Next Article in Journal
Engineered Bacteria-Nano Hybrid System: The Intelligent Drug Factory for Next-Generation Cancer Immunotherapy
Previous Article in Journal
Design of Polycation-Functionalized Resveratrol Nanocrystals for Intranasal Administration
Previous Article in Special Issue
Innovative Approaches in Cancer Treatment: Emphasizing the Role of Nanomaterials in Tyrosine Kinase Inhibition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Kinase Suppressor of Ras 1 (KSR1) for Cancer Therapy

Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2025, 17(10), 1348; https://doi.org/10.3390/pharmaceutics17101348
Submission received: 29 August 2025 / Revised: 15 October 2025 / Accepted: 17 October 2025 / Published: 19 October 2025

Abstract

Carcinogenesis is driven by aberrant activation of molecular signaling pathways governing cell proliferation, apoptosis, and differentiation. Among these, the RAS/RAF/MEK/ERK (RAS/MAPK) cascade is one of the most frequently dysregulated oncogenic pathways, driving tumor initiation and progression across diverse cancer types. Although inhibitors of BRAF and MEK have achieved clinical success in selected malignancies, adaptive resistance often undermines therapeutic durability. This has spurred interest in alternative nodes within the pathway. The kinase suppressor of Ras (KSR) is a scaffold protein that organizes RAF, MEK, and ERK into functional complexes, ensuring efficient and sustained signal transmission. Once regarded as a passive structural component, KSR1 is now recognized as an active regulator of pathway dynamics. Emerging evidence indicates that KSR1 overexpression promotes cancer cell proliferation and survival, while genetic or pharmacologic inhibition of KSR1 attenuates RAS/MAPK signaling and suppresses tumor growth in preclinical models. In this review, we provide a comprehensive overview of accessory and scaffold proteins modulating the RAS/MAPK pathway, with a particular focus on KSR1. We highlight its structural and functional properties, summarize preclinical evidence for KSR1-targeted interventions, and discuss its therapeutic potential in cancer, with emphasis on hepatocellular carcinoma (HCC).

1. Introduction

Carcinogenesis is a multistep process driven by the accumulation of genetic and epigenetic alterations leading to a constitutive activation of intracellular molecular signaling networks that control proliferation, survival, and differentiation [1,2,3]. Several core oncogenic signaling pathways are recurrently dysregulated in human malignancies, most notably the RAS/RAF/MEK/ERK (RAS/MAPK) cascade, the PI3K/AKT/mTOR pathway, and the JAK/STAT pathway [4,5,6]. Among these, the RAS/MAPK signaling cascade is one of the most frequently activated across cancer types. In most cases, sustained activation of this pathway arises from activating mutations in members of the RAS gene family (HRAS, KRAS, NRAS) or RAF kinases (ARAF, BRAF, CRAF) [4]. For instance, constitutively activating mutations in KRAS are detected in more than 90% of pancreatic ductal adenocarcinomas [7]. As well, over half of colorectal adenocarcinoma harbor activating mutations in RAS or RAF genes, maintaining constitutive RAS/MAPK pathway activation [4]. In papillary thyroid carcinoma, BRAF mutations are particularly prevalent, occurring in up to 70% of patients, and serve as a major driver of aberrant pathway activation in this malignancy [8].
Owing to its pivotal role in tumor initiation and progression, the RAS/MAPK signaling pathway has been a major focus for targeted therapy development [9,10]. For example, small-molecule inhibitors of BRAF have demonstrated significant clinical benefit, particularly in tumors harboring specific activating mutations. However, treatment efficacy is often limited by the emergence of drug-resistant tumor cells, which compromise the durability of therapeutic responses [4,11,12]. These challenges have stimulated interest in targeting alternative nodes within the pathway, including non-catalytic regulators such as scaffold proteins. In particular, kinase suppressor of Ras 1 (KSR1) has emerged as a key modulator of RAS/MAPK signaling and a novel therapeutic target for cancer with pathway activation.

2. The RAS/MAPK Signaling Pathway

The RAS/MAPK signaling cascade is a highly conserved intracellular signaling pathway that governs fundamental cellular processes, including proliferation, differentiation, survival, and migration. Its activation is typically initiated at the cell surface through ligand binding to transmembrane receptors such as receptor tyrosine kinases (RTKs) and, in some contexts, G-protein-coupled receptors (GPCRs) [13,14]. Upon ligand engagement, RTKs undergo conformational changes that promote dimerization, leading to trans-autophosphorylation of specific tyrosine residues within their cytoplasmic domains (Figure 1). These phosphorylated tyrosines serve as docking sites for adaptor proteins such as growth factor receptor-bound protein 2 (Grb2). Grb2 recruits the guanine nucleotide exchange factor (GEF), son of sevenless (SOS), to the plasma membrane, where it catalyzes the exchange of GDP for GTP on the small GTPase RAS. This molecular switch activates RAS by inducing a conformational change that enables its interaction with downstream effectors, RAF kinases [14,15,16].
Recruited by activated RAS to the plasma membrane, RAF becomes activated through dimerization. Once activated, RAF in turn phosphorylates the mitogen-activated protein kinase kinases, MEK1 and MEK2, on conserved serine residues. MEKs are dual-specificity kinases, capable of phosphorylating both threonine and tyrosine residues, a feature that distinguishes them from most other kinases [17,18]. Activated MEK1/2 phosphorylates extracellular signal-regulated kinases ERK1 and ERK2, which are the terminal effectors of the pathway. Once phosphorylated, ERKs translocate from the cytoplasm into the nucleus, where they phosphorylate a wide array of nuclear substrates, including key transcription factors such as c-Fos and c-Jun—components of the activator protein-1 (AP-1) complex [19,20]. AP-1 regulates gene expression by binding to specific DNA sequences within promoter regions of target genes involved in cell cycle progression, survival, angiogenesis, and epithelial-mesenchymal transition [13,14].

3. Accessory and Scaffold Proteins in the RAS/MAPK Signaling Pathway

The RAS/MAPK signaling pathway is not simply a linear cascade of kinase activation; rather, it is a highly regulated signaling network orchestrated by numerous non-enzymatic proteins that spatially and temporally coordinate signaling dynamics. Among these, accessory and scaffold proteins play indispensable roles [21,22]. Though they are not catalytic components of the pathway, they serve as molecular integrators that assemble multi-protein complexes, regulate subcellular localization, and fine-tune the magnitude, duration, and specificity of signal propagation. These proteins ensure that signaling responses are precisely modulated in accordance with extracellular stimuli and the physiological context. Accessory proteins in the RAS/MAPK pathway can be broadly categorized into four functional groups based on their structural characteristics and regulatory mechanisms: anchoring proteins, docking proteins, adaptor proteins, and scaffold proteins [4,21]. Each class contributes to organizing and orchestrating signal flow at distinct cellular compartments and stages of the cascade (Figure 2).

3.1. Anchoring Proteins: Spatial Regulation of Kinase Activity

Anchoring proteins primarily regulate the spatial distribution of signaling complexes by tethering pathway components to specific membrane domains or organelles. These proteins enable the spatial compartmentalization of signaling, which is increasingly recognized as a critical determinant of signaling specificity. CNK1 (Connector Enhancer of KSR1) is a well-characterized anchoring protein that supports RAF membrane recruitment and dimerization upon RAS activation. CNK1 interacts with RAF isoforms and is required for efficient RAF activation. Notably, CNK1 contains a pleckstrin homology (PH) domain that facilitates membrane association. Pharmacological inhibition of CNK1 using small molecules such as PHT-7.3 impairs KRAS4B-driven signaling by disrupting CNK1-RAS colocalization at the plasma membrane, thereby reducing downstream RAF/MEK/ERK activity [4,21,23]. Flotillins (FLOT1 and FLOT2) are lipid raft-associated proteins that localize to membrane microdomains via their prohibitin homology (PHB) domains. Flotillins mediate clustering of receptors such as EGFR and are directly implicated in enhancing MAPK pathway signaling by interacting with CRAF, MEK, and ERK following stimulation. Their function underscores the importance of membrane microdomain composition in modulating signaling efficiency [21,24,25]. LAT (Linker for Activation of T cells) and NTAL (Non–T cell Activation Linker) are transmembrane scaffold proteins involved in immune cell signaling but also participate in MAPK regulation by anchoring receptor complexes to the membrane and facilitating the recruitment of cytoplasmic signaling proteins [4,21,26].
Spatial regulation also occurs at intracellular sites. For example, PAQR10 and PAQR11, members of the progestin and adipoQ receptor family, localize RAS to the Golgi apparatus via their N-terminal cytoplasmic tails. This non-canonical localization of RAS is functionally significant, as Golgi-localized RAS can engage distinct downstream effectors compared to membrane-localized RAS. Likewise, SEF interacts with phosphorylated MEK on the Golgi and modulates MAPK activity, potentially shaping sustained ERK signaling that is critical for certain cellular outcomes [27,28,29].

3.2. Docking Proteins: Bridging Receptors and Signaling Modules

Docking proteins act as platforms that physically link activated receptors to intracellular signaling components. They typically interact with both membrane-localized receptors and cytoplasmic effectors, allowing rapid assembly of signaling complexes. DOK1 and DOK2 (Downstream of Kinase proteins) function in RTK signaling by recruiting RAS effectors and modulating RAS activation [30]. They contain PH and PTB (phosphotyrosine-binding) domains that confer membrane localization and enable specific interactions with phosphotyrosine residues on activated receptors. IRS2 (Insulin receptor substrate 2), a classical docking protein in insulin/IGF signaling, also plays roles in RAS/MAPK activation by facilitating the recruitment of Grb2. Its dual-domain architecture supports both membrane localization and interaction with activated RTKs [21,31].
β-arrestins 1 and 2, traditionally known for their roles in GPCR desensitization and internalization, also serve as docking proteins for MAPK signaling. Upon GPCR activation, β-arrestins can recruit and stabilize RAF-MEK-ERK complexes, allowing signal propagation not only at the plasma membrane but also from endosomal compartments. This endosomal signaling allows for sustained ERK activation and can influence specific transcriptional outcomes [21,32,33]. FGF Receptor Substrate 2 (FRS2) is a docking protein that binds activated FGFRs via its PTB domain. FRS2 becomes tyrosine-phosphorylated upon receptor stimulation and subsequently recruits adapter proteins GRB2 and SHP2 (Src homology region 2 domain-containing phosphatase-2), forming a critical link between activated RTKs and the RAS/MAPK signaling axis [4,34].

3.3. Adaptor Proteins: Signal Integration and Branching

Adaptor proteins function as molecular bridges, bringing together upstream activators and downstream effectors. Their modular domains allow combinatorial interactions, enabling signal branching and integration [35]. CRK and CRK-like protein (CRKL) contain an N-terminal SH2 domain followed by two SH3 domains (SH3n and SH3c). These proteins link phosphotyrosine motifs on activated receptors to various proline-rich effectors. Interestingly, the SH2 domain of CRK promotes oncogenic transformation, whereas its SH3 domains can exert context-dependent negative regulatory effects [36]. GRB2 is one of the most critical adaptors in RAS signaling. Its SH2 domain binds to phosphotyrosine residues on RTKs or docking proteins such as FRS2 and SHC (Src Homology and Collagen), while its two SH3 domains interact with proline-rich regions of SOS1 (a guanine nucleotide exchange factor), enabling the activation of RAS. GRB2 is structurally organized with two α-helices flanking a central β-sheet in the SH2 domain, providing specificity for phosphorylated motifs [37,38,39].
SHC acts upstream of GRB2 and facilitates its recruitment to receptor complexes. SHC possesses a CH2-PTB-CH1-SH2 topology and undergoes phosphorylation at multiple sites following receptor activation, acting as a versatile hub that bridges diverse upstream signals to the RAS pathway [40]. SHP2 is a protein tyrosine phosphatase with two SH2 domains and a catalytic domain that is autoinhibited via intramolecular interactions. Upon phosphorylation at Y542 by RTKs, this inhibition is relieved, and SHP2 becomes active. SHP2 plays a key positive role in RAS activation by promoting GRB2-SOS complex assembly and counteracting inhibitory phosphatases [41]. Paxillin is a focal adhesion adapter containing binding sites for SH2 and SH3 domain-containing proteins. It forms complexes with kinases and scaffold proteins involved in RAS/MAPK signaling, contributing to cytoskeletal dynamics and cell migration in a signaling-dependent manner [42].

3.4. Scaffold Proteins: Assembling and Modulating the MAPK Module

Scaffold proteins coordinate signaling by physically assembling multiple kinases into pre-formed complexes. This organization enhances the efficiency and specificity of signal propagation while also preventing inappropriate cross-talk. KSR1 and KSR2 are prototypical RAS/MAPK scaffolds that organize RAF, MEK, and ERK into signaling-competent complexes [43,44]. KSR1 facilitates MEK phosphorylation by RAF and promotes ERK activation, thereby acting as a positive regulator of MAPK output. Its functional homolog KSR2 performs similar roles, though it is more restricted in tissue distribution. Importantly, both KSR1 and KSR2 interact with 14-3-3 proteins, which modulate their subcellular localization and scaffold activity, thereby fine-tuning pathway dynamics [45].
IQGAP1, a multifunctional scaffold, interacts with RTKs such as EGFR and HER2 (Human Epidermal growth factor Receptor 2). It links receptor activation to the MAPK cascade either through direct interaction or via intermediate adaptors like ShcA. IQGAP1 binds both inactive and activated receptors and contributes to receptor trafficking, signal duration, and ERK nuclear translocation [46]. SHOC2 represents another important scaffold. It forms a complex with the catalytic subunit of protein phosphatase 1 (PP1) to mediate RAF dephosphorylation at Ser259, an essential step for RAF dimerization and full activation [47]. Through this function, SHOC2 acts as a gatekeeper of RAS/MAPK pathway activation and has been implicated in both developmental disorders (Noonan-like syndromes) and cancer [48].

