MedPath

Binimetinib Advanced Drug Monograph

Published:Jun 5, 2025

Generic Name

Binimetinib

Brand Names

Mektovi

Drug Type

Small Molecule

Chemical Formula

C17H15BrF2N4O3

CAS Number

606143-89-9

Associated Conditions

Metastatic Melanoma, Metastatic Non-Small Cell Lung Cancer, Unresectable Melanoma

Binimetinib: A Comprehensive Review of its Pharmacology, Clinical Efficacy, and Safety

1. Introduction to Binimetinib

1.1. Overview, Chemical Properties, and DrugBank Identification

Binimetinib, identified by DrugBank ID DB11967 and CAS Number 606143-89-9, is an orally administered small molecule kinase inhibitor.[1] It is also marketed under the brand name Mektovi® and has been referred to by investigational codes such as ARRY-162, MEK162, and ARRY-438162.[2]

Chemically, binimetinib belongs to the class of benzimidazoles. Its structure is specifically 1-methyl-1H-benzimidazole substituted at positions 4, 5, and 6 by fluorine, (4-bromo-2-fluorophenyl)nitrilo, and N-(2-hydroxyethoxy)aminocarbonyl groups, respectively.[1] The molecular formula for binimetinib is C17​H15​BrF2​N5​O3​, and it has a molecular weight of approximately 441.2 g/mol.[1] The complex heterocyclic structure, featuring a benzimidazole core common in kinase inhibitors, along with fluorine and bromine substitutions (which can modulate pharmacokinetic properties like lipophilicity and metabolic stability) and an N-(2-hydroxyethoxy)aminocarbonyl group (potentially involved in target binding), underpins its specific and potent biological activity.[1]

Table 1: Key Characteristics of Binimetinib

CharacteristicDetail
DrugBank IDDB11967
CAS Number606143-89-9
Common SynonymsMektovi, ARRY-162, MEK162, ARRY-438162
TypeSmall Molecule
Molecular FormulaC17​H15​BrF2​N5​O3​
Molecular Weight~441.2 g/mol
Therapeutic ClassKinase Inhibitor, MEK Inhibitor
Key Approved Indications- Unresectable or metastatic melanoma with BRAF V600E or V600K mutations (in combination with encorafenib) <br> - Metastatic non-small cell lung cancer (NSCLC) with BRAF V600E mutation (in combination with encorafenib)

Sources: User Query,.[1]

1.2. Therapeutic Class and General Use

Binimetinib is classified as an antineoplastic agent, functioning as an inhibitor of Mitogen-activated protein kinase kinase 1 and 2 (MEK1/2).[1] Its primary clinical application is in combination with encorafenib, a BRAF inhibitor. This combination is approved for the treatment of patients with unresectable or metastatic melanoma harboring BRAF V600E or V600K mutations. More recently, this combination has also been approved for adult patients with metastatic non-small cell lung cancer (NSCLC) that has a BRAF V600E mutation. The presence of these specific mutations must be confirmed by an FDA-approved diagnostic test prior to initiating therapy.[1]

The rationale for combining binimetinib with a BRAF inhibitor stems from the understanding of MAPK pathway signaling and resistance mechanisms. The MAPK pathway, when constitutively activated by BRAF mutations, drives tumor growth. While BRAF inhibitors are effective, resistance often develops through various mechanisms, including reactivation of the MAPK pathway downstream of BRAF or through feedback loops. By inhibiting MEK, a kinase downstream of BRAF, binimetinib provides a dual blockade of the pathway. This approach has been shown to be more effective than BRAF inhibitor monotherapy in delaying resistance and improving clinical outcomes, as demonstrated in pivotal trials such as the COLUMBUS study.[9]

2. Mechanism of Action

2.1. Targeting the MAPK/ERK Pathway

The Mitogen-Activated Protein Kinase (MAPK)/Extracellular signal-Regulated Kinase (ERK) pathway is a fundamental intracellular signaling cascade. It plays a crucial role in transducing extracellular signals to the nucleus, thereby regulating a wide array of cellular processes including proliferation, differentiation, survival, migration, and angiogenesis.[4] In normal physiology, this pathway is tightly regulated. However, in many human cancers, the MAPK/ERK pathway is aberrantly activated due to mutations in its upstream components, most commonly in RAS or BRAF oncogenes.[4] This constitutive activation leads to uncontrolled cell growth, resistance to apoptosis, and ultimately, malignant transformation and progression. Given its central role in driving oncogenesis in numerous tumor types, the MAPK/ERK pathway is a well-established and critical target for anticancer therapies.

MEK1 and MEK2 (Mitogen-activated protein kinase kinase 1 and 2, also known as MAP2K1 and MAP2K2) are highly specific dual-specificity kinases that occupy a pivotal position within this pathway. They are directly phosphorylated and activated by RAF kinases (ARAF, BRAF, CRAF) and, in turn, phosphorylate and activate ERK1 and ERK2 (also known as MAPK3 and MAPK1).[1] Activated ERK then translocates to the nucleus to regulate the activity of numerous transcription factors, leading to changes in gene expression that drive the cancer phenotype.

2.2. Role as a MEK1/2 Inhibitor

Binimetinib functions as a potent, selective, orally bioavailable, and reversible inhibitor of the kinase activity of MEK1 and MEK2.[1] A distinguishing feature of binimetinib is its mechanism of inhibition; it is an ATP-noncompetitive, allosteric inhibitor.[1] This means that binimetinib binds to a site on the MEK enzymes distinct from the ATP-binding pocket. This allosteric binding induces a conformational change in MEK1/2 that prevents their catalytic activity, thereby blocking the phosphorylation and subsequent activation of their downstream substrates, ERK1 and ERK2.[1] Allosteric inhibition can offer advantages in terms of higher selectivity for the target kinase over other ATP-utilizing enzymes and may also be effective against kinases that have developed resistance to ATP-competitive inhibitors.

The inhibition of ERK activation by binimetinib effectively abrogates the downstream signaling cascade. This leads to a reduction in tumor cell proliferation, an increase in apoptosis (programmed cell death), and an inhibition of the production of various inflammatory cytokines, such as interleukin-1 (IL-1), IL-6, and tumor necrosis factor (TNF), which can contribute to the tumor microenvironment and cancer-related symptoms.[1] The half-maximal inhibitory concentration (IC50​) of binimetinib for MEK1/2 is approximately 12 nM, indicating its high potency.[1] By targeting MEK1/2, binimetinib effectively shuts down a critical node in the hyperactive MAPK pathway that drives the growth and survival of cancer cells, particularly those with activating BRAF mutations.

3. Pharmacology

3.1. Pharmacodynamics

The pharmacodynamic effects of binimetinib confirm its mechanism of action and on-target activity. In vitro, binimetinib has been shown to effectively inhibit the phosphorylation of ERK in cell-free assays. Furthermore, it demonstrates cytotoxic activity by reducing the viability of human melanoma cell lines that harbor BRAF mutations.[3] These preclinical findings are corroborated by in vivo studies, where binimetinib inhibited ERK phosphorylation and suppressed tumor growth in murine xenograft models derived from BRAF-mutant human melanomas.[3]

Clinical pharmacodynamic assessments have further validated these effects in patients. Studies involving patients with advanced solid tumors have demonstrated target inhibition, as evidenced by reduced levels of phosphorylated ERK (pERK1/2) in serum samples and skin punch biopsies following binimetinib administration.[11] This direct evidence of target engagement in humans is crucial for linking the drug's mechanism to its clinical effects.

Significantly, when binimetinib is co-administered with the BRAF inhibitor encorafenib, a synergistic or enhanced anti-tumor effect is observed. In vitro studies on BRAF mutation-positive cell lines show greater anti-proliferative activity with the combination compared to either agent alone. This enhanced activity is also seen in vivo, where the combination resulted in greater tumor growth inhibition in BRAF V600E mutant human melanoma xenograft models.[12] This preclinical and clinical pharmacodynamic synergy provides a strong mechanistic basis for the improved efficacy observed with the combination therapy in clinical trials.

3.2. Pharmacokinetics

The pharmacokinetic profile of binimetinib has been characterized in healthy subjects and patients with solid tumors.

