551D or R117H-5T on the second CFTR allele.

Table 3 lists mutations that are responsive to ivacaftor based on 1) a positive clinical response and/or 2) in vitro data in FRT cells indicating that ivacaftor increases chloride transport to at least 10% over baseline (% of normal).

Table 3: List of CFTR Gene Mutations that Produce CFTR Protein and are Responsive to KALYDECO

E56K G178R S549R S977F F1074L 2789+5G→A
P67L E193K G551D F1052V D1152H 3272-26A→G
R74W L206W G551S K1060T G1244E 3849+10kbC→T
D110E R347H D579G A1067T S1251N  
D110H R352Q 711+3A→G G1069R S1255P  
R117C A455E E831X R1070Q D1270N  
R117H S549N S945L R1070W G1349D  

Pharmacodynamics

Sweat Chloride Evaluation

Changes in sweat chloride (a biomarker) response to KALYDECO were evaluated in seven clinical trials [see Clinical Studies]. In a two-part, randomized, double-blind, placebo-controlled, crossover clinical trial in patients with CF who had a G1244E, G1349D, G178R, G551S, G970R, S1251N, S1255P, S549N, or S549R mutation in the CFTR gene (Trial 4), the treatment difference in mean change in sweat chloride from baseline through 8 weeks of treatment was -49 mmol/L (95% CI -57, -41). The mean changes in sweat chloride for the mutations for which KALYDECO is indicated ranged from -51 to -8, whereas the range for individual subjects with the G970R mutation was -1 to -11 mmol/L. In an open-label clinical trial in 34 patients ages 2 to less than 6 years administered either 50 mg or 75 mg of ivacaftor twice daily (Trial 6), the mean absolute change from baseline in sweat chloride through 24 weeks of treatment was -45 mmol/L (95% CI -53, -38) [see Use In Specific Populations]. In a randomized, double-blind, placebo controlled, 2-period, 3-treatment, 8-week crossover study in patients with CF age 12 years and older who were heterozygous for the F508del mutation and with a second CFTR mutation predicted to be responsive to ivacaftor (Trial 7), the treatment difference in mean change in sweat chloride from study baseline to the average of week 4 and week 8 of treatment for KALYDECO treated patients was -4.5 mmol/L (95% CI -6.7, -2.3).

There was no direct correlation between decrease in sweat chloride levels and improvement in lung function (FEV1).

Cardiac Electrophysiology

The effect of multiple doses of ivacaftor 150 mg and 450 mg twice daily on QTc interval was evaluated in a randomized, placebo-and active-controlled (moxifloxacin 400 mg) four-period crossover thorough QT study in 72 healthy subjects. In a study with demonstrated ability to detect small effects, the upper bound of the one-sided 95% confidence interval for the largest placebo adjusted, baseline-corrected QTc based on Fridericia's correction method (QTcF) was below 10 ms, the threshold for regulatory concern.

Pharmacokinetics

The pharmacokinetics of ivacaftor is similar between healthy adult volunteers and patients with CF.

After oral administration of a single 150 mg dose to healthy volunteers in a fed state, peak plasma concentrations (Tmax) occurred at approximately 4 hours, and the mean (±SD) for AUC and Cmax were 10600 (5260) ng*hr/mL and 768 (233) ng/mL, respectively.

After every 12-hour dosing, steady-state plasma concentrations of ivacaftor were reached by days 3 to 5, with an accumulation ratio ranging from 2.2 to 2.9.

Absorption

The exposure of ivacaftor increased approximately 2.5-to 4-fold when given with food that contains fat. Therefore, KALYDECO should be administered with fat-containing food. Examples of fat-containing foods include eggs, butter, peanut butter, cheese pizza, whole-milk dairy products (such as whole milk, cheese, and yogurt), etc. The median (range) Tmax is approximately 4.0 (3.0; 6.0) hours in the fed state.

KALYDECO granules (2 x 75 mg) had similar bioavailability as the 150 mg tablet when given with fat-containing food in adult subjects. The effect of food on ivacaftor absorption is similar for KALYDECO granules and the 150 mg tablet formulation.

Distribution

Ivacaftor is approximately 99% bound to plasma proteins, primarily to alpha 1-acid glycoprotein and albumin. Ivacaftor does not bind to human red blood cells.

After oral administration of 150 mg every 12 hours for 7 days to healthy volunteers in a fed state, the mean (±SD) for apparent volume of distribution was 353 (122) L.

Metabolism

Ivacaftor is extensively metabolized in humans. In vitro and clinical studies indicate that ivacaftor is primarily metabolized by CYP3A. M1 and M6 are the two major metabolites of ivacaftor in humans. M1 has approximately one-sixth the potency of ivacaftor and is considered pharmacologically active. M6 has less than one-fiftieth the potency of ivacaftor and is not considered pharmacologically active.

Elimination

Following oral administration, the majority of ivacaftor (87.8%) is eliminated in the feces after metabolic conversion. The major metabolites M1 and M6 accounted for approximately 65% of the total dose eliminated with 22% as M1 and 43% as M6. There was negligible urinary excretion of ivacaftor as unchanged parent. The apparent terminal half-life was approximately 12 hours following a single dose. The mean apparent clearance (CL/F) of ivacaftor was similar for healthy subjects and patients with CF. The CL/F (SD) for the 150 mg dose was 17.3 (8.4) L/hr in healthy subjects.

Specific populations

Pediatric Patients

The following conclusions about exposures between adults and the pediatric population are based on population PK analyses:

Pediatric Patients 2 To Less Than 6 Years Of Age Who Weigh Less Than 14 kg

Following oral administration of KALYDECO granules, 50 mg every 12 hours, the mean (±SD) steady state AUC (AUCss) was 10500 (4260) ng/mL*h and is similar to the mean AUCss of 10700 (4100) ng/mL*h in adult patients administered KALYDECO tablets, 150 mg every 12 hours.

Pediatric Patients 2 To Less Than 6 Years Of Age Who Weigh 14 kg Or Greater

Following oral administration of KALYDECO granules, 75 mg every 12 hours, the mean (±SD) AUC (AUCss) was 11300 (3820) ng/mL*h and is similar to the mean AUC in adult patients administered KALYDECO tablets, 150 mg every 12 hours.

Pediatric Patients 6 To Less Than 12 Years Of Age

Following oral administration of KALYDECO tablets, 150 mg every 12 hours, the mean (±SD) AUCss was 20000 (8330) ng/mL*h and is 87% higher than the mean AUC in adult patients administered KALYDECO tablets, 150 mg every 12 hours.

Pediatric Patients 12 To Less Than 18 Years Of Age

Following oral administration of KALYDECO tablets, 150 mg every 12 hours, the mean (±SD) AUCss was 9240 (3420) ng/mL*h and is similar to the mean AUCss in adult patients administered KALYDECO tablets, 150 mg every 12 hours.

