Next Article in Journal
Comparative Mechanistic Insights and Therapeutic Potential of Pembrolizumab, Durvalumab, and Ipilimumab as Immune Checkpoint Inhibitors in the Targeted Management of Oral and Head and Neck Squamous Cell Carcinoma
Next Article in Special Issue
Advances in Precision Oncology: From Molecular Profiling to Regulatory-Approved Targeted Therapies
Previous Article in Journal
KRAS G12C Inhibition in Solid Tumors: Biological Breakthroughs, Clinical Evidence, and Open Challenges
Previous Article in Special Issue
TOPK Drives IL19-Mediated Crosstalk Between Cancer Cells and Fibroblasts to Promote Solar UV-Induced Skin Damage and Carcinogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-EGFR Therapy in Metastatic Colorectal Cancer: Identifying, Tracking, and Overcoming Resistance

by
Luís Felipe Leite
1,
Mariana Macambira Noronha
2,
Junior Samuel Alonso de Menezes
3,
Lucas Diniz da Conceição
1,
Luiz F. Costa Almeida
1,
Anelise Poluboiarinov Cappellaro
4,
Marcos Belotto
5,
Tiago Biachi de Castria
6,7,
Renata D’Alpino Peixoto
8 and
Thais Baccili Cury Megid
9,*
1
Department of Medical Sciences, Universidade Federal Fluminense, Niteroi 24220-140, RJ, Brazil
2
Department of Medical Sciences, Universidade Federal do Ceará, Fortaleza 60020-181, CE, Brazil
3
Department of Medical Sciences, Universidade Federal da Bahia, Salvador 40170-110, BA, Brazil
4
Department of Medical Sciences, Centro Universitário Maurício de Nassau de Barreiras, Barreiras 47808-180, BA, Brazil
5
Department of Surgery, Hospital 9 de Julho, São Paulo 01409-002, SP, Brazil
6
Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
7
Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
8
Department of Medical Oncology, BC Cancer Agency, Vancouver, BC V5Z 4E6, Canada
9
Centre de Formation Médicale du Nouveau-Brunswick, Université de Sherbrooke, Moncton, NB E1C 2Z3, Canada
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(17), 2804; https://doi.org/10.3390/cancers17172804
Submission received: 28 June 2025 / Revised: 19 August 2025 / Accepted: 26 August 2025 / Published: 27 August 2025
(This article belongs to the Special Issue The Advance of Biomarker-Driven Targeted Therapies in Cancer)

Simple Summary

Epidermal growth factor receptor (EGFR) inhibitors are used to treat some patients with metastatic colorectal cancer, but many tumors do not respond or develop resistance during therapy. This review explores how understanding the molecular mechanisms of resistance can help guide treatment decisions. We highlight the concept of negative hyperselection, which uses extended genetic profiling to exclude tumors unlikely to benefit from EGFR inhibitors. In addition, we discuss the role of circulating tumor DNA, a blood-based test that monitors tumor evolution in real time. Together, these strategies form a precision oncology platform that supports more personalized and effective care. While these advances offer promise, challenges remain regarding cost, access, and demonstrating meaningful survival benefit in clinical practice.

Abstract

Epidermal growth factor receptor (EGFR) inhibitors remain a cornerstone in the treatment of metastatic colorectal cancer with RAS and BRAF wild-type cancer. Yet, primary and acquired resistance limit their benefit for many patients. A growing body of evidence reveals that resistance is not random but rather driven by a complex network of molecular alterations that sustain tumor growth independent of EGFR signaling. These include amplification of ERBB2 (HER2) and MET, activation of the PI3K and AKT pathways, EGFR extracellular domain mutations, and rare kinase fusions. The concept of negative hyperselection has emerged as a powerful strategy to refine patient selection by excluding tumors with these resistance drivers. Multiple clinical trials have consistently shown that patients who are hyperselected based on comprehensive molecular profiling achieve significantly higher response rates and improved survival compared to those selected by RAS and BRAF status alone. Liquid biopsy through circulating tumor DNA has further transformed this landscape, offering a noninvasive tool to capture tumor heterogeneity, monitor clonal evolution in real time, and guide rechallenge strategies after resistance emerges. Together, negative hyperselection, ctDNA-guided monitoring, and emerging therapeutics define a precision-oncology framework for identifying, tracking, and overcoming resistance to anti-EGFR therapy in mCRC, moving the field toward more effective and individualized care. Looking ahead, the development of innovative therapeutics such as bispecific antibodies, antibody drug conjugates, and RNA-based therapies promises to further expand in this challenging clinical scenario. These advances move precision oncology in colorectal cancer from concept to clinical reality, reshaping the standard of care through molecular insights.

1. Introduction

Colorectal cancer (CRC) is a major global health concern. GLOBOCAN 2020 estimates report close to 2,000,000 new cases and more than 900,000 deaths worldwide in a single year [1]. In the United States, the American Cancer Society projects 153,000 new diagnoses and 52,550 for the year 2023, including almost 20,000 cases under the age of fifty [2]. Although overall mortality continues to decline at roughly 2% per year, incidence in adults younger than 55 years has been rising by about one to two percent annually since the mid-nineteen nineties, and late-stage presentation is again increasing after years of progress toward earlier detection [3]. A gradual shift toward left-sided primary tumors has been noted, with rectal cancer now accounting for approximately one-third of new cases in the United States. This trend is both concerning and clinically relevant in the context of this review, as left-sided tumors typically demonstrate greater responsiveness to anti-EGFR therapy [3,4].
These changing epidemiologic patterns mirror a deeper reality: CRC is not a single homogeneous disease but a spectrum of molecularly distinct entities. This molecular heterogeneity has reshaped how metastatic CRC (mCRC) is treated, moving beyond one-size-fits-all approaches toward strategies guided by tumor biology [5,6]. International guidelines from societies such as the American Society of Clinical Oncology (ASCO), European Society of Clinical Oncology (ESMO), and National Comprehensive Cancer Network (NCCN) recommend routine testing for mutations in KRAS, NRAS, and BRAF [7,8,9]. Tumors that retain the wild-type presentation of these genes are considered eligible for epidermal growth factor receptor (EGFR) blockade with cetuximab or panitumumab, particularly in left-sided tumors [10,11]. Randomized trials demonstrate that the addition of an anti-EGFR antibody to fluoropyrimidine-based doublet or triplet chemotherapy improves response rate and progression-free survival (PFS) compared with chemotherapy alone, and can extend overall survival (OS) in selected patients [12,13].
Identifying the patients most likely to benefit from anti-EGFR antibodies and continuously monitoring the emergence of resistance drivers represents one important frontier in mCRC research and care. This approach allows for the selection of patients who are more likely to respond, while avoiding the administration of ineffective and potentially toxic therapy to those unlikely to benefit. A considerable fraction of patients with KRAS, NRAS, and BRAF wild-type tumors derive little or no advantage from anti-EGFR therapy, and acquired resistance commonly emerges within months of treatment initiation [14,15]. Multiple biological mechanisms account for this variability [15]. The growing recognition of resistance mechanisms has led to the development of more refined strategies to optimize patient selection.
Negative hyperselection is an emerging approach designed to refine patient selection for anti-EGFR therapy in mCRC. It expands molecular profiling beyond the standard evaluation of RAS and BRAF, aiming to identify additional biomarkers of primary resistance, such as ERBB2 amplification, MET amplification, and rare oncogenic fusions before treatment begins [16]. By excluding tumors harboring these alterations, negative hyperselection seeks to enrich the treatment population for tumors with true EGFR dependency, improving the precision of therapeutic decisions. In parallel, real-time analysis of circulating tumor DNA offers a complementary strategy to monitor the emergence of resistance during therapy, supporting decisions on rechallenge and informing treatment adaptation over time [17].
This review examines how molecular profiling has established a precision oncology platform for EGFR-targeted therapy in mCRC, with a particular focus on the evolving paradigm of negative hyperselection. We discuss the biological basis of primary and acquired resistance, explore how ctDNA complements tissue-based testing, and reflect on how these advances are shaping the broader landscape of mCRC care.

2. The EGFR Pathway in Colorectal Cancer

The EGFR plays a central role in regulating cell growth, survival, and proliferation in mCRC. EGFR is a single-pass membrane glycoprotein that couples extracellular growth factor cues to intracellular programmers of proliferation and survival [18]. In its inactive state, the extracellular domain adopts a folded conformation that holds the receptor in a monomeric and catalytically silent configuration [19,20].
Activation begins when ligands such as epidermal growth factor, transforming growth factor alpha, amphiregulin, or epiregulin bind to the extracellular domain. This binding triggers an untethering of the receptor structure, enabling it to form dimers, either with another EGFR molecule or with a partner in the ERBB family, such as HER2. Dimerization brings together the intracellular kinase domains into an asymmetric alignment, where one kinase lobe activates the other, leading to autophosphorylation of tyrosine residues on the cytoplasmic tail [21,22].
These phosphorylated tyrosines act as docking sites for a variety of adaptor proteins that initiate multiple downstream signaling cascades [23]. One major route involves the adaptor protein GRB2 and the guanine exchange factor SOS, which together activate RAS by facilitating GTP loading. This triggers the RAF–MEK–ERK cascade, driving gene transcription necessary for cell cycle progression and proliferation [24]. Concurrently, the PI3K–AKT pathway is engaged. The p85 subunit of phosphatidylinositol 3-kinase binds to other phosphorylated tyrosines, leading to the production of phosphatidylinositol trisphosphate and subsequent activation of AKT. This arm supports metabolic reprogramming, protein synthesis, and resistance to apoptosis, which are key features of tumor survival [25].
Additional branches of the pathway contribute to tumor behavior. Recruitment of phospholipase C gamma generates diacylglycerol and inositol trisphosphate, elevating intracellular calcium and activating protein kinase C, which is involved in regulating cell motility [26]. Simultaneously, phosphorylation events mediated by SRC family kinases facilitate STAT protein docking. Activated STAT3 dimers then translocate to the nucleus, where they promote the expression of key genes including MYC, BCL-xL, and vascular endothelial growth factor (VEGF), further enhancing proliferation, survival, and angiogenesis [18,27].
These convergent routes provide a powerful engine for malignant transformation when feedback checks are lost. Immunohistochemistry detects EGFR protein in 95% of CRCs and intense staining in nearly 80%, whereas true gene amplification is found in fewer than 10%. Overexpression together with abundant autocrine ligand promotes sustained signaling that correlates with poor differentiation, early relapse, and reduced survival [28]. However, despite its frequent expression, EGFR is not directly tested to guide anti-EGFR therapy, as expression levels do not predict response. Instead, the absence of mutations in KRAS, NRAS, and BRAF and other mechanisms determine tumor dependency on EGFR signaling.
Activating mutations in KRAS or NRAS bypass the need for EGFR signaling by locking the RAF, MEK ERK pathway in a permanently active state. Similarly, the BRAFV600E mutation acts further downstream to provide a continuous growth signal independent of receptor activation [29,30]. In parallel, alterations in PIK3CA or loss of PTEN hyperactivate the AKT pathway, promoting survival and resistance to apoptosis [31,32]. These molecular alterations not only drive tumor progression but also predict poor response to EGFR-targeted therapies, significantly influencing both prognosis and treatment outcomes [33,34].

Targeting the EGFR Pathway

To understand how a precision oncology paradigm for EGFR inhibitors has emerged, we must first revisit the mechanism of action of these drugs. Recognizing that EGFR’s extracellular domain is both accessible and central to this cascade provided the rationale for developing high-affinity monoclonal antibodies that occupy the ligand pocket, prevent dimerization, and promote receptor down-regulation [35]. Cetuximab is a chimeric immunoglobulin G-one molecule; panitumumab is a fully human immunoglobulin G-two molecule with lower intrinsic immunogenicity [36,37]. Both bind domain III and mediate their antitumor effect through three complementary mechanisms: blockade of ligand-induced signaling, lysosomal degradation of cell-surface receptors, and engagement of immune effector cells through the Fc region (antibody-dependent cellular cytotoxicity is more pronounced with cetuximab because of its IgG-one backbone) [36,38].
In the past decades, early single-arm studies demonstrated activity in chemotherapy-refractory disease, paving the way for randomized evaluations [39,40]. The BOND trial showed that adding cetuximab to irinotecan after prior failure doubled objective response to 23% and tripled PFS to 4.1 months [41]. In the first-line CRYSTAL study, cetuximab combined with FOLFIRI raised response from 40% to 59% and extended PFS from 8 to 10 months among tumors harboring wild-type KRAS [42]. The PRIME study established a similar benefit for panitumumab plus FOLFOX, where PFS improved from 7.4 to 9.6 months and overall survival from 19.7 to 23.9 months in extended RAS-wild-type disease [43]. Direct comparison in the ASPECCT trial confirmed similar outcomes for these drugs, with OS of 10.4 months for panitumumab and 10 months for cetuximab, and fewer severe infusion reactions with the fully human antibody [44].
However, it was noted that the clinical benefit was strictly contingent on molecular context [45]. Analyses of every pivotal study showed no advantage in tumors carrying activating mutations in KRAS or NRAS exons 2, 3, or 4, or BRAFV600E [46,47]. In left-sided primaries that retain wild-type RAS and BRAF, cetuximab or panitumumab combined with doublet chemotherapy achieves median OS beyond 35 months, surpassing outcomes obtained with bevacizumab-containing regimens [48]. In right-sided disease, the same antibodies failed to outperform the vascular–endothelial–growth-factor blockade, even when molecularly selected, an observation ascribed to differential embryologic origin, microbiome composition, and cytokine milieu [49,50].
Nowadays, for patients with left-sided, RAS-wild-type, and BRAF-wild-type metastatic disease, doublet chemotherapy combined with cetuximab or panitumumab is a category-one option. Right-sided disease or tumors harboring RAS mutations are better served by chemotherapy plus bevacizumab or, when microsatellite instability is present, immune checkpoint blockade. Tumors with BRAF V600E mutations and without microsatellite instability should be treated with FOLFOX, encorafenib, and cetuximab according to the recently published BREAKWATER trial [51]. Additionally, antibody–drug conjugates (ADCs) that link EGFR recognition to cytotoxic payloads, bispecific antibodies that co-target EGFR and c-MET or EGFR and HER3, and small interfering RNA nanocomplexes that selectively silence mutant RAS are entering early-phase trials, while their results are awaited [52,53,54,55,56,57].

