MedPath

Lumacaftor Advanced Drug Monograph

Published:Sep 22, 2025

Generic Name

Lumacaftor

Brand Names

Orkambi

Drug Type

Small Molecule

Chemical Formula

C24H18F2N2O5

CAS Number

936727-05-8

Associated Conditions

Cystic Fibrosis (CF)

A Comprehensive Monograph on Lumacaftor (VX-809): Pharmacology, Clinical Efficacy, and Therapeutic Role in Cystic Fibrosis

Introduction

Cystic fibrosis (CF) is a life-shortening, autosomal recessive genetic disorder that precipitates a complex, multi-organ pathology. The disease arises from mutations in the gene encoding the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein.[1] This protein functions as a cyclic adenosine monophosphate (cAMP)-regulated anion channel, primarily facilitating the transport of chloride (

Cl−) and bicarbonate (HCO3−​) ions across the apical membrane of epithelial cells.[2] In healthy individuals, this ion movement is crucial for regulating the viscosity of secretions and maintaining hydration of epithelial surfaces in various organs, including the lungs, pancreas, intestines, and sweat glands.[1] Dysfunction of the CFTR protein disrupts this delicate hydro-electrolyte balance, leading to the production of abnormally thick, viscous mucus. This hallmark of CF pathology results in airway obstruction, chronic bacterial infections, progressive lung damage, pancreatic insufficiency, malnutrition, and other systemic complications.[1]

The genetic basis of CF is heterogeneous, with over 2,000 identified variants in the CFTR gene; however, a single mutation is responsible for the majority of cases worldwide. This mutation, known as F508del (or p.Phe508del), involves the deletion of a single phenylalanine residue at position 508 of the CFTR protein.[1] The F508del mutation is classified as a Class II defect, characterized by severe disruption of the protein's normal folding pathway within the endoplasmic reticulum. This misfolding triggers the cell's quality control mechanisms, which recognize the aberrant protein and target it for premature ubiquitination and proteasomal degradation.[1] The primary consequence is a drastic reduction in the quantity of functional CFTR channels that successfully traffic to and are integrated into the cell surface. The small amount of F508del-CFTR that does escape this degradation pathway exhibits additional functional impairments, including decreased stability at the cell membrane and a defective channel gating mechanism, further compounding the loss of function.[1]

Prior to the advent of targeted molecular therapies, the management of CF was entirely supportive, focusing on symptom control, mucus clearance, nutritional support, and treatment of infections.[6] The development of Lumacaftor represented a paradigm shift in this approach. As a first-in-class CFTR "corrector," Lumacaftor was specifically engineered to address the fundamental molecular defect of the F508del mutation: the protein trafficking failure.[2] By partially rescuing the misfolded protein and promoting its delivery to the cell surface, Lumacaftor established the therapeutic principle of correcting the underlying protein defect. This innovation laid the groundwork for the corrector-potentiator combination strategy, a therapeutic paradigm that has fundamentally altered the natural history of cystic fibrosis for a significant portion of the patient population.

1. Molecular Profile and Physicochemical Properties

A thorough understanding of Lumacaftor's chemical identity and physical characteristics is foundational to appreciating its pharmacological behavior, formulation requirements, and therapeutic application. This section provides a definitive summary of its molecular and physicochemical profile.

1.1 Chemical Identification and Nomenclature

Lumacaftor is a synthetic organic small molecule with a complex structure that is precisely defined by its systematic chemical name and a series of unique identifiers used across scientific, regulatory, and commercial domains.

  • Systematic (IUPAC) Name: The International Union of Pure and Applied Chemistry (IUPAC) name for Lumacaftor is 3-{6-{amino}-3-methylpyridin-2-yl}benzoic acid.[1] This name systematically deconstructs the molecule into its core components, identifying it as an aromatic amide formed through the condensation of a 1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropane-1-carboxylic acid moiety with a 3-(6-amino-3-methylpyridin-2-yl)benzoic acid moiety.[5]
  • CAS Number: The unique Chemical Abstracts Service (CAS) Registry Number for Lumacaftor is 936727-05-8.[1] This identifier is universally used in chemical literature and databases to unambiguously identify the substance.
  • DrugBank ID: In the DrugBank database, a comprehensive resource for drug and drug target information, Lumacaftor is assigned the accession number DB09280.[1]
  • Developmental Codes and Synonyms: During its development by Vertex Pharmaceuticals, Lumacaftor was primarily known by the code VX-809. Other related developmental codes include VRT 826809 and VRT-826809.[1] These codes are frequently encountered in the preclinical and early clinical trial literature.
  • Other Database Identifiers: To facilitate comprehensive data retrieval and cross-referencing, Lumacaftor is cataloged in numerous other major databases. Key identifiers include its PubChem Compound ID (CID) 16678941, ChEMBL ID CHEMBL2103870, Unique Ingredient Identifier (UNII) EGP8L81APK, and Kyoto Encyclopedia of Genes and Genomes (KEGG) ID D10134.[6]

1.2 Structural and Physicochemical Characteristics

The physical and chemical properties of Lumacaftor dictate its behavior in biological systems, influencing its absorption, distribution, and formulation.

  • Chemical Formula and Molecular Weight: The molecular formula for Lumacaftor is C24​H18​F2​N2​O5​.[6] This corresponds to an average molecular weight of 452.414 g/mol and a more precise monoisotopic mass of 452.118378014 g/mol.[1]
  • Physical Form and Appearance: At room temperature, Lumacaftor is a white to off-white solid.[5]
  • Solubility: Lumacaftor is characterized by its very low solubility in aqueous media, with reported values of less than 0.05 µg/mL.[11] This poor water solubility is a critical physicochemical property that significantly impacts its oral bioavailability and necessitates specific administration guidelines. The compound exhibits slight solubility in organic solvents such as dimethyl sulfoxide (DMSO) and methanol.[5]
  • Melting and Boiling Points: The melting point of Lumacaftor is reported to be in the range of 200–205°C. Its predicted boiling point is approximately 653.0±55.0 °C.[5]
  • Predicted pKa: The predicted acid dissociation constant (pKa) for Lumacaftor is 3.95±0.10.[5] This value is primarily attributed to the carboxylic acid group on the benzoic acid portion of the molecule, indicating that it will be predominantly ionized at physiological pH.

The key chemical and physical properties of Lumacaftor are summarized in Table 1 for ease of reference.

Table 1: Chemical and Physical Properties of Lumacaftor

PropertyValueSource(s)
Systematic (IUPAC) Name3-{6-{amino}-3-methylpyridin-2-yl}benzoic acid1
CAS Number936727-05-86
DrugBank IDDB092801
Developmental CodesVX-809, VRT 826809, VRT-8268091
Chemical FormulaC24​H18​F2​N2​O5​6
Average Molecular Weight452.414 g/mol1
Physical FormWhite to Off-White Solid5
Aqueous Solubility<0.05 µg/mL11
Organic SolubilitySlightly soluble in DMSO and Methanol5
Predicted pKa3.95±0.105

2. Mechanism of Action: A Pharmacological Chaperone for F508del-CFTR

Lumacaftor's therapeutic effect is derived from its function as a CFTR corrector, a class of molecules designed to address the primary molecular defect associated with the F508del mutation. Its mechanism is best understood in the context of both the specific pathology it targets and the residual defects it fails to address, which provides the essential rationale for its use in combination therapy.

