This site is intended for healthcare professionals
EMC Drug Feed
Drug information

Kalydeco

POM
Read time: 37 mins
Last updated: 26 Jun 2020

Summary of product characteristics


1. Name of the medicinal product

Kalydeco 150 mg film-coated tablets


2. Qualitative and quantitative composition

Each film-coated tablet contains 150 mg of ivacaftor.

Excipient with known effect

Each film-coated tablet contains 167.2 mg of lactose (as monohydrate)

For the full list of excipients, see section 6.1.


3. Pharmaceutical form

Film-coated tablet (tablet)

Light blue, capsule-shaped film-coated tablets, printed with “V 150” in black ink on one side and plain on the other (16.5 mm x 8.4 mm in modified tablet shape).


4.1. Therapeutic indications

Kalydeco tablets are indicated for the treatment of adults, adolescents, and children aged 6 years and older and weighing 25 kg or more with cystic fibrosis (CF) who have an R117H CFTR mutation or one of the following gating (class III) mutations in the CFTR gene: G551D, G1244E, G1349D, G178R, G551S, S1251N, S1255P, S549N or S549R (see sections 4.4 and 5.1).

Kalydeco tablets are also indicated in a combination regimen with tezacaftor 100 mg/ivacaftor 150 mg tablets for the treatment of adults and adolescents aged 12 years and older with cystic fibrosis (CF) who are homozygous for the F508del mutation or who are heterozygous for the F508del mutation and have one of the following mutations in the CFTR gene: P67L, R117C, L206W, R352Q, A455E, D579G, 711+3A→G, S945L, S977F, R1070W, D1152H, 2789+5G→A, 3272-26A→G, and 3849+10kbC→T.


4.2. Posology and method of administration

Kalydeco should only be prescribed by physicians with experience in the treatment of cystic fibrosis. If the patient's genotype is unknown, an accurate and validated genotyping method should be performed before starting treatment to confirm the presence of an indicated mutation in the CFTR gene (see section 4.1). The phase of the poly-T variant identified with the R117H mutation should be determined in accordance with local clinical recommendations.

Posology

Kalydeco monotherapy in adults, adolescents, and children aged 6 years and older and weighing 25 kg or more

The recommended dose is one Kalydeco 150 mg tablet taken orally every 12 hours (300 mg total daily dose) with fat-containing food (see Method of administration).

Kalydeco in a combination regimen with tezacaftor/ivacaftor in adults and adolescents aged 12 years and older

The recommended dose is one tezacaftor 100 mg/ivacaftor 150 mg tablet taken in the morning and one Kalydeco 150 mg tablet taken in the evening, approximately 12 hours apart with fat-containing food (see Method of administration).

Missed dose

If a dose is missed within 6 hours of the time it is usually taken, the patient should be told to take it as soon as possible and then take the next dose at the regularly scheduled time. If more than 6 hours have passed since the time the dose is usually taken, the patient should be told to wait until the next scheduled dose.

Patients receiving Kalydeco in combination with tezacaftor/ivacaftor should be told not to take more than one dose of either tablet at the same time.

Concomitant use of CYP3A inhibitors

Kalydeco monotherapy

When co-administered with strong inhibitors of CYP3A, the Kalydeco dose should be reduced to 150 mg twice a week (see sections 4.4 and 4.5).

When co-administered with moderate inhibitors of CYP3A, the Kalydeco dose should be reduced to 150 mg once daily (see sections 4.4 and 4.5).

Kalydeco in a combination regimen with tezacaftor/ivacaftor

When co-administered with strong inhibitors of CYP3A, the Kalydeco evening dose should not be administered (see sections 4.4 and 4.5). One tablet of tezacaftor 100 mg/ivacaftor 150 mg should be taken twice a week, taken approximately 3 to 4 days apart.

When co-administered with moderate inhibitors of CYP3A, the dose of Kalydeco and tezacaftor/ivacaftor should be adjusted according to the recommendations in Table 1 (see section 4.4 and 4.5).

Table 1: Dosing recommendations for concomitant use with moderate CYP3A inhibitors

Day 1

Day 2

Day 3

Day 4*

Morning Dose

Tezacaftor 100 mg/ ivacaftor 150 mg tablet

-

-

Ivacaftor 150 mg tablet

-

-

Evening Dose

Ivacaftor 150 mg tablet

-

-

-

-

* Continue dosing with tezacaftor 100 mg/ivacaftor 150 mg or ivacaftor 150 mg tablets on alternate days.

Special populations

Elderly

Very limited data are available for elderly patients treated with ivacaftor (administered as monotherapy or in a combination regimen with tezacaftor/ivacaftor). No dose adjustment is considered necessary unless moderate hepatic impairment exists. Caution is recommended for patients with severe renal impairment or end-stage renal disease (see section 5.2).

Renal impairment

No dose adjustment is necessary for patients with mild to moderate renal impairment. Caution is recommended in patients with severe renal impairment (creatinine clearance less than or equal to 30 mL/min) or end-stage renal disease (see sections 4.4 and 5.2).

Hepatic impairment

No dose adjustment is necessary for patients with mild hepatic impairment (Child-Pugh Class A). For patients with moderate hepatic impairment (Child-Pugh Class B), the dose should be reduced (see Table 2). There is no experience of the use of Kalydeco monotherapy or in combination with tezacaftor/ivacaftor in patients with severe hepatic impairment. Therefore, its use is not recommended unless the benefits outweigh the risks (see Table 2 and sections 4.4 and 5.2).

Table 2: Dosing recommendations for patients with hepatic impairment for Kalydeco monotherapy and in a combination regimen with tezacaftor/ivacaftor

Kalydeco monotherapy

Kalydeco in a combination regimen with tezacaftor/ivacaftor

Mild

(Child-Pugh Class A)

No dose adjustment

No dose adjustment

Moderate

(Child-Pugh Class B)

One tablet of ivacaftor 150 mg once daily

One tablet of tezacaftor 100 mg/ivacaftor 150 mg once daily

Severe

(Child-Pugh Class C)

Starting dose: One tablet of ivacaftor 150 mg every other day.

Dosing intervals should be modified according to clinical response and tolerability.

Starting dose: One tablet of tezacaftor 100 mg/ivacaftor 150 mg once daily.

Dosing intervals should be modified according to clinical response and tolerability.

Paediatric population

The safety and efficacy of Kalydeco monotherapy in children aged less than 6 months have not been established. No data are available.

An appropriate dose for children under 6 years of age and weighing less than 25 kg cannot be achieved with Kalydeco tablets.

Limited data are available in patients less than 6 years of age with an R117H mutation in the CFTR gene. Available data in patients aged 6 years and older are described in sections 4.8, 5.1 and 5.2.

The safety and efficacy of Kalydeco in a combination regimen with tezacaftor/ivacaftor in children aged less than 12 years have not been established. No data are available.

Method of administration

For oral use.

Patients should be instructed to swallow the tablets whole. The tablets should not be chewed, crushed, or broken before swallowing.

Kalydeco tablets should be taken with fat-containing food.

Food or drink containing grapefruit or Seville oranges should be avoided during treatment (see section 4.5).


4.3. Contraindications

Hypersensitivity to the active substance or to any of the excipients listed in section 6.1.


4.4. Special warnings and precautions for use

Only patients with CF who had a G551D, G1244E, G1349D, G178R, G551S, S1251N, S1255P, S549N, S549R gating (class III), G970R or R117H mutation in at least one allele of the CFTR gene were included in studies 1, 2, 5 and 6 (see section 5.1).

In study 5, four patients with the G970R mutation were included. In three of four patients the change in the sweat chloride test was < 5 mmol/L and this group did not demonstrate a clinically relevant improvement in FEV1 after 8 weeks of treatment. Clinical efficacy in patients with the G970R mutation of the CFTR gene could not be established (see section 5.1).

Efficacy results from a Phase 2 study in patients with CF who are homozygous for the F508del mutation in the CFTR gene showed no statistically significant difference in FEV1 over 16 weeks of ivacaftor treatment compared to placebo (see section 5.1). Therefore, use of Kalydeco monotherapy in these patients is not recommended.

Less evidence of a positive effect of ivacaftor has been shown for patients with an R117H-7T mutation associated with less severe disease in study 6 (see section 5.1).

Kalydeco in combination with tezacaftor/ivacaftor should not be prescribed in patients with CF who are heterozygous for the F508del mutation and have a second CFTR mutation not listed in section 4.1.

Effect on liver function tests

Moderate transaminase (alanine transaminase [ALT] or aspartate transaminase [AST]) elevations are common in subjects with CF. Transaminase elevations have been observed in some patients treated with ivacaftor monotherapy and in a combination regimen with tezacaftor/ivacaftor. Therefore, liver function tests are recommended for all patients prior to initiating ivacaftor, every 3 months during the first year of treatment and annually thereafter. For all patients with a history of transaminase elevations, more frequent monitoring of liver function tests should be considered. In the event of significant elevations of transaminases (e.g., patients with ALT or AST > 5 x the upper limit of normal (ULN), or ALT or AST > 3 x ULN with bilirubin > 2 x ULN), dosing should be interrupted and laboratory tests closely followed until the abnormalities resolve. Following resolution of transaminase elevations, the benefits and risks of resuming treatment should be considered (see section 4.8).

Hepatic impairment

Use of ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, is not recommended in patients with severe hepatic impairment unless the benefits are expected to outweigh the risks (see sections 4.2 and 5.2).

Renal impairment

Caution is recommended while using ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, in patients with severe renal impairment or end-stage renal disease (see sections 4.2 and 5.2).

Patients after organ transplantation

Ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, has not been studied in patients with CF who have undergone organ transplantation. Therefore, use in transplanted patients is not recommended. See section 4.5 for interactions with ciclosporin or tacrolimus.