3.5. Negative Modulators of RAS/MAPK Signaling

SPRED (Sprouty-related EVH1 domain-containing protein) promotes the function of RasGAPs (Ras GTPase activating proteins), thereby accelerating RAS inactivation and suppressing RAS/MAPK signaling [49]. Expression of SPRED proteins, SPRED1 in particular, is frequently reduced in various human cancers, compared with adjacent non-tumorous tissues [49,50]. Sprouty family (SPRY1–4) is a feedback inhibitor of receptor tyrosine kinase (RTK) signaling. The precise molecular mechanism by which Spry proteins restrain RTK signaling remains unclear; however, it has been proposed that they sequester GRB2 from SOS by binding to its SH2 domain, thereby impairing RAS activation [51]. RKIP (Raf Kinase Inhibitory Protein) is a prototypical negative regulator of RAS/MAPK signaling. RKIP binds to the N-terminal region of RAF, competitively blocking the RAF–MEK interaction, thereby preventing MEK phosphorylation and downstream ERK activation [52]. Beyond this direct inhibitory role, RKIP functions as a signaling switch: under certain stimuli, protein kinase C (PKC)–mediated phosphorylation of RKIP induces its dissociation from RAF, relieving inhibition and allowing MAPK signaling to proceed [53]. Loss or downregulation of RKIP has been observed in various cancers, including prostate, breast, and hepatocellular carcinoma, where it correlates with enhanced metastatic potential and poor prognosis, underscoring its role as a metastasis suppressor [54,55,56,57].
Erbin, a LAP (leucine-rich repeat and PDZ domain) family protein, also participates in fine-tuning RAS/MAPK signaling activity (Table 1). Erbin directly binds the C-terminal tail of HER2 and, through its interaction with SHOC2, regulates the activation status of RAF. As well, by disrupting the interaction between RAS and RAF, Erbin can dampen RAS/MAPK pathway activation [58]. Dual-Specificity Phosphatases (DUSPs) not only directly dephosphorylate ERK, but they also physically interact with RAS/MAPK components to tether them in inactive complexes [59,60]. DUSP6 (MKP-3), for instance, preferentially targets ERK1/2 and has been reported to restrain MAPK-driven proliferation in multiple tissues [61]. Reduced expression of DUSP7 has been observed in cervical cancer, permitting sustained MAPK activation [62].

4. KSR: A Central Scaffold in RAS/MAPK Signaling

The Kinase Suppressor of Ras (KSR) represents a unique class of regulatory proteins that function predominantly as molecular scaffolds within the RAS/MAPK signaling pathway. Initially identified through classical genetic screens in Drosophila melanogaster and Caenorhabditis elegans, KSR was discovered not as a conventional kinase but rather as a modulator essential for Ras-driven phenotypes [64,65]. This finding shifted the paradigm of signal transduction from a sole emphasis on enzymatic phosphorylation events to a broader understanding of the critical role played by scaffolding proteins. Unlike traditional kinases that catalyze phosphorylation, KSR proteins exhibit limited intrinsic kinase activity. Instead, KSR exerts its regulatory function by physically assembling the core components of the RAS/MAPK cascade—RAF, MEK, and ERK—into macromolecular complexes (Figure 3a). This assembly not only increases the efficiency of signal transmission but also ensures signaling specificity by restricting cross-talk and inappropriate pathway activation. Through its modular domain structure and tightly regulated subcellular localization, KSR acts as a precision controller that modulates the amplitude, duration, and localization of MAPK signaling outputs, thereby influencing diverse cellular outcomes such as proliferation, differentiation, and survival.

4.1. Structure of KSR Proteins

In mammals, two paralogs of the Kinase Suppressor of Ras (KSR) have been identified, namely KSR1 and KSR2, which share significant structural similarity. Both KSR1 and 2 proteins exhibit a domain organization reminiscent of RAF kinases. This shared architecture consists of five conserved regions, termed CA1 through CA5, each contributing distinct functional roles critical for KSR’s scaffolding activity and regulatory control within the MAPK pathway (Figure 3b) [44,69].

4.1.1. CA1 (Coiled-Coil/SAM Domain)

Located at the N-terminus, the CA1 domain encompasses approximately 40 amino acids, forming a sterile alpha motif (SAM). This domain is essential for membrane association and heterodimerization with RAF kinases, with a particular affinity for B-RAF. The SAM domain facilitates the spatial organization of signaling complexes at the plasma membrane, an essential step during pathway activation, thereby enabling effective signal transmission [70,71].

4.1.2. CA2 (Proline-Rich Domain)

Although less characterized, the proline-rich CA2 domain likely mediates interactions with SH3 domain-containing adaptor or signaling proteins. The region adjacent to CA2 has been shown to interact with AMP-activated protein kinase (AMPK), suggesting a novel role for KSR in integrating cellular energy status with MAPK signaling output, potentially linking metabolic cues to proliferative signaling [72].

4.1.3. CA3 (Atypical C1 Domain)

Homologous to the CR1 domain of RAF kinases, the domain diverges functionally by lacking robust diacylglycerol (DAG) binding. Instead, it retains the capacity to bind negatively charged phospholipids, anchoring KSR to anionic lipid-enriched microdomains at the plasma membrane. This membrane tethering is critical for colocalizing MEK and RAF kinases within a confined membrane environment, facilitating efficient phosphorylation events [73].

4.1.4. CA4 (Serine/Threonine-Rich Domain)

This region contains an FXFP motif, a canonical docking site recognized by activated ERK. By retaining ERK within the scaffolded complex, KSR regulates the kinetics and localization of ERK activation. Moreover, this facilitates feedback phosphorylation of KSR itself or other scaffold components by ERK, thus modulating ERK substrate specificity and fine-tuning downstream signaling responses [44].

4.1.5. CA5 (Kinase/Pseudokinase Domain)

Structurally analogous to the CR3 kinase domain of RAF, CA5 is notable for its ability to bind MEK despite lacking robust kinase activity. Although KSR has been known as a pseudokinase, emerging evidence reveals that KSR can exhibit context-dependent catalytic function. Mutations or substitutions within CA5 have been shown to severely impair RAS/MAPK pathway activation, underscoring the critical scaffold-kinase interface [44,74].

4.2. Dynamic Regulation of KSR1 Localization

KSR1 is regulated by a multilayered mechanism involving phosphorylation events, interactions with regulatory proteins, and controlled intracellular trafficking, collectively ensuring signaling precision. Under resting conditions, C-TAK1 kinase phosphorylates KSR1 at serine residues Ser297 and Ser392. These phosphorylations promote high-affinity binding of 14-3-3 proteins, resulting in the cytosolic sequestration of KSR1 in an inactive conformation, thus preventing unwarranted MAPK activation [45]. Upon extracellular stimuli such as growth factor binding, activation of upstream RAS triggers protein phosphatase 2A (PP2A) to dephosphorylate Ser392. This dephosphorylation disrupts the 14-3-3 interaction, enabling KSR1 to translocate to the plasma membrane. At the membrane, KSR1 assembles the RAF–MEK–ERK signaling module into an active complex, thereby potentiating signal transduction [45].
The regulation of KSR1 localization restricts KSR1′s scaffolding function to appropriate signaling contexts, preventing aberrant RAS/MAPK pathway activation. Importantly, the subcellular localization of KSR1 is a critical determinant of RAS/MAPK pathway dynamics: membrane-associated KSR1 facilitates rapid and robust ERK activation, whereas cytoplasmic retention dampens signaling intensity and duration.

4.3. Beyond Scaffolding: Alternative Functions of KSR1 in RAS/MAPK Signaling

Traditionally, KSR1 is a major scaffold protein of the RAS/MAPK signaling pathway, forming a complex with the major components and facilitating phosphorylation events among them. For example, by simultaneously binding RAF and MEK, KSR1 spatially aligns these kinases and enhances MEK phosphorylation by RAF. In addition to the scaffolding role, several alternative mechanisms have been proposed on how KSR1 amplifies and modulates RAS/MAPK pathway signaling.
Research from Therrien and colleagues demonstrated that MEK binding to the kinase domain of KSR1 promotes asymmetric heterodimerization with BRAF, which allosterically enhances BRAF catalytic activity toward unbound MEK substrates [75]. These findings indicate that KSR–MEK complexes can allosterically activate BRAF via interactions between KSR’s N-terminal regulatory region and kinase domain, challenging the conventional view of KSR solely as a scaffold for recruiting MEK to RAF. As well, KSR1 likely protects MEK and ERK from dephosphorylation, extending active signaling—consistent with scaffold-mediated insulation of kinases and the prolonged ERK activation [76].

5. Roles of KSR1 in Cancer

Kinase suppressor of Ras 1 (KSR1) occupies a unique position within the RAS/MAPK signaling network, serving as both a facilitator and, under certain circumstances, a restrainer of pathway output. This functional duality—tumor-promoting in some contexts, tumor-suppressive in others—presents both a conceptual challenge and an opportunity for therapeutic innovation. While most experimental evidence underscores KSR1′s role in scaffolding RAF, MEK, and ERK to enhance oncogenic signaling, a subset of studies demonstrates that excessive KSR1 expression can paradoxically attenuate RAS/MAPK activity [77]. This duality in KSR1 function highlights its complexity as a signaling regulator. The same molecular scaffold can either enhance or restrain oncogenic signaling, depending on dosage, post-translational modifications, interaction partners, and cellular context. For therapeutic development, such biphasic behavior underscores the importance of precisely modulating KSR1 activity (Figure 4).

5.1. KSR1 as a Tumor-Promoter

Accumulating evidence from diverse experimental models indicates that KSR1 functions as a tumor-promoter, largely through its role in facilitating persistent activation of the RAS/MAPK signaling cascade. Functional analyses in mouse embryonic fibroblasts (MEFs) have provided direct evidence for KSR1′s oncogenic potential. Ectopic expression of KSR1 in MEFs induced sustained ERK phosphorylation, accelerated S-phase entry, and enhanced cell proliferation [78]. Conversely, KSR1-deficient MEFs displayed attenuated ERK activation and impaired transformation, phenotypes that were restored in a dose-dependent manner upon KSR1 re-expression, with ERK activation and transformation increasing up to 14-fold [76]. Notably, in Ras-deficient MEFs, ectopic KSR1 expression restored colony-forming ability, underscoring KSR1′s capacity to drive RAS/MAPK pathway activation independently of upstream RAS activity [79].
Loss-of-function studies in various cancer contexts further support KSR1′s tumor-promoting role. Targeted knockdown of KSR1 using shRNA markedly impairs anchorage-independent growth in soft agar assays and reduces tumor formation in xenograft mouse models, confirming its role in promoting tumorigenesis [69,80]. In pancreatic cancer, KSR1 deletion decreased RAS/MAPK signaling, suppressed cellular transformation, and modestly prolonged survival in mouse models [81]. In hematologic malignancies, KSR1 deficiency attenuated Myc activation via the RAS/MAPK pathway, reduced B cell proliferation, increased apoptosis, and delayed lymphoma onset [82]. In melanoma, KSR1 deletion impaired ERK phosphorylation, leading to reduced proliferation, invasion, and metastasis, along with increased senescence and apoptosis [83]. In skin tumor models, KSR1 deletion alone did not abolish papilloma formation, but significantly reduced tumorigenesis in the presence of oncogenic Ras mutations [84]. Similarly, KSR1 knockout mice displayed a significant reduction in mammary tumor burden in models expressing the polyomavirus middle T antigen, indicating the in vivo tumor-promoting function of KSR1 [85]. Taken together, these findings firmly establish KSR1 as a pivotal amplifier of oncogenic RAS/MAPK signaling. By modulating the intensity and persistence of ERK activation, KSR1 influences the threshold for tumor initiation and progression

5.2. KSR as a Tumor-Suppressor

Although the name Kinase Suppressor of Ras (KSR1) historically suggested an inhibitory role in RAS-mediated oncogenesis, evidence for its tumor-suppressive function is comparatively limited and context-dependent. Early studies reported that murine KSR1 could antagonize oncogenic Ras activity. In NIH3T3 cells, KSR1 expression suppressed Ras-induced transformation, while in embryonic neuroretina cells, it inhibited proliferation [86]. These findings provided the first experimental support for a potential negative regulatory function. In human embryonic kidney cells stimulated with insulin or phorbol ester—both potent activators of the RAS/MAPK pathway through protein kinase C—ectopic expression of KSR1 attenuated pathway activation [87,88].
Clinical evidence also points to a potential tumor-suppressive role for KSR1 in certain contexts. In breast cancer, higher KSR1 expression correlated with improved patient survival [89], suggesting that in specific tumor settings, KSR1 may attenuate rather than enhance oncogenic RAS/MAPK signaling. Supporting this notion, KSR1 expression has been reported to be significantly reduced in pancreatic ductal adenocarcinoma (PDAC) and papillary thyroid carcinoma (PTC) compared with adjacent non-tumorous tissue [90]. In contrast, hepatocellular carcinoma (HCC) and clear cell renal cell carcinoma (ccRCC) display marked upregulation of KSR1. It is noteworthy that activating mutations in the RAS and RAF genes are found in more than 90% of PDAC [91,92,93] and 70% of PTC cases [94], respectively, whereas such mutations are rarely detected in HCC and ccRCC.
The mechanisms underlying KSR1 downregulation in PDAC and PTC remain unclear, but one possibility is that reduced KSR1 expression serves to limit excessive RAS/MAPK pathway activation in tumors already driven by RAS or RAF mutations. Recent studies have demonstrated that hyperactivation of the RAS/MAPK cascade can be detrimental to tumor cell survival [95,96]. For example, enforced ERK expression in melanoma cells carrying an activating mutation in BRAF (BRAFV600E) triggered cell death in vitro and tumor regression in xenograft models [95]. Similarly, colorectal cancer cell lines harboring KRAS or BRAF mutations exhibited marked sensitivity to PP2A inhibition, which caused further amplification of MAPK signaling and led to cell death [96]. Together, these findings raise the intriguing possibility that elevated KSR1 expression may function as a tumor suppressive mechanism in cancers harboring constitutively active RAS or RAF mutations by driving RAS/MAPK signaling into a state of toxic hyperactivation, whereas in tumors lacking such mutations (e.g., HCC, ccRCC), increased KSR1 levels instead enhance oncogenic signaling and promote tumorigenesis.

6. Targeting KSR1

Although KSR1 has been well-characterized as a scaffold protein that coordinates RAS/MAPK cascade activation, its potential as a therapeutic target in oncology remains underexplored.

6.1. Preclinical Studies Targeting KSR1

Preclinical studies directly investigating KSR1 as a therapeutic target remain limited, largely due to the lack of potent and selective KSR1 inhibitors. Nevertheless, several lines of evidence suggest that KSR1 plays a critical role in tumor progression and may represent a tractable vulnerability. In endometrial cancer cell lines, KSR1 knockdown reduced ERK phosphorylation and suppressed cell proliferation [97]. Similarly, siRNA-mediated silencing of KSR1 in colorectal cancer cells triggered apoptosis and inhibited tumor growth in xenograft models [80]. In melanoma harboring the oncogenic BRAFV600E mutation, KSR1 knockout impaired cell proliferation, induced cell cycle arrest, and promoted apoptosis, highlighting a context-dependent synthetic vulnerability in BRAF-driven tumors [83]. Collectively, these findings point to KSR1 as a potentially valuable therapeutic target across multiple tumor types. However, the current evidence base is limited, emphasizing the urgent need for the development of effective KSR1-directed inhibitors, as well as optimized delivery strategies to achieve therapeutic efficacy in vivo.
While most current efforts focus on KSR1 inhibition, it is also conceivable that enhancing KSR1 activity could yield therapeutic benefits in specific tumor contexts where KSR exerts tumor-suppressive effects. Although it is theoretically possible to boost KSR’s activity in cancer via, for example, delivering a viral vector expressing KSR1, targeted activation of KSR1 remains technically challenging and untested in cancer. Moreover, given the context-dependent and potentially oncogenic consequences of KSR1 activation, current translational strategies are more feasibly directed toward KSR1 inhibition.

