MedPath

Afatinib Advanced Drug Monograph

Published:Jul 15, 2025

Generic Name

Afatinib

Brand Names

Gilotrif, Giotrif

Drug Type

Small Molecule

Chemical Formula

C24H25ClFN5O3

CAS Number

850140-72-6

Associated Conditions

Metastatic Non-Small Cell Lung Cancer, Refractory, metastatic squamous cell Non-small cell lung cancer

Comprehensive Monograph: Afatinib (DB08916)

Executive Summary

Afatinib is a second-generation, orally administered, irreversible tyrosine kinase inhibitor (TKI) belonging to the ErbB family blocker class of antineoplastic agents. Developed by Boehringer Ingelheim and marketed under brand names including Gilotrif and Giotrif, it represents a significant milestone in the targeted therapy of cancer.[1] Its primary clinical application is in the treatment of metastatic non-small cell lung cancer (NSCLC).[1] Specifically, it is indicated as a first-line treatment for patients whose tumors harbor non-resistant epidermal growth factor receptor (EGFR) mutations and as a second-line treatment for patients with metastatic squamous NSCLC that has progressed following platinum-based chemotherapy.[4]

The pharmacological distinction of Afatinib lies in its mechanism of action. Unlike first-generation reversible TKIs, Afatinib forms a covalent bond with the kinase domains of EGFR (ErbB1), human epidermal growth factor receptor 2 (HER2/ErbB2), and HER4 (ErbB4). This irreversible binding results in a potent, sustained, and broad-spectrum blockade of signaling pathways critical for tumor cell proliferation, survival, and differentiation.[6] This mechanism confers activity not only against common activating EGFR mutations but also against certain mutations that are resistant to first-generation agents.[1]

The clinical efficacy of Afatinib is robustly supported by the extensive LUX-Lung clinical trial program. In the first-line setting for EGFR mutation-positive (EGFR M+) NSCLC, pivotal Phase III trials demonstrated a significant improvement in progression-free survival (PFS) compared to standard chemotherapy.[9] A key finding from these trials is a statistically significant overall survival (OS) benefit in the large patient subgroup with tumors harboring an exon 19 deletion (del19) mutation.[1] Furthermore, Afatinib has shown a survival advantage over erlotinib in the second-line treatment of squamous NSCLC.[12]

The safety profile of Afatinib is well-characterized and is a direct consequence of its mechanism of action. The potent inhibition of wild-type EGFR in normal epithelial tissues leads to a high incidence of predictable, mechanism-based adverse events, most notably diarrhea and rash/acneiform dermatitis.[1] The successful use of Afatinib is therefore intrinsically linked to proactive and aggressive management of these toxicities, often requiring dose modifications to maintain treatment continuity and patient quality of life.[5]

Afatinib has secured regulatory approval from major global agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), solidifying its role in oncology practice.[15] It holds a distinct position in the therapeutic armamentarium for NSCLC, particularly for patients with del19 or specific uncommon EGFR mutations, and as a component of sequential therapy strategies in an evolving landscape of targeted treatments.

Drug Identity and Physicochemical Properties

Nomenclature and Identifiers

The small molecule drug Afatinib is identified by a comprehensive set of names and international codes to ensure unambiguous reference in clinical, research, and regulatory contexts. It was developed by Boehringer Ingelheim under the internal code BIBW 2992.[1] Upon commercialization, it was assigned distinct brand names for different markets, most notably Gilotrif in the United States and Giotrif in the European Union, with other names including Afanix also in use.[1]

The standardized nomenclature and identifiers are crucial for global pharmacovigilance, scientific literature indexing, and database integration. These are systematically cataloged across major chemical and pharmacological databases.

  • Generic Name (INN): Afatinib [1]
  • Brand Names: Gilotrif, Giotrif, Afanix, Tomtovok, Tovok, Xovoltib [1]
  • Developmental Code: BIBW 2992 [1]
  • DrugBank ID: DB08916 [1]
  • CAS Number (Free Base): 850140-72-6 [1]
  • IUPAC Name: N-[(3-Chloro-4-fluorophenyl)amino]-7-oxy]-6-quinazolinyl]-4(dimethylamino)-2-butenamide [1]
  • Other Key Identifiers:
  • PubChem CID: 10184653 [1]
  • ChemSpider: 8360155 [1]
  • UNII: 41UD74L59M [1]
  • KEGG: D09724 [1]
  • ChEBI: CHEBI:61390 [1]
  • ChEMBL: ChEMBL1173655 [1]

Chemical Structure and Formula

Afatinib is a synthetic organic compound classified as a quinazoline derivative, specifically a 4-anilinoquinazoline.[17] Its structure comprises a quinazoline core substituted at key positions to optimize target binding and pharmacological properties. The key structural features include a 3-chloro-4-fluoroanilino group at the 4-position, which anchors the molecule in the ATP-binding pocket of the target kinase, and a reactive butenamide side chain at the 6-position, which contains a Michael acceptor moiety responsible for its irreversible covalent binding.[1] A (S)-tetrahydrofuran-3-yloxy group is present at the 7-position.[17]

  • Chemical Class: Aniline-quinazoline derivative [17]
  • Molecular Formula: C24​H25​ClFN5​O3​ [1]
  • Molecular Weight: 485.94 g/mol [1]
  • Canonical SMILES: CN(CC=CC(=O)Nc1cc2c(ncnc2cc1OC1COCC1)Nc1ccc(c(c1)Cl)F)C [19]
  • InChIKey: ULXXDDBFHOBEHA-CWDCEQMOSA-N [1]

Afatinib Dimaleate Salt Form

For clinical use, Afatinib is formulated as a dimaleate salt, Afatinib dimaleate.[6] This pharmaceutical formulation strategy is critical for the drug's utility as an oral agent. The free base form of Afatinib is strongly lipophilic, with a calculated logP of 4.7, which would typically result in poor aqueous solubility and, consequently, low and erratic oral absorption.[22]

The conversion to a dimaleate salt dramatically improves the drug's physicochemical properties. The salt form is described as highly soluble throughout the physiological pH range of 1 to 7.5, with solubility greater than 50 mg/mL.[6] This enhanced solubility is a direct prerequisite for achieving consistent dissolution in the gastrointestinal tract, which in turn enables the high relative oral bioavailability (92%) observed for the tablet formulation.[7] Therefore, the decision to develop the dimaleate salt was a pivotal step in translating the potent molecule into a clinically viable oral therapy.

  • CAS Number (Dimaleate): 850140-73-7 [24]
  • Molecular Formula (Dimaleate): C32​H33​ClFN5​O11​ [24]
  • Molecular Weight (Dimaleate): 718.08 g/mol [24]
  • Solubility: Highly soluble in aqueous buffers (pH 1-7.5).[6]

The following table consolidates the key identification and physicochemical properties of Afatinib and its clinically relevant salt form.