Interactions with medicinal products

CYP3A inducers

Exposure to ivacaftor may be reduced by the concomitant use of CYP3A inducers, potentially resulting in the loss of ivacaftor efficacy. Therefore, co-administration of Kalydeco (monotherapy or in combination with tezacaftor/ivacaftor) with strong CYP3A inducers is not recommended (see section 4.5).

CYP3A inhibitors

The dose of Kalydeco (monotherapy or in combination with tezacaftor/ivacaftor) must be adjusted when used concomitantly with strong or moderate CYP3A inhibitors (see sections 4.2 and 4.5).

Cataracts

Cases of non-congenital lens opacities without impact on vision have been reported in paediatric patients treated with ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor. Although other risk factors were present in some cases (such as corticosteroid use and exposure to radiation), a possible risk attributable to treatment cannot be excluded. Baseline and follow-up ophthalmological examinations are recommended in paediatric patients initiating ivacaftor treatment, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor (see section 5.3).

Lactose content

Kalydeco contains lactose. Patients with rare hereditary problems of galactose intolerance, total lactase deficiency or glucose-galactose malabsorption should not take this medicine.

Sodium content

This medicine contains less than 1 mmol sodium (23 mg) per dose, that is to say essentially 'sodium-free'.


4.5. Interaction with other medicinal products and other forms of interaction

Ivacaftor is a substrate of CYP3A4 and CYP3A5. It is a weak inhibitor of CYP3A and P-gp and a potential inhibitor of CYP2C9. In vitro studies showed that ivacaftor is not a substrate for P-gp.

Medicinal products affecting the pharmacokinetics of ivacaftor

CYP3A inducers

Co-administration of ivacaftor with rifampicin, a strong CYP3A inducer, decreased ivacaftor exposure (AUC) by 89% and decreased hydroxymethyl ivacaftor (M1) to a lesser extent than ivacaftor. Co-administration of Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) with strong CYP3A inducers, such as rifampicin, rifabutin, phenobarbital, carbamazepine, phenytoin and St. John's wort (Hypericum perforatum), is not recommended (see section 4.4).

No dose adjustment is recommended when Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is used with moderate or weak CYP3A inducers.

CYP3A inhibitors

Ivacaftor is a sensitive CYP3A substrate. Co-administration with ketoconazole, a strong CYP3A inhibitor, increased ivacaftor exposure (measured as area under the curve [AUC]) by 8.5-fold and increased M1 to a lesser extent than ivacaftor. A reduction of the Kalydeco dose (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is recommended for co-administration with strong CYP3A inhibitors, such as ketoconazole, itraconazole, posaconazole, voriconazole, telithromycin and clarithromycin (see sections 4.2 and 4.4).

Co-administration with fluconazole, a moderate inhibitor of CYP3A, increased ivacaftor exposure by 3-fold and increased M1 to a lesser extent than ivacaftor. A reduction of the Kalydeco dose (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is recommended for patients taking concomitant moderate CYP3A inhibitors, such as fluconazole and erythromycin (see sections 4.2 and 4.4).

Co-administration of ivacaftor with grapefruit juice, which contains one or more components that moderately inhibit CYP3A, may increase exposure to ivacaftor. Food or drink containing grapefruit or Seville oranges should be avoided during treatment with Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor, see section 4.2).

Ciprofloxacin

Co-administration of ciprofloxacin with ivacaftor did not affect the exposure of ivacaftor. No dose adjustment is required when Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is co-administered with ciprofloxacin.

Medicinal products affected by ivacaftor:

Administration of ivacaftor may increase systemic exposure of medicinal products that are sensitive substrates of CYP2C9, and/or P-gp, and/or CYP3A which may increase or prolong their therapeutic effect and adverse reactions.

CYP2C9 substrates

Ivacaftor may inhibit CYP2C9. Therefore, monitoring of the international normalised ratio (INR) is recommended during co-administration of warfarin with Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor). Other medicinal products for which exposure may be increased include glimepiride and glipizide; these medicinal products should be used with caution.

Digoxin and other P-gp substrates

Co-administration with digoxin, a sensitive P-gp substrate, increased digoxin exposure by 1.3-fold, consistent with weak inhibition of P-gp by ivacaftor. Administration of Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) may increase systemic exposure of medicinal products that are sensitive substrates of P-gp, which may increase or prolong their therapeutic effect and adverse reactions. When used concomitantly with digoxin or other substrates of P-gp with a narrow therapeutic index, such as ciclosporin, everolimus, sirolimus or tacrolimus, caution and appropriate monitoring should be used.

CYP3A substrates

Co-administration with (oral) midazolam, a sensitive CYP3A substrate, increased midazolam exposure 1.5-fold, consistent with weak inhibition of CYP3A by ivacaftor. No dose adjustment of CYP3A substrates, such as midazolam, alprazolam, diazepam or triazolam, is required when these are co-administered with ivacaftor (as monotherapy or in a combination regimen with tezacaftor/ivacaftor).

Hormonal contraceptives

Ivacaftor (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) has been studied with an oestrogen/progesterone oral contraceptive and was found to have no significant effect on the exposures of the oral contraceptive. Therefore, no dose adjustment of oral contraceptives is necessary.

Potential for ivacaftor to interact with transporters

In vitro studies showed that ivacaftor is not a substrate for OATP1B1 or OATP1B3. Ivacaftor and its metabolites are substrates of BCRP in vitro. Due to its high intrinsic permeability and low likelihood of being excreted intact, co-administration of BCRP inhibitors is not expected to alter exposure of ivacaftor and M1-IVA, while any potential changes in M6-IVA exposures are not expected to be clinically relevant.

Paediatric population

Interaction studies have only been performed in adults.


4.6. Fertility, pregnancy and lactation

Pregnancy

There are no or limited amount of data (less than 300 pregnancy outcomes) from the use of ivacaftor in pregnant women. Animals studies do not indicate direct or indirect harmful effects with respect to reproductive toxicity (see section 5.3). As a precautionary measure, it is preferable to avoid the use of Kalydeco during pregnancy.

Breast-feeding

It is unknown whether ivacaftor and/or its metabolites are excreted in human milk. Available pharmacokinetic data in animals have shown excretion of ivacaftor into the milk of lactating female rats. As such, a risk to the newborns/infants cannot be excluded. A decision must be made whether to discontinue breast-feeding or to discontinue/abstain from Kalydeco therapy taking into account the benefit of breast-feeding for the child and the benefit of therapy for the woman.

Fertility

There are no data available on the effect of ivacaftor on fertility in humans. Ivacaftor had an effect on fertility in rats (see section 5.3).


4.7. Effects on ability to drive and use machines

Kalydeco has minor influence on the ability to drive or use machines. Ivacaftor may cause dizziness (see section 4.8) and, therefore, patients experiencing dizziness should be advised not to drive or use machines until symptoms abate.


4.8. Undesirable effects

Summary of the safety profile

The most common adverse reactions experienced by patients aged 6 years and older who received ivacaftor in the pooled 48-week placebo-controlled Phase 3 studies that occurred with an incidence of at least 3% and up to 9% higher than in the placebo arm were headache (23.9%), oropharyngeal pain (22.0%), upper respiratory tract infection (22.0%), nasal congestion (20.2%), abdominal pain (15.6%), nasopharyngitis (14.7%), diarrhoea (12.8%), dizziness (9.2%), rash (12.8%) and bacteria in sputum (12.8%). Transaminase elevations occurred in 12.8% of ivacaftor-treated patients versus 11.5% of placebo-treated patients.

In patients aged 2 to less than 6 years the most common adverse reactions were nasal congestion (26.5%), upper respiratory tract infection (23.5%), transaminase elevations (14.7%), rash (11.8%), and bacteria in sputum (11.8%).

Serious adverse reactions in patients who received ivacaftor included abdominal pain and transaminase elevations (see section 4.4).

Tabulated list of adverse reactions

Table 3 reflects the adverse reactions observed with ivacaftor monotherapy in clinical trials (placebo-controlled and uncontrolled studies) in which the length of exposure to ivacaftor ranged from 16 weeks to 144 weeks. Additional adverse reactions observed with ivacaftor in combination with tezacaftor/ivacaftor are also provided in Table 3. The frequency of adverse reactions is defined as follows: very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000); not known (cannot be estimated from the available data). Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness.

Table 3: Adverse reactions in patients treated with ivacaftor monotherapy or in combination with tezacaftor

System organ class

Adverse reactions

Frequency

Infections and infestations

Upper respiratory tract infection

very common

Nasopharyngitis

very common

Rhinitis

common

Nervous system disorders

Headache

very common

Dizziness

very common

Ear and labyrinth disorders

Ear pain

common

Ear discomfort

common

Tinnitus

common

Tympanic membrane hyperaemia

common

Vestibular disorder

common

Ear congestion

uncommon

Respiratory, thoracic and mediastinal disorders

Oropharyngeal pain

very common

Nasal congestion

very common

Sinus congestion

common

Pharyngeal erythema

common

Gastrointestinal disorders

Abdominal pain

very common

Diarrhoea

very common

Nausea *

common

Hepatobiliary disorders

Transaminase elevations

very common

Skin and subcutaneous tissue disorders

Rash

very common

Reproductive system and breast disorders

Breast mass

common

Breast inflammation

uncommon

Gynaecomastia

uncommon

Nipple disorder

uncommon

Nipple pain

uncommon

Investigations

Bacteria in sputum

very common

* Adverse reaction and frequency reported only in clinical studies with ivacaftor in combination with tezacaftor/ivacaftor.