Patients With Hepatic Impairment

Adult subjects with moderately impaired hepatic function (Child-Pugh Class B, score 7-9) had similar ivacaftor Cmax, but an approximately two-fold increase in ivacaftor AUC0-∞ compared with healthy subjects matched for demographics. Based on simulations of these results, a reduced KALYDECO dose to one tablet or packet of granules once daily is recommended for patients with moderate hepatic impairment. The impact of mild hepatic impairment (Child-Pugh Class A) on the pharmacokinetics of ivacaftor has not been studied, but the increase in ivacaftor AUC0-∞ is expected to be less than two-fold. Therefore, no dose adjustment is necessary for patients with mild hepatic impairment. The impact of severe hepatic impairment (Child-Pugh Class C, score 10-15) on the pharmacokinetics of ivacaftor has not been studied. The magnitude of increase in exposure in these patients is unknown, but is expected to be substantially higher than that observed in patients with moderate hepatic impairment. When benefits are expected to outweigh the risks, KALYDECO should be used with caution in patients with severe hepatic impairment at a dose of one tablet or one packet of granules given once daily or less frequently [see DOSAGE AND ADMINISTRATION and Use In Specific Populations].

Patients With Renal Impairment

KALYDECO has not been studied in patients with mild, moderate, or severe renal impairment (creatinine clearance less than or equal to 30 mL/min) or in patients with end-stage renal disease. No dose adjustments are recommended for mild and moderate renal impairment patients because of minimal elimination of ivacaftor and its metabolites in urine (only 6.6% of total radioactivity was recovered in the urine in a human PK study); however, caution is recommended when administering KALYDECO to patients with severe renal impairment or end-stage renal disease.

Male And Female Patients

The effect of gender on KALYDECO pharmacokinetics was evaluated using population pharmacokinetics of data from clinical studies of KALYDECO. No dose adjustments are necessary based on gender.

Drug Interaction Studies

Drug interaction studies were performed with KALYDECO and other drugs likely to be co-administered or drugs commonly used as probes for pharmacokinetic interaction studies [see DRUG INTERACTIONS].

Dosing recommendations based on clinical studies or potential drug interactions with KALYDECO are presented below.

Potential For Ivacaftor To Affect Other Drugs

Based on in vitro results, ivacaftor and metabolite M1 have the potential to inhibit CYP3A and P-gp. Clinical studies showed that KALYDECO is a weak inhibitor of CYP3A and P-gp, but not an inhibitor of CYP2C8. In vitro studies suggest that ivacaftor and M1 may inhibit CYP2C9. In vitro, ivacaftor, M1, and M6 were not inducers of CYP isozymes. Dosing recommendations for co-administered drugs with KALYDECO are shown in Figure 2.

Figure 2: Impact of KALYDECO on Other Drugs

Note: The data obtained with substrates but without co-administration of KALYDECO are used as reference.
*NE: Norethindrone; **EE: Ethinyl Estradiol
The vertical lines are at 0.8, 1.0, and 1.25, respectively.

Potential For Other Drugs To Affect Ivacaftor

In vitro studies showed that ivacaftor and metabolite M1 were substrates of CYP3A enzymes (i.e., CYP3A4 and CYP3A5). Exposure to ivacaftor is reduced by concomitant CYP3A inducers and increased by concomitant CYP3A inhibitors [see DOSAGE AND ADMINISTRATION and DRUG INTERACTIONS]. KALYDECO dosing recommendations for co-administration with other drugs are shown in Figure 3.

Figure 3: Impact of Other Drugs on KALYDECO

Note: The data obtained for KALYDECO without co-administration of inducers or inhibitors are used as reference.
The vertical lines are at 0.8, 1.0, and 1.25, respectively.

Clinical Studies

Trials In Patients With CF Who Have A G551D Mutation In The CFTR Gene

Dose Ranging

Dose ranging for the clinical program consisted primarily of one double-blind, placebo-controlled, crossover trial in 39 adult (mean age 31 years) Caucasian patients with CF who had FEV1 ≥40% predicted. Twenty patients with median predicted FEV1 at baseline of 56% (range: 42% to 109%) received KALYDECO 25, 75, 150 mg or placebo every 12 hours for 14 days and 19 patients with median predicted FEV1 at baseline of 69% (range: 40% to 122%) received KALYDECO 150, 250 mg, or placebo every 12 hours for 28 days. The selection of the 150 mg every 12 hours dose was primarily based on nominal improvements in lung function (pre-dose FEV1) and changes in pharmacodynamic parameters (sweat chloride and nasal potential difference). The twice-daily dosing regimen was primarily based on an apparent terminal plasma half-life of approximately 12 hours.

Efficacy

The efficacy of KALYDECO in patients with CF who have a G551D mutation in the CFTR gene was evaluated in two randomized, double-blind, placebo-controlled clinical trials in 213 clinically stable patients with CF (109 receiving KALYDECO 150 mg twice daily). All eligible patients from these trials were rolled over into an open-label extension study.

Trial 1 evaluated 161 patients with CF who were 12 years of age or older (mean age 26 years) with FEV1 at screening between 40-90% predicted [mean FEV1 64% predicted at baseline (range: 32% to 98%)]. Trial 2 evaluated 52 patients who were 6 to 11 years of age (mean age 9 years) with FEV1 at screening between 40-105% predicted [mean FEV1 84% predicted at baseline (range: 44% to 134%)]. Patients who had persistent Burkholderia cenocepacia, Burkholderia dolosa, or Mycobacterium abscessus isolated from sputum at screening and those with abnormal liver function defined as 3 or more liver function tests (ALT, AST, AP, GGT, total bilirubin) ≥3 times the upper limit of normal were excluded.

Patients in both trials were randomized 1:1 to receive either 150 mg of KALYDECO or placebo every 12 hours with food containing fat for 48 weeks in addition to their prescribed CF therapies (e.g., tobramycin, dornase alfa). The use of inhaled hypertonic saline was not permitted.

The primary efficacy endpoint in both studies was improvement in lung function as determined by the mean absolute change from baseline in percent predicted pre-dose FEV1 through 24 weeks of treatment.

In both studies, treatment with KALYDECO resulted in a significant improvement in FEV1. The treatment difference between KALYDECO and placebo for the mean absolute change in percent predicted FEV1 from baseline through Week 24 was 10.6 percentage points (P<0.0001) in Trial 1 and 12.5 percentage points (P<0.0001) in Trial 2 (Figure 4). These changes persisted through 48 weeks. Improvements in percent predicted FEV1 were observed regardless of age, disease severity, sex, and geographic region.

Figure 4: Mean Absolute Change from Baseline in Percent Predicted FEV1*

*Primary endpoint was assessed at the 24-week time point.

Other efficacy variables included absolute change from baseline in sweat chloride [see CLINICAL PHARMACOLOGY], time to first pulmonary exacerbation (Trial 1 only), absolute change from baseline in weight, and improvement from baseline in Cystic Fibrosis Questionnaire Revised (CFQ-R) respiratory domain score, a measure of respiratory symptoms relevant to patients with CF such as cough, sputum production, and difficulty breathing. For the purpose of the study, a pulmonary exacerbation was defined as a change in antibiotic therapy (IV, inhaled, or oral) as a result of 4 or more of 12 pre-specified sino-pulmonary signs/symptoms. Patients treated with KALYDECO demonstrated statistically significant improvements in risk of pulmonary exacerbations, CF symptoms (in Trial 1 only), and gain in body weight (Table 4). Weight data, when expressed as body mass index normalized for age and sex in patients <20 years of age, were consistent with absolute change from baseline in weight.