3. Mechanisms of Primary Resistance to Anti-EGFR Therapy

Following the demonstration of clinical efficacy with EGFR-targeted agents in selected mCRC patients, efforts were made toward mapping the mechanisms of intrinsic resistance to Cetuximab and Panitumumab. Primary resistance is the failure to achieve clinical benefit from anti-EGFR therapy by pre-existing tumor features or genetic alterations that activate downstream signaling independent of EGFR stimulation (Figure 1). Primary resistance is often driven by mutations in the RAS gene family (KRAS and NRAS). Activating mutations in KRAS or NRAS occur in approximately 40% of mCRC cases, with strong concordance between primary tumors and distant metastases [18,58]. Activating mutations at KRAS codons 12 and 13 (exon 2), as well as in KRAS exons 3–4 and NRAS exons 2–4, impair GTP hydrolysis and lock RAS in its active, GTP-bound form [59]. This constitutive activation drives continuous signaling through the RAF–MEK–ERK (MAPK) pathway, bypassing any blockade of EGFR at the cell surface. Consequently, even if cetuximab or panitumumab successfully inhibits EGFR, the MAPK pathway remains fully active and continues to drive CRC tumor proliferation [60]. Currently, an ongoing phase 3 first-line trial (NCT06252649, CodeBreaK 301) is evaluating whether the addition of anti-KRAS targeted therapy (Sotorasib) to the first line setting is a possible strategy to overcome resistance (Table 1) [61]
Similarly, the BRAFV600E mutation bypasses EGFR inhibition by locking the RAF–MEK–ERK cascade in an active state, marking a constitutive resistance. A meta-analysis of 10 randomized control trial demonstrated that neither cetuximab nor panitumumab, alone or combined with chemotherapy, improved OS (HR, 0.91; 95% CI, 0.62–1.34; p = 0.63), PFS (HR, 0.88; 95% confidence interval (CI), 0.67–1.14; p = 0.33) or (relative risk, 1.31; 95% CI 0.83–2.08, p = 0.25) in BRAF-mutant mCRC compared with chemotherapy ± bevacizumab [62]. In a retrospective subgroup of the FIRE-3 trial, patients with BRAF-mutant tumors achieved almost identical median OS and PFS whether treated with FOLFIRI plus cetuximab or FOLFIRI plus bevacizumab [63]. The phase 1 FIRE-4.5 study then directly compared first-line FOLFOXIRI plus bevacizumab against FOLFOXIRI plus cetuximab in BRAFV600E-mutant mCRC, finding significantly longer PFS (6.7 months v 10.7 months; HR, 1.89; 95% CI, 1.19 to 3.01, 1.89; p = 0.006) with bevacizumab despite similar response rates [64]. In recognition of this clinical significance difference, NCCN and ESMO guidelines now recommend testing for KRAS, NRAS, and BRAFV600E mutations to guide the selection of first-line and subsequent therapies [9,65].

3.1. HER2 (ERBB2) Amplification and Mutations

HER2 amplification is a well-established mechanism of resistance to anti-EGFR therapy, occurring in approximately 5–7% of RAS/BRAF wild-type mCRC cases [66]. HER2 overexpression actiular sequencing, such as NGSvates downstream signaling, independently of EGFR, bypassed the necessity for EGFR signaling and thus rendered EGFR inhibitors ineffective [67,68]. This pathway redundancy resistance strategy occurs when an amplification of a parallel receptor compensates for the inhibition of EGFR [69]. Clinically, this resistance manifests as a marked reduction in the efficacy of anti-EGFR agents among patients harboring HER2 amplification, correlating inferior response compared to HER2-negative counterparts. A recent meta-analysis of five retrospective cohort studies involving 594 patients with RAS wild-type mCRC demonstrated that HER2-positive tumors treated with anti-EGFR agents exhibited significantly worse PFS (HR 2.84; 95% CI, 1.44–5.60) and ORR (HR 1.96; 95% CI 1.10–3.48) [70].
In response to this clinical challenge, recent trials have explored targeted therapies against HER2 in mCRC. These include antibody-drug conjugates such as trastuzumab deruxtecan (T-DXd), evaluated in the DESTINY-CRC01 trial, which demonstrated an overall response rate of 30.2% and a median PFS of 5.6 months in patients with HER2-positive mCRC refractory to standard therapies, including anti-EGFR agents [71]. Additionally, the HERACLES trial evaluated trastuzumab plus lapatinib, the MyPathway study investigated trastuzumab plus pertuzumab, and the MOUNTAINEER trial assessed trastuzumab plus tucatinib, all demonstrating meaningful responses in heavily pretreated HER2-positive mCRC [72,73,74]. Furthermore, the phase II TRIUMPH (EPOC1602) trial evaluated pertuzumab plus trastuzumab in RAS wild-type, HER2-amplified mCRC identified by tissue or ctDNA and met its primary endpoint, with confirmed ORRs of 30% (95% CI, 14–50) in the tissue-positive cohort and 28% (12–49) in the ctDNA-positive cohort; median PFS/OS were 4.0/10.1 and 3.1/8.8 months, respectively [75,76].
Collectively, these studies support the clinical validity of HER2 as a therapeutic target in CRC and reinforce the critical importance of routine HER2 testing to guide treatment decisions. HER2-directed therapies are rapidly becoming a new standard of care for this molecularly defined subset [77].

3.2. PI3K Pathway Alterations (PIK3CA Mutations, PTEN Loss)

PIK3CA mutations and PTEN loss activate the PI3K/AKT pathway, promoting tumor survival through mechanisms independent of EGFR [78,79]. These alterations lead to the activation of alternative survival pathways that undermine the efficacy of EGFR-targeted therapies. They are found in approximately 10–20% of CRC, with hotspots in exon 9 and exon 20 [80]. Additionally, loss of function of the tumor suppressor PTEN, which normally antagonizes PI3K signaling, occurs in about 30% of sporadic CRCs and similarly predicts a lack of response to anti-EGFR treatment [81,82].
The prognostic role of PIK3CA mutations were evaluated in a recent meta-analysis of four randomized controlled trials [83]. PIK3CA mutations showed no prognostic significance in mCRC. In the control arms without anti-EGFR therapy, OS (HR = 1.11; 95% CI, 0.80–1.55; p = 0.54) and PFS (HR = 1.10; 95% CI, 0.72–1.68; p = 0.66) did not differ significantly between PIK3CA mutant and wild-type tumors. These findings indicate that PIK3CA mutations do not impact the natural history of the disease, independent of treatment.
In contrast, the predictive value of PIK3CA mutations emerged in the context of anti-EGFR therapy. Patients harboring wild-type PIK3CA tumors experienced a significant improvement in PFS with anti-EGFR antibodies (HR = 0.57; 95% CI, 0.38–0.87; p < 0.01). Conversely, patients with PIK3CA mutant tumors derived no significant PFS benefit from anti-EGFR treatment (HR = 0.70; 95% CI, 0.26–1.88; p = 0.48). No significant OS benefit was observed in either wild-type (HR = 0.81; p = 0.29) or mutant (HR = 0.87; p = 0.62) subgroups. These data indicate that PIK3CA mutations may modulate sensitivity to anti-EGFR therapy by affecting PFS. Therefore, while PIK3CA mutations do not alter overall prognosis, they may serve as biomarkers of resistance to anti-EGFR agents.

3.3. MET Amplification

Amplification of the MET proto-oncogene, encoding another receptor tyrosine kinase, represents an additional mechanism of bypass signaling [84]. This amplification functions as a parallel signaling route that effectively bypasses the blockade of EGFR, sustaining tumor growth despite anti-EGFR therapy. Interestingly, the traditional expansion of ‘MET’ as ‘mesenchymal–epithelial transition factor’ is a common misnomer [85]; the official HGNC designation is ‘MET proto-oncogene,’ and the original ‘met’ acronym was a neutral label from the MNNG-HOS transforming gene [86].
Similarly to HER2, MET overexpression can activate downstream pathways independently of EGFR, leading to resistance. MET activation triggers essential signaling pathways, such as RAS-ERK-MAPK, PI3K-AKT-mTOR, Wnt/β-catenin, and STAT, that drive cellular survival and proliferation and has been proposed as a mechanism to bypass the EGFR-blockade [84]. Therefore, MET amplification is included as a resistance marker in hyperselection panels like PRESSING [87].
MET amplification is rare, occurring in 1–2% of CRC tumors [14,88]. However, in patients resistant to anti-EGFR therapy, this mutation prevalence is significantly higher, reaching 22.6% (95% CI: 13.31–35.67%) [89]. A phase II study tested tivantinib plus cetuximab in 41 patients with anti-EGFR resistance and MET overexpression. The initial phase showed a disease control rate (DCR) of 52.4%, but the trial missed its primary endpoint in the second phase, with only four patients achieving an objective response versus the expected five. Despite this, survival outcomes were encouraging, with a median PFS of 2.6 months and a median OS of 9.2 months [90].

3.4. MAP2K1 (MEK1) Mutations

MAP2K1 is the gene that encodes the protein MEK1, a serine/threonine protein kinase that acts downstream of RAS in the RAS-RAF-MEK-ERK pathway [91]. These mutations drive resistance to anti-EGFR therapies by constitutively activating the MAPK pathway independent of EGFR-inhibition [92]. A recent real-world study screened 246 mCRC patients and found that 2 out of 112 (1.8%) MSS, RAS/BRAF wild-type patients harbored the MAP2K1 K57N mutation. Both patients received panitumumab monotherapy as third-line treatment and showed primary resistance, with clinical benefit observed only when anti-EGFR agents were combined with cytotoxic chemotherapy [93]. Additionally, although clinical trials have shown the efficacy of combining anti-MEK and anti-EGFR therapies in mCRC with MEK-driven resistance, data specifically addressing resistance in patients with MAP2K1 mutations remain scarce [94].

3.5. AKT1 Mutations

AKT1 is a serine/threonine kinase involved in the PI3K-AKT-mTOR pathway. Activating mutations in AKT1 are rare in mCRC, found in less than 2% of cases [95]. These mutations enable alternative signaling pathways that bypass EGFR dependency, contributing to resistance against anti-EGFR treatments [96]. AKT1 mutations frequently co-occur with other genomic alterations within the PI3K-AKT pathway or gene amplifications, further complicating therapeutic strategies. A retrospective cohort study of the clinicopathologic and molecular profile to analysis of AKT1 E17K-mutated CRC revealed a strong association with mucinous histology (p = 0.0006) and frequent co-occurrence with BRAF V600E and KRAS mutations (p = 0.0001) [97]. In this study, 83.3% of AKT1-mutated cases harbored concurrent RAS/RAF alterations, and two patients with wild-type RAS and BRAF demonstrated primary resistance to cetuximab, suggesting that AKT1 mutations may independently contribute to anti-EGFR resistance.
Patients harboring AKT1 activating mutations often exhibit inferior response outcomes [98]. Many AKT inhibitors, AZD5363, GDC-0068, and MK2006, have been tested in early clinical trials [99,100,101]. Investigation of these drugs has focused on PTEN-deficient cancers as well as AKT1- and PIK3CA-mutant tumors.

3.6. Gene Fusions

Gene fusions are structural genomic alterations where segments from two different genes become abnormally joined, typically resulting in the expression of a hybrid oncogenic protein. NTRK and RET fusions are distinct gene rearrangements involving receptor tyrosine kinases that lead to constitutive activation of downstream signaling pathways in mCRC [102,103]. NTRK fusions involve the TRK family (NTRK1–3) and are pan-tumor biomarkers effectively targeted by TRK-inhibitors such as larotrectinib and entrectinib [104]. For example, larotrectinib has shown tumor-agnostic efficacy in a pooled analysis of three trials (adult phase 1, pediatric phase 1–2, adult/adolescent phase 2), with an overall response rate of 75% (95% CI, 61–85) by independent review; at 1 year, 71% of responses were ongoing and 55% of patients remained progression-free, with no discontinuations due to drug-related adverse events [103].
RET fusions involve the RET (rearranged during transfection) kinase and are targeted by selective RET-inhibitors, including selpercatinib and pralsetinib [105]. Both gene rearrangements drive ligand-independent activation of the MAPK and PI3K-AKT signaling pathways, bypassing EGFR-inhibition and contributing to resistance to anti-EGFR therapies [106].
Supporting this idea, a large circulating tumor DNA (ctDNA) analysis of 3808 patients with advanced CRC detected gene fusions in 1.1% of cases, most commonly involving RET, FGFR3, and ALK [107]. These fusions were frequently subclonal and often co-occurred with RAS and EGFR mutations associated with anti-EGFR resistance. Notably, 78% of patients with clinical data had received EGFR-targeted therapy before fusion detection, reinforcing the hypothesis that these fusions may emerge as novel mechanisms of acquired resistance to anti-EGFR treatment in mCRC. Overall, identifying targetable gene fusions offers a path forward to expand options in mCRC—allowing a small but meaningful subset of patients to active, tumor-agnostic therapies [108].
Table 1. Primary mechanisms of intrinsic resistance to anti-EGFR therapy in metastatic colorectal cancer (mCRC).
Table 1. Primary mechanisms of intrinsic resistance to anti-EGFR therapy in metastatic colorectal cancer (mCRC).
Mechanism of Primary ResistancePrevalenceClinical Impact in Patients Treated with Anti-EGFR Therapy (Effect Size, 95% CI)Possible Strategies to Overcome ResistanceReferences
KRAS/NRAS activating mutations 40%OS: HR 1.06 (0.96–1.17)Addition of anti-KRAS targeted therapy (e.g., Code BreaK 301 trial)[61,109,110]
BRAF V600E mutations8–12%OS: HR 0.91 (0.62–1.34); PFS: HR 0.88 (0.67–1.14); ORR: RR 1.31 (0.83–2.08)Combined EGFR and BRAF inhibitors (e.g., BREAKWATER)[47,111,112]
HER2 (ERBB2) amplification5–7%PFS: HR 2.84 (1.44–5.60); ORR: OR 1.96 (1.10–3.48)HER2-targeted therapy (e.g., HERACLES-B trial, TRIUMPH trial)[71,72,75,113]
PIK3CA mutations/PTEN loss10–20%PFS: WT HR 0.57 (0.38–0.87) ; mutant HR 0.70 (0.26–1.88)Addition of PI3K or mTor inhibitors[73,81,82,114,115]
MET amplification1–2% baseline; 22% after anti-EGFROS: HR 1.33 (1.06–1.59); PFS: HR 1.47 (1.03–1.91) Dual EGFR/MET Inhibition (e.g., Tivantinib + Cetuximab)[85,90,116,117]
MAP2K1 (MEK1) mutations1.8% (MSS, RAS/BRAF WT cohort)Data remains limited to small case series/reportsCombined BRAF + EGFR + MEK1 inhibitiors[118,119,120]
AKT1 mutations<2%Data remains limited to small case series/reportsAKT inhibitors[94,121]
Oncogenic fusions (NTRK/RET/ALK/ROS1/FGFR3)1%Data remains limited to small case series/reportsFusion-targeted therapies (e.g., TRK, RET inhibitors)[96]
The table summarizes prevalence, clinical impact in anti-EGFR–treated cohorts (reported OS, PFS, ORR effect sizes with 95% CIs), and example strategies to overcome resistance. Abbreviations: OS, overall survival; PFS, progression-free survival; ORR, objective response rate; WT, wild type; NR, not reported; mTOR, mechanistic target of rapamycin; MSS, microsatellite stable. Notes: For PIK3CA, PFS benefit with anti-EGFR therapy is confined to WT tumors; no significant benefit is seen in PIK3CA-mutant disease. For MET, the listed HRs reflect the prognostic impact of high c-MET expression in CRC overall and are not anti-EGFR-specific estimates.