Description of selected adverse reactions

Hepatobiliary disorders

Transaminase elevations

During the 48-week placebo-controlled studies 1 and 2 of ivacaftor monotherapy in patients aged 6 years and older, the incidence of maximum transaminase (ALT or AST) > 8, > 5 or > 3 x ULN was 3.7%, 3.7% and 8.3% in ivacaftor-treated patients and 1.0%, 1.9% and 8.7% in placebo-treated patients, respectively. Two patients, one on placebo and one on ivacaftor permanently discontinued treatment for elevated transaminases, each > 8 x ULN. No ivacaftor-treated patients experienced a transaminase elevation > 3 x ULN associated with elevated total bilirubin > 1.5 x ULN. In ivacaftor-treated patients, most transaminase elevations up to 5 x ULN resolved without treatment interruption. Ivacaftor dosing was interrupted in most patients with transaminase elevations > 5 x ULN. In all instances where dosing was interrupted for elevated transaminases and subsequently resumed, ivacaftor dosing was able to be resumed successfully (see section 4.4).

During the placebo controlled Phase 3 studies (up to 24 weeks) of ivacaftor in a combination regimen with tezacaftor/ivacaftor, the incidence of maximum transaminase (ALT or AST) > 8, > 5, or > 3 x ULN were similar between tezacaftor/ivacaftor in combination with ivacaftor and placebo-treated patients; 0.2%, 1.0%, and 3.4% in tezacaftor/ivacaftor in combination with ivacaftor-treated patients, and 0.4%, 1.0%, and 3.4% in placebo-treated patients. One patient (0.2%) on therapy and 2 patients (0.4%) on placebo permanently discontinued treatment for elevated transaminases. No patients treated with tezacaftor/ivacaftor discontinued treatment for elevated transaminases.

Paediatric population

The safety data of ivacaftor monotherapy were evaluated in 11 patients between 6 months to less than 12 months of age, 19 patients between 12 months to less than 24 months of age, 34 patients between 2 to less than 6 years of age, 61 patients between 6 to less than 12 years of age and 94 patients between 12 to less than 18 years of age. The safety data of ivacaftor in combination with tezacaftor/ivacaftor was evaluated in 98 patients between 12 to less than 18 years of age.

The safety profile is generally consistent among paediatric patients aged 6 months and older and adult patients.

During the 24-week open-label Phase 3 clinical study of ivacaftor monotherapy in 34 patients aged 2 to less than 6 years (study 7), 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 dosing with ivacaftor granules. Ivacaftor was permanently discontinued in one patient. In children aged 6 to less than 12 years, the incidence of patients experiencing transaminase elevations (ALT or AST) > 3 x ULN was 15.0% (6/40) in ivacaftor-treated patients and 14.6% (6/41) in patients who received placebo. A single ivacaftor-treated patient (2.5%) in this age range had an elevation of ALT and AST > 8 x ULN. Peak LFT (ALT or AST) elevations were generally higher in paediatric patients than in older patients. In almost all instances where dosing was interrupted for elevated transaminases and subsequently resumed, ivacaftor dosing was able to be resumed successfully (see section 4.4). Cases suggestive of positive rechallenge were observed.

During the 24-week, open-label, Phase 3 clinical study of ivacaftor in patients aged less than 24 months (study 8), the incidence of patients experiencing transaminase elevations (ALT or AST) > 3, > 5, and > 8 x ULN in the cohort of patients aged 12 months to less than 24 months was 27.8% (5/18), 11.1% (2/18) and 11.1% (2/18), respectively. In the cohort of patients aged 6 months to less than 12 months one patient (9.1%) had elevated ALT of > 3 to ≤5 x ULN. No patients had elevations in total bilirubin or discontinued ivacaftor treatment due to transaminase elevations in either cohort. The two patients with elevations of ALT or AST > 8 x ULN interrupted treatment and subsequently resumed ivacaftor successfully (see section 4.4 for management of elevated transaminases).

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via the

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard or search for MHRA Yellow Card in the Google Play or Apple App Store


4.9. Overdose

No specific antidote is available for overdose with ivacaftor. Treatment of overdose consists of general supportive measures including monitoring of vital signs, liver function tests and observation of the clinical status of the patient.


5.1. Pharmacodynamic properties

Pharmacotherapeutic group: Other respiratory system products, ATC code: R07AX02

Mechanism of action

Ivacaftor is a potentiator of the CFTR protein, i.e., in vitro ivacaftor increases CFTR channel gating to enhance chloride transport in specified gating mutations (as listed in section 4.1) with reduced channel-open probability compared to normal CFTR. Ivacaftor also potentiated the channel-open probability of R117H-CFTR, which has both low channel-open probability (gating) and reduced channel current amplitude (conductance). The G970R mutation causes a splicing defect resulting in little-to-no CFTR protein at the cell surface which may explain the results observed in subjects with this mutation in study 5 (see Pharmacodynamic effects and Clinical efficacy data).

In vitro responses seen in single channel patch clamp experiments using membrane patches from rodent cells expressing mutant CFTR forms do not necessarily correspond to in vivo pharmacodynamic response (e.g., sweat chloride) or clinical benefit. The exact mechanism leading ivacaftor to potentiate the gating activity of normal and some mutant CFTR forms in this system has not been completely elucidated.

Pharmacodynamic effects

Ivacaftor monotherapy

In studies 1 and 2 in patients with the G551D mutation in one allele of the CFTR gene, ivacaftor led to rapid (15 days), substantial (the mean change in sweat chloride from baseline through week 24 was -48 mmol/L [95% CI -51, -45] and -54 mmol/L [95% CI -62, -47], respectively) and sustained (through 48 weeks) reductions in sweat chloride concentration.

In study 5, part 1 in patients who had a non-G551D gating mutation in the CFTR gene, treatment with ivacaftor led to a rapid (15 days) and substantial mean change from baseline in sweat chloride of -49 mmol/L (95% CI -57, -41) through 8 weeks of treatment. However, in patients with the G970R-CFTR mutation, the mean (SD) absolute change in sweat chloride at week 8 was -6.25 (6.55) mmol/L. Similar results to part 1 were seen in part 2 of the study. At the 4-week follow-up visit (4 weeks after dosing with ivacaftor ended), mean sweat chloride values for each group were trending to pre-treatment levels.

In study 6 in patients aged 6 years or older with CF who had an R117H mutation in the CFTR gene, the treatment difference in mean change in sweat chloride from baseline through 24 weeks of treatment was -24 mmol/L (95% CI -28, -20). In subgroup analyses by age, the treatment difference was 21.87 mmol/L (95% CI: 26.46, 17.28) in patients aged 18 years or older, and 27.63 mmol/L (95% CI: 37.16, 18.10) in patients aged 6 11 years. Two patients 12 to 17 years of age were enrolled in this study.

Ivacaftor in a combination regimen with tezacaftor/ivacaftorIn patients homozygous for the F508del mutation, the treatment difference between ivacaftor in combination with tezacaftor/ivacaftor and placebo in mean absolute change from baseline in sweat chloride through week 24, was -10.1 mmol/L (95% CI: -11.4, -8.8).

In patients heterozygous for the F508del mutation and a second mutation associated with residual CFTR activity, the treatment difference in mean absolute change from baseline in sweat chloride through week 8 was -9.5 mmol/L (95% CI: -11.7, -7.3) between ivacaftor in combination with tezacaftor/ivacaftor and placebo, and -4.5 mmol/L (95% CI: -6.7, -2.3) between ivacaftor and placebo.

Clinical efficacy and safety

Kalydeco monotherapy

Study 1 and 2: studies in patients with CF with G551D gating mutations

The efficacy of Kalydeco has been evaluated in two Phase 3 randomised, double-blind, placebo-controlled, multi-centre studies of clinically stable patients with CF who had the G551D mutation in the CFTR gene on at least 1 allele and had FEV1 ≥ 40% predicted.

Patients in both studies were randomised 1:1 to receive either 150 mg of ivacaftor 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 sodium chloride was not permitted.

Study 1 evaluated 161 patients who were 12 years of age or older; 122 (75.8%) patients had the F508del mutation in the second allele. At the start of the study, patients in the placebo group used some medicinal products at a higher frequency than the ivacaftor group. These medications included dornase alfa (73.1% versus 65.1%), salbutamol (53.8% versus 42.2%), tobramycin (44.9% versus 33.7%) and salmeterol/fluticasone (41.0% versus 27.7%). At baseline, mean predicted FEV1 was 63.6% (range: 31.6% to 98.2%) and mean age was 26 years (range: 12 to 53 years).

Study 2 evaluated 52 patients who were 6 to 11 years of age at screening; mean (SD) body weight was 30.9 (8.63) kg; 42 (80.8%) patients had the F508del mutation in the second allele. At baseline, mean predicted FEV1 was 84.2% (range: 44.0% to 133.8%) and mean age was 9 years (range: 6 to 12 years); 8 (30.8%) patients in the placebo group and 4 (15.4%) patients in the ivacaftor group had an FEV1 less than 70% predicted at baseline.

The primary efficacy endpoint in both studies was the mean absolute change from baseline in percent predicted FEV1 through 24 weeks of treatment.

The treatment difference between ivacaftor and placebo for the mean absolute change (95% CI) in percent predicted FEV1 from baseline through week 24 was 10.6 percentage points (8.6, 12.6) in study 1 and 12.5 percentage points (6.6, 18.3) in study 2. The treatment difference between ivacaftor and placebo for the mean relative change (95% CI) in percent predicted FEV1 from baseline through week 24 was 17.1% (13.9, 20.2) in study 1 and 15.8% (8.4, 23.2) in study 2. The mean change from baseline through week 24 in FEV1 (L) was 0.37 L in the ivacaftor group and 0.01 L in the placebo group in study 1 and 0.30 L in the ivacaftor group and 0.07 L in the placebo group in study 2. In both studies, improvements in FEV1 were rapid in onset (day 15) and durable through 48 weeks.