6.2. Potential Strategies for KSR1 Targeting

Given its central role as a scaffold in the RAS/MAPK cascade, KSR1 presents a unique set of opportunities and challenges for therapeutic targeting. Unlike kinases with well-defined catalytic pockets, KSR1 primarily functions through protein–protein interactions (PPIs) with upstream and downstream effectors, including RAF, MEK, and ERK. This structural role necessitates innovative strategies that disrupt its scaffolding function without broadly impairing MAPK signaling required for normal physiology (Figure 5).

6.2.1. Small-Molecule Disruptors of KSR1–RAF/MEK Interactions

Structural studies have identified multiple domains within KSR1 that mediate its binding to RAF kinases and MEK. Small molecules or peptidomimetics that selectively block these interfaces could effectively uncouple KSR1 from the MAPK module, reducing signal amplification in oncogenic contexts. Although no clinically approved compounds exist, high-throughput screening for PPI inhibitors has yielded candidate molecules capable of displacing KSR1 from RAF in vitro, leading to attenuated ERK phosphorylation. One of the most significant advances in the pharmacological targeting of KSR has been the discovery of APS-2-79, a small-molecule allosteric inhibitor that interferes with KSR and RAF heterodimerization, and inhibits oncogenic RAS/MAPK signaling [98]. By targeting a specific pocket within the pseudokinase domain, APS-2-79 stabilizes KSR in an inactive conformation, thereby preventing the formation of a functional RAF–KSR–MEK complex [69].
APS-2-79 demonstrated only modest activity in reducing cell viability in RAS-mutant cancer cell lines and showed little to no effect in RAF-mutant cancer cells [98]. This limited efficacy contrasts with the more pronounced tumor-suppressive effects observed following genetic depletion of KSR1, where siRNA- or knockout-mediated suppression of KSR1 markedly impaired cell proliferation, induced apoptosis, and reduced tumor growth in both in vitro and in vivo models [80,83,97]. These findings highlight an important gap between genetic validation of KSR1 as a therapeutic target and the current pharmacological tools available to inhibit its function. Consequently, there is a pressing need for the development of next-generation KSR1 inhibitors with improved potency and pharmacokinetic properties.

6.2.2. Targeted Protein Degradation Approaches

Targeted protein degradation strategies such as PROTACs (proteolysis-targeting chimeras) or molecular glues may be effective [99,100,101]. By recruiting KSR1 to E3 ubiquitin ligases, these agents could selectively deplete the protein in tumor cells while sparing non-essential paralogs (e.g., KSR2) or unrelated RAS/MAPK components.

6.2.3. RNA-Based Silencing Methods

RNA interference (RNAi) or antisense oligonucleotide (ASO) strategies have already demonstrated preclinical efficacy in suppressing KSR1 expression. siRNA-mediated knockdown has been shown to reduce proliferation, impair ERK signaling, and induce apoptosis in multiple tumor cell types, as noted above. Such approaches may be especially suitable for localized delivery to tumors, where nanoparticle-based carriers could be employed for targeted tissue uptake [102,103].

7. Targeting KSR1 in Hepatocellular Carcinoma

Hepatocellular carcinoma (HCC) accounts for more than 80% of primary liver cancers and ranks as the third leading cause of cancer-related death worldwide [104,105]. Global incidence is projected to rise in the coming decade, driven by persistent risk factors such as chronic hepatitis B and C infection, alcohol-related liver disease, and the growing prevalence of metabolic-associated steatohepatitis [106,107,108].
One particularly intriguing feature of hepatocellular carcinoma (HCC) is that activating mutations in RAS and RAF genes—common oncogenic drivers in many other solid tumors—are rare. Despite this low mutational frequency, the RAS/MAPK signaling pathway is frequently hyperactivated in HCC, with more than half of cases exhibiting elevated ERK activity [109,110]. This paradox suggests that pathway activation in HCC is largely driven by alternative mechanisms, including overexpression or constitutive activation of upstream receptor tyrosine kinases (such as EGFR, FGFR, and c-Met), amplification of autocrine or paracrine growth factor signaling, and suppression of negative regulators like Sprouty and dual-specificity phosphatases (DUSPs). These non-mutational mechanisms converge to sustain RAS/MAPK pathway signaling, promoting tumor cell proliferation, survival, invasion, and therapeutic resistance, and they underscore the importance of targeting pathway modulators beyond the canonical mutational hotspots.

7.1. Preclinical Studies Targeting RAS/MAPK Signaling in HCC

RAS/MAPK signaling is frequently upregulated in hepatocellular carcinoma (HCC). Comparative analyses have shown that in more than 50% of HCC tissues, RAS/MAPK activity is elevated relative to adjacent non-malignant liver tissues [13]. Pharmacologic inhibition of RAS/MAPK signaling with rigosertib suppresses HCC cell proliferation and induces apoptosis, accompanied by marked decreases in ERK phosphorylation [111,112]. However, the precise downstream mechanisms of rigosertib-mediated RAS inhibition remain incompletely understood. At the RAF node, RAF1 is directly targeted by miR-4510, leading to ERK inactivation and downregulation of ERK-dependent genes. Silencing RAF1 with siRNA similarly reduces ERK activity and proliferation in HCC cells [113].
MEK inhibition has also demonstrated antitumor efficacy in preclinical HCC models. The MEK inhibitor cobimetinib significantly reduced proliferation and induced apoptosis in sorafenib-resistant Hep3B cells. In xenograft mice bearing sorafenib-resistant HCC, cobimetinib treatment suppressed ERK phosphorylation and slowed tumor progression [114]. Likewise, siRNA-mediated knockdown of ERK2 in rat HCC cells reduced DNA synthesis, and pharmacologic inhibition with the MEK inhibitor U0126 produced comparable reductions in proliferative cell populations [115].
In addition to the core signaling kinases, several accessory proteins modulate RAS/MAPK signaling in HCC. The adaptor protein GRB2 links receptor tyrosine kinases to RAS activation via SOS [116]. Overexpression of RNF173 promotes GRB2 degradation, thereby suppressing RAF/MEK/ERK signaling and reducing HCC malignancy [117]. Another adaptor, SHP2, facilitates RAS/MAPK activation [118,119]. SHP2 knockdown in HCC cells markedly reduces tumorigenic potential, whereas reintroduction of wild-type SHP2 restores proliferation and tumor growth. In xenograft models, SHP2 silencing significantly suppressed tumor formation, while SHP2 overexpression accelerated tumor progression [120].
Negative regulators of RAS/MAPK signaling also play critical roles in HCC. SPRED1 overexpression suppresses ERK activation and proliferation, whereas dominant-negative SPRED1 mutants or SPRED1-targeting miRNAs enhance ERK phosphorylation [121,122]. In xenograft models, SPRED1 expression attenuated ERK activity, particularly when combined with sorafenib treatment. Similarly, SPRED2 overexpression inhibited proliferation and induced apoptosis, while SPRED2 knockdown increased ERK phosphorylation and accelerated tumor growth in vivo [123,124]. Another negative regulator, RKIP, binds RAF to block RAS/MAPK signaling [125]. In HCC cells, RKIP expression inversely correlates with MEK/ERK phosphorylation, and RKIP overexpression reduces HCC cell proliferation and migration by suppressing IGF-I–mediated MAPK activation [126].
Collectively, these findings underscore the central role of RAS/MAPK signaling in HCC pathogenesis and highlight multiple nodes—including RAF, MEK, ERK, adaptor proteins, and regulators—that represent viable targets for therapeutic intervention (Table 2).

7.2. Resistance to Drugs Targeting the RAS/MAPK Signaling Pathway in HCC

Although considerable progress has been made in developing targeted therapies for hepatocellular carcinoma (HCC), their long-term clinical efficacy remains limited due to the emergence of drug resistance [127,128,129,130]. Such resistance can be intrinsic—present before treatment initiation—or acquired during therapy as tumor cells adapt to sustained drug pressure [131]. Consequently, most patients with advanced HCC fail to achieve durable responses to systemic therapies, highlighting the urgent need to better understand and overcome resistance mechanisms.
Multiple mechanisms have been identified by which HCC cells evade the tumoricidal effects of molecularly targeted therapies. For example, inhibition of fibroblast growth factor receptor (FGFR) by the treatment with lenvatinib can trigger activation of the EGFR–PAK2–ERK5 signaling axis [128,132,133]. Accordingly, combined treatment with the EGFR inhibitor gefitinib and lenvatinib exerts strong anti-proliferative effects in EGFR-expressing liver cancer cell lines in vitro, as well as in xenografted cell lines, immunocompetent mouse models, and patient-derived HCC xenografts in vivo [134]. Using a CRISPR-based library screen, Lu et al. identified NF1 and DUSP9 as key drivers of lenvatinib resistance in HCC and further demonstrated that NF1 loss reactivates both the PI3K/AKT and RAS/MAPK pathways, whereas DUSP9 loss selectively activates the RAS/MAPK pathway, collectively promoting resistance [135].
Several mechanisms have been implicated in sorafenib resistance in HCC. Overexpression of fibroblast growth factors (FGFs) can promote resistance, thereby sustaining cancer cell survival [136,137,138]. Similarly, activation of the hepatocyte growth factor (HGF)/c-Met signaling axis has been shown to facilitate resistance [139]. Aberrant activation of EGFR and HER3 receptors, together with overexpression of multiple EGFR ligands, has also been observed in sorafenib-resistant HCC, providing an additional mechanism for drug evasion [140]. Intriguingly, enrichment of cancer stem cells (CSCs) represents another route of resistance, as CSCs possess intrinsic survival advantages and reduced drug uptake, enabling them to withstand sorafenib-induced tumor suppression [141,142]. Additionally, compensatory activation of the PI3K/AKT pathway provides an alternative survival route, enabling HCC cells to bypass sorafenib-induced apoptosis [143]. Consistent with this, combined treatment with sorafenib and MK-2206, an Akt inhibitor, has been shown to overcome the emergence of sorafenib resistance [144].
Regorafenib has been established as a second-line treatment option for advanced HCC following failure of first-line sorafenib or lenvatinib therapy [145,146,147,148,149]. However, in vitro studies have shown that long-term exposure to regorafenib can lead to the emergence of regorafenib-resistant HCC cells characterized by enhanced TGF-β signaling activity [150]. Inhibition of the TGF-β receptor in combination with regorafenib significantly increased cancer cell death and senescence. In another study, regorafenib-resistant HCC cells exhibited high expression of SphK2. Ectopic overexpression of SphK2 reduced sensitivity to regorafenib via concomitant activation of nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) signaling pathways [151]. Collectively, these findings highlight the multifaceted mechanisms by which HCC cells develop resistance to current RTK- and RAS/MAPK-targeted therapies and emphasize the need for novel strategies to overcome resistance within this signaling network (Table 3).

7.3. KSR as a Therapeutic Target in HCC

The Kinase Suppressor of Ras (KSR) plays a pivotal role in regulating the RAS/MAPK signaling cascade [69,79]. Overexpression of KSR has been shown to directly activate MAPK signaling [79] and to contribute to MAPK reactivation and ERK phosphorylation following pharmacologic inhibition of BRAF or MEK [152]. Furthermore, elevated KSR1 expression has been linked to reduced sensitivity to KRAS inhibitors [79]. In several malignancies—including pancreatic ductal adenocarcinoma, melanoma, and skin carcinoma—KSR1 has been identified as an oncogenic facilitator of RAS/MAPK signaling, and its genetic or pharmacological inhibition suppresses tumor growth [79]. In contrast, studies investigating KSR1 in hepatocellular carcinoma (HCC) have been remarkably limited until recently. In HCC models, KSR2 knockdown suppresses proliferation and migration, whereas KSR2 overexpression enhances malignant phenotypes, increases MEK and ERK phosphorylation, and attenuates the growth-inhibitory effects of sorafenib [153].
The recent work by Moon et al. represents the first comprehensive study to establish a functional link between KSR1 and HCC pathogenesis [90], offering new insights into targeting RAS/MAPK signaling. KSR1 overexpression was frequently observed in HCC patient samples and strongly correlated with RAS/MAPK pathway activity. Functionally, KSR1 overexpression selectively increased pMEK1/2 and pERK1/2 levels both in vitro and in vivo, underscoring its role as a positive regulator of RAS/MAPK signaling in HCC. Importantly, overexpression of KSR1, when cooperated with p53 inactivation or c-Myc overexpression, drives efficient hepatocarcinogenesis, achieving tumorigenic potency comparable to that induced by activating mutations in RAS or RAF.
Intriguingly, inhibition of KSR1—either by shRNA or by APS-2-79, a pharmacological inhibitor of KSR—significantly reduced RAS/MAPK pathway activity [90]. Treatment with APS-2-79 suppressed HCC cell proliferation in vitro and prolonged survival in HCC-bearing mice. Together, these findings demonstrate that KSR1 can drive RAS/MAPK pathway activation in the liver independently of upstream RAS or RAF mutations, highlighting KSR1 as a promising therapeutic target in HCC with aberrant activation of RAS/MAPK signaling (Figure 6).
The significance of the Moon et al. study lies in its pioneering demonstration that KSR1 inhibition represents a viable therapeutic strategy for HCC, thereby broadening the spectrum of actionable nodes within the RAS/MAPK signaling network. While most prior studies of KSR1 were performed in non-hepatic cancer models, this work establishes a critical proof-of-concept in liver cancer, underscoring KSR1′s role as a potential oncogenic driver. Looking forward, this discovery is expected to stimulate further research into KSR biology in HCC, including the development of next-generation KSR1 inhibitors with improved selectivity, bioavailability, and potency. Moreover, these findings position KSR1 as an emerging molecular vulnerability in hepatocellular carcinoma, representing a novel therapeutic target.