Table 4: Effect of KALYDECO on Other Efficacy Endpoints in Trials 1 and 2

Endpoint Trial 1 Trial 2
Treatment differencea
(95% CI)
P value Treatment differencea
(95% CI)
P value
Mean absolute change from baseline in CFQ-R respiratory domain score (points)
Through Week 24 8.1
(4.7, 11.4)
<0.0001 6.1
(-1.4, 13.5)
0.1092
Through Week 48 8.6
(5.3, 11.9)
<0.0001 5.1
(-1.6, 11.8)
0.1354
Relative risk of pulmonary exacerbation
Through Week 24 0.40b 0.0016 NA NA
Through Week 48 0.46b 0.0012 NA NA
Mean absolute change from baseline in body weight (kg)
At Week 24 2.8
(1.8, 3.7)
<0.0001 1.9
(0.9, 2.9)
0.0004
At Week 48 2.7
(1.3, 4.1)
0.0001 2.8
(1.3, 4.2)
0.0002
Absolute change in sweat chloride (mmol/L)
Through Week 24 -48
(-51, -45)
<0.0001 -54
(-62, -47)
<0.0001
Through Week 48 -48
(-51, -45)
<0.0001 -53
(-61, -46)
<0.0001
CI: confidence interval; NA: not analyzed due to low incidence of events
aTreatment difference = effect of KALYDECO – effect of Placebo
bHazard ratio for time to first pulmonary exacerbation

Trial In Patients With A G1244E, G1349D, G178R, G551S, G970R, S1251N, S1255P, S549N, Or S549R Mutation In The CFTR Gene

The efficacy and safety of KALYDECO in patients with CF who have a G1244E, G1349D, G178R, G551S, G970R, S1251N, S1255P, S549N, or S549R mutation in the CFTR gene were evaluated in a two-part, randomized, double-blind, placebo-controlled, crossover design clinical trial in 39 patients with CF (Trial 4). Patients who completed Part 1 of this trial continued into the 16-week open-label Part 2 of the study. The mutations studied were G178R, S549N, S549R, G551S, G970R, G1244E, S1251N, S1255P, and G1349D. See Clinical Studies for efficacy in patients with a G551D mutation.

Patients were 6 years of age or older (mean age 23 years) with FEV1 ≥40% at screening [mean FEV1 at baseline 78% predicted (range: 43% to 119%)]. Patients with evidence of colonization with Burkholderia cenocepacia, Burkholderia dolosa, or Mycobacterium abscessus and those with abnormal liver function defined as 3 or more liver function tests (ALT, AST, AP, GGT, total bilirubin) ≥3 times the upper limit of normal at screening were excluded.

Patients were randomized 1:1 to receive either 150 mg of KALYDECO or placebo every 12 hours with food containing fat for 8 weeks in addition to their prescribed CF therapies during the first treatment period and crossed over to the other treatment for the second 8 weeks. The two 8-week treatment periods were separated by a 4-to 8-week washout period. The use of inhaled hypertonic saline was not permitted.

The primary efficacy endpoint was improvement in lung function as determined by the mean absolute change from baseline in percent predicted FEV1 through 8 weeks of treatment. Other efficacy variables included absolute change from baseline in sweat chloride through 8 weeks of treatment [see CLINICAL PHARMACOLOGY], absolute change from baseline in body mass index (BMI) at 8 weeks of treatment (including body weight at 8 weeks), and improvement in CFQ-R respiratory domain score through 8 weeks of treatment. For the overall population of the 9 mutations studied, treatment with KALYDECO compared to placebo resulted in significant improvement in percent predicted FEV1 [10.7 through Week 8 (P<0.0001)], BMI [0.66 kg/m² at Week 8 (P<0.0001)], and CFQ-R respiratory domain score [9.6 through Week 8 (P=0.0004)]; however, there was a high degree of variability of efficacy responses among the 9 mutations (Table 5).

Table 5: Effect of KALYDECO for Efficacy Variables in the Overall Populations and for Specific CFTR Mutations

Mutation (n) Absolute change in percent predicted FEV1 BMI (kg/m²) CFQ-R Respiratory Domain Score (Points) Absolute Change in Sweat Chloride (mmol/L)
At Week 2 At Week 4 At Week 8 At Week 8 At Week 8 At Week 8
All patients (n=39)
Results shown as mean (95% CI) change from baseline KALYDECO vs. placebo-treated patients:
  8.3 (4.5, 12.1) 10.0 (6.2, 13.8) 13.8 (9.9, 17.6) 0.66† (0.34, 0.99) 12.8 (6.7, 18.9) -50 (-58, -41)*
Patients grouped under mutation types (n)
Results shown as mean (minimum, maximum) for change from baseline for KALYDECO-treated patients**:
G1244E (5) 11 (-5, 25) 6 (-5, 13) 8 (-1, 18) 0.63 (0.34, 1.32) 3.3 (-27.8, 22.2) -55 (-75, -34)
G1349D (2) 19 (5, 33) 18 (2, 35) 20 (3, 36) 1.15 (1.07, 1.22) 16.7 (-11.1, 44.4) -80 (-82, -79)
G178R (5) 7 (1, 17) 10 (-2, 21) 8 (-1, 18) 0.85 (0.33, 1.46) 20.0 (5.6, 50.0) -53 (-65, -35)
G551S (2) 0 (-5, 5) 0.3 (-5, 6) 3†† 0.16†† 16.7†† -68††
G970R (4) 7 (1, 13) 7 (1, 14) 3 (-1, 5) 0.48 (-0.38, 1.75) 1.4 (-16.7, 16.7) -6 (-16, -2)
S1251N (8) 2 (-23, 20) 8 (-13, 26) 9 (-20, 21) 0.73 (0.08, 1.83) 23.3 (5.6, 50.0) -54 (-84, -7)
S1255P (2) 11 (8, 14) 9 (5, 13) 3 (-1, 8) 1.62 (1.39, 1.84) 8.3 (5.6, 11.1) -78 (-82, -74)
S549N (6) 11 (5, 16) 8 (-9, 19) 11 (-2, 20) 0.79 (0.00, 1.91) 8.8 (-8.3, 27.8) -74 (-93, -53)
S549R (4) 3 (-4, 8) 4 (-4, 10) 5 (-3, 13) 0.53 (0.33, 0.80) 6.9 (0.0, 11.1) -61††† (-71, -54)
* n=36 for the analysis of absolute change in sweat chloride.
** Statistical testing was not performed due to small numbers for individual mutations.
† Result for weight gain as a component of body mass index was consistent with BMI.
†† Reflects results from the one patient with the G551S mutation with data at the 8-week time point.
††† n=3 for the analysis of absolute change in sweat chloride.