The treatment difference between ivacaftor and placebo for the mean absolute change (95% CI) in percent predicted FEV1 from baseline through week 24 in patients 12 to 17 years of age in study 1 was 11.9 percentage points (5.9, 17.9). The treatment difference between ivacaftor and placebo for the mean absolute change (95% CI) in percent predicted FEV1 from baseline through week 24 in patients with baseline predicted FEV1 greater than 90% in study 2 was 6.9 percentage points (-3.8, 17.6).

The results for clinically relevant secondary endpoints are shown in Table 4.

Table 4: Effect of ivacaftor on other efficacy endpoints in studies 1 and 2

Endpoint

Study 1

Study 2

Treatment differencea

(95% CI)

P value

Treatment differencea

(95% CI)

P value

Mean absolute change from baseline in CFQ-Rb respiratory domain score (points)c

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

0.40d

0.0016

NA

NA

Through week 48

0.46d

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

Mean absolute change from baseline in BMI (kg/m2)

At week 24

0.94

(0.62, 1.26)

< 0.0001

0.81

(0.34, 1.28)

0.0008

At week 48

0.93

(0.48, 1.38)

< 0.0001

1.09

(0.51, 1.67)

0.0003

Mean change from baseline in z-scores

Weight-for-age z-score at week 48e

0.33

(0.04, 0.62)

0.0260

0.39

(0.24, 0.53)

< 0.0001

BMI-for-age z-score at week 48e

0.33

(0.002, 0.65)

0.0490

0.45

(0.26, 0.65)

< 0.0001

CI: confidence interval; NA: not analysed due to low incidence of events

a Treatment difference = effect of ivacaftor – effect of placebo

b CFQ-R: Cystic Fibrosis Questionnaire-Revised is a disease-specific, health-related quality-of-life measure for CF.

c Study 1 data were pooled from CFQ-R for adults/adolescents and CFQ-R for children 12 to 13 years of age; Study 2 data were obtained from CFQ-R for children 6 to 11 years of age.

d Hazard ratio for time to first pulmonary exacerbation

e In subjects under 20 years of age (CDC growth charts)

Study 5: study in patients with CF with non-G551D gating mutations

Study 5 was a Phase 3, two-part, randomised, double-blind, placebo-controlled, crossover study (part 1) followed by a 16-week open-label extension period (part 2) to evaluate the efficacy and safety of ivacaftor in patients with CF aged 6 years and older who have a G970R or non-G551D gating mutation in the CFTR gene (G178R, S549N, S549R, G551S, G1244E, S1251N, S1255P or G1349D).

In part 1, patients were randomised 1:1 to receive either 150 mg of ivacaftor or placebo every 12 hours with fat-containing food for 8 weeks in addition to their prescribed CF therapies and crossed over to the other treatment for the second 8 weeks after a 4- to 8-week washout period. The use of inhaled hypertonic saline was not permitted. In part 2, all patients received ivacaftor as indicated in part 1 for 16 additional weeks. The duration of continuous ivacaftor treatment was 24 weeks for patients randomised to part 1 placebo/ivacaftor treatment sequence and 16 weeks for patients randomised to part 1 ivacaftor/placebo treatment sequence.

Thirty-nine patients (mean age 23 years) with baseline FEV1 ≥ 40% predicted (mean FEV1 78% predicted [range: 43% to 119%]) were enrolled. Sixty-two percent (24/39) of them carried the F508del-CFTR mutation in the second allele. A total of 36 patients continued into part 2 (18 per treatment sequence).

In part 1 of study 5, the mean FEV1 percent predicted at baseline in placebo-treated patients was 79.3% while in ivacaftor-treated patients this value was 76.4%. The mean overall post-baseline value was 76.0% and 83.7%, respectively. The mean absolute change from baseline through week 8 in percent predicted FEV1 (primary efficacy endpoint) was 7.5% in the ivacaftor period and -3.2% in the placebo period. The observed treatment difference (95% CI) between ivacaftor and placebo was 10.7% (7.3, 14.1) (P < 0.0001).

The effect of ivacaftor in the overall population of study 5 (including the secondary endpoints absolute change in BMI at 8 weeks of treatment and absolute change in the respiratory domain score of the CFQ-R through 8 weeks of treatment) and by individual mutation (absolute change in sweat chloride and in percent predicted FEV1 at week 8) is shown in Table 5. Based on clinical (percent predicted FEV1) and pharmacodynamic (sweat chloride) responses to ivacaftor, efficacy in patients with the G970R mutation could not be established.

Table 5: Effect of ivacaftor for efficacy variables in the overall population and for specific CFTR mutations

Absolute change in percent predicted FEV1

BMI

(kg/m2)

CFQ-R respiratory domain score (points)

Through week 8

At week 8

Through week 8

All patients (N = 39)

Results shown as mean (95% CI) change from baseline ivacaftor vs placebo-treated patients:

10.7 (7.3, 14.1)

0.66 (0.34, 0.99)

9.6 (4.5, 14.7)

Patients grouped under mutation types (n)

Results shown as mean (minimum, maximum) change from baseline for ivacaftor-treated patients at week 8*:

Mutation (n)

Absolute change in sweat chloride (mmol/L)

Absolute change in percent predicted FEV1 (percentage points)

At week 8

At week 8

G1244E (5)

G1349D (2)

G178R (5)

G551S (2)

G970R# (4)

S1251N (8)

S1255P (2)

S549N (6)

S549R (4)

‑55 (-75, -34)

-80 (-82, -79)

-53 (-65, -35)

-68

-6 (-16, -2)

-54 (-84, -7)

-78 (-82, -74)

-74 (-93, -53)

-61†† (-71, -54)

8 (-1, 18)

20 (3, 36)

8 (-1, 18)

3

3 (-1, 5)

9 (-20, 21)

3 (-1, 8)

11 (-2, 20)

5 (-3, 13)

* Statistical testing was not performed due to small numbers for individual mutations.

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.

# Causes a splicing defect resulting in little-to-no CFTR protein at the cell surface

In part 2 of study 5, the mean (SD) absolute change in percent predicted FEV1 following 16 weeks (patients randomised to the ivacaftor/placebo treatment sequence in part 1) of continuous ivacaftor treatment was 10.4% (13.2%). At the follow-up visit, 4 weeks after ivacaftor dosing had ended, the mean (SD) absolute change in percent predicted FEV1 from part 2 week 16 was -5.9% (9.4%). For patients randomised to the placebo/ivacaftor treatment sequence in part 1 there was a further mean (SD) change of 3.3% (9.3%) in percent predicted FEV1 after the additional 16 weeks of treatment with ivacaftor. At the follow up visit, 4 weeks after ivacaftor dosing had ended, the mean (SD) absolute change in percent predicted FEV1 from part 2 week 16 was -7.4% (5.5%).

Study 3: study in patients with CF with the F508del mutation in the CFTR gene

Study 3 (part A) was a 16-week, 4:1 randomised, double-blind, placebo-controlled, parallel-group Phase 2 study of ivacaftor (150 mg every 12 hours) 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.

The mean absolute change from baseline through week 16 in percent predicted FEV1 (primary efficacy endpoint) was 1.5 percentage points in the ivacaftor group and -0.2 percentage points in the placebo group. The estimated treatment difference for ivacaftor versus placebo was 1.7 percentage points (95% CI -0.6, 4.1); this difference was not statistically significant (P = 0.15).

Study 4: open-label extension study

In study 4 patients who completed treatment in studies 1 and 2 with placebo were switched to ivacaftor while patients on ivacaftor continued to receive it for a minimum of 96 weeks, i.e., the length of treatment with ivacaftor was at least 96 weeks for patients in the placebo/ivacaftor group and at least 144 weeks for patients in the ivacaftor/ivacaftor group.

One hundred and forty-four (144) patients from study 1 were rolled over in study 4, 67 in the placebo/ivacaftor group and 77 in the ivacaftor/ivacaftor group. Forty-eight (48) patients from study 2 were rolled over in study 4, 22 in the placebo/ivacaftor group and 26 in the ivacaftor/ivacaftor group.

Table 6 shows the results of the mean (SD) absolute change in percent predicted FEV1 for both groups of patients. For patients in the placebo/ivacaftor group baseline percent predicted FEV1 is that of study 4 while for patients in the ivacaftor/ivacaftor group the baseline value is that of studies 1 and 2.

Table 6: Effect of ivacaftor on percent predicted FEV1 in study 4

Original study and treatment group

Duration of ivacaftor treatment (weeks)

Absolute change from baseline in percent predicted FEV1 (percentage points)

N

Mean (SD)

Study 1

Ivacaftor

48*

77

9.4 (8.3)

144

72

9.4 (10.8)

Placebo

0*

67

-1.2 (7.8)

96

55

9.5 (11.2)

Study 2

Ivacaftor

48*

26

10.2 (15.7)

144

25

10.3 (12.4)

Placebo

0*

22

-0.6 (10.1)

96

21

10.5 (11.5)

* Treatment occurred during blinded, controlled, 48-week Phase 3 study.

Change from prior study baseline after 48 weeks of placebo treatment.

When the mean (SD) absolute change in percent predicted FEV1 is compared from study 4 baseline for patients in the ivacaftor/ivacaftor group (n = 72) who rolled over from study 1, the mean (SD) absolute change in percent predicted FEV1 was 0.0% (9.05), while for patients in the ivacaftor/ivacaftor group (n = 25) who rolled over from study 2 this figure was 0.6% (9.1). This shows that patients in the ivacaftor/ivacaftor group maintained the improvement seen at week 48 of the initial study (day 0 through week 48) in percent predicted FEV1 through week 144. There were no additional improvements in study 4 (week 48 through week 144).

