Fact-checked by Grok 2 weeks ago

c-Raf

c-Raf, also known as RAF1 or Raf-1, is a encoded by the RAF1 proto- on human , serving as a critical mediator in the RAS-RAF-MEK-ERK (MAPK) signaling cascade that regulates cellular processes such as , , survival, and . Originally identified in the as the cellular homolog of the v-raf viral , c-Raf functions downstream of , where binding to active Ras-GTP recruits it to the plasma membrane for via a series of events and dimerization with other RAF family members like B-Raf. Structurally, the 648-amino-acid protein (~73 kDa) comprises an N-terminal regulatory with Ras-binding and cysteine-rich regions, a central hinge, and a C-terminal , allowing tight control through inhibitory intramolecular interactions that are relieved upon stimulation; recent cryo-EM studies as of 2025 have provided deeper insights into these dynamics. In physiological contexts, c-Raf integrates signals from receptor tyrosine kinases and G-protein-coupled receptors to phosphorylate and activate MEK1/2, which in turn stimulate ERK1/2 to influence and cytoskeletal dynamics, with studies in mice revealing its essential role in embryonic development and vascular integrity, as embryos lacking c-Raf die around E10.5-E12.5 due to defective placental and hematopoietic functions. Beyond normal signaling, c-Raf is implicated in oncogenesis, where it is frequently overexpressed in various cancers including , liver, and malignancies, facilitating tumor progression by amplifying RAS-driven signals even in the absence of direct RAF1 mutations, which are rare compared to those in B-RAF; emerging evidence also highlights RAF1 fusions and amplifications in some tumors. Its dysregulation also contributes to therapeutic resistance, such as paradoxical activation of the MAPK pathway in response to B-RAF inhibitors in wild-type cells, highlighting c-Raf's role in adaptive signaling networks.

History and Discovery

Initial Identification of Raf Kinases

The Raf kinase family was first identified through the discovery of the viral oncogene in 1983, transduced by the acutely transforming murine sarcoma retrovirus 3611-MSV. This retrovirus, isolated from a tumor, efficiently transformed NIH 3T3 fibroblasts upon with its proviral DNA, producing foci at a rate of approximately 4 per nanogram, and the recovered virus induced fibrosarcomas in inoculated . The v-raf was characterized as a novel sequence without close homology to previously known viral , though its transforming potential suggested an initial association with activity, akin to other retroviral oncoproteins like v-src. This finding, reported by U.R. Rapp and colleagues, marked the initial recognition of the Raf family as key players in cellular transformation. The cellular homolog of v-raf, termed c-Raf (also known as c-Raf-1 or Raf-1), was subsequently identified in mammalian cells in the mid-1980s. In 1984, chromosomal mapping studies located the human c-raf locus on , confirming its distinction from other families and its conservation across species. The full coding of human c-raf was cloned and reported in 1986, revealing a 648-amino-acid protein with conserved motifs indicative of a , thus establishing its biochemical classification within the superfamily. This cellular counterpart demonstrated structural similarity to v-raf, particularly in its kinase domain, underscoring the retroviral oncogene's origin as a captured and altered version of a normal cellular involved in . Initial functional studies of Raf kinases in the late 1980s began to elucidate their role in signaling pathways. Experiments demonstrated that c-Raf undergoes rapid in response to growth factors such as and , as well as upon expression of membrane-bound oncogenes like v-src, indicating activation within minutes of stimulation in transformed cells. Complementary work with temperature-sensitive mutants of avian retroviruses carrying related Raf-like oncogenes, such as v-mil in the MH2 virus, showed that shifting to permissive temperatures restored transforming activity and morphological changes in infected cells, linking Raf members to sustained proliferative signaling. These early investigations, building on the transforming properties observed with v-raf, positioned the Raf kinases as critical intermediaries in mitogenic signal relay from cell surface receptors to intracellular effectors.

Specific Characterization of c-Raf

The human c-Raf, also known as RAF1, was cloned as a cDNA from a fetal liver library, revealing an encoding a 648-amino acid protein containing a domain. The activity of c-Raf was demonstrated in 1991 through assays, in which immunoprecipitated c-Raf from stimulated cells phosphorylated exogenous substrates including basic protein, confirming its role as an active . Early subcellular localization studies utilizing in revealed that c-Raf is predominantly distributed in the under basal conditions but translocates to the plasma membrane upon stimulation with growth factors such as colony-stimulating factor 1, highlighting its dynamic localization in response to signaling cues. studies in the early , building on initial data, confirmed the chromosomal assignment of the RAF1 gene to the 3p25 region in humans, positioning it within a locus implicated in certain cancers and providing a genetic framework for further functional analyses.

Structure

Domain Organization

c-Raf, also known as RAF1, is a 648-amino-acid serine/threonine that consists of an N-terminal regulatory region spanning residues 1-257 and a C-terminal encompassing residues 306-648. The N-terminal region maintains the protein in an autoinhibited state through intramolecular interactions with the , while the C-terminal harbors the catalytic activity essential for in the MAPK/ERK pathway. The regulatory domain includes several key components that mediate interactions with upstream regulators. The conserved region 1 (CR1), corresponding to residues 51-131, contains a zinc finger-like structure that facilitates binding to GTP-bound Ras, thereby recruiting c-Raf to the plasma membrane for activation. Adjacent to CR1 is the conserved region 2 (CR2), a serine-rich segment from residues 256-269, which serves as a binding site for 14-3-3 proteins upon phosphorylation at serine 259, stabilizing the inactive conformation. The kinase domain adopts a conserved bilobal typical of eukaryotic protein kinases, with an N-lobe involved in ATP and a C-lobe responsible for recognition. The ATP-binding site features a glycine-rich loop (G-loop) at residues 377-382, which positions the for phosphoryl transfer. The activation loop, spanning residues 491-502, includes critical sites serine 497 and serine 499, which are targeted by and modulate catalytic competence upon modification. Connecting the regulatory and kinase domains is a flexible inter-domain linker region (residues 258-305), which plays a role in autoinhibition by allowing the N-terminal region to occlude the kinase and can influence dimerization interfaces.