Trial In Patients With CF Who Have An R117H Mutation In The CFTR Gene

The efficacy and safety of KALYDECO in patients with CF who have an R117H mutation in the CFTR gene were evaluated in a randomized, double-blind, placebo-controlled, parallel-group clinical trial (Trial 5). Fifty-nine of 69 patients completed 24 weeks of treatment. Two patients discontinued and 8 patients did not complete treatment due to study termination. Trial 5 evaluated 69 clinically stable patients with CF who were 6 years of age or older (mean age 31 years). Patients who were 12 years and older had FEV1 at screening between 40-90% predicted, and patients who were 6-11 years of age had FEV1 at screening between 40-105% predicted. The overall mean FEV1 was 73% predicted at baseline (range: 33% to 106%). The patients had well preserved BMIs (mean overall: 23.76 kg/m²) and a high proportion were pancreatic sufficient as assessed by a low rate of pancreatic enzyme replacement therapy use (pancreatin: 11.6%; pancrelipase: 5.8%). Patients who had persistent Burkholderia cenocepacia, Burkholderia dolosa, or Mycobacterium abscessus isolated from sputum at screening, and those with abnormal liver function defined as 3 or more liver function tests (ALT, AST, AP, GGT, total bilirubin) ≥3 times the ULN, were excluded.

Patients were randomized 1:1 to receive either 150 mg of KALYDECO (n=34) or placebo (n=35) every 12 hours with food containing fat for 24 weeks in addition to their prescribed CF therapies.

The primary efficacy endpoint was improvement in lung function as determined by the mean absolute change from baseline in percent predicted FEV1 through 24 weeks of treatment. The treatment difference for absolute change in percent predicted FEV1 through Week 24 was 2.1 percentage points (analysis conducted with the full analysis set which included all 69 patients), and did not reach statistical significance (Table 6).

Other efficacy variables that were analyzed included absolute change in sweat chloride from baseline through Week 24, improvement in cystic fibrosis respiratory symptoms through Week 24 as assessed by the CFQ-R respiratory domain score (Table 6), absolute change in body mass index (BMI) at Week 24, and time to first pulmonary exacerbation. The overall treatment difference for the absolute change from baseline in BMI at Week 24 was 0.3 kg/m² and the calculated hazard ratio for time to first pulmonary exacerbation was 0.93, which were not statistically significant.

Statistically significant improvements in clinical efficacy (FEV1, CFQ-R respiratory domain) were seen in several subgroup analyses, and decreases in sweat chloride were observed in all subgroups. The mean baseline sweat chloride for all patients was 70 mmol/L. Subgroups analyzed included those based on age, lung function, and poly-T status (Table 6).

Table 6: Effect of KALYDECO on Overall Population (Percent Predicted FEV1, CFQ-R Respiratory Domain Score, and Sweat Chloride) and in Relevant Subgroups Through 24 Weeks

Subgroup Parameter Study Drug % Predicted FEV1(Percentage Points) CFQ-R Respiratory Domain Score (Points) Sweat Chloride (mmol/L)
n Mean Treatment Difference (95% CI) n Mean Treatment Difference (95% CI) n Mean Treatment Difference (95% CI)
R117H-AII Patients
  Placebo 35 0.5 2.1 34 -0.8 8.4 35 -2.3 -24.0
Kalydeco 34 2.6 (-1.1, 5.4) 33 7.6 (2.2, 14.6) 32 -26.3 (-28.0, -19.9)
Subgroup by Age
6-11 Placebo 8 3.5 -6.3 7 -1.6 -6.1 8 1.0 -27.6
Kalydeco 9 -2.8 (-12.0, -0.7) 8 -7.7 (-15.7, 3.4) 8 -26.6 (-37.2, -18.1)
12-17 Placebo 1 - - 1 - - 1 - -
Kalydeco 1 - - 1 1 - -
≥18 Placebo 26 -0.5 5.0 26 -0.5 12.6 26 -4.0 -21.9
Kalydeco 24 4.5 (1.1, 8.8) 24 12.2 (5.0, 20.3) 23 -25.9 (-26.5, -17.3)
Subgroup by Poly-T Status†
5T Placebo 24 0.7 5.3 24 -0.6 15.3 24 -4.6 -24.2
Kalydeco 14 6.0 (1.3, 9.3) 14 14.7 (7.7, 23.0) 13 -28.7 (-30.2, -18.2)
7T Placebo 5 -0.9 0.2 5 -6.0 5.2 5 3.9 -24.1
Kalydeco 11 -0.7 (-8.1, 8.5) 11 -0.7 (-13.0, 23.4) 10 -20.2 (-33.9, -14.3)
Subgroup by Baseline FEV1 % Predicted
<70% Placebo 15 0.4 4.0 15 3.0 11.4 15 -3.8 -25.5
Kalydeco 13 4.5 (-2.1, 10.1) 13 14.4 (1.2, 21.6) 12 -29.3 (-31.8, -19.3)
70-90% Placebo 14 0.2 2.6 13 -3.6 8.8 14 -3.1 -20.0
Kalydeco 14 2.8 (-2.3, 7.5) 14 5.2 (-2.6, 20.2) 14 -23.0 (-26.9, -12.9)
>90% Placebo 6 2.2 -4.3 6 -2.5 -0.7 6 1.0 -26.8
Kalydeco 7 -2.1 (-9.9, 1.3) 6 -3.2 (-10.4, 9.0) 6 -25.9 (-39.5, -14.1)
* MMRM analysis with fixed effects for treatment, age, week, baseline value, treatment by week, and subject as a random effect
† (n=54) Poly-T status confirmed by genotyping

Trial In Patients With CF Heterozygous For The F508del Mutation And A Second Mutation Predicted To Be Responsive To Ivacaftor

The efficacy and safety of KALYDECO and an ivacaftor-containing combination product in 246 patients with CF was evaluated in a randomized, double-blind, placebo-controlled, 2-period, 3-treatment, 8-week crossover design clinical trial (Trial 7). Mutations predicted to be responsive to ivacaftor were selected for the study based on the clinical phenotype (pancreatic sufficiency), biomarker data (sweat chloride), and in vitro responsiveness to ivacaftor.

Eligible patients were heterozygous for the F508del mutation with a second mutation predicted to be responsive to ivacaftor. Of the 244 patients included in the efficacy analysis, who were randomized and dosed, 146 patients had a splice mutation and 98 patients had a missense mutation, as the second allele. 156 patients received KALYDECO and 161 patients received placebo. Patients were aged 12 years and older (mean age 35 years [range 12-72]) and had a percent predicted FEV1 at screening between 40-90 [mean ppFEV1 at study baseline 62 (range: 35 to 94)]. Patients with evidence of colonization with organisms associated with a more rapid decline in pulmonary status (e.g. Burkholderia cenocepacia, Burkholderia dolosa, or Mycobacterium abscessus) and those with abnormal liver function at screening were excluded. Abnormal liver function was defined as 2 or more liver function tests (ALT, AST, ALP, GGT) ≥3 times the upper limit of normal or total bilirubin ≥2 times the upper limit of normal, or a single increase in ALT/AST ≥5 times the upper limit of normal.