7.4. Clinical Studies Targeting Other Dysregulated Elements of the RAS/MAPK Pathway

Although clinical trials targeting KSR1 have yet to be initiated, clinical studies to target the RAS/MAPK signaling cascade at multiple levels in various cancers could inform the therapeutic effect of targeting KSR1 in human cancers, including HCC. Inhibiting farnesylation—essential for RAS membrane localization and activity—initially showed promise, but farnesyltransferase inhibitors like lonafarnib yielded disappointing results in clinical trials due to compensatory prenylation by geranylgeranyl transferase I [154,155]. Consequently, downstream components of the RAS/MAPK signaling pathway have emerged as more tractable targets. Selective BRAF inhibitors, including vemurafenib and dabrafenib, have significantly improved overall and progression-free survival in patients with melanoma [156,157,158].
Notably, MEK inhibitors have shown remarkable efficacy in certain cancers. For example, selumetinib, recently approved by the FDA, demonstrated clinical benefit in relapsed low-grade serous ovarian cancer and neurofibromatosis type 1 [159,160]. As well, trametinib and cobimetinib have shown meaningful improvements in progression-free survival and overall response rates in various types of cancers [161,162].
With recognition that ERK reactivation underlies resistance to RAF or MEK inhibitors, several clinical studies have been performed by targeting ERK directly. Selective ERK1/2 inhibitors such as BVD-523 are in clinical development for RAS- or RAF-mutant tumors (Figure 7). BVD-523 has shown potent anti-proliferative activity in colorectal and pancreatic cancers harboring BRAF or KRAS mutations [9,163]. Nevertheless, adaptive resistance remains a major obstacle to durable responses with RAS/MAPK-targeted therapies. This persistent challenge underscores the need for novel therapeutic approaches that go beyond single-agent inhibition. Future strategies may include rational combination therapies integrating KSR1 inhibition with RAF, MEK, or ERK inhibitors to enhance antitumor efficacy and overcome adaptive resistance in HCC and other RAS/MAPK-driven malignancies.

8. Perspectives and Conclusions

KSR1 has emerged as a pivotal scaffold protein within the RAS/MAPK signaling cascade, orchestrating the spatial and temporal assembly of pathway components to ensure efficient and sustained activation of MEK and ERK [43,44]. Beyond its canonical scaffolding function, KSR1 is increasingly recognized as a dynamic regulator of signaling intensity, duration, and subcellular localization. By modulating the proximity and phosphorylation state of RAF, MEK, and ERK, KSR1 fine-tunes the signaling output of the RAS/MAPK pathway in a highly context-dependent manner [43,44]. At physiological levels, KSR1 facilitates balanced MAPK signaling that supports controlled proliferation and survival. However, its overexpression or dysregulation can amplify signaling flux, thereby promoting oncogenic transformation.

8.1. Targeting KSR1 in HCC

From a therapeutic standpoint, we believe that targeting KSR1 represents a conceptually distinct and underexplored strategy for modulating oncogenic RAS/MAPK signaling. By selectively destabilizing or disrupting the scaffold architecture that coordinates RAS/MAPK signaling, it may be possible to attenuate oncogenic signaling in tumor cells while preserving the physiological signaling needed in normal tissues. Thus, pharmacological manipulation of KSR1 could allow fine-tuned pathway control. In HCC, a malignancy characterized by frequent RAS/MAPK pathway activation despite the rarity of RAS or RAF mutations, KSR1 appears to serve as a non-mutational driver of oncogenic signaling. Elevated KSR1 expression in HCC correlates with enhanced MEK and ERK phosphorylation and aggressive clinical behavior, suggesting that KSR1 contributes to the constitutive activation of RAS/MAPK signaling that underpins HCC progression. Functional studies have shown that KSR1 silencing in HCC, either through RNA interference or pharmacological blockade, attenuates RAS/MAPK signaling and inhibits tumor growth both in vitro and in vivo [43,44]. Notably, KSR1-deficient mice develop normally and remain viable, implying that KSR1 may be dispensable for normal tissue homeostasis yet essential for tumor maintenance [84]. This observation strengthens the case for KSR1 as a therapeutically tractable and cancer-selective vulnerability.
The development of small molecules such as APS-2-79, which allosterically stabilize KSR1 in an inactive conformation and prevent RAF–KSR–MEK complex formation, provides proof-of-principle that KSR1 is druggable [43,44]. However, these early agents show modest potency and limited selectivity, underscoring the need for more sophisticated approaches. In our view, the next frontier for KSR1 research lies in integrating structural biology, proteomics, and chemical innovation to design compounds that more precisely modulate its scaffolding function or promote its selective degradation.

8.2. Future Research Directions

Several key directions warrant attention in future research. First, high-resolution structural characterization of KSR1 in its active and inactive states will be essential for rational drug design. In particular, the dynamic transitions that occur during complex formation with RAF and MEK remain poorly understood. For example, hydrogen–deuterium exchange mass spectrometry could illuminate these conformational landscapes, revealing transient pockets or surfaces amenable to allosteric inhibition [164]. Second, the development of next-generation KSR1 inhibitors should expand beyond traditional small molecules. Emerging technologies such as proteolysis-targeting chimeras (PROTACs) or molecular glues offer the potential to achieve selective and complete degradation of KSR1 in cancer cells [99,100,101]. These approaches could circumvent the limitations of occupancy-driven inhibition by eliminating the protein entirely, providing more durable pathway suppression. Parallel optimization of pharmacokinetic and delivery properties—such as liver-specific nanoparticle formulations—will be crucial for achieving effective targeting in HCC. Finally, rational combination strategies with MEK inhibitors, tyrosine kinase inhibitors, or immunotherapies may also reveal synergistic effects capable of overcoming adaptive resistance to current treatments.

8.3. Combination Therapies as Possible Therapeutic Strategies

Given KSR1′s central role in organizing the RAF–MEK–ERK signaling module, targeting KSR1 alone may not fully suppress oncogenic signaling due to compensatory feedback loops and parallel pathway activation. Therefore, rational combination therapies that simultaneously inhibit KSR1 and other critical signaling nodes hold strong promise for achieving durable antitumor responses.

8.3.1. Combination with MEK or ERK Inhibitors

Pharmacological inhibition of KSR1 could sensitize tumors to downstream blockade of MEK or ERK. Because KSR1 facilitates MEK phosphorylation and stabilizes active MEK–ERK complexes, its disruption may enhance the efficacy of MEK/ERK inhibitors by weakening pathway reactivation and reducing the emergence of resistant clones. Preclinical evidence indicates that genetic suppression of KSR1 potentiates the cytotoxic effects of MEK inhibitors, suggesting a synergistic interaction [165]. This approach could be particularly relevant in HCC and RAS-driven tumors, where persistent ERK activity underlies adaptive drug resistance.

8.3.2. Combination with Receptor Tyrosine Kinase (RTK) Inhibitors

Upstream RTKs such as EGFR, FGFR, and c-Met are major activators of RAS signaling in HCC. RTK inhibitors (e.g., sorafenib, lenvatinib) often show transient efficacy due to compensatory RAS/MAPK reactivation [134,136,139]. Co-targeting KSR1 could suppress this rebound activation by disrupting scaffold-mediated pathway reassembly. Such vertical blockade of the RTK–RAS–KSR–MEK–ERK axis might enhance tumor control and delay resistance development. Moreover, inhibiting KSR1 could help overcome the escape mechanisms mediated by cross-talk between MAPK and PI3K/AKT signaling cascades.

8.3.3. Combination with Immunotherapies

Emerging evidence links MAPK pathway activation to an immunosuppressive tumor microenvironment, partly through upregulation of PD-L1 and suppression of cytotoxic T-cell infiltration [166,167]. Inhibiting KSR1 might attenuate these immunosuppressive signals by reducing ERK-driven transcriptional outputs, thereby enhancing the efficacy of immune checkpoint inhibitors such as anti-PD-1/PD-L1 antibodies. The immunomodulatory potential warrants systematic evaluation of KSR1 blockade in combination with immunotherapy, particularly in HCC, where immune evasion remains a major therapeutic challenge.
In summary, KSR1 constitutes an emerging molecular vulnerability in human cancers. Its scaffold function positions it as a master regulator of RAS/MAPK oncogenic signaling, offering novel opportunities for targeted intervention even in patients lacking canonical RAS or RAF mutations. Advances in structure-based drug design, proteolysis-targeting technologies, and rational combination strategies hold considerable promise for translating KSR1 inhibition into clinically viable therapies—potentially reshaping treatment paradigms for patients with limited options.