Structural Dynamics and Recent Insights

In the autoinhibited state of c-Raf, the N-lobe of the kinase domain engages with the regulatory domain, particularly through interactions involving the cysteine-rich domain (CRD), which nestles against the kinase domain and helps bury the activation segment in an autoinhibitory conformation. This configuration locks the C-helix in an outward, inactive position, as revealed by cryo-EM structures at 3.4 Å resolution. Earlier NMR studies from 2017 provided initial insights into the dynamic regulatory interactions contributing to this autoinhibition, while a 2022 further corroborated the burial of the activation segment in related Raf family members. Dimerization-induced of c-Raf involves the formation of a side-to-side between kinase domains, which relieves autoinhibition by rearranging the C-helix into an active inward position and exposing the dimerization surface. This process is particularly asymmetric in c-Raf/B-Raf heterodimers, where one protomer adopts a more active conformation to drive the partner's , as detailed in 2022 cryo-EM studies of the monomer-to-dimer transition. The side-to-side dimer , often mediated by 14-3-3 binding to phosphorylated serine residues, promotes back-to-back alignment in the active state, enabling subsequent MEK phosphorylation. Recent cryo-EM structures from 2025 of the c-Raf/MEK1/14-3-3 ternary complex, resolved at 3.4 Å for the autoinhibited state, illustrate how 14-3-3 stabilizes the inactive state by the CR2 , specifically the phosphorylated Ser259 site, which sequesters the kinase domain and prevents dimerization. In this complex, the 14-3-3 dimer engages both segments (pSer259 and pSer621) on opposite sides, maintaining the CRD in a cradle-like position that blocks regulatory domain release. These structures highlight two open-monomer intermediates where pSer259 is released, facilitating and progression toward activation. The Hsp90 chaperone complex, known as the RHC (Raf--Cdc37), plays a critical role in stabilizing the nascent kinase domain of c-Raf during folding, as shown in structural data. Cryo-EM analysis at 3.7 resolution reveals an asymmetric interaction where the unfolded N-lobe β5-strand of c-Raf threads through the luminal cavity, engaging via van der Waals contacts with 's src-loops and helical hairpins, while Cdc37 bridges the N- and C-lobes to the chaperone. This configuration ensures proper maturation of the kinase domain, with molecular simulations confirming strong electrostatic stabilization (e.g., -171.11 kcal/mol for the pS13-K405 interaction).

Evolutionary Relationships

Conservation Across Species

c-Raf, encoded by the RAF1 gene, displays high sequence conservation across vertebrate species, particularly in its kinase domain. The human Raf1 protein shares 98.9% amino acid sequence identity with its ortholog, reflecting strong evolutionary preservation of core catalytic functions. This high similarity extends to the kinase domain, where identity exceeds 90% among vertebrates, enabling conserved roles in . In contrast, the N-terminal regulatory domain shows lower conservation, approximately 70%, allowing for species-specific regulatory nuances while maintaining essential Ras-binding motifs. In , c-Raf orthologs exhibit moderate sequence similarity, underscoring functional homology in Ras-MAPK signaling. The Raf homolog, known as Polehole (Raf or D-Raf), shares about 40% overall identity with human c-Raf, with higher conservation in the kinase domain. This ortholog is essential for torso receptor tyrosine kinase (RTK) signaling during embryonic patterning. Similarly, the lin-45 gene encodes a Raf homolog required for vulval induction downstream of let-60 , displaying significant sequence similarity in key domains despite overall lower identity compared to vertebrates. Functional conservation is evident from genetic studies across species. In mice, targeted disruption of the Raf1 gene results in embryonic lethality around mid-gestation, characterized by growth retardation and defects in placental and fetal liver development, as reported in late analyses. This phenotype mirrors the developmental defects observed in upon Raf loss, including disruptions in where the /Ras/ pathway specifies fates. Such parallels highlight the preserved necessity of c-Raf for RTK-mediated signaling in . The evolutionary roots of c-Raf trace to the tyrosine kinase-like (TKL) family, which emerged in the last eukaryotic common ancestor approximately 1.5 billion years ago, with partial homologs appearing in early eukaryotes but no direct Raf kinases in . This ancient supports the broad conservation of Raf-mediated pathways from unicellular organisms to complex metazoans.

Relations to Other Raf Family Members

The Raf family of serine/ in humans consists of three isoforms: c-Raf (encoded by RAF1), B-Raf (encoded by BRAF), and A-Raf (encoded by ARAF). These genes are located on different chromosomes: RAF1 on 3p25.2, BRAF on 7q34, and ARAF on Xp11.2. Despite these distinct genomic positions, the isoforms share approximately 75% identity in their kinase domains, while exhibiting notable differences in their N-terminal regulatory regions that influence activation and specificity. In terms of intrinsic activity, c-Raf displays the lowest basal activity among the isoforms and typically requires dimerization for full , whereas B-Raf possesses the highest basal activity and can as a under certain conditions. A-Raf exhibits the weakest overall activity toward MEK substrates and shows tissue-restricted expression, predominantly in urogenital tissues. In contrast, c-Raf maintains the broadest expression pattern across tissues, enabling its involvement in diverse cellular contexts.48650-7/fulltext) Functional redundancy exists among the isoforms, particularly through the formation of c-Raf/B-Raf heterodimers, which are prevalent in Ras-mediated MAPK signaling and enhance pathway activation. studies in mice reveal compensatory mechanisms; for instance, c-Raf can partially compensate for B-Raf loss in cardiac development, mitigating some hypertrophic responses despite B-Raf's dominant role in certain contexts.01054-4.pdf) The evolutionary divergence of the Raf family stems from events associated with the two rounds of whole-genome duplication (2R hypothesis) early in evolution, approximately 500 million years ago, leading to the three paralogs observed in mammals; c-Raf has retained the most ubiquitous expression profile among them.

Regulation of Activity

Activation Pathways

The activation of c-Raf, a serine/ central to the MAPK/ERK pathway, is initiated primarily through -dependent recruitment to the plasma membrane. GTP-bound interacts with high affinity (approximately 10 nM) to the cysteine-rich domain (CR1) of c-Raf, specifically via the (RBD), which facilitates the translocation of cytosolic c-Raf to the membrane. This binding involves key residues R89 and Y91 within the CR1 region of c-Raf, enabling the disruption of autoinhibitory interactions that maintain c-Raf in an inactive state. Consequently, recruits c-Raf to lipid rafts enriched in signaling molecules, positioning it for subsequent activating events. Upstream signals from receptor , such as , trigger the GDP-to-GTP exchange on via guanine nucleotide exchange factors (GEFs), thereby activating the Ras-c-Raf interaction. This recruitment enables Src family to c-Raf at 341 (Y341) within its domain, a critical step that enhances activity and promotes downstream signaling. Src-mediated at Y341 occurs in a Ras-dependent manner, stabilizing an open conformation conducive to further activation. Scaffold proteins like kinase suppressor of Ras 1 and 2 (KSR1/2) further augment c-Raf activation by assembling a multiprotein complex at the membrane, recruiting c-Raf alongside , MEK, and other components to increase signaling efficiency. Discovered in mammalian systems in , KSR1 acts as a docking platform that coordinates these interactions without intrinsic kinase activity, thereby amplifying the localized activation of c-Raf. This scaffold-mediated recruitment ensures spatial organization, preventing cross-talk and optimizing the phosphorylation cascade. Dimerization represents another key , where c-Raf forms homodimers or heterodimers with B-Raf through a conserved in their domains. Studies using fluorescence resonance energy transfer () in the 2010s demonstrated that this dimerization, induced by binding, enables allosteric of the receiver protomer independent of ATP binding, thereby boosting catalytic efficiency. The asymmetric nature of these dimers allows one subunit to allosterically relieve autoinhibition in the partner, a process essential for full competence.

Inhibitory and Feedback Mechanisms

One key inhibitory mechanism of c-Raf involves the binding of 14-3-3 proteins, which is facilitated by at serine 259 (S259) in the conserved region 2 (CR2) and serine 621 (S621) in the conserved region 3 (CR3). This binding stabilizes the autoinhibited monomeric conformation of c-Raf, preventing premature and ensuring signaling fidelity by antagonizing recruitment to the plasma membrane in response to weak stimuli. Recent cryo-electron microscopy (cryo-EM) structures of the CRAF₂/14-3-3₂ complex at 3.4 Å resolution reveal that the dimeric 14-3-3 proteins bridge the CR2 and CR3 regions, enforcing an autoinhibitory state that is disrupted upon RAS-mediated membrane recruitment. A 2025 study further provides cryo-EM structures of CRAF/MEK1/14-3-3 complexes at resolutions up to 2.3 Å, elucidating autoinhibited and open-monomer states and additional regulatory features. Negative regulation of c-Raf also occurs through inhibitory at specific sites, including S259 and threonine 401 (T401). of S259 by () in response to elevated levels directly suppresses c-Raf activity, rendering it resistant to and correlating with deactivation of downstream ERK signaling. Similarly, at T401 inhibits c-Raf function by disrupting key protein interactions within the MAPK cascade. For to proceed, of these sites, particularly S259, is essential and is mediated by 2A (PP2A), which associates with c-Raf to promote membrane localization and relieve inhibition. Feedback inhibition further fine-tunes c-Raf activity through downstream ERK-mediated . Activated ERK phosphorylates c-Raf at T401 (and homologous sites such as S289, S296, and S301), which dampens pathway signaling by promoting from upstream activators like . This negative feedback loop also targets the kinase suppressor of (KSR) scaffold, where ERK at sites like T260, T274, S320, and S443 disrupts the KSR-c-Raf complex, attenuating signal transmission to MEK. Studies from the in cellular models, including fibroblasts, demonstrated this ERK-dependent as a rapid mechanism to limit sustained MAPK activation following stimulation. To prevent prolonged signaling, c-Raf undergoes proteasomal degradation via ubiquitination, particularly after extended activation periods. The E3 ubiquitin ligase targets misfolded or kinase-inactive forms of c-Raf for polyubiquitination, leading to their clearance by the 26S ; this process is autophosphorylation-dependent at S621, as unphosphorylated c-Raf is rapidly degraded to maintain . inhibitors such as MG132 confirm this pathway by stabilizing c-Raf levels, highlighting its role in terminating signaling to avoid aberrant cellular responses.

Biological Functions

Role in MAPK/ERK Signaling

c-Raf, also known as Raf-1, serves as a key kinase (MAP3K) in the canonical Ras-Raf-MEK-ERK signaling cascade, where it directly phosphorylates and activates MEK1/2 by targeting serine residues at positions 218 and 222 in their activation loops. This phosphorylation event, characterized by a Michaelis constant (Km) of approximately 0.8-1 μM for MEK as substrate, enables MEK1/2 to subsequently phosphorylate and activate ERK1/2 at and residues in their TEY . Activated ERK1/2 then translocates to the to phosphorylate transcription factors such as Elk-1, thereby regulating programs involved in and . In response to extracellular stimuli like growth factors including (PDGF) and (EGF), c-Raf integrates signals primarily through upstream activation by GTP-bound at the plasma membrane, leading to rapid amplification of mitogenic responses. This activation typically manifests as transient pulses of ERK , peaking within 5-10 minutes post-stimulation before returning to baseline due to feedback mechanisms, ensuring precise temporal control of downstream effects. Among Raf isoforms, c-Raf predominantly transduces stress-related signals, such as those induced by tumor necrosis factor-α (TNF-α), whereas B-Raf is the primary mediator of growth factor-driven ERK activation; notably, c-Raf/B-Raf heterodimers enhance signaling efficiency by promoting allosteric activation and increased output within the cascade. Systems-level modeling using flux control analysis has revealed that only about 10-20% activation of the total cellular c-Raf pool is sufficient to achieve maximal ERK signaling flux, underscoring the pathway's ultrasensitive response and the isoform's role as a tunable rather than a strict rate-limiter. This quantitative aspect highlights how events, such as those at c-Raf's serine 338 and 341 residues, fine-tune its contribution to overall pathway dynamics.

Involvement in Other Cellular Processes

Beyond its canonical role in the MAPK/ERK pathway, c-Raf contributes to the regulation of through post-translational modifications of pro-apoptotic proteins. Specifically, of c-Raf stimulates the of the BH3-only protein BAD at serine 112 (Ser112), which promotes its sequestration in the by 14-3-3 proteins, thereby preventing BAD from binding and inhibiting the anti-apoptotic protein at the mitochondria. This mechanism enhances cell survival in a kinase-dependent manner via downstream effectors like RSK. Studies in cultured cerebellar granule neurons have demonstrated that growth factor-induced c-Raf leads to BAD at Ser112 and Ser136, cooperatively inactivating BAD and protecting neurons from induced by trophic factor withdrawal. c-Raf also participates in cytoskeletal organization through direct protein-protein interactions independent of its activity toward MEK. In particular, c-Raf binds to Rho-associated α (Rok-α, also known as ROCK1), inhibiting its activity and thereby modulating RhoA signaling to facilitate proper dynamics. This interaction is essential for the formation of stress fibers in fibroblasts, as evidenced by studies showing that c-Raf-deficient embryonic fibroblasts exhibit hyperactivation of Rok-α, resulting in excessive cortical bundling and impaired stress fiber assembly; restoring c-Raf expression or inhibiting Rok-α reverses this defect and promotes stress fiber formation and cell spreading. These MEK-independent effects highlight c-Raf's function in maintaining cytoskeletal integrity during and . In the context of angiogenesis, c-Raf supports vascular development by influencing endothelial cell behavior, including the regulation of vascular endothelial growth factor (VEGF) expression through crosstalk with NF-κB signaling. In mouse models of growth plate maturation, conditional deletion of c-Raf in chondrocytes disrupts VEGF-A secretion, delaying endothelial invasion and vascularization, underscoring c-Raf's role in coordinating angiogenic cues. Although primarily studied in non-endothelial compartments, emerging evidence from endothelial cell lines indicates that c-Raf activation enhances NF-κB-mediated transcription of VEGF in response to inflammatory stimuli, fostering autocrine loops that promote endothelial proliferation and tube formation in vitro. c-Raf links to metabolic regulation in hepatic cells by integrating with insulin signaling pathways that control homeostasis. Through the Raf-1/MEK/ERK/p90RSK , c-Raf indirectly modulates activity by promoting the and inactivation of glycogen synthase kinase-3 (GSK-3), which otherwise inhibits via phosphorylations at multiple sites. This effect is particularly relevant in liver hepatocytes, where stimuli like AICAR activate c-Raf to enhance GSK-3 inhibition, thereby increasing synthesis in coordination with insulin-induced Akt signaling, though the pathway's contribution is context-dependent and secondary to PI3K/Akt.

Disease Associations

Non-Cancerous Disorders

c-Raf, encoded by the RAF1 gene, has been implicated in several non-cancerous disorders through its dysregulation in the MAPK/ERK signaling pathway, particularly in genetic developmental conditions known as and in inflammatory and neurological pathologies. Gain-of-function mutations in RAF1 lead to hyperactivation of downstream effectors, disrupting normal cellular processes during development and immune responses. In , a characterized by facial dysmorphology, , and congenital heart defects, rare RAF1 mutations cause gain-of-function effects that enhance ERK signaling, resulting in and other cardiac abnormalities. For instance, the S257L mutation in the CR2 domain of c-Raf impairs inhibitory at serine 259, promoting constitutive activity and leading to severe cardiac defects in affected individuals. These mutations were first identified in 2007, and RAF1 variants account for approximately 3-17% of cases, with a notable ~5% prevalence in cohorts lacking PTPN11 mutations. Patients with RAF1-associated exhibit a higher incidence of (up to 95%) compared to other genetic subtypes. Similarly, activating variants in RAF1 contribute to cardio-facio-cutaneous (CFC) syndrome, another involving ectodermal, cardiac, and craniofacial anomalies that impair postnatal development. The P261Q variant, located in the CR2 domain, exemplifies these changes by reducing autoinhibitory interactions and elevating MAPK pathway activity, which disrupts cellular proliferation and differentiation during embryogenesis. RAF1 mutations represent 10-15% of cases across , including CFC, where they are less common than BRAF variants but still drive overlapping phenotypes like and . These genetic alterations highlight c-Raf's conserved role in developmental signaling, as seen in evolutionary studies of MAPK conservation. Beyond genetic disorders, c-Raf participates in inflammatory responses, particularly in TLR4-mediated signaling that exacerbates sepsis. In mouse models of lipopolysaccharide (LPS)-induced sepsis, c-Raf activation downstream of TLR4 promotes phosphorylation of MEK and ERK, culminating in excessive cytokine release such as TNF-α and IL-6, which drives systemic inflammation and organ failure. Studies from the 2010s using Raf inhibitors like sorafenib demonstrated reduced cytokine production and improved survival in LPS-challenged mice, underscoring c-Raf's role in amplifying the innate immune response to bacterial endotoxins. This pathway's overactivation contributes to the cytokine storm observed in human sepsis, linking c-Raf to acute inflammatory disorders. Neurological associations involve c-Raf overactivation in models, where amyloid-β (Aβ) peptides induce activation, leading to c-Raf-mediated ERK signaling and subsequent hyper. In Aβ-exposed neuronal cultures and transgenic mouse models, this cascade activates GSK-3β, a key , promoting formation and synaptic dysfunction. Inhibition of the -Raf-ERK pathway reduces at disease-relevant sites like Ser396/404, suggesting c-Raf as a contributor to Aβ-driven neurodegeneration without directly causing oncogenesis.

Oncogenic Roles and Alterations

c-Raf, encoded by the RAF1 gene, contributes to oncogenesis primarily through hyperactivation rather than frequent direct mutations, often amplifying MAPK/ERK signaling in various cancers. Amplification and copy number gains of RAF1 occur in approximately 16% of cutaneous melanomas according to (TCGA) data, with higher rates up to 29% observed in some cohorts of metastatic samples, and these alterations are associated with worse . In lung cancers, RAF1 alterations including amplifications are less common, detected in about 2-3% of non-small cell lung carcinoma cases in pan-cancer analyses, yet overexpression of c-Raf correlates with disease progression across multiple tumor types. Such genomic gains lead to elevated c-Raf protein levels, enhancing tumor and survival independent of its kinase activity in certain contexts. Activating in c-Raf are rare in cancers, occurring in less than 1% of cases overall due to its intrinsically low basal activity compared to B-Raf. In , point in the c-Raf domain, such as those mimicking sites like S338, are infrequent, comprising around 2% of alterations in some subtypes, and are more prevalent in tumors with concurrent where they potentiate downstream signaling. These typically enhance c-Raf's responsiveness to upstream activation, promoting oncogenic transformation in RAS-mutant backgrounds rather than acting as primary drivers. c-Raf engages in significant with other oncogenes, amplifying malignant phenotypes through integrated pathway activation. In NRAS-driven melanomas, c-Raf serves as the primary effector of NRAS, enhancing PI3K/AKT signaling to support tumor growth and resistance to targeted therapies. Additionally, c-Raf compensates for BRAF inhibition by forming heterodimers with wild-type or B-Raf, leading to paradoxical ERK activation that drives resistance in BRAF- cancers. Recent studies highlight c-Raf's evolving role in KRAS- pancreatic cancer, where it promotes tumor invasion through both ERK-dependent and independent mechanisms, such as kinase-independent scaffolding functions that sustain pro-survival signaling. A 2023 review emphasizes c-Raf's context-specific contributions in KRAS-driven malignancies, underscoring its potential as a therapeutic vulnerability in these aggressive tumors.

Therapeutic Targeting

Development of Inhibitors

The development of c-Raf inhibitors includes early multikinase agents like , a type II ATP-competitive inhibitor approved by the FDA in 2005 for advanced , which binds the kinase's conserved ATP-binding pocket and extends into an allosteric pocket to stabilize the inactive DFG-out conformation, with an of approximately 6 nM for c-Raf while also inhibiting B-Raf (IC50 ~22 nM) and other kinases such as VEGFR2, resulting in a pan-Raf profile with notable off-target effects that limit selectivity. Subsequent advancements in type II inhibitors include compounds that further stabilize the inactive DFG-out conformation of the . PLX-4032 (), clinically approved for BRAF-mutant , represents this approach but paradoxically induces c-Raf in wild-type cells through enhanced Raf dimerization and of the MAPK pathway. To circumvent issues with orthosteric binding and paradoxical , researchers have explored allosteric modulators that target the Ras-binding site within the CR1 regulatory of c-Raf, disrupting upstream recruitment by active Ras-GTP. Preclinical compounds developed in the exemplify this strategy by binding to interfaces that prevent Ras-c-Raf interaction, thereby inhibiting without directly engaging the . More recently, proteolysis-targeting chimeras (PROTACs) have emerged as a degradation-based for c-Raf, recruiting ubiquitin ligases to induce -mediated proteasomal breakdown. For example, MS934, a VHL-recruiting MEK1/2 PROTAC, collaterally degrades CRAF in KRAS-mutant cells, offering a complementary mechanism to traditional inhibition by eliminating the protein entirely rather than merely blocking its activity.

Clinical Applications and Challenges

Sorafenib, a multikinase inhibitor targeting c-Raf among other kinases, received FDA approval in 2007 for the treatment of unresectable hepatocellular carcinoma (HCC), based on the SHARP trial demonstrating improved overall survival from 7.9 to 10.7 months compared to placebo. The objective response rate in this trial was approximately 2%, primarily attributed to Raf pathway inhibition, though the drug's broader effects on angiogenesis also contribute. Similarly, sorafenib was approved by the FDA in 2005 for advanced renal cell carcinoma (RCC), where it extended progression-free survival to 5.5 months versus 2.8 months with placebo in the TARGET trial, with c-Raf inhibition playing a key role in suppressing tumor proliferation. In BRAF-mutant , combination therapies incorporating c-Raf modulation have shown enhanced efficacy; for instance, the BRAF inhibitor combined with the improved median to 14.9 months in the phase III trial, compared to 7.3 months with monotherapy, by mitigating paradoxical c-Raf activation. These 2018 results, with updates through the 2020s confirming durable benefits, highlight how dual inhibition addresses c-Raf-dependent resistance in BRAF V600-mutant settings. A major clinical challenge is to Raf-targeted therapies, often arising from feedback reactivation of c-Raf dimers following B-Raf inhibition, a mechanism identified in the 2010s that sustains MAPK signaling in and other cancers. RAS mutations further predict poor response to c-Raf inhibitors by enhancing wild-type Raf dimerization and pathway reactivation, limiting efficacy in RAS-mutant tumors. Emerging strategies as of 2025 target complexes that regulate c-Raf activation, with structural studies revealing potential disruption sites to overcome dimer-mediated . Ongoing preclinical efforts include development of imaging probes for pan-Raf kinases to better select patients and monitor therapeutic engagement, as well as type I pan-RAF inhibitors like ELV-3111 that combine safely with MEK inhibitors for enhanced activity in NRAS and BRAF mutant cancers.

Protein Interactions

Core Binding Partners

c-Raf, also known as Raf-1, interacts directly with members of the family of small , including H-Ras, K-Ras, and N-Ras, primarily through its Ras-binding domain (RBD) within the conserved region 1 (CR1). These GTP-bound Ras proteins bind to the RBD with high affinity, characterized by a (Kd) of approximately 20 nM for H-Ras, facilitating the recruitment and translocation of cytosolic c-Raf to the plasma membrane where activation occurs. This interaction is essential for c-Raf's membrane localization and subsequent signaling, and the Ras-c-Raf interface has been extensively studied in over 100 publications, highlighting key residues such as Ras Q61 and Y32 that contribute to specificity and binding stability. 14-3-3 proteins, particularly isoforms σ and ζ, bind to phosphorylated residues on c-Raf, including serine 259 (S259) and serine 621 (S621), thereby inhibiting its kinase activity by maintaining an autoinhibited conformation. This was first identified in the mid-1990s through two-hybrid screens and co-immunoprecipitation assays, revealing that 14-3-3 with the N-terminal regulatory of c-Raf prevents premature activation and promotes cytoplasmic retention. Pulldown experiments from that era confirmed the phosphorylation-dependent nature of these interactions, with S259 phosphorylation by kinases like AKT enhancing 14-3-3 to block Ras recruitment. As the primary substrate of c-Raf, MEK1 and MEK2 interact via a docking motif known as the D- on MEK, which engages the domain of c-Raf to position it for efficient at activation sites S217 and S221. The Michaelis constant () for this reaction is approximately 0.5-1 μM, reflecting moderate substrate affinity that supports rapid signal propagation in the MAPK pathway. Structural studies have delineated the D- as a basic stretch of residues on MEK that binds an acidic patch on c-Raf, ensuring specificity in the kinase-substrate complex. c-Raf stability is maintained through interaction with the chaperone , mediated by the co-chaperone Cdc37, which binds the kinase domain of immature c-Raf to prevent and facilitate maturation. Co-immunoprecipitation studies have demonstrated that disruption of the -Cdc37-c-Raf leads to ubiquitination and proteasomal of c-Raf, underscoring 's role in stabilizing the protein under physiological conditions. This binary interaction, often observed in early biosynthetic stages, ensures proper folding without directly impacting catalytic activity once c-Raf is fully assembled.

Regulatory Complexes

c-Raf, also known as RAF1, is regulated through dynamic protein complexes that control its activation, localization, and signaling output in the MAPK/ERK pathway. These complexes primarily involve upstream activators like , scaffolding proteins such as KSR, regulatory adapters like 14-3-3, and inhibitory factors including RKIP, which collectively modulate c-Raf's autoinhibited state, membrane recruitment, and dimerization. Structural studies have revealed that c-Raf's N-terminal regulatory region, comprising the and cysteine-rich domain (CRD), interacts with these partners to relieve intramolecular inhibition and facilitate domain activation. A central regulatory complex forms upon RAS-GTP binding to c-Raf's , which recruits c-Raf to the plasma membrane and disrupts autoinhibitory interactions between the N-terminal region and the . This -c-Raf engagement is stabilized by the CRD's membrane interactions and is essential for subsequent dimerization with other RAF isoforms or KSR. Cryo-EM structures show that in the autoinhibited state, binding initiates a conformational shift, exposing sites for and enabling c-Raf's transition to an open ready for . Additionally, the scaffold protein KSR1 assembles a multiprotein complex including c-Raf, MEK, and ERK, promoting efficient ; KSR1's pseudokinase heterodimerizes with c-Raf in a side-to-side manner, allosterically positioning c-Raf's catalytic αC for MEK . The 14-3-3 proteins form a critical regulatory complex with c-Raf by binding phosphorylated serine residues (pSer259 in the N-region and pSer621 in the kinase domain), stabilizing the autoinhibited monomer and preventing premature dimerization. Upon signaling, dephosphorylation of pSer259 by the SHOC2-MRAS-PP1C phosphatase complex releases this inhibition, allowing 14-3-3 to reorient and bridge two c-Raf protomers via their pSer621 sites, thereby promoting active back-to-back dimers. This 14-3-3-mediated dimerization is further supported by N-terminal phosphorylation (e.g., at Ser338 and Tyr341 by PAK or Src kinases), which enhances complex stability and c-Raf activity. In parallel, the chaperone complex HSP90-CDC37 binds c-Raf to maintain its folding and stability, while associated PP5 dephosphorylates regulatory sites post-activation to facilitate signal termination. Inhibitory regulatory complexes also play key roles; for instance, Inhibitory Protein (RKIP) binds the N-terminal region of c-Raf (residues ~Tyr340-Lys615), forming a stable complex via hydrogen bonds and hydrophobic interactions at hotspots like Tyr340, Arg398, and RKIP's Lys80 and Trp84. This binding prevents c-Raf at activating sites (Ser338, Tyr340/341) and disrupts c-Raf-MEK , thereby suppressing MAPK signaling. simulations indicate a binding free energy of approximately -174 kJ/mol for the RKIP-c-Raf complex, underscoring its potency. Scaffolds like CNK1 can either enhance or inhibit c-Raf activation depending on context, often by modulating Src-mediated within these assemblies. Overall, these complexes ensure precise spatiotemporal control of c-Raf, integrating positive and negative inputs to fine-tune cellular responses.

References

  1. [1]
    Raf Kinases: Function, Regulation and Role in Human Cancer - PMC
    In this review, we focus on the physiological roles of Raf kinases in normal and disease conditions, specifically cancer, and the current thoughts on Raf ...
  2. [2]
    RAF protein-serine/threonine kinases: structure and regulation
    Aug 27, 2010 · A-RAF, B-RAF, and C-RAF are a family of three protein-serine/threonine kinases that participate in the RAS-RAF-MEK-ERK signal transduction cascade.
  3. [3]
    Discovery of Raf Family Is a Milestone in Deciphering the Ras ... - PMC
    The Ras-Raf-MEK-ERK signaling pathway, the first well-established MAPK pathway, plays essential roles in cell proliferation, survival, differentiation and ...Missing: temperature- | Show results with:temperature-
  4. [4]
    The complete coding sequence of the human raf oncogene ... - NIH
    Abstract. The complete 648 amino acid sequence of the human raf oncogene was deduced from the 2977 nucleotide sequence of a fetal liver cDNA.Missing: et al. title
  5. [5]
    Retroviral Oncogenes: A Historical Primer - PMC - PubMed Central
    Retroviruses are the original source of oncogenes. The discovery and characterization of these genes were made possible by the introduction of quantitative cell ...
  6. [6]
    Involvement of the RAF1 locus, at band 3p25, in the 3p ... - PubMed
    The cytogenetic and RFLP data suggest that the RAF1 locus at 3p25 is involved in the chromosomal deletion of SCLC. Publication types. Comparative Study ...Missing: mapped early 1990s
  7. [7]
    Mechanistic principles of RAF kinase signaling - PMC - NIH
    The RAF family of kinases are key components acting downstream of receptor tyrosine kinases and cells employ several distinct mechanisms to strictly control ...
  8. [8]
    KRAS interaction with RAF1 RAS-binding domain and cysteine-rich ...
    Feb 19, 2021 · The N-terminal CR1 region contains the RAS-binding domain (RBD) and the cysteine-rich domain (CRD), the CR2 region contains a 14-3-3 recognition ...
  9. [9]
    Structural Determinants of Ras-Raf Interaction Analyzed in Live Cells
    The sequence responsible for the interaction of Raf-1 with the GTP-bound form of Ras (Ras binding domain, RBD) has been narrowed to residues 51–131 ... c-Raf as a ...
  10. [10]
    B- and C-RAF Display Essential Differences in Their Binding to Ras
    Both RAF kinases co-localized with H-Ras12V at the plasma membrane, with C-RAF exhibiting a considerably higher degree of co-localization with farnesylated ...
  11. [11]
    Structural snapshots of RAF kinase interactions - Portland Press
    Human RAF paralogs share evolutionarily conserved regions (Figure 1), which are functionally split into a regulatory N-terminal half, comprising a RAS-binding ...Ras-Binding Domains · Cysteine-Rich Domain · Catalytic Kinase Domain<|control11|><|separator|>
  12. [12]
    Positive and negative regulation of Raf kinase activity and function ...
    Activating and inhibitory phosphorylation mechanisms play an essential role in regulating Raf kinase activity. Here we demonstrate that phosphorylation of C-Raf ...
  13. [13]
    Cryo-EM structures of CRAF/MEK1/14-3-3 complexes in ... - Nature
    Sep 1, 2025 · CRAF (RAF1) is one of three RAF-family kinases that initiate MAP kinase signaling in response to activated RAS and is essential for ...
  14. [14]
    Autoinhibition in Ras effectors Raf, PI3Kα, and RASSF5
    Sep 29, 2018 · The absence of a crystal (or NMR) structure of Raf's autoinhibited conformation suggests an ensemble of “closed” monomeric states with the N- ...
  15. [15]
    Structural insights into the BRAF monomer-to-dimer transition ...
    Jan 25, 2022 · For example, the published cryo-EM structure of an autoinhibited, monomeric BRAF complex confirmed that a 14-3-3 dimer can bind simultaneously ...
  16. [16]
    Structural dynamics of RAF1-HSP90-CDC37 and HSP90 complexes ...
    Mar 2, 2024 · Our quantitative analysis reveals that CDC37 bridges the HSP90-RAF1 interaction, RAF1 binds HSP90 asymmetrically, and that HSP90 structural elements engage RAF ...
  17. [17]
    Raf1 Antibody | Biorbyt
    E. coli-derived human Raf1 recombinant protein (Position: Y364-Q451). Human Raf1 shares 98.9% amino acid (aa) sequence identity with both mouse and rat Raf1.
  18. [18]
    Novel raf kinase protein–protein interactions found by an exhaustive ...
    This C-terminus has 96% identity among human isoforms and 95% identity between human and mouse enzymes. In contrast, the similarity among Raf isoforms in their ...
  19. [19]
    [PDF] Genetic and molecular analysis of Draf conserved regions in ...
    Draf shares 40% amino acid sequence identity with human Raf-1 and human Raf-. 1 can substitute for Draf in both Torso (Baek et al. 1996) and DER (Brand and ...
  20. [20]
    C. elegans lin-45 raf gene participates in let-60 ras-stimulated vulval ...
    May 13, 1993 · The lin-45 gene encodes a homologue of the raf family of serine/threonine kinases and is necessary for vulval differentiation.Missing: identity | Show results with:identity
  21. [21]
    Craf-1 protein kinase is essential for mouse development
    We report here that Craf-1 is essential for mouse development, as its mutation results in embryonic lethality.
  22. [22]
    Role of the EGFR/Ras/Raf pathway in specification of photoreceptor ...
    Apr 1, 2001 · ABSTRACT. The Drosophila EGF receptor is required for differentiation of many cell types during eye development.
  23. [23]
    The first eukaryotic kinome tree illuminates the dynamic history of ...
    Jan 31, 2020 · Fourteen human kinases with unresolved placement in the human kinome tree were found to originate from three known ePK superfamilies. Further ...
  24. [24]
    RAF1 Raf-1 proto-oncogene, serine/threonine kinase [ (human)]
    Sep 27, 2025 · This gene is the cellular homolog of viral raf gene (v-raf). The encoded protein is a MAP kinase kinase kinase (MAP3K), which functions downstream of the Ras ...
  25. [25]
    BRAF B-Raf proto-oncogene, serine/threonine kinase [ (human)]
    Sep 27, 2025 · This gene encodes a protein belonging to the RAF family of serine/threonine protein kinases. This protein plays a role in regulating the MAP kinase/ERK ...
  26. [26]
    ARAF A-Raf proto-oncogene, serine/threonine kinase [ (human)]
    Enables protein serine/threonine kinase activity. Involved in negative regulation of apoptotic process; regulation of TOR signaling; ...Missing: chromosome | Show results with:chromosome
  27. [27]
    Inhibition of RAF dimers: it takes two to tango - Portland Press
    Dec 24, 2020 · R. (. 2005. ) Mutations of C-RAF are rare in human cancer because C-RAF has a low basal kinase activity compared with B-RAF . Cancer Res. 65.
  28. [28]
    Discovery of Raf Family Is a Milestone in Deciphering the Ras ...
    Regulation of Raf Kinase by Phosphorylation. The intrinsic kinase activity of C-Raf and A-Raf is tightly controlled until they are transported to the plasma ...
  29. [29]
    ARAF - Serine/threonine-protein kinase A-Raf - UniProt
    Involved in the transduction of mitogenic signals from the cell membrane to the nucleus. May also regulate the TOR signaling cascade.
  30. [30]
    Conferring specificity on the ubiquitous Raf/MEK signalling pathway
    ... A-Raf and B-Raf, feature a more restricted expression. A-Raf is mainly expressed in urogenital tissues and B-Raf expression is highest in neuronal tissues ...
  31. [31]
    Central role for BRAF in cardiac hypertrophy - Portland Press
    Jan 18, 2023 · Heart targeted knockout studies have implicated BRAF in pathological hypertrophy and interstitial fibrosis. These studies were performed on male ...
  32. [32]
    Ras-Mediated Activation of the Raf Family Kinases - PMC
    The Raf kinases interact directly with active, membrane-localized Ras, and this interaction is often the first step in the Raf activation process.B-Raf--Mek1 Complex · The Raf Activation Process · Raf Dimerization And...
  33. [33]
    Activation of c-Raf-1 by Ras and Src through different mechanisms
    c-Raf-1 binds to the proto-oncogene Ras in a GTP-dependent manner, but the exact mechanism of activation of c-Raf-1 by Ras is still unclear. We have established ...
  34. [34]
    [PDF] Kinase suppressor of Ras (KSR) Is a scaffold which facilitates ...
    Kinase suppressor of Ras (KSR) is a scaffold that facilitates mitogen-activated protein kinase activation and enhances signaling in the Ras/MAPK pathway.
  35. [35]
    KSR is a scaffold required for activation of the ERK/MAPK module
    KSR was originally identified in RAS-dependent genetic screens inDrosophila and C. elegans (Kornfeld et al. 1995;Sundaram and Han 1995; Therrien et al. 1995).
  36. [36]
    Allosteric Activation of Functionally Asymmetric RAF Kinase Dimers
    Here we show that the dimer is functionally asymmetric with one kinase functioning as an activator to stimulate activity of the partner, receiver kinase.
  37. [37]
    14-3-3 Antagonizes Ras-Mediated Raf-1 Recruitment to the Plasma ...
    We show that 14-3-3 binding, rather than Raf-1 phosphorylation, is required for the correct regulation of kinase activity.
  38. [38]
    Cyclic AMP-dependent kinase regulates Raf-1 kinase mainly by ...
    Serine 338 phosphorylation of a RafS259A mutant was unaffected by PKA. Using RafS259 mutants we also demonstrate that Raf-1 is the sole target for PKA ...
  39. [39]
    Article Regulation of Raf-1 by Direct Feedback Phosphorylation
    Through a negative feedback mechanism involving ERK, Raf-1 is phosphorylated ... Similarly, Raf-1 activity is elevated in cells overexpressing IMP, an inhibitor ...Article · Results · Raf-1 Inactivation By...
  40. [40]
    Dephosphorylation of Ser-259 regulates Raf-1 membrane association
    We show that mitogens stimulate Ser-259 dephosphorylation and Raf-1/protein phosphatase 2A association concomitantly with membrane accumulation and activation ...Missing: S259 T401 PP2A
  41. [41]
    Signaling dynamics of the KSR1 scaffold complex - PNAS
    Jul 7, 2009 · We also find that docking of active ERK to the KSR1 scaffold allows ERK to phosphorylate KSR1 and B-Raf on feedback S/TP sites. Strikingly, ...Missing: fibroblasts | Show results with:fibroblasts
  42. [42]
    Strong negative feedback from Erk to Raf confers robustness to ...
    May 24, 2011 · Feedback acts on c‐Raf. (A) Phosphorylation of Erk‐specific phosphorylation sites in Raf‐1 changes strongly after Mek inhibition. HT29 and ...Missing: T401 KSR
  43. [43]
    CRAF Autophosphorylation of Serine 621 Is Required to Prevent Its ...
    Sep 26, 2008 · The degradation of misfolded CRAF is thought to occur by the recruitment of the E3 ubiquitin ligase CHIP (C terminus of Hsp70-interacting ...Results · Kinase-Inactive Craf Is... · Phosphorylation Of S621 Is...
  44. [44]
    Enzymatic characteristics of the c-Raf-1 protein kinase. - PNAS
    The Km values of c-Raf-1 for ATP and MAPKK were 11.6 microM and 0.8 microM, respectively, and the stoichiometry of phosphorylation of MAPKK by c-Raf-1 was 1.67 ...
  45. [45]
    ROLES OF THE RAF/MEK/ERK PATHWAY IN CELL GROWTH ...
    Raf, either through downstream MEK and ERK, or independently of MEK and ERK, can induce the phosphorylation of proteins, which control apoptosis. Additional ...
  46. [46]
    Targeting the RAS/RAF/MAPK pathway for cancer therapy - Nature
    Dec 18, 2023 · The inhibition of BRAF or CRAF, together with ATG7 inhibition, was found to be a viable treatment strategy for RAS-driven tumors. Understanding ...<|separator|>
  47. [47]
    Kinetics of receptor tyrosine kinase activation define ERK signaling ...
    In comparing responses of PC12 cells to epidermal growth factor (EGF) and nerve growth factor (NGF), transient activation of ERK was associated with ...
  48. [48]
    Mathematical Models of Protein Kinase Signal Transduction
    We have developed a mathematical theory that describes the regulation of signaling pathways as a function of a limited number of key parameters.
  49. [49]
    Regulation of BAD phosphorylation at serine 112 by the ... - PubMed
    Nov 18, 1999 · We demonstrate that activated Ras and Raf, which are upstream of mitogen-activated protein kinases (MAPK), stimulate selective phosphorylation of BAD at Ser- ...
  50. [50]
    14-3-3 Proteins and Survival Kinases Cooperate to Inactivate BAD ...
    Phosphorylation of BAD at Ser-112 and Ser-136 ... BAD and various phosphorylation site mutants to induce apoptosis in cultured cerebellar granule neurons.
  51. [51]
    Raf-1 regulates Rho signaling and cell migration
    Raf kinases relay signals inducing proliferation, differentiation, and survival. The Raf-1 isoform has been extensively studied as the upstream kinase link.
  52. [52]
    c-Raf promotes angiogenesis during normal growth plate maturation
    Summary: The chondrocyte-specific ablation of c-Raf in mice results in a delay in vascular invasion and growth plate maturation due to increased.
  53. [53]
    Critical role for NF‐κB‐induced JunB in VEGF regulation and tumor ...
    Regulation of vascular endothelial growth factor (VEGF) expression is a complex process involving a plethora of transcriptional regulators.Missing: Raf crosstalk
  54. [54]
    AICAR positively regulate glycogen synthase activity and LDL ...
    The signal connection between AICAR and GSK-3 is indirect and involves activation of Raf-1/MEK/p42/44MAPK/p90RSK signaling cascade as pharmacologic inhibition ...
  55. [55]
    Germline gain-of-function mutations in RAF1 cause Noonan syndrome
    Jul 1, 2007 · Our findings implicate RAF1 gain-of-function mutations as a causative agent of a human developmental disorder, representing a new genetic mechanism.Missing: S257L 2006
  56. [56]
    PTPN11, SOS1, KRAS, and RAF1 gene analysis, and genotype ...
    Dec 1, 2008 · After 2006, germline mutations in the KRAS, SOS1, and RAF1 genes were reported to cause Noonan syndrome (NS), in addition to the PTPN11 gene ...
  57. [57]
    Molecular and clinical analysis of RAF1 in Noonan syndrome and ...
    In the current study, we identified eight RAF1 mutations in 18 of 119 patients with NS and related conditions without mutations in known genes. We summarized ...Missing: S257L 2006
  58. [58]
    Cardiofaciocutaneous Syndrome - GeneReviews® - NCBI Bookshelf
    Jan 18, 2007 · The