2.1 The Molecular Pathology of the F508del Mutation

The F508del mutation is a Class II mutation, meaning it primarily impairs protein maturation and trafficking.[3] The deletion of the phenylalanine residue at position 508 in the nucleotide-binding domain 1 (NBD1) disrupts a critical folding checkpoint during the protein's biosynthesis. This structural perturbation causes the nascent polypeptide chain to misfold, preventing it from attaining its correct three-dimensional conformation.[1]

This misfolded protein is recognized as defective by the robust cellular quality control machinery located in the endoplasmic reticulum (ER). Consequently, the vast majority of F508del-CFTR protein is retained within the ER, ubiquitinated, and targeted for premature degradation by the proteasome.[1] This results in the defining feature of the F508del mutation: a profound quantitative defect, with a severely reduced number of CFTR channels successfully trafficking through the Golgi apparatus to the apical membrane of epithelial cells.[1]

Furthermore, the small fraction of F508del-CFTR protein that does manage to escape ER-associated degradation and reach the cell surface is not fully functional. It exhibits two additional intrinsic defects:

  1. Defective Channel Gating: The protein displays a significantly reduced channel-open probability, meaning it does not open as frequently as wild-type CFTR to allow chloride ion passage.[1]
  2. Decreased Stability: The protein is less stable at the plasma membrane and is subject to accelerated turnover and removal.[1]

These combined defects—a primary and severe trafficking failure coupled with secondary gating and stability issues—result in a near-total loss of CFTR function in individuals homozygous for the F508del mutation.

2.2 Lumacaftor as a CFTR Corrector

Lumacaftor functions as a pharmacological chaperone, a small molecule that directly interacts with the misfolded F508del-CFTR protein to facilitate its proper conformation.[7] While the precise binding site is complex, evidence suggests that Lumacaftor stabilizes the N-terminal portion of the protein, particularly the first membrane-spanning domain (MSD1).[5] By binding to and stabilizing this region, Lumacaftor aids the conformational maturation of the protein, effectively shielding it from recognition by the ER's quality control system.[1]

This chaperone activity partially rescues the protein from its default path of degradation. As a result, a greater proportion of F508del-CFTR is able to complete its processing, undergo complex glycosylation in the Golgi, and traffic to the cell surface.[1] The net effect of Lumacaftor is an increase in the density of F508del-CFTR channels at the apical membrane, thereby addressing the primary quantitative defect of the mutation.[2] Preclinical

in vitro studies using Fischer rat thyroid (FRT) cells expressing F508del-CFTR provided early validation of this mechanism. In these assays, Lumacaftor (VX-809) was shown to improve the maturation of the F508del-CFTR protein and enhance its chloride transport function, with half-maximal effective concentrations (EC50​) of 0.1 µM and 0.5 µM, respectively.[5]

2.3 The Rationale for Combination Therapy

The mechanism of Lumacaftor reveals a critical limitation that defines its therapeutic use. While it successfully increases the quantity of F508del-CFTR channels at the cell surface, it does not correct the intrinsic functional defects of the rescued protein. The channels that arrive at the membrane, courtesy of Lumacaftor's chaperone activity, still suffer from the characteristic low channel-open probability (gating defect) inherent to the F508del mutation.[1] Delivering more channels to the surface is a necessary first step, but it is therapeutically insufficient if those channels remain predominantly closed.

This insufficiency created the absolute requirement for a second, mechanistically distinct agent: a CFTR "potentiator." Ivacaftor (DB08820) is a potentiator that acts directly on CFTR channels already present at the cell surface. It binds to the channel and allosterically modulates its function, increasing the probability that the channel will be in an open conformation, thereby facilitating a greater flow of chloride ions.[2]

The combination of Lumacaftor and Ivacaftor, commercialized as Orkambi, thus represents a synergistic, dual-mechanism approach to treating the F508del mutation. Lumacaftor, the corrector, increases the number of channels at the destination, while Ivacaftor, the potentiator, ensures those channels can perform their function more effectively once they arrive.[5] This two-pronged strategy partially restores CFTR function to a level—estimated to be approximately 10–20% of normal

in vivo—sufficient to produce clinical benefit.[2] The development of Lumacaftor and the elucidation of its mechanism did more than just produce a single drug; it established the corrector-potentiator paradigm as the foundational therapeutic strategy for CF, a principle that has guided all subsequent CFTR modulator development.

3. Clinical Pharmacology and Pharmacokinetics

The disposition of Lumacaftor within the human body—its absorption, distribution, metabolism, and excretion (ADME)—is characterized by several key features that are critical for its safe and effective clinical use. Its pharmacokinetic profile explains the specific dosing requirements, the potential for variability in patient response, and the basis for certain drug interactions.

3.1 Absorption, Distribution, and the Critical Role of Fat-Containing Foods

  • Absorption: Lumacaftor is administered orally and is systemically absorbed. In both healthy subjects and patients with CF, its systemic exposure, as measured by the area under the concentration-time curve (AUC), increases in a roughly dose-proportional manner within the clinically relevant dose range.[14] Following oral administration in a fed state, the time to reach maximum plasma concentration ( Tmax​) is approximately 4 hours.[14]
  • Food Effect: The absorption of Lumacaftor is profoundly influenced by the presence of food, a direct consequence of its lipophilic nature and poor aqueous solubility. Administration with a fat-containing meal is essential for optimal bioavailability. Clinical studies have consistently demonstrated that taking Lumacaftor with fatty foods increases its systemic exposure by approximately 2-fold compared to administration in a fasted state.[11] This significant food effect underscores the critical importance of patient adherence to administration instructions, as failure to take the medication with an appropriate meal can lead to sub-therapeutic drug levels.
  • Steady State and Accumulation: With a twice-daily dosing regimen, Lumacaftor reaches steady-state plasma concentrations within approximately one week of initiation. A moderate accumulation factor of about 1.9 is observed, indicating that concentrations at steady state are nearly double those after the first dose.[14]
  • Distribution: Once absorbed into the systemic circulation, Lumacaftor is extensively bound to plasma proteins, with approximately 99% bound, primarily to albumin.[14] This high degree of protein binding limits the amount of free, pharmacologically active drug. Lumacaftor also has a large apparent volume of distribution, with estimates for the central and peripheral compartments being 23.5 L and 33.3 L, respectively, suggesting significant distribution into tissues outside of the bloodstream.[14]

3.2 Metabolism and Elimination Pathways

  • Metabolism: In humans, Lumacaftor does not undergo extensive metabolism. The primary metabolic pathways are minor and involve oxidation and glucuronidation.[14] The major metabolite identified in the blood, designated M28-LUM, is present at concentrations less than 10% of the parent drug and is not considered to be pharmacologically active at these levels.[14]
  • Elimination: The primary route of elimination for Lumacaftor is through the feces. The majority of an administered dose is excreted as unchanged parent drug in the feces, with studies reporting values ranging from 51% to nearly 90%.[14] Renal elimination is negligible, with less than 0.2% of the dose recovered as unchanged drug in the urine.[14] This excretion profile suggests that dose adjustments are not necessary for patients with mild to moderate renal impairment.
  • Half-Life: When administered as part of the combination product Orkambi, Lumacaftor exhibits a relatively long terminal half-life of approximately 26 hours.[11] This pharmacokinetic property supports a convenient twice-daily (every 12 hours) dosing schedule to maintain therapeutic concentrations.