The primary efficacy endpoint was the mean absolute change from study baseline in percent predicted FEV1 averaged at Weeks 4 and 8 of treatment. The key secondary efficacy endpoint was absolute change in CFQ-R respiratory domain score from study baseline averaged at Weeks 4 and 8 of treatment. For the overall population, treatment with KALYDECO compared to placebo resulted in significant improvement in ppFEV1 [4.7 percent points from study baseline to average of Week 4 and Week 8 (P<0.0001)] and CFQ-R respiratory domain score [9.7 points from study baseline to average of Week 4 and Week 8 (P<0.0001)]. Statistically significant improvements compared to placebo were also observed in the subgroup of patients with splice mutations and missense mutations (Table 7).

Table 7: Effect of KALYDECO for Efficacy Variables

Mutation (n) Absolute Change in percent predicted FEV1*† Absolute Change in CFQ-R Respiratory Domain Score (Points)*§ Absolute Change in Sweat Chloride (mmol/L)*§
Splice mutations (n=94 for IVA and n=97 for PBO)
Results shown as difference in mean (95% CI) change from study baseline for KALYDECO vs. placebo-treated patients:
  5.4 (4.1, 6.8) 8.5 (5.3, 11.7) -2.4 (-5.0, 0.3)
By individual splice mutation (n). Results shown as mean (minimum, maximum) for change from study baseline for KALYDECO-treated patients
2789+5G→A (28) 5.1 (-7.1, 17.0) 8.6 (-5.6, 27.8) 0.4 (-7.5, 8.8)
3272-26A→G (23) 3.5 (-9.1, 16.0) 8.0 (-11.1, 27.8) -2.3 (-25.0, 11.8)
3849+10kBc→T (40) 5.1 (-6.8, 16.2) 7.5 (-30.6, 55.6) -4.6 (-80.5, 23.0)
711+3A→G (2) 9.2 (8.9, 9.6) -8.3 (-13.9, -2.8) -9.9 (-13.5, -6.3)
E831X (1) 7.1 (7.1, 7.1) 0.0 (0.0, 0.0) -7.8 (-7.8, -7.8)
Missense mutations (n=62 for IVA and n=63 for PBO)
Results shown as difference in mean (95% CI) change from study baseline for KALYDECO vs. placebo-treated patients:
  3.6 (1.9, 5.2) 11.5 (7.5, 15.4) -7.8 (-11.2, -4.5)
By individual missense mutation (n). Results shown as mean (minimum, maximum) for change from study baseline for KALYDECO-treated patients
D579G (2) 13.3 (12.4, 14.1) 15.3 (-2.8, 33.3) -30.8 (-36.0, -25.5)
D1152H (15) 2.4 (-5.0, 10.2) 13.7 (-16.7, 50.0) -4.8 (-22.0, 3.0)
A455E (14) 3.7 (-6.6, 19.7) 6.8 (-13.9, 33.3) 7.5 (-16.8, 16.0)
L206W (2) 4.2 (2.5, 5.9) 12.5 (-5.6, 30.6) 3.9 (-8.3, 16.0)
P67L (12) 4.3 (-2.5, 25.7) 10.8 (-12.5, 36.1) -10.5 (-34.8, 9.8)
R1070W (1) 2.9 (2.9, 2.9) 44.4 (44.4, 44.4) 0.3 (0.3, 0.3)
R117C (1) 3.5 (3.5, 3.5) 22.2 (22.2, 22.2) -36.0 (-36.0, -36.0)
R347H (3) 2.5 (-0.6, 6.9) 6.5 (5.6, 8.3) -19.2 (-25.8, -7.0)
R352Q (2) 4.4 (3.5, 5.3) 9.7 (8.3, 11.1) -21.9 (-45.5, 1.8)
S945L (9) 8.8 (-0.2, 20.5) 10.6 (-25.0, 27.8) -30.8 (-50.8, -17.3)
S977F (1) 4.3 (4.3, 4.3) -2.8 (-2.8, -2.8) -19.5 (-19.5, -19.5)
*Average of Week 4 and 8 values
†Absolute change in ppFEV1 by individual mutations is an ad hoc analysis.
§Absolute change in CFQ-R respiratory domain score and absolute change in sweat chloride by mutation subgroups and by individual mutations are ad hoc analyses.

In an analysis of BMI at Week 8, an exploratory end-point, patients treated with KALYDECO had a mean improvement of 0.28 kg/m² [95% CI (0.14, 0.43)], 0.24 kg/m² [95% CI (0.06, 0.43)], and 0.35 kg/m² [95% CI (0.12, 0.58)] versus placebo for the overall, splice, and missense mutation populations of patients, respectively.

Trial In Patients Homozygous For The F508del Mutation In The CFTR Gene

Trial 3 was a 16-week, randomized, double-blind, placebo-controlled, parallel-group trial in 140 patients with CF age 12 years and older who were homozygous for the F508del mutation in the CFTR gene and who had FEV1 ≥40% predicted. Patients were randomized 4:1 to receive KALYDECO 150 mg (n=112) every 12 hours or placebo (n=28) in addition to their prescribed CF therapies. The mean age of patients enrolled was 23 years and the mean baseline FEV1 was 79% predicted (range 40% to 129%). As in Trials 1 and 2, patients who had persistent Burkholderia cenocepacia, Burkholderia dolosa, or Mycobacterium abscessus isolated from sputum at screening and those with abnormal liver function defined as 3 or more liver function tests (ALT, AST, AP, GGT, total bilirubin) ≥3 times the upper limit of normal were excluded. The use of inhaled hypertonic saline was not permitted.

The primary endpoint was improvement in lung function as determined by the mean absolute change from baseline through Week 16 in percent predicted FEV1. The treatment difference from placebo for the mean absolute change in percent predicted FEV1 through Week 16 in patients with CF homozygous for the F508del mutation in the CFTR gene was 1.72 percentage points (1.5% and -0.2% for patients in the KALYDECO and placebo-treated groups, respectively) and did not reach statistical significance (Table 8).

Other efficacy variables that were analyzed included absolute change in sweat chloride from baseline through Week 16, change in cystic fibrosis respiratory symptoms through Week 16 as assessed by the CFQ-R respiratory domain score (Table 8), change in weight through Week 16, and rate of pulmonary exacerbation. The overall treatment difference for change from baseline in weight through Week 16 was -0.16 kg (95% CI -1.06, 0.74); the rate ratio for pulmonary exacerbation was 0.677 (95% CI 0.33, 1.37).

Table 8: Effect of KALYDECO on Overall Population (Percent Predicted FEV1, CFQ-R Respiratory Domain Score, and Sweat Chloride) Through 16 Weeks

Subgroup Parameter Study Drug Absolute Change through Week 16 - Full Analysis Set
% Predicted FEV1(Percentage Points) CFQ-R Respiratory Domain Score (Points) Sweat Chloride (mmol/L)
n Mean Treatment Difference (95% CI) n Mean Treatment Difference (95% CI) n Mean Treatment Difference (95% CI)
F508del homozygous
  Placebo 28 -0.2 1.72 28 -1.44 1.3 28 0.13 -2.9
  Kalydeco 111 1.5 (-0.6, 4.1) 111 -0.12 (-2.9, 5.6) 109 -2.74 (-5.6, -0.2)
* MMRM analysis with fixed effects for treatment, age week, baseline value, treatment by week, and subject as a random effect

Side Effects of Ivacaftor

Ivacaftor can cause serious side effects. See "Drug Precautions".