4. Mechanisms of Acquired Resistance to Anti-EGFR Therapy

Even among patients initially responsive to anti-EGFR therapy and without primary resistance, disease progression occurs. Intratumoral heterogeneity is a central feature of colorectal cancer and decisively influences this acquired resistance [108]. In fact, CRC tumor consists of a complex population of distinct cellular clones that, over time, accumulate various genetic and epigenetic alterations, including mutations in oncogenes and tumor suppressor genes, DNA methylation changes, and variations in microRNA expression [122,123]. This diversity results from a branched evolutionary process driven by genomic instability, which generates clonal variation, and by the natural selection of more adapted clones, favored by the pressures of the tumor microenvironment and applied therapies (Figure 2).
In the context of KRAS wild-type mCRC, we detailed previously that anti-EGFR therapy demonstrates significant efficacy [8]. However, intrinsic tumor heterogeneity and genomic instability promote the rapid emergence of resistant subclones, leading to a progressive decline in response and disease progression, with acquired resistance typically manifesting within 12 months. This phenomenon reflects a dynamic tumor ecosystem in which pre-existing or newly emerged subpopulations, endowed with adaptive advantages, expand under the selective pressure imposed by therapy [124]. The identification of the main molecular alterations responsible for this resistance has been a priority for the development of personalized therapeutic strategies, prompting a series of clinical trials aimed at understanding the mechanisms involved in acquired resistance [125,126,127].
Genetic alterations in KRAS, such as mutations and amplifications, are present in approximately 50% of cases and represent the main mechanism of acquired resistance in this scenario [128]. These alterations may arise from the expansion of pre-existing mutated clones or from the emergence of new mutations induced by the selective pressure exerted by prolonged use of agents such as cetuximab or panitumumab [129,130]. Moreover, they may coexist with other genomic alterations that promote the activation of alternative signaling pathways, further compromising the efficacy of the anti-EGFR blockade [131]. Although less frequent, secondary mutations also contribute significantly to acquired resistance to anti-EGFR therapy [132]. Among these, mutations in the extracellular domain III of EGFR are particularly notable, as this region is crucial for cetuximab recognition; such mutations impair antibody binding to the receptor and reduce its therapeutic efficacy [133]. Additionally, mutations in BRAF, a member of the RAF serine/threonine kinase family, promote continuous activation of the RAF-ERK pathway, constituting an important mechanism of acquired resistance [134,135]. ERBB2 amplifications, which encode the HER2 receptor, may also be induced by cetuximab treatment, resulting in persistent activation of the ERK1/2 pathway [136]. In parallel, the selective pressure exerted by anti-EGFR therapies favors the expansion of subclones with MET amplification, further contributing to acquired resistance [84]. The coexistence of these mechanisms underscores the need for a deeper understanding to develop more effective therapeutic approaches. Beyond genomic alterations, there is evidence that acquired resistance may also be mediated by transcriptomic profiles associated with epithelial–mesenchymal transition, characterizing a pattern of cross-resistance [137]. Additional preclinical data indicate that cancer-associated fibroblasts (CAF), through the secretion of various soluble factors, play a central role in inducing resistance to EGFR blockade [137,138]. Key CAF-derived factors implicated in anti-EGFR resistance include HGF (activates MET and sustains MAPK/PI3K–AKT signaling), TGF-β (promotes EMT and adaptive tolerance), IL-6/IL-8 (STAT3-mediated survival/inflammation), FGF2 (FGFR signaling), and neuregulin-1 (HER3/HER2 activation) [139]. By sustaining parallel pathways despite EGFR blockade, these stromal interactions highlight the importance of considering the tumor microenvironment as a therapeutic target [140,141]. These findings support the notion that therapeutic resistance mechanisms are not limited to genomic alterations in the molecular target but also include transcriptomic adaptations and complex interactions with the tumor microenvironment [60].

5. Hyperselection of Patients for Anti-EGFR Therapy

After elucidating the diverse and complex mechanisms underlying resistance to anti-EGFR therapy, it became clear that traditional selection criteria were insufficient to fully identify patients who would benefit from these agents. This realization prompted the emergence of a new precision oncology paradigm: negative hyperselection. Rather than selecting patients based solely on the presence of favorable biomarkers, this approach seeks to exclude tumors harboring known resistance drivers, even when conventional biomarkers suggest eligibility [10]. In other words, even after the traditional selection based on RAS and BRAF and tumor sidedness, approximately one-third of patients do not respond to EGFR-inhibition, indicating that additional molecular escape mechanisms limit therapeutic efficacy [142]. This approach exemplifies how a deep understanding of molecular mechanisms can directly inform clinical decision-making, bridging the gap between genomic insight and patient care.
As detailed in previous sections, alterations such as HER2 and MET amplifications, NTRK, ROS1, ALK, and RET fusions, PIK3CA exon 20 and MAP2K1 mutations, as well as PTEN loss, activate alternative signaling pathways that circumvent EGFR-blockade [143]. Mapping the presence of these alterations before treatment initiation could enhance patient selection.
In this context, the PRESSING case–control study was landmark in expanding the understanding of molecular mechanisms behind resistance to this therapy. It evaluated whether a selected panel of genomic alterations (the PRESSING panel) could predict resistance to anti-EGFR antibodies beyond common RAS and BRAF mutations. The panel included HER2 and MET amplifications, fusions involving NTRK, ROS1, ALK, and RET, and PIK3CA exon 20 mutations. The key finding was that PRESSING-negative patients (without these alterations) showed significantly greater clinical benefit, with response rates potentially increasing from 50% to 67% and longer PFS (median 6.3 vs. 2.7 months). Although OS was not significantly affected, these alterations appear important in predicting treatment resistance.
Furthermore, tumor sidedness retained its prognostic impact even after molecular hyperselection: right-sided tumors displayed a higher frequency of resistance-associated alterations (41.4% vs. 20%) and poorer response to treatment (PFS of 3.3 vs. 7.3 months). The combination of tumor sidedness with the PRESSING panel increased the predictive accuracy to approximately 80%, underscoring the importance of integrating both anatomical and molecular variables in therapeutic decision-making. The PRESSING panel also demonstrated that microsatellite instability (MSI-high), present exclusively in resistant patients, is strongly associated with these alterations and predominates in right-sided tumors, highlighting the molecular complexity of mCRC.
Building on these promising results, new studies have explored the clinical role of negative hyperselection in managing mCRC. Notably, the randomized phase II PANDA trial evaluated chemotherapy plus anti-EGFR therapy in elderly patients with RAS/BRAF wild-type mCRC, stratified by a modified PRESSING panel that included MAP2K1 mutations and PTEN loss expression [16]. Molecular hyperselection was independently linked to significant improvements in PFS (12.8 vs. 7.6 months) and OS (29.5 vs. 20 months), a benefit not seen in the original PRESSING study. These findings confirm the clinical importance of these rare mutations in EGFR resistance and support hyperselection as a strong molecular strategy to identify ideal candidates for anti-EGFR therapy in clinical practice.
Exploratory analyses of the randomized phase II PanaMa trial evaluated the role of negative hyperselection in maintenance therapy with panitumumab in patients with RAS wild-type mCRC [140]. All participants received induction with mFOLFOX6 combined with panitumumab, followed by randomization to maintenance with chemotherapy alone or in combination with the anti-EGFR agent. The results confirmed the prognostic value of negative hyperselection, evidenced by superior PFS and OS in patients without mutations compared to those with at least one identified alteration (median OS: 28.7 vs. 22.2 months; significant). In a comparative context of maintenance with or without panitumumab, negative hyperselection proved to be an effective predictive biomarker capable of identifying patients with the greatest potential benefit.
Taking a step further, current studies aim to enhance both the precision and the breadth of detectable mutations. The PRESSING2 study employed an expanded molecular panel to identify uncommon genomic alterations in RAS wild-type mCRC patients undergoing anti-EGFR therapy [87]. This panel incorporated potential novel oncogenic alterations relative to the original PRESSING panel, including mutations associated with the MAPK pathway, PIK3CA, EGFR-independent tyrosine kinase signaling routes, and EGFR rearrangements affecting the tyrosine kinase domain. PRESSING2-positive patients exhibited significantly worse survival, suggesting that an expanded molecular panel may refine the selection process for anti-EGFR therapy. Table 2 presents the main clinical studies evaluating the role of negative hyperselection.

6. The Role of Circulating Tumor DNA in Tracking Resistance to Anti-EGFR Therapy

While negative hyperselection enhances baseline patient stratification, resistance to anti-EGFR therapy often emerges dynamically under treatment pressure. This has positioned liquid biopsy and ctDNA as a critical component of the precision oncology platform for mCRC treatment.
Historically, molecular profiling for CRC has relied on analyzing tumor tissue obtained via biopsy or surgical resection (from either the primary tumor or a metastatic site) [85]. Formalin-fixed paraffin-embedded samples remain the reference for detecting oncogenic drivers and copy number alterations, such as HER2 or MET amplification. Yet tissue captures only one spatial and temporal frame of a disease that is intrinsically heterogeneous and continually evolving under therapeutic pressure. Repeated biopsy is often impractical, sometimes impossible, and it cannot reveal the dynamic rise and fall of subclones that ultimately determine clinical outcome [147]. Despite its capabilities, tissue-based next generation sequencing (NGS) has several inherent limitations, such as intratumor heterogeneity, lack of analysis of dynamic changes and clonal evolution that occur over time (particularly in the presence of therapy), and practical execution difficulties due to challenges in obtaining sufficient quantity and quality of tumor tissue, often compounded by tumor location or the patient’s condition [122,148].
ctDNA, small DNA fragments shed by tumors into the bloodstream, allows for non-invasive liquid biopsies for genomic profiling via molecular sequencing, such as NGS [149]. Compared to tissue biopsies, ctDNA analysis offers several advantages: it is non-invasive and repeatable, can better capture tumor heterogeneity by sampling DNA from multiple sites, enables real-time monitoring of tumor evolution and treatment response (including early detection of resistance), serves as a sensitive marker for minimal residual disease (MRD) post-treatment to predict recurrence, and potentially offers faster turnaround times [150,151,152,153].
In anti-EGFR therapy for mCRC, ctDNA is valuable for baseline stratification (detecting primary resistance like RAS or BRAF when tissue is limited), monitoring response and resistance (detecting changes in mutation frequency and emergence of new resistance mechanisms like EGFR ECD mutations), and guiding rechallenge strategies based on the presence or absence of prior resistance mutations [60,154,155] (Figure 3). In a recent meta-analysis including 402 patients retreated with cetuximab or panitumumab after an EGFR-free interval, the pooled objective response rate was 20.5% and the disease control rate reached 67.4%. The median PFS and OS were estimated at 3.5 and 9.8 months, respectively. Notably, patients selected for rechallenge based on the absence of RAS mutations in ctDNA experienced significantly improved survival, with a pooled hazard ratio for death of 0.41 compared to those with detectable RAS mutations. These findings underscore the clinical utility of ctDNA not only as a biomarker but as a real-time decision-making instrument in the management of anti-EGFR resistance [17].
Despite its promise, ctDNA analysis presents several limitations currently. Its sensitivity is affected by tumor biology and assay performance [156,157,158]. False-negative results may occur in cases with low tumor burden, limited tumor shedding, or metastases located in compartments with restricted access to the bloodstream, such as the peritoneum or central nervous system [159,160]. Turnaround time, although generally shorter than that of tissue-based sequencing, may still be suboptimal due to logistical delays in sample handling and reporting [161,162]. Financial and infrastructural barriers also limit access to ctDNA testing in many healthcare settings [163].
Clinically, the interpretation of ctDNA results is not always straightforward. The presence of resistance mutations does not uniformly indicate a lack of benefit, and absence of detectable alterations does not ensure durable response. Outside of validated settings such as RAS testing for anti-EGFR therapy or minimal residual disease detection, the impact of ctDNA-guided interventions on overall survival remains to be established [164].
In summary, resistance to anti-EGFR therapy, whether intrinsic or acquired, remains a major obstacle in the treatment of metastatic colorectal cancer. This challenge has driven the development of more refined molecular stratification strategies, including the concept of negative hyperselection, which seeks to exclude tumors that harbor additional resistance drivers beyond RAS and BRAF mutations. In parallel, circulating tumor DNA has emerged as a valuable tool for real-time monitoring of clonal evolution during therapy, capturing the dynamic emergence of acquired resistance and informing decisions such as anti-EGFR rechallenge. Concurrently, emerging modalities, including bispecific antibodies, EGFR-directed antibody–drug conjugates, and RNA-based therapeutics will expand the options for patients with resistant disease [53,158,165,166]. Linking the targetable molecular alterations to matched strategies enables more personalized care. Together, these approaches illustrate how precision oncology is transforming the management of colorectal cancer by replacing static, one-time molecular assessments with a continuous and adaptive model of care.
Selecting patients most likely to benefit from EGFR blockade, directing others to alternative targeted options, and ultimately aiming for improved survival and quality of life while minimizing unnecessary toxicity seems to be the goal in this setting. This paradigm shift must be accompanied by careful consideration of cost effectiveness, demonstrated survival benefit, and equitable access. As the field advances, integrating molecular complexity into routine practice must be balanced with the responsibility to ensure that innovations translate into meaningful improvements in patient outcomes.