Author Contributions

Conceptualization, H.M. and S.W.R.; investigation, H.M., H.P., S.L. (Soyun Lee) and S.L. (Sangjik Lee); visualization, H.M. and H.P.; writing—original draft preparation, H.M., H.P., S.L. (Soyun Lee), S.L. (Sangjik Lee) and S.W.R.; writing—review and editing, H.M. and S.W.R.; supervision, S.W.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2021R1C1C2006182) (awarded to H.M.) and the National Research Foundation of Korea (NRF) grant 2019R1A2C2009518 (awarded to S.W.R.), which was funded by the Korea government (MSIP).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhang, S.; Xiao, X.; Yi, Y.; Wang, X.; Zhu, L.; Shen, Y.; Lin, D.; Wu, C. Tumor initiation and early tumorigenesis: Molecular mechanisms and interventional targets. Signal Transduct. Target. Ther. 2024, 9, 149. [Google Scholar] [CrossRef]
  2. Cheng, Y.; He, C.; Wang, M.; Ma, X.; Mo, F.; Yang, S.; Han, J.; Wei, X. Targeting epigenetic regulators for cancer therapy: Mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 2019, 4, 62. [Google Scholar] [CrossRef]
  3. Herceg, Z.; Hainaut, P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Mol. Oncol. 2007, 1, 26–41. [Google Scholar] [CrossRef] [PubMed]
  4. Bahar, M.E.; Kim, H.J.; Kim, D.R. Targeting the RAS/RAF/MAPK pathway for cancer therapy: From mechanism to clinical studies. Signal Transduct. Target. Ther. 2023, 8, 455. [Google Scholar] [CrossRef]
  5. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef] [PubMed]
  6. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
  7. Varghese, A.M.; Perry, M.A.; Chou, J.F.; Nandakumar, S.; Muldoon, D.; Erakky, A.; Zucker, A.; Fong, C.; Mehine, M.; Nguyen, B.; et al. Clinicogenomic landscape of pancreatic adenocarcinoma identifies KRAS mutant dosage as prognostic of overall survival. Nat. Med. 2025, 31, 466–477. [Google Scholar] [CrossRef] [PubMed]
  8. Li, M.; Li, Q.; Zou, C.; Huang, Q.; Chen, Y. Application and recent advances in conventional biomarkers for the prognosis of papillary thyroid carcinoma. Front. Oncol. 2025, 15, 1598934. [Google Scholar] [CrossRef]
  9. Liu, F.; Yang, X.; Geng, M.; Huang, M. Targeting ERK, an Achilles’ Heel of the MAPK pathway, in cancer therapy. Acta Pharm. Sin. B 2018, 8, 552–562. [Google Scholar] [CrossRef]
  10. Drosten, M.; Barbacid, M. Targeting the MAPK Pathway in KRAS-Driven Tumors. Cancer Cell 2020, 37, 543–550. [Google Scholar] [CrossRef]
  11. Anaya, Y.A.; Bracho, R.P.; Chauhan, S.C.; Tripathi, M.K.; Bandyopadhyay, D. Small Molecule B-RAF Inhibitors as Anti-Cancer Therapeutics: Advances in Discovery, Development, and Mechanistic Insights. Int. J. Mol. Sci. 2025, 26, 2676. [Google Scholar] [CrossRef]
  12. Kubatka, P.; Bojkova, B.; Nosalova, N.; Huniadi, M.; Samuel, S.M.; Sreenesh, B.; Hrklova, G.; Kajo, K.; Hornak, S.; Cizkova, D.; et al. Targeting the MAPK signaling pathway: Implications and prospects of flavonoids in 3P medicine as modulators of cancer cell plasticity and therapeutic resistance in breast cancer patients. Epma J. 2025, 16, 437–463. [Google Scholar] [CrossRef] [PubMed]
  13. Ito, Y.; Sasaki, Y.; Horimoto, M.; Wada, S.; Tanaka, Y.; Kasahara, A.; Ueki, T.; Hirano, T.; Yamamoto, H.; Fujimoto, J.; et al. Activation of mitogen-activated protein kinases/extracellular signal-regulated kinases in human hepatocellular carcinoma. Hepatology 1998, 27, 951–958. [Google Scholar] [CrossRef]
  14. Delire, B.; Stärkel, P. The Ras/MAPK pathway and hepatocarcinoma: Pathogenesis and therapeutic implications. Eur. J. Clin. Investig. 2015, 45, 609–623. [Google Scholar] [CrossRef]
  15. Chardin, P.; Camonis, J.H.; Gale, N.W.; van Aelst, L.; Schlessinger, J.; Wigler, M.H.; Bar-Sagi, D. Human Sos1: A guanine nucleotide exchange factor for Ras that binds to GRB2. Science 1993, 260, 1338–1343. [Google Scholar] [CrossRef] [PubMed]
  16. Seiler, C.; Stainthorp, A.K.; Ketchen, S.; Jones, C.M.; Marks, K.; Quirke, P.; Ladbury, J.E. The Grb2 splice variant, Grb3-3, is a negative regulator of RAS activation. Commun. Biol. 2022, 5, 1029. [Google Scholar] [CrossRef]
  17. Dimri, M.; Satyanarayana, A. Molecular Signaling Pathways and Therapeutic Targets in Hepatocellular Carcinoma. Cancers 2020, 12, 491. [Google Scholar] [CrossRef] [PubMed]
  18. Cseh, B.; Doma, E.; Baccarini, M. “RAF” neighborhood: Protein-protein interaction in the Raf/Mek/Erk pathway. FEBS Lett. 2014, 588, 2398–2406. [Google Scholar] [CrossRef]
  19. Dhillon, A.S.; Hagan, S.; Rath, O.; Kolch, W. MAP kinase signalling pathways in cancer. Oncogene 2007, 26, 3279–3290. [Google Scholar] [CrossRef]
  20. Maik-Rachline, G.; Hacohen-Lev-Ran, A.; Seger, R. Nuclear ERK: Mechanism of Translocation, Substrates, and Role in Cancer. Int. J. Mol. Sci. 2019, 20, 1194. [Google Scholar] [CrossRef]
  21. Pudewell, S.; Wittich, C.; Kazemein Jasemi, N.S.; Bazgir, F.; Ahmadian, M.R. Accessory proteins of the RAS-MAPK pathway: Moving from the side line to the front line. Commun. Biol. 2021, 4, 696. [Google Scholar] [CrossRef]
  22. Martin-Vega, A.; Cobb, M.H. Navigating the ERK1/2 MAPK Cascade. Biomolecules 2023, 13, 1555. [Google Scholar] [CrossRef]
  23. Fischer, A.; Mühlhäuser, W.W.D.; Warscheid, B.; Radziwill, G. Membrane localization of acetylated CNK1 mediates a positive feedback on RAF/ERK signaling. Sci. Adv. 2017, 3, e1700475. [Google Scholar] [CrossRef]
  24. Zhan, Z.; Ye, M.; Jin, X. The roles of FLOT1 in human diseases (Review). Mol. Med. Rep. 2023, 28, 212. [Google Scholar] [CrossRef] [PubMed]
  25. Saldaña-Villa, A.K.; Lara-Lemus, R. The Structural Proteins of Membrane Rafts, Caveolins and Flotillins, in Lung Cancer: More Than Just Scaffold Elements. Int. J. Med. Sci. 2023, 20, 1662–1670. [Google Scholar] [CrossRef] [PubMed]
  26. Thomé, C.H.; Ferreira, G.A.; Pereira-Martins, D.A.; Dos Santos, G.A.; Ortiz, C.A.; de Souza, L.E.B.; Sobral, L.M.; Silva, C.L.A.; Scheucher, P.S.; Gil, C.D.; et al. NTAL is associated with treatment outcome, cell proliferation and differentiation in acute promyelocytic leukemia. Sci. Rep. 2020, 10, 10315. [Google Scholar] [CrossRef] [PubMed]
  27. Jin, T.; Ding, Q.; Huang, H.; Xu, D.; Jiang, Y.; Zhou, B.; Li, Z.; Jiang, X.; He, J.; Liu, W.; et al. PAQR10 and PAQR11 mediate Ras signaling in the Golgi apparatus. Cell Res. 2012, 22, 661–676. [Google Scholar] [CrossRef]
  28. Brown, M.D.; Sacks, D.B. Protein scaffolds in MAP kinase signalling. Cell. Signal. 2009, 21, 462–469. [Google Scholar] [CrossRef]
  29. Philips, M.R. Sef: A MEK/ERK catcher on the Golgi. Mol. Cell 2004, 15, 168–169. [Google Scholar] [CrossRef]
  30. Skinner, M. A new role for Dok. Nat. Rev. Cancer 2010, 10, 239. [Google Scholar] [CrossRef]
  31. Saltiel, A.R. Insulin signaling in health and disease. J. Clin. Investig. 2021, 131, e142241. [Google Scholar] [CrossRef]
  32. Costa-Neto, C.M.; Parreiras, E.S.L.T. Deciphering complexity of GPCR signaling and modulation: Implications and perspectives for drug discovery. Clin. Sci. 2025, 139, 463–477. [Google Scholar] [CrossRef]
  33. Kahsai, A.W.; Shah, K.S.; Shim, P.J.; Lee, M.A.; Shreiber, B.N.; Schwalb, A.M.; Zhang, X.; Kwon, H.Y.; Huang, L.Y.; Soderblom, E.J.; et al. Signal transduction at GPCRs: Allosteric activation of the ERK MAPK by β-arrestin. Proc. Natl. Acad. Sci. USA 2023, 120, e2303794120. [Google Scholar] [CrossRef]
  34. Xie, Y.; Su, N.; Yang, J.; Tan, Q.; Huang, S.; Jin, M.; Ni, Z.; Zhang, B.; Zhang, D.; Luo, F.; et al. FGF/FGFR signaling in health and disease. Signal Transduct. Target. Ther. 2020, 5, 181. [Google Scholar] [CrossRef]
  35. Pawson, T. Dynamic control of signaling by modular adaptor proteins. Curr. Opin. Cell Biol. 2007, 19, 112–116. [Google Scholar] [CrossRef]
  36. Shigeno-Nakazawa, Y.; Kasai, T.; Ki, S.; Kostyanovskaya, E.; Pawlak, J.; Yamagishi, J.; Okimoto, N.; Taiji, M.; Okada, M.; Westbrook, J.; et al. A pre-metazoan origin of the CRK gene family and co-opted signaling network. Sci. Rep. 2016, 6, 34349. [Google Scholar] [CrossRef] [PubMed]
  37. Malagrinò, F.; Puglisi, E.; Pagano, L.; Travaglini-Allocatelli, C.; Toto, A. GRB2: A dynamic adaptor protein orchestrating cellular signaling in health and disease. Biochem. Biophys. Rep. 2024, 39, 101803. [Google Scholar] [CrossRef]
  38. Kazemein Jasemi, N.S.; Herrmann, C.; Magdalena Estirado, E.; Gremer, L.; Willbold, D.; Brunsveld, L.; Dvorsky, R.; Ahmadian, M.R. The intramolecular allostery of GRB2 governing its interaction with SOS1 is modulated by phosphotyrosine ligands. Biochem. J. 2021, 478, 2793–2809. [Google Scholar] [CrossRef] [PubMed]
  39. Kazemein Jasemi, N.S.; Ahmadian, M.R. Allosteric regulation of GRB2 modulates RAS activation. Small GTPases 2022, 13, 282–286. [Google Scholar] [CrossRef]
  40. Ahmed, S.B.M.; Prigent, S.A. Insights into the Shc Family of Adaptor Proteins. J. Mol. Signal 2017, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  41. Chen, H.; Libring, S.; Ruddraraju, K.V.; Miao, J.; Solorio, L.; Zhang, Z.Y.; Wendt, M.K. SHP2 is a multifunctional therapeutic target in drug resistant metastatic breast cancer. Oncogene 2020, 39, 7166–7180. [Google Scholar] [CrossRef]
  42. Schaller, M.D. Paxillin: A focal adhesion-associated adaptor protein. Oncogene 2001, 20, 6459–6472. [Google Scholar] [CrossRef]
  43. Roy, F.; Laberge, G.; Douziech, M.; Ferland-McCollough, D.; Therrien, M. KSR is a scaffold required for activation of the ERK/MAPK module. Genes Dev. 2002, 16, 427–438. [Google Scholar] [CrossRef]
  44. Frodyma, D.; Neilsen, B.; Costanzo-Garvey, D.; Fisher, K.; Lewis, R. Coordinating ERK signaling via the molecular scaffold Kinase Suppressor of Ras. F1000Research 2017, 6, 1621. [Google Scholar] [CrossRef]
  45. Ory, S.; Zhou, M.; Conrads, T.P.; Veenstra, T.D.; Morrison, D.K. Protein phosphatase 2A positively regulates Ras signaling by dephosphorylating KSR1 and Raf-1 on critical 14-3-3 binding sites. Curr. Biol. 2003, 13, 1356–1364. [Google Scholar] [CrossRef]
  46. Thines, L.; Roushar, F.J.; Hedman, A.C.; Sacks, D.B. The IQGAP scaffolds: Critical nodes bridging receptor activation to cellular signaling. J. Cell Biol. 2023, 222, e202205062. [Google Scholar] [CrossRef]
  47. Jang, H.; Stevens, P.; Gao, T.; Galperin, E. The leucine-rich repeat signaling scaffolds Shoc2 and Erbin: Cellular mechanism and role in disease. Febs J. 2021, 288, 721–739. [Google Scholar] [CrossRef] [PubMed]
  48. You, F.; Sun, H.; Zhou, X.; Sun, W.; Liang, S.; Zhai, Z.; Jiang, Z. PCBP2 mediates degradation of the adaptor MAVS via the HECT ubiquitin ligase AIP4. Nat. Immunol. 2009, 10, 1300–1308. [Google Scholar] [CrossRef] [PubMed]
  49. Lorenzo, C.; McCormick, F. SPRED proteins and their roles in signal transduction, development, and malignancy. Genes Dev. 2020, 34, 1410–1421. [Google Scholar] [CrossRef] [PubMed]
  50. Kawazoe, T.; Taniguchi, K. The Sprouty/Spred family as tumor suppressors: Coming of age. Cancer Sci. 2019, 110, 1525–1535. [Google Scholar] [CrossRef]
  51. Puranik, N.; Jung, H.; Song, M. SPROUTY2, a Negative Feedback Regulator of Receptor Tyrosine Kinase Signaling, Associated with Neurodevelopmental Disorders: Current Knowledge and Future Perspectives. Int. J. Mol. Sci. 2024, 25, 11043. [Google Scholar] [CrossRef] [PubMed]
  52. Touboul, R.; Baritaki, S.; Zaravinos, A.; Bonavida, B. RKIP Pleiotropic Activities in Cancer and Inflammatory Diseases: Role in Immunity. Cancers 2021, 13, 6247. [Google Scholar] [CrossRef] [PubMed]
  53. Lorenz, K.; Lohse, M.J.; Quitterer, U. Protein kinase C switches the Raf kinase inhibitor from Raf-1 to GRK-2. Nature 2003, 426, 574–579. [Google Scholar] [CrossRef]
  54. Kumari, S.; Peela, S.; Bramhachari, P.V.; Mallikarjuna, K.; Nagaraju, G.P.; Srilatha, M. Raf-kinase inhibitor protein (RKIP): A therapeutic target in colon cancer. Biochim. Biophys. Acta Rev. Cancer 2025, 1880, 189387. [Google Scholar] [CrossRef]
  55. Lai, T.H.; Ahmed, M.; Hwang, J.S.; Bahar, M.E.; Pham, T.M.; Yang, J.; Kim, W.; Maulidi, R.F.; Lee, D.K.; Kim, D.H.; et al. Manipulating RKIP reverses the metastatic potential of breast cancer cells. Front. Oncol. 2023, 13, 1189350. [Google Scholar] [CrossRef] [PubMed]
  56. Cessna, H.; Baritaki, S.; Zaravinos, A.; Bonavida, B. The Role of RKIP in the Regulation of EMT in the Tumor Microenvironment. Cancers 2022, 14, 4596. [Google Scholar] [CrossRef]
  57. Figy, C.; Guo, A.; Fernando, V.R.; Furuta, S.; Al-Mulla, F.; Yeung, K.C. Changes in Expression of Tumor Suppressor Gene RKIP Impact How Cancers Interact with Their Complex Environment. Cancers 2023, 15, 958. [Google Scholar] [CrossRef]
  58. Huang, Y.Z.; Zang, M.; Xiong, W.C.; Luo, Z.; Mei, L. Erbin suppresses the MAP kinase pathway. J. Biol. Chem. 2003, 278, 1108–1114. [Google Scholar] [CrossRef]
  59. Khoubai, F.Z.; Grosset, C.F. DUSP9, a Dual-Specificity Phosphatase with a Key Role in Cell Biology and Human Diseases. Int. J. Mol. Sci. 2021, 22, 11538. [Google Scholar] [CrossRef]
  60. Chen, H.F.; Chuang, H.C.; Tan, T.H. Regulation of Dual-Specificity Phosphatase (DUSP) Ubiquitination and Protein Stability. Int. J. Mol. Sci. 2019, 20, 2668. [Google Scholar] [CrossRef]
