Tezacaftor

Tezacaftor for the treatment of cystic fibrosis

Marc Sala & Manu Jain

To cite this article: Marc Sala & Manu Jain (2018): Tezacaftor for the treatment of cystic fibrosis, Expert Review of Respiratory Medicine, DOI: 10.1080/17476348.2018.1507741
To link to this article: https://doi.org/10.1080/17476348.2018.1507741

Accepted author version posted online: 03 Aug 2018.

Submit your article to this journal

Article views: 2

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=ierx20

Drug Profile
Tezacaftor for the treatment of cystic fibrosis

Marc Sala and Manu Jain*

Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA

*Corresponding author:

Manu Jain

Email: [email protected]

Abstract
Introduction: Cystic fibrosis (CF) is the most common, life-limiting autosomal recessive disease in Caucasians, and is caused by defects in production of the CFTR ion channel. Until recently, there were no available treatments targeting the disease-causing defects in CFTR but newly developed CFTR modulators are changing the course of disease in CF. The newest modulator, tezacaftor, is a CFTR corrector that was recently approved by the FDA to be used in combination with the first approved CFTR potentiator, ivacaftor.
Areas Covered: A detailed review of the clinical trials and published literature, focusing on safety and efficacy, leading to the approval of tezacaftor in cystic fibrosis.
Expert commentary: Recent trials have demonstrated that the combination of tezacaftor- ivacaftor is a slightly superior combination to its predecessor, lumacaftor-ivacaftor, with respect to an increase in FEV1, adverse event profile and drug-drug interactions. It is also approved for a large number of non-F508del, residual function mutations that are predicted to respond based on in vitro testing. The horizon for continued improvements in CFTR-targeted treatments is promising, with three-drug combinations currently in Phase 3 clinical trials, and other drugs with novel mechanisms of action being studied. Within the next 5 years, the vast majority of patients with CF are expected to have a modulator approved for their genotype.

Keywords: cystic fibrosis, tezacaftor, CFTR, ivacaftor

⦁ Introduction:
Cystic fibrosis (CF) is a genetic disease caused by absent or dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) protein [1]. The gene encoding CFTR was discovered in 1989 by two independent laboratories [2-4]. CFTR is expressed predominantly at the epithelial apical cell membrane where it primarily functions as a cAMP-dependent ion channel for chloride and bicarbonate movement, but also acts as a regulator of other proteins involved in ion transport, such as the epithelial sodium channel, ENaC, and alternative chloride channels. Disruption of the production, transport, or function of CFTR leads to inadequate chloride and bicarbonate movement, which in turn results in impaired chloride reabsorption and elevated chloride levels in sweat, the measurement of which is a common diagnostic test for cystic fibrosis [5]. In the airway epithelium, inadequate chloride transport leads to a cycle of viscous mucous secretions, chronic airway infection, and an exuberant inflammatory response that results in bronchiectasis that often progresses to respiratory failure [6]. While cystic fibrosis manifests in multiple organ systems, lung disease accounts for the majority of the morbidity and mortality in CF [1].

Over 2000 CF-causing mutations in the CFTR gene have been described [201]. Traditionally, CFTR mutations are grouped into six classes depending on the specific type of defect in CFTR protein synthesis, transport, or function [1], although alternative schemes have been proposed [7,8]. Class I mutations involve a premature stop codon, leading to little or no normal CFTR mRNA synthesis, and a near complete absence of CFTR protein at the cell membrane. These are primarily nonsense mutations which create a premature stop codon in the CFTR gene, but also include splicing abnormalities leading to severely reduced protein production.

Class II mutations result in impaired folding of CFTR protein, leading to premature degradation by the ubiquitin-proteasome system and increased proteostatic stress [1,9-11]. As a result, little or no CFTR is trafficked to the cell membrane. This class includes the Phe508del (F508del) mutation, which is the most common CF mutation worldwide. F508del is present in at least

one allele in over 85% of CF patients in the United States (US), and nearly 50% of US CF patients are homozygous for this mutation [201].

Class III mutations typically result in amino acid substitutions that produce CFTR protein with a “gating” defect (causing a low probability of channel opening) in the plasma membrane.[1] The dysfunctional CFTR protein is produced and trafficked to the cell membrane normally, but chloride transport is severely impaired. The most common class III mutation, Gly551Asp (G551D) is present in approximately 4% of CF patients in the US [201].

Class IV CFTR mutations cause intrinsic channel dysfunction and a reduction in ion transport less severe than that in class III mutations [1]. Relatively normal amounts of CFTR protein are found at the cell surface. Arg117His (R117H) is the most common class IV mutation (though it also leads to gating defects), and is present in approximately 2.8% of CF patients in the US [12].

Class V mutations result in attenuated synthesis or maturation of the CFTR protein resulting in reduced amounts of normally-functioning CFTR at the cell membrane, often due to RNA splicing defects [1]. The most common mutation from this class is 3849+10kb CT [12].

Class VI mutations destabilize CFTR at the cell surface (via increased endocytosis or decreased recycling back to the cell surface). This class is represented by mutations such as c.120del23 and rescued F508del (rF508del).

It should be noted that one mutation may fall into multiple classes. Examples include the F508del (classically class II, but also III) and R117H (classically class IV, but also III) mutations.

Historically, most CF therapies have targeted lung disease and other end organ manifestations in an attempt to disrupt the cycle of mucous obstruction, inflammation, and infection.
However, in the past decade the focus of emerging therapies has shifted toward discovering drugs that target the underlying defect in CF, collectively referred to as CFTR modulators.