7. Conclusions

Optimizing anti-EGFR therapy in mCRC demands more than RAS and BRAF testing. A growing body of evidence shows that resistance arises from a complex network of alterations, including HER2 and MET amplification, PI3K pathway activation, and rare oncogenic fusions. The concept of negative hyperselection, which systematically excludes these resistance drivers, has consistently translated into improved response rates and survival across multiple clinical trials. Liquid biopsy has further transformed this landscape. Circulating tumor DNA enables real-time tracking of clonal evolution, early detection of resistance, and more effective rechallenge strategies. This approach offers a dynamic complement to tissue-based profiling and a pragmatic framework for real-world decision-making, with the potential to improve outcomes while reducing ineffective exposure, toxicity, and costs.
The future of EGFR-targeted therapy lies at the intersection of deeper molecular refinement and adaptive treatment strategies. Emerging modalities such as bispecific antibodies, antibody drug conjugates, and RNA-based therapeutics promise to expand treatment options for patients previously considered resistant. Successful implementation of liquid biopsy will require validated and standardized assays, timely turnaround, equitable access, and prospective evidence demonstrating survival benefit and cost-effectiveness. Precision oncology in CRC is no longer aspirational. It is becoming the standard, driven by molecular insight and continuous innovation.

Author Contributions

L.F.L.: conceptualization, data curation, methodology, writing—original draft preparation. M.M.N.: conceptualization, writing—original draft preparation. L.F.C.A.: methodology, writing—original draft, software. L.D.d.C.: methodology. J.S.A.d.M.: data curation. A.P.C.: data curation, visualization. M.B.: supervision. T.B.d.C.: writing—reviewing and editing. R.D.P.: visualization, supervision, writing—reviewing and editing. T.B.C.M.: visualization, supervision, writing—reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study is a narrative review of published data and does not involve human participants. Therefore, ethical approval was not required.

Informed Consent Statement

This study is a narrative review of published data and does not involve human participants. Therefore, informed consent was not required.

Data Availability Statement

The data underlying this study are extracted from publicly available studies.

Conflicts of Interest

The authors declare no competing interests related to this work. All authors take responsibility for the accuracy and integrity of the data presented and their interpretation. Tiago Biachi de Castria receives an honorarium from Bristol Myers Squibb, Eli Lilly, Merck Sharp & Dohme Corp., AstraZeneca and A2Bio.