For patients in the placebo/ivacaftor group from study 1, the annualised rate of pulmonary exacerbations was higher in the initial study when patients were on placebo (1.34 events/year) than during the subsequent study 4 when patients rolled over to ivacaftor (0.48 events/year across day 1 to week 48, and 0.67 events/year across weeks 48 to 96). For patients in the ivacaftor/ivacaftor group from study 1, the annualised rate of pulmonary exacerbations was 0.57 events/year across day 1 to week 48 when patients were on ivacaftor. When they rolled over into study 4, the rate of annualised pulmonary exacerbations was 0.91 events/year across day 1 to week 48 and 0.77 events/year across weeks 48 to 96.

For patients who rolled over from study 2 the number of events was, overall, low.

Study 6: study in patients with CF with an R117H mutation in the CFTR gene

Study 6 evaluated 69 patients who were 6 years of age or older; 53 (76.8%) patients had the F508del mutation in the second allele. The confirmed R117H poly-T variant was 5T in 38 patients and 7T in 16 patients. At baseline, mean predicted FEV1 was 73% (range: 32.5% to 105.5%) and mean age was 31 years (range: 6 to 68 years). The mean absolute change from baseline through week 24 in percent predicted FEV1 (primary efficacy endpoint) was 2.57 percentage points in the ivacaftor group and 0.46 percentage points in the placebo group. The estimated treatment difference for ivacaftor versus placebo was 2.1 percentage points (95% CI -1.1, 5.4).

A pre-planned subgroup analysis was conducted in patients 18 years and older (26 patients on placebo and 24 on ivacaftor). Treatment with ivacaftor resulted in a mean absolute change in percent predicted FEV1 through week 24 of 4.5 percentage points in the ivacaftor group versus -0.46 percentage points in the placebo group. The estimated treatment difference for ivacaftor versus placebo was 5.0 percentage points (95% CI 1.1, 8.8).

In a subgroup analysis in patients with a confirmed R117H-5T genetic variant, the difference in the mean absolute change from baseline through week 24 in percent predicted FEV1 between ivacaftor and placebo was 5.3% (95% CI 1.3, 9.3). In patients with a confirmed R117H-7T genetic variant, the treatment difference between ivacaftor and placebo was 0.2% (95% CI -8.1, 8.5).

For secondary efficacy variables, no treatment differences were observed for ivacaftor versus placebo in absolute change from baseline in BMI at week 24 or time to first pulmonary exacerbation.Treatment differences were observed in absolute change in CFQ R respiratory domain score through week 24 (treatment difference of ivacaftor versus placebo was 8.4 [95% CI 2.2, 14.6] points) and for the mean change from baseline in sweat chloride (see Pharmacodynamic effects).

Kalydeco in a combination regimen with tezacaftor/ivacaftor

The efficacy and safety of Kalydeco in a combination regimen with tezacaftor/ivacaftor in patients with CF was assessed in two clinical studies; a 24 week, randomized, double-blind, placebo-controlled study with 504 patients aged 12 years and older who were homozygous for the F508del mutation; and a randomized, double-blind, placebo-controlled and ivacaftor controlled, 2 period, 3 treatment, 8-week crossover study with 244 patients aged 12 years and older who were heterozygous for the F508del mutation and a second mutation associated with residual CFTR activity. An open-label, rollover, 96 week study is on-going to evaluate the long-term safety and efficacy of the combination regimen in both patient populations. Refer to the Summary of Product Characteristics of tezacaftor/ivacaftor for additional data.

Paediatric population

The European Medicines Agency has deferred the obligation to submit the results of studies with Kalydeco in one or more subsets of the paediatric population in cystic fibrosis (see section 4.2 for information on paediatric use).


5.2. Pharmacokinetic properties

The pharmacokinetics of ivacaftor are 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, 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

Following multiple oral dose administrations of ivacaftor, the exposure of ivacaftor generally increased with dose from 25 mg every 12 hours to 450 mg every 12 hours. The exposure of ivacaftor increased approximately 2.5- to 4-fold when given with food containing fat. The AUC of ivacaftor when given in combination with tezacaftor increased approximately 3-fold when given with fat containing food. Ivacaftor, administered as monotherapy or in combination with tezacaftor, should be administered with fat-containing food. The median (range) tmax is approximately 4.0 (3.0; 6.0) hours in the fed state.

Ivacaftor granules (2 x 75 mg sachets) had similar bioavailability as the 150 mg tablet when given with fat-containing food to healthy adult subjects. The geometric least squares mean ratio (90% CI) for the granules relative to tablets was 0.951 (0.839, 1.08) for AUC0-∞ and 0.918 (0.750, 1.12) for Cmax. The effect of food on ivacaftor absorption is similar for both formulations, i.e., tablets and granules.

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 ivacaftor150 mg every 12 hours for 7 days in healthy volunteers in a fed state, the mean (± SD) apparent volume of distribution was 353 (122) L.

After oral administration of ivacaftor 150 mg every 12 hours in combination with tezacaftor 100 mg once daily in patients with CF in the fed state, the mean (± SD) for apparent volume of distribution of ivacaftor was 206 (82.9) L.

Biotransformation

Ivacaftor is extensively metabolised in humans. In vitro and in vivo data indicate that ivacaftor is primarily metabolised 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.

The effect of the potentially reduced activity of CYP3A4 in patients carrying the CYP3A4*22 variant on ivacaftor exposure is not known.

Elimination

Following oral administration in healthy volunteers, the majority of ivacaftor (87.8%) was eliminated in the faeces 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 in the fed state. The apparent clearance (CL/F) of ivacaftor was similar for healthy subjects and patients with CF. The mean (± SD) CL/F for a single 150 mg dose was 17.3 (8.4) L/hr in healthy subjects.

After oral administration of ivacaftor 150 mg every 12 hours in combination with tezacaftor 100 mg once daily in patients with CF in the fed state, the mean (± SD) for apparent clearance of ivacaftor was 15.7 (6.38) L/h. After steady-state dosing of ivacaftor in combination with tezacaftor in CF patients, the mean (SD) terminal half-life of ivacaftor was approximately 9.3 (1.7) hours.

Linearity/non-linearity

The pharmacokinetics of ivacaftor are generally linear with respect to time or dose ranging from 25 mg to 250 mg.

Special populations

Hepatic impairment

Following a single dose of 150 mg of ivacaftor, adult subjects with moderately impaired hepatic function (Child-Pugh Class B, score 7 to 9) had similar ivacaftor Cmax (mean [± SD] of 735 [331] ng/mL) but an approximately two-fold increase in ivacaftor AUC0-∞ (mean [± SD] of 16800 [6140] ng*hr/mL) compared with healthy subjects matched for demographics. Simulations for predicting the steady-state exposure of ivacaftor showed that by reducing the dosage from 150 mg q12h to 150 mg once daily, adults with moderate hepatic impairment would have comparable steady-state Cmin values as those obtained with a dose of 150 mg q12h in adults without hepatic impairment. Based on these results, a modified regimen of Kalydeco monotherapy is recommended for patients with moderate hepatic impairment (see Table 2 in section 4.2).

Following multiple doses of ivacaftor and tezacaftor for 10 days, subjects with moderately impaired hepatic function (Child-Pugh Class B, score 7 to 9) had a 50% increase in ivacaftor AUC, and an approximate 36% and 10% increase in AUC and Cmax for tezacaftor, respectively. Based on these results, a modified regimen for Kalydeco in a combination regimen with tezacaftor/ivacaftor is recommended for patients with moderate hepatic impairment (see Table 2 in section 4.2).

The impact of severe hepatic impairment (Child Pugh Class C, score 10 to15) on the pharmacokinetics of ivacaftor monotherapy or in combination with tezacaftor/ivacaftor have not been studied. The magnitude of increase in exposure in these patients is unknown but is expected to be higher than that observed in patients with moderate hepatic impairment. The use of Kalydeco monotherapy or in combination with tezacaftor/ivacaftor in patients with severe hepatic impairment is therefore not recommended unless the benefits outweigh the risks (see Table 2 in section 4.2 and section 4.4).

No dose adjustment is considered necessary for patients with mild hepatic impairment.

Renal impairment

Pharmacokinetic studies have not been performed with ivacaftor in patients with renal impairment, either as monotherapy or in a combination regimen with tezacaftor/ivacaftor. In a human pharmacokinetic study with ivacaftor monotherapy, there was minimal elimination of ivacaftor and its metabolites in urine (only 6.6% of total radioactivity was recovered in the urine). There was negligible urinary excretion of ivacaftor as unchanged parent (less than 0.01% following a single oral dose of 500 mg).

No dose adjustments are recommended for mild and moderate renal impairment. Caution is recommended when administering ivacaftor, either as monotherapy or in combination with tezacaftor, to patients with severe renal impairment (creatinine clearance less than or equal to 30 mL/min) or end-stage renal disease (see sections 4.2 and 4.4).

Race

Race had no clinically meaningful effect on the PK of ivacaftor in white (n = 379) and non-white (n = 29) patients based on a population PK analysis.

Gender

The pharmacokinetic parameters of ivacaftor, either as monotherapy or in combination with tezacaftor, are similar in males and females.

Elderly

Clinical studies of ivacaftor monotherapy did not include sufficient numbers of patients age 65 years and older to determine whether pharmacokinetic parameters are similar or not to those in younger adults.

Clinical studies of ivacaftor in combination regimen with tezacaftor did not include patients over 75 years of age. The pharmacokinetic parameters of ivacaftor in combination with tezacaftor in the elderly patients (65-72 years) are comparable to those in younger adults.

Paediatric population

Predicted ivacaftor exposure based on observed ivacaftor concentrations in Phase 2 and 3 studies as determined using population PK analysis is presented by age group in Table 7.