Absorption:

Binimetinib is administered orally. At least 50% of an administered dose is absorbed, with some evidence suggesting that actual absorption may be considerably higher due to the pharmacokinetic properties of the drug and the instability of its glucuronide conjugates within the gastrointestinal tract.1 The median time to reach maximum plasma concentration (Tmax) is approximately 1.6 hours.1

The administration of binimetinib is not significantly affected by food. A single 45 mg dose taken with a high-fat, high-calorie meal (approximately 150 calories from protein, 350 from carbohydrate, and 500 from fat) did not result in clinically relevant changes to binimetinib exposure (AUC and Cmax).1 Consequently, binimetinib can be administered with or without food, offering flexibility for patients.7

Following twice-daily dosing, binimetinib exhibits an accumulation of approximately 1.5-fold, and its systemic exposure (AUC) is roughly dose-proportional.1 The coefficient of variation (CV%) for AUC at steady state is less than 40%.1

Distribution:

Binimetinib has an apparent volume of distribution (Vd/F) with a geometric mean of 92 Liters (CV 45%).1 It is highly bound to human plasma proteins, with a binding percentage of 97%.12 The blood-to-plasma concentration ratio for binimetinib is 0.72.12

Metabolism:

The primary metabolic pathway for binimetinib is glucuronidation, with the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme system being the major contributor, accounting for up to 61% of its metabolism.12 Other metabolic pathways include N-dealkylation, amide hydrolysis, and the loss of an ethane-diol moiety from its side chain.12

An active metabolite, designated M3, is formed through oxidative N-desmethylation catalyzed by cytochrome P450 (CYP) enzymes CYP1A2 and CYP2C19. This active metabolite accounts for approximately 8.6% of the total binimetinib exposure.12 Following a single oral dose of 45 mg radiolabeled binimetinib, approximately 60% of the circulating radioactivity AUC in plasma was attributable to the parent compound.12

Elimination:

Binimetinib is eliminated from the body through both fecal and renal routes. After a single oral 45 mg dose of radiolabeled binimetinib in healthy subjects, approximately 62.3% of the administered radioactivity was recovered in the feces (with 32% as unchanged drug), and 31.4% was recovered in the urine (with 6.5% as unchanged drug). The overall recovery of radioactivity in excreta was high, at 93.6%.1

The mean terminal elimination half-life (t1/2​) of binimetinib is approximately 3.5 hours (CV 28.5%).12 The apparent clearance (CL/F) has a mean value of 20.2 L/h (CV 24%).1 The relatively short half-life supports the twice-daily dosing schedule required to maintain therapeutic plasma concentrations.

Special Populations:

Patient-specific factors such as age (ranging from 20 to 94 years), sex, or body weight do not appear to have a clinically significant impact on the systemic exposure of binimetinib.12 The effect of race or ethnicity on binimetinib pharmacokinetics has not been fully elucidated.12

Hepatic impairment, however, does affect binimetinib exposure. Patients with moderate (total bilirubin >1.5 and ≤3 × ULN and any AST) or severe (total bilirubin levels >3 × ULN and any AST) hepatic impairment exhibit a 2-fold increase in binimetinib AUC. This necessitates a dose reduction in these patient populations.7

Transporters:

Binimetinib is a substrate for the efflux transporters P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP).12 It is not a substrate for the uptake transporters OATP1B1, OATP1B3, OATP2B1, or OCT1.12

Table 2: Summary of Binimetinib Pharmacokinetic Parameters

ParameterValue / Description
Absorption
Oral Bioavailability≥50% absorbed; Tmax ~1.6 hours
Effect of FoodNo clinically significant effect on exposure
Accumulation (BID dosing)~1.5-fold
Distribution
Apparent Volume of Distribution (Vd​/F)92 L (Geometric Mean)
Protein Binding97%
Blood-to-Plasma Ratio0.72
Metabolism
Primary PathwayGlucuronidation (UGT1A1 accounts for ~61%)
Other PathwaysN-dealkylation, amide hydrolysis, loss of ethane-diol
Active Metabolite(s)M3 (via CYP1A2, CYP2C19); accounts for ~8.6% of exposure
Elimination
Terminal Half-life (t1/2​)~3.5 hours
Apparent Clearance (CL/F)~20.2 L/h
Excretion~62% in feces (32% unchanged); ~31% in urine (6.5% unchanged)
Impact of Hepatic ImpairmentModerate or Severe: ~2-fold increase in AUC; dose reduction required
Key Transporter Interactions (Substrate)P-gp, BCRP

Sources:.[1]

4. Clinical Efficacy in Approved Indications

4.1. Metastatic Melanoma (BRAF V600E/K Mutation) in Combination with Encorafenib

Binimetinib, in combination with encorafenib, is indicated for the treatment of patients with unresectable or metastatic melanoma characterized by a BRAF V600E or V600K mutation, as confirmed by an FDA-approved diagnostic test.[1]

The cornerstone evidence supporting this indication comes from the COLUMBUS trial (NCT01909453), a large, randomized, active-controlled, open-label, two-part Phase 3 study.[9] Part 1 of the COLUMBUS trial (N=577) was pivotal, comparing the combination of encorafenib 450 mg once daily (QD) plus binimetinib 45 mg twice daily (BID) (referred to as the COMBO arm) against two monotherapy arms: encorafenib 300 mg QD (ENCO arm) and vemurafenib 960 mg BID (VEMU arm).[9] The primary efficacy endpoint for Part 1 was progression-free survival (PFS) for the COMBO arm versus the VEMU arm, assessed by blinded independent central review.[23]

Key Efficacy Results from COLUMBUS Part 1:

Comparing the COMBO arm to vemurafenib monotherapy (VEMU):

  • Progression-Free Survival (PFS): The combination of encorafenib plus binimetinib demonstrated a statistically significant and clinically meaningful improvement in median PFS. Initial reports indicated a median PFS of 14.9 months for the COMBO arm versus 7.3 months for the VEMU arm (Hazard Ratio 0.54; 95% Confidence Interval [CI]: 0.41-0.71; P<0.001).[9]
  • Objective Response Rate (ORR): The ORR was substantially higher in the COMBO arm at 63% (including an 8% Complete Response rate) compared to 40% in the VEMU arm (including a 6% CR rate).[9]
  • Overall Survival (OS): The combination therapy also led to a significant improvement in median OS. The 5-year update of the COLUMBUS trial reported a median OS of 33.6 months for the COMBO arm versus 16.9 months for the VEMU arm (HR 0.61; 95% CI: 0.47-0.79; P<0.001).[10] The 5-year OS rate for the COMBO arm was 35%.[21]

Comparing the COMBO arm to encorafenib monotherapy (ENCO):

  • Progression-Free Survival (PFS): The median PFS was 14.9 months for the COMBO arm versus 9.6 months for the ENCO arm.[9]
  • Objective Response Rate (ORR): The ORR was 63% for the COMBO arm compared to 51% for the ENCO arm.[9]

The results of the COLUMBUS trial, initially published in The Lancet Oncology [10] and with subsequent long-term updates (e.g., 5-year data in Future Oncology [21]), firmly established the encorafenib plus binimetinib combination as a highly effective first-line treatment option for patients with BRAF V600-mutant melanoma. This regimen is now considered a new benchmark in this setting due to its superior efficacy outcomes.[9]

An important aspect of the combination therapy is its tolerability profile. The COLUMBUS trial suggested that the combination of encorafenib and binimetinib did not lead to an overall increase in toxicity compared to BRAF inhibitor monotherapy. In fact, fewer patients in the combination arm required dose reductions or treatment interruptions due to adverse events than those in the single-agent encorafenib arm.[10] The side effect profile of the combination was considered more acceptable, and side effects tended to decrease over time with continued treatment.[9] This improved tolerability, alongside superior efficacy, contributes significantly to the favorable clinical profile of the combination. While BRAF inhibitor monotherapy can lead to certain toxicities like cutaneous squamous cell carcinomas or pyrexia, the addition of a MEK inhibitor can sometimes mitigate these, although MEK inhibitor-specific toxicities (such as ocular issues or rhabdomyolysis) may emerge.

4.2. Metastatic Non-Small Cell Lung Cancer (BRAF V600E Mutation) in Combination with Encorafenib

Binimetinib, in combination with encorafenib, is also approved for the treatment of adult patients with metastatic non-small cell lung cancer (NSCLC) harboring a BRAF V600E mutation, as detected by an FDA-approved test.[5] BRAF V600E mutations occur in a small percentage of NSCLC cases but represent an actionable genomic target.