3.3 Pharmacokinetic Profile and Factors Influencing Variability

The pharmacokinetic profile of Lumacaftor is marked by considerable inter-individual variability, which can influence patient response and tolerability.[11] Studies have identified patient-specific factors that contribute to this variability. Notably, both patient age and body weight have been shown to have a statistically significant effect on the peak plasma concentration (

Cmax​) of Lumacaftor.[11] This finding provides the rationale for the age- and weight-based dosing strategies employed, particularly in the pediatric population, to achieve consistent therapeutic exposures across different age groups.

An interesting observation is that the systemic exposure to Lumacaftor is approximately 2-fold higher in healthy volunteers than in patients with CF.[11] The reasons for this difference are not fully elucidated but may be related to physiological differences in the CF population, such as altered gastrointestinal transit, fat malabsorption, or differences in body composition, which could affect drug absorption and distribution.

The combination of low aqueous solubility, a pronounced food effect, and high inter-patient variability gives Lumacaftor a somewhat "fragile" pharmacokinetic profile. This implies that its clinical effectiveness in a real-world setting is highly dependent on factors beyond the drug molecule itself, particularly patient education and strict adherence to the administration requirement of co-ingestion with fat-containing food. Any deviation from this instruction could easily lead to insufficient drug exposure, potentially rendering an expensive and otherwise beneficial therapy ineffective. This reliance on extrinsic factors for consistent bioavailability may partly explain the gap sometimes observed between the tightly controlled outcomes of clinical trials and the effectiveness seen in routine clinical practice.

Table 2: Summary of Lumacaftor Pharmacokinetic Parameters

ParameterValueClinical RelevanceSource(s)
Time to Peak (Tmax​)~4.0 hours (fed state)Reached within a few hours of oral dosing.14
Food Effect (AUC)~2-fold increase with fatAdministration with fat-containing food is mandatory for adequate absorption.14
Protein Binding~99% (primarily albumin)High binding limits free drug concentration; potential for displacement interactions.14
Apparent Volume of Distribution (Vd​)Central: 23.5 L; Peripheral: 33.3 LIndicates extensive distribution into tissues beyond the plasma.14
Terminal Half-life (T1/2​)~26 hours (in combination)Long half-life supports a twice-daily (q12h) dosing regimen.11
Apparent Clearance (CL/F)2.38 L/hrReflects the rate of drug elimination from the body.16
Primary Route of EliminationFeces (~51-90% as unchanged drug)Minimal renal excretion; no dose adjustment needed for mild-moderate renal impairment.14
Accumulation Factor~1.9Concentrations nearly double from first dose to steady state.14

4. Drug-Drug Interaction Profile: The Clinical Impact of CYP3A4 Induction

The drug-drug interaction (DDI) profile of Lumacaftor is one of its most clinically significant characteristics. As a potent enzyme inducer, it has the potential to alter the pharmacokinetics of a wide range of co-administered medications, including its own partner drug, Ivacaftor. This necessitates careful medication management for patients with CF, who often require polypharmacy to manage their complex disease.

4.1 Potent Induction of Cytochrome P450 Enzymes and P-glycoprotein

Lumacaftor is a strong and broad-spectrum inducer of drug-metabolizing enzymes and transporters. Its most prominent effect is the potent induction of the Cytochrome P450 3A (CYP3A) subfamily, which is responsible for the metabolism of a large proportion of clinically used drugs.[14]

In vitro data and clinical studies have confirmed this strong inductive effect.[11]

Beyond CYP3A, Lumacaftor also has the potential to induce several other important CYP isoenzymes, including CYP2B6, CYP2C8, CYP2C9, and CYP2C19.[6] Furthermore, it is an inducer of the efflux transporter P-glycoprotein (P-gp), which plays a key role in limiting the absorption and promoting the excretion of many drugs.[6] This wide-ranging inductive activity means that the initiation of Lumacaftor therapy can significantly accelerate the clearance of any co-administered drug that serves as a substrate for these pathways. This can lead to decreased systemic exposure and potential therapeutic failure of the affected medications.[16]

4.2 Intramolecular Interaction: Impact on Ivacaftor Exposure

A unique and critical drug interaction occurs within the Orkambi combination product itself. Ivacaftor, the potentiator component, is a sensitive substrate of CYP3A enzymes.[11] When Lumacaftor is co-administered, its powerful induction of CYP3A leads to a substantial increase in the metabolic clearance of Ivacaftor. This results in a marked reduction in Ivacaftor's plasma concentration and overall systemic exposure (AUC) compared to when Ivacaftor is administered alone.[3]

This intramolecular pharmacokinetic antagonism presents a significant formulation challenge. To counteract the inductive effect of Lumacaftor and ensure that Ivacaftor achieves therapeutic concentrations, the daily dose of Ivacaftor in the Orkambi fixed-dose combination is significantly higher (500 mg total daily dose) than the dose used in Ivacaftor monotherapy (Kalydeco®, 300 mg total daily dose).[3] This dose adjustment is a direct consequence of Lumacaftor's DDI profile.

This inherent pharmacological conflict—where one component of a combination therapy actively reduces the concentration of its essential partner—represents a therapeutic paradox. It places a ceiling on the potential efficacy of the combination and creates a complex web of external interactions that complicates patient management. This significant clinical liability was a primary driver for the pharmaceutical industry's quest to develop next-generation CFTR correctors, such as Tezacaftor and Elexacaftor, which were specifically designed to have cleaner DDI profiles and lack the potent CYP3A induction property of Lumacaftor.

4.3 Clinically Significant Interactions and Management

The potent inductive properties of Lumacaftor lead to a large number of clinically significant drug interactions, requiring careful review of a patient's medication list before and during therapy. The interactions can be broadly categorized based on the interacting agent.