References

  1. GBD 2019 Colorectal Cancer Collaborators. Global, regional, and national burden of colorectal cancer and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol. Hepatol. 2022, 7, 627–647. [Google Scholar] [CrossRef]
  2. Siegel, R.L.; Wagle, N.S.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 233–254. [Google Scholar] [CrossRef]
  3. Sung, H.; Siegel, R.L.; Laversanne, M.; Jiang, C.; Morgan, E.; Zahwe, M.; Cao, Y.; Bray, F.; Jemal, A. Colorectal cancer incidence trends in younger versus older adults: An analysis of population-based cancer registry data. Lancet Oncol. 2025, 26, 51–63. [Google Scholar] [CrossRef]
  4. Szostek, J.; Serafin, M.; Mąka, M.; Jabłońska, B.; Mrowiec, S. Right-Sided Versus Left-Sided Colon Cancer-A 5-Year Single-Center Observational Study. Cancers 2025, 17, 537. [Google Scholar] [CrossRef]
  5. Guo, L.; Wang, Y.; Yang, W.; Wang, C.; Guo, T.; Yang, J.; Shao, Z.; Cai, G.; Cai, S.; Zhang, L.; et al. Molecular Profiling Provides Clinical Insights into Targeted and Immunotherapies as Well as Colorectal Cancer Prognosis. Gastroenterology 2023, 165, 414–428.E7. [Google Scholar] [CrossRef]
  6. Napolitano, S.; Martini, G.; Ciardiello, D.; Tufo, S.D.; Martinelli, E.; Troiani, T.; Ciardiello, F. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol. Hepatol. 2024, 9, 664–676. [Google Scholar] [CrossRef]
  7. Morris, V.K.; Kennedy, E.B.; Baxter, N.N.; Benson, A.B.; Cercek, A.; Cho, M.; Ciombor, K.K.; Cremolini, C.; Davis, A.; Deming, D.A.; et al. Treatment of Metastatic Colorectal Cancer: ASCO Guideline. J. Clin. Oncol. 2023, 41, 678–700. [Google Scholar] [CrossRef] [PubMed]
  8. Cervantes, A.; Adam, R.; Roselló, S.; Arnold, D.; Normanno, N.; Taïeb, J.; Seligmann, J.; De Baere, T.; Osterlund, P.; Yoshino, T.; et al. Metastatic colorectal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 10–32. [Google Scholar] [CrossRef] [PubMed]
  9. Benson, A.B.; Venook, A.P.; Adam, M.; Chang, G.; Chen, Y.-J.; Ciombor, K.K.; Cohen, S.A.; Cooper, H.S.; Deming, D.; Garrido-Laguna, I.; et al. Colon Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2024, 22, e240029. [Google Scholar] [CrossRef] [PubMed]
  10. Kasi, P.M.; Afable, M.G.; Herting, C.; Lukanowski, M.; Jin, Z. Anti-EGFR Antibodies in the Management of Advanced Colorectal Cancer. Oncologist 2023, 28, 1034–1048. [Google Scholar] [CrossRef]
  11. Tejpar, S.; Stintzing, S.; Ciardiello, F.; Tabernero, J.; Van Cutsem, E.; Beier, F.; Esser, R.; Lenz, H.-J.; Heinemann, V. Prognostic and Predictive Relevance of Primary Tumor Location in Patients with RAS Wild-Type Metastatic Colorectal Cancer: Retrospective Analyses of the CRYSTAL and FIRE-3 Trials. JAMA Oncol. 2017, 3, 194–201. [Google Scholar] [CrossRef]
  12. Van Cutsem, E.; Lenz, H.-J.; Köhne, C.-H.; Heinemann, V.; Tejpar, S.; Melezínek, I.; Beier, F.; Stroh, C.; Rougier, P.; van Krieken, J.H.; et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol. 2015, 33, 692–700. [Google Scholar] [CrossRef]
  13. Bokemeyer, C.; Van Cutsem, E.; Rougier, P.; Ciardiello, F.; Heeger, S.; Schlichting, M.; Celik, I.; Köhne, C.-H. Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: Pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur. J. Cancer 2012, 48, 1466–1475. [Google Scholar] [CrossRef]
  14. Misale, S.; Di Nicolantonio, F.; Sartore-Bianchi, A.; Siena, S.; Bardelli, A. Resistance to anti-EGFR therapy in colorectal cancer: From heterogeneity to convergent evolution. Cancer Discov. 2014, 4, 1269–1280. [Google Scholar] [CrossRef]
  15. Martinelli, E.; Ciardiello, D.; Martini, G.; Troiani, T.; Cardone, C.; Vitiello, P.P.; Normanno, N.; Rachiglio, A.M.; Maiello, E.; Latiano, T.; et al. Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: Challenges and future perspectives. Ann. Oncol. 2020, 31, 30–40. [Google Scholar] [CrossRef]
  16. Dienstmann, R.; Salazar, R.; Tabernero, J. Overcoming Resistance to Anti-EGFR Therapy in Colorectal Cancer. Am. Soc. Clin. Oncol. Educ. Book 2015, 35, e149–e156. [Google Scholar] [CrossRef] [PubMed]
  17. Pietrantonio, F.; Bergamo, F.; Rossini, D.; Ghelardi, F.; De Grandis, M.C.; Germani, M.M.; Barsotti, G.; Formica, V.; Frassineti, G.L.; Boscolo, G.; et al. Negative hyperselection of elderly patients with RAS and BRAF wild-type metastatic colorectal cancer receiving initial panitumumab plus FOLFOX or 5-FU/LV. Eur. J. Cancer 2023, 195, 113396. [Google Scholar] [CrossRef] [PubMed]
  18. da Silva, L.F.L.; Saldanha, E.F.; da Conceição, L.D.; Noronha, M.M.; da Silva, M.V.M.G.; Peixoto, R.D. ’Alpino Anti-EGFR Rechallenge in Metastatic Colorectal Cancer and the Role of ctDNA: A Systematic Review and Meta-analysis. J. Gastrointest. Cancer 2024, 56, 28. [Google Scholar] [CrossRef] [PubMed]
  19. Li, Q.; Geng, S.; Luo, H.; Wang, W.; Mo, Y.-Q.; Luo, Q.; Wang, L.; Song, G.-B.; Sheng, J.-P.; Xu, B. Signaling pathways involved in colorectal cancer: Pathogenesis and targeted therapy. Signal Transduct. Target. Ther. 2024, 9, 266. [Google Scholar] [CrossRef]
  20. Uribe, M.L.; Marrocco, I.; Yarden, Y. EGFR in Cancer: Signaling Mechanisms, Drugs, and Acquired Resistance. Cancers 2021, 13, 2748. [Google Scholar] [CrossRef]
  21. Krasinskas, A.M. EGFR Signaling in Colorectal Carcinoma. Pathol. Res. Int. 2011, 2011, 932932. [Google Scholar] [CrossRef]
  22. Schultz, D.F.; Billadeau, D.D.; Jois, S.D. EGFR trafficking: Effect of dimerization, dynamics, and mutation. Front. Oncol. 2023, 13, 1258371. [Google Scholar] [CrossRef]
  23. Chung, I.; Akita, R.; Vandlen, R.; Toomre, D.; Schlessinger, J.; Mellman, I. Spatial control of EGF receptor activation by reversible dimerization on living cells. Nature 2010, 464, 783–787. [Google Scholar] [CrossRef] [PubMed]
  24. Levantini, E.; Maroni, G.; Del Re, M.; Tenen, D.G. EGFR signaling pathway as therapeutic target in human cancers. Semin. Cancer Biol. 2022, 85, 253–275. [Google Scholar] [CrossRef]
  25. Wee, P.; Wang, Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers 2017, 9, 52. [Google Scholar] [CrossRef]
  26. Koveitypour, Z.; Panahi, F.; Vakilian, M.; Peymani, M.; Seyed Forootan, F.; Nasr Esfahani, M.H.; Ghaedi, K. Signaling pathways involved in colorectal cancer progression. Cell Biosci. 2019, 9, 97. [Google Scholar] [CrossRef]
  27. Cruz-Duarte, R.; Rebelo de Almeida, C.; Negrão, M.; Fernandes, A.; Borralho, P.; Sobral, D.; Gallego-Paez, L.M.; Machado, D.; Gramaça, J.; Vílchez, J.; et al. Predictive and Therapeutic Implications of a Novel PLCγ1/SHP2-Driven Mechanism of Cetuximab Resistance in Metastatic Colorectal Cancer. Clin. Cancer Res. 2022, 28, 1203–1216. [Google Scholar] [CrossRef]
  28. Quesnelle, K.M.; Boehm, A.L.; Grandis, J.R. STAT-mediated EGFR signaling in cancer. J. Cell. Biochem. 2007, 102, 311–319. [Google Scholar] [CrossRef]
  29. Spano, J.-P.; Lagorce, C.; Atlan, D.; Milano, G.; Domont, J.; Benamouzig, R.; Attar, A.; Benichou, J.; Martin, A.; Morere, J.-F.; et al. Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann. Oncol. 2005, 16, 102–108. [Google Scholar] [CrossRef] [PubMed]
  30. Napolitano, S.; Ciardiello, D.; Cioli, E.; Martinelli, E.; Troiani, T.; Giulia Zampino, M.; Fazio, N.; De Vita, F.; Ciardiello, F.; Martini, G. BRAFV600E mutant metastatic colorectal cancer: Current advances in personalized treatment and future perspectives. Cancer Treat. Rev. 2025, 134, 102905. [Google Scholar] [CrossRef] [PubMed]
  31. Yaeger, R.; Corcoran, R.B. Targeting Alterations in the RAF-MEK Pathway. Cancer Discov. 2019, 9, 329–341. [Google Scholar] [CrossRef]
  32. Samuels, Y.; Diaz, L.A.; Schmidt-Kittler, O.; Cummins, J.M.; Delong, L.; Cheong, I.; Rago, C.; Huso, D.L.; Lengauer, C.; Kinzler, K.W.; et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell 2005, 7, 561–573. [Google Scholar] [CrossRef]
  33. Jhawer, M.; Goel, S.; Wilson, A.J.; Montagna, C.; Ling, Y.-H.; Byun, D.-S.; Nasser, S.; Arango, D.; Shin, J.; Klampfer, L.; et al. PIK3CA mutation/PTEN expression status predicts response of colon cancer cells to the epidermal growth factor receptor inhibitor cetuximab. Cancer Res. 2008, 68, 1953–1961. [Google Scholar] [CrossRef]
  34. Lièvre, A.; Ouine, B.; Canet, J.; Cartier, A.; Amar, Y.; Cacheux, W.; Mariani, O.; Guimbaud, R.; Selves, J.; Lecomte, T.; et al. Protein biomarkers predictive for response to anti-EGFR treatment in RAS wild-type metastatic colorectal carcinoma. Br. J. Cancer 2017, 117, 1819–1827. [Google Scholar] [CrossRef] [PubMed]
  35. Ruzzo, A.; Graziano, F.; Canestrari, E.; Magnani, M. Molecular predictors of efficacy to anti-EGFR agents in colorectal cancer patients. Curr. Cancer Drug Targets 2010, 10, 68–79. [Google Scholar] [CrossRef] [PubMed]
  36. Halder, S.; Basu, S.; Lall, S.P.; Ganti, A.K.; Batra, S.K.; Seshacharyulu, P. Targeting the EGFR signaling pathway in cancer therapy: What’s new in 2023? Expert Opin. Ther. Targets 2023, 27, 305–324. [Google Scholar] [CrossRef] [PubMed]
  37. Kast, J.; Dutta, S.; Upreti, V.V. Panitumumab: A Review of Clinical Pharmacokinetic and Pharmacology Properties After over a Decade of Experience in Patients with Solid Tumors. Adv. Ther. 2021, 38, 3712–3723. [Google Scholar] [CrossRef]
  38. Vincenzi, B.; Schiavon, G.; Silletta, M.; Santini, D.; Tonini, G. The biological properties of cetuximab. Crit. Rev. Oncol. Hematol. 2008, 68, 93–106. [Google Scholar] [CrossRef]
  39. Kurai, J.; Chikumi, H.; Hashimoto, K.; Yamaguchi, K.; Yamasaki, A.; Sako, T.; Touge, H.; Makino, H.; Takata, M.; Miyata, M.; et al. Antibody-dependent cellular cytotoxicity mediated by cetuximab against lung cancer cell lines. Clin. Cancer Res. 2007, 13, 1552–1561. [Google Scholar] [CrossRef]
  40. Fracasso, P.M.; Burris, H.; Arquette, M.A.; Govindan, R.; Gao, F.; Wright, L.P.; Goodner, S.A.; Greco, F.A.; Jones, S.F.; Willcut, N.; et al. A phase 1 escalating single-dose and weekly fixed-dose study of cetuximab: Pharmacokinetic and pharmacodynamic rationale for dosing. Clin. Cancer Res. 2007, 13, 986–993. [Google Scholar] [CrossRef][Green Version]
  41. Weiner, L.M.; Belldegrun, A.S.; Crawford, J.; Tolcher, A.W.; Lockbaum, P.; Arends, R.H.; Navale, L.; Amado, R.G.; Schwab, G.; Figlin, R.A. Dose and schedule study of panitumumab monotherapy in patients with advanced solid malignancies. Clin. Cancer Res. 2008, 14, 502–508. [Google Scholar] [CrossRef]
  42. Saltz, L.B.; Lenz, H.-J.; Kindler, H.L.; Hochster, H.S.; Wadler, S.; Hoff, P.M.; Kemeny, N.E.; Hollywood, E.M.; Gonen, M.; Quinones, M.; et al. Randomized phase II trial of cetuximab, bevacizumab, and irinotecan compared with cetuximab and bevacizumab alone in irinotecan-refractory colorectal cancer: The BOND-2 study. J. Clin. Oncol. 2007, 25, 4557–4561. [Google Scholar] [CrossRef]
  43. Van Cutsem, E.; Lang, I.; Folprecht, G.; Nowacki, M.; Barone, C.; Shchepotin, I.; Maurel, J.; Cunningham, D.; Celik, I.; Kohne, C. Cetuximab plus FOLFIRI: Final data from the CRYSTAL study on the association of KRAS and BRAF biomarker status with treatment outcome. J. Clin. Oncol. 2010, 28, 3570. [Google Scholar] [CrossRef]
  44. Douillard, J.Y.; Siena, S.; Cassidy, J.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Final results from PRIME: Randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann. Oncol. 2014, 25, 1346–1355. [Google Scholar] [CrossRef] [PubMed]
  45. Price, T.J.; Peeters, M.; Kim, T.W.; Li, J.; Cascinu, S.; Ruff, P.; Suresh, A.S.; Thomas, A.; Tjulandin, S.; Zhang, K.; et al. Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): A randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 2014, 15, 569–579. [Google Scholar] [CrossRef] [PubMed]
  46. Di Fiore, F.; Blanchard, F.; Charbonnier, F.; Le Pessot, F.; Lamy, A.; Galais, M.P.; Bastit, L.; Killian, A.; Sesboüé, R.; Tuech, J.J.; et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br. J. Cancer 2007, 96, 1166–1169. [Google Scholar] [CrossRef] [PubMed]
  47. Rowland, A.; Dias, M.M.; Wiese, M.D.; Kichenadasse, G.; McKinnon, R.A.; Karapetis, C.S.; Sorich, M.J. Meta-analysis comparing the efficacy of anti-EGFR monoclonal antibody therapy between KRAS G13D and other KRAS mutant metastatic colorectal cancer tumours. Eur. J. Cancer 2016, 55, 122–130. [Google Scholar] [CrossRef]
  48. Mao, C.; Liao, R.-Y.; Qiu, L.-X.; Wang, X.-W.; Ding, H.; Chen, Q. BRAF V600E mutation and resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer: A meta-analysis. Mol. Biol. Rep. 2011, 38, 2219–2223. [Google Scholar] [CrossRef]
  49. Watanabe, J.; Muro, K.; Shitara, K.; Yamazaki, K.; Shiozawa, M.; Ohori, H.; Takashima, A.; Yokota, M.; Makiyama, A.; Akazawa, N.; et al. Panitumumab vs Bevacizumab Added to Standard First-Line Chemotherapy and Overall Survival Among Patients with RAS Wild-type, Left-Sided Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 2023, 329, 1271–1282. [Google Scholar] [CrossRef]
  50. Holch, J.W.; Ricard, I.; Stintzing, S.; Modest, D.P.; Heinemann, V. The relevance of primary tumour location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur. J. Cancer 2017, 70, 87–98. [Google Scholar] [CrossRef]
  51. Baran, B.; Mert Ozupek, N.; Yerli Tetik, N.; Acar, E.; Bekcioglu, O.; Baskin, Y. Difference Between Left-Sided and Right-Sided Colorectal Cancer: A Focused Review of Literature. Gastroenterol. Res. 2018, 11, 264–273. [Google Scholar] [CrossRef]
  52. Kopetz, S.; Yoshino, T.; Van Cutsem, E.; Eng, C.; Kim, T.W.; Wasan, H.S.; Desai, J.; Ciardiello, F.; Yaeger, R.; Maughan, T.S.; et al. Encorafenib, cetuximab and chemotherapy in BRAF-mutant colorectal cancer: A randomized phase 3 trial. Nat. Med. 2025, 31, 901–908. [Google Scholar] [CrossRef]
  53. Jacob, J.; Anami, Y.; High, P.C.; Liang, Z.; Subramanian, S.; Ghosh, S.C.; AghaAmiri, S.; Guernsey-Biddle, C.; Tran, H.; Rowe, J.; et al. Antibody-Drug Conjugates Targeting the EGFR Ligand Epiregulin Elicit Robust Antitumor Activity in Colorectal Cancer. Cancer Res. 2025, 85, 973–986. [Google Scholar] [CrossRef]
  54. Pisheh, L.; Matis, S.; Taglieri, M.; Di Gregorio, L.; Benelli, R.; Poggi, A. EGFR-Targeted Antibody-Drug Conjugate to Different Aminobisphosphonates: Direct and Indirect Antitumor Effects on Colorectal Carcinoma Cells. Cancers 2024, 16, 1256. [Google Scholar] [CrossRef]
  55. Kopetz, S.; Boni, V.; Kato, K.; Raghav, K.P.S.; Pallis, A.; Habermehl, C.; Galipelli, S.; Courlet, P.; Rodriguez Rivera, I. First-in-human trial of M9140, an anti-CEACAM5 antibody drug conjugate (ADC) with exatecan payload, in patients (pts) with metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2024, 42, 3000. [Google Scholar] [CrossRef]
  56. Shen, L.; Parseghian, C.M.; Zhang, Y.; Chen, Z.; Qin, Y.; Zhang, W.; Deng, Y.; Pan, H.; Gu, S.; Li, C.; et al. Abstract CT168: EMB-01, an EGFR/cMET bispecific antibody, in metastatic colorectal cancer: Results from an international phase Ib/II study. Cancer Res. 2025, 85, CT168. [Google Scholar] [CrossRef]
  57. Argiles, G.; Jungels, C.; Garcia-Carbonero, R.; Diez Garcia, M.; Bendell, J.C.; Tabernero, J.; Bekradda, M.; Lammerts van Bueren, J.; Bol, K.; Stalbovskaya, V.; et al. Phase I dose-escalation study of MCLA-158, a first-in-class bispecific antibody targeting EGFR and LGR5, in metastatic colorectal cancer (CRC). J. Clin. Oncol. 2021, 39, 62. [Google Scholar] [CrossRef]
  58. Lenz, H.-J.; Overman, M.J.; Van Cutsem, E.; Limon, M.L.; Wong, K.Y.M.; Hendlisz, A.; Aglietta, M.; Garcia-Alfonso, P.; Neyns, B.; Gelsomino, F.; et al. First-line (1L) nivolumab (NIVO) + ipilimumab (IPI) in patients (pts) with microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC): 64-month (mo) follow-up from CheckMate 142. J. Clin. Oncol. 2024, 42, 97. [Google Scholar] [CrossRef]
  59. Abdelgadir, O.; Kuo, Y.-F.; Okorodudu, A.O.; Khan, M.F.; Cheng, Y.-W.; Dong, J. KRAS, NRAS, and BRAF Hot-Spot Mutations in Relation to Sidedness of Primary Colorectal Cancer: A Retrospective Cohort Study. Diagnostics 2025, 15, 142. [Google Scholar] [CrossRef]
  60. Ranđelović, I.; Nyíri, K.; Koppány, G.; Baranyi, M.; Tóvári, J.; Kigyós, A.; Tímár, J.; Vértessy, B.G.; Grolmusz, V. Gluing GAP to RAS Mutants: A New Approach to an Old Problem in Cancer Drug Development. Int. J. Mol. Sci. 2024, 25, 2572. [Google Scholar] [CrossRef] [PubMed]
  61. Ríos-Hoyo, A.; Monzonís, X.; Vidal, J.; Linares, J.; Montagut, C. Unveiling acquired resistance to anti-EGFR therapies in colorectal cancer: A long and winding road. Front. Pharmacol. 2024, 15, 1398419. [Google Scholar] [CrossRef]
  62. Kim, T.W.; Price, T.; Grasselli, J.; Strickler, J.H.; Masuishi, T.; Kwok, G.W.; Yalcin, S.; Obiozor, C.C.; Chan, E.; Gokani, P.; et al. A phase 3 study of first-line sotorasib, panitumumab, and FOLFIRI versus FOLFIRI with or without bevacizumab-awwb for patients with KRAS G12C–mutated metastatic colorectal cancer (CodeBreaK 301). J. Clin. Oncol. 2025, 43, TPS326. [Google Scholar] [CrossRef]
  63. Pietrantonio, F.; Petrelli, F.; Coinu, A.; Di Bartolomeo, M.; Borgonovo, K.; Maggi, C.; Cabiddu, M.; Iacovelli, R.; Bossi, I.; Lonati, V.; et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A meta-analysis. Eur. J. Cancer 2015, 51, 587–594. [Google Scholar] [CrossRef]
  64. Stintzing, S.; Miller-Phillips, L.; Modest, D.P.; Fischer von Weikersthal, L.; Decker, T.; Kiani, A.; Vehling-Kaiser, U.; Al-Batran, S.-E.; Heintges, T.; Kahl, C.; et al. Impact of BRAF and RAS mutations on first-line efficacy of FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab: Analysis of the FIRE-3 (AIO KRK-0306) study. Eur. J. Cancer 2017, 79, 50–60. [Google Scholar] [CrossRef]
  65. Stintzing, S.; Heinrich, K.; Tougeron, D.; Modest, D.P.; Schwaner, I.; Eucker, J.; Pihusch, R.; Stauch, M.; Kaiser, F.; Kahl, C.; et al. FOLFOXIRI Plus Cetuximab or Bevacizumab as First-Line Treatment of BRAFV600E-Mutant Metastatic Colorectal Cancer: The Randomized Phase II FIRE-4.5 (AIO KRK0116) Study. J. Clin. Oncol. 2023, 41, 4143–4153. [Google Scholar] [CrossRef] [PubMed]