The approval for this indication was primarily based on the results of the PHAROS trial (NCT03915951), a Phase 2, single-arm, open-label, multicenter study.[24] The trial enrolled adult patients with BRAF V600E-mutated metastatic NSCLC, including cohorts of both treatment-naive and previously treated patients.[26] The primary endpoint of the PHAROS trial was the ORR as assessed by Independent Radiology Review (IRR) in the treatment-naive patient cohort.[26]

Key Efficacy Results from PHAROS:

The FDA-approved prescribing information, likely reflecting the final analysis for approval, provides the following efficacy data 7:

  • Treatment-Naive Patients:
  • ORR by IRR: 64% (95% CI: 54%-74%).
  • Median Duration of Response (DoR) by IRR: Not reached (95% CI: 20.7, Not Estimable).
  • Median PFS by IRR: 14.8 months (95% CI: 9.3, Not Estimable).
  • Previously Treated Patients (had received prior systemic therapy):
  • ORR by IRR: 50% (95% CI: 36%-64%).
  • Median DoR by IRR: 16.7 months (95% CI: 7.4, Not Estimable).
  • Median PFS by IRR: 9.3 months (95% CI: 5.5, 15.7).

Earlier reports from the PHAROS study highlighted an ORR of 75% (95% CI, 62%-85%) in treatment-naive patients (n=59) and 46% in previously treated patients (n=39).[26] One source mentioned a median PFS of 30.2 months in treatment-naive patients from an earlier analysis with a median follow-up of 33.3 months.[28] Discrepancies in reported outcomes often arise from different data cutoff dates and patient populations included in specific analyses; the FDA label data [7] generally represents the pivotal findings supporting approval.

The PHAROS trial demonstrated that the combination of encorafenib and binimetinib yields substantial and durable responses in patients with BRAF V600E-mutant NSCLC. This is particularly significant as this genomic subtype represents an unmet need, and the high response rates observed, especially in the treatment-naive setting, led to regulatory approvals based on these Phase 2 data. Such approvals are common for targeted therapies in oncogene-addicted tumors that show compelling activity.

Table 3: Efficacy Outcomes from Pivotal Clinical Trials of Binimetinib + Encorafenib

IndicationTrial (Phase)Patient PopulationTreatment Arms & DosingPrimary Endpoint(s)Key Efficacy Results (Combination Arm vs Comparator if applicable)
Unresectable or Metastatic MelanomaCOLUMBUS (Phase 3)BRAF V600E/K-mutantEncorafenib 450mg QD + Binimetinib 45mg BID (COMBO) <br> vs. <br> Vemurafenib 960mg BID (VEMU) <br> vs. <br> Encorafenib 300mg QD (ENCO)PFS (COMBO vs VEMU)COMBO vs VEMU: <br> - Median PFS: 14.9 mo vs 7.3 mo (HR 0.54; P<0.001) <br> - ORR: 63% (8% CR) vs 40% (6% CR) <br> - Median OS: 33.6 mo vs 16.9 mo (HR 0.61; P<0.001) <br><br> COMBO vs ENCO: <br> - Median PFS: 14.9 mo vs 9.6 mo <br> - ORR: 63% vs 51%
Metastatic Non-Small Cell Lung Cancer (NSCLC)PHAROS (Phase 2)BRAF V600E-mutantEncorafenib 450mg QD + Binimetinib 45mg BID (Single Arm)ORR (Treatment-Naive)Treatment-Naive (IRR): <br> - ORR: 64% <br> - Median DoR: Not Reached <br> - Median PFS: 14.8 mo <br><br> Previously Treated (IRR): <br> - ORR: 50% <br> - Median DoR: 16.7 mo <br> - Median PFS: 9.3 mo

Sources:.[7] QD=once daily, BID=twice daily, CR=complete response, HR=hazard ratio, IRR=Independent Radiology Review, mo=months, ORR=objective response rate, OS=overall survival, PFS=progression-free survival.

5. Investigational Uses and Clinical Data in Other Malignancies

5.1. NRAS-Mutated Cancers (excluding melanoma) - NCI-MATCH Subprotocol Z1A (NCT02465060)

The National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) trial included a subprotocol (Z1A) evaluating single-agent binimetinib (45 mg BID) in patients with various advanced solid tumors (excluding melanoma) harboring activating mutations in NRAS codons 12, 13, or 61.[30] Melanoma was excluded as binimetinib has been extensively studied in that population.

The primary endpoint of this subprotocol, an objective response rate (ORR) indicative of promising activity, was not met. The observed ORR was only 2.1% (1 confirmed partial response out of 47 evaluable patients), with a 90% CI of 0.1–9.7%. The single confirmed PR occurred in a patient with NRAS Q61R-mutated malignant ameloblastoma, who remained on treatment for 26 months.[30] An unconfirmed PR was noted in a colorectal cancer (CRC) patient with an NRAS Q61R mutation. The median PFS for the overall efficacy population was 3.5 months, and the 6-month PFS rate was 29.2%. The median OS was 10.5 months.[30]

Interestingly, a post-hoc analysis suggested potential differential activity based on the specific NRAS mutation codon. Patients with tumors harboring NRAS codon 61 mutations, particularly those with colorectal cancer, exhibited significantly longer OS and PFS compared to patients whose tumors had NRAS codon 12 or 13 mutations. For all tumor types combined, the median OS for codon 61 NRAS-mutated tumors was 13.1 months versus 5.5 months for codon 12/13 mutations (p=0.04), and median PFS was 5.8 months versus 1.8 months, respectively (p=0.006). However, when CRC patients were excluded from this analysis, the survival differences by codon were no longer statistically significant.[30]

Regarding safety, toxicities were consistent with the known profile of MEK inhibitors. Dose reductions were required in 44% of patients, and 30% of patients discontinued treatment due to adverse events. One death (multiorgan failure) was considered possibly related to binimetinib.[30]

These findings indicate that binimetinib monotherapy has limited efficacy across a broad range of NRAS-mutated cancers. The observation of potential codon-specific sensitivity, especially the better outcomes in NRAS codon 61 mutated tumors (driven largely by CRC cases in this cohort), is an important finding. It suggests that not all NRAS mutations confer the same biological characteristics or sensitivity to MEK inhibition alone, and that further research is needed to understand these differences and potentially refine patient selection for MEK-targeted strategies in NRAS-mutant settings.

5.2. Advanced Colorectal Cancer (RAS-mutant) - MErCuRIC trial (NCT02510001)

The MErCuRIC trial was a Phase 1b, open-label, single-arm study designed to evaluate the combination of binimetinib with crizotinib (a MET inhibitor) in patients with advanced RAS-mutant colorectal cancer (CRC).[31] The rationale was based on preclinical evidence suggesting potential synergy between MEK and MET pathway inhibition in RAS-driven CRC.

Unfortunately, the combination did not demonstrate clinical efficacy. Among 29 evaluable patients in the dose expansion phase, no objective responses were observed. Seven patients (24%) achieved disease stabilization as their best response. The median PFS on treatment was short at 1.81 months, and the median OS was 5.62 months.[31]

The combination of binimetinib and crizotinib was also poorly tolerated. The maximum tolerated dose (MTD) was established as binimetinib 30 mg BID (administered on days 1–21 of a 28-day cycle) plus crizotinib 250 mg once daily (OD) continuously. Dose-limiting toxicities (DLTs) included Grade ≥3 transaminitis (elevated ALT/AST), Grade ≥3 creatinine phosphokinase (CPK) increases, and Grade 3 fatigue. Drug-related adverse events were frequent, with common Grade ≥3 events including CPK and ALT/AST elevations, vomiting, rash, edema, and decreased left ventricular ejection fraction (LVEF).[31]

The MErCuRIC trial underscores the significant challenges in developing effective targeted therapy combinations for RAS-mutant CRC. The poor tolerability, likely due to overlapping toxicities of the two agents, limited the ability to administer both drugs at optimal continuous doses, which may have compromised potential efficacy. An exploratory analysis did suggest that patients with a high baseline RAS-mutant allele frequency in circulating tumor DNA (ctDNA) had a significantly shorter median OS, pointing to a potential prognostic or predictive biomarker that warrants further investigation in this difficult-to-treat patient population.[31]

5.3. Other KRAS-Mutant Tumors

Binimetinib has also been investigated in other contexts of KRAS-mutant cancers. A Phase 2 clinical trial (NCT04735068) evaluated the combination of binimetinib and hydroxychloroquine in patients with advanced KRAS-mutant non-small cell lung cancer.[32] Hydroxychloroquine is an inhibitor of autophagy, a cellular recycling process that cancer cells can utilize as a survival mechanism when under therapeutic stress from agents like MEK inhibitors. The strategy of combining MEK inhibition with autophagy inhibition aims to block this potential escape route and enhance anti-tumor activity. The specific results from this completed trial were not detailed in the provided materials.

