In the relentless pursuit of more effective treatments for colorectal cancer (CRC), a groundbreaking study has recently shed light on the challenges faced when targeting RAS mutations, which remain some of the most elusive drivers of this disease. Published in BMC Cancer, the Phase Ia/b trial known as MErCuRIC represents a comprehensive effort to evaluate the dual inhibition of MEK1/2 and MET pathways using binimetinib and crizotinib, respectively, in patients with advanced RAS mutant colorectal cancer. This multidisciplinary investigation offers crucial insights into the biological complexities and therapeutic hurdles associated with this aggressive cancer subtype.
RAS mutations, particularly in KRAS, have long been recognized as pivotal contributors to the pathogenesis and progression of colorectal cancer. However, effective therapeutic strategies to counteract RAS-driven oncogenic signaling have remained limited, largely due to the intricate network of feedback mechanisms that sustain tumor growth. The study zeroes in on the RAS/MEK pathway, which forms a critical axis in tumor cell proliferation, survival, and resistance. Prior preclinical models suggested that MET signaling and its downstream STAT3 activation may serve as alternative survival pathways that cancer cells exploit to bypass MEK inhibition, prompting the rationale for a combined MEK and MET blockade.
The MErCuRIC study utilized binimetinib, a potent small-molecule inhibitor of MEK1/2, in conjunction with crizotinib, a well-characterized MET inhibitor approved for other malignancies. The trial began with a dose escalation phase involving patients with advanced solid tumors to establish the maximum tolerated dose (MTD) and examine safety profiles. This was followed by a dose expansion phase focused specifically on patients harboring RAS mutant metastatic colorectal cancer, aiming to scrutinize therapeutic efficacy and biological response markers.
Twenty patients participated in the dose escalation cohorts, where dosing regimens were meticulously adjusted through a rolling-6 design to optimize tolerability. The investigators determined the MTD to be binimetinib at 30 mg twice daily on days 1 through 21 every 28 days, combined with continuous daily dosing of crizotinib at 250 mg. Notably, dose-limiting toxicities prominently featured hepatotoxicity, as evidenced by grade 3 or higher transaminitis, along with elevations in creatinine phosphokinase and fatigue. These toxicities highlighted the systemic impact and narrow therapeutic window of this pharmacological combination.
Following the identification of the MTD, the trial expanded to include 36 patients diagnosed with RAS mutant metastatic colorectal cancer. Detailed pharmacokinetic and pharmacodynamic analyses performed on blood samples revealed compelling evidence of target pathway engagement, affirming that both binimetinib and crizotinib effectively reached their intended molecular targets in vivo. Complementary analyses of skin and tumor biopsies using c-MET immunohistochemistry and in situ hybridization techniques allowed researchers to interrogate the expression patterns and amplification status of MET, a critical determinant of treatment response.
However, despite optimal dosing and proven target inhibition, the clinical outcomes painted a sobering picture. The most common treatment-related adverse events (TR-AEs) reported were rash, fatigue, and diarrhea, converging with the toxicities noted in earlier cohorts. Alarmingly, nearly 45% of patients experienced grade 3 or higher adverse events, reflecting considerable tolerability challenges that may limit the clinical utility of this regimen. Objective tumor responses were conspicuously absent, with the best observed result being stable disease in just under a quarter of patients, underscoring the limited anti-tumor efficacy of the combined MEK and MET blockade in this setting.
A particularly intriguing molecular observation was the subgroup of patients exhibiting MET “super-expression,” characterized by high immunohistochemical H-scores exceeding 180 and strong MET in situ hybridization signals. Although this biomarker subset accounted for about 24% of patients, only one individual displayed true MET amplification. Unfortunately, this patient discontinued treatment early due to toxicity, precluding any assessment of potential benefit in this biologically defined group. These findings raise important questions about the predictive value of MET overexpression and amplification in guiding MET inhibitor use within RAS mutant colorectal cancer.
Moreover, circulating tumor DNA (ctDNA) analyses revealed that patients presenting with high baseline RAS mutant allele frequencies faced significantly shorter overall survival compared to those with lower mutant allele burdens. This correlation between molecular tumor load and survival outcomes reinforces the aggressive nature of high-terminal RAS mutation burdens and highlights the necessity of considering tumor heterogeneity and clonality in treatment design and prognostication.
The MErCuRIC study thus offers a rigorous and highly detailed examination of the therapeutic landscape for RAS mutant advanced colorectal cancer, shedding light on the intricacies of pathway interplay, resistance mechanisms, and toxicity management. It underscores the enduring challenge of directly targeting RAS-driven cancers, even when sophisticated combination approaches are employed. Importantly, it calls for a deeper understanding of the biology underpinning resistance, and the identification of novel biomarkers to stratify patients who might benefit from targeted therapeutic strategies.
From a clinical trial design perspective, the findings illustrate the critical importance of comprehensive biomarker-driven patient selection and the integration of translational science to unravel resistance pathways. The high incidence of adverse events associated with binimetinib and crizotinib combination therapy suggests that careful attention must be paid to dosing strategies, supportive care, and perhaps the exploration of alternative scheduling or sequencing to mitigate toxicity without compromising efficacy.
While the results are sobering, the endeavor to disentangle RAS mutant colorectal cancer’s complex signaling networks and develop effective targeted therapies remains an urgent priority. This study serves as a pivotal reference point, guiding future research efforts toward more innovative and nuanced therapeutic interventions. It points to the potential value of combination regimens that can overcome resistance pathways, including MET-mediated escape mechanisms, but also highlights the necessity of balancing efficacy with patient quality of life.
In conclusion, the MErCuRIC trial marks a significant milestone in the ongoing battle against RAS mutant colorectal cancer. It demonstrates the formidable challenges inherent in overcoming biological redundancy and therapeutic resistance, and the critical role of precise molecular characterization in guiding clinical decision-making. As research advances, continued exploration of alternative targets, combination strategies, and personalized medicine approaches will be essential to transform the therapeutic landscape for this challenging and prevalent malignancy.
—
Subject of Research: Therapeutic targeting of RAS mutant advanced colorectal cancer using MEK1/2 and MET inhibitors.
Article Title: A Phase Ia/b study of MEK1/2 inhibitor binimetinib with MET inhibitor crizotinib in patients with RAS mutant advanced colorectal cancer (MErCuRIC).
Article References:
Aroldi, F., Elez, E., André, T. et al. A Phase Ia/b study of MEK1/2 inhibitor binimetinib with MET inhibitor crizotinib in patients with RAS mutant advanced colorectal cancer (MErCuRIC).
BMC Cancer 25, 658 (2025). https://doi.org/10.1186/s12885-025-14068-1
Image Credits: Scienmag.com
DOI: https://doi.org/10.1186/s12885-025-14068-1
Tags: advanced RAS mutant CRCbinimetinib and crizotinib trialbiological complexities of colorectal cancerClinical Trials in Oncologydual pathway targeting in cancer treatmentinnovative treatments for colorectal cancerKRAS mutations and cancerMEK and MET inhibitionmultidisciplinary approach to cancer researchovercoming resistance in cancer therapyRAS mutations in colorectal cancertherapeutic challenges in colorectal cancer