Table 1: Drug Identification and Physicochemical Properties

PropertyValueSource(s)
Generic NameAfatinib1
Brand NamesGilotrif (US), Giotrif (EU)1
Developmental CodeBIBW 29921
DrugBank IDDB089161
Chemical ClassAniline-quinazoline17
IUPAC NameN-[(3-Chloro-4-fluorophenyl)amino]-7-oxy]-6-quinazolinyl]-4(dimethylamino)-2-butenamide1
CAS Number (Free Base)850140-72-61
Molecular Formula (Free Base)C24​H25​ClFN5​O3​1
Molecular Weight (Free Base)485.94 g/mol1
CAS Number (Dimaleate Salt)850140-73-724
Molecular Formula (Dimaleate Salt)C32​H33​ClFN5​O11​24
Molecular Weight (Dimaleate Salt)718.08 g/mol24
Solubility (Dimaleate Salt)Highly soluble in aqueous buffers (pH 1-7.5)6
pKa (Basic Groups)8.2 (dimethylamine), 5.0 (quinazoline)22

Pharmacology and Mechanism of Action

Classification

Afatinib is classified as a targeted antineoplastic agent. More specifically, it is a second-generation, orally bioavailable tyrosine kinase inhibitor (TKI).[1] Its key distinction within the TKI class is that it functions as an irreversible ErbB Family Blocker, a classification that sets it apart from both first-generation (reversible) and third-generation (mutant-selective) inhibitors.[6]

Molecular Target Engagement and Irreversible Inhibition

The primary therapeutic activity of Afatinib stems from its potent and selective inhibition of the ErbB family of receptor tyrosine kinases. This family, which includes the epidermal growth factor receptor (EGFR, also known as ErbB1), HER2 (ErbB2), HER3 (ErbB3), and HER4 (ErbB4), plays a central role in regulating cell growth, proliferation, and survival. In many cancers, these pathways are hyperactivated due to receptor overexpression or activating mutations.[6]

Afatinib exerts its effect by binding to the intracellular kinase domains of EGFR, HER2, and HER4, thereby blocking the autophosphorylation and subsequent downstream signaling necessary for tumor cell function.[6] Although it does not directly bind to the kinase-impaired ErbB3, it effectively neutralizes its signaling by inhibiting the transphosphorylation of ErbB3 by its binding partners (e.g., EGFR, HER2), thus achieving a complete blockade of signaling from all ErbB family dimers.[6]

The defining feature of Afatinib's mechanism is its irreversibility. The molecule contains an electrophilic acrylamide moiety that functions as a Michael acceptor. This group forms a stable, covalent bond with a specific cysteine residue located within the ATP-binding site of the target kinase.[1] For EGFR, this critical residue is Cysteine 797 (Cys797), and for HER2, it is Cysteine 805 (Cys805).[1] This covalent binding leads to a prolonged and essentially permanent inhibition of kinase activity, a mode of action that persists long after the drug has been cleared from systemic circulation.[6]

Potency and Selectivity

Afatinib demonstrates high potency against its target kinases, with inhibitory concentrations in the low nanomolar range. This potency is maintained across both wild-type and common mutated forms of EGFR, a key feature of its second-generation design.

  • Wild-Type EGFR: The half-maximal inhibitory concentration (IC50​) for wild-type EGFR is approximately 0.5 nM.[6]
  • Activating EGFR Mutations: Potency is maintained or even slightly enhanced against common activating mutations found in NSCLC, such as the exon 21 L858R substitution (IC50​ = 0.2–0.4 nM).[6]
  • Acquired Resistance Mutations: Crucially, Afatinib retains activity against the T790M "gatekeeper" mutation, which is the most common mechanism of acquired resistance to first-generation TKIs. While its potency is reduced against the L858R/T790M double mutant (IC50​ = 9–10 nM), this level of inhibition is still achievable at clinical doses and represents a significant advantage over its predecessors.[6]
  • HER2 (ErbB2): Afatinib also potently inhibits HER2 with an IC50​ of 14 nM.[21]

Comparative Pharmacology

The pharmacological properties of Afatinib are best understood in the context of the evolution of EGFR TKIs.

  • Versus First-Generation TKIs (e.g., Gefitinib, Erlotinib): The primary advantages of Afatinib are its irreversible binding and broader target profile. First-generation agents bind reversibly and are largely selective for EGFR.[8] Afatinib's covalent binding provides a more sustained and complete blockade of the signaling pathway.[8] Furthermore, its pan-ErbB activity inhibits signaling from multiple family members, potentially overcoming redundancy in signaling pathways. Most importantly, first-generation TKIs are rendered ineffective by the T790M resistance mutation, whereas Afatinib maintains clinically relevant activity.[30]
  • Versus Third-Generation TKIs (e.g., Osimertinib): The development of third-generation TKIs represents a further refinement in targeted therapy. These agents are designed to be highly selective for the mutated forms of EGFR (including T790M) while largely sparing the wild-type receptor.[23] This mutant-selectivity provides a significantly wider therapeutic window, resulting in fewer mechanism-based, on-target/off-tumor toxicities. Afatinib, by contrast, is equally potent against wild-type EGFR as it is against the common activating mutations.[19] This lack of selectivity is the direct cause of its prominent side effect profile.

The pharmacological design of Afatinib as a pan-ErbB, irreversible inhibitor is a double-edged sword that dictates its entire clinical profile. This design was a rational strategy to overcome the limitations of first-generation agents, particularly acquired resistance mediated by T790M.[23] The broad and potent blockade is responsible for its robust efficacy in EGFR M+ NSCLC, its activity in HER2-driven cancers (an investigational use), and its ability to inhibit some resistant clones.[1] However, this same mechanism is the direct cause of its most common and dose-limiting toxicities. ErbB receptors, especially wild-type EGFR, are essential for the homeostatic maintenance of normal epithelial tissues, particularly in the skin and gastrointestinal tract.[8] The potent and irreversible inhibition of wild-type EGFR by Afatinib inevitably disrupts these normal physiological processes, leading directly to the high incidence of diarrhea (>90%) and dermatologic reactions.[1] This inextricable link between the drug's primary strength and its principal weakness creates a narrow therapeutic index and elevates toxicity management to a central role in its clinical application.[19]

Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The pharmacokinetic profile of Afatinib describes its movement into, through, and out of the body. It is characterized by time-independent characteristics, minimal enzymatic metabolism, and a primary dependence on transporter proteins for disposition.[6]

Absorption

Following oral administration, Afatinib is well absorbed, although its exposure is significantly influenced by the presence of food.