6. Ongoing Clinical Development and Future Directions

6.1. BRAF Fusion-Positive Low-Grade Glioma (LGG) and Pancreatic Cancer (PC) - Perfume Trial (NCCH2101/MK011; NCT06159478; jRCT2031230007)

The Perfume trial is an ongoing Phase 2, open-label, parallel, two-cohort, multicenter, investigator-initiated registration-directed clinical trial evaluating the efficacy and safety of binimetinib in patients with advanced or recurrent low-grade glioma (LGG; WHO Grade 1 and 2) or pancreatic cancer (PC) that harbors a BRAF fusion or rearrangement.[33] BRAF fusions, as opposed to the more common V600 point mutations, represent another class of oncogenic alterations that lead to constitutive activation of the MAPK pathway. MEK inhibitors have shown preclinical activity against BRAF fusion-positive cell lines, and early clinical trials with MEK inhibitors (including binimetinib and selumetinib) have suggested efficacy in patients with BRAF fusion-positive LGG.[33]

Patients enrolled in the Perfume trial (age ≥12 years) receive binimetinib at a dose of 45 mg BID. Key eligibility for LGG includes a Karnofsky/Lansky Performance Status (KPS/LPS) ≥70, regardless of prior cancer drug therapy. For PC, patients must have an ECOG PS of 0-1 and be refractory or intolerant to at least one prior systemic therapy.[33] The primary endpoint is the objective response rate (ORR) based on RECIST 1.1 criteria, as assessed by independent central review. Secondary endpoints include investigator-assessed ORR, ORR by RANO criteria in LGG, progression-free survival, overall survival, disease control rate, duration of response, and safety.[33]

Enrollment for the Perfume trial began in March 2023 and is ongoing at six facilities in Japan. As of December 2024, six patients with LGG and three patients with PC had been enrolled.[33] This trial is significant as it aims to expand the utility of MEK inhibitors like binimetinib to cancers driven by non-V600 BRAF alterations, specifically fusions, which are prevalent in certain rare cancers like pilocytic astrocytoma (a type of LGG) and pancreatic acinar cell carcinoma. Success in this trial could provide a much-needed targeted therapy option for these patient populations. The study also incorporates a decentralized clinical trial system to reduce the burden on patients living in remote areas.[33]

6.2. Melanoma with Brain Metastases

Brain metastases are a frequent and prognostically unfavorable complication in patients with advanced melanoma, representing a significant unmet medical need. Several clinical trials are investigating the role of binimetinib in combination with encorafenib in this challenging patient population.

A recruiting Phase 2 trial (NCT06887088) is evaluating a sequential treatment strategy: encorafenib plus binimetinib followed by the immunotherapy combination of cemiplimab (anti-PD-1) and fianlimab (anti-LAG-3) in patients with BRAF-mutant melanoma who have symptomatic brain metastases.[35] Another Phase 2 trial, sponsored by M.D. Anderson Cancer Center, is specifically assessing the efficacy of binimetinib plus encorafenib in patients with melanoma (harboring a BRAFV600 mutation) that has metastasized to the brain.[20] These studies are crucial for determining the activity of BRAF/MEK inhibition within the central nervous system (CNS) and how best to integrate targeted therapies with immunotherapies for patients with CNS involvement. The ability of these oral agents to penetrate the blood-brain barrier and elicit responses in intracranial lesions is a key area of investigation.

7. Safety Profile and Tolerability (when used with Encorafenib)

The safety profile of binimetinib is primarily characterized from its use in combination with encorafenib. The following adverse reactions (ARs) are based on data from pivotal trials such as COLUMBUS (melanoma) and PHAROS (NSCLC).

7.1. Common Adverse Reactions (≥20-25%)

The most frequently reported ARs (all grades) with the combination of binimetinib and encorafenib include:

  • General: Fatigue (asthenia), pyrexia (fever).[7]
  • Gastrointestinal: Nausea, diarrhea (colitis), vomiting, abdominal pain (upper abdominal pain, abdominal discomfort, epigastric discomfort), constipation.[7]
  • Musculoskeletal: Musculoskeletal pain (back pain, pain in extremity, myalgia, non-cardiac chest pain, neck pain), arthralgia.[7]
  • Ocular: Visual impairment (blurred vision, serous retinopathy/retinal pigment epithelial detachment).[7]
  • Respiratory: Dyspnea, cough.[7]
  • Dermatologic: Rash (dermatitis acneiform), hyperkeratosis, dry skin, pruritus, alopecia, photosensitivity, palmar-plantar erythrodysesthesia syndrome (PPES), erythema, panniculitis.[7]
  • Neurologic: Headache, dizziness, dysgeusia.[18]
  • Hematologic: Anemia, leukopenia, neutropenia, lymphopenia.[18]
  • Metabolic/Laboratory Abnormalities: Increased creatine phosphokinase (CPK), increased transaminases (AST, ALT), increased gamma-glutamyl transferase (GGT), increased alkaline phosphatase, increased blood creatinine, hyponatremia.[18]
  • Other: Peripheral edema.[18]

Many of these common adverse events are low grade and manageable with supportive care or dose modifications. The profile reflects expected toxicities from both MEK inhibition (e.g., rash, diarrhea, ocular issues, CPK elevation) and BRAF inhibition (e.g., pyrexia, fatigue, certain cutaneous AEs).

7.2. Serious Adverse Reactions and Warnings/Precautions

The combination of binimetinib and encorafenib is associated with several potentially serious adverse reactions that require careful monitoring and management:

  • Cardiomyopathy: Decreases in left ventricular ejection fraction (LVEF), both asymptomatic and symptomatic, have been reported. In one study, an absolute decrease of ≥10% LVEF below baseline occurred in 7% of patients receiving the combination.[3] LVEF should be assessed by echocardiogram or MUGA scan at baseline, after one month of treatment, and then every 2 to 3 months thereafter. The safety of binimetinib has not been established in patients with a baseline LVEF below 50%.[7]
  • Venous Thromboembolism (VTE): Incidents of deep vein thrombosis (DVT) and pulmonary embolism (PE) have occurred in patients receiving the combination, with VTE reported in 6-7% of patients in clinical trials.[7]
  • Ocular Toxicities:
  • Serous Retinopathy / Retinal Pigment Epithelial Detachment (RPED): This is a common ocular toxicity associated with MEK inhibitors. In the COLUMBUS trial, serous retinopathy occurred in 20% of patients, which included retinal detachment (8%) and macular edema (6%).[7] These events often occur early in treatment.
  • Retinal Vein Occlusion (RVO): RVO is a known, serious class-related adverse reaction of MEK inhibitors and can potentially lead to blindness.[7]
  • Uveitis: Inflammation of the uvea has also been reported.[7] Regular ophthalmologic evaluations are recommended at baseline, at regular intervals during treatment, and promptly for any new or worsening visual disturbances.[7]
  • Interstitial Lung Disease (ILD) / Pneumonitis: Rare but potentially severe cases of ILD or pneumonitis have been observed.[7] New or progressive unexplained pulmonary symptoms should be assessed promptly.
  • Hepatotoxicity: Elevations in liver function tests (AST, ALT, bilirubin) can occur. Liver function tests should be monitored before initiating treatment, monthly during treatment, and as clinically indicated.[7]
  • Rhabdomyolysis: Marked elevations in CPK and symptoms of muscle pain or weakness consistent with rhabdomyolysis have been reported. CPK levels should be monitored periodically, especially in patients reporting muscle symptoms.[7]
  • Hemorrhage: Hemorrhagic events, including serious events like intracranial hemorrhage (which was fatal in 1% of patients in one study), can occur.[7]
  • New Primary Malignancies: The development of new primary malignancies, both cutaneous (e.g., cutaneous squamous cell carcinoma, keratoacanthoma, basal cell carcinoma, new primary melanoma) and non-cutaneous, is a risk associated with BRAF inhibitors due to paradoxical activation of the MAPK pathway in BRAF wild-type cells. Regular dermatologic examinations are required before treatment, every 2 months during treatment, and for up to 6 months after discontinuation.[3]
  • Embryo-Fetal Toxicity: Binimetinib, like other MEK inhibitors, can cause fetal harm when administered to a pregnant woman. Patients of reproductive potential should be advised of this risk and the need for effective contraception during treatment and for a period after the last dose.[7]
  • QT Prolongation: Encorafenib, the combination partner, is associated with dose-dependent QTc interval prolongation.[19]
  • Hypersensitivity: Binimetinib is contraindicated in patients with hypersensitivity to the active substance or any of its excipients.[18]

The extensive range of potential serious adverse events necessitates a proactive and vigilant approach to patient monitoring. This includes regular LVEF assessments, ophthalmologic examinations, liver function tests, CPK measurements, and dermatologic screenings. Early detection and prompt management, often involving dose modifications, are critical for patient safety and to allow for continued therapy where possible.