  66. Pizzamiglio, M.; Soulabaille, A.; Lahlou, W.; Pilla, L.; Zaanan, A.; Taieb, J. Advances and challenges in targeted therapies for HER2-amplified colorectal cancer. Eur. J. Cancer 2025, 222, 115471. [Google Scholar] [CrossRef] [PubMed]
  67. García-Alfonso, P.; Valladares-Ayerbes, M.; Muñoz Martín, A.J.; Morales Herrero, R.; Galvez Muñoz, E.; Prat-Llorens, G. State of the art of the molecular hyperselection to guide treatment with anti-EGFR antibodies in RAS WT mCRC: Implications for clinical practice and future perspectives. Expert Opin. Biol. Ther. 2025, 25, 413–423. [Google Scholar] [CrossRef] [PubMed]
  68. Yonesaka, K.; Zejnullahu, K.; Okamoto, I.; Satoh, T.; Cappuzzo, F.; Souglakos, J.; Ercan, D.; Rogers, A.; Roncalli, M.; Takeda, M.; et al. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci. Transl. Med. 2011, 3, 99ra86. [Google Scholar] [CrossRef]
  69. Ciardiello, F.; Normanno, N. HER2 Signaling and Resistance to the Anti-EGFR Monoclonal Antibody Cetuximab: A Further Step toward Personalized Medicine for Patients with Colorectal Cancer. Cancer Discov. 2011, 1, 472–474. [Google Scholar] [CrossRef]
  70. Raghav, K.; Loree, J.M.; Morris, J.S.; Overman, M.J.; Yu, R.; Meric-Bernstam, F.; Menter, D.; Korphaisarn, K.; Kee, B.; Muranyi, A.; et al. Validation of HER2 Amplification as a Predictive Biomarker for Anti-Epidermal Growth Factor Receptor Antibody Therapy in Metastatic Colorectal Cancer. JCO Precis. Oncol. 2019, 3, 1–13. [Google Scholar] [CrossRef]
  71. Bekaii-Saab, T.S.; Lach, K.; Hsu, L.-I.; Siadak, M.; Stecher, M.; Ward, J.; Beckerman, R.; Strickler, J.H. Impact of Anti-EGFR Therapies on HER2-Positive Metastatic Colorectal Cancer: A Systematic Literature Review and Meta-Analysis of Clinical Outcomes. Oncologist 2023, 28, 885–893. [Google Scholar] [CrossRef]
  72. Yoshino, T.; Di Bartolomeo, M.; Raghav, K.; Masuishi, T.; Loupakis, F.; Kawakami, H.; Yamaguchi, K.; Nishina, T.; Wainberg, Z.; Elez, E.; et al. Final results of DESTINY-CRC01 investigating trastuzumab deruxtecan in patients with HER2-expressing metastatic colorectal cancer. Nat. Commun. 2023, 14, 3332. [Google Scholar] [CrossRef]
  73. Sartore-Bianchi, A.; Lonardi, S.; Martino, C.; Fenocchio, E.; Tosi, F.; Ghezzi, S.; Leone, F.; Bergamo, F.; Zagonel, V.; Ciardiello, F.; et al. Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: The phase II HERACLES-B trial. ESMO Open 2020, 5, e000911. [Google Scholar] [CrossRef]
  74. Strickler, J.H.; Cercek, A.; Siena, S.; André, T.; Ng, K.; Van Cutsem, E.; Wu, C.; Paulson, A.S.; Hubbard, J.M.; Coveler, A.L.; et al. Tucatinib plus trastuzumab for chemotherapy-refractory, HER2-positive, RAS wild-type unresectable or metastatic colorectal cancer (MOUNTAINEER): A multicentre, open-label, phase 2 study. Lancet Oncol. 2023, 24, 496–508. [Google Scholar] [CrossRef]
  75. Meric-Bernstam, F.; Hurwitz, H.; Raghav, K.P.S.; McWilliams, R.R.; Fakih, M.; VanderWalde, A.; Swanton, C.; Kurzrock, R.; Burris, H.; Sweeney, C.; et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): An updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019, 20, 518–530. [Google Scholar] [CrossRef] [PubMed]
  76. Nakamura, Y.; Okamoto, W.; Sawada, K.; Komatsu, Y.; Kato, K.; Taniguchi, H.; Kato, T.; Nishina, T.; Esaki, T.; Nomura, H.; et al. TRIUMPH Study: A multicenter Phase II study to evaluate efficacy and safety of combination therapy with trastuzumab and pertuzumab in patients with HER2-positive metastatic colorectal cancer (EPOC1602). Ann. Oncol. 2017, 28, v207. [Google Scholar] [CrossRef]
  77. Nakamura, Y.; Okamoto, W.; Kato, T.; Esaki, T.; Kato, K.; Komatsu, Y.; Yuki, S.; Masuishi, T.; Nishina, T.; Ebi, H.; et al. Circulating tumor DNA-guided treatment with pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer: A phase 2 trial. Nat. Med. 2021, 27, 1899–1903. [Google Scholar] [CrossRef] [PubMed]
  78. Gupta, A.; Michelini, F.; Shao, H.; Yeh, C.; Drago, J.Z.; Liu, D.; Rosiek, E.; Romin, Y.; Ghafourian, N.; Thyparambil, S.; et al. EGFR-directed antibodies promote HER2 ADC internalization and efficacy. Cell Rep. Med. 2024, 5, 101792. [Google Scholar] [CrossRef]
  79. Wang, H.; Tang, R.; Jiang, L.; Jia, Y. The role of PIK3CA gene mutations in colorectal cancer and the selection of treatment strategies. Front. Pharmacol. 2024, 15, 1494802. [Google Scholar] [CrossRef]
  80. Day, F.L.; Jorissen, R.N.; Lipton, L.; Mouradov, D.; Sakthianandeswaren, A.; Christie, M.; Li, S.; Tsui, C.; Tie, J.; Desai, J.; et al. PIK3CA and PTEN Gene and Exon Mutation-Specific Clinicopathologic and Molecular Associations in Colorectal Cancer. Clin. Cancer Res. 2013, 19, 3285–3296. [Google Scholar] [CrossRef]
  81. Sartore-Bianchi, A.; Martini, M.; Molinari, F.; Veronese, S.; Nichelatti, M.; Artale, S.; Di Nicolantonio, F.; Saletti, P.; De Dosso, S.; Mazzucchelli, L.; et al. PIK3CA Mutations in Colorectal Cancer Are Associated with Clinical Resistance to EGFR-Targeted Monoclonal Antibodies. Cancer Res. 2009, 69, 1851–1857. [Google Scholar] [CrossRef]
  82. Laurent-Puig, P.; Cayre, A.; Manceau, G.; Buc, E.; Bachet, J.-B.; Lecomte, T.; Rougier, P.; Lievre, A.; Landi, B.; Boige, V.; et al. Analysis of PTEN, BRAF, and EGFR status in determining benefit from cetuximab therapy in wild-type KRAS metastatic colon cancer. J. Clin. Oncol. 2009, 27, 5924–5930. [Google Scholar] [CrossRef]
  83. Perrone, F.; Lampis, A.; Orsenigo, M.; Di Bartolomeo, M.; Gevorgyan, A.; Losa, M.; Frattini, M.; Riva, C.; Andreola, S.; Bajetta, E.; et al. PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Ann. Oncol. 2009, 20, 84–90. [Google Scholar] [CrossRef]
  84. Lu, X.; Li, Y.; Li, Y.; Zhang, X.; Shi, J.; Feng, H.; Yu, Z.; Gao, Y. Prognostic and predictive biomarkers for anti-EGFR monoclonal antibody therapy in RAS wild-type metastatic colorectal cancer: A systematic review and meta-analysis. BMC Cancer 2023, 23, 1117. [Google Scholar] [CrossRef] [PubMed]
  85. Bardelli, A.; Corso, S.; Bertotti, A.; Hobor, S.; Valtorta, E.; Siravegna, G.; Sartore-Bianchi, A.; Scala, E.; Cassingena, A.; Zecchin, D.; et al. Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013, 3, 658–673. [Google Scholar] [CrossRef] [PubMed]
  86. Religa, P.; Łazarczyk, M.; Mickael, M.-E.; Skiba, D.S. Misleading Interpretations of the MET Gene Acronym: Over 15 Years of Confusion in Scientific Publications and a Call for Reaffirming the Original Gene Nomenclature. J. Thorac. Oncol. 2025, 20, 1032–1034. [Google Scholar] [CrossRef] [PubMed]
  87. Cooper, C.S.; Park, M.; Blair, D.G.; Tainsky, M.A.; Huebner, K.; Croce, C.M.; Vande Woude, G.F. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 1984, 311, 29–33. [Google Scholar] [CrossRef]
  88. Randon, G.; Maddalena, G.; Germani, M.M.; Pircher, C.C.; Manca, P.; Bergamo, F.; Giordano, M.; Sposetti, C.; Montagna, A.; Vetere, G.; et al. Negative Ultraselection of Patients with RAS/BRAF Wild-Type, Microsatellite-Stable Metastatic Colorectal Cancer Receiving Anti-EGFR-Based Therapy. JCO Precis. Oncol. 2022, 6, e2200037. [Google Scholar] [CrossRef]
  89. Zhang, M.; Li, G.; Sun, X.; Ni, S.; Tan, C.; Xu, M.; Huang, D.; Ren, F.; Li, D.; Wei, P.; et al. MET amplification, expression, and exon 14 mutations in colorectal adenocarcinoma. Hum. Pathol. 2018, 77, 108–115. [Google Scholar] [CrossRef]
  90. Raghav, K.; Morris, V.; Tang, C.; Morelli, P.; Amin, H.M.; Chen, K.; Manyam, G.C.; Broom, B.; Overman, M.J.; Shaw, K.; et al. MET amplification in metastatic colorectal cancer: An acquired response to EGFR inhibition, not a de novo phenomenon. Oncotarget 2016, 7, 54627–54631. [Google Scholar] [CrossRef]
  91. Eng, C.; Bessudo, A.; Hart, L.L.; Severtsev, A.; Gladkov, O.; Müller, L.; Kopp, M.V.; Vladimirov, V.; Langdon, R.; Kotiv, B.; et al. A randomized, placebo-controlled, phase 1/2 study of tivantinib (ARQ 197) in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with wild-type KRAS who have received first-line systemic therapy. Int. J. Cancer 2016, 139, 177–186. [Google Scholar] [CrossRef]
  92. Roberts, P.J.; Der, C.J. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 2007, 26, 3291–3310. [Google Scholar] [CrossRef]
  93. Wu, P.-K.; Park, J.-I. MEK1/2 Inhibitors: Molecular Activity and Resistance Mechanisms. Semin. Oncol. 2015, 42, 849–862. [Google Scholar] [CrossRef]
  94. Mauri, G.; Patelli, G.; Gori, V.; Lauricella, C.; Mussolin, B.; Amatu, A.; Bencardino, K.; Tosi, F.; Bonazzina, E.; Bonoldi, E.; et al. Case Report: MAP2K1 K57N mutation is associated with primary resistance to anti-EGFR monoclonal antibodies in metastatic colorectal cancer. Front. Oncol. 2022, 12, 1030232. [Google Scholar] [CrossRef]
  95. Tian, J.; Chen, J.H.; Chao, S.X.; Pelka, K.; Giannakis, M.; Hess, J.; Burke, K.; Jorgji, V.; Sindurakar, P.; Braverman, J.; et al. Combined PD-1, BRAF and MEK inhibition in BRAFV600E colorectal cancer: A phase 2 trial. Nat. Med. 2023, 29, 458–466. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, M.S.; Jeong, E.G.; Yoo, N.J.; Lee, S.H. Mutational analysis of oncogenic AKT E17K mutation in common solid cancers and acute leukaemias. Br. J. Cancer 2008, 98, 1533–1535. [Google Scholar] [CrossRef] [PubMed]
  97. Leiphrakpam, P.D.; Rajappa, S.J.; Krishnan, M.; Batra, R.; Murthy, S.S.; Are, C. Colorectal cancer: Review of signaling pathways and associated therapeutic strategies. J. Surg. Oncol. 2023, 127, 1277–1295. [Google Scholar] [CrossRef]
  98. Hechtman, J.F.; Sadowska, J.; Huse, J.T.; Borsu, L.; Yaeger, R.; Shia, J.; Vakiani, E.; Ladanyi, M.; Arcila, M.E. AKT1 E17K in Colorectal Carcinoma Is Associated with BRAF V600E but Not MSI-H Status: A Clinicopathologic Comparison to PIK3CA Helical and Kinase Domain Mutants. Mol. Cancer Res. 2015, 13, 1003–1008. [Google Scholar] [CrossRef][Green Version]
  99. Mayer, E.L.; Ren, Y.; Wagle, N.; Mahtani, R.; Ma, C.; DeMichele, A.; Cristofanilli, M.; Meisel, J.; Miller, K.D.; Abdou, Y.; et al. PACE: A Randomized Phase II Study of Fulvestrant, Palbociclib, and Avelumab After Progression on Cyclin-Dependent Kinase 4/6 Inhibitor and Aromatase Inhibitor for Hormone Receptor–Positive/Human Epidermal Growth Factor Receptor–Negative Metastatic Breast Cancer. J. Clin. Oncol. 2024, 42, 2050–2060. [Google Scholar] [CrossRef] [PubMed]
  100. Kim, J.S.; Kim, J.E.; Kim, K.; Lee, J.; Park, J.O.; Lim, H.Y.; Park, Y.S.; Kang, W.K.; Kim, S.T. The Impact of Cetuximab Plus AKT- or mTOR- Inhibitor in a Patient-Derived Colon Cancer Cell Model with Wild-Type RAS and PIK3CA Mutation. J. Cancer 2017, 8, 2713–2719. [Google Scholar] [CrossRef]
  101. Sun, L.; Huang, Y.; Liu, Y.; Zhao, Y.; He, X.; Zhang, L.; Wang, F.; Zhang, Y. Ipatasertib, a novel Akt inhibitor, induces transcription factor FoxO3a and NF-κB directly regulates PUMA-dependent apoptosis. Cell Death Dis. 2018, 9, 911. [Google Scholar] [CrossRef]
  102. Malkomes, P.; Lunger, I.; Luetticke, A.; Oppermann, E.; Haetscher, N.; Serve, H.; Holzer, K.; Bechstein, W.O.; Rieger, M.A. Selective AKT Inhibition by MK-2206 Represses Colorectal Cancer-Initiating Stem Cells. Ann. Surg. Oncol. 2016, 23, 2849–2857. [Google Scholar] [CrossRef]
  103. Olsen, A.S.F.; Gundestrup, A.K.; Kleif, J.; Thanon, T.; Bertelsen, C.A. Accuracy of preoperative staging with multidetector computed tomography in colon cancer. Color. Dis. 2021, 23, 680–688. [Google Scholar] [CrossRef]
  104. Daly, C.; Castanaro, C.; Zhang, W.; Zhang, Q.; Wei, Y.; Ni, M.; Young, T.M.; Zhang, L.; Burova, E.; Thurston, G. FGFR3-TACC3 fusion proteins act as naturally occurring drivers of tumor resistance by functionally substituting for EGFR/ERK signaling. Oncogene 2017, 36, 471–481. [Google Scholar] [CrossRef] [PubMed]
  105. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  106. Subbiah, V.; Cassier, P.A.; Siena, S.; Garralda, E.; Paz-Ares, L.; Garrido, P.; Nadal, E.; Vuky, J.; Lopes, G.; Kalemkerian, G.P.; et al. Pan-cancer efficacy of pralsetinib in patients with RET fusion-positive solid tumors from the phase 1/2 ARROW trial. Nat. Med. 2022, 28, 1640–1645. [Google Scholar] [CrossRef] [PubMed]
  107. Ratti, M.; Grizzi, G.; Passalacqua, R.; Lampis, A.; Cereatti, F.; Grassia, R.; Hahne, J.C. NTRK fusions in colorectal cancer: Clinical meaning and future perspective. Expert Opin. Ther. Targets 2021, 25, 677–683. [Google Scholar] [CrossRef] [PubMed]
  108. Clifton, K.; Rich, T.A.; Parseghian, C.; Raymond, V.M.; Dasari, A.; Pereira, A.A.L.; Willis, J.; Loree, J.M.; Bauer, T.M.; Chae, Y.K.; et al. Identification of Actionable Fusions as an Anti-EGFR Resistance Mechanism Using a Circulating Tumor DNA Assay. JCO Precis. Oncol. 2019, 3, PO.19.00141. [Google Scholar] [CrossRef]
  109. Subbiah, V. Tissue-agnostic cancer therapies: Promise, reality, and the path forward. Nat. Commun. 2025, 16, 4972. [Google Scholar] [CrossRef]
  110. Loree, J.M.; Wang, Y.; Syed, M.A.; Sorokin, A.V.; Coker, O.; Xiu, J.; Weinberg, B.A.; Vanderwalde, A.M.; Tesfaye, A.; Raymond, V.M.; et al. Clinical and functional characterization of atypical KRAS/NRAS mutations in metastatic colorectal cancer. Clin. Cancer Res. 2021, 27, 4587–4598. [Google Scholar] [CrossRef]
  111. Tabernero, J.; Ros, J.; Élez, E. The Evolving Treatment Landscape in BRAF-V600E–Mutated Metastatic Colorectal Cancer. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 254–263. [Google Scholar] [CrossRef] [PubMed]
  112. Elez, E.; Yoshino, T.; Shen, L.; Lonardi, S.; Cutsem, E.V.; Eng, C.; Kim, T.W.; Wasan, H.S.; Desai, J.; Ciardiello, F.; et al. Encorafenib, Cetuximab, and mFOLFOX6 in BRAF-Mutated Colorectal Cancer. N. Engl. J. Med. 2025, 392, 2425–2437. [Google Scholar] [CrossRef]
  113. Tan, L.; Tran, B.; Tie, J.; Markman, B.; Ananda, S.; Tebbutt, N.C.; Michael, M.; Link, E.; Wong, S.Q.; Chandrashekar, S.; et al. A Phase Ib/II Trial of Combined BRAF and EGFR Inhibition in BRAF V600E Positive Metastatic Colorectal Cancer and Other Cancers: The EVICT (Erlotinib and Vemurafenib In Combination Trial) Study. Clin. Cancer Res. 2023, 29, 1017–1030. [Google Scholar] [CrossRef] [PubMed]
  114. Goodwin, R.; Jonker, D.; Chen, E.; Kennecke, H.; Cabanero, M.; Tsao, M.-S.; Vickers, M.; Bohemier, C.; Lim, H.; Ritter, H.; et al. A phase Ib study of a PI3Kinase inhibitor BKM120 in combination with panitumumab in patients with KRAS wild-type advanced colorectal cancer. Investig. New Drugs 2020, 38, 1077–1084. [Google Scholar] [CrossRef]
  115. Zheng, Y.; Lou, S.; Lu, J.; Zheng, F.; Tang, Y.; Zhang, E.; Cui, S.; Zhao, H. Selective PI3Kδ inhibitor TYM-3-98 suppresses AKT/mTOR/SREBP1-mediated lipogenesis and promotes ferroptosis in KRAS-mutant colorectal cancer. Cell Death Dis. 2024, 15, 474. [Google Scholar] [CrossRef] [PubMed]
  116. Foley, T.M.; Payne, S.N.; Pasch, C.A.; Yueh, A.E.; Van De Hey, D.R.; Korkos, D.P.; Clipson, L.; Maher, M.E.; Matkowskyj, K.A.; Newton, M.A.; et al. Dual PI3K/mTOR Inhibition in Colorectal Cancers with APC and PIK3CA Mutations. Mol. Cancer Res. 2017, 15, 317–327. [Google Scholar] [CrossRef]
  117. Liu, Y.; Yu, X.-F.; Zou, J.; Luo, Z.-H. Prognostic value of c-Met in colorectal cancer: A meta-analysis. World J. Gastroenterol. 2015, 21, 3706–3710. [Google Scholar] [CrossRef]
  118. Rimassa, L.; Bozzarelli, S.; Pietrantonio, F.; Cordio, S.; Lonardi, S.; Toppo, L.; Zaniboni, A.; Bordonaro, R.; Di Bartolomeo, M.; Tomasello, G.; et al. Phase II Study of Tivantinib and Cetuximab in Patients with KRAS Wild-type Metastatic Colorectal Cancer with Acquired Resistance to EGFR Inhibitors and Emergence of MET Overexpression: Lesson Learned for Future Trials with EGFR/MET Dual Inhibition. Clin. Color. Cancer 2019, 18, 125–132.E2. [Google Scholar] [CrossRef]
  119. Chuang, J.; Wang, C.; Guo, Y.; Valenzuela, V.; Wu, J.; Fakih, M. MAP2K1 Mutations in Advanced Colorectal Cancer Predict Poor Response to Anti-EGFR Therapy and to Vertical Targeting of MAPK Pathway. Clin. Color. Cancer 2021, 20, 72–78. [Google Scholar] [CrossRef]
  120. Dankner, M.; Rousselle, E.; Petrecca, S.; Fabi, F.; Nowakowski, A.; Lazaratos, A.-M.; Rajadurai, C.V.; Stein, A.J.B.; Bian, D.; Tai, P.; et al. Clinical Activity of Mitogen-Activated Protein Kinase Inhibitors in Patients with MAP2K1 (MEK1)-Mutated Metastatic Cancers. JCO Precis. Oncol. 2025, 9, e2400199. [Google Scholar] [CrossRef]
  121. Kim, S.T.; Lee, S.J.; Park, J.O.; Lim, H.Y.; Kang, W.K.; Park, Y.S.; Lee, J. The impact of cetuximab plus AKT- or mTOR- inhibitor in patient-derived colon cancer cell model with RAS wild type and PIK3CA mutation. J. Clin. Oncol. 2016, 34, e15153. [Google Scholar] [CrossRef]
  122. Zhai, Z.; Yu, X.; Yang, B.; Zhang, Y.; Zhang, L.; Li, X.; Sun, H. Colorectal cancer heterogeneity and targeted therapy: Clinical implications, challenges and solutions for treatment resistance. Semin. Cell Dev. Biol. 2017, 64, 107–115. [Google Scholar] [CrossRef]
  123. Siravegna, G.; Mussolin, B.; Buscarino, M.; Corti, G.; Cassingena, A.; Crisafulli, G.; Ponzetti, A.; Cremolini, C.; Amatu, A.; Lauricella, C.; et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 2015, 21, 795–801. [Google Scholar] [CrossRef] [PubMed]
  124. Sartore-Bianchi, A.; Siena, S.; Tonini, G.; Bardelli, A.; Santini, D. Overcoming dynamic molecular heterogeneity in metastatic colorectal cancer: Multikinase inhibition with regorafenib and the case of rechallenge with anti-EGFR. Cancer Treat. Rev. 2016, 51, 54–62. [Google Scholar] [CrossRef] [PubMed]
  125. Niida, A.; Mimori, K.; Shibata, T.; Miyano, S. Modeling colorectal cancer evolution. J. Hum. Genet. 2021, 66, 869–878. [Google Scholar] [CrossRef]
  126. Ciardiello, F.; Bianco, R.; Caputo, R.; Caputo, R.; Damiano, V.; Troiani, T.; Melisi, D.; De Vita, F.; De Placido, S.; Bianco, A.R.; et al. Antitumor activity of ZD6474, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in human cancer cells with acquired resistance to antiepidermal growth factor receptor therapy. Clin. Cancer Res. 2004, 10, 784–793. [Google Scholar] [CrossRef] [PubMed]