  • Bioavailability: The relative oral bioavailability of a 20 mg Afatinib tablet is high, at 92% when compared to an oral solution, indicating efficient absorption of the dissolved drug.[7]
  • Time to Peak Concentration (Tmax​): Maximum plasma concentrations (Cmax​) are typically achieved between 2 and 5 hours post-dose.[6]
  • Food Effect: The absorption of Afatinib is highly sensitive to food. Co-administration with a high-fat meal results in a substantial reduction in drug exposure, decreasing Cmax​ by 50% and the area under the concentration-time curve (AUC) by 39%. Even consuming food within 3 hours before or 1 hour after taking Afatinib can reduce the AUC by an average of 26%.[7] This pronounced food effect necessitates strict administration guidelines, requiring patients to take the medication on an empty stomach.[3]

Distribution

Once absorbed, Afatinib distributes extensively throughout the body and binds avidly to proteins in both plasma and blood cells.

  • Plasma Protein Binding: Afatinib is highly bound to human plasma proteins, with a bound fraction of approximately 95%.[1]
  • Volume of Distribution (Vd​): The apparent volume of distribution is large, reported at 2770 L, which indicates extensive penetration into peripheral tissues beyond the plasma compartment.[7]
  • Red Blood Cell Partitioning: A notable characteristic of Afatinib is its rapid and extensive partitioning into red blood cells, where it is highly associated.[28]
  • Accumulation: The drug's effective elimination half-life of approximately 37 hours supports once-daily dosing and leads to predictable accumulation upon reaching steady state. Drug exposure (based on AUC) increases by a factor of 2.5 to 3.4 after multiple doses compared to a single dose.[6]

Metabolism

Afatinib undergoes minimal enzymatic metabolism, a key feature that distinguishes it from many other orally administered drugs.

  • Metabolic Pathway: Enzyme-catalyzed metabolic reactions, particularly those mediated by the cytochrome P450 (CYP) system, play a negligible role in the clearance of Afatinib.[1]
  • Metabolites: The primary "metabolites" of Afatinib are not products of enzymatic conversion but are rather covalently bound adducts formed via non-enzymatic Michael addition reactions. These adducts are formed with nucleophilic groups on endogenous proteins, primarily plasma proteins like albumin and hemoglobin.[6] This is a direct consequence of the drug's reactive chemistry, which is essential for its irreversible binding to target kinases. While this covalent binding is central to its therapeutic mechanism, the formation of adducts with other endogenous proteins has been noted as a theoretical concern for potential idiosyncratic drug reactions.[28]

Excretion

The elimination of Afatinib from the body occurs primarily through the feces as unchanged drug.

  • Primary Route: The major route of elimination is fecal excretion.[6]
  • Fecal Excretion: Following a single oral dose of radiolabeled Afatinib, 85.4% of the dose was recovered in the feces, almost entirely as the parent drug.[7]
  • Renal Excretion: Renal clearance is a minor pathway, accounting for only about 4-5% of the total dose being excreted in the urine.[1]
  • Elimination Half-Life (t1/2​): The effective terminal elimination half-life is approximately 37 hours, consistent with its accumulation profile and suitability for once-daily administration.[1]

The pharmacokinetic profile is largely consistent across different patient populations, with factors like age, ethnicity, and hepatic function having no clinically significant influence. However, exposure is correlated with renal function, and is higher in females and patients with low body weight, which may necessitate closer monitoring in these groups.[31] The minimal involvement of CYP enzymes in its metabolism is a significant clinical advantage, as it greatly reduces the potential for drug-drug interactions with the many medications that act as CYP inhibitors or inducers. Instead, the focus of interaction management shifts entirely to the P-glycoprotein (P-gp) efflux transporter, for which Afatinib is a substrate.[6]

Table 2: Summary of Key Pharmacokinetic Parameters

ParameterValueSource(s)
Route of AdministrationOral1
Relative Bioavailability92% (tablet vs. oral solution)7
Time to Peak (Tmax​)2–5 hours6
Food Effect (High-Fat Meal)Cmax​ decreased by 50%, AUC decreased by 39%7
Plasma Protein Binding~95%1
Apparent Volume of Distribution (Vd​)2770 L7
Primary MetabolismMinimal; non-enzymatic covalent adduct formation6
Primary Excretion RouteFeces (as unchanged drug)6
Fraction Excreted in Feces85.4%7
Fraction Excreted in Urine4.3%17
Terminal Half-Life (t1/2​)~37 hours1

Clinical Efficacy and Therapeutic Applications

The clinical utility of Afatinib has been established through a comprehensive development program, primarily in non-small cell lung cancer (NSCLC). Its efficacy is demonstrated in specific, biomarker-defined patient populations, reflecting the targeted nature of the therapy.

First-Line Treatment of EGFR Mutation-Positive (EGFR M+) NSCLC

The cornerstone of Afatinib's approval as a first-line therapy for EGFR M+ metastatic NSCLC rests on two pivotal, randomized Phase III trials: LUX-Lung 3 (a global study) and LUX-Lung 6 (conducted in an Asian population).[11] These trials compared Afatinib against standard-of-care platinum-based chemotherapy regimens (pemetrexed/cisplatin in LUX-Lung 3 and gemcitabine/cisplatin in LUX-Lung 6).[9]

  • Progression-Free Survival (PFS): Both trials successfully met their primary endpoint, showing a statistically significant and clinically meaningful improvement in PFS for patients treated with Afatinib.
  • In LUX-Lung 3, the median PFS was 11.1 months in the Afatinib arm versus 6.9 months in the chemotherapy arm (Hazard Ratio for progression or death, 0.58; 95% CI, 0.43 to 0.78; p<0.001).[9]
  • In LUX-Lung 6, the results were similarly robust, with a median PFS of 11.0 months for Afatinib versus 5.6 months for chemotherapy (HR, 0.28; 95% CI, 0.20 to 0.39; p<0.0001).[10]
  • When analysis was restricted to patients with the two most common EGFR mutations (exon 19 deletion [del19] or exon 21 L858R substitution), the benefit was even more pronounced, with a median PFS of 13.6 months in the Afatinib arm of LUX-Lung 3.[9]
  • Overall Survival (OS) by Mutation Type: While the initial trials did not show an OS benefit in the overall EGFR M+ population, a pre-planned combined analysis of OS data from LUX-Lung 3 and LUX-Lung 6 revealed a critical finding. Patients whose tumors harbored the del19 mutation experienced a significant OS benefit when treated with first-line Afatinib compared to chemotherapy. These patients lived a median of over one year longer, establishing a clear survival advantage for this specific subgroup.[1] In contrast, a similar OS benefit was not observed for patients with the L858R mutation. This discovery highlights a profound biological heterogeneity between the two most common "activating" EGFR mutations. It suggests that tumors driven by del19 mutations may exhibit a stronger "oncogene addiction" that is more durably disrupted by the irreversible, pan-ErbB blockade of Afatinib, translating a PFS benefit into a significant OS advantage. This finding has direct clinical implications, making Afatinib a compelling first-line choice for patients with del19-mutant NSCLC.
  • Uncommon EGFR Mutations: Beyond the common mutations, Afatinib has demonstrated consistent clinical activity against a range of non-resistant, uncommon EGFR mutations, such as G719X, L861Q, and S768I.[1] In a large, pooled analysis of over 1000 patients with uncommon mutations treated with Afatinib, those who were TKI-naïve and had major uncommon mutations achieved a median Time to Treatment Failure (TTF) of 12.6 months and an objective response rate (ORR) of 59.0%.[34] This robust dataset establishes Afatinib as a preferred TKI for many of these rarer mutation subtypes, for which prospective data from other TKIs is often lacking.[34]