7.3. Management Strategies for Adverse Events

The management of adverse events associated with binimetinib (in combination with encorafenib) typically involves dose interruption, dose reduction, or, in cases of severe or persistent toxicity, permanent discontinuation of one or both drugs.[7] Specific guidelines for managing common and serious toxicities such as LVEF decline, VTE, ocular events, ILD, hepatotoxicity, rhabdomyolysis, and various dermatologic reactions are detailed in the official prescribing information.[7]

Table 4: Common (≥20%) and Selected Serious Adverse Reactions Associated with Binimetinib + Encorafenib (Melanoma - COLUMBUS Trial Data)

Adverse Reaction (System Organ Class)Frequency Category (All Grades)% Patients (All Grades)% Patients (Grade 3/4)
Gastrointestinal Disorders
NauseaVery Common41%2%
DiarrheaVery Common36%5%
VomitingVery Common30%2%
Abdominal painCommon18%2%
ConstipationCommon17%0%
General Disorders
FatigueVery Common43%6%
PyrexiaVery Common18% (BRAF inhibitor effect)2%
Peripheral edemaVery Common13%<1%
Skin and Subcutaneous Tissue Disorders
RashCommon22%2%
HyperkeratosisCommon10% (BRAF inhibitor effect)<1%
Dry SkinCommon10% (BRAF inhibitor effect)0%
PruritusCommon10%<1%
Musculoskeletal Disorders
ArthralgiaCommon24% (BRAF inhibitor effect)2%
Myopathy/Muscular Disorder (incl. CPK incr.)Very Common26% (CPK incr.)7% (CPK incr.)
Back painCommon10%1%
Nervous System Disorders
HeadacheCommon22% (BRAF inhibitor effect)<1%
DizzinessCommon15%<1%
Eye Disorders
Visual impairment (e.g., blurred vision, RPED)Very Common20% (MEK inhibitor effect)1%
Investigations
Blood CPK increasedVery Common26%7%
GGT increasedVery Common24%10%
AST increasedCommon17%2%
ALT increasedCommon14%3%
Vascular Disorders
HypertensionCommon11%4%
HemorrhageCommon13%3%
Venous ThromboembolismCommon6%3%
Cardiac Disorders
Left Ventricular Dysfunction (LVEF decrease)Common7%2%
Neoplasms
Cutaneous Squamous Cell CarcinomaCommon3% (BRAF inhibitor effect)2%

Frequencies and percentages are approximate based on prescribing information for Mektovi (binimetinib) in combination with Braftovi (encorafenib) from the COLUMBUS trial. "Very Common" typically ≥10%, "Common" ≥1% to <10%. Specific percentages for Grade 3/4 events are provided where available. Note that some AEs are more characteristic of BRAF inhibitors (e.g., pyrexia, hyperkeratotic rashes) while others are more characteristic of MEK inhibitors (e.g., serous retinopathy, CPK elevation, certain gastrointestinal AEs). Sources:.[7]

8. Dosage, Administration, and Formulations

8.1. Recommended Dosing Regimens for Approved Indications

For both approved indications—unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, and metastatic non-small cell lung cancer (NSCLC) with a BRAF V600E mutation—the recommended dosage of binimetinib (Mektovi) is 45 mg taken orally twice daily.[7] Doses should be administered approximately 12 hours apart. Binimetinib is always used in combination with encorafenib (Braftovi), and treatment is continued until disease progression or the development of unacceptable toxicity.[17] The prescribing information for encorafenib should be consulted for its specific dosing recommendations.

8.2. Administration Guidelines

Binimetinib tablets may be taken with or without food.[3]

  • Missed Dose: If a dose of binimetinib is missed, the patient should take it as soon as they remember. However, if the next scheduled dose is due within 6 hours, the missed dose should be skipped, and the patient should resume the regular dosing schedule. Patients should not take two doses at the same time or an extra dose to compensate for a missed one.[15]
  • Vomiting: If vomiting occurs after taking a dose of binimetinib, an additional dose should not be taken. The patient should continue with the next scheduled dose at the regular time.[15]
  • Swallowing Tablets: The tablets should ideally be swallowed whole. Although manufacturer guidance on crushing or splitting Mektovi tablets is not explicitly provided, it is generally advisable to swallow film-coated tablets whole unless otherwise specified.[15]

8.3. Dose Modifications for Adverse Reactions

Management of adverse reactions may require dose interruption, reduction, or permanent discontinuation of binimetinib.

  • The first dose reduction for binimetinib is to 30 mg orally twice daily.[7]
  • If a patient is unable to tolerate binimetinib at 30 mg twice daily, subsequent modification involves permanent discontinuation of the drug.[7] Detailed guidelines for dose modifications are provided in the prescribing information for specific adverse reactions, including but not limited to:
  • Cardiomyopathy (based on LVEF decrease)
  • Venous thromboembolism
  • Ocular toxicities (e.g., serous retinopathy, RVO, uveitis)
  • Interstitial lung disease
  • Hepatotoxicity (AST/ALT elevations)
  • Rhabdomyolysis or CPK elevations
  • Other dermatologic reactions.[7] It is important to note that if encorafenib (the combination partner) is permanently discontinued for any reason, binimetinib must also be discontinued.[17] This structured approach to toxicity management, with clear criteria for dose adjustments, is crucial for maintaining patient safety while attempting to optimize therapeutic benefit.

8.4. Dosage Adjustments in Special Populations

  • Hepatic Impairment: For patients with moderate hepatic impairment (defined as total bilirubin >1.5 to ≤3 times the upper limit of normal [ULN] and any AST) or severe hepatic impairment (total bilirubin >3 × ULN and any AST), the recommended dosage of binimetinib is reduced to 30 mg orally twice daily.[7] No dose adjustment is typically needed for mild hepatic impairment.
  • Renal Impairment: Specific dose adjustment guidelines for renal impairment are not prominently highlighted in the provided snippets, suggesting that mild to moderate renal impairment may not significantly alter binimetinib pharmacokinetics to a clinically relevant extent requiring dose changes. However, severe renal impairment or end-stage renal disease data may be limited, and caution is generally advised.
  • Pediatric Use: The safety and efficacy of binimetinib have not been established in pediatric patients.[1]
  • Geriatric Use: When used in combination with encorafenib, no overall differences in the safety or effectiveness of binimetinib were observed in older patients (typically ≥65 years) compared to younger patients.[13] However, elderly patients may be more susceptible to certain side effects, and careful monitoring is always warranted.

8.5. Available Formulations

Binimetinib (Mektovi) is available as 15 mg film-coated tablets for oral administration.[1]

Table 5: Recommended Dosage Modifications for Binimetinib due to Adverse Reactions

Adverse ReactionSeverity / GradeRecommended Action for Binimetinib
CardiomyopathyAsymptomatic, absolute LVEF decrease >10% from baseline AND LVEF below LLNWithhold for up to 4 weeks. Evaluate LVEF every 2 weeks. Resume at reduced dose (30mg BID) if LVEF ≥LLN AND absolute decrease from baseline ≤10% AND patient asymptomatic. If LVEF does not recover within 4 weeks, permanently discontinue.
Symptomatic CHF OR Absolute LVEF decrease >20% from baseline AND LVEF below LLNPermanently discontinue.
Venous Thromboembolism (VTE)Uncomplicated DVT or PEWithhold for up to 4 weeks. If improves, resume at same dose. If no improvement, permanently discontinue.
Life-threatening PEPermanently discontinue.
Ocular Toxicities
Serous Retinopathy/RPEDSymptomaticWithhold for up to 10 days. If improves and asymptomatic, resume at same dose. If not improved, resume at reduced dose (30mg BID) or permanently discontinue.
Retinal Vein Occlusion (RVO)Any gradePermanently discontinue.
UveitisGrade 1-2Treat with topical steroids. If no improvement in 1 week or Grade 2 recurs, withhold for up to 4 weeks. If improves, resume at same or reduced dose. If no improvement, permanently discontinue.
Grade 3-4Permanently discontinue.
Interstitial Lung Disease (ILD) / PneumonitisAny gradePermanently discontinue.
Hepatotoxicity (AST/ALT increased)Recurrent Grade 2 OR First occurrence of Grade 3Withhold for up to 4 weeks. If improves to Grade 0-1 or baseline, resume at reduced dose (30mg BID). If no improvement, permanently discontinue.
First occurrence of Grade 4 OR Grade 3 with bilirubin >2x ULNPermanently discontinue OR Withhold for up to 4 weeks; if improves to Grade 0-1 or baseline, resume at reduced dose (30mg BID); if no improvement, permanently discontinue.
Rhabdomyolysis or CPK ElevationsGrade 4 asymptomatic CPK elevation OR Any grade CPK elevation with symptoms OR with renal impairmentWithhold for up to 4 weeks. If improves to Grade 0-1, resume at reduced dose (30mg BID). If not resolved within 4 weeks, permanently discontinue.
Other Adverse Reactions (e.g., Dermatologic excluding PPES, other Grade 2-4)Grade 2 not improving within 2 weeks; First occurrence of Grade 3; Recurrent Grade 3; First occurrence Grade 4Depending on specific AE and grade: Withhold until Grade 0-1, then resume at same or reduced dose. For Grade 4 or intolerable Grade 2/3, may require permanent discontinuation. Refer to full prescribing information for detailed management of other specific ARs.

This table summarizes general principles. Clinicians must refer to the full, current prescribing information for complete and specific dose modification instructions. LLN = Lower Limit of Normal. CHF = Congestive Heart Failure. Sources:.[7]

9. Drug Interactions

The metabolism and transport of binimetinib can be influenced by concomitant medications, and binimetinib itself may affect other drugs. Understanding these interactions is crucial for safe and effective therapy, especially considering it is used in combination with encorafenib, which has its own distinct drug interaction profile.

9.1. Effect of Other Drugs on Binimetinib

Binimetinib's primary metabolic pathway is glucuronidation mediated by UGT1A1, which accounts for approximately 61% of its metabolism.[12] Minor contributions to its metabolism come from CYP1A2 and CYP2C19, which form the active metabolite M3.[12] Binimetinib is also a substrate of the efflux transporters P-gp and BCRP.[12]

  • UGT1A1 Modulators:
  • UGT1A1 Inducers (e.g., rifampicin, phenobarbital, smoking): These agents have the potential to decrease binimetinib exposure by increasing its glucuronidation. However, some prescribing information suggests that UGT1A1 genotype and smoking (a UGT1A1 inducer) do not have a clinically important effect on binimetinib exposure.[5] Nevertheless, caution is advised when co-administering strong UGT1A1 inducers.[18]
  • UGT1A1 Inhibitors (e.g., atazanavir, indinavir, sorafenib): These may increase binimetinib plasma concentrations. Pharmacokinetic simulations predict that atazanavir (a UGT1A1 inhibitor) does not significantly alter the Cmax of binimetinib.[7] Co-administration with UGT1A1 inhibitors should be done with caution.[18]
  • CYP1A2 Inducers (e.g., carbamazepine, rifampicin): Since CYP1A2 is involved in forming the active metabolite M3, and potentially in other minor pathways, strong inducers of CYP1A2 could decrease binimetinib exposure and potentially its active metabolite levels, possibly reducing efficacy.[18]
  • P-gp Inducers (e.g., St. John's Wort, phenytoin): As binimetinib is a P-gp substrate, inducers of P-gp may decrease its plasma concentrations and efficacy.[18]
  • P-gp/BCRP Inhibitors: While binimetinib is a substrate for these transporters, its moderate to high passive permeability suggests that inhibition of P-gp or BCRP is unlikely to lead to clinically important increases in binimetinib concentrations.[18]
  • Encorafenib: Encorafenib is a reversible inhibitor of UGT1A1. However, clinical data indicate no significant differences in binimetinib exposure when it is co-administered with encorafenib.[12] This is an important observation given their routine combination.
  • Acid-Reducing Agents (e.g., proton pump inhibitors like rabeprazole, H2-receptor antagonists, antacids): Co-administration of a gastric acid-reducing agent (rabeprazole) did not alter the extent of binimetinib exposure (AUC).[12]

9.2. Effect of Binimetinib on Other Drugs

Binimetinib itself has a relatively modest impact on major CYP enzymes and transporters.

  • CYP3A4 Substrates (e.g., midazolam, hormonal contraceptives): Clinical studies showed that binimetinib did not alter the exposure of midazolam, a sensitive CYP3A4 substrate.[12] This suggests binimetinib is not a significant inhibitor or inducer of CYP3A4. However, this point requires careful clinical consideration because binimetinib is almost exclusively used with encorafenib. Encorafenib IS a moderate inducer of CYP3A4 and can significantly decrease the exposure of CYP3A4 substrates, including hormonal contraceptives, potentially leading to reduced efficacy.[19] Therefore, when the combination is used, the drug interaction profile of encorafenib for CYP3A4 substrates is clinically dominant.
  • CYP1A2 Substrates (e.g., duloxetine, theophylline): In vitro data suggest binimetinib is a potential inducer of CYP1A2. Therefore, co-administration with sensitive CYP1A2 substrates should be done with caution as their efficacy might be reduced.[18]
  • OAT3 Substrates (e.g., pravastatin, ciprofloxacin): Binimetinib is a weak inhibitor of the organic anion transporter 3 (OAT3) in vitro. Caution is advised when co-administering binimetinib with sensitive OAT3 substrates, as their exposure might be increased.[18]
  • CYP2C9 Substrates: In vitro, binimetinib is a weak reversible inhibitor of CYP2C9.[18] The clinical significance of this is likely low but warrants consideration with narrow therapeutic index CYP2C9 substrates.
  • Other Transporters: Binimetinib is not expected to cause clinically significant drug-drug interactions via inhibition of other common drug transporters.[18]

The drug interaction profile of binimetinib itself appears relatively manageable. The primary considerations for binimetinib involve UGT1A1 and potentially CYP1A2 induction. However, the clinical context of its use almost exclusively with encorafenib means that encorafenib's more pronounced effects on CYP3A4 (as an inducer, and being affected by strong CYP3A4 inhibitors/inducers) often dictate the overall drug interaction management strategy for the combination therapy. Clinicians must always consult the prescribing information for both drugs when considering potential interactions.

Table 6: Clinically Significant Drug Interactions with Binimetinib (and Key Considerations for Encorafenib Combination)

Interacting Agent/ClassEffect on Binimetinib or Concomitant DrugMechanism (if known)Clinical Recommendation
UGT1A1 Inducers (e.g., rifampicin, phenobarbital)Potential decrease in binimetinib exposureInduction of UGT1A1-mediated glucuronidation of binimetinibCo-administer with caution. Some labels suggest no clinically important effect from smoking (UGT1A1 inducer).
UGT1A1 Inhibitors (e.g., atazanavir, indinavir)Potential increase in binimetinib exposureInhibition of UGT1A1-mediated glucuronidation of binimetinibCo-administer with caution. Simulations with atazanavir predict similar binimetinib Cmax.
CYP1A2 Inducers (e.g., carbamazepine, rifampicin)Potential decrease in binimetinib exposure (and active metabolite M3)Induction of CYP1A2May decrease binimetinib efficacy; consider alternatives if possible.
P-gp Inducers (e.g., St. John's Wort, phenytoin)Potential decrease in binimetinib exposureInduction of P-gp efflux of binimetinibMay decrease binimetinib efficacy; avoid if possible.
Sensitive CYP1A2 Substrates (e.g., duloxetine, theophylline)Potential decrease in exposure of CYP1A2 substrateBinimetinib is a potential inducer of CYP1A2Exercise caution; monitor efficacy of CYP1A2 substrate.
Sensitive OAT3 Substrates (e.g., pravastatin, ciprofloxacin)Potential increase in exposure of OAT3 substrateBinimetinib is a weak inhibitor of OAT3Exercise caution; monitor for toxicity of OAT3 substrate.
Encorafenib (Combination Partner)No clinically significant effect on binimetinib exposure.Encorafenib is a UGT1A1 inhibitor, but clinically no net effect on binimetinib PK.Standard combination dosing.
Strong or Moderate CYP3A4 Inhibitors (e.g., ketoconazole, clarithromycin, grapefruit juice)Significant increase in encorafenib exposure. (No direct major effect on binimetinib)Inhibition of CYP3A4 metabolism of encorafenib.Avoid coadministration with BRAFTOVI (encorafenib). If unavoidable, reduce encorafenib dose.
Strong CYP3A4 Inducers (e.g., rifampin, phenytoin, St. John's Wort)Significant decrease in encorafenib exposure. (No direct major effect on binimetinib)Induction of CYP3A4 metabolism of encorafenib.Avoid coadministration with BRAFTOVI (encorafenib).
Sensitive CYP3A4 Substrates (including hormonal contraceptives)Significant decrease in exposure of CYP3A4 substrate. (Binimetinib itself has minimal effect)Encorafenib is a moderate inducer of CYP3A4.Avoid coadministration of BRAFTOVI (encorafenib) with sensitive CYP3A4 substrates where decreased efficacy is critical. Use alternative non-hormonal contraception.