  • Interaction with Strong CYP3A Inducers: Co-administration of Orkambi with other strong CYP3A inducers (e.g., rifampin, carbamazepine, phenobarbital, St. John's Wort) is not recommended. While these agents have minimal effect on Lumacaftor itself, they can further decrease the already-reduced exposure of Ivacaftor, potentially rendering the combination therapy ineffective.[20]
  • Interaction with CYP3A Inhibitors: Strong CYP3A inhibitors (e.g., ketoconazole, itraconazole, clarithromycin, voriconazole) block the metabolism of Ivacaftor, leading to a significant increase in its plasma concentrations and a higher risk of adverse effects. To manage this, if Orkambi is initiated in a patient already taking a strong CYP3A inhibitor, the Orkambi dose must be reduced (e.g., to one tablet daily for patients ≥6 years) for the first week of treatment. After this initial period, the full dose can be resumed. No dose adjustment is needed if an inhibitor is started in a patient already stable on Orkambi.[16] Patients should also be advised to avoid grapefruit and Seville oranges, as they contain natural CYP3A inhibitors.[24]
  • Interaction with CYP3A Substrates: As a strong inducer, Lumacaftor can significantly reduce the exposure and efficacy of drugs metabolized by CYP3A. This is a major concern for sensitive substrates or those with a narrow therapeutic index.
  • Hormonal Contraceptives: Oral, injectable, transdermal, and implantable hormonal contraceptives are CYP3A substrates. Their effectiveness is substantially reduced by Lumacaftor, and they should not be relied upon as a method of contraception. Alternative or barrier methods are recommended. Furthermore, an increased incidence of menstrual abnormalities has been reported in patients using hormonal contraceptives with Orkambi.[16]
  • Immunosuppressants: Drugs like cyclosporine, tacrolimus, sirolimus, and everolimus are sensitive CYP3A substrates. Co-administration with Orkambi is not recommended due to the high risk of decreased immunosuppressant levels, which could lead to organ rejection in transplant recipients.[20]
  • Other Sensitive Substrates: Co-administration with many other sensitive CYP3A substrates, such as certain benzodiazepines (midazolam, alprazolam), HMG-CoA reductase inhibitors (statins), and antipsychotics, is either not recommended or requires careful monitoring and potential dose adjustments of the substrate drug.[20]
  • Antifungal Agents: A particularly complex interaction exists with triazole antifungals. While strong inhibitors like itraconazole increase Ivacaftor levels, Lumacaftor simultaneously induces the metabolism of the antifungal itself. Studies have shown that co-administration of Orkambi with itraconazole, posaconazole, or voriconazole can result in sub-therapeutic levels of the antifungal agent, risking treatment failure for fungal infections common in CF.[26]

Table 3: Clinically Significant Drug Interactions with Lumacaftor/Ivacaftor (Orkambi)

Interacting Drug/ClassMechanism of InteractionEffect on Interacting DrugEffect on OrkambiClinical Management RecommendationSource(s)
Part A: Drugs Affecting Orkambi
Strong CYP3A Inducers (e.g., rifampin, carbamazepine, St. John's Wort)CYP3A InductionN/A↓ Ivacaftor exposureCo-administration is not recommended due to risk of reduced Orkambi efficacy.22
Strong CYP3A Inhibitors (e.g., itraconazole, ketoconazole, clarithromycin)CYP3A InhibitionN/A↑ Ivacaftor exposure by ~4.3-foldIf initiating Orkambi, reduce dose for the first week. No adjustment if patient is already on Orkambi. Avoid grapefruit.22
Part B: Orkambi Affecting Other Drugs
Sensitive CYP3A Substrates (general)Lumacaftor is a strong CYP3A inducer↓ ExposureN/ACo-administration not recommended, especially for drugs with a narrow therapeutic index. May require dose increase of substrate.16
Hormonal Contraceptives (oral, injectable, implantable)CYP3A Induction↓ Exposure, reduced efficacyN/AShould not be relied upon for contraception. Alternative methods are required. Increased menstrual abnormalities reported.16
Immunosuppressants (e.g., cyclosporine, tacrolimus)CYP3A Induction↓ ExposureN/ACo-administration is not recommended due to risk of sub-therapeutic levels and organ rejection.20
Benzodiazepines (e.g., midazolam, alprazolam)CYP3A Induction↓ Exposure, reduced efficacyN/AConsider alternative agents. Co-administration with midazolam is not recommended.20
Triazole Antifungals (e.g., itraconazole, voriconazole)CYP3A Induction↓ Exposure, sub-therapeutic levels↑ Ivacaftor exposure (inhibitor effect)Complex interaction. Monitor for breakthrough fungal infections. Dose of antifungal may need to be increased.20
WarfarinInduction of CYP2C9↓ Exposure (potential)N/AMonitor International Normalized Ratio (INR) closely upon initiation of Orkambi.20
Part C: Contraindicated Combinations
Various (e.g., bosentan, cariprazine, cobimetinib, doravirine, elbasvir/grazoprevir)Multiple, primarily CYP3A induction by Lumacaftor or induction of Orkambi metabolism.VariesVariesCo-administration is contraindicated due to significant reduction in efficacy of one or both agents.22

5. Clinical Efficacy in Cystic Fibrosis

The clinical development program for Lumacaftor, as part of the fixed-dose combination Orkambi, was extensive, encompassing pivotal trials in adults and adolescents, followed by studies in progressively younger pediatric populations. The results of these trials established its efficacy profile, characterized by a consistent and robust effect on a key biomarker of CFTR function but a more modest impact on clinical measures of lung function.

5.1 Pivotal Phase III Trials in Adults and Adolescents (≥12 years): TRAFFIC and TRANSPORT

The cornerstone of the regulatory submission for Orkambi consisted of two identically designed, Phase III, randomized, double-blind, placebo-controlled, 24-week studies named TRAFFIC (VX13-809-103) and TRANSPORT (VX13-809-104).[27] These landmark trials enrolled a combined total of 1,108 patients with CF aged 12 years and older who were homozygous for the F508del mutation.[12]