Second-Line Treatment of Metastatic Squamous NSCLC

Afatinib's indication was expanded to include the treatment of patients with metastatic squamous NSCLC progressing on or after platinum-based chemotherapy. This approval was based on the results of the LUX-Lung 8 trial, a large, randomized, open-label Phase III study.[12]

  • Pivotal Trial (LUX-Lung 8): This trial directly compared Afatinib against Erlotinib, a first-generation EGFR TKI that was a standard second-line option at the time.[12]
  • Key Outcomes: Afatinib demonstrated superiority over Erlotinib for both the primary endpoint of PFS and the key secondary endpoint of OS. Treatment with Afatinib reduced the risk of cancer progression by 19% and, more importantly, reduced the risk of death by 19% compared to Erlotinib. Median OS was 7.9 months for patients in the Afatinib arm versus 6.8 months for those in the Erlotinib arm.[12] This was the first trial to show a survival benefit of one EGFR TKI over another in this setting and established Afatinib as an important therapeutic option for this patient population.

Investigational Uses and Real-World Evidence

  • HER2-Positive Breast Cancer: Emerging evidence has supported the investigation of Afatinib in other cancers driven by ErbB family members. Phase II results in patients with HER2-positive breast cancer were described as promising, with 19 of 41 patients (46%) deriving clinical benefit, prompting further investigation in Phase III trials.[1]
  • Sequential Therapy (GioTag Study): The approval of highly effective third-generation TKIs like osimertinib, particularly for T790M-mediated resistance, altered the treatment landscape. This could have rendered second-generation agents obsolete. However, Boehringer Ingelheim strategically supported the GioTag study, a global real-world evidence initiative.[38] This study investigated the clinical outcomes of a specific treatment sequence: first-line Afatinib followed by osimertinib upon the development of T790M-mediated resistance. The results were striking, showing a median OS of nearly four years (47.6 months in the U.S. cohort).[39] This finding repositioned Afatinib not as a competitor to osimertinib, but as a potential partner in a long-term sequencing strategy designed to maximize overall survival. This represents a sophisticated example of product life-cycle management, adapting to a changing competitive environment by generating data to answer new and relevant clinical questions.

Table 3: Summary of Pivotal Phase III Clinical Trials

Trial NameIndicationNTreatment ArmsPrimary EndpointKey Results (Median PFS / OS)Source(s)
LUX-Lung 31L EGFR M+ NSCLC (Global)345Afatinib vs. Pemetrexed/CisplatinPFSPFS: 11.1 vs. 6.9 months (HR 0.58)OS (del19): 33.3 vs. 21.1 months (HR 0.54)1
LUX-Lung 61L EGFR M+ NSCLC (Asia)364Afatinib vs. Gemcitabine/CisplatinPFSPFS: 11.0 vs. 5.6 months (HR 0.28)OS (del19): 31.4 vs. 18.4 months (HR 0.64)10
LUX-Lung 82L Squamous NSCLC795Afatinib vs. ErlotinibPFSPFS: 2.6 vs. 1.9 months (HR 0.81)OS: 7.9 vs. 6.8 months (HR 0.81)12

Safety Profile and Management of Adverse Events

Overview

The safety profile of Afatinib is well-defined and consistent with its mechanism of action as a potent, irreversible pan-ErbB inhibitor. The adverse events (AEs) are largely predictable, class-specific, and manageable with appropriate supportive care and dose adjustments.[13] The most prominent toxicities are gastrointestinal and dermatologic, arising directly from the inhibition of wild-type EGFR in normal tissues.[8]

Common Adverse Reactions

The most frequently reported AEs in patients treated with Afatinib are listed below. The high incidence rates underscore the importance of patient education and proactive management from the initiation of therapy.

  • Diarrhea: This is the most common AE, reported in over 90% of patients in clinical trials. While usually Grade 1 or 2, it can be severe (Grade 3 or 4) and may lead to dehydration, hypokalemia, and subsequent renal impairment. Fatal cases have been reported.[1] Proactive management with anti-diarrheal agents like loperamide is a cornerstone of treatment.[5]
  • Rash/Acneiform Dermatitis: A very common cutaneous reaction, manifesting as an erythematous and acneiform rash. It typically occurs on the face, neck, and upper torso and can be exacerbated by sun exposure. Patients should be advised to use sunscreen and wear protective clothing.[1]
  • Stomatitis/Mucositis: Inflammation and ulceration of the oral mucosa are frequent, causing pain and difficulty eating, which can impact nutrition and quality of life.[1]
  • Paronychia: Inflammation of the nail folds is common and can be painful, affecting the fingers and toes.[1]
  • Other Common AEs (Incidence >20%): Decreased appetite, dry skin (xerosis), and itching (pruritus) are also frequently observed.[1]

Serious and Life-Threatening Adverse Reactions

While less common, several serious AEs require immediate medical attention and may necessitate permanent discontinuation of the drug.

  • Interstitial Lung Disease (ILD)/Pneumonitis: A rare but potentially fatal AE (incidence ~1.5%). Patients must be monitored for acute onset or worsening of pulmonary symptoms such as dyspnea (shortness of breath), cough, and fever. If ILD is suspected, Afatinib must be withheld, and if confirmed, it should be permanently discontinued.[1]
  • Severe Cutaneous Reactions: Life-threatening bullous, blistering, and exfoliative skin conditions, including rare cases suggestive of Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), have been reported. The drug must be discontinued if SJS or TEN is suspected.[14]
  • Hepatotoxicity: Abnormalities in liver function tests occur in approximately 9.7% of patients, with rare (0.2%) fatal cases of hepatic failure. Periodic monitoring of liver enzymes (ALT, AST) is mandatory during treatment. The drug should be withheld for worsening liver function and discontinued for severe drug-induced hepatic impairment.[4]
  • Gastrointestinal Perforation: Though rare (0.2%), GI perforation has been reported and can be fatal. Patients with a history of GI ulceration, diverticular disease, or those receiving concomitant corticosteroids or NSAIDs are at increased risk.[4]
  • Keratitis/Corneal Ulceration: Inflammation of the cornea can occur. Patients with a history of keratitis, ulcerative keratitis, or severe dry eye, as well as contact lens wearers, are at higher risk. Symptoms like eye pain, redness, or blurred vision require prompt ophthalmologic evaluation.[1]

The successful clinical administration of Afatinib is fundamentally dependent on proactive toxicity mitigation. Because the side effects are a direct and predictable extension of the drug's pharmacology, their management can be planned in advance. This transforms the treatment paradigm from a reactive "prescribe and monitor" model to an active, collaborative effort between the clinician and an educated patient. Patients should be equipped with supportive care medications (e.g., loperamide) and instructed on their use at the earliest sign of an AE.[5] This approach is crucial for maintaining dose intensity, which is linked to efficacy, and preserving the patient's quality of life, thereby enabling them to remain on this effective therapy for as long as possible.