Sources:.[7] This table highlights interactions primarily related to binimetinib and crucial interactions related to its combination partner encorafenib that dominate the clinical DDI management.

10. Regulatory Status

10.1. Approvals by Major Regulatory Agencies

Binimetinib (Mektovi), in combination with encorafenib (Braftovi), has received marketing authorization from several major regulatory bodies worldwide for specific oncological indications.

  • Food and Drug Administration (FDA), USA:
  • Melanoma: Approved on June 27, 2018, for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, in combination with encorafenib.[7]
  • Non-Small Cell Lung Cancer (NSCLC): Approved on October 11, 2023, for adult patients with metastatic NSCLC with a BRAF V600E mutation, in combination with encorafenib.[5]
  • European Medicines Agency (EMA), European Union:
  • Melanoma: Approved for the treatment of adult patients with unresectable or metastatic melanoma with a BRAF V600 mutation, in combination with encorafenib.[8]
  • Non-Small Cell Lung Cancer (NSCLC): Received a positive opinion from the Committee for Medicinal Products for Human Use (CHMP) in July 2024, followed by European Commission (EC) approval on August 30, 2024, for adult patients with advanced NSCLC with a BRAF V600E mutation, in combination with encorafenib.[25]
  • Health Canada:
  • Melanoma: Approved on March 2, 2021, for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600 mutation, as detected by a validated test, in combination with encorafenib.[23]
  • Therapeutic Goods Administration (TGA), Australia:
  • Melanoma: Approved for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation, in combination with encorafenib.[41] While approved by the TGA, a 2019 report noted that the pharmaceutical company had not proceeded with Pharmaceutical Benefits Scheme (PBS) listing at that time for this indication.[42]
  • Pharmaceuticals and Medical Devices Agency (PMDA), Japan:
  • Melanoma: Approved for unresectable or metastatic melanoma with a BRAF V600 mutation, in combination with encorafenib.[20]
  • Other Regions: Regulatory applications for binimetinib in combination with encorafenib for its approved indications have been submitted or granted in various other countries around the world.[20]

10.2. Approved Companion Diagnostics

The use of binimetinib in combination with encorafenib is contingent upon the presence of specific BRAF mutations in the tumor. Therefore, FDA-approved (or similarly validated in other regions) companion diagnostic tests are required to identify eligible patients.

  • For melanoma, an FDA-approved test must confirm the presence of a BRAF V600E or V600K mutation in tumor specimens prior to initiating therapy.[5]
  • For NSCLC, an FDA-approved test must confirm the presence of a BRAF V600E mutation in tumor or plasma specimens. If no mutation is detected in a plasma specimen, tumor tissue should be tested.[5] Examples of FDA-approved companion diagnostics for the NSCLC indication include the FoundationOne CDx (tissue-based) and FoundationOne Liquid CDx (plasma-based) assays.[25]

The widespread regulatory approvals underscore the significant clinical benefit offered by the binimetinib and encorafenib combination in these genetically defined patient populations. The mandatory use of companion diagnostics is a critical component of this targeted therapy approach, ensuring that treatment is directed towards patients who are most likely to respond, thereby optimizing efficacy and minimizing unnecessary exposure to potentially toxic drugs.

11. Conclusion and Expert Insights

11.1. Summary of Binimetinib's Role in Targeted Cancer Therapy

Binimetinib, a potent and selective MEK1/2 inhibitor, has established a significant role in targeted cancer therapy, primarily through its combination with the BRAF inhibitor encorafenib. This dual blockade of the MAPK signaling pathway has proven to be a highly effective strategy for patients with BRAF V600-mutant unresectable or metastatic melanoma and, more recently, for BRAF V600E-mutant metastatic non-small cell lung cancer. The therapeutic efficacy of this combination stems from its ability to more comprehensively inhibit the oncogenic signaling driven by BRAF mutations, thereby overcoming or delaying the onset of resistance mechanisms that often limit the durability of BRAF inhibitor monotherapy. The clinical development of binimetinib, particularly the outcomes of the COLUMBUS and PHAROS trials, has demonstrated substantial improvements in progression-free survival, overall survival (in melanoma), and objective response rates compared to previous standards of care or in settings with limited options. Coupled with a safety profile that, while notable for specific MEK inhibitor-class effects, is generally manageable through proactive monitoring and appropriate dose modifications, binimetinib in combination with encorafenib represents a valuable therapeutic advancement.

11.2. Key Differentiators and Clinical Implications

Several factors contribute to the clinical importance of binimetinib:

  • Efficacy of the Combination Strategy: The cornerstone of binimetinib's success is its synergistic action when combined with encorafenib. The dual targeting of two critical nodes (BRAF and MEK) in the MAPK pathway provides a more profound and sustained inhibition of downstream signaling. This is particularly evident in the COLUMBUS trial, where the combination significantly outperformed BRAF inhibitor monotherapy in terms of PFS and OS in melanoma.[9] This mechanistic advantage translates directly into improved patient outcomes.
  • Tolerability Profile of the Combination: While combination therapies often raise concerns about additive toxicities, the encorafenib/binimetinib regimen has demonstrated a manageable safety profile. Some evidence from the COLUMBUS trial even suggested that the combination did not increase the overall rate of adverse events compared to encorafenib monotherapy and may have a more acceptable profile than older BRAF inhibitor regimens or other BRAF/MEK combinations, with fewer patients requiring dose reductions or interruptions due to toxicity.[9] This aspect is crucial for maintaining quality of life and allowing patients to remain on effective therapy for longer durations.
  • Favorable Pharmacokinetic Characteristics: Binimetinib's pharmacokinetic profile, including oral administration, the lack of a significant food effect on exposure, and well-defined metabolic pathways (primarily UGT1A1-mediated glucuronidation with minor CYP involvement for an active metabolite), contributes to its clinical utility and predictability.[1] While binimetinib itself has a relatively modest drug-drug interaction profile, the co-administration with encorafenib necessitates careful attention to encorafenib's significant interactions, particularly those involving CYP3A4.

11.3. Unmet Needs and Future Perspectives

Despite the advances made with binimetinib and encorafenib, several challenges and areas for future research remain:

  • Mechanisms of Acquired Resistance: Acquired resistance to BRAF/MEK inhibitor combinations eventually occurs in most patients. Understanding the diverse molecular mechanisms underlying this resistance is critical for developing subsequent lines of therapy or strategies to prevent or overcome it. This includes investigating triplet therapies (e.g., adding immunotherapy or other targeted agents) or optimizing treatment sequencing.
  • Expanding to Other Tumor Types and BRAF Alterations: The MAPK pathway is dysregulated in many cancers beyond BRAF V600-mutant melanoma and NSCLC. Ongoing research, such as the Perfume trial evaluating binimetinib in BRAF fusion-positive low-grade gliomas and pancreatic cancer [33], is exploring the broader applicability of MEK inhibition. The NCI-MATCH trial results in NRAS-mutant cancers, while largely negative for monotherapy, hinted at potential codon-specific sensitivities that warrant further exploration [30], emphasizing the need for more refined biomarker-driven patient selection.
  • Treatment of Brain Metastases: CNS metastases remain a significant challenge in melanoma and other cancers. Clinical trials specifically evaluating the efficacy and safety of binimetinib in combination with encorafenib in patients with brain metastases are crucial for addressing this high unmet need.[20] Determining the CNS penetration of these agents and their activity in the brain microenvironment is a key research focus.
  • Biomarker Development: Beyond the presence of BRAF V600 mutations, there is a need for additional biomarkers that can predict response, duration of benefit, or likelihood of specific toxicities with MEK inhibitor-based therapies. The exploratory finding of ctDNA RAS-mutant allele frequency as a potential prognostic marker in the MErCuRIC trial is an example of such efforts.[31] Further research into genomic, transcriptomic, or proteomic markers could help personalize treatment strategies more effectively.

In summary, binimetinib, as part of a dual MAPK pathway blockade strategy, has significantly improved outcomes for patients with specific BRAF-mutant cancers. Its continued investigation in other oncogenic contexts and in combination with novel agents holds the promise of further expanding its therapeutic reach and addressing ongoing challenges in cancer treatment.