The most common side effects include:

  • headache
  • upper respiratory tract infection (common cold), including:
    • sore throat
    • nasal or sinus congestion
    • runny nose
  • stomach (abdominal) pain
  • diarrhea
  • rash
  • nausea
  • dizziness

Tell your doctor if you have any side effect that bothers you or that does not go away.

These are not all the possible side effects of ivacaftor. For more information, ask your doctor or pharmacist.

Call your doctor for medical advice about side effects. You may report side effects to the FDA at 1-800-FDA-1088.

 

Ivacaftor Dosage

Take ivacaftor exactly as prescribed. Follow the directions on your prescription label carefully.

The recommended dose of ivacaftor for both adults and pediatric patients age 6 years and older is one 150 mg tablet taken orally every 12 hours (300 mg total daily dose) with fat-containing food. 

The recommended dose of ivacaftor in children 2 to less than 6 years of age and less than 14 kg is one 50 mg packet mixed with 1 teaspoon (5 mL) of soft food or liquid and administered orally every 12 hours with fat-containing food.

The recommended dose of ivacaftor in children 2 to less than 6 years of age and 14 kg or greater is one 75 mg packet mixed with 1 teaspoon (5 mL) of soft food or liquid and administered orally every 12 hours with fat-containing food. 

Your doctor may reduce your dose if you have liver disease or are taking certain medications. 

Ivacaftor Overdose

If you take too much ivacaftor, call your local Poison Control Center or seek emergency medical attention right away.

Other Requirements

  • Store ivacaftor at room temperature between 68°F to 77°F (20°C to 25°C).
  • Do not use ivacaftor after the expiration date on the package.

Keep ivacaftor and all medicines out of the reach of children.

 

What should i discuss with my healthcare provider before taking ivacaftor (kalydeco)?

You should not use ivacaftor if you are allergic to it.

To make sure you can safely use ivacaftor, tell your doctor if you have any of these other conditions:

  • liver disease; or
  • kidney disease.

FDA pregnancy category B. Ivacaftor is not expected to harm an unborn baby. Tell your doctor if you are pregnant or plan to become pregnant during treatment.

It is not known whether ivacaftor passes into breast milk or if it could harm a nursing baby. Do not use this medication without telling your doctor if you are breast-feeding a baby.

Where can i get more information?

Your pharmacist can provide more information about ivacaftor.

Remember, keep this and all other medicines out of the reach of children, never share your medicines with others, and use this medication only for the indication prescribed.

Every effort has been made to ensure that the information provided by Cerner Multum, Inc. ('Multum') is accurate, up-to-date, and complete, but no guarantee is made to that effect. Drug information contained herein may be time sensitive. Multum information has been compiled for use by healthcare practitioners and consumers in the United States and therefore Multum does not warrant that uses outside of the United States are appropriate, unless specifically indicated otherwise. Multum's drug information does not endorse drugs, diagnose patients or recommend therapy. Multum's drug information is an informational resource designed to assist licensed healthcare practitioners in caring for their patients and/or to serve consumers viewing this service as a supplement to, and not a substitute for, the expertise, skill, knowledge and judgment of healthcare practitioners. The absence of a warning for a given drug or drug combination in no way should be construed to indicate that the drug or drug combination is safe, effective or appropriate for any given patient. Multum does not assume any responsibility for any aspect of healthcare administered with the aid of information Multum provides. The information contained herein is not intended to cover all possible uses, directions, precautions, warnings, drug interactions, allergic reactions, or adverse effects. If you have questions about the drugs you are taking, check with your doctor, nurse or pharmacist.

Copyright 1996-2013 Cerner Multum, Inc. Version: 1.01. Revision date: 5/14/2012.

Your use of the content provided in this service indicates that you have read,understood and agree to the End-User License Agreement,which can be accessed by clicking on this link.

What happens if I overdose?

Seek emergency medical attention or call the Poison Help line at 1-800-222-1222.

Introduction

A cystic fibrosis transmembrane conductance regulator (CFTR) potentiator.1 2 4 5 8 10 11 12

Cautions for Ivacaftor

Contraindications

  • Manufacturer states none known.1

Warnings/Precautions

Hepatic Effects

Elevated ALT or AST concentrations reported.1

Assess serum ALT and AST concentrations prior to initiation of therapy, every 3 months during first year of therapy, and annually thereafter.1

Closely monitor patients who develop increased ALT or AST concentrations until abnormalities resolve.1

Interrupt therapy in patients with ALT or AST elevations >5 times ULN.1 Following resolution, consider benefits and risks of resuming therapy.1

Interactions with CYP3A Inducers

Concomitant use with potent CYP3A inducers (e.g., carbamazepine, phenobarbital, phenytoin, rifabutin, rifampin, St. John’s wort [Hypericum perforatum]) substantially decreases systemic exposure of ivacaftor possibly reducing efficacy of the drug.1 Concomitant use not recommended.1 (See Specific Drugs and Foods under Interactions.)

Specific Populations

Pregnancy

Category B.1

Lactation

Distributed into milk in rats; likely distributed into human milk.1 Use with caution in nursing women.1

Pediatric Use

Safety and efficacy established in pediatric patients 6–17 years of age with cystic fibrosis and a G551D mutation in the CFTR gene.1

Safety and efficacy not established in pediatric patients <6 years of age.1

Geriatric Use

Insufficient experience in patients ≥65 years of age to determine whether geriatric patients respond differently than younger adults; cystic fibrosis is generally a disease of children and young adults.1

Hepatic Impairment

Effect of mild hepatic impairment (Child-Pugh class A) on pharmacokinetics not studied but minimal effects expected; dosage adjustment not necessary.1 (See Special Populations under Pharmacokinetics.)

Increased AUC in patients with moderate hepatic impairment (Child-Pugh class B, score 7–9); dosage reduction recommended.1 (See Hepatic Impairment under Dosage and Administration and Special Populations under Pharmacokinetics.)

Effect of severe hepatic impairment (Child-Pugh class C, score 10–15) on pharmacokinetics not studied but increased AUC expected; use with caution and at reduced dosage after weighing risks and benefits of therapy.1

Renal Impairment

Not studied in patients with mild, moderate, or severe renal impairment or in those with ESRD.1

Dosage adjustment not necessary in patients with mild or moderate renal impairment because of minimal urinary excretion of drug and metabolites.1 (See Elimination Route under Pharmacokinetics.)

Use with caution in patients with severe renal impairment (Clcr ≤30 mL/minute) or in those with ESRD.1

Common Adverse Effects

Headache, oropharyngeal pain, upper respiratory tract infection, nasal congestion, abdominal pain, nasopharyngitis, diarrhea, rash, nausea, dizziness.1

Before Using ivacaftor

In deciding to use a medicine, the risks of taking the medicine must be weighed against the good it will do. This is a decision you and your doctor will make. For ivacaftor, the following should be considered:

Allergies

Tell your doctor if you have ever had any unusual or allergic reaction to ivacaftor or any other medicines. Also tell your health care professional if you have any other types of allergies, such as to foods, dyes, preservatives, or animals. For non-prescription products, read the label or package ingredients carefully.