Table 4: Common and Serious Adverse Reactions with Management Strategies

Adverse ReactionIncidenceNCI-CTCAE Grade Triggering ActionRecommended Management/InterventionSource(s)
Diarrhea>90%Grade 2 lasting >48 hrs; ≥Grade 3Initiate loperamide at first sign. Withhold Afatinib until resolves to ≤Grade 1. Resume at reduced dose. Ensure hydration.1
Rash/DermatitisVery CommonGrade 2 lasting >7 days; Intolerable Grade 2; ≥Grade 3Use emollients, topical antibiotics/steroids. Advise sun protection. Withhold Afatinib until resolves to ≤Grade 1. Resume at reduced dose.1
Stomatitis/MucositisCommon≥Grade 3Supportive care (e.g., mouthwashes). Withhold Afatinib until resolves to ≤Grade 1. Resume at reduced dose.1
Interstitial Lung Disease (ILD)~1.5%Any suspected/confirmed casePermanently discontinue Afatinib.1
Hepatotoxicity~9.7% (any grade)Worsening liver function; Severe impairmentObtain periodic liver tests. Withhold for worsening function. Permanently discontinue for severe impairment.14
GI Perforation0.2%Any suspected/confirmed casePermanently discontinue Afatinib.7
KeratitisUncommonUlcerative keratitis confirmedInterrupt or permanently discontinue Afatinib.1
Severe Skin Reactions (SJS/TEN)RareAny suspected/confirmed casePermanently discontinue Afatinib.14

Dosage, Administration, and Special Populations

Recommended Dosing and Administration

The dosing and administration of Afatinib are standardized to optimize efficacy while allowing for modifications to manage toxicity.

  • Standard Recommended Dose: The standard starting dosage is 40 mg taken orally once daily. Treatment should continue until disease progression or unacceptable toxicity.[5]
  • Administration Instructions: Afatinib must be taken on an empty stomach. Patients should be instructed to take their dose at least 1 hour before a meal or 2 hours after a meal. Tablets should be swallowed whole with water.[3] For patients with difficulty swallowing, the tablet may be dispersed in approximately 100 mL of non-carbonated water.[40]
  • Missed Dose: If a dose is missed, it should be taken as soon as remembered, unless the next scheduled dose is due within 12 hours. Patients should not take a double dose to make up for a missed one.[5]

Dosage Modifications for Adverse Reactions

A structured dose reduction strategy is essential for managing AEs and maintaining patients on therapy.

  • Dose Reduction Schedule: The dose can be reduced in 10 mg increments, from 40 mg to 30 mg, and then to a minimum of 20 mg per day.[5]
  • Withholding Therapy: Treatment should be temporarily withheld for prolonged or intolerable Grade 2 AEs (e.g., diarrhea lasting >48 hours, cutaneous reactions lasting >7 days) or for any Grade 3 or higher AE.[5]
  • Resuming Therapy: Once the AE has fully resolved or improved to Grade 1 or baseline, Afatinib can be resumed at a reduced dose (10 mg per day less than the dose at which the AE occurred).[5]
  • Permanent Discontinuation: Afatinib should be permanently discontinued for life-threatening conditions (e.g., confirmed ILD, severe blistering skin lesions, GI perforation, severe hepatic impairment) or if a severe or intolerable AE occurs at a dose of 20 mg per day.[40]

Use in Special Populations

Dosage adjustments or specific precautions are recommended for certain patient populations.

  • Renal Impairment:
  • Mild to Moderate (eGFR ≥30 mL/min/1.73 m²): No starting dose adjustment is necessary.[32]
  • Severe (eGFR 15 to 29 mL/min/1.73 m²): The recommended starting dose is reduced to 30 mg orally once daily. Patients should be monitored closely.[5]
  • End-Stage Renal Disease (eGFR <15 mL/min/1.73 m²) or Dialysis: Afatinib has not been studied in this population, and no dosing recommendation is available.[40]
  • Hepatic Impairment:
  • Mild (Child-Pugh A) to Moderate (Child-Pugh B): No starting dose adjustment is necessary.[40]
  • Severe (Child-Pugh C): Afatinib has not been studied in this population. Patients should be monitored closely, and the dose should be adjusted if not tolerated.[37]
  • Pregnancy and Lactation:
  • Pregnancy: Afatinib is designated as Pregnancy Category D and can cause fetal harm. Females of reproductive potential must be advised of the risk and should use effective contraception during treatment and for at least 2 weeks after the final dose.[4]
  • Lactation: It is not known if Afatinib is excreted in human milk, but it is present at high concentrations in the milk of lactating rats. Breastfeeding is not recommended during treatment with Afatinib.[28]
  • Pediatric Use: The safety and efficacy of Afatinib have not been established in patients younger than 18 years. Its use is not recommended in children or adolescents.[32]
  • Other Factors: Clinical studies have shown that female patients and those with lower body weight tend to have higher exposure to Afatinib. This may increase their risk of developing AEs, particularly diarrhea, rash, and stomatitis. Closer monitoring is recommended for these patients.[31]

Table 5: Recommended Dosage Adjustments

ConditionRecommended ActionSource(s)
Grade ≥3 Adverse ReactionWithhold therapy. When reaction resolves to ≤Grade 1, resume at a dose reduced by 10 mg/day.5
Prolonged/Intolerable Grade 2 AEWithhold therapy. When reaction resolves to ≤Grade 1, resume at a dose reduced by 10 mg/day.5
Severe or Intolerable AE at 20 mg/dayPermanently discontinue.40
Severe Renal Impairment (eGFR 15-29 mL/min)Reduce starting dose to 30 mg once daily.5
Concomitant Chronic P-gp InhibitorReduce daily dose by 10 mg if not tolerated.5
Concomitant Chronic P-gp InducerIncrease daily dose by 10 mg as tolerated.5

Drug-Drug Interactions

The potential for drug-drug interactions (DDIs) with Afatinib is primarily driven by its interaction with drug transporters, rather than metabolic enzymes.