  127. Montagut, C.; Dalmases, A.; Bellosillo, B.; Crespo, M.; Pairet, S.; Iglesias, M.; Salido, M.; Gallen, M.; Marsters, S.; Tsai, S.P.; et al. Identification of a mutation in the extracellular domain of the Epidermal Growth Factor Receptor conferring cetuximab resistance in colorectal cancer. Nat. Med. 2012, 18, 221–223. [Google Scholar] [CrossRef]
  128. Arena, S.; Bellosillo, B.; Siravegna, G.; Martínez, A.; Cañadas, I.; Lazzari, L.; Ferruz, N.; Russo, M.; Misale, S.; González, I.; et al. Emergence of Multiple EGFR Extracellular Mutations during Cetuximab Treatment in Colorectal Cancer. Clin. Cancer Res. 2015, 21, 2157–2166. [Google Scholar] [CrossRef] [PubMed]
  129. Ali, M.; Kaltenbrun, E.; Anderson, G.R.; Stephens, S.J.; Arena, S.; Bardelli, A.; Counter, C.M.; Wood, K.C. Codon bias imposes a targetable limitation on KRAS-driven therapeutic resistance. Nat. Commun. 2017, 8, 15617. [Google Scholar] [CrossRef]
  130. Diaz, L.A.; Williams, R.T.; Wu, J.; Kinde, I.; Hecht, J.R.; Berlin, J.; Allen, B.; Bozic, I.; Reiter, J.G.; Nowak, M.A.; et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012, 486, 537–540. [Google Scholar] [CrossRef]
  131. Misale, S.; Yaeger, R.; Hobor, S.; Scala, E.; Janakiraman, M.; Liska, D.; Valtorta, E.; Schiavo, R.; Buscarino, M.; Siravegna, G.; et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012, 486, 532–536. [Google Scholar] [CrossRef]
  132. Russo, M.; Siravegna, G.; Blaszkowsky, L.S.; Corti, G.; Crisafulli, G.; Ahronian, L.G.; Mussolin, B.; Kwak, E.L.; Buscarino, M.; Lazzari, L.; et al. Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal Cancer. Cancer Discov. 2016, 6, 147–153. [Google Scholar] [CrossRef]
  133. García-Albéniz, X.; Alonso, V.; Escudero, P.; Méndez, M.; Gallego, J.; Rodríguez, J.R.; Salud, A.; Fernández-Plana, J.; Manzano, H.; Zanui, M.; et al. Prospective Biomarker Study in Advanced RAS Wild-Type Colorectal Cancer: POSIBA Trial (GEMCAD 10-02). Oncologist 2019, 24, e1115–e1122. [Google Scholar] [CrossRef]
  134. Montagut, C.; Argilés, G.; Ciardiello, F.; Poulsen, T.T.; Dienstmann, R.; Kragh, M.; Kopetz, S.; Lindsted, T.; Ding, C.; Vidal, J.; et al. Efficacy of Sym004 in Patients with Metastatic Colorectal Cancer with Acquired Resistance to Anti-EGFR Therapy and Molecularly Selected by Circulating Tumor DNA Analyses: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2018, 4, e175245. [Google Scholar] [CrossRef] [PubMed]
  135. Subbiah, V.; Baik, C.; Kirkwood, J.M. Clinical Development of BRAF plus MEK Inhibitor Combinations. Trends Cancer 2020, 6, 797–810. [Google Scholar] [CrossRef] [PubMed]
  136. Bray, S.M.; Lee, J.; Kim, S.T.; Hur, J.Y.; Ebert, P.J.; Calley, J.N.; Wulur, I.H.; Gopalappa, T.; Wong, S.S.; Qian, H.-R.; et al. Genomic characterization of intrinsic and acquired resistance to cetuximab in colorectal cancer patients. Sci. Rep. 2019, 9, 15365. [Google Scholar] [CrossRef]
  137. Mohan, S.; Heitzer, E.; Ulz, P.; Lafer, I.; Lax, S.; Auer, M.; Pichler, M.; Gerger, A.; Eisner, F.; Hoefler, G.; et al. Changes in colorectal carcinoma genomes under anti-EGFR therapy identified by whole-genome plasma DNA sequencing. PLoS Genet. 2014, 10, e1004271. [Google Scholar] [CrossRef] [PubMed]
  138. Woolston, A.; Khan, K.; Spain, G.; Barber, L.J.; Griffiths, B.; Gonzalez-Exposito, R.; Hornsteiner, L.; Punta, M.; Patil, Y.; Newey, A.; et al. Genomic and Transcriptomic Determinants of Therapy Resistance and Immune Landscape Evolution during Anti-EGFR Treatment in Colorectal Cancer. Cancer Cell 2019, 36, 35–50.E9. [Google Scholar] [CrossRef]
  139. Garvey, C.M.; Lau, R.; Sanchez, A.; Sun, R.X.; Fong, E.J.; Doche, M.E.; Chen, O.; Jusuf, A.; Lenz, H.-J.; Larson, B.; et al. Anti-EGFR Therapy Induces EGF Secretion by Cancer-Associated Fibroblasts to Confer Colorectal Cancer Chemoresistance. Cancers 2020, 12, 1393. [Google Scholar] [CrossRef]
  140. Zhou, J.; Ji, Q.; Li, Q. Resistance to anti-EGFR therapies in metastatic colorectal cancer: Underlying mechanisms and reversal strategies. J. Exp. Clin. Cancer Res. 2021, 40, 328. [Google Scholar] [CrossRef] [PubMed]
  141. Shah, R.; Johnson, K.A.; Lippert, A.E.L.; Kraus, S.G.; Emmerich, P.B.; Pasch, C.A.; Zhang, W.; Matkowskyj, K.A.; LeBeau, A.M.; Deming, D.A. Cancer-Associated Fibroblast Proteins as Potential Targets against Colorectal Cancers. Cancers 2024, 16, 3158. [Google Scholar] [CrossRef]
  142. Cao, Z.; Quazi, S.; Arora, S.; Osellame, L.D.; Burvenich, I.J.; Janes, P.W.; Scott, A.M. Cancer-associated fibroblasts as therapeutic targets for cancer: Advances, challenges, and future prospects. J. Biomed. Sci. 2025, 32, 7. [Google Scholar] [CrossRef]
  143. Vaquero, J.; Pavy, A.; Gonzalez-Sanchez, E.; Meredith, M.; Arbelaiz, A.; Fouassier, L. Genetic alterations shaping tumor response to anti-EGFR therapies. Drug Resist. Updates 2022, 64, 100863. [Google Scholar] [CrossRef] [PubMed]
  144. Stahler, A.; Kind, A.J.; Sers, C.; Mamlouk, S.; Müller, L.; Karthaus, M.; Fruehauf, S.; Graeven, U.; Fischer Von Weikersthal, L.; Sommerhäuser, G.; et al. Negative Hyperselection of Resistance Mutations for Panitumumab Maintenance in RAS Wild-Type Metastatic Colorectal Cancer (PanaMa Phase II Trial, AIO KRK 0212). Clin. Cancer Res. 2024, 30, 1256–1263. [Google Scholar] [CrossRef]
  145. Fucà, G.; Raimondi, A.; Prisciandaro, M.; Lonardi, S.; Cremolini, C.; Ratti, M.; Clavarezza, M.; Murialdo, R.; Sartore-Bianchi, A.; Smiroldo, V.; et al. Reinduction of an Anti-EGFR-based First-line Regimen in Patients with RAS Wild-type Metastatic Colorectal Cancer Enrolled in the Valentino Study. Oncologist 2022, 27, e29–e36. [Google Scholar] [CrossRef]
  146. Shitara, K.; Muro, K.; Watanabe, J.; Yamazaki, K.; Ohori, H.; Shiozawa, M.; Yasui, H.; Oki, E.; Sato, T.; Naito, T.; et al. Negative hyperselection of patients with RAS wild-type metastatic colorectal cancer for panitumumab: A biomarker study of the phase III PARADIGM trial. J. Clin. Oncol. 2023, 41, 11. [Google Scholar] [CrossRef]
  147. Morano, F.; Corallo, S.; Lonardi, S.; Raimondi, A.; Cremolini, C.; Rimassa, L.; Murialdo, R.; Zaniboni, A.; Sartore-Bianchi, A.; Tomasello, G.; et al. Negative Hyperselection of Patients with RAS and BRAF Wild-Type Metastatic Colorectal Cancer Who Received Panitumumab-Based Maintenance Therapy. J. Clin. Oncol. 2019, 37, 3099–3110. [Google Scholar] [CrossRef]
  148. Randon, G.; Pietrantonio, F. Towards Multiomics-Based Dissection of Anti-EGFR Sensitivity in Colorectal Cancer. Clin. Cancer Res. 2023, 29, 4021–4023. [Google Scholar] [CrossRef] [PubMed]
  149. Gupta, R.G.; Somer, R.A. Intratumor Heterogeneity: Novel Approaches for Resolving Genomic Architecture and Clonal Evolution. Mol. Cancer Res. 2017, 15, 1127–1137. [Google Scholar] [CrossRef]
  150. Assi, T.; Khoury, R.; Ibrahim, R.; Baz, M.; Ibrahim, T.; Le Cesne, A. Overview of the role of liquid biopsy in cancer management. Transl. Oncol. 2023, 34, 101702. [Google Scholar] [CrossRef]
  151. Petrillo, A.; Salati, M.; Trapani, D.; Ghidini, M. Circulating Tumor DNA as a Biomarker for Outcomes Prediction in Colorectal Cancer Patients. Curr. Drug Targets 2021, 22, 1010–1020. [Google Scholar] [CrossRef]
  152. Larribère, L.; Martens, U.M. Advantages and Challenges of Using ctDNA NGS to Assess the Presence of Minimal Residual Disease (MRD) in Solid Tumors. Cancers 2021, 13, 5698. [Google Scholar] [CrossRef] [PubMed]
  153. Oliveira, K.C.S.; Ramos, I.B.; Silva, J.M.C.; Barra, W.F.; Riggins, G.J.; Palande, V.; Pinho, C.T.; Frenkel-Morgenstern, M.; Santos, S.E.B.; Assumpcao, P.P.; et al. Current Perspectives on Circulating Tumor DNA, Precision Medicine, and Personalized Clinical Management of Cancer. Mol. Cancer Res. 2020, 18, 517–528. [Google Scholar] [CrossRef]
  154. Kato, S.; Okamura, R.; Mareboina, M.; Lee, S.; Goodman, A.; Patel, S.P.; Fanta, P.T.; Schwab, R.B.; Vu, P.; Raymond, V.M.; et al. Revisiting Epidermal Growth Factor Receptor (EGFR) Amplification as a Target for Anti-EGFR Therapy: Analysis of Cell-Free Circulating Tumor DNA in Patients with Advanced Malignancies. JCO Precis. Oncol. 2019, 3, 1–14. [Google Scholar] [CrossRef]
  155. van ’t Erve, I.; Greuter, M.J.E.; Bolhuis, K.; Vessies, D.C.L.; Leal, A.; Vink, G.R.; van den Broek, D.; Velculescu, V.E.; Punt, C.J.A.; Meijer, G.A.; et al. Diagnostic Strategies toward Clinical Implementation of Liquid Biopsy RAS/BRAF Circulating Tumor DNA Analyses in Patients with Metastatic Colorectal Cancer. J. Mol. Diagn. 2020, 22, 1430–1437. [Google Scholar] [CrossRef]
  156. Sartore-Bianchi, A.; Pietrantonio, F.; Lonardi, S.; Mussolin, B.; Rua, F.; Crisafulli, G.; Bartolini, A.; Fenocchio, E.; Amatu, A.; Manca, P.; et al. Circulating tumor DNA to guide rechallenge with panitumumab in metastatic colorectal cancer: The phase 2 CHRONOS trial. Nat. Med. 2022, 28, 1612–1618. [Google Scholar] [CrossRef]
  157. Leite da Silva, L.F.; Saldanha, E.F.; de Menezes, J.S.A.; Halamy Pereira, L.; de Bragança Dos Santos, J.A.R.; Buonopane, I.R.; de Souza, E.M.; de Menezes, C.U.G.; Lopes, G. Plasma ctDNA kinetics as a predictor of systemic therapy response for advanced non-small cell lung cancer: A systematic review and meta-analysis. Oncologist 2025, 30, oyae344. [Google Scholar] [CrossRef]
  158. Malla, M.; Loree, J.M.; Kasi, P.M.; Parikh, A.R. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving Practices. J. Clin. Oncol. 2022, 40, 2846–2857. [Google Scholar] [CrossRef] [PubMed]
  159. Valenza, C.; Saldanha, E.F.; Gong, Y.; De Placido, P.; Gritsch, D.; Ortiz, H.; Trapani, D.; Conforti, F.; Cremolini, C.; Peters, S.; et al. Circulating tumor DNA clearance as a predictive biomarker of pathologic complete response in patients with solid tumors treated with neoadjuvant immune checkpoint inhibitors: A systematic review and meta-analysis. Ann. Oncol. 2025, 36, 726. [Google Scholar] [CrossRef]
  160. Napolitano, S.; Parikh, A.R.; Henry, J.; Parseghian, C.M.; Willis, J.; Raghav, K.P.; Morris, V.K.; Johnson, B.; Kee, B.K.; Dasari, A.N.; et al. Novel Clinical Tool to Estimate Risk of False-Negative KRAS Mutations in Circulating Tumor DNA Testing. JCO Precis. Oncol. 2023, 7, e2300228. [Google Scholar] [CrossRef] [PubMed]
  161. Keller, L.; Belloum, Y.; Wikman, H.; Pantel, K. Clinical relevance of blood-based ctDNA analysis: Mutation detection and beyond. Br. J. Cancer 2021, 124, 345–358. [Google Scholar] [CrossRef]
  162. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.; et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat. Med. 2020, 26, 1859–1864. [Google Scholar] [CrossRef] [PubMed]
  163. Pathak, P.S.; Chan, G.; Deming, D.A.; Chee, C.E. State-of-the-Art Management of Colorectal Cancer: Treatment Advances and Innovation. Am. Soc. Clin. Oncol. Educ. Book 2024, 44, e438466. [Google Scholar] [CrossRef]
  164. Medford, A.J.; Ellisen, L.W. Optimizing Access to Liquid Biopsy in the Present and Future Cancer Landscape. JCO Precis. Oncol. 2024, 8, e2300609. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, J.; Guo, B. RNA-based therapeutics for colorectal cancer: Updates and future directions. Pharmacol. Res. 2020, 152, 104550. [Google Scholar] [CrossRef] [PubMed]
  166. Boustany, L.M.; LaPorte, S.L.; Wong, L.; White, C.; Vinod, V.; Shen, J.; Yu, W.; Koditek, D.; Winter, M.B.; Moore, S.J.; et al. A Probody T Cell-Engaging Bispecific Antibody Targeting EGFR and CD3 Inhibits Colon Cancer Growth with Limited Toxicity. Cancer Res. 2022, 82, 4288–4298. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of primary resistance to anti-EGFR therapy in metastatic colorectal cancer. Several genetic alterations drive intrinsic resistance to EGFR-targeted monoclonal antibodies such as cetuximab and panitumumab. These include activating mutations in KRAS and BRAF, amplification or mutation of HER2 and MET, extracellular domain mutations in EGFR, and alterations in downstream signaling components such as PIK3CA, AKT1, MEK1, and PTEN. These molecular events activate parallel or downstream signaling cascades, including the RAS RAF MEK ERK and PI3K AKT mTOR pathways, which sustain tumor cell survival, proliferation, and disease progression independent of EGFR signaling blockade.
Figure 1. Mechanisms of primary resistance to anti-EGFR therapy in metastatic colorectal cancer. Several genetic alterations drive intrinsic resistance to EGFR-targeted monoclonal antibodies such as cetuximab and panitumumab. These include activating mutations in KRAS and BRAF, amplification or mutation of HER2 and MET, extracellular domain mutations in EGFR, and alterations in downstream signaling components such as PIK3CA, AKT1, MEK1, and PTEN. These molecular events activate parallel or downstream signaling cascades, including the RAS RAF MEK ERK and PI3K AKT mTOR pathways, which sustain tumor cell survival, proliferation, and disease progression independent of EGFR signaling blockade.
Cancers 17 02804 g001
Figure 2. Mechanisms of Acquired Resistance to Anti-EGFR Therapy in Metastatic Colorectal Cancer. EGFR-targeted monoclonal antibodies (cetuximab or panitumumab) initially reduce tumor burden in RAS/BRAF wild-type colorectal cancer. However, under treatment pressure, resistant clones (e.g., with KRAS mutations) are positively selected or newly emerge through acquired mutations. These resistant subclones expand over subsequent treatment cycles, ultimately leading to disease progression despite continued anti-EGFR therapy.
Figure 2. Mechanisms of Acquired Resistance to Anti-EGFR Therapy in Metastatic Colorectal Cancer. EGFR-targeted monoclonal antibodies (cetuximab or panitumumab) initially reduce tumor burden in RAS/BRAF wild-type colorectal cancer. However, under treatment pressure, resistant clones (e.g., with KRAS mutations) are positively selected or newly emerge through acquired mutations. These resistant subclones expand over subsequent treatment cycles, ultimately leading to disease progression despite continued anti-EGFR therapy.
Cancers 17 02804 g002
Figure 3. Precision Oncology Framework for Anti-EGFR Therapy in Metastatic Colorectal Cancer. Liquid biopsy enables noninvasive, real-time molecular profiling across the treatment course. At baseline, panels assessing RAS/BRAF status and additional negative hyperselection markers (e.g., HER2, MET, MAP2K1, PIK3CA, PTEN, gene fusions) identify patients most likely to benefit from EGFR blockade. During treatment and at disease progression, serial ctDNA analysis detects acquired resistance mechanisms, supporting rechallenge decisions or the addition of targeted agents. This dynamic strategy integrates predictive and adaptive molecular insights to optimize anti-EGFR therapy.
Figure 3. Precision Oncology Framework for Anti-EGFR Therapy in Metastatic Colorectal Cancer. Liquid biopsy enables noninvasive, real-time molecular profiling across the treatment course. At baseline, panels assessing RAS/BRAF status and additional negative hyperselection markers (e.g., HER2, MET, MAP2K1, PIK3CA, PTEN, gene fusions) identify patients most likely to benefit from EGFR blockade. During treatment and at disease progression, serial ctDNA analysis detects acquired resistance mechanisms, supporting rechallenge decisions or the addition of targeted agents. This dynamic strategy integrates predictive and adaptive molecular insights to optimize anti-EGFR therapy.
Cancers 17 02804 g003
Table 2. Clinical Studies evaluating the role of negative hyperselection in mCRC.
Table 2. Clinical Studies evaluating the role of negative hyperselection in mCRC.
StudyPatient PopulationBiomarkers EvaluatedOverall Response Rate (%)Median PFS (Months)Median OS (Months)Key Findings
PanaMa Trial [140]mCRC, RAS WTBRAF V600E/PIK3CA/AKT1/ALK/ERBB2/PTEN MUT and HER2/neu35.8% in hyperselected vs. 25%7.5 months in hyperselected vs. 4.4 months28.7 months in hyperselected vs. 22.2 monthsHyperselected patients had significantly better PFS and OS with panitumumab maintenance.
Valentino Study [144]mCRC, RAS WTHER2/MET PIK3CA/PTEN mutationsN/A10.5 months in hyperselected vs. 6.03 months33.3 months in hyperselected vs. 14.1 monthsReinduction with panitumumab was more effective in hyperselected patients.
PANDA Trial [145]Elderly mCRC, RAS WT, BRAF WTHER2/MET NTRK/ROS1/ALK/RET PIK3CA PTEN AKT1 MAP2K171% in hyperselected vs. 51%12.8 months in hyperselected vs. 7.6 months29.5 months in hyperselected vs. 20 monthsElderly hyperselected patients showed better PFS and OS with anti-EGFR therapy.
PRESSING2 Study [87]mCRC, RAS WT, MSS, POLE WTERBB2/MET NTRKs/RET/ROS1/ALK AKT1/PTEN/PIK3CA79% in hyperselected vs. 56%12.8 months in hyperselected vs. 6.4 months49.9 months in hyperselected vs. 22.6 monthsUltraselected patients (without rare mutations) had significantly better survival outcomes.
PARADIGM Study [17]mCRC, RAS WTPTEN/EGFR HER2/MET ALK/RET/NTRK1N/AN/A41.4 months in hyperselected vs. 18.7Exploratory analyses suggest that ctDNA is useful to identify gene alterations that predict resistance to EGFR inhibition.
Morano et al. [146]mCRC, RAS/BRAF WTALK/ROS1/NTRKs/RET HER2/PIK3CAex.20/PTEN/AKT175.3% in hyperselected vs. 59.2%12.1 months in hyperselected vs. 7.768.1 months in hyperselected vs. 48.1Patients with right-sided tumors and those without hyperselected tumors had significantly poor outcomes in terms of overall ORR, PFS, and OS.
BRAF WT: BRAF Wild Type; ctDNA: Circulating Tumor DNA; FOLFOX: (Oxaliplatin, Leucovorin, and 5-Fluorouracil); mFOLFOX6: Modified FOLFOX6 (Oxaliplatin, Leucovorin, and 5-Fluorouracil); mCRC: Metastatic Colorectal Cancer; Median OS: Median Overall Survival; Median PFS: Median Progression-Free Survival; MSS: Microsatellite Stable; 5-FU/FA: 5-Fluorouracil/Folinic Acid; POLE WT: POLE Wild Type; RAS WT: RAS Wild Type; N/A: Not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Leite, L.F.; Noronha, M.M.; de Menezes, J.S.A.; da Conceição, L.D.; Almeida, L.F.C.; Cappellaro, A.P.; Belotto, M.; Biachi de Castria, T.; Peixoto, R.D.; Megid, T.B.C. Anti-EGFR Therapy in Metastatic Colorectal Cancer: Identifying, Tracking, and Overcoming Resistance. Cancers 2025, 17, 2804. https://doi.org/10.3390/cancers17172804