Table 7: Mean (SD) ivacaftor exposure by age group

Age group

Dose

Cmin, ss (ng/mL)

AUC, ss (ng*h/mL)

6 months to less than 12 months (5 kg to < 7 kg) *

25 mg q12h

336

5410

6 months to less than 12 months (7 kg to < 14 kg)

50 mg q12h

508 (252)

9140 (4200)

12 months to less than 24 months (7 kg to < 14 kg)

50 mg q12h

440 (212)

9050 (3050)

12 months to less than 24 months (≥ 14 kg to < 25 kg)

75 mg q12h

451 (125)

9600 (1800)

2- to 5-year-olds (< 14 kg)

50 mg q12h

577 (317)

10500 (4260)

2- to 5-year-olds (≥ 14 kg to < 25 kg)

75 mg q12h

629 (296)

11300 (3820)

6- to 11-year-olds (≥ 14 kg to < 25 kg)

75 mg q12h

641 (329)

10760 (4470)

6- to 11-year-olds (≥ 25 kg)

150 mg q12h

958 (546)

15300 (7340)

12- to 17-year-olds

150 mg q12h

564 (242)

9240 (3420)

Adults (≥ 18 years old)

150 mg q12h

701 (317)

10700 (4100)

* Values based on data from a single patient; standard deviation not reported.

Exposures in 6- to 11-year-olds are predictions based on simulations from the population PK model using data obtained for this age group.

The pharmacokinetic parameters of ivacaftor in combination with tezacaftor in adolescent patients (12 to 17 year-olds) are comparable to those in adults (see Table 8).

Table 8: Mean (SD) of ivacaftor exposure when used in combination with tezacaftor, by age group

Age group

Dose

Ivacaftor Mean (SD)

Cmin, ss (ng/mL)

Ivacaftor Mean (SD)

AUC,ss (ng*h/mL)

12- to 17-year-olds

n = 97

tezacaftor 100 mg qd / ivacaftor 150 mg q12h

700 (413)

11400 (5500)

Adults (≥ 18 years old)

n = 389

tezacaftor 100 mg qd / ivacaftor 150 mg q12h

738 (318)

11400 (4140)


5.3. Preclinical safety data

Non-clinical data reveal no special hazard for humans based on conventional studies of safety pharmacology, repeated dose toxicity, genotoxicity, and carcinogenic potential.

Pregnancy and fertility

Ivacaftor was associated with slight decreases of the seminal vesicle weights, a decrease of overall fertility index and number of pregnancies in females mated with treated males and significant reductions in number of corpora lutea and implantation sites with subsequent reductions in the average litter size and average number of viable embryos per litter in treated females. The No-Observed-Adverse-Effect-Level (NOAEL) for fertility findings provides an exposure level of approximately 4 times the systemic exposure of ivacaftor and its metabolites when administered as ivacaftor monotherapy in adult humans at the maximum recommended human dose (MRHD). Placental transfer of ivacaftor was observed in pregnant rats and rabbits.

Peri- and post-natal development

Ivacaftor decreased survival and lactation indices and caused a reduction in pup body weights. The NOAEL for viability and growth in the offspring provides an exposure level of approximately 3 times the systemic exposure of ivacaftor and its metabolites when administered as ivacaftor monotherapy in adult humans at the MRHD.

Juvenile animal studies

Findings of cataracts were observed in juvenile rats dosed from postnatal day 7 through 35 at ivacaftor exposure levels of 0.22 times the MRHD based on systemic exposure of ivacaftor and its metabolites when administered as ivacaftor monotherapy. This finding has not been observed in foetuses derived from rat dams treated with ivacaftor on gestation days 7 to 17, in rat pups exposed to ivacaftor through milk ingestion up to postnatal day 20, in 7-week old rats, nor in 3.5 to 5-month old dogs treated with ivacaftor. The potential relevance of these findings in humans is unknown.


6.1. List of excipients

Tablet core

Cellulose, microcrystalline

Lactose monohydrate

Hypromellose acetate succinate

Croscarmellose sodium

Sodium laurilsulfate (E487)

Silica, colloidal anhydrous

Magnesium stearate

Tablet film coat

Polyvinyl alcohol

Titanium dioxide (E171)

Macrogol (PEG 3350)

Talc

Indigo carmine aluminium lake (E132)

Carnauba wax

Printing ink

Shellac

Iron oxide black (E172)

Propylene glycol (E1520)

Ammonia solution, concentrated


6.2. Incompatibilities

Not applicable.


6.3. Shelf life

4 years.


6.4. Special precautions for storage

This medicinal product does not require any special storage conditions.


6.5. Nature and contents of container

The film-coated tablets are packed in a thermoform (PolyChloroTriFluoroEthylene [PCTFE]/foil) blister or a High-Density PolyEthylene (HDPE) bottle with a polypropylene child-resistant closure, foil-lined induction seal and molecular sieve desiccant.

The following pack sizes are available:

- Blister card pack containing 28 film-coated tablets

- Blister pack containing 56 film-coated tablets

- Bottle containing 56 film-coated tablets

Not all pack sizes may be marketed


6.6. Special precautions for disposal and other handling

Any unused medicinal product or waste material should be disposed of in accordance with local requirements.


7. Marketing authorisation holder

Vertex Pharmaceuticals (Ireland) Limited

28-32 Pembroke Street Upper

Dublin 2, D02 EK84

Ireland


8. Marketing authorisation number(s)

EU/1/12/782/001

EU/1/12/782/002

EU/1/12/782/005


9. Date of first authorisation/renewal of the authorisation

Date of first authorisation: 23 July 2012

Date of latest renewal: 28 April 2017


10. Date of revision of the text

06/2020

Detailed information on this medicinal product is available on the website of the European Medicines Agency http://www.ema.europa.eu.

4.1 Therapeutic indications

Kalydeco tablets are indicated for the treatment of adults, adolescents, and children aged 6 years and older and weighing 25 kg or more with cystic fibrosis (CF) who have an R117H CFTR mutation or one of the following gating (class III) mutations in the CFTR gene: G551D, G1244E, G1349D, G178R, G551S, S1251N, S1255P, S549N or S549R (see sections 4.4 and 5.1).

Kalydeco tablets are also indicated in a combination regimen with tezacaftor 100 mg/ivacaftor 150 mg tablets for the treatment of adults and adolescents aged 12 years and older with cystic fibrosis (CF) who are homozygous for the F508del mutation or who are heterozygous for the F508del mutation and have one of the following mutations in the CFTR gene: P67L, R117C, L206W, R352Q, A455E, D579G, 711+3A→G, S945L, S977F, R1070W, D1152H, 2789+5G→A, 3272-26A→G, and 3849+10kbC→T.

4.2 Posology and method of administration

Kalydeco should only be prescribed by physicians with experience in the treatment of cystic fibrosis. If the patient's genotype is unknown, an accurate and validated genotyping method should be performed before starting treatment to confirm the presence of an indicated mutation in the CFTR gene (see section 4.1). The phase of the poly-T variant identified with the R117H mutation should be determined in accordance with local clinical recommendations.

Posology

Kalydeco monotherapy in adults, adolescents, and children aged 6 years and older and weighing 25 kg or more

The recommended dose is one Kalydeco 150 mg tablet taken orally every 12 hours (300 mg total daily dose) with fat-containing food (see Method of administration).

Kalydeco in a combination regimen with tezacaftor/ivacaftor in adults and adolescents aged 12 years and older

The recommended dose is one tezacaftor 100 mg/ivacaftor 150 mg tablet taken in the morning and one Kalydeco 150 mg tablet taken in the evening, approximately 12 hours apart with fat-containing food (see Method of administration).

Missed dose

If a dose is missed within 6 hours of the time it is usually taken, the patient should be told to take it as soon as possible and then take the next dose at the regularly scheduled time. If more than 6 hours have passed since the time the dose is usually taken, the patient should be told to wait until the next scheduled dose.

Patients receiving Kalydeco in combination with tezacaftor/ivacaftor should be told not to take more than one dose of either tablet at the same time.

Concomitant use of CYP3A inhibitors

Kalydeco monotherapy

When co-administered with strong inhibitors of CYP3A, the Kalydeco dose should be reduced to 150 mg twice a week (see sections 4.4 and 4.5).

When co-administered with moderate inhibitors of CYP3A, the Kalydeco dose should be reduced to 150 mg once daily (see sections 4.4 and 4.5).

Kalydeco in a combination regimen with tezacaftor/ivacaftor

When co-administered with strong inhibitors of CYP3A, the Kalydeco evening dose should not be administered (see sections 4.4 and 4.5). One tablet of tezacaftor 100 mg/ivacaftor 150 mg should be taken twice a week, taken approximately 3 to 4 days apart.

When co-administered with moderate inhibitors of CYP3A, the dose of Kalydeco and tezacaftor/ivacaftor should be adjusted according to the recommendations in Table 1 (see section 4.4 and 4.5).

Table 1: Dosing recommendations for concomitant use with moderate CYP3A inhibitors

Day 1

Day 2

Day 3

Day 4*

Morning Dose

Tezacaftor 100 mg/ ivacaftor 150 mg tablet

-

-

Ivacaftor 150 mg tablet

-

-

Evening Dose

Ivacaftor 150 mg tablet

-

-

-

-

* Continue dosing with tezacaftor 100 mg/ivacaftor 150 mg or ivacaftor 150 mg tablets on alternate days.

Special populations

Elderly

Very limited data are available for elderly patients treated with ivacaftor (administered as monotherapy or in a combination regimen with tezacaftor/ivacaftor). No dose adjustment is considered necessary unless moderate hepatic impairment exists. Caution is recommended for patients with severe renal impairment or end-stage renal disease (see section 5.2).