  61. Ahmad, M.K.; Abdollah, N.A.; Shafie, N.H.; Yusof, N.M.; Razak, S.R.A. Dual-specificity phosphatase 6 (DUSP6): A review of its molecular characteristics and clinical relevance in cancer. Cancer Biol. Med. 2018, 15, 14–28. [Google Scholar] [CrossRef]
  62. Bai, H.; Song, M.; Jiao, R.; Li, W.; Zhao, J.; Xiao, M.; Jin, M.; Zhang, Z.; Deng, H. DUSP7 inhibits cervical cancer progression by inactivating the RAS pathway. J. Cell. Mol. Med. 2021, 25, 9306–9318. [Google Scholar] [CrossRef] [PubMed]
  63. Melillo, R.M.; Santoro, M.; Ong, S.H.; Billaud, M.; Fusco, A.; Hadari, Y.R.; Schlessinger, J.; Lax, I. Docking protein FRS2 links the protein tyrosine kinase RET and its oncogenic forms with the mitogen-activated protein kinase signaling cascade. Mol. Cell Biol. 2001, 21, 4177–4187. [Google Scholar] [CrossRef] [PubMed]
  64. Kornfeld, K.; Hom, D.B.; Horvitz, H.R. The ksr-1 gene encodes a novel protein kinase involved in Ras-mediated signaling in C. elegans. Cell 1995, 83, 903–913. [Google Scholar] [CrossRef]
  65. Sundaram, M.; Han, M. The C. elegans ksr-1 gene encodes a novel Raf-related kinase involved in Ras-mediated signal transduction. Cell 1995, 83, 889–901. [Google Scholar] [CrossRef]
  66. Young, L.C.; Hartig, N.; Boned Del Río, I.; Sari, S.; Ringham-Terry, B.; Wainwright, J.R.; Jones, G.G.; McCormick, F.; Rodriguez-Viciana, P. SHOC2-MRAS-PP1 complex positively regulates RAF activity and contributes to Noonan syndrome pathogenesis. Proc. Natl. Acad. Sci. USA 2018, 115, E10576–E10585. [Google Scholar] [CrossRef]
  67. Hauseman, Z.J.; Fodor, M.; Dhembi, A.; Viscomi, J.; Egli, D.; Bleu, M.; Katz, S.; Park, E.; Jang, D.M.; Porter, K.A.; et al. Structure of the MRAS-SHOC2-PP1C phosphatase complex. Nature 2022, 609, 416–423. [Google Scholar] [CrossRef] [PubMed]
  68. Boned Del Río, I.; Young, L.C.; Sari, S.; Jones, G.G.; Ringham-Terry, B.; Hartig, N.; Rejnowicz, E.; Lei, W.; Bhamra, A.; Surinova, S.; et al. SHOC2 complex-driven RAF dimerization selectively contributes to ERK pathway dynamics. Proc. Natl. Acad. Sci. USA 2019, 116, 13330–13339. [Google Scholar] [CrossRef]
  69. Neilsen, B.K.; Frodyma, D.E.; Lewis, R.E.; Fisher, K.W. KSR as a therapeutic target for Ras-dependent cancers. Expert. Opin. Ther. Targets 2017, 21, 499–509. [Google Scholar] [CrossRef]
  70. Koveal, D.; Schuh-Nuhfer, N.; Ritt, D.; Page, R.; Morrison, D.K.; Peti, W. A CC-SAM, for coiled coil-sterile α motif, domain targets the scaffold KSR-1 to specific sites in the plasma membrane. Sci. Signal. 2012, 5, ra94. [Google Scholar] [CrossRef]
  71. Kolch, W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat. Rev. Mol. Cell Biol. 2005, 6, 827–837. [Google Scholar] [CrossRef]
  72. Costanzo-Garvey, D.L.; Pfluger, P.T.; Dougherty, M.K.; Stock, J.L.; Boehm, M.; Chaika, O.; Fernandez, M.R.; Fisher, K.; Kortum, R.L.; Hong, E.G.; et al. KSR2 is an essential regulator of AMP kinase, energy expenditure, and insulin sensitivity. Cell Metab. 2009, 10, 366–378. [Google Scholar] [CrossRef]
  73. Zhou, M.; Horita, D.A.; Waugh, D.S.; Byrd, R.A.; Morrison, D.K. Solution structure and functional analysis of the cysteine-rich C1 domain of kinase suppressor of Ras (KSR). J. Mol. Biol. 2002, 315, 435–446. [Google Scholar] [CrossRef]
  74. Cacace, A.M.; Michaud, N.R.; Therrien, M.; Mathes, K.; Copeland, T.; Rubin, G.M.; Morrison, D.K. Identification of constitutive and ras-inducible phosphorylation sites of KSR: Implications for 14-3-3 binding, mitogen-activated protein kinase binding, and KSR overexpression. Mol. Cell. Biol. 1999, 19, 229–240. [Google Scholar] [CrossRef] [PubMed]
  75. Lavoie, H.; Sahmi, M.; Maisonneuve, P.; Marullo, S.A.; Thevakumaran, N.; Jin, T.; Kurinov, I.; Sicheri, F.; Therrien, M. MEK drives BRAF activation through allosteric control of KSR proteins. Nature 2018, 554, 549–553. [Google Scholar] [CrossRef]
  76. Kortum, R.L.; Lewis, R.E. The molecular scaffold KSR1 regulates the proliferative and oncogenic potential of cells. Mol. Cell. Biol. 2004, 24, 4407–4416. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, H.; Koo, C.Y.; Stebbing, J.; Giamas, G. The dual function of KSR1: A pseudokinase and beyond. Biochem. Soc. Trans. 2013, 41, 1078–1082. [Google Scholar] [CrossRef]
  78. Razidlo, G.L.; Kortum, R.L.; Haferbier, J.L.; Lewis, R.E. Phosphorylation regulates KSR1 stability, ERK activation, and cell proliferation. J. Biol. Chem. 2004, 279, 47808–47814. [Google Scholar] [CrossRef] [PubMed]
  79. Paniagua, G.; Jacob, H.K.C.; Brehey, O.; García-Alonso, S.; Lechuga, C.G.; Pons, T.; Musteanu, M.; Guerra, C.; Drosten, M.; Barbacid, M. KSR induces RAS-independent MAPK pathway activation and modulates the efficacy of KRAS inhibitors. Mol. Oncol. 2022, 16, 3066–3081. [Google Scholar] [CrossRef]
  80. Fisher, K.W.; Das, B.; Kim, H.S.; Clymer, B.K.; Gehring, D.; Smith, D.R.; Costanzo-Garvey, D.L.; Fernandez, M.R.; Brattain, M.G.; Kelly, D.L.; et al. AMPK Promotes Aberrant PGC1β Expression to Support Human Colon Tumor Cell Survival. Mol. Cell. Biol. 2015, 35, 3866–3879. [Google Scholar] [CrossRef]
  81. Germino, E.A.; Miller, J.P.; Diehl, L.; Swanson, C.J.; Durinck, S.; Modrusan, Z.; Miner, J.H.; Shaw, A.S. Homozygous KSR1 deletion attenuates morbidity but does not prevent tumor development in a mouse model of RAS-driven pancreatic cancer. PLoS ONE 2018, 13, e0194998. [Google Scholar] [CrossRef]
  82. Gramling, M.W.; Eischen, C.M. Suppression of Ras/Mapk pathway signaling inhibits Myc-induced lymphomagenesis. Cell Death Differ. 2012, 19, 1220–1227. [Google Scholar] [CrossRef]
  83. Liu, Z.; Krstic, A.; Neve, A.; Casalou, C.; Rauch, N.; Wynne, K.; Cassidy, H.; McCann, A.; Kavanagh, E.; McCann, B.; et al. Kinase Suppressor of RAS 1 (KSR1) Maintains the Transformed Phenotype of BRAFV600E Mutant Human Melanoma Cells. Int. J. Mol. Sci. 2023, 24, 11821. [Google Scholar] [CrossRef] [PubMed]
  84. Lozano, J.; Xing, R.; Cai, Z.; Jensen, H.L.; Trempus, C.; Mark, W.; Cannon, R.; Kolesnick, R. Deficiency of kinase suppressor of Ras1 prevents oncogenic ras signaling in mice. Cancer Res. 2003, 63, 4232–4238. [Google Scholar]
  85. Nguyen, A.; Burack, W.R.; Stock, J.L.; Kortum, R.; Chaika, O.V.; Afkarian, M.; Muller, W.J.; Murphy, K.M.; Morrison, D.K.; Lewis, R.E.; et al. Kinase suppressor of Ras (KSR) is a scaffold which facilitates mitogen-activated protein kinase activation in vivo. Mol. Cell. Biol. 2002, 22, 3035–3045. [Google Scholar] [CrossRef]
  86. Denouel-Galy, A.; Douville, E.M.; Warne, P.H.; Papin, C.; Laugier, D.; Calothy, G.; Downward, J.; Eychène, A. Murine Ksr interacts with MEK and inhibits Ras-induced transformation. Curr. Biol. 1998, 8, 46–55. [Google Scholar] [CrossRef] [PubMed]
  87. Joneson, T.; Fulton, J.A.; Volle, D.J.; Chaika, O.V.; Bar-Sagi, D.; Lewis, R.E. Kinase suppressor of Ras inhibits the activation of extracellular ligand-regulated (ERK) mitogen-activated protein (MAP) kinase by growth factors, activated Ras, and Ras effectors. J. Biol. Chem. 1998, 273, 7743–7748. [Google Scholar] [CrossRef]
  88. Therrien, M.; Michaud, N.R.; Rubin, G.M.; Morrison, D.K. KSR modulates signal propagation within the MAPK cascade. Genes Dev. 1996, 10, 2684–2695. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, H.; Angelopoulos, N.; Xu, Y.; Grothey, A.; Nunes, J.; Stebbing, J.; Giamas, G. Proteomic profile of KSR1-regulated signalling in response to genotoxic agents in breast cancer. Breast Cancer Res. Treat. 2015, 151, 555–568. [Google Scholar] [CrossRef]
  90. Moon, H.; Park, H.; Lee, S.; Lee, S.; Jun, M.; Lee, J.; Baek, J.; Park, H.; Bang, J.; Ro, S.W. Kinase Suppressor of Ras 1 (KSR1) Promotes Liver Carcinogenesis via Activation of the RAS/RAF/MEK/ERK Signaling Pathway. JHEP Rep 2025, 7, 101536. [Google Scholar] [CrossRef]
  91. Drizyte-Miller, K.; Talabi, T.; Somasundaram, A.; Cox, A.D.; Der, C.J. KRAS: The Achilles’ heel of pancreas cancer biology. J. Clin. Investig. 2025, 135, e191939. [Google Scholar] [CrossRef]
  92. Zhang, Z.; Zhang, H.; Liao, X.; Tsai, H.I. KRAS mutation: The booster of pancreatic ductal adenocarcinoma transformation and progression. Front. Cell Dev. Biol. 2023, 11, 1147676. [Google Scholar] [CrossRef] [PubMed]
  93. Halbrook, C.J.; Lyssiotis, C.A.; Pasca di Magliano, M.; Maitra, A. Pancreatic cancer: Advances and challenges. Cell 2023, 186, 1729–1754. [Google Scholar] [CrossRef]
  94. Leonardi, G.C.; Candido, S.; Carbone, M.; Raiti, F.; Colaianni, V.; Garozzo, S.; Cinà, D.; McCubrey, J.A.; Libra, M. BRAF mutations in papillary thyroid carcinoma and emerging targeted therapies (review). Mol. Med. Rep. 2012, 6, 687–694. [Google Scholar] [CrossRef] [PubMed]
  95. Leung, G.P.; Feng, T.; Sigoillot, F.D.; Geyer, F.C.; Shirley, M.D.; Ruddy, D.A.; Rakiec, D.P.; Freeman, A.K.; Engelman, J.A.; Jaskelioff, M.; et al. Hyperactivation of MAPK Signaling Is Deleterious to RAS/RAF-mutant Melanoma. Mol. Cancer Res. 2019, 17, 199–211. [Google Scholar] [CrossRef] [PubMed]
  96. Dias, M.H.; Friskes, A.; Wang, S.; Fernandes Neto, J.M.; van Gemert, F.; Mourragui, S.; Papagianni, C.; Kuiken, H.J.; Mainardi, S.; Alvarez-Villanueva, D.; et al. Paradoxical Activation of Oncogenic Signaling as a Cancer Treatment Strategy. Cancer Discov. 2024, 14, 1276–1301. [Google Scholar] [CrossRef]
  97. Llobet, D.; Eritja, N.; Domingo, M.; Bergada, L.; Mirantes, C.; Santacana, M.; Pallares, J.; Macià, A.; Yeramian, A.; Encinas, M.; et al. KSR1 is overexpressed in endometrial carcinoma and regulates proliferation and TRAIL-induced apoptosis by modulating FLIP levels. Am. J. Pathol. 2011, 178, 1529–1543. [Google Scholar] [CrossRef]
  98. Dhawan, N.S.; Scopton, A.P.; Dar, A.C. Small molecule stabilization of the KSR inactive state antagonizes oncogenic Ras signalling. Nature 2016, 537, 112–116. [Google Scholar] [CrossRef]
  99. Tan, X.; Huang, Z.; Pei, H.; Jia, Z.; Zheng, J. Molecular glue-mediated targeted protein degradation: A novel strategy in small-molecule drug development. iScience 2024, 27, 110712. [Google Scholar] [CrossRef]
  100. Alabi, S.B.; Crews, C.M. Major advances in targeted protein degradation: PROTACs, LYTACs, and MADTACs. J. Biol. Chem. 2021, 296, 100647. [Google Scholar] [CrossRef]
  101. Zhao, L.; Zhao, J.; Zhong, K.; Tong, A.; Jia, D. Targeted protein degradation: Mechanisms, strategies and application. Signal Transduct. Target. Ther. 2022, 7, 113. [Google Scholar] [CrossRef] [PubMed]
  102. Liang, X.H.; Sun, H.; Nichols, J.G.; Crooke, S.T. RNase H1-Dependent Antisense Oligonucleotides Are Robustly Active in Directing RNA Cleavage in Both the Cytoplasm and the Nucleus. Mol. Ther. 2017, 25, 2075–2092. [Google Scholar] [CrossRef]
  103. Kole, R.; Krainer, A.R.; Altman, S. RNA therapeutics: Beyond RNA interference and antisense oligonucleotides. Nat. Rev. Drug Discov. 2012, 11, 125–140. [Google Scholar] [CrossRef] [PubMed]
  104. Gabbia, D.; De Martin, S. Insights into Hepatocellular Carcinoma: From Pathophysiology to Novel Therapies. Int. J. Mol. Sci. 2024, 25, 4188. [Google Scholar] [CrossRef] [PubMed]
  105. Chakraborty, E.; Sarkar, D. Emerging Therapies for Hepatocellular Carcinoma (HCC). Cancers 2022, 14, 2798. [Google Scholar] [CrossRef]
  106. Nevola, R.; Tortorella, G.; Rosato, V.; Rinaldi, L.; Imbriani, S.; Perillo, P.; Mastrocinque, D.; La Montagna, M.; Russo, A.; Di Lorenzo, G.; et al. Gender Differences in the Pathogenesis and Risk Factors of Hepatocellular Carcinoma. Biology 2023, 12, 984. [Google Scholar] [CrossRef]
  107. Konyn, P.; Ahmed, A.; Kim, D. Current epidemiology in hepatocellular carcinoma. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 1295–1307. [Google Scholar] [CrossRef]
  108. Lampimukhi, M.; Qassim, T.; Venu, R.; Pakhala, N.; Mylavarapu, S.; Perera, T.; Sathar, B.S.; Nair, A. A Review of Incidence and Related Risk Factors in the Development of Hepatocellular Carcinoma. Cureus 2023, 15, e49429. [Google Scholar] [CrossRef]
  109. Dillon, M.; Lopez, A.; Lin, E.; Sales, D.; Perets, R.; Jain, P. Progress on Ras/MAPK Signaling Research and Targeting in Blood and Solid Cancers. Cancers 2021, 13, 5059. [Google Scholar] [CrossRef]
  110. Moon, H.; Ro, S.W. MAPK/ERK Signaling Pathway in Hepatocellular Carcinoma. Cancers 2021, 13, 3026. [Google Scholar] [CrossRef]
  111. Athuluri-Divakar, S.K.; Vasquez-Del Carpio, R.; Dutta, K.; Baker, S.J.; Cosenza, S.C.; Basu, I.; Gupta, Y.K.; Reddy, M.V.; Ueno, L.; Hart, J.R.; et al. A Small Molecule RAS-Mimetic Disrupts RAS Association with Effector Proteins to Block Signaling. Cell 2016, 165, 643–655. [Google Scholar] [CrossRef] [PubMed]
  112. Dietrich, P.; Freese, K.; Mahli, A.; Thasler, W.E.; Hellerbrand, C.; Bosserhoff, A.K. Combined effects of PLK1 and RAS in hepatocellular carcinoma reveal rigosertib as promising novel therapeutic “dual-hit” option. Oncotarget 2018, 9, 3605–3618. [Google Scholar] [CrossRef]