⦁ Overview of the Market
There are currently two broad classes of FDA-approved CFTR modulators, categorized by their molecular mechanism of action: potentiators and correctors. Potentiators bind to CFTR protein in the plasma membrane to increase the open probability of the CFTR channel, thereby increasing ion conductance [13]. Ivacaftor, the only approved CFTR potentiator, was first approved for patients with the G551D mutation. Ivacaftor has been clinically shown to improve FEV1, decrease sweat chloride, improve quality of life metrics (CFQ-R score), and increase weight in patients with at least one copy of G551D [14]. Follow-up studies suggest that ivacaftor also attenuates FEV1 decline by approximately 50% [15]. Subsequent clinical trials showed similar efficacy in patients 6-11 years old with one copy of G551D [16], additional non- G551D gating mutations [17], and the R117H mutation (a class III and IV mutation)[18]. Based on these clinical data, the FDA expanded the indications for ivacaftor to include non-G551D gating mutations. The FDA also approved ivacaftor for several residual function mutations based on Fischer rat thyroid cell in vitro data alone [19], opening the door to a mechanism of approval of modulators for rarer CFTR mutations which could not enroll for a formal clinical trial [20]. Currently, roughly 10% of the US CF population has a mutation that qualifies for ivacaftor [12].

CFTR correctors target the protein folding defect that results from the most prevalent CF- causing mutation, F508del [21]. Until recently, lumacaftor was the only clinically approved corrector. The addition of ivacaftor to lumacaftor as a combination therapy addresses both the folding and gating defects of F508del protein and increases conductance more than each compound singly. The combination was shown to improve ppFEV1 (2.8% above placebo) and reduce pulmonary exacerbation frequency for F508del homozygotes in two concurrent Phase 3 studies [22]. A Phase 3 extension study following TRAFFIC/TRANSPORT (i.e., PROGRESS) demonstrated that lumacaftor-ivacaftor had sustained benefits in ppFEV1 and pulmonary exacerbation rate, increased BMI, and was associated with an estimated a 42% reduction in annual rate of lung function decline compared to matched registry control group [23]. Of concern, however, Orkambi was reported to cause dyspnea/chest tightness or bronchospasm

at a higher frequency than placebo in these Phase 3 trials, where the enrollment criteria included a percent predicted FEV1 (ppFEV1) between 40-90%. Subsequent subgroup analyses
[24] and additional observational studies [25-28] have confirmed this effect and further reported that the incidence and severity of bronchospasm to be higher in patients with ppFEV1<40%.

Drug-drug interactions must be monitored closely when taking ivacaftor due to its weak CYP3A4 and P-glycoprotein (P-gp) inhibition, increasing the exposure to medications that are substrates for CYP3A4 or P-gp, including benzodiazepines, cyclosporine, and tacrolimus [29]. Ivacaftor itself is a substrate of CYP3A4, and therefore requires dose adjustments when co- administered with CYP3A4 inhibitors (e.g., the “azole” antifungals, grapefruit, or Seville oranges) or inducers (e.g., rifampin). In addition to being a substrate of CYP3A4 like ivacaftor, lumacaftor also acts on several CYP450 enzymes, including a strong net induction of CYP3A4. When taking the lumacaftor-ivacaftor combination, there is likely to be reduced concentrations of corticosteroids, the “azole” antifungals, midazolam, cyclosporine, tacrolimus, everolimus, and sirolimus, all of which are CYP3A4 substrates [30]. This has particular relevance to patients with CF who have undergone solid organ transplants and require therapeutic levels of immunosuppression with these agents to prevent graft rejection.

Taken together, these data demonstrate that while lumacaftor-ivacaftor combination was a significant advance for F508del homozygous patients, there remains an unmet need for a better-tolerated, small molecule CFTR corrector/potentiator combination with fewer drug-drug interactions, which is particularly relevant in the CF population that must take multiple other medications.

⦁ Tezacaftor 2.1: Chemistry
CFTR is a 1480-amino acid ATP-binding cassette transporter glycoprotein that consists of two membrane spaning domains (MSD1 and MSD2), two cytoplasmic nucleotide-binding domains

(NBD1 and NBD2), and a regulatory (R) domain [6]. The F508del mutation in NBD1 leads to accumulation of F508del-CFTR intermediates that are degraded by the proteasome. Tezacaftor is structurally related to lumacaftor (Fig 1) [31]. These compounds were identified by using the method of high-throughput screening (HTS) in Fischer rat thyroid (FRT) epithelial cells co- expressing F508del-CFTR and a yellow fluorescent protein (YFP) halide sensor [32]. Compounds which increased iodide influx (and quenched the YFP sensor) through F508del-CFTR in the presence of extracellular iodide and the CFTR potentiators forskolin and genistein were considered “hits.” In the case of lumacaftor, experiments indicate that this small molecule binds to F508del-CFTR directly in order to modulate the conformation of MSD1 and thereby partially mitigate the folding defects caused by the F508del mutation in NBD1 [33,34]. Whether the molecular mechanism of action for tezacaftor differs significantly from that lumacaftor has not been well documented.