Renal impairment

No dose adjustment is necessary for patients with mild to moderate renal impairment. Caution is recommended in patients with severe renal impairment (creatinine clearance less than or equal to 30 mL/min) or end-stage renal disease (see sections 4.4 and 5.2).

Hepatic impairment

No dose adjustment is necessary for patients with mild hepatic impairment (Child-Pugh Class A). For patients with moderate hepatic impairment (Child-Pugh Class B), the dose should be reduced (see Table 2). There is no experience of the use of Kalydeco monotherapy or in combination with tezacaftor/ivacaftor in patients with severe hepatic impairment. Therefore, its use is not recommended unless the benefits outweigh the risks (see Table 2 and sections 4.4 and 5.2).

Table 2: Dosing recommendations for patients with hepatic impairment for Kalydeco monotherapy and in a combination regimen with tezacaftor/ivacaftor

Kalydeco monotherapy

Kalydeco in a combination regimen with tezacaftor/ivacaftor

Mild

(Child-Pugh Class A)

No dose adjustment

No dose adjustment

Moderate

(Child-Pugh Class B)

One tablet of ivacaftor 150 mg once daily

One tablet of tezacaftor 100 mg/ivacaftor 150 mg once daily

Severe

(Child-Pugh Class C)

Starting dose: One tablet of ivacaftor 150 mg every other day.

Dosing intervals should be modified according to clinical response and tolerability.

Starting dose: One tablet of tezacaftor 100 mg/ivacaftor 150 mg once daily.

Dosing intervals should be modified according to clinical response and tolerability.

Paediatric population

The safety and efficacy of Kalydeco monotherapy in children aged less than 6 months have not been established. No data are available.

An appropriate dose for children under 6 years of age and weighing less than 25 kg cannot be achieved with Kalydeco tablets.

Limited data are available in patients less than 6 years of age with an R117H mutation in the CFTR gene. Available data in patients aged 6 years and older are described in sections 4.8, 5.1 and 5.2.

The safety and efficacy of Kalydeco in a combination regimen with tezacaftor/ivacaftor in children aged less than 12 years have not been established. No data are available.

Method of administration

For oral use.

Patients should be instructed to swallow the tablets whole. The tablets should not be chewed, crushed, or broken before swallowing.

Kalydeco tablets should be taken with fat-containing food.

Food or drink containing grapefruit or Seville oranges should be avoided during treatment (see section 4.5).

4.3 Contraindications

Hypersensitivity to the active substance or to any of the excipients listed in section 6.1.

4.4 Special warnings and precautions for use

Only patients with CF who had a G551D, G1244E, G1349D, G178R, G551S, S1251N, S1255P, S549N, S549R gating (class III), G970R or R117H mutation in at least one allele of the CFTR gene were included in studies 1, 2, 5 and 6 (see section 5.1).

In study 5, four patients with the G970R mutation were included. In three of four patients the change in the sweat chloride test was < 5 mmol/L and this group did not demonstrate a clinically relevant improvement in FEV1 after 8 weeks of treatment. Clinical efficacy in patients with the G970R mutation of the CFTR gene could not be established (see section 5.1).

Efficacy results from a Phase 2 study in patients with CF who are homozygous for the F508del mutation in the CFTR gene showed no statistically significant difference in FEV1 over 16 weeks of ivacaftor treatment compared to placebo (see section 5.1). Therefore, use of Kalydeco monotherapy in these patients is not recommended.

Less evidence of a positive effect of ivacaftor has been shown for patients with an R117H-7T mutation associated with less severe disease in study 6 (see section 5.1).

Kalydeco in combination with tezacaftor/ivacaftor should not be prescribed in patients with CF who are heterozygous for the F508del mutation and have a second CFTR mutation not listed in section 4.1.

Effect on liver function tests

Moderate transaminase (alanine transaminase [ALT] or aspartate transaminase [AST]) elevations are common in subjects with CF. Transaminase elevations have been observed in some patients treated with ivacaftor monotherapy and in a combination regimen with tezacaftor/ivacaftor. Therefore, liver function tests are recommended for all patients prior to initiating ivacaftor, every 3 months during the first year of treatment and annually thereafter. For all patients with a history of transaminase elevations, more frequent monitoring of liver function tests should be considered. In the event of significant elevations of transaminases (e.g., patients with ALT or AST > 5 x the upper limit of normal (ULN), or ALT or AST > 3 x ULN with bilirubin > 2 x ULN), dosing should be interrupted and laboratory tests closely followed until the abnormalities resolve. Following resolution of transaminase elevations, the benefits and risks of resuming treatment should be considered (see section 4.8).

Hepatic impairment

Use of ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, is not recommended in patients with severe hepatic impairment unless the benefits are expected to outweigh the risks (see sections 4.2 and 5.2).

Renal impairment

Caution is recommended while using ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, in patients with severe renal impairment or end-stage renal disease (see sections 4.2 and 5.2).

Patients after organ transplantation

Ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor, has not been studied in patients with CF who have undergone organ transplantation. Therefore, use in transplanted patients is not recommended. See section 4.5 for interactions with ciclosporin or tacrolimus.

Interactions with medicinal products

CYP3A inducers

Exposure to ivacaftor may be reduced by the concomitant use of CYP3A inducers, potentially resulting in the loss of ivacaftor efficacy. Therefore, co-administration of Kalydeco (monotherapy or in combination with tezacaftor/ivacaftor) with strong CYP3A inducers is not recommended (see section 4.5).

CYP3A inhibitors

The dose of Kalydeco (monotherapy or in combination with tezacaftor/ivacaftor) must be adjusted when used concomitantly with strong or moderate CYP3A inhibitors (see sections 4.2 and 4.5).

Cataracts

Cases of non-congenital lens opacities without impact on vision have been reported in paediatric patients treated with ivacaftor, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor. Although other risk factors were present in some cases (such as corticosteroid use and exposure to radiation), a possible risk attributable to treatment cannot be excluded. Baseline and follow-up ophthalmological examinations are recommended in paediatric patients initiating ivacaftor treatment, either in monotherapy or in a combination regimen with tezacaftor/ivacaftor (see section 5.3).

Lactose content

Kalydeco contains lactose. Patients with rare hereditary problems of galactose intolerance, total lactase deficiency or glucose-galactose malabsorption should not take this medicine.

Sodium content

This medicine contains less than 1 mmol sodium (23 mg) per dose, that is to say essentially 'sodium-free'.

4.5 Interaction with other medicinal products and other forms of interaction

Ivacaftor is a substrate of CYP3A4 and CYP3A5. It is a weak inhibitor of CYP3A and P-gp and a potential inhibitor of CYP2C9. In vitro studies showed that ivacaftor is not a substrate for P-gp.

Medicinal products affecting the pharmacokinetics of ivacaftor

CYP3A inducers

Co-administration of ivacaftor with rifampicin, a strong CYP3A inducer, decreased ivacaftor exposure (AUC) by 89% and decreased hydroxymethyl ivacaftor (M1) to a lesser extent than ivacaftor. Co-administration of Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) with strong CYP3A inducers, such as rifampicin, rifabutin, phenobarbital, carbamazepine, phenytoin and St. John's wort (Hypericum perforatum), is not recommended (see section 4.4).

No dose adjustment is recommended when Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is used with moderate or weak CYP3A inducers.

CYP3A inhibitors

Ivacaftor is a sensitive CYP3A substrate. Co-administration with ketoconazole, a strong CYP3A inhibitor, increased ivacaftor exposure (measured as area under the curve [AUC]) by 8.5-fold and increased M1 to a lesser extent than ivacaftor. A reduction of the Kalydeco dose (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is recommended for co-administration with strong CYP3A inhibitors, such as ketoconazole, itraconazole, posaconazole, voriconazole, telithromycin and clarithromycin (see sections 4.2 and 4.4).

Co-administration with fluconazole, a moderate inhibitor of CYP3A, increased ivacaftor exposure by 3-fold and increased M1 to a lesser extent than ivacaftor. A reduction of the Kalydeco dose (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is recommended for patients taking concomitant moderate CYP3A inhibitors, such as fluconazole and erythromycin (see sections 4.2 and 4.4).

Co-administration of ivacaftor with grapefruit juice, which contains one or more components that moderately inhibit CYP3A, may increase exposure to ivacaftor. Food or drink containing grapefruit or Seville oranges should be avoided during treatment with Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor, see section 4.2).

Ciprofloxacin

Co-administration of ciprofloxacin with ivacaftor did not affect the exposure of ivacaftor. No dose adjustment is required when Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) is co-administered with ciprofloxacin.

Medicinal products affected by ivacaftor:

Administration of ivacaftor may increase systemic exposure of medicinal products that are sensitive substrates of CYP2C9, and/or P-gp, and/or CYP3A which may increase or prolong their therapeutic effect and adverse reactions.

CYP2C9 substrates

Ivacaftor may inhibit CYP2C9. Therefore, monitoring of the international normalised ratio (INR) is recommended during co-administration of warfarin with Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor). Other medicinal products for which exposure may be increased include glimepiride and glipizide; these medicinal products should be used with caution.

Digoxin and other P-gp substrates

Co-administration with digoxin, a sensitive P-gp substrate, increased digoxin exposure by 1.3-fold, consistent with weak inhibition of P-gp by ivacaftor. Administration of Kalydeco (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) may increase systemic exposure of medicinal products that are sensitive substrates of P-gp, which may increase or prolong their therapeutic effect and adverse reactions. When used concomitantly with digoxin or other substrates of P-gp with a narrow therapeutic index, such as ciclosporin, everolimus, sirolimus or tacrolimus, caution and appropriate monitoring should be used.