  • Primary Endpoint (Lung Function): The primary efficacy endpoint in both studies was the absolute change from baseline in the percent predicted forced expiratory volume in one second (ppFEV₁) through week 24.[29]
  • Treatment with the approved dose of Lumacaftor 400 mg / Ivacaftor 250 mg every 12 hours resulted in improvements that were statistically superior to placebo, but modest in magnitude.
  • In the TRAFFIC study, the least squares mean treatment difference (Orkambi vs. placebo) for the absolute change in ppFEV₁ was 2.6 percentage points (95% CI: 1.2, 4.0; P=0.0003).[1]
  • In the TRANSPORT study, the treatment difference was 3.0 percentage points (95% CI: 1.6, 4.4; P<0.0001).[1]
  • While statistically significant, the clinical relevance of this magnitude of improvement was debated, as a substantial portion of patients—over 70% in one analysis—did not achieve an absolute improvement in ppFEV₁ of 5% or more, a threshold often considered to be clinically meaningful.[1]
  • Key Secondary Endpoints:
  • Pulmonary Exacerbations: One of the most compelling findings from the trials was the effect on pulmonary exacerbations, which are key drivers of morbidity and lung function decline in CF. A pooled analysis of the data from both trials demonstrated a clinically meaningful and statistically significant reduction in the rate of exacerbations. The hazard ratio for having an exacerbation was significantly lower in the treatment group, corresponding to a 30% to 39% reduction in events compared to the placebo group.[14] The rate of exacerbations requiring hospitalization was also significantly reduced.
  • Nutritional Status (BMI): The results for Body Mass Index (BMI), a crucial indicator of overall health in CF, were inconsistent between the two trials. A statistically significant improvement in BMI was observed in the TRANSPORT study (treatment difference of 0.4 kg/m²) but not in the TRAFFIC study.[32]
  • Patient-Reported Outcomes (CFQ-R): Despite improvements in lung function and exacerbation rates, the trials failed to show a statistically significant improvement in the respiratory domain score of the Cystic Fibrosis Questionnaire-Revised (CFQ-R), a validated measure of health-related quality of life from the patient's perspective.[29]

5.2 Efficacy in Pediatric Populations

Following the initial approval for adolescents and adults, a series of open-label studies were conducted to evaluate the safety, pharmacokinetics, and efficacy of Orkambi in younger children. These studies supported the sequential expansion of the drug's indication down to the age of one year.[27]

  • Ages 6–11 Years: A 24-week, placebo-controlled study (VX15-809-109) in 204 children in this age group demonstrated efficacy using a different primary endpoint. The Lung Clearance Index (LCI2.5​), a more sensitive measure of ventilation inhomogeneity and small airway disease, showed a statistically significant improvement in the Orkambi group compared to placebo (mean change of -1.01 vs. +0.08).[33]
  • Ages 2–5 Years and 1–2 Years: For the youngest pediatric cohorts, efficacy was established primarily through safety and pharmacokinetic data, with pharmacodynamic endpoints serving as key supportive evidence. These studies demonstrated that Orkambi was generally safe and that age- and weight-based dosing achieved target drug exposures. The primary evidence of biological activity came from significant reductions in sweat chloride concentration and improvements in markers of growth (e.g., BMI-for-age z-score) and pancreatic function (fecal elastase), which were considered reasonably likely to predict clinical benefit.[33]

5.3 Key Outcomes Across Trials: Biomarkers

  • Sweat Chloride: Across all age groups studied, treatment with Lumacaftor/Ivacaftor resulted in a rapid, substantial, and sustained reduction in sweat chloride concentration.[1] This biomarker provides direct evidence of the drug's effect on restoring CFTR protein function in the sweat duct. Mean absolute reductions from baseline were consistently in the range of 25 to 32 mmol/L from pathologically high starting values (typically >100 mmol/L).[1] Importantly, in studies that included a washout period, sweat chloride levels quickly returned toward pre-treatment values upon cessation of the drug, confirming that the observed effect was directly attributable to the medication.[1]

The clinical data for Orkambi reveals a notable disconnect between its effect on a direct biomarker of CFTR function (sweat chloride) and its impact on a key clinical outcome of lung disease (ppFEV₁). The large and consistent drop in sweat chloride provides unequivocal proof that the drug combination is working at the molecular level to restore a significant degree of CFTR channel activity. However, this robust pharmacodynamic effect does not translate into a proportionally large improvement in lung function. This therapeutic gap suggests that restoring CFTR function to the level achieved by Lumacaftor/Ivacaftor (~15-20% of normal) is sufficient to substantially normalize ion transport in the sweat gland but is not enough to overcome the complex, downstream pathophysiology of established CF lung disease, which includes accumulated mucus, chronic inflammation, and a persistent cycle of infection. This observation highlighted the need for even more effective modulators capable of restoring a higher level of CFTR function, a need that was eventually met by triple-combination therapies. It also provided a strong rationale for initiating modulator therapy as early in life as possible, with the goal of preventing the onset of irreversible lung damage.

5.4 Long-Term and Real-World Evidence

The open-label extension study of TRAFFIC and TRANSPORT, known as PROGRESS, provided longer-term data. Results suggested that the modest improvements in ppFEV₁ were maintained for up to 96 weeks and, more importantly, that treatment with Orkambi was associated with a slower rate of annual lung function decline compared to matched untreated patients from the U.S. CF Patient Registry.[12]

However, post-marketing studies have illuminated the challenges of translating clinical trial results into real-world practice. These studies have reported significantly higher rates of treatment discontinuation than seen in the pivotal trials, with one multicenter cohort study finding that 18.2% of patients stopped the drug within the first year.[40] The primary reason for discontinuation was adverse events, particularly respiratory symptoms. For patients who were able to tolerate and continue the treatment, the clinical benefits in lung function and nutritional status were generally consistent with those observed in the trials. Conversely, patients who discontinued therapy often experienced a significant decline in their clinical status, highlighting the risks associated with treatment intolerance.[40]

Table 4: Summary of Efficacy Outcomes from Pivotal TRAFFIC and TRANSPORT Trials (Lumacaftor 400 mg/Ivacaftor 250 mg q12h vs. Placebo)

EndpointTRAFFIC StudyTRANSPORT Study
Orkambi (n=184)Placebo (n=188)
Absolute Change in ppFEV₁ at Week 24 (percentage points)
LS Mean Treatment Difference (95% CI)2.6 (1.2, 4.0)3.0 (1.6, 4.4)
P-valueP=0.0003P<0.0001
Relative Change in ppFEV₁ at Week 24 (%)
LS Mean Treatment Difference (95% CI)4.3 (1.9, 6.8)5.3 (2.7, 7.8)
P-valueP=0.0006P<0.0001
Absolute Change in BMI at Week 24 (kg/m²)
LS Mean Treatment Difference (95% CI)0.1 (-0.1, 0.3)0.4 (0.2, 0.5)
P-valueNot SignificantP=0.0001
Absolute Change in CFQ-R Respiratory Domain Score at Week 24 (points)
LS Mean Treatment Difference (95% CI)1.5 (-1.7, 4.7)2.9 (-0.3, 6.0)
P-valueNot SignificantNot Significant
Pulmonary Exacerbations (Pooled Analysis)
Hazard Ratio (95% CI)0.61 (0.45, 0.83)
P-valueP=0.0014
Data sourced from.1 ppFEV₁ = percent predicted forced expiratory volume in one second; BMI = Body Mass Index; CFQ-R = Cystic Fibrosis Questionnaire-Revised; LS = Least Squares; CI = Confidence Interval.

6. Safety, Tolerability, and Risk Management

The safety and tolerability profile of Lumacaftor/Ivacaftor is a critical factor in its clinical use, influencing patient selection, monitoring requirements, and real-world adherence. While generally considered to have an acceptable profile, several key safety concerns require diligent clinical management.