Pediatric

Appropriate studies performed to date have not demonstrated pediatric-specific problems that would limit the usefulness of ivacaftor in children. However, safety and efficacy have not been established in children younger than 2 years of age.

Geriatric

Although appropriate studies on the relationship of age to the effects of ivacaftor have not been performed in the geriatric population, no geriatric-specific problems have been documented to date.

Pregnancy

Pregnancy Category Explanation
All Trimesters B Animal studies have revealed no evidence of harm to the fetus, however, there are no adequate studies in pregnant women OR animal studies have shown an adverse effect, but adequate studies in pregnant women have failed to demonstrate a risk to the fetus.

Breast Feeding

There are no adequate studies in women for determining infant risk when using this medication during breastfeeding. Weigh the potential benefits against the potential risks before taking this medication while breastfeeding.

Interactions with Medicines

Although certain medicines should not be used together at all, in other cases two different medicines may be used together even if an interaction might occur. In these cases, your doctor may want to change the dose, or other precautions may be necessary. When you are taking ivacaftor, it is especially important that your healthcare professional know if you are taking any of the medicines listed below. The following interactions have been selected on the basis of their potential significance and are not necessarily all-inclusive.

Using ivacaftor with any of the following medicines is usually not recommended, but may be required in some cases. If both medicines are prescribed together, your doctor may change the dose or how often you use one or both of the medicines.

  • Aprepitant
  • Atazanavir
  • Boceprevir
  • Carbamazepine
  • Clarithromycin
  • Cobicistat
  • Conivaptan
  • Crizotinib
  • Diltiazem
  • Dronedarone
  • Eliglustat
  • Erythromycin
  • Fluconazole
  • Fosaprepitant
  • Fosphenytoin
  • Idelalisib
  • Imatinib
  • Indinavir
  • Itraconazole
  • Ketoconazole
  • Lopinavir
  • Morphine
  • Morphine Sulfate Liposome
  • Nefazodone
  • Nelfinavir
  • Phenytoin
  • Posaconazole
  • Rifampin
  • Ritonavir
  • Saquinavir
  • St John's Wort
  • Telaprevir
  • Telithromycin
  • Topotecan
  • Verapamil
  • Voriconazole

Interactions with Food/Tobacco/Alcohol

Certain medicines should not be used at or around the time of eating food or eating certain types of food since interactions may occur. Using alcohol or tobacco with certain medicines may also cause interactions to occur. The following interactions have been selected on the basis of their potential significance and are not necessarily all-inclusive.

Other Medical Problems

The presence of other medical problems may affect the use of ivacaftor. Make sure you tell your doctor if you have any other medical problems, especially:

  • Cataracts—Use with caution. May make this condition worse.
  • Kidney disease, severe or
  • Liver disease, moderate or severe—Use with caution. The effects may be increased because of slower removal of the medicine from the body.

Brand Names U.S.

  • Kalydeco

Pharmacology

Potentiates epithelial cell chloride ion transport of defective (G551D mutant) cell-surface CFTR protein thereby improving the regulation of salt and water absorption and secretion in various tissues (eg, lung, GI tract).

Absorption

Variable; increased (by 2.5- to 4-fold) with fatty foods

Distribution

Vd: 353 L ± 122 L

Metabolism

Hepatic; extensive via CYP3A; forms 2 major metabolites (M1 [active; 1/6 potency] and M6 [inactive])

Excretion

Feces (87.8%, ~65% of administered dose as metabolites); urine (minimal, as unchanged drug)

Dosing Pediatric

Cystic fibrosis: Oral:

Granules:

Children 2 to <6 years of age:

<14 kg: 50 mg packet every 12 hours

≥14 kg: 75 mg packet every 12 hours

Tablet: Children ≥6 years of age and Adolescents: Refer to adult dosing.

Dosage adjustment for ivacaftor with concomitant medications:

CYP3A strong inhibitors (eg, clarithromycin, itraconazole, ketoconazole, posaconazole, telithromycin, voriconazole):

Children 2 to <6 years of age:

<14 kg: 50 mg granule packet twice weekly

≥14 kg: 75 mg granule packet twice weekly

Children ≥6 years of age and Adolescents: Refer to adult dosing.

CYP3A moderate inhibitors (eg, erythromycin, fluconazole):

Children 2 to <6 years of age:

<14 kg: 50 mg granule packet once daily

≥14 kg: 75 mg granule packet once daily

Children ≥6 years of age and Adolescents: Refer to adult dosing.

CYP3A strong inducers (eg, carbamazepine, phenobarbital, phenytoin, rifabutin, rifampin, St. John's wort): Refer to adult dosing.

Missed dose: Refer to adult dosing.

Dietary Considerations

Avoid grapefruit or Seville oranges.

Drug Interactions

Afatinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Afatinib. Management: Per US labeling: reduce afatinib by 10mg if not tolerated. Per Canadian labeling: avoid combination if possible; if used, administer the P-gp inhibitor simultaneously with or after the dose of afatinib. Consider therapy modification

Amodiaquine: CYP2C8 Inhibitors may increase the serum concentration of Amodiaquine. Avoid combination

ARIPiprazole: CYP3A4 Inhibitors (Weak) may increase the serum concentration of ARIPiprazole. Management: Monitor for increased aripiprazole pharmacologic effects. Aripiprazole dose adjustments may or may not be required based on concomitant therapy and/or indication. Consult full interaction monograph for specific recommendations. Monitor therapy

Betrixaban: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Betrixaban. Management: Decrease the betrixaban dose to an initial single dose of 80 mg followed by 40 mg once daily if combined with a P-glycoprotein inhibitor. Consider therapy modification

Bilastine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Bilastine. Management: Consider alternatives when possible; bilastine should be avoided in patients with moderate to severe renal insufficiency who are receiving p-glycoprotein inhibitors. Consider therapy modification

Bitter Orange: May increase the serum concentration of Ivacaftor. Avoid combination

Bosentan: May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

Brentuximab Vedotin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Brentuximab Vedotin. Specifically, concentrations of the active monomethyl auristatin E (MMAE) component may be increased. Monitor therapy

Celiprolol: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Celiprolol. Monitor therapy

Colchicine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Colchicine. Colchicine distribution into certain tissues (e.g., brain) may also be increased. Management: Colchicine is contraindicated in patients with impaired renal or hepatic function who are also receiving a p-glycoprotein inhibitor. In those with normal renal and hepatic function, reduce colchicine dose as directed. Consider therapy modification

Conivaptan: May increase the serum concentration of CYP3A4 Substrates. Avoid combination

CYP3A4 Inducers (Moderate): May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

CYP3A4 Inducers (Strong): May decrease the serum concentration of Ivacaftor. Avoid combination

CYP3A4 Inhibitors (Moderate): May increase the serum concentration of Ivacaftor. Management: Ivacaftor dose reductions are required; consult full monograph content for specific age- and weight-based recommendations. No dose adjustment is needed when using ivacaftor/lumacaftor with a moderate CYP3A4 inhibitor. Consider therapy modification