P-glycoprotein (P-gp) Pathway Interactions

Afatinib is a substrate of the P-glycoprotein (P-gp, also known as ABCB1) efflux transporter, which is found in the intestines, liver, and other tissues and plays a key role in limiting drug absorption and distribution.[6] Consequently, its plasma concentrations are highly susceptible to modulation by concomitant medications that inhibit or induce P-gp activity.

  • P-gp Inhibitors: Co-administration of Afatinib with potent P-gp inhibitors (e.g., ritonavir, cyclosporine A, ketoconazole, itraconazole, verapamil, amiodarone) can increase Afatinib exposure and the risk of toxicity.[6] For patients requiring chronic therapy with a P-gp inhibitor, the daily dose of Afatinib should be reduced by 10 mg if the standard dose is not tolerated. The original dose can be resumed after the P-gp inhibitor is discontinued.[5] It is recommended to administer P-gp inhibitors with staggered dosing, separated from the Afatinib dose by several hours.[32]
  • P-gp Inducers: Co-administration with potent P-gp inducers (e.g., rifampicin, carbamazepine, phenytoin, phenobarbital, St. John's wort) can decrease Afatinib plasma concentrations, potentially reducing its efficacy.[6] For patients requiring chronic therapy with a P-gp inducer, the daily dose of Afatinib may be increased by 10 mg as tolerated. The original dose should be resumed 2 to 3 days after the P-gp inducer is discontinued.[40]

Cytochrome P450 (CYP) System

Afatinib has a very low potential for clinically relevant DDIs mediated by the CYP450 enzyme system. Pharmacokinetic studies have shown that enzyme-catalyzed metabolism is a negligible pathway for Afatinib clearance, and the drug is not a significant inhibitor or inducer of major CYP enzymes.[6] This is a notable clinical advantage, as it simplifies the management of co-medications compared to drugs that are heavily metabolized by or interact with the CYP system.

Interactions with Other Transporters

  • Breast Cancer Resistance Protein (BCRP): In addition to P-gp, Afatinib is also a substrate of BCRP. Therefore, potent BCRP inhibitors could also potentially increase Afatinib exposure.[32]
  • Effect of Afatinib on Other Substrates: Afatinib itself is a moderate inhibitor of P-gp and BCRP in vitro. Clinically, it may increase the bioavailability of other orally administered substrates of these transporters, such as rosuvastatin (a BCRP substrate).[32] Caution is advised when co-administering Afatinib with sensitive P-gp or BCRP substrates.

Pharmacodynamic Interactions

There is a potential for additive or synergistic pharmacodynamic effects when Afatinib is combined with other agents that target the ErbB signaling pathway. For instance, combination therapy with the anti-EGFR monoclonal antibody Cetuximab has been explored in clinical trials for patients with acquired resistance to TKIs, with promising activity observed.[30]

Regulatory and Development History

Developer and Clinical Program

Afatinib was discovered and developed by the global pharmaceutical company Boehringer Ingelheim.[2] It marked a significant entry for the company into the field of oncology, becoming its first FDA-approved oncology product.[17]

The foundation for Afatinib's regulatory submissions was the comprehensive LUX-Lung clinical trial program. This extensive series of studies, which included eight major trials enrolling over 3,760 patients worldwide, was designed to investigate the efficacy and safety of Afatinib across various NSCLC patient populations and treatment settings.[11] A key component of the development strategy was the parallel development of a companion diagnostic test. Boehringer Ingelheim collaborated with QIAGEN to develop the therascreen® EGFR RGQ PCR Kit, which was approved by the FDA alongside Afatinib to identify patients with the specific EGFR mutations eligible for treatment.[33]

Regulatory Approval Timeline

Afatinib received approvals from major regulatory agencies worldwide based on the robust data from the LUX-Lung program. The regulatory journey reflects a strategic, multi-phase approach to establish its role in cancer therapy.

  • July 12, 2013 (U.S. FDA): The FDA granted its initial approval for Gilotrif (afatinib) for the first-line treatment of patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitution mutations, as detected by an FDA-approved test. This approval was primarily based on the data from the LUX-Lung 3 trial.[15]
  • September 25, 2013 (European Medicines Agency - EMA): The European Commission granted a marketing authorization for Giotrif (afatinib) for the monotherapy of EGFR TKI-naïve adult patients with locally advanced or metastatic NSCLC with activating EGFR mutation(s). The positive opinion from the Committee for Medicinal Products for Human Use (CHMP) was based on the LUX-Lung 3 data.[9]
  • April 15, 2016 (U.S. FDA): The FDA expanded Afatinib's indication to include the treatment of patients with metastatic, squamous NSCLC progressing after platinum-based chemotherapy. This approval was based on the superior OS demonstrated in the LUX-Lung 8 trial compared to erlotinib.[12]
  • March 31, 2016 (EMA): The EMA extended the indication for Giotrif to include the treatment of patients with locally advanced or metastatic NSCLC of squamous histology progressing on or after platinum-based chemotherapy, also based on the LUX-Lung 8 results.[36]
  • January 12, 2018 (U.S. FDA): The FDA further broadened the first-line indication for Afatinib to include patients with metastatic NSCLC whose tumors have other non-resistant EGFR mutations (e.g., G719X, L861Q, S768I). This expansion was based on pooled analyses of data from the LUX-Lung 2, 3, and 6 trials.[15]

The regulatory and post-marketing strategy for Afatinib illustrates a sophisticated approach to life-cycle management. The initial, tightly focused approval in a biomarker-defined population de-risked the launch. This was followed by a classic indication expansion into squamous NSCLC based on head-to-head superiority data. Finally, the strategy evolved into indication refinement (broadening to uncommon mutations) and the generation of real-world evidence (e.g., the GioTag study) to inform clinical practice and defend its position in a competitive and evolving treatment landscape.[35] This demonstrates a long-term commitment to maximizing the drug's clinical value.

Table 6: Key Regulatory Milestones (FDA & EMA)

DateRegulatory AgencyActionBasis for Action
July 12, 2013U.S. FDAInitial Approval1L treatment of metastatic NSCLC with common EGFR mutations (del19/L858R). Based on LUX-Lung 3.
Sept 25, 2013EMAInitial Marketing Authorization1L treatment of EGFR TKI-naïve NSCLC with activating EGFR mutations. Based on LUX-Lung 3.
April 15, 2016U.S. FDAIndication ExpansionTreatment of metastatic squamous NSCLC progressing after platinum chemotherapy. Based on LUX-Lung 8.
March 31, 2016EMAIndication ExtensionTreatment of locally advanced/metastatic squamous NSCLC progressing after platinum chemotherapy. Based on LUX-Lung 8.
Jan 12, 2018U.S. FDAIndication Broadening1L treatment of metastatic NSCLC with other non-resistant EGFR mutations (e.g., G719X, L861Q, S768I). Based on LUX-Lung 2, 3, & 6.