6.1 Common and Significant Adverse Events Profile

The most frequently reported adverse events in clinical trials of Orkambi were respiratory and gastrointestinal in nature.[12]

  • Common Side Effects: In pooled analyses of the pivotal trials, adverse events occurring in ≥5% of patients and at a higher rate than placebo included dyspnea (shortness of breath, 13%), nasopharyngitis (13%), nausea (13%), diarrhea (12%), upper respiratory tract infection (10%), fatigue (9%), respiration abnormal/chest tightness (9%), increased blood creatine phosphokinase (9%), rash (7%), and flatulence (7%).[24]
  • Menstrual Abnormalities: An increased incidence of menstrual abnormalities, such as irregular periods, dysmenorrhea, or menorrhagia, has been observed in female patients, particularly those concomitantly using hormonal contraceptives. This is attributed to the drug's interaction with and reduction of contraceptive hormone levels.[16]
  • Increased Blood Pressure: Modest increases in blood pressure have been reported in patients receiving Orkambi. Regular blood pressure monitoring is recommended during treatment.[24]

6.2 In-Depth Review of Key Safety Concerns

Beyond the common side effects, several specific safety concerns warrant particular attention and have led to specific risk management strategies in the drug's labeling.

  • Hepatotoxicity (Liver-related Events): The potential for liver injury is a significant safety concern. Elevated liver transaminases (ALT and AST) have been observed in clinical trials.[32] More seriously, post-marketing reports include cases of serious liver-related adverse reactions, some associated with concomitant elevations in total serum bilirubin. There have been reports of worsening liver function, including hepatic encephalopathy, in patients with pre-existing advanced liver disease (e.g., cirrhosis).[24]
  • Risk Management: To mitigate this risk, liver function tests (ALT, AST, and total bilirubin) are mandatory for all patients prior to initiating Orkambi. These tests must be repeated every 3 months during the first year of treatment and annually thereafter. For patients with a history of transaminase elevations, more frequent monitoring is recommended. Dosing must be interrupted if ALT or AST levels rise to >5 times the upper limit of normal (ULN), or if they rise to >3 times ULN with concurrent bilirubin >2 times ULN. The decision to resume dosing after resolution should be based on a careful benefit-risk assessment.[24]
  • Respiratory Events: A distinct pattern of respiratory adverse events, including dyspnea, chest tightness, and abnormal respiration, is characteristic of Orkambi, particularly upon treatment initiation.[24] These symptoms are most common within the first week of therapy and typically resolve with continued treatment.[29]
  • The incidence and severity of these respiratory events are notably higher in patients with more advanced lung disease, specifically those with a baseline ppFEV₁ of less than 40%.[24] This specific adverse event profile represents a major barrier to treatment initiation and adherence in a vulnerable patient population. In real-world settings, these early-onset respiratory symptoms are a primary contributor to the high rates of treatment discontinuation, creating a subset of "non-tolerant" patients who are unable to access the drug's potential long-term benefits. This issue is particularly problematic as it disproportionately affects patients with severe disease who are in greatest need of effective therapy. This tolerability challenge was a key factor motivating the development of next-generation modulators with improved safety profiles.
  • Cataracts: Cases of non-congenital lens opacities, or cataracts, have been reported in pediatric patients receiving Orkambi and other CFTR modulators. While a direct causal link has not been definitively established, the observation has led to specific monitoring recommendations.[24]
  • Risk Management: It is recommended that pediatric patients undergo baseline and follow-up ophthalmological examinations to monitor for the development of cataracts.[24]

7. Regulatory and Commercial Landscape

The regulatory journey and market placement of Lumacaftor, as the central component of Orkambi, mark a significant chapter in the history of cystic fibrosis therapeutics. Its approval represented the first therapy to target the underlying defect in the largest CF patient population, establishing a new standard of care and a major commercial franchise.

7.1 Global Regulatory Approval History and Approved Indications

Orkambi received marketing authorization from major regulatory agencies worldwide, with a consistent and specific indication.

  • U.S. Food and Drug Administration (FDA): Orkambi was first approved by the FDA in July 2015 for use in patients with CF aged 12 years and older.[1] This initial approval was a landmark event, offering the first disease-modifying therapy for the F508del homozygous population. Following this, the indication was systematically expanded to younger age groups based on subsequent clinical data:
  • 2016: Approval extended to children aged 6 through 11 years.[36]
  • 2018: Approval extended to children aged 2 through 5 years.[36]
  • 2022: Approval further extended to children aged 1 to less than 2 years.[36]
  • European Medicines Agency (EMA): The EMA granted a marketing authorization for Orkambi in November 2015, initially for patients aged 12 and older.[33] Similar to the FDA, the indication was later expanded to include younger children, now down to the age of one year.[33] In its assessment, the EMA acknowledged that the clinical benefits, particularly in lung function, were smaller than might be expected for a drug targeting the disease mechanism, but concluded that they were clinically relevant given the severity of the disease and the absence of alternative treatments for this genotype.[33]
  • Therapeutic Goods Administration (TGA) of Australia: Orkambi is approved by the TGA and is also listed on the Pharmaceutical Benefits Scheme (PBS), which provides subsidized access for eligible patients. The reimbursed indication in Australia covers patients aged one year and older who are homozygous for the F508del mutation.[46]
  • Approved Indication: Across all regulatory jurisdictions, the indication for the Lumacaftor/Ivacaftor combination (Orkambi) is highly specific: it is for the treatment of cystic fibrosis in patients who are homozygous for the F508del mutation in the CFTR gene.[1] An FDA-cleared or equivalent CF mutation test is required to confirm the patient's genotype prior to initiating treatment.[47] The efficacy and safety of Orkambi have not been established in patients with other CF-causing mutations.

7.2 Dosing, Administration, and Available Formulations of Orkambi®

To accommodate different age groups and ensure appropriate dosing, Orkambi is available in multiple formulations and strengths.

  • Formulations: The drug is supplied as fixed-dose combination tablets for older children and adults, and as oral granules in unit-dose packets for infants and young children who cannot swallow tablets.[23]
  • Administration: A critical aspect of administration for all formulations is that Orkambi must be taken orally every 12 hours with a fat-containing meal or snack. This is essential to maximize the absorption of the lipophilic active ingredients.[14]
  • Dosage Regimens:
  • Tablets:
  • Patients aged 12 years and older: The recommended dose is two tablets, each containing Lumacaftor 200 mg and Ivacaftor 125 mg, taken every 12 hours (total daily dose: Lumacaftor 800 mg / Ivacaftor 500 mg).[14]
  • Patients aged 6 through 11 years: The recommended dose is two tablets, each containing Lumacaftor 100 mg and Ivacaftor 125 mg, taken every 12 hours (total daily dose: Lumacaftor 400 mg / Ivacaftor 250 mg).[23]
  • Oral Granules:
  • Patients aged 1 to 5 years: Dosing is based on body weight, using packets of varying strengths (Lumacaftor/Ivacaftor: 75 mg/94 mg, 100 mg/125 mg, or 150 mg/188 mg) administered every 12 hours.[23]
  • Dose Adjustments: Dose reductions are specified for certain populations. Patients with moderate hepatic impairment (Child-Pugh Class B) require a reduced dose, and the drug should be used with caution in patients with severe hepatic impairment (Child-Pugh Class C).[14] As previously detailed, a temporary dose reduction is also required when initiating therapy in patients who are already taking strong CYP3A inhibitors.[23]

7.3 Patent Status and Market Context

Orkambi is a key product of Vertex Pharmaceuticals and is protected by a robust portfolio of intellectual property.