  113. Ghousein, A.; Mosca, N.; Cartier, F.; Charpentier, J.; Dupuy, J.W.; Raymond, A.A.; Bioulac-Sage, P.; Grosset, C.F. miR-4510 blocks hepatocellular carcinoma development through RAF1 targeting and RAS/RAF/MEK/ERK signalling inactivation. Liver Int. 2020, 40, 240–251. [Google Scholar] [CrossRef]
  114. Tong, X.; Wang, Q.; Wu, D.; Bao, L.; Yin, T.; Chen, H. MEK inhibition by cobimetinib suppresses hepatocellular carcinoma and angiogenesis in vitro and in vivo. Biochem. Biophys. Res. Commun. 2020, 523, 147–152. [Google Scholar] [CrossRef]
  115. Bessard, A.; Frémin, C.; Ezan, F.; Fautrel, A.; Gailhouste, L.; Baffet, G. RNAi-mediated ERK2 knockdown inhibits growth of tumor cells in vitro and in vivo. Oncogene 2008, 27, 5315–5325. [Google Scholar] [CrossRef]
  116. Rozakis-Adcock, M.; Fernley, R.; Wade, J.; Pawson, T.; Bowtell, D. The SH2 and SH3 domains of mammalian Grb2 couple the EGF receptor to the Ras activator mSos1. Nature 1993, 363, 83–85. [Google Scholar] [CrossRef]
  117. Zhou, J.; Tu, D.; Peng, R.; Tang, Y.; Deng, Q.; Su, B.; Wang, S.; Tang, H.; Jin, S.; Jiang, G.; et al. RNF173 suppresses RAF/MEK/ERK signaling to regulate invasion and metastasis via GRB2 ubiquitination in Hepatocellular Carcinoma. Cell Commun. Signal. 2023, 21, 224. [Google Scholar] [CrossRef]
  118. Hadari, Y.R.; Kouhara, H.; Lax, I.; Schlessinger, J. Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation. Mol. Cell. Biol. 1998, 18, 3966–3973. [Google Scholar] [CrossRef]
  119. Cunnick, J.M.; Meng, S.; Ren, Y.; Desponts, C.; Wang, H.G.; Djeu, J.Y.; Wu, J. Regulation of the mitogen-activated protein kinase signaling pathway by SHP2. J. Biol. Chem. 2002, 277, 9498–9504. [Google Scholar] [CrossRef] [PubMed]
  120. Deng, R.; Zhao, X.; Qu, Y.; Chen, C.; Zhu, C.; Zhang, H.; Yuan, H.; Jin, H.; Liu, X.; Wang, Y.; et al. Shp2 SUMOylation promotes ERK activation and hepatocellular carcinoma development. Oncotarget 2015, 6, 9355–9369. [Google Scholar] [CrossRef] [PubMed]
  121. Yoshida, T.; Hisamoto, T.; Akiba, J.; Koga, H.; Nakamura, K.; Tokunaga, Y.; Hanada, S.; Kumemura, H.; Maeyama, M.; Harada, M.; et al. Spreds, inhibitors of the Ras/ERK signal transduction, are dysregulated in human hepatocellular carcinoma and linked to the malignant phenotype of tumors. Oncogene 2006, 25, 6056–6066. [Google Scholar] [CrossRef]
  122. Tan, W.; Lin, Z.; Chen, X.; Li, W.; Zhu, S.; Wei, Y.; Huo, L.; Chen, Y.; Shang, C. miR-126-3p contributes to sorafenib resistance in hepatocellular carcinoma via downregulating SPRED1. Ann. Transl. Med. 2021, 9, 38. [Google Scholar] [CrossRef] [PubMed]
  123. Ma, X.N.; Liu, X.Y.; Yang, Y.F.; Xiao, F.J.; Li, Q.F.; Yan, J.; Zhang, Q.W.; Wang, L.S.; Li, X.Y.; Wang, H. Regulation of human hepatocellular carcinoma cells by Spred2 and correlative studies on its mechanism. Biochem. Biophys. Res. Commun. 2011, 410, 803–808. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, T.; Gao, T.; Fujisawa, M.; Ohara, T.; Sakaguchi, M.; Yoshimura, T.; Matsukawa, A. SPRED2 Is a Novel Regulator of Autophagy in Hepatocellular Carcinoma Cells and Normal Hepatocytes. Int. J. Mol. Sci. 2024, 25, 6269. [Google Scholar] [CrossRef]
  125. Yeung, K.; Seitz, T.; Li, S.; Janosch, P.; McFerran, B.; Kaiser, C.; Fee, F.; Katsanakis, K.D.; Rose, D.W.; Mischak, H.; et al. Suppression of Raf-1 kinase activity and MAP kinase signalling by RKIP. Nature 1999, 401, 173–177. [Google Scholar] [CrossRef]
  126. Lee, H.C.; Tian, B.; Sedivy, J.M.; Wands, J.R.; Kim, M. Loss of Raf kinase inhibitor protein promotes cell proliferation and migration of human hepatoma cells. Gastroenterology 2006, 131, 1208–1217. [Google Scholar] [CrossRef] [PubMed]
  127. Xie, X.; Wang, Y.; Wang, Z.; Zhang, L.; Li, J.; Li, Y. Hepatocellular carcinoma drug resistance models. Cancer Cell Int. 2025, 25, 195. [Google Scholar] [CrossRef]
  128. Zheng, J.; Wang, S.; Xia, L.; Sun, Z.; Chan, K.M.; Bernards, R.; Qin, W.; Chen, J.; Xia, Q.; Jin, H. Hepatocellular carcinoma: Signaling pathways and therapeutic advances. Signal Transduct. Target. Ther. 2025, 10, 35. [Google Scholar] [CrossRef]
  129. Lei, Y.R.; He, X.L.; Li, J.; Mo, C.F. Drug Resistance in Hepatocellular Carcinoma: Theoretical Basis and Therapeutic Aspects. Front. Biosci. (Landmark Ed.) 2024, 29, 52. [Google Scholar] [CrossRef]
  130. Jing, F.; Li, X.; Jiang, H.; Sun, J.; Guo, Q. Combating drug resistance in hepatocellular carcinoma: No awareness today, no action tomorrow. Biomed. Pharmacother. 2023, 167, 115561. [Google Scholar] [CrossRef]
  131. Ladd, A.D.; Duarte, S.; Sahin, I.; Zarrinpar, A. Mechanisms of drug resistance in HCC. Hepatology 2024, 79, 926–940. [Google Scholar] [CrossRef]
  132. Zheng, S.; Chan, S.W.; Liu, F.; Liu, J.; Chow, P.K.H.; Toh, H.C.; Hong, W. Hepatocellular Carcinoma: Current Drug Therapeutic Status, Advances and Challenges. Cancers 2024, 16, 1582. [Google Scholar] [CrossRef]
  133. Jiang, L.; Li, L.; Liu, Y.; Lu, L.; Zhan, M.; Yuan, S.; Liu, Y. Drug resistance mechanism of kinase inhibitors in the treatment of hepatocellular carcinoma. Front. Pharmacol. 2023, 14, 1097277. [Google Scholar] [CrossRef]
  134. Jin, H.; Shi, Y.; Lv, Y.; Yuan, S.; Ramirez, C.F.A.; Lieftink, C.; Wang, L.; Wang, S.; Wang, C.; Dias, M.H.; et al. EGFR activation limits the response of liver cancer to lenvatinib. Nature 2021, 595, 730–734. [Google Scholar] [CrossRef] [PubMed]
  135. Lu, Y.; Shen, H.; Huang, W.; He, S.; Chen, J.; Zhang, D.; Shen, Y.; Sun, Y. Genome-scale CRISPR-Cas9 knockout screening in hepatocellular carcinoma with lenvatinib resistance. Cell Death Discov. 2021, 7, 359. [Google Scholar] [CrossRef]
  136. Gao, L.; Wang, X.; Tang, Y.; Huang, S.; Hu, C.A.; Teng, Y. FGF19/FGFR4 signaling contributes to the resistance of hepatocellular carcinoma to sorafenib. J. Exp. Clin. Cancer Res. 2017, 36, 8. [Google Scholar] [CrossRef]
  137. Wang, H.; Yang, J.; Zhang, K.; Liu, J.; Li, Y.; Su, W.; Song, N. Advances of Fibroblast Growth Factor/Receptor Signaling Pathway in Hepatocellular Carcinoma and its Pharmacotherapeutic Targets. Front. Pharmacol. 2021, 12, 650388. [Google Scholar] [CrossRef]
  138. Zhu, Y.J.; Zheng, B.; Wang, H.Y.; Chen, L. New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol. Sin. 2017, 38, 614–622. [Google Scholar] [CrossRef]
  139. Yu, J.; Chen, G.G.; Lai, P.B.S. Targeting hepatocyte growth factor/c-mesenchymal-epithelial transition factor axis in hepatocellular carcinoma: Rationale and therapeutic strategies. Med. Res. Rev. 2021, 41, 507–524. [Google Scholar] [CrossRef] [PubMed]
  140. Blivet-Van Eggelpoël, M.J.; Chettouh, H.; Fartoux, L.; Aoudjehane, L.; Barbu, V.; Rey, C.; Priam, S.; Housset, C.; Rosmorduc, O.; Desbois-Mouthon, C. Epidermal growth factor receptor and HER-3 restrict cell response to sorafenib in hepatocellular carcinoma cells. J. Hepatol. 2012, 57, 108–115. [Google Scholar] [CrossRef] [PubMed]
  141. Xin, H.W.; Ambe, C.M.; Hari, D.M.; Wiegand, G.W.; Miller, T.C.; Chen, J.Q.; Anderson, A.J.; Ray, S.; Mullinax, J.E.; Koizumi, T.; et al. Label-retaining liver cancer cells are relatively resistant to sorafenib. Gut 2013, 62, 1777–1786. [Google Scholar] [CrossRef] [PubMed]
  142. Rezayatmand, H.; Razmkhah, M.; Razeghian-Jahromi, I. Drug resistance in cancer therapy: The Pandora’s Box of cancer stem cells. Stem Cell Res. Ther. 2022, 13, 181. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, K.F.; Chen, H.L.; Tai, W.T.; Feng, W.C.; Hsu, C.H.; Chen, P.J.; Cheng, A.L. Activation of phosphatidylinositol 3-kinase/Akt signaling pathway mediates acquired resistance to sorafenib in hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther. 2011, 337, 155–161. [Google Scholar] [CrossRef]
  144. Tan, X.P.; Xiong, B.H.; Zhang, Y.X.; Wang, S.L.; Zuo, Q.; Li, J. FXYD5 promotes sorafenib resistance through the Akt/mTOR signaling pathway in hepatocellular carcinoma. Eur. J. Pharmacol. 2022, 931, 175186. [Google Scholar] [CrossRef]
  145. Merle, P.; Kudo, M.; Krotneva, S.; Ozgurdal, K.; Su, Y.; Proskorovsky, I. Regorafenib versus Cabozantinib as a Second-Line Treatment for Advanced Hepatocellular Carcinoma: An Anchored Matching-Adjusted Indirect Comparison of Efficacy and Safety. Liver Cancer 2023, 12, 145–155. [Google Scholar] [CrossRef]
  146. Lu, F.; Zhao, K.; Ye, M.; Xing, G.; Liu, B.; Li, X.; Ran, Y.; Wu, F.; Chen, W.; Hu, S. Efficacy and safety of second-line therapies for advanced hepatocellular carcinoma: A network meta-analysis of randomized controlled trials. BMC Cancer 2024, 24, 1023. [Google Scholar] [CrossRef]
  147. Shen, Y.; Bai, Y. Effectiveness of regorafenib in second-line therapy for advanced hepatocellular carcinoma: A systematic review and meta-analysis. Medicine 2025, 104, e41356. [Google Scholar] [CrossRef]
  148. Yu, J.; Bai, Y.; Cui, Z.; Cheng, C.; Ladekarl, M.; Cheng, K.C.; Chan, K.S.; Yarmohammadi, H.; Mauriz, J.L.; Zhang, Y. Efficacy and safety of regorafenib as a first-line agent alone or in combination with an immune checkpoint inhibitor for advanced hepatocellular carcinoma: A retrospective cohort study. J. Gastrointest. Oncol. 2024, 15, 1072–1081. [Google Scholar] [CrossRef]
  149. Li, Z.; Wang, J.; Zhao, J.; Leng, Z. Regorafenib plus programmed death-1 inhibitors vs. regorafenib monotherapy in second-line treatment for advanced hepatocellular carcinoma: A systematic review and meta-analysis. Oncol. Lett. 2024, 28, 318. [Google Scholar] [CrossRef] [PubMed]
  150. Karabicici, M.; Azbazdar, Y.; Ozhan, G.; Senturk, S.; Firtina Karagonlar, Z.; Erdal, E. Changes in Wnt and TGF-β Signaling Mediate the Development of Regorafenib Resistance in Hepatocellular Carcinoma Cell Line HuH7. Front. Cell Dev. Biol. 2021, 9, 639779. [Google Scholar] [CrossRef]
  151. Shi, W.; Zhang, S.; Ma, D.; Yan, D.; Zhang, G.; Cao, Y.; Wang, Z.; Wu, J.; Jiang, C. Targeting SphK2 Reverses Acquired Resistance of Regorafenib in Hepatocellular Carcinoma. Front. Oncol. 2020, 10, 694. [Google Scholar] [CrossRef]
  152. Ojha, R.; Leli, N.M.; Onorati, A.; Piao, S.; Verginadis, I.I.; Tameire, F.; Rebecca, V.W.; Chude, C.I.; Murugan, S.; Fennelly, C.; et al. ER Translocation of the MAPK Pathway Drives Therapy Resistance in BRAF-Mutant Melanoma. Cancer Discov. 2019, 9, 396–415. [Google Scholar] [CrossRef]
  153. Gao, C.; Wang, S.W.; Lu, J.C.; Chai, X.Q.; Li, Y.C.; Zhang, P.F.; Huang, X.Y.; Cai, J.B.; Zheng, Y.M.; Guo, X.J.; et al. KSR2-14-3-3ζ complex serves as a biomarker and potential therapeutic target in sorafenib-resistant hepatocellular carcinoma. Biomark. Res. 2022, 10, 25. [Google Scholar] [CrossRef]
  154. Baranyi, M.; Buday, L.; Hegedűs, B. K-Ras prenylation as a potential anticancer target. Cancer Metastasis Rev. 2020, 39, 1127–1141. [Google Scholar] [CrossRef]
  155. Stalnecker, C.A.; Der, C.J. RAS, wanted dead or alive: Advances in targeting RAS mutant cancers. Sci. Signal. 2020, 13, eaay6013. [Google Scholar] [CrossRef]
  156. Blay, J.Y.; Cropet, C.; Mansard, S.; Loriot, Y.; De La Fouchardière, C.; Haroche, J.; Topart, D.; Tougeron, D.; You, B.; Italiano, A.; et al. Long term activity of vemurafenib in cancers with BRAF mutations: The ACSE basket study for advanced cancers other than BRAF(V600)-mutated melanoma. ESMO Open 2023, 8, 102038. [Google Scholar] [CrossRef]
  157. Kim, A.; Cohen, M.S. The discovery of vemurafenib for the treatment of BRAF-mutated metastatic melanoma. Expert. Opin. Drug Discov. 2016, 11, 907–916. [Google Scholar] [CrossRef] [PubMed]
  158. Hauschild, A.; Grob, J.J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H., Jr.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365. [Google Scholar] [CrossRef] [PubMed]
  159. Farley, J.; Brady, W.E.; Vathipadiekal, V.; Lankes, H.A.; Coleman, R.; Morgan, M.A.; Mannel, R.; Yamada, S.D.; Mutch, D.; Rodgers, W.H.; et al. Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: An open-label, single-arm, phase 2 study. Lancet Oncol. 2013, 14, 134–140. [Google Scholar] [CrossRef]
  160. Gross, A.M.; Wolters, P.L.; Dombi, E.; Baldwin, A.; Whitcomb, P.; Fisher, M.J.; Weiss, B.; Kim, A.; Bornhorst, M.; Shah, A.C.; et al. Selumetinib in Children with Inoperable Plexiform Neurofibromas. N. Engl. J. Med. 2020, 382, 1430–1442. [Google Scholar] [CrossRef] [PubMed]
  161. Gershenson, D.M.; Miller, A.; Brady, W.E.; Paul, J.; Carty, K.; Rodgers, W.; Millan, D.; Coleman, R.L.; Moore, K.N.; Banerjee, S.; et al. Trametinib versus standard of care in patients with recurrent low-grade serous ovarian cancer (GOG 281/LOGS): An international, randomised, open-label, multicentre, phase 2/3 trial. Lancet 2022, 399, 541–553. [Google Scholar] [CrossRef] [PubMed]
  162. Diamond, E.L.; Durham, B.H.; Ulaner, G.A.; Drill, E.; Buthorn, J.; Ki, M.; Bitner, L.; Cho, H.; Young, R.J.; Francis, J.H.; et al. Efficacy of MEK inhibition in patients with histiocytic neoplasms. Nature 2019, 567, 521–524. [Google Scholar] [CrossRef] [PubMed]
  163. Buchbinder, E.I.; Cohen, J.V.; Tarantino, G.; Lian, C.G.; Liu, D.; Haq, R.; Hodi, F.S.; Lawrence, D.P.; Giobbie-Hurder, A.; Knoerzer, D.; et al. A Phase II Study of ERK Inhibition by Ulixertinib (BVD-523) in Metastatic Uveal Melanoma. Cancer Res. Commun. 2024, 4, 1321–1327. [Google Scholar] [CrossRef]