⦁ : Mechanism of action
Tezacaftor is a novel CFTR corrector, structurally similar to lumacaftor, which improves processing and trafficking of F508del-CFTR and increases chloride transport from 2.5% to 8.1% of normal levels in human bronchial epithelial (HBE) cells derived from F508del/F508del donors. Chloride transport was further enhanced by co-treatment with ivacaftor to 15.7% of normal.
Additional effects of combination treatment included improved ciliary beat frequency and fluid transport. Chloride transport was also assayed, and found to be improved, in a panel of additional CFTR mutant forms, including those associated with gating defects and residual CFTR function [35].

⦁ : Pharmacokinetics/pharmacodynamics: The pharmacokinetics of oral tezacaftor monotherapy and tezacaftor-ivacaftor have been evaluated in a Phase 2 trial [36]. These data showed that oral tezacaftor was rapidly absorbed and reached steady state by approximately two weeks. Tezacaftor at 50 mg twice daily had a similar AUC at steady state as compared to 100 mg once daily dosing. Mean AUC ratios and Cmax ratios for tezacaftor-ivacaftor relative to tezacaftor were 0.998 and 1.07, respectively. Exposures of tezacaftor and its metabolites were

dose-proportional and similar between tezacaftor monotherapy and tezacaftor-ivacaftor groups. The recommended dose of the tezacaftor-ivacaftor combination is one tablet (tezacaftor 100 mg/ivacaftor 150 mg) taken in the morning and one tablet (ivacaftor 150 mg) taken in the evening, approximately 12 hours apart. At this FDA-approved dosing, the tezacaftor Cmax is 5.95 mcg/mL, T1/2 15 hrs, and AUC0-24h 84.5 mcg*h/mL. Exposure estimates of tezacaftor were similar between F508del homozygotes and F508del/G551D compound heterozygotes. Fat-containing foods had no effect on tezacaftor exposure but did increase ivacaftor exposure by 3 times compared to a fasting state [37].

⦁ : Drug-drug interactions
Unlike lumacaftor, tezacaftor is not an inducer of CYP3A4 enzymes (unpublished data per Donaldson et al study). However, tezacaftor is a substrate for metabolism by the CYP3A4 system (like ivacaftor and lumacaftor as delineated above), where CYP3A4 inducers may decrease exposure to tezacaftor and CYP3A4 inhibitors may increase tezacaftor exposure. Tezacaftor-ivacaftor has been studied with hormonal oral contraceptive ethinyl estradiol/norethindrone and no significant effects on the exposure to the contraceptive was observed [37]. This is in contrast to lumacaftor, which may decrease the efficacy of oral contraceptives, the mechanism of which is unknown, however, it is noted that estrogen is hydroxylated to inactive metabolites by hepatic CYP3A4 and that many antiepileptic drugs (e.g., carbamazepine, phenytoin, phenobarbital) – also inducers of CYP3A4 – are associated with contraceptive failure [38].

⦁ : Clinical efficacy
⦁ : Phase 2 trial
A Phase 2 trial was conducted to compare the efficacy and safety of tezacaftor-ivacaftor, tezacaftor alone, or placebo, in F508del homozygotes or F508del/G551D compound heterozygotes [36]. This Phase 2 trial was a randomized, placebo-controlled design using escalating doses of tezacaftor monotherapy before testing in combination with ivacaftor. The trial applied 28-day treatment, 28-day washout periods and all subjects were required to have a

confirmed diagnosis of CF, an FEV1 between 40-90% predicted, and a BMI of at least 18.5 kg/m2. F508del homozygotes were 18 years or older and compound heterozygotes were 12 years or older. The compound heterozygotes patient had to have been taking ivacaftor for at least 28 days prior to study enrollment. The study design included 14 arms, where F508del homozygotes received tezacaftor as monotherapy (10 mg – 150 mg once daily), in combination with ivacaftor (150 mg twice daily), or placebo. Compound heterozygotes received tezacaftor 100 mg daily or placebo in addition to their previously prescribed ivacaftor 150 mg twice daily. Primary endpoints included safety metrics through Day 56 (e.g., adverse events, laboratory values, EKGs); secondary endpoints included change in ppFEV1 from baseline through day 28, pharmacokinetic parameters, and change in Cystic Fibrosis Questionnaire-Revised (CFQ-R) score. 185 of 190 subjects (97.4%) completed the study and follow-up. Adverse events reported were similar across the study arms, the majority (81.4%) being mild-moderate in nature. Serious adverse events occurred in pooled tezacaftor monotherapy, pooled tezacaftor- ivacaftor combination therapy, and pooled placebo at 6.1%, 7.5%, and 15.2%, respectively.
Discontinuations occurred in these same groups at a frequency of 3.0%, 2.8%, and 0%, respectively. In the dose escalation phase for F508del homozygotes, the largest within-group improvement in absolute ppFEV1 was observed in the tezacaftor (100 mg daily)/ivacaftor (150 mg twice daily) arm and was 3.75%. Sweat chloride decreased by 6.04 mmol/L. Meanwhile, in subjects with the F508del/G551D genotype, the addition of tezacaftor 100 mg daily or placebo to physician prescribed ivacaftor 150 mg twice daily resulted in an absolute within-group increase in ppFEV1 of 4.60% and a sweat chloride decrease of 7.02 mmol/L.

⦁ : Phase 3 trials
In follow-up to the Phase II trial [36], tezacaftor-ivacaftor was tested in two concomitantly conducted Phase 3 trials to test the efficacy of tezacaftor-ivacaftor in F508del homozygote CF patients as well as compound heterozygote patients with one F508del allele and the other allele with a “residual function” (RF) mutation (the EVOLVE and EXPAND trails, respectively) [39,40].