CYP3A4 Inhibitors (Strong): May increase the serum concentration of Ivacaftor. Management: Ivacaftor dose reductions are required; consult full monograph content for specific age- and weight-based recommendations. Consider therapy modification

Dabigatran Etexilate: P-glycoprotein/ABCB1 Inhibitors may increase serum concentrations of the active metabolite(s) of Dabigatran Etexilate. Management: Dabigatran dose reductions may be needed. Specific recommendations vary considerably according to US vs Canadian labeling, specific P-gp inhibitor, renal function, and indication for dabigatran treatment. Refer to full monograph or dabigatran labeling. Consider therapy modification

Dabrafenib: May decrease the serum concentration of CYP3A4 Substrates. Management: Seek alternatives to the CYP3A4 substrate when possible. If concomitant therapy cannot be avoided, monitor clinical effects of the substrate closely (particularly therapeutic effects). Consider therapy modification

Dasatinib: May increase the serum concentration of CYP3A4 Substrates. Monitor therapy

Deferasirox: May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

Dofetilide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Dofetilide. Monitor therapy

DOXOrubicin (Conventional): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of DOXOrubicin (Conventional). Management: Seek alternatives to P-glycoprotein inhibitors in patients treated with doxorubicin whenever possible. One U.S. manufacturer (Pfizer Inc.) recommends that these combinations be avoided. Consider therapy modification

Edoxaban: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Edoxaban. Management: See full monograph for details. Reduced doses are recommended for patients receiving edoxaban for venous thromboembolism in combination with certain inhibitors. Similar dose adjustment is not recommended for edoxaban use in atrial fibrillation. Consider therapy modification

Everolimus: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Everolimus. Management: Everolimus dose reductions are required for patients being treated for subependymal giant cell astrocytoma or renal cell carcinoma. See prescribing information for specific dose adjustment and monitoring recommendations. Consider therapy modification

Flibanserin: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Flibanserin. Monitor therapy

Fosaprepitant: May increase the serum concentration of CYP3A4 Substrates. Monitor therapy

Fusidic Acid (Systemic): May increase the serum concentration of CYP3A4 Substrates. Avoid combination

Grapefruit Juice: May increase the serum concentration of Ivacaftor. Avoid combination

HYDROcodone: CYP3A4 Inhibitors (Weak) may increase the serum concentration of HYDROcodone. Monitor therapy

Idelalisib: May increase the serum concentration of CYP3A4 Substrates. Avoid combination

Lomitapide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Lomitapide. Management: Patients on lomitapide 5 mg/day may continue that dose. Patients taking lomitapide 10 mg/day or more should decrease the lomitapide dose by half. The lomitapide dose may then be titrated up to a max adult dose of 30 mg/day. Consider therapy modification

MiFEPRIStone: May increase the serum concentration of CYP3A4 Substrates. Management: Minimize doses of CYP3A4 substrates, and monitor for increased concentrations/toxicity, during and 2 weeks following treatment with mifepristone. Avoid cyclosporine, dihydroergotamine, ergotamine, fentanyl, pimozide, quinidine, sirolimus, and tacrolimus. Consider therapy modification

Naldemedine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Naldemedine. Monitor therapy

Naloxegol: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Naloxegol. Monitor therapy

NiMODipine: CYP3A4 Inhibitors (Weak) may increase the serum concentration of NiMODipine. Monitor therapy

Palbociclib: May increase the serum concentration of CYP3A4 Substrates. Monitor therapy

PAZOPanib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of PAZOPanib. Avoid combination

P-glycoprotein/ABCB1 Substrates: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of P-glycoprotein/ABCB1 Substrates. P-glycoprotein inhibitors may also enhance the distribution of p-glycoprotein substrates to specific cells/tissues/organs where p-glycoprotein is present in large amounts (e.g., brain, T-lymphocytes, testes, etc.). Monitor therapy

Pimozide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Pimozide. Avoid combination

Prucalopride: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Prucalopride. Monitor therapy

Ranolazine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Ranolazine. Monitor therapy

RifAXIMin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of RifAXIMin. Monitor therapy

Sarilumab: May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

Silodosin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Silodosin. Avoid combination

Siltuximab: May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

Simeprevir: May increase the serum concentration of CYP3A4 Substrates. Monitor therapy

St John's Wort: May decrease the serum concentration of Ivacaftor. Avoid combination

Stiripentol: May increase the serum concentration of CYP3A4 Substrates. Management: Use of stiripentol with CYP3A4 substrates that are considered to have a narrow therapeutic index should be avoided due to the increased risk for adverse effects and toxicity. Any CYP3A4 substrate used with stiripentol requires closer monitoring. Consider therapy modification

Tocilizumab: May decrease the serum concentration of CYP3A4 Substrates. Monitor therapy

Topotecan: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Topotecan. Avoid combination

Venetoclax: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Venetoclax. Management: Reduce the venetoclax dose by at least 50% in patients requiring these combinations. Consider therapy modification

VinCRIStine (Liposomal): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of VinCRIStine (Liposomal). Avoid combination

Warnings/Precautions

Concerns related to adverse effects:

• Cataracts: Noncongenital lens opacities and cataracts have been reported in pediatric patients treated with ivacaftor; other risk factors were present in some cases (eg, corticosteroid use, exposure to radiation), but a possible risk related to ivacaftor cannot be excluded. Baseline and follow-up ophthalmological examinations are recommended in pediatric patients.

• CNS effects: May cause dizziness, which may impair physical or mental abilities; patients must be cautioned about performing tasks that require mental alertness (eg, operating machinery or driving).

• Hepatic effects: May increase hepatic transaminases. Monitor liver function; increased monitoring may be necessary in patients with a history of elevated hepatic transaminases. Temporarily discontinue treatment if ALT or AST >5 times ULN. Following resolution of transaminase elevations, consider the benefits and risks of resuming therapy.

Disease-related concerns:

• Hepatic impairment: Use with caution in patients with hepatic impairment; dosage adjustment recommended in patients with moderate to severe (Child-Pugh class B or C) impairment.

• Renal impairment: Use with caution in patients with severe renal impairment (CrCl ≤30 mL/minute) or ESRD.

Concurrent drug therapy issues:

• Drug-drug interactions: Potentially significant interactions may exist, requiring dose or frequency adjustment, additional monitoring, and/or selection of alternative therapy. Consult drug interactions database for more detailed information.

Usual Adult Dose for Cystic Fibrosis

150 mg orally every 12 hours

Comments:
-This drug should be taken with fat-containing food (e.g., eggs, butter, peanut butter, cheese pizza, whole milk, cheese, yogurt).
-If the patient's genotype is unknown, an approved CF mutation test should be used to detect the presence of a CFTR mutation followed by verification with bi-directional sequencing when recommended by the mutation test instructions for use.
-The tablet formulation is recommended for patients 6 years and older.

Uses:
-For the treatment of cystic fibrosis (CF) in patients age 2 years and older who have one mutation in the CFTR gene that is responsive to ivacaftor potentiation based on clinical and/or in vitro assay

(web3)