  164. Ozohanics, O.; Ambrus, A. Hydrogen-Deuterium Exchange Mass Spectrometry: A Novel Structural Biology Approach to Structure, Dynamics and Interactions of Proteins and Their Complexes. Life 2020, 10, 286. [Google Scholar] [CrossRef] [PubMed]
  165. Daley, B.R.; Vieira, H.M.; Rao, C.; Hughes, J.M.; Beckley, Z.M.; Huisman, D.H.; Chatterjee, D.; Sealover, N.E.; Cox, K.; Askew, J.W.; et al. SOS1 and KSR1 modulate MEK inhibitor responsiveness to target resistant cell populations based on PI3K and KRAS mutation status. Proc. Natl. Acad. Sci. USA 2023, 120, e2313137120. [Google Scholar] [CrossRef]
  166. Atefi, M.; Avramis, E.; Lassen, A.; Wong, D.J.; Robert, L.; Foulad, D.; Cerniglia, M.; Titz, B.; Chodon, T.; Graeber, T.G.; et al. Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin. Cancer Res. 2014, 20, 3446–3457. [Google Scholar] [CrossRef]
  167. Bedognetti, D.; Roelands, J.; Decock, J.; Wang, E.; Hendrickx, W. The MAPK hypothesis: Immune-regulatory effects of MAPK-pathway genetic dysregulations and implications for breast cancer immunotherapy. Emerg. Top. Life Sci. 2017, 1, 429–445. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of RAS/MAPK Signaling Pathway. Ligand-induced receptor dimerization promotes phosphorylation of tyrosine residues within the C-terminal cytoplasmic domain of receptor tyrosine kinases (RTKs). Adaptor proteins bind to these phosphotyrosine sites and recruit guanine nucleotide exchange factors (GEFs), which catalyze the exchange of GDP for GTP on RAS. Activated RAS triggers RAF dimerization, leading to RAF kinase activation and subsequent phosphorylation of MEK. MEK then phosphorylates and activates ERK (MAPK), which translocates into the nucleus to phosphorylate diverse transcription factors, thereby regulating gene expression programs involved in proliferation, survival, and differentiation.
Figure 1. Schematic illustration of RAS/MAPK Signaling Pathway. Ligand-induced receptor dimerization promotes phosphorylation of tyrosine residues within the C-terminal cytoplasmic domain of receptor tyrosine kinases (RTKs). Adaptor proteins bind to these phosphotyrosine sites and recruit guanine nucleotide exchange factors (GEFs), which catalyze the exchange of GDP for GTP on RAS. Activated RAS triggers RAF dimerization, leading to RAF kinase activation and subsequent phosphorylation of MEK. MEK then phosphorylates and activates ERK (MAPK), which translocates into the nucleus to phosphorylate diverse transcription factors, thereby regulating gene expression programs involved in proliferation, survival, and differentiation.
Pharmaceutics 17 01348 g001
Figure 2. Schematic illustration of accessory and scaffold proteins regulating the RAS/MAPK signaling cascade. A variety of non-enzymatic proteins spatially and temporally organize the assembly and activity of pathway components of RAS/MAPK signaling. Anchoring, scaffold, adapter, and docking proteins coordinate the efficient activation of RAS/MAPK signaling pathway, whereas negative modulators act to attenuate the signal (see text for details).
Figure 2. Schematic illustration of accessory and scaffold proteins regulating the RAS/MAPK signaling cascade. A variety of non-enzymatic proteins spatially and temporally organize the assembly and activity of pathway components of RAS/MAPK signaling. Anchoring, scaffold, adapter, and docking proteins coordinate the efficient activation of RAS/MAPK signaling pathway, whereas negative modulators act to attenuate the signal (see text for details).
Pharmaceutics 17 01348 g002
Figure 3. Involvement of kinase suppressor of Ras 1 (KSR1) in RAS/MAPK pathway activation and structural organization of KSR proteins. (a) KSR1 functions as a scaffold protein that facilitates the assembly of RAF, MEK, and ERK, thereby promoting activation of the RAS/MAPK signaling cascade and driving oncogenic signaling. (b) Schematic representation of the conserved domain architecture of KSR proteins. Both human (hKSR1, hKSR2) and murine (mKSR1) KSR share five conserved domains (CA1–CA5), which mediate interactions with signaling partners and regulate pathway activity.
Figure 3. Involvement of kinase suppressor of Ras 1 (KSR1) in RAS/MAPK pathway activation and structural organization of KSR proteins. (a) KSR1 functions as a scaffold protein that facilitates the assembly of RAF, MEK, and ERK, thereby promoting activation of the RAS/MAPK signaling cascade and driving oncogenic signaling. (b) Schematic representation of the conserved domain architecture of KSR proteins. Both human (hKSR1, hKSR2) and murine (mKSR1) KSR share five conserved domains (CA1–CA5), which mediate interactions with signaling partners and regulate pathway activity.
Pharmaceutics 17 01348 g003
Figure 4. Dual roles of KSR1 in cancer. KSR1 can exert either tumor-promoting or tumor-suppressive effects depending on the cellular and genetic context. KSR1 overexpression facilitates RAS/MAPK pathway activation that supports proliferation in many cancers; however, KSR1 overexpression may lead to hyperactivation of RAS/MAPK signaling in cancers harboring active RAS or RAF mutations, resulting in growth arrest or apoptosis.
Figure 4. Dual roles of KSR1 in cancer. KSR1 can exert either tumor-promoting or tumor-suppressive effects depending on the cellular and genetic context. KSR1 overexpression facilitates RAS/MAPK pathway activation that supports proliferation in many cancers; however, KSR1 overexpression may lead to hyperactivation of RAS/MAPK signaling in cancers harboring active RAS or RAF mutations, resulting in growth arrest or apoptosis.
Pharmaceutics 17 01348 g004
Figure 5. Potential strategies for KSR1 targeting. KSR1 activity in cancer can be therapeutically suppressed through multiple approaches, including RNA interference or antisense oligonucleotides, small-molecule inhibitors that disrupt KSR1–RAF/MEK interactions, and targeted protein degradation strategies such as PROTACs.
Figure 5. Potential strategies for KSR1 targeting. KSR1 activity in cancer can be therapeutically suppressed through multiple approaches, including RNA interference or antisense oligonucleotides, small-molecule inhibitors that disrupt KSR1–RAF/MEK interactions, and targeted protein degradation strategies such as PROTACs.
Pharmaceutics 17 01348 g005
Figure 6. KSR1 in hepatocellular carcinoma (HCC). KSR1 is highly expressed in HCC and positively linked to RAS/MAPK activity. KSR1 overexpression leads to RAS/MAPK pathway activation via enhanced phosphorylation of MEK and ERK. In murine models, KSR1 drives hepatocarcinogenesis in cooperation with P53 inactivation or c-Myc overexpression. Conversely, inhibition of KSR1—via shRNA-mediated knockdown or pharmacological blockade using APS-2-79—attenuates RAS/MAPK pathway activation and suppresses tumor progression [90].
Figure 6. KSR1 in hepatocellular carcinoma (HCC). KSR1 is highly expressed in HCC and positively linked to RAS/MAPK activity. KSR1 overexpression leads to RAS/MAPK pathway activation via enhanced phosphorylation of MEK and ERK. In murine models, KSR1 drives hepatocarcinogenesis in cooperation with P53 inactivation or c-Myc overexpression. Conversely, inhibition of KSR1—via shRNA-mediated knockdown or pharmacological blockade using APS-2-79—attenuates RAS/MAPK pathway activation and suppresses tumor progression [90].
Pharmaceutics 17 01348 g006
Figure 7. Chemical structures of small-molecule inhibitors cited in this review. The binding mode of APS-2-79 with KSR2 is also illustrated below the chemical (adapted from reference [98]).
Figure 7. Chemical structures of small-molecule inhibitors cited in this review. The binding mode of APS-2-79 with KSR2 is also illustrated below the chemical (adapted from reference [98]).
Pharmaceutics 17 01348 g007
Table 1. Accessory and Scaffold Proteins for RAS/MAPK Signaling Pathway.
Table 1. Accessory and Scaffold Proteins for RAS/MAPK Signaling Pathway.
ClassProteinBinding PartnerFunctional ImpactReference
AnchoringCNK1 (CNKSR1)RAF-1, Src, KSRUpon RAS activation, promotes RAF relocalization/oligomerization → enhances RAF/MEK/ERK activation[4,21]
FLOT1/2 (Flotillins)CRAF, MEK1/2, ERK1/2Support EGFR clustering/activation; upon stimulation enhance CRAF/MEK/ERK signaling[4,21]
LAT, NTALGrb2, Gads, PLCγ1, Sos1Nucleate membrane-proximal signalosomes[4,21]
PAQR10/11H-, N-, KRAS4A, RasGRP1Anchor RAS to the Golgi → spatial control of signaling[27]
SEF (IL17RD)p-MEK, p-ERKBinds p-MEK at Golgi; potentiates RAF/MEK/ERK signaling[21]
DockingDOK1/2Activated RTKs (EGFR, RET) via PTBDocking adaptors that assemble signaling complexes[21]
IRS2Insulin receptor (RTKs)Couples RTKs to downstream effectors[21]
β-arrestin1/2GPCRs; RAF/MEK/ERKScaffolds RAF/MEK/ERK at plasma membrane and endosomes[21]
FRS2RTKs, SHP2, GRB2Central docking adaptor linking RTKs to Ras/ERK[63]
AdapterCRK/CRKLMultiple pTyr ligandsPromotes ERK signaling by coupling pTyr scaffolds to C3G → Rap1 → B-Raf → MEK → ERK[36]
Grb2pTyr proteins (SH2); SOS1 (N-SH3); Gab1 (C-SH3)Core adaptor coupling RTKs to RAS via SOS[37]
Shc (ShcA/SHC1)RTKs/integrins → Grb2RTK-induced phosphorylation enables Grb2 binding to activate RAS[40]
SHP2 (PTPN11)Gab1 (EGFR and other RTKs), FRS2 (FGFRs)Tyr phosphorylation (e.g., Y542) relieves autoinhibition → supports RAS pathway[41]
PaxillinERK/MAPK module componentsRecruits/organizes ERK module at adhesions[42]
ScaffoldKSR1 and 2RAF (BRAF, CRAF), MEK1/2, ERK1/2Scaffold RAF–MEK–ERK at the membrane to boost efficient and precise RAS signaling[64,65]
IQGAP1EGFR (direct/via ShcA), HER2 heterodimersBridges EGFR/HER2 to MAPK; tunes receptor trafficking and signal amplitude[46]
SHOC2MRAS, PP1, RAF-1Dephosphorylates inhibitory RAF-1 Ser259 site → enables RAF activation[46,66,67,68]
Negative
Modulator
SPRED family (SPRED1 and 2)NF1Negative regulation of RTK→RAS–MAPK by modulating RasGAPs[49]
Sprouty family (SPRY 1–4)GRB2 (SH2)Sequesters GRB2 away from SOS → suppresses RAS activation[51]
RKIP (PEBP1)Raf-1, MEKBlocks Raf-1/MEK interaction or binds Raf-1 N-region to inhibit MEK phosphorylation[52]
ErbinERBB2, Shoc2Modulates Shoc2–ERK signal strength[47]
DUSPERK1/2Acts as negative feedback regulators → directly dephosphorylates MAPKs (especially ERK)[59,60]
Table 2. Targeting Components and Regulators of RAS/MAPK Signaling in HCC.
Table 2. Targeting Components and Regulators of RAS/MAPK Signaling in HCC.
AgentTargetModelResultReference
rigosertibRASin vitro (I)Reduced HCC cell proliferation[112]
miR-4510RAFin vitro (I)Suppressed HCC cell proliferation[113]
in vivo (I)Decreased tumor size and proliferation
cobimetinibMEKin vitro (I)Reduced HCC cell proliferation[114]
in vivo (I)Decreased tumor development
siRNAERKin vitro (I)Decreased HCC cell proliferation[115]
RNF173GRB2in vitro (I)Inhibited RAS/MAPK signaling[117]
shRNA
SUMO1
SHP2in vitro (I)
in vivo (A)
Inhibited RAS/MAPK signaling
Increased tumor growth
[120]
miR-126
miR-126 inhibitor
SPRED1in vitro (I)
in vivo (A)
Increased proliferation and ERK phosphorylation
Reduced tumor growth
[122]
SPRED2
siRNA
SPRED2in vitro (A)
in vivo (I)
Reduced proliferation and ERK phosphorylation
Increased tumor growth
[123]
siRNA
RKIP
RKIPin vitro (I)
in vitro (A)
Increased MEK phosphorylation
Reduced HCC cell proliferation and migration
[126]
(I) Inhibition of target; (A) Activation of target.
Table 3. Resistance to Molecular Target Therapy in HCC.
Table 3. Resistance to Molecular Target Therapy in HCC.
DrugTargetResistance MechanismReference
Lenvatinib VEGFR, FGFR, PDGFREGFR–PAK2–ERK5 signaling activation[134]
Loss of NF1 and DUSP9[135]
SorafenibVEGFR, FGFR, PDGFR, RAFOverexpression of FGF[136]
Activation of HGF/c-Met axis[139]
Activation of EGFR and HER3[140]
Enrichment of cancer stem cells[141]
Compensatory activation of the PI3K/AKT pathway[143]
RegorafenibVEGFR, FGFR, PDGFRActivation of TGF-β signaling[150]
NF-κB and STAT3 activation (SphK2 overexpression)[151]
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

Moon, H.; Park, H.; Lee, S.; Lee, S.; Ro, S.W. Targeting Kinase Suppressor of Ras 1 (KSR1) for Cancer Therapy. Pharmaceutics 2025, 17, 1348. https://doi.org/10.3390/pharmaceutics17101348

AMA Style

Moon H, Park H, Lee S, Lee S, Ro SW. Targeting Kinase Suppressor of Ras 1 (KSR1) for Cancer Therapy. Pharmaceutics. 2025; 17(10):1348. https://doi.org/10.3390/pharmaceutics17101348

Chicago/Turabian Style

Moon, Hyuk, Hyunjung Park, Soyun Lee, Sangjik Lee, and Simon Weonsang Ro. 2025. "Targeting Kinase Suppressor of Ras 1 (KSR1) for Cancer Therapy" Pharmaceutics 17, no. 10: 1348. https://doi.org/10.3390/pharmaceutics17101348

APA Style

Moon, H., Park, H., Lee, S., Lee, S., & Ro, S. W. (2025). Targeting Kinase Suppressor of Ras 1 (KSR1) for Cancer Therapy. Pharmaceutics, 17(10), 1348. https://doi.org/10.3390/pharmaceutics17101348

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