The EVOLVE trial [40] was a double-blind, randomized, placebo-controlled Phase III trial comparing the therapeutic effects of combination tezacaftor-ivacaftor (100 mg tezacaftor once daily + 150 mg ivacaftor twice daily) versus placebo in F508del homozygotes. The study involved 91 international sites. Eligibility criteria included CF patients genetically confirmed to be F508del homozygotes, age 12 or older, and a ppFEV1 between 40-90%. The primary endpoint was absolute change in ppFEV1 from baseline to 24 weeks. Secondary endpoints included relative change in ppFEV1 at 24 weeks, number of pulmonary exacerbations, change in BMI, change in sweat chloride level, and change in CFQ-R symptom score. 509 enrolled patients received at least one dose of drug or placebo, of whom 504 patients were included in the efficacy analysis. Mean ppFEV1 at baseline was 60% and the primary endpoint showed a difference of 4.0% in ppFEV1 in the tezacaftor-ivacaftor group compared to placebo (3.4% in the tezacaftor-ivacaftor group versus -0.6% in the placebo group) at 24 weeks. Subjects receiving tezacaftor-ivacaftor had a lower pulmonary exacerbation rate (0.64 vs. 0.99 events per year). The CFQ-R scores favored tezacaftor-ivacaftor (mean difference of 5.1 points).
Sweat chloride levels were reduced in the tezacaftor-ivacaftor group compared to placebo (between-group difference -10.1 mmol/L). There was no significant difference in absolute increase in BMI between treatment and placebo groups at 24 weeks (0.18 vs 0.12, P = 0.41).

The EXPAND trial [39] was a double-blind, randomized, placebo-controlled Phase 3 trial with three arms: 1) combination tezacaftor 100 mg once daily with ivacaftor 150 mg twice daily; 2) monotherapy with ivacaftor 150 mg twice daily; and 3) placebo. Each patient received two of the three regimens over two 8 week intervention periods, each separated by a washout period of 8 weeks. The study involved 86 international sites. Eligible CF patients included genetically- confirmed F508del status on one allele and a residual function mutation on the other CFTR allele, age 12 or older, a ppFEV1 between 40-90%, and a sweat chloride of at least 60 mmol/L or documented evidence of chronic sinopulmonary disease. The residual function mutations included in this study are represented in Table 1, and included class IV noncanonical splice mutations and Class II-IV missense mutations. Similar to the EVOLVE trial, the primary endpoint was absolute change in the ppFEV1 from baseline to an average of week 4 and week 8 of each

intervention. Also similar to the EVOLVE trial, secondary endpoints included relative change in the ppFEV1, absolute change in sweat chloride level, and change in CFQ-R score, all from baseline to an average of week 4 and week 8 of each intervention. Exploratory endpoints included rate of pulmonary exacerbations, change in fecal elastase-1 level, change in trypsinogen level, and change in BMI. 248 patients were enrolled and 234 completed both intervention periods. The result of the primary endpoint with either tezacaftor-ivacaftor (N = 161) or ivacaftor alone (N = 156) was a significant increase in ppFEV1 compared to placebo (N = 161) (6.8% for tezacaftor-ivacaftor and 4.7% for ivacaftor). The difference between tezacaftor- ivacaftor and ivacaftor was statistically significant. Benefits were rapid (observed by day 15) and durable up to week 8 of the intervention, and were observed irrespective of the class of residual function mutation. The change in CFQ-R secondary endpoint favored both tezacaftor- ivacaftor and ivacaftor compared to placebo (+11.1 points and +9.7 points, respectively), but was not statistically different between tezacaftor-ivacaftor and ivacaftor alone. Similarly, these treatment groups both showed benefit compared to placebo in terms of relative change in ppFEV1 and sweat chloride level (-9.5 mmol/L for tezacaftor-ivacaftor and -4.5 mmol/L for ivacaftor). Change in trypsinogen, pulmonary exacerbations, and BMI appeared to favor treatment groups, but these endpoints were not reported with respect to statistical significance versus placebo.

For patients with CF who have one copy of F508del and the other allele with a minimal function (MF) mutation (i.e., mutation classes I and II), a Phase 3 study (NCT02516410) found there was no difference in ppFEV1 after 12 weeks of treatment with tezacaftor-ivacaftor compared to placebo. The study was terminated early based on pre-determined futility criteria [41].

Finally, a fourth Phase 3 study (NCT02412111) was performed in patients with CF who have one copy of F508del and one copy of a gating mutation (e.g., G551D). In this study tezacaftor was added to ivacaftor that had been started previously clinically. The results were divergent from the phase 2 study, in that this population of patients did not meet its primary endpoint. The absolute change in ppFEV1 at 8 weeks with the addition of tezacaftor was a 0.5% increase,

compared to placebo which was a 0.2% increase (p = 0.5846). Sweat chloride did decrease by
5.8 mmol/L in the dual therapy group compared to ivacaftor alone. There was no difference in CFQ-R scores with the addition of tezacaftor [42].

The first interim analysis for the tezacaftor-ivacaftor Phase 3, 96-week open label extension study (EXTEND, NCT02565914) were recently reported [43]. This showed a durable benefits at
≥48 weeks in absolute change from baseline in ppFEV1 and CFQ-R among subjects who received treatment drug in EVOLVE and EXPAND. There were no new safety signals. Compared to baseline, there was also a decreased estimated annual pulmonary exacerbation rate among F508del homozygotes and F508del/RF subjects in EXTEND.

⦁ Regulatory Affairs
The efficacy and safety data from EVOLVE and EXPAND led to FDA-approval of tezacaftor- ivacaftor (Symdeko®) for people ages 12 or older with CF who are either F508del homozygotes or who have a single residual function mutation (without consideration of the other mutation) as indicated in Tables 1 and 2. This is in contrast with the clinical trials where efficacy was demonstrated in individuals who were F508del/RF compound heterozygotes. A Phase 3 trial is ongoing to evaluate the safety and efficacy of tezacaftor-ivacaftor in children age 6-11 (NCT02953314).

⦁ : Safety and tolerability
In the Phase III EVOLVE trial, 92.7% of patients reported at least one adverse event (90.4% in the treatment group, 95.0% in the placebo group), most of which were mild or moderate severity events. The tezacaftor-ivacaftor group had fewer serious adverse events (12.4% tezacaftor-ivacaftor versus 18.2% placebo) or discontinuations of the trial regimen due to adverse events (2.8% tezacaftor-ivacaftor versus 3.1% placebo). Importantly, the incidence of respiratory adverse events was lower and occurred later in the tezacaftor-ivacaftor group compared to placebo (13.1% versus 15.9%, respectively), including upon acute initiation of drug and irrespective of baseline ppFEV1. This was in contrast to what was observed in the

lumacaftor-ivacaftor phase 3 trials (TRAFFIC/TRANSPORT), where although the rate of serious adverse events was similar between the lumacaftor-ivacaftor and placebo arms (17.3% and 28.6% respectively), the rate of discontinuation was higher in the treatment group (4.2% lumacaftor-ivacaftor versus 1.6% placebo) and respiratory adverse effects occurred more frequently with lumacaftor-ivacaftor than placebo. These included chest tightness (8.7% versus 5.9% in placebo) and dyspnea (13.0% versus 7.8% in placebo).

Meanwhile, in the Phase III EXPAND trial, the tezacaftor-ivacaftor group had fewer serious adverse events than the ivacaftor or placebo groups (2%, 5%, and 9%, respectively) or discontinuations of the trial regimen due to adverse events (0%, 1%, and <1%, respectively). As in the EVOLVE trial, and again in contrast to lumacaftor-ivacaftor, the incidence of respiratory adverse events was no greater in the tezacaftor-ivacaftor group than in the placebo group (6.8% versus 9.9% respectively), including upon acute initiation of drug.

⦁ Conclusion
Tezacaftor-ivacaftor is an effective, well-tolerated treatment for cystic fibrosis patients who are F508del homozygotes or have a subset of qualifying residual function mutations. Compared to Lumacaftor-ivacaftor, it offers a similar efficacy profile but better tolerability and fewer drug- drug interactions. In addition it is approved for an additional 22 residual function mutations.
The most convincing endpoints include an increased ppFEV1, improved symptoms (CFR-Q score), and decreased sweat chloride. Long term data on efficacy and safety are still being accumulated and analyzed.

⦁ Expert Commentary and Five Year Review:
Based on the Phase 2 and Phase 3 data, tezacaftor-ivacaftor appears to have notable advantages over lumacaftor-ivacaftor: 1) improved side effect profile; 2) fewer drug-drug interactions; 3) expanded indications (i.e., it is approved for either F508del homozygotes or for those with qualifying residual function mutations).

Most patients with an F508del homozygous genotype who qualify for lumacaftor-ivacaftor are already taking it, raising the question whether to switch to tezacaftor-ivacaftor. While there are no data to guide this decision, in our practice, we integrate several factors: 1) did the patient have a significant objective response to lumacaftor-ivacaftor (durable increase in ppFEV1 or BMI, or fewer exacerbations in the past 12 months)?; 2) are there drug-drug interactions with lumacaftor-ivacaftor that could be avoided by using tezacaftor (especially among patients who are prescribed the “azole” antifungals)?; and 3) patient preference (some patients are eager to try new drugs while others would like to await further safety data before switching therapies).

In the next five years, we are almost certainly going to see the approval of improved modulators (or combinations of modulators), both from Vertex and other pharmaceutical companies, in an effort to fill the unmet needs in the CF patient community.

For example, there are upcoming Phase 3 trials for “triple combination” CFTR modulators (two correctors and one potentiator) to serve two groups of patients: F508del homozygotes and those with F508del and a second allele with a minimal function mutation, the latter group having no approved modulator therapy available. Two triple combinations are currently being studied. VX-659 in combination with tezacaftor and ivacaftor, and VX-445 in combination with tezacaftor and ivacaftor. This certainly suggests that most patients on lumacaftor-ivacaftor combination will eventually stop and switch to tezacaftor-ivacaftor now or the “triple” if and when it becomes available.

The VX-659 triple combination Phase 3 trial is currently recruiting F508del homozygotes and F508del/MF compound heterozygotes (NCT03447249, ECLIPSE F/F and ECLIPSE F/MF) [44].
The Phase 2 trials showed an absolute improvement in ppFEV1 of 9.7% and a decrease in sweat chloride by 42.2 mmol/L in F508del homozygotes when VX-659 was added to tezacaftor- ivacaftor [44]. There was an absolute improvement in ppFEV1 of 13.3% and a decrease in sweat chloride by 51.4 mmol/L in the F508del/MF cohort compared to placebo [45]. The

overall safety profile in both Phase 2 trials was favorable, with no serious adverse events and no discontinuations or interruptions due to adverse events in the F508del homozygote arm. In the F508del/MF arm, there were no treatment-related serious adverse events, but one patient had treatment interrupted due to rash.