  • Patent Protection: The drug is covered by numerous patents in the United States and other countries. These patents cover the drug substance (the active molecules), pharmaceutical compositions and formulations, and specific methods of use for treating cystic fibrosis in defined patient populations.[49] Key patents protecting Orkambi are expected to expire in the late 2020s and early 2030s. For example, patents related to the composition and methods of administration extend to dates such as December 2028, August 2029, and December 2030.[49]
  • Market Exclusivity: In addition to patent protection, Orkambi has benefited from periods of regulatory exclusivity, including pediatric exclusivity, which can extend market protection beyond the patent expiration dates. Based on an analysis of the existing patents and regulatory exclusivities, the earliest potential date for a generic version of Orkambi to enter the market is projected to be June 11, 2031.[49]
  • Market Position: The launch of Orkambi solidified Vertex Pharmaceuticals' position as the leader in the cystic fibrosis therapeutic market.[52] It was a flagship product that generated substantial revenue and served as the commercial and clinical foundation for the company's subsequent, more advanced CFTR modulators, including Symdeko (Tezacaftor/Ivacaftor) and the highly effective triple-combination therapy Trikafta (Elexacaftor/Tezacaftor/Ivacaftor).[53]

Conclusion: Lumacaftor's Legacy and the Evolution of CFTR Modulator Therapy

Lumacaftor, the corrector component of the combination therapy Orkambi, holds a pivotal place in the history of medicine. Its development and clinical application represent a fundamental shift in the treatment of cystic fibrosis, moving from purely symptomatic management to a strategy that targets the underlying molecular cause of the disease. The successful clinical validation of Lumacaftor served as the essential proof-of-concept for the corrector-potentiator therapeutic paradigm, demonstrating for the first time that it was possible to partially rescue the defective F508del-CFTR protein and achieve clinical benefit in the largest segment of the CF population.[2] This achievement irrevocably changed the natural history of the disease for eligible patients and validated a novel drug development pathway focused on correcting protein-folding defects.

This landmark success, however, must be viewed through the lens of the drug's significant limitations, which ultimately defined its role as a first-generation agent. The clinical trial data consistently showed that its efficacy, while statistically significant, was clinically modest, particularly with respect to improvements in lung function.[1] This was coupled with a challenging safety and tolerability profile, most notably the early-onset respiratory adverse events that led to high rates of treatment discontinuation in real-world settings, especially among patients with more advanced lung disease.[40] Perhaps its greatest liability was its potent induction of the CYP3A enzyme system, which created a therapeutic paradox by accelerating the metabolism of its essential partner, Ivacaftor, and generating a complex and problematic profile of drug-drug interactions that complicated the care of a patient population characterized by polypharmacy.[18]

Ultimately, the legacy of Lumacaftor is that of a foundational but transitional therapy. The very limitations that constrained its clinical utility—modest efficacy, tolerability issues, and a difficult DDI profile—provided the clear and compelling impetus for the rapid development of its successors. The search for a corrector with an improved safety profile and a cleaner interaction footprint led directly to the development of Tezacaftor. The need for a much greater magnitude of clinical efficacy drove the discovery of the next-generation corrector Elexacaftor and the advent of highly effective triple-combination therapy. In this context, Lumacaftor was not an endpoint but a crucial stepping stone. It was the indispensable first step that illuminated the path forward, fundamentally altering the therapeutic landscape for cystic fibrosis and paving the way for the more transformative and life-altering medicines that have followed.

Works cited

  1. Lumacaftor: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 22, 2025, https://go.drugbank.com/drugs/DB09280
  2. Lumacaftor-ivacaftor in the treatment of cystic fibrosis: design ..., accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6650604/
  3. Introduction - Clinical Review Report: Lumacaftor/Ivacaftor (Orkambi) - NCBI Bookshelf, accessed September 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK540353/
  4. Efficacy of lumacaftor-ivacaftor for the treatment of cystic fibrosis patients homozygous for the F508del-CFTR mutation - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4963025/
  5. Lumacaftor (VX-809) | 936727-05-8 - ChemicalBook, accessed September 22, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB32489655.htm
  6. Lumacaftor | C24H18F2N2O5 | CID 16678941 - PubChem, accessed September 22, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Lumacaftor
  7. Lumacaftor - Wikipedia, accessed September 22, 2025, https://en.wikipedia.org/wiki/Lumacaftor
  8. Lumacaftor (VX-809) | CFTR Modulator | MedChemExpress, accessed September 22, 2025, https://www.medchemexpress.com/VX-809.html
  9. lumacaftor | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 22, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7481
  10. Compound: LUMACAFTOR (CHEMBL2103870) - ChEMBL - EMBL-EBI, accessed September 22, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL2103870
  11. Insights Into Patient Variability During Ivacaftor-Lumacaftor Therapy in Cystic Fibrosis, accessed September 22, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2021.577263/full
  12. Lumacaftor/Ivacaftor: A Review in Cystic Fibrosis - ResearchGate, accessed September 22, 2025, https://www.researchgate.net/publication/305110523_LumacaftorIvacaftor_A_Review_in_Cystic_Fibrosis
  13. How ORKAMBI® (lumacaftor/ivacaftor) Works | Watch the Video, accessed September 22, 2025, https://www.orkambi.com/how-orkambi-works
  14. 206038Orig1s000 - accessdata.fda.gov, accessed September 22, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/0206038Orig1s000ClinPharmR.pdf
  15. Demonstrating Bioequivalence for a Lumacaftor Monosubstance Formulation Versus Orkambi® (Lumacaftor/Ivacaftor) in Healthy Subjects | springermedizin.de, accessed September 22, 2025, https://www.springermedizin.de/demonstrating-bioequivalence-for-a-lumacaftor-monosubstance-form/51138500
  16. Reference ID: 4212367 This label may not be the latest approved by ..., accessed September 22, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206038s007lbl.pdf
  17. A retrospective observational study: Bidirectional pharmacokinetic interactions between ivacaftor-lumacaftor in cystic fibrosis - ERS Publications, accessed September 22, 2025, https://publications.ersnet.org/content/erj/56/suppl64/362
  18. Cytochrome P450 3A4 Induction: Lumacaftor versus Ivacaftor Potentially Resulting in Significantly Reduced Plasma Concentration of Ivacaftor - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/29595119/
  19. Cytochrome P450 3A4 Induction: Lumacaftor versus Ivacaftor Potentially Resulting in Significantly Reduced Plasma Concentration of Ivacaftor - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6350194/
  20. Drug Interactions | ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambihcp.com/drug-interactions
  21. Ivacaftor and Lumacaftor Pathway, Pharmacokinetics/Pharmacodynamics - ClinPGx, accessed September 22, 2025, https://www.clinpgx.org/pathway/PA166153178
  22. Orkambi (lumacaftor/ivacaftor) dosing, indications, interactions, adverse effects, and more, accessed September 22, 2025, https://reference.medscape.com/drug/orkambi-lumacaftor-ivacaftor-1000000
  23. Dosing and Administration | ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambihcp.com/dosing-and-administration
  24. Orkambi (lumacaftor / ivacaftor): Uses, Side Effects, Dosage & Reviews, accessed September 22, 2025, https://www.goodrx.com/orkambi/what-is
  25. Orkambi®– Lumacaftor/Ivacaftor Drug Drug Interaction a case study - FDA, accessed September 22, 2025, https://www.fda.gov/media/94945/download
  26. Subtherapeutic triazole concentrations as result of a drug-drug interaction with lumacaftor/ivacaftor. - ClinPGx, accessed September 22, 2025, https://www.clinpgx.org/literature/15153288
  27. Executive Summary - Clinical Review Report: Lumacaftor/Ivacaftor (Orkambi) - NCBI, accessed September 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK540359/
  28. Clinical Trials Finder | CFF Clinical Trials Tool - Find a Care Center, accessed September 22, 2025, https://apps.cff.org/Trials/Finder?search=%7BPipelineDrugId%3A%2786%27%2CPipelineDrugDisplayName%3A%27Lumacaftor+%2B+ivacaftor+(Orkambi)+%27%7D
  29. Results - Clinical Review Report: Lumacaftor/Ivacaftor (Orkambi ..., accessed September 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK540360/
  30. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR - PubMed, accessed September 22, 2025, https://pubmed.ncbi.nlm.nih.gov/25981758/
  31. Orkambi (lumacaftor/ivacaftor) for the Treatment of Cystic Fibrosis (CF) - Clinical Trials Arena, accessed September 22, 2025, https://www.clinicaltrialsarena.com/projects/orkambi-lumacaftor-ivacaftor-cystic-fibrosis/
  32. Clinical Trials for Age 12 Years and Older - ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambihcp.com/clinical-data-trials-1-2
  33. Orkambi | European Medicines Agency (EMA), accessed September 22, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/orkambi
  34. CDR Clinical Review Report for Orkambi - Canada's Drug Agency, accessed September 22, 2025, https://www.cda-amc.ca/sites/default/files/cdr/clinical/SR0471_Orkambi_CL_Report.pdf
  35. Clinical Studies and Results - ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambi.com/clinical-studies
  36. History of ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambi.com/orkambi-history
  37. Safety and efficacy of treatment with lumacaftor in combination with ivacaftor in younger patients with cystic fibrosis - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6482058/
  38. Effects of Lumacaftor/Ivacaftor on Cystic Fibrosis Disease Progression in Children 2 through 5 Years of Age Homozygous for F508del-CFTR, accessed September 22, 2025, https://www.atsjournals.org/doi/pdf/10.1513/AnnalsATS.202208-684OC?download=true
  39. Treatment of Cystic Fibrosis Patients Homozygous for F508del with Lumacaftor-Ivacaftor (Orkambi®) Restores Defective CFTR Channel Function in Circulating Mononuclear Cells - MDPI, accessed September 22, 2025, https://www.mdpi.com/1422-0067/21/7/2398
  40. Clinical Effectiveness of Lumacaftor/Ivacaftor in Patients with Cystic Fibrosis Homozygous for F508del-CFTR. A Clinical Trial - PMC, accessed September 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7780982/
  41. Real-Life Safety and Effectiveness of Lumacaftor–Ivacaftor in Patients with Cystic Fibrosis, accessed September 22, 2025, https://www.atsjournals.org/doi/10.1164/rccm.201906-1227OC
  42. Orkambi (lumacaftor/ivacaftor): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed September 22, 2025, https://www.webmd.com/drugs/2/drug-169200/orkambi-oral/details
  43. Orkambi Interactions Checker - Drugs.com, accessed September 22, 2025, https://www.drugs.com/drug-interactions/ivacaftor-lumacaftor,orkambi.html
  44. highlights of prescribing information - Vertex Pharmaceuticals, accessed September 22, 2025, https://pi.vrtx.com/files/uspi_lumacaftor_ivacaftor.pdf
  45. Annotation of EMA Label for ivacaftor / lumacaftor and CFTR - ClinPGx, accessed September 22, 2025, https://www.clinpgx.org/labelAnnotation/PA166182953
  46. CFTR Modulators – Cystic Fibrosis Australia, accessed September 22, 2025, https://www.cysticfibrosis.org.au/cftr-modulators/
  47. Drugs@FDA: Drug Product ORKAMBI (lumacaftor and ivacaftor), NDA206038, Vertex Pharmaceuticals Incorporated - ClinPGx, accessed September 22, 2025, https://www.clinpgx.org/literature/15101996
  48. Mechanism of Action - ORKAMBI® (lumacaftor/ivacaftor), accessed September 22, 2025, https://www.orkambihcp.com/mechanism-of-action
  49. Generic Orkambi Availability - Drugs.com, accessed September 22, 2025, https://www.drugs.com/availability/generic-orkambi.html
  50. Drug Patents containing Ivacaftor; Lumacaftor - Pharsight, accessed September 22, 2025, https://pharsight.greyb.com/ingredient/ivacaftor;-lumacaftor-patent-expiration
  51. When will the patents on ORKAMBI expire, and when will generic ORKAMBI be available? - DrugPatentWatch, accessed September 22, 2025, https://www.drugpatentwatch.com/p/tradename/ORKAMBI
  52. Vertex Pharmaceuticals (VRTX) Secures Italian Reimbursement for Gene-Edited Therapy, accessed September 22, 2025, https://www.gurufocus.com/news/3112746/vertex-pharmaceuticals-vrtx-secures-italian-reimbursement-for-geneedited-therapy
  53. Orkambi* (lumacaftor/ivacaftor) vs Trikafta* (elexacaftor/tezacaftor/ivacaftor - Everyone.org, accessed September 22, 2025, https://everyone.org/explore/compare?id1=20&id2=315
  54. Biotech Stocks To Follow Today - September 21st - MarketBeat, accessed September 22, 2025, https://www.marketbeat.com/instant-alerts/biotech-stocks-to-follow-today-september-21st-2025-09-21/

Published at: September 22, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.