Clinical Considerations and Practice Implications

Patient Selection and Biomarker Testing

The use of Afatinib as a first-line therapy for NSCLC is strictly contingent upon the patient's tumor biomarker status. It is imperative that an FDA-approved or otherwise validated test is used to determine the presence of EGFR mutations in a tumor specimen prior to initiating treatment.[4] This precision medicine approach ensures that the therapy is directed only to those patients who are likely to benefit.

Furthermore, a detailed understanding of the specific EGFR mutation is clinically valuable. As demonstrated by the LUX-Lung trials, the OS benefit of Afatinib is most pronounced in patients with the del19 mutation.[1] For patients with less common mutations like G719X, L861Q, or S768I, Afatinib is a key evidence-based option, supported by both the expanded FDA label and extensive pooled analyses where data for other TKIs may be limited.[1]

Positioning in the Treatment Landscape

The choice of a first-line TKI for EGFR M+ NSCLC is a complex clinical decision influenced by mutation type, efficacy data, and toxicity profiles.

  • For Del19-Mutant NSCLC: Afatinib's proven OS benefit over chemotherapy makes it a very strong consideration in this subgroup.[11]
  • For L858R-Mutant NSCLC: In the absence of a proven OS benefit for Afatinib in this subgroup, the treatment decision may be more heavily influenced by factors such as PFS and comparative tolerability. Third-generation TKIs, with their more favorable safety profiles, are often preferred.
  • For Uncommon Mutations: Afatinib holds a critical niche for patients with major uncommon mutations, where it has the most robust supporting data.[34]
  • The Sequencing Strategy: The findings from the GioTag real-world study have introduced a compelling new paradigm. The "Afatinib followed by Osimertinib" sequence, which yielded a median OS of nearly four years, presents a viable long-term strategy that challenges the default use of a third-generation TKI first.[38] This approach leverages the distinct mechanisms and resistance patterns of different TKI generations to potentially maximize the duration of disease control.

Optimizing Therapeutic Outcomes

The clinical success of Afatinib therapy is inseparable from the effective management of its side effects. The high incidence and potential severity of AEs like diarrhea and rash mean that proactive toxicity management is not an ancillary task but a core component of the treatment itself. Clinicians must prioritize comprehensive patient education before starting therapy, ensuring patients understand the likelihood of these AEs, can recognize them early, and are equipped with supportive medications (e.g., loperamide) and clear instructions for their use. Prompt implementation of dose modifications according to established guidelines is crucial for mitigating toxicity, maintaining dose intensity, and enabling patients to continue benefiting from the therapy for as long as possible.

Future Perspectives

The future role of Afatinib will likely be in well-defined clinical niches. It will remain a primary treatment option for NSCLC harboring specific uncommon EGFR mutations and a strong consideration for del19-positive tumors. Its place in sequential therapy strategies will continue to be explored and refined. Further research may focus on novel combination therapies, potentially pairing Afatinib's potent ErbB blockade with immunotherapy or other targeted agents to overcome resistance and further improve patient outcomes. Its activity in other tumor types, such as HER2-driven breast cancer and head and neck cancer, remains an area of ongoing investigation that could lead to future indication expansions.[1]

Conclusion

Afatinib (DB08916) is a potent, second-generation, irreversible ErbB family blocker that has secured a definitive role in the treatment of specific subsets of non-small cell lung cancer. Its unique pharmacological profile, characterized by covalent binding and broad-spectrum inhibition of EGFR, HER2, and HER4, provides a durable blockade of key oncogenic signaling pathways. This mechanism underpins its proven clinical efficacy, most notably the significant progression-free survival benefit over chemotherapy in first-line EGFR M+ NSCLC and the overall survival advantage in patients with del19 mutations and in the second-line treatment of squamous NSCLC.

However, the drug's potent, non-selective mechanism is also the direct cause of its challenging but manageable safety profile, dominated by high rates of diarrhea and rash. The successful use of Afatinib in the clinic is therefore a testament to a treatment paradigm that integrates proactive toxicity management and dose modification as essential components of care.

Developed by Boehringer Ingelheim, Afatinib's regulatory and post-marketing history demonstrates a strategic approach to establishing its value, from initial biomarker-driven approvals to indication expansions and the generation of critical real-world evidence for sequential therapy. As the landscape of targeted oncology therapies continues to evolve, Afatinib remains a vital therapeutic option, particularly for patients with uncommon EGFR mutations and as a key component in long-term treatment strategies designed to maximize patient survival.

Works cited

  1. en.wikipedia.org, accessed July 15, 2025, https://en.wikipedia.org/wiki/Afatinib
  2. Boehringer Ingelheim - Afatinib - AdisInsight - Springer, accessed July 15, 2025, https://adisinsight.springer.com/drugs/800023647
  3. Afatinib: MedlinePlus Drug Information, accessed July 15, 2025, https://medlineplus.gov/druginfo/meds/a613044.html
  4. Learn About Certain Types of NSCLC | GILOTRIF® (afatinib) tablets - Boehringer Ingelheim, accessed July 15, 2025, https://patient.boehringer-ingelheim.com/us/products/gilotrif/
  5. HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights ..., accessed July 15, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/201292s017lbl.pdf
  6. Clinical Pharmacokinetics and Pharmacodynamics of Afatinib - PMC, accessed July 15, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5315738/
  7. DRUG NAME: Afatinib - BC Cancer, accessed July 15, 2025, http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Afatinib_monograph.pdf
  8. Afatinib | Marnett Research Laboratory - Vanderbilt University, accessed July 15, 2025, https://lab.vanderbilt.edu/marnett-lab/afatinib/
  9. Afatinib for patients with EGFR mutation positive lung cancer - Hospital Pharmacy Europe, accessed July 15, 2025, https://hospitalpharmacyeurope.com/clinical-zones/oncology/afatinib-for-patients-with-egfr-mutation-positive-lung-cancer/
  10. Boehringer Ingelheim Presents Progression-Free Survival Data for Investigational Afatinib and Nintedanib in Advanced NSCLC - PR Newswire, accessed July 15, 2025, https://www.prnewswire.com/news-releases/boehringer-ingelheim-presents-progression-free-survival-data-for-investigational-afatinib-and-nintedanib-in-advanced-nsclc-207613401.html
  11. AFATINIB* - Boehringer Ingelheim, accessed July 15, 2025, https://pro.boehringer-ingelheim.com/inoncology/sites/default/files/2022-01/afatinibbackgrounder.pdf
  12. FDA Expands Use of Lung Cancer Therapeutic | The AACR, accessed July 15, 2025, https://www.aacr.org/patients-caregivers/progress-against-cancer/fda-expands-use-lung-cancer-drug-afatinib/
  13. Practical Management of Adverse Events Associated with Afatinib - PMC, accessed July 15, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9116182/
  14. Important Safety Information | GILOTRIF® (afatinib) tablets - Boehringer Ingelheim, accessed July 15, 2025, https://pro.boehringer-ingelheim.com/us/products/gilotrif/important-safety-information
  15. Gilotrif (afatinib) FDA Approval History - Drugs.com, accessed July 15, 2025, https://www.drugs.com/history/gilotrif.html
  16. Giotrif | European Medicines Agency (EMA), accessed July 15, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/giotrif
  17. Afatinib | C24H25ClFN5O3 | CID 10184653 - PubChem, accessed July 15, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/10184653
  18. Gilotrif (afatinib): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing - WebMD, accessed July 15, 2025, https://www.webmd.com/drugs/2/drug-164950/gilotrif-oral/details
  19. afatinib | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed July 15, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=5667
  20. Afatinib (BIBW 2992, CAS Number: 850140-72-6) | Cayman Chemical, accessed July 15, 2025, https://www.caymanchem.com/product/11492/afatinib
  21. Afatinib ≥98% (HPLC) - Sigma-Aldrich, accessed July 15, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml3109
  22. Afatinib dimaleate: Chemical properties, Indications, Mechanism of action and Side effects, accessed July 15, 2025, https://www.chemicalbook.com/article/afatinib-dimaleate-chemical-properties-indications-mechanism-of-action-and-side-effects.htm
  23. afatinib - PharmGKB, accessed July 15, 2025, https://www.pharmgkb.org/chemical/PA165981154
  24. Afatinib dimaleate | DrugBank Online, accessed July 15, 2025, https://go.drugbank.com/salts/DBSALT000005
  25. Afatinib Dimaleate | C32H33ClFN5O11 | CID 15606394 - PubChem, accessed July 15, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Afatinib-Dimaleate
  26. Afatinib dimaleate | EGFR - Tocris Bioscience, accessed July 15, 2025, https://www.tocris.com/products/afatinib-dimaleate_6812
  27. Afatinib - StatPearls - NCBI Bookshelf, accessed July 15, 2025, https://www.ncbi.nlm.nih.gov/books/NBK542248/
  28. 201292Orig1s000 - accessdata.fda.gov, accessed July 15, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/201292orig1s000pharmr.pdf
  29. Afatinib (BIBW 2992), Her2/ErbB 2 and EGFR kinase inhibitor - Abcam, accessed July 15, 2025, https://www.abcam.com/en-us/products/biochemicals/afatinib-bibw-2992-her2-erbb-2-and-egfr-kinase-inhibitor-ab142018
  30. Afatinib—new therapy option for EGFR-mutant lung cancer - PMC, accessed July 15, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4665630/
  31. Clinical Pharmacokinetics and Pharmacodynamics of Afatinib - PubMed, accessed July 15, 2025, https://pubmed.ncbi.nlm.nih.gov/27470518/
  32. Giotrif, INN-afatinib - EMA, accessed July 15, 2025, https://www.ema.europa.eu/en/documents/product-information/giotrif-epar-product-information_en.pdf
  33. FDA approves GILOTRIF™ (afatinib) as first-line treatment for metastatic non-small cell lung cancer with common EGFR mutations - PR Newswire, accessed July 15, 2025, https://www.prnewswire.com/news-releases/fda-approves-gilotrif-afatinib-as-first-line-treatment-for-metastatic-non-small-cell-lung-cancer-with-common-egfr-mutations-215313431.html
  34. Afatinib for the Treatment of Non-Small Cell Lung Cancer Harboring Uncommon EGFR Mutations: An Updated Database of 1023 Cases Brief Report - PubMed Central, accessed July 15, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9104339/
  35. New uncommon EGFR mutations database published featuring clinical outcomes of NSCLC patients treated with afatinib - Boehringer Ingelheim, accessed July 15, 2025, https://www.boehringer-ingelheim.com/human-health/cancer/lung-cancer/uncommonmutationsdatabase
  36. Afatinib (Gilotrif) | HemOnc.org - A Hematology Oncology Wiki, accessed July 15, 2025, https://hemonc.org/wiki/Afatinib_(Gilotrif)
  37. FDA and EMA accept regulatory applications for Boehringer Ingelheim's Gilotrif® (afatinib) for treatment of advanced squamous cell carcinoma of the lung - PR Newswire, accessed July 15, 2025, https://www.prnewswire.com/news-releases/fda-and-ema-accept-regulatory-applications-for-boehringer-ingelheims-gilotrif-afatinib-for-treatment-of-advanced-squamous-cell-carcinoma-of-the-lung-300132550.html
  38. Real World Evidence | GILOTRIF® (afatinib) tablets - Boehringer Ingelheim, accessed July 15, 2025, https://pro.boehringer-ingelheim.com/us/products/gilotrif/real-world-evidence
  39. Additional analysis of real-world data confirms sequential Gilotrif® followed by osimertinib provided a median overall survival of nearly four years in U.S.-treated patients with EGFR mutation-positive NSCLC - Boehringer Ingelheim, accessed July 15, 2025, https://www.boehringer-ingelheim.com/us/media/press-releases/additional-analysis-real-world-data-confirms-sequential-gilotrif-followed-osimertinib-provided
  40. Afatinib Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 15, 2025, https://www.drugs.com/dosage/afatinib.html
  41. Gilotrif Dosage Guide - Drugs.com, accessed July 15, 2025, https://www.drugs.com/dosage/gilotrif.html
  42. Showing BioInteractions for Afatinib (DB08916) | DrugBank Online, accessed July 15, 2025, https://go.drugbank.com/drugs/DB08916/biointeractions
  43. Afatinib Completed Phase 1 Trials for Neoplasms Treatment | DrugBank Online, accessed July 15, 2025, https://go.drugbank.com/drugs/DB08916/clinical_trials?conditions=DBCOND0027948&phase=1&purpose=treatment&status=completed
  44. Boehringer Ingelheim Release: European Committee For Medicinal Products For Human Use Recommends Approval Of Giotrif (Afatinib) For Advanced Squamous Cell Cancer Of The Lung - BioSpace, accessed July 15, 2025, https://www.biospace.com/boehringer-ingelheim-release-european-committee-for-medicinal-products-for-human-use-recommends-approval-of-giotrif-afatinib-for-advanced-squamous
  45. Boehringer's Giotrif wins new NSCLC licence in Europe - PMLiVE, accessed July 15, 2025, https://pmlive.com/pharma_news/boehringers_giotrif_wins_new_nsclc_licence_in_europe_972872/
  46. FDA broadens afatinib indication to previously untreated, metastatic, accessed July 15, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-broadens-afatinib-indication-previously-untreated-metastatic-nsclc-other-non-resistant-egfr

Published at: July 15, 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.