Analogous to the VX-659 program, the VX-445 triple combination will begin recruiting in the summer of 2018 for Phase 3 clinical trial in F508del homozygotes and F508del/MF compound heterozygotes (AURORA F/F and AURORA F/MF) [46]. The Phase 2 trials (NCT03227471) evaluated VX-445 (dose range 50-200 mg) in combination with tezacaftor-ivacaftor in F508del homozygotes and F508del/MF. The triple combination achieved an increase in ppFEV1 of 11.0 and a decrease in sweat chloride by 39.6 mmol/L in the F508del homozygote group [46], and an increase in ppFEV1 of 13.8 and a decrease in sweat chloride by 39.1 mmol/L in the F508del/MF group [45]. The 445 triple combination was generally well-tolerated. In the F508del homozygote triple combination arm, no serious adverse events were noted, but discontinuation or interruption due to chest pain (1 subject), increased bilirubin (1 subject), and myopathy with increased CPK, ALT, AST (1 subject) were reported. In the F508del/MF triple combination arm, there were no reported treatment-related serious AEs, however, treatment was discontinued or interrupted due to rash (1 subject), constipation (1 subject), and increased bilirubin (2 subjects).

In December 2018, another pharmaceutical company, Proteostasis Therapeutics, announced positive Phase 2 trials results using a novel CFTR modulator (referred to as a CFTR amplifier, which increases mRNA throughput), PTI-428, which increased absolute ppFEV1 among patients already taking Orkambi by 5.2% [47].

Key issues:
⦁ The combination regimen of tezacaftor-ivacaftor is FDA approved for CF patients who are F508del homozygotes or have a qualifying residual function mutation (Tables 1 and 2).

⦁ In F508del homozygotes, tezacaftor-ivacaftor improves ppFEV1, sweat chloride, pulmonary exacerbation frequency, and CFQ-R scores. In CF patients with residual function mutations, tezacaftor-ivacaftor improves ppFEV1, sweat chloride, and CFQ-R scores.
⦁ Tezacaftor-ivacaftor is generally well tolerated from a safety standpoint, but liver function tests should be monitored as they are with ivacaftor monotherapy. Pediatric patients should also be monitored for cataracts, similar to the recommendation for ivacaftor.
⦁ For F508del homozygotes, tezacaftor has several advantages over lumacaftor:
⦁ Fewer drug-drug interactions, owing to absence of CYP3A4 induction
⦁ An improved respiratory side effects profile
⦁ The CFTR drug development pipeline includes “triple combination” regimens by adding a novel corrector to tezacaftor-ivacaftor to achieve greater improvements in ppFEV1 for F508del homozygotes and to provide access to modulator therapy for individuals with non-F508del minimal function mutations.
⦁ CFTR amplifiers are a novel class of modulators which increase mRNA throughput and may offer another therapeutic option for CF patients in addition to correctors and potentiators.

Information Resources:
⦁ Clinical trial details and enrollment status are available at https://clinicaltrials.gov.
⦁ More information on cystic fibrosis epidemiology and care guidelines can be found at https://cff.org.
⦁ Press releases related to tezacaftor-ivacaftor or other Vertex trial results can be found at https://investors.vrtx.com.

Funding
This paper was not funded.

Declaration of interest
M Jain is a consultant for Vertex and Proteostasis. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures
A reviewer on this manuscript has disclosed that they have received grants and honoraria from Vertex Pharmaceuticals, as well as payment from Proteostasis for being part of an advisory board. Peer reviewers on this manuscript have no other relevant financial or other relationships to disclose.

References
Papers of special note have been highlighted as of interest (*) to readers

⦁ O'Sullivan BP, Freedman SD. Cystic fibrosis. Lancet, 373(9678), 1891-1904 (2009).
*Broad overview of cystic fibrosis disease epidemiology, disease manifestations, and traditinoal mutation classification scheme for reader unfamiliar with CF.
⦁ Riordan JR, Rommens JM, Kerem B et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science, 245(4922), 1066-1073 (1989).
⦁ Rommens JM, Iannuzzi MC, Kerem B et al. Identification of the cystic fibrosis gene: chromosome walking and jumping. Science, 245(4922), 1059-1065 (1989).
⦁ Kerem B, Rommens JM, Buchanan JA et al. Identification of the cystic fibrosis gene: genetic analysis. Science, 245(4922), 1073-1080 (1989).
⦁ Collie JT, Massie RJ, Jones OA, LeGrys VA, Greaves RF. Sixty-five years since the New York heat wave: advances in sweat testing for cystic fibrosis. Pediatric pulmonology, 49(2), 106-117 (2014).
⦁ Rowe SM, Miller S, Sorscher EJ. Cystic fibrosis. N Engl J Med, 352(19), 1992-2001 (2005).
⦁ De Boeck K, Amaral MD. Progress in therapies for cystic fibrosis. Lancet Respir Med, 4(8), 662-674 (2016).
⦁ Marson FAL, Bertuzzo CS, Ribeiro JD. Classification of CFTR mutation classes. Lancet Respir Med, 4(8), e37-e38 (2016).
⦁ Lukacs GL, Verkman AS. CFTR: folding, misfolding and correcting the DeltaF508 conformational defect. Trends in molecular medicine, 18(2), 81-91 (2012).
⦁ Balch WE, Roth DM, Hutt DM. Emergent properties of proteostasis in managing cystic fibrosis. Cold Spring Harb Perspect Biol, 3(2) (2011).
⦁ Roth DM, Hutt DM, Tong J et al. Modulation of the maladaptive stress response to manage diseases of protein folding. PLoS Biol, 12(11), e1001998 (2014).
⦁ Cystic Fibrosis Foundation Patient Registry: 2016 annual data report. Bethesda, MD: Cystic Fibrosis Foundation; 2017 [accessed 2018 May 1]. Available from:

https://www.cff.org/Research/Researcher-Resources/Patient-Registry/2016-Patient- Registry-Annual-Data-Report.pdf
⦁ Van Goor F, Hadida S, Grootenhuis PD et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci U S A, 106(44), 18825-18830 (2009).
⦁ Ramsey BW, Davies J, McElvaney NG et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med, 365(18), 1663-1672 (2011).
⦁ Sawicki GS, McKone EF, Pasta DJ et al. Sustained Benefit from ivacaftor demonstrated by combining clinical trial and cystic fibrosis patient registry data. Am J Respir Crit Care Med, 192(7), 836-842 (2015).
⦁ Davies JC, Wainwright CE, Canny GJ et al. Efficacy and safety of ivacaftor in patients
aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med, 187(11), 1219-1225 (2013).
⦁ De Boeck K, Munck A, Walker S et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros, 13(6), 674-680 (2014).
⦁ Moss RB, Flume PA, Elborn JS et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med, 3(7), 524-533 (2015).
⦁ Van Goor F, Yu H, Burton B, Hoffman BJ. Effect of ivacaftor on CFTR forms with missense mutations associated with defects in protein processing or function. J Cyst Fibros, 13(1), 29-36 (2014).
⦁ Durmowicz AG, Lim R, Rogers H, Rosebraugh CJ, Chowdhury BA. The U.S. Food and Drug Administration's Experience with Ivacaftor in Cystic Fibrosis. Establishing Efficacy Using In Vitro Data in Lieu of a Clinical Trial. Ann Am Thorac Soc, 15(1), 1-2 (2018).
⦁ Van Goor F, Hadida S, Grootenhuis PD et al. Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. Proc Natl Acad Sci U S A, 108(46), 18843-18848 (2011).
⦁ Wainwright CE, Elborn JS, Ramsey BW. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med, 373(18), 1783-1784 (2015).

⦁ Konstan MW, McKone EF, Moss RB et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): a phase 3, extension study. Lancet Respir Med, 5(2), 107-118 (2017).
⦁ Elborn JS, Ramsey BW, Boyle MP et al. Efficacy and safety of lumacaftor/ivacaftor combination therapy in patients with cystic fibrosis homozygous for Phe508del CFTR by pulmonary function subgroup: a pooled analysis. Lancet Respir Med, 4(8), 617-626 (2016).
⦁ Taylor-Cousar JL, Jain M, Barto TL et al. Lumacaftor/ivacaftor in patients with cystic fibrosis and advanced lung disease homozygous for F508del-CFTR. J Cyst Fibros, 17(2), 228-235 (2018).
⦁ Hubert D, Chiron R, Camara B et al. Real-life initiation of lumacaftor/ivacaftor combination in adults with cystic fibrosis homozygous for the Phe508del CFTR mutation and severe lung disease. J Cyst Fibros, 16(3), 388-391 (2017).
⦁ Jennings MT, Dezube R, Paranjape S et al. An Observational Study of Outcomes and Tolerances in Patients with Cystic Fibrosis Initiated on Lumacaftor/Ivacaftor. Ann Am Thorac Soc, 14(11), 1662-1666 (2017).
⦁ Popowicz N, Wood J, Tai A, Morey S, Mulrennan S. Immediate effects of lumacaftor/ivacaftor administration on lung function in patients with severe cystic fibrosis lung disease. J Cyst Fibros, 16(3), 392-394 (2017).
⦁ Robertson SM, Luo X, Dubey N et al. Clinical drug-drug interaction assessment of ivacaftor as a potential inhibitor of cytochrome P450 and P-glycoprotein. J Clin Pharmacol, 55(1), 56-62 (2015).
⦁ Jordan CL, Noah TL, Henry MM. Therapeutic challenges posed by critical drug-drug interactions in cystic fibrosis. Pediatr Pulmonol, 51(S44), S61-S70 (2016). *A review of drug-drug interactions that occur between CFTR modulators and other frequently prescribed medications in cystic fibrosis clinics.
⦁ Rudashevskaya EL, Stockner T, Trauner M, Freissmuth M, Chiba P. Pharmacological correction of misfolding of ABC proteins. Drug Discov Today Technol, 12, e87-94 (2014).

⦁ Pedemonte N, Lukacs GL, Du K et al. Small-molecule correctors of defective DeltaF508- CFTR cellular processing identified by high-throughput screening. J Clin Invest, 115(9), 2564-2571 (2005).
⦁ Ren HY, Grove DE, De La Rosa O et al. VX-809 corrects folding defects in cystic fibrosis transmembrane conductance regulator protein through action on membrane-spanning domain 1. Mol Biol Cell, 24(19), 3016-3024 (2013).
⦁ Mijnders M, Kleizen B, Braakman I. Correcting CFTR folding defects by small-molecule correctors to cure cystic fibrosis. Curr Opin Pharmacol, 34, 83-90 (2017).
⦁ Van Goor F. Nonclinical profile of the CFTR corrector VX-661. Pediatric Pulmonology, 51(S45), S194-S485 (2016).
⦁ Donaldson SH, Pilewski JM, Griese M et al. Tezacaftor/Ivacaftor in Subjects with Cystic Fibrosis and F508del/F508del-CFTR or F508del/G551D-CFTR. Am J Respir Crit Care Med, 197(2), 214-224 (2018). *The Phase 2 study of tezacaftor-ivacaftor in F508del homozygotes and F508del/G551D heterozygotes which established the superior safety profile for tezacaftor.
⦁ Symdeko [package insert]. Boston, MA: Vertex Pharmaceuticals Incorporated; 2018 [accessed 2018 May 1]. Available from: ⦁ https://www.accessdata.fd⦁ a.gov/drugsatfda_d⦁ ocs/label/20⦁ 18/210491lbl.pdf *Vertex drug insert for Tezacaftor-Ivacaftor (Symdeko) which contains indications, adverse effects, and basis for the approved mutations.
⦁ Sabers A. Pharmacokinetic interactions between contraceptives and antiepileptic drugs.
Seizure, 17(2), 141-144 (2008).
⦁ Rowe SM, Daines C, Ringshausen FC et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med, 377(21), 2024-2035 (2017). *The Phase 3 study of tezacaftor-ivacaftor in F508del / residual function compound heterozygotes which demonstrated efficacy in absolute increase in FEV1 independent of their specific residual function mutation.
⦁ Taylor-Cousar JL, Munck A, McKone EF et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med, 377(21), 2013-2023 (2017). *The

Phase 3 study of tezacaftor-ivacaftor in F508del / F508del homozygotes which demonstrated efficacy in absolute increase in FEV1 and reduced pulmonary exacerbation frequency.

⦁ Vertex Provides Update on Ongoing Phase 3 Program for VX-661 in Combination with Ivacaftor for the Treatment of Cystic Fibrosis; 2016 [accessed 2018 May 1]. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-provides- update-ongoing-phase-3-program-vx-661
⦁ Vertex Reports Third-Quarter 2017 Financial Results; 2017 [accessed 2018 May 1]. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex- reports-third-quarter-2017-financial-results
⦁ Flume, PA, Owen CA, Brown CD, Wang L et al. Long-term Safety and Efficacy of Tezacaftor/Ivacaftor in Patients With Cystic Fibrosis Homozygous for F508del-CFTR or Heterozygous for F508del and a Residual Function Mutation: First Interim Analysis Results of a Phase 3, Open-label, Rollover Study. Am J Respir Crit Care Med, 197, A7807 (2018).
⦁ Vertex Initiates Phase 3 Study of VX-659, Tezacaftor and Ivacaftor as a Triple Combination Regimen for People with Cystic Fibrosis Who Have Two Copies of the F508del Mutation; 2018 [accessed 2018 May 1]. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-initiates-phase-3- study-vx-659-tezacaftor-and-ivacaftor
⦁ Vertex Selects Two Next-Generation Correctors, VX-659 and VX-445, to Advance into Phase 3 Development as Part of Two Different Triple Combination Regimens for People with Cystic Fibrosis; 2018 [accessed 2018 May 1]. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-selects-two-next- generation-correctors-vx-659-and-vx-445

⦁ Vertex Initiates Phase 3 Studies of VX-445, Tezacaftor and Ivacaftor as a Triple Combination Regimen for People with Cystic Fibrosis; 2018 [accessed 2018 May 1]. Available from: ⦁ https://www.vrtx⦁ .com/story/vertex-initiate⦁ s-phase-3-vx-445-tr⦁ iple-combination-studies
⦁ Proteostasis Therapeutics Announces FDA Grants Breakthrough Therapy Designation for PTI-428 in Cystic Fibrosis; 2018 [accessed 2018 May 1]. Available from: http://ir.proteostasis.com/news-releases/news-release-details/proteostasis- therapeutics-announces-fda-grants-breakthrough

Table 1 – Specific residual function mutations included in EXPAND trial [39]

2789+5GA D110E R352Q R1070W
3849+10kbCT D110H A455E F1074L
3272-26AG R117C D579G D1152H
711+3AG E831X S945L D1270N
E56K L206W S977F
P67L R347H F1052V
R74W E193K K1060T

Table 2 – Genotype and Tezacaftor-Ivacaftor approval status

Mutation 1
Mutation 2 Approved for Tezacaftor-
Ivacaftor
Comments
F508del F508del Yes
Any CF- defining
mutation Residual Function Yes Residual function mutations approved for Symdeko are as listed in Table 1
F508del Minimal
Function No MF includes mutations from classes I
and II
F508del Class III Gating mutations
(e.g., G551D) No

May 15, 2018

Sophie Fagg [email protected] Commissioning Editor
Expert Review of Respiratory Medicine

Dear Dr. Fagg:

We are pleased to submit the invited manuscript entitled “Tezacaftor for the Treatment of Cystic Fibrosis” to be considered for publication in Expert Review of Respiratory Medicine. We believe that our manuscript achieves the mission of Expert Review to provide thorough but succinct coverage of an important topic, accompanied by expert commentary from leaders in the field, for a multidisciplinary audience.

As requested, below, you will find the authors’ details, separate from the submitted manuscript:
Authors: Marc Sala1, Manu Jain1
Affiliations: 1Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA.

Thank you very much for your consideration.

Sincerely,

Manu Jain Professor
Division of Pulmonary and Critical Care Medicine Feinberg School of Medicine
Northwestern University