CYP3A substrates

Co-administration with (oral) midazolam, a sensitive CYP3A substrate, increased midazolam exposure 1.5-fold, consistent with weak inhibition of CYP3A by ivacaftor. No dose adjustment of CYP3A substrates, such as midazolam, alprazolam, diazepam or triazolam, is required when these are co-administered with ivacaftor (as monotherapy or in a combination regimen with tezacaftor/ivacaftor).

Hormonal contraceptives

Ivacaftor (as monotherapy or in a combination regimen with tezacaftor/ivacaftor) has been studied with an oestrogen/progesterone oral contraceptive and was found to have no significant effect on the exposures of the oral contraceptive. Therefore, no dose adjustment of oral contraceptives is necessary.

Potential for ivacaftor to interact with transporters

In vitro studies showed that ivacaftor is not a substrate for OATP1B1 or OATP1B3. Ivacaftor and its metabolites are substrates of BCRP in vitro. Due to its high intrinsic permeability and low likelihood of being excreted intact, co-administration of BCRP inhibitors is not expected to alter exposure of ivacaftor and M1-IVA, while any potential changes in M6-IVA exposures are not expected to be clinically relevant.

Paediatric population

Interaction studies have only been performed in adults.

4.6 Fertility, pregnancy and lactation

Pregnancy

There are no or limited amount of data (less than 300 pregnancy outcomes) from the use of ivacaftor in pregnant women. Animals studies do not indicate direct or indirect harmful effects with respect to reproductive toxicity (see section 5.3). As a precautionary measure, it is preferable to avoid the use of Kalydeco during pregnancy.

Breast-feeding

It is unknown whether ivacaftor and/or its metabolites are excreted in human milk. Available pharmacokinetic data in animals have shown excretion of ivacaftor into the milk of lactating female rats. As such, a risk to the newborns/infants cannot be excluded. A decision must be made whether to discontinue breast-feeding or to discontinue/abstain from Kalydeco therapy taking into account the benefit of breast-feeding for the child and the benefit of therapy for the woman.

Fertility

There are no data available on the effect of ivacaftor on fertility in humans. Ivacaftor had an effect on fertility in rats (see section 5.3).

4.7 Effects on ability to drive and use machines

Kalydeco has minor influence on the ability to drive or use machines. Ivacaftor may cause dizziness (see section 4.8) and, therefore, patients experiencing dizziness should be advised not to drive or use machines until symptoms abate.

4.8 Undesirable effects

Summary of the safety profile

The most common adverse reactions experienced by patients aged 6 years and older who received ivacaftor in the pooled 48-week placebo-controlled Phase 3 studies that occurred with an incidence of at least 3% and up to 9% higher than in the placebo arm were headache (23.9%), oropharyngeal pain (22.0%), upper respiratory tract infection (22.0%), nasal congestion (20.2%), abdominal pain (15.6%), nasopharyngitis (14.7%), diarrhoea (12.8%), dizziness (9.2%), rash (12.8%) and bacteria in sputum (12.8%). Transaminase elevations occurred in 12.8% of ivacaftor-treated patients versus 11.5% of placebo-treated patients.

In patients aged 2 to less than 6 years the most common adverse reactions were nasal congestion (26.5%), upper respiratory tract infection (23.5%), transaminase elevations (14.7%), rash (11.8%), and bacteria in sputum (11.8%).

Serious adverse reactions in patients who received ivacaftor included abdominal pain and transaminase elevations (see section 4.4).

Tabulated list of adverse reactions

Table 3 reflects the adverse reactions observed with ivacaftor monotherapy in clinical trials (placebo-controlled and uncontrolled studies) in which the length of exposure to ivacaftor ranged from 16 weeks to 144 weeks. Additional adverse reactions observed with ivacaftor in combination with tezacaftor/ivacaftor are also provided in Table 3. The frequency of adverse reactions is defined as follows: very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000); not known (cannot be estimated from the available data). Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness.

Table 3: Adverse reactions in patients treated with ivacaftor monotherapy or in combination with tezacaftor

System organ class

Adverse reactions

Frequency

Infections and infestations

Upper respiratory tract infection

very common

Nasopharyngitis

very common

Rhinitis

common

Nervous system disorders

Headache

very common

Dizziness

very common

Ear and labyrinth disorders

Ear pain

common

Ear discomfort

common

Tinnitus

common

Tympanic membrane hyperaemia

common

Vestibular disorder

common

Ear congestion

uncommon

Respiratory, thoracic and mediastinal disorders

Oropharyngeal pain

very common

Nasal congestion

very common

Sinus congestion

common

Pharyngeal erythema

common

Gastrointestinal disorders

Abdominal pain

very common

Diarrhoea

very common

Nausea *

common

Hepatobiliary disorders

Transaminase elevations

very common

Skin and subcutaneous tissue disorders

Rash

very common

Reproductive system and breast disorders

Breast mass

common

Breast inflammation

uncommon

Gynaecomastia

uncommon

Nipple disorder

uncommon

Nipple pain

uncommon

Investigations

Bacteria in sputum

very common

* Adverse reaction and frequency reported only in clinical studies with ivacaftor in combination with tezacaftor/ivacaftor.

Description of selected adverse reactions

Hepatobiliary disorders

Transaminase elevations

During the 48-week placebo-controlled studies 1 and 2 of ivacaftor monotherapy in patients aged 6 years and older, the incidence of maximum transaminase (ALT or AST) > 8, > 5 or > 3 x ULN was 3.7%, 3.7% and 8.3% in ivacaftor-treated patients and 1.0%, 1.9% and 8.7% in placebo-treated patients, respectively. Two patients, one on placebo and one on ivacaftor permanently discontinued treatment for elevated transaminases, each > 8 x ULN. No ivacaftor-treated patients experienced a transaminase elevation > 3 x ULN associated with elevated total bilirubin > 1.5 x ULN. In ivacaftor-treated patients, most transaminase elevations up to 5 x ULN resolved without treatment interruption. Ivacaftor dosing was interrupted in most patients with transaminase elevations > 5 x ULN. In all instances where dosing was interrupted for elevated transaminases and subsequently resumed, ivacaftor dosing was able to be resumed successfully (see section 4.4).

During the placebo controlled Phase 3 studies (up to 24 weeks) of ivacaftor in a combination regimen with tezacaftor/ivacaftor, the incidence of maximum transaminase (ALT or AST) > 8, > 5, or > 3 x ULN were similar between tezacaftor/ivacaftor in combination with ivacaftor and placebo-treated patients; 0.2%, 1.0%, and 3.4% in tezacaftor/ivacaftor in combination with ivacaftor-treated patients, and 0.4%, 1.0%, and 3.4% in placebo-treated patients. One patient (0.2%) on therapy and 2 patients (0.4%) on placebo permanently discontinued treatment for elevated transaminases. No patients treated with tezacaftor/ivacaftor discontinued treatment for elevated transaminases.

Paediatric population

The safety data of ivacaftor monotherapy were evaluated in 11 patients between 6 months to less than 12 months of age, 19 patients between 12 months to less than 24 months of age, 34 patients between 2 to less than 6 years of age, 61 patients between 6 to less than 12 years of age and 94 patients between 12 to less than 18 years of age. The safety data of ivacaftor in combination with tezacaftor/ivacaftor was evaluated in 98 patients between 12 to less than 18 years of age.

The safety profile is generally consistent among paediatric patients aged 6 months and older and adult patients.

During the 24-week open-label Phase 3 clinical study of ivacaftor monotherapy in 34 patients aged 2 to less than 6 years (study 7), 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 dosing with ivacaftor granules. Ivacaftor was permanently discontinued in one patient. In children aged 6 to less than 12 years, the incidence of patients experiencing transaminase elevations (ALT or AST) > 3 x ULN was 15.0% (6/40) in ivacaftor-treated patients and 14.6% (6/41) in patients who received placebo. A single ivacaftor-treated patient (2.5%) in this age range had an elevation of ALT and AST > 8 x ULN. Peak LFT (ALT or AST) elevations were generally higher in paediatric patients than in older patients. In almost all instances where dosing was interrupted for elevated transaminases and subsequently resumed, ivacaftor dosing was able to be resumed successfully (see section 4.4). Cases suggestive of positive rechallenge were observed.

During the 24-week, open-label, Phase 3 clinical study of ivacaftor in patients aged less than 24 months (study 8), the incidence of patients experiencing transaminase elevations (ALT or AST) > 3, > 5, and > 8 x ULN in the cohort of patients aged 12 months to less than 24 months was 27.8% (5/18), 11.1% (2/18) and 11.1% (2/18), respectively. In the cohort of patients aged 6 months to less than 12 months one patient (9.1%) had elevated ALT of > 3 to ≤5 x ULN. No patients had elevations in total bilirubin or discontinued ivacaftor treatment due to transaminase elevations in either cohort. The two patients with elevations of ALT or AST > 8 x ULN interrupted treatment and subsequently resumed ivacaftor successfully (see section 4.4 for management of elevated transaminases).

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via the

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard or search for MHRA Yellow Card in the Google Play or Apple App Store

Learning Zones

The Learning Zones are an educational resource for healthcare professionals that provide medical information on the epidemiology, pathophysiology and burden of disease, as well as diagnostic techniques and treatment regimens.

 

 

Disclaimer

The drug SPC information (indications, contra-indications, interactions, etc), has been developed in collaboration with eMC (www.medicines.org.uk/emc/). Medthority offers the whole library of SPC documents from eMC.

Medthority will not be held liable for explicit or implicit errors, or missing data.

Reporting of suspected adverse reactions 

Drug Licencing

Drugs appearing in this section are approved by UK Medicines & Healthcare Products Regulatory Agency (MHRA), & the European Medicines Agency (EMA).