Works cited

  1. Binimetinib | C17H15BrF2N4O3 | CID 10288191 - PubChem, accessed June 5, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Binimetinib
  2. Binimetinib | ARRY-162 | MEK1/2 inhibitor | antitumor | TargetMol, accessed June 5, 2025, https://www.targetmol.com/compound/binimetinib
  3. Binimetinib (MEKTOVI) National Drug Monograph Jun 2022 - VA.gov, accessed June 5, 2025, https://www.va.gov/formularyadvisor/DOC_PDF/MON_Binimetinib_MEKTOVI_Jun_2022.pdf
  4. What is the mechanism of Binimetinib? - Patsnap Synapse, accessed June 5, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-binimetinib
  5. Drugs@FDA: Drug Product MEKTOVI (BINIMETINIB), NDA210498, Array BioPharma Inc., accessed June 5, 2025, https://www.pharmgkb.org/literature/15101911
  6. MEKTOVI® (binimetinib) Indications and Usage | Pfizer Medical Information - US, accessed June 5, 2025, https://www.pfizermedicalinformation.com/mektovi/indications-usage
  7. www.accessdata.fda.gov, accessed June 5, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/210498s010lbl.pdf
  8. Mektovi | European Medicines Agency (EMA), accessed June 5, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/mektovi
  9. A trial of encorafenib, binimetinib and vemurafenib for melanoma (COLUMBUS), accessed June 5, 2025, https://www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-of-encorafenib-binimetinib-and-vemurafenib-for-melanoma-columbus
  10. Encorafenib and Binimetinib: A New Benchmark in Metastatic Melanoma Therapy?, accessed June 5, 2025, https://ascopost.com/issues/december-10-2018/encorafenib-and-binimetinib/
  11. A phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitor, accessed June 5, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5344293/
  12. MEKTOVI® (binimetinib) Clinical Pharmacology | Pfizer Medical Information - US, accessed June 5, 2025, https://www.pfizermedicalinformation.com/mektovi/clinical-pharmacology
  13. Binimetinib: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed June 5, 2025, https://go.drugbank.com/drugs/DB11967
  14. The Absorption, Distribution, Metabolism, and Excretion of Binimetinib Following a Single Oral Dose of [ 14C]Binimetinib 45 mg in Healthy Male Participants - PMC, accessed June 5, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11780716/
  15. Mektovi: Uses, dosage, side effects, interactions, and more - MedicalNewsToday, accessed June 5, 2025, https://www.medicalnewstoday.com/articles/drugs-mektovi
  16. Binimetinib (oral route) - Mayo Clinic, accessed June 5, 2025, https://www.mayoclinic.org/drugs-supplements/binimetinib-oral-route/description/drg-20443876
  17. MEKTOVI® (binimetinib) Dosage and Administration | Pfizer Medical Information - US, accessed June 5, 2025, https://www.pfizermedicalinformation.com/mektovi/dosage-admin
  18. Mektovi 15 mg film-coated tablets - Summary of Product ..., accessed June 5, 2025, https://www.medicines.org.uk/emc/product/9501/smpc
  19. Drug Interactions | BRAFTOVI® (encorafenib) + MEKTOVI® (binimetinib) | Safety Info, accessed June 5, 2025, https://braftovi.pfizerpro.com/mektovi-m/drug-interactions
  20. Binimetinib + Encorafenib for Melanoma · 2025 Clinical Trial · Phase 2 | Power, accessed June 5, 2025, https://www.withpower.com/trial/phase-3-skin-neoplasms-8-2021-d9fc0?condition=Cutaneous+Melanoma&overallStatus=Recruiting&hasNoPlacebo=false
  21. A plain language summary of the 5-year update from part 1 of the COLUMBUS study: encorafenib and binimetinib for people with BRAF V600-mutant melanoma: Future Oncology - Taylor & Francis Online: Peer-reviewed Journals, accessed June 5, 2025, https://www.tandfonline.com/doi/abs/10.2217/fon-2022-1258
  22. Real-life evidence of encorafenib plus binimetinib in patients with unresectable advanced or metastatic BRAF V600 -mutant melanoma in Spain: the BECARE (GEM-2002) trial - Frontiers, accessed June 5, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2025.1466185/full
  23. Summary Basis of Decision for Braftovi - Drug and Health Products Portal, accessed June 5, 2025, https://dhpp.hpfb-dgpsa.ca/review-documents/resource/SBD00534
  24. Drug Interactions | BRAFTOVI® (encorafenib) + MEKTOVI® (binimetinib) | Safety Info, accessed June 5, 2025, https://braftovi.pfizerpro.com/mektovi-n/drug-interactions
  25. Encorafenib (Braftovi®) in combination with binimetinib (Mektovi®) for the treatment of advanced non-small cell lung cancer (NSCLC), accessed June 5, 2025, https://eprints.aihta.at/1540/1/Fact%20Sheet%20Nr.187.pdf
  26. Encorafenib/Binimetinib Combo Approved in Europe for BRAF V600E+ NSCLC - OncLive, accessed June 5, 2025, https://www.onclive.com/view/encorafenib-binimetinib-combo-approved-in-europe-for-braf-v600e-nsclc
  27. Adverse Reactions | BRAFTOVI® (encorafenib) + MEKTOVI® (binimetinib) | Safety Info, accessed June 5, 2025, https://braftovi.pfizerpro.com/mektovi-n/adverse-reactions
  28. SOC Encorafenib/Binimetinib Sets a Precedent for Therapeutic Improvements in BRAF V600E–Mutant NSCLC - OncLive, accessed June 5, 2025, https://www.onclive.com/view/soc-encorafenib-binimetinib-sets-a-precedent-for-therapeutic-improvements-in-braf-v600e-mutant-nsclc
  29. (PDF) A plain language summary of the PHAROS study: the combination of encorafenib and binimetinib for people with BRAF V600E-mutant metastatic non-small cell lung cancer - ResearchGate, accessed June 5, 2025, https://www.researchgate.net/publication/378232492_A_plain_language_summary_of_the_PHAROS_study_the_combination_of_encorafenib_and_binimetinib_for_people_with_BRAF_V600E-mutant_metastatic_non-small_cell_lung_cancer
  30. Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated ..., accessed June 5, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8542423/
  31. A Phase Ia/b study of MEK1/2 inhibitor binimetinib with MET inhibitor ..., accessed June 5, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11984268/
  32. Binimetinib Completed Phase 2 Trials for KRAS Mutation-Related Tumors / Non-Small Cell Lung Cancer (NSCLC) Treatment - DrugBank, accessed June 5, 2025, https://go.drugbank.com/drugs/DB11967/clinical_trials?conditions=DBCOND0132579%2CDBCOND0034130&phase=2&purpose=treatment&status=completed
  33. Perfume trial: Phase II trial of binimetinib in patients with BRAF fusion-positive low-grade glioma or pancreatic cancer. - ASCO Publications, accessed June 5, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.TPS3188
  34. Abstract - ASCO Meeting Program Guide, accessed June 5, 2025, https://meetings.asco.org/abstracts-presentations/252289/abstracts
  35. Binimetinib Recruiting Phase 2 Trials for Melanoma and Brain Metastases / Melanoma BRAF V600E/K Mutated Treatment - DrugBank, accessed June 5, 2025, https://go.drugbank.com/drugs/DB11967/clinical_trials?conditions=DBCOND0050210%2CDBCOND0071768&phase=2&purpose=treatment&status=recruiting
  36. Binimetinib: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed June 5, 2025, https://www.rxlist.com/binimetinib/generic-drug.htm
  37. Adjuvant Encorafenib/Binimetinib Is Well Tolerated and May Reduce Recurrence Risk in BRAF V600–Mutant Melanoma - OncLive, accessed June 5, 2025, https://www.onclive.com/view/adjuvant-encorafenib-binimetinib-is-well-tolerated-and-may-reduce-recurrence-risk-in-braf-v600-mutant-melanoma
  38. Binimetinib: MedlinePlus Drug Information, accessed June 5, 2025, https://medlineplus.gov/druginfo/meds/a618041.html
  39. Binimetinib Oral: Uses, Side Effects, Interactions, Pictures, Warnings & Dosing - WebMD, accessed June 5, 2025, https://www.webmd.com/drugs/2/drug-175525/binimetinib-oral/details
  40. Braftovi (Encorafenib) + Mektovi (Binimetinib) | Drug Therapies - Melanoma Canada, accessed June 5, 2025, https://melanomacanada.ca/diagnosis-and-treatment/melanoma/targeted-therapy/braftovimektovi/
  41. The POLARIS Trial - Australian Clinical Trials, accessed June 5, 2025, https://australianclinicaltrials.com/study/the-polaris-trial/
  42. Hansard - House of Representatives 24/02/2020 Parliament of Australia, accessed June 5, 2025, https://www.aph.gov.au/Parliamentary_Business/Hansard/Hansard_Display?bid=chamber/hansardr/f3ed3760-e998-492f-9f3e-11b9802d8a98/&sid=0022

Published at: June 5, 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.