AMA Style

Leite LF, Noronha MM, de Menezes JSA, da Conceição LD, Almeida LFC, Cappellaro AP, Belotto M, Biachi de Castria T, Peixoto RD, Megid TBC. Anti-EGFR Therapy in Metastatic Colorectal Cancer: Identifying, Tracking, and Overcoming Resistance. Cancers. 2025; 17(17):2804. https://doi.org/10.3390/cancers17172804

Chicago/Turabian Style

Leite, Luís Felipe, Mariana Macambira Noronha, Junior Samuel Alonso de Menezes, Lucas Diniz da Conceição, Luiz F. Costa Almeida, Anelise Poluboiarinov Cappellaro, Marcos Belotto, Tiago Biachi de Castria, Renata D’Alpino Peixoto, and Thais Baccili Cury Megid. 2025. "Anti-EGFR Therapy in Metastatic Colorectal Cancer: Identifying, Tracking, and Overcoming Resistance" Cancers 17, no. 17: 2804. https://doi.org/10.3390/cancers17172804

APA Style

Leite, L. F., Noronha, M. M., de Menezes, J. S. A., da Conceição, L. D., Almeida, L. F. C., Cappellaro, A. P., Belotto, M., Biachi de Castria, T., Peixoto, R. D., & Megid, T. B. C. (2025). Anti-EGFR Therapy in Metastatic Colorectal Cancer: Identifying, Tracking, and Overcoming Resistance. Cancers, 17(17), 2804. https://doi.org/10.3390/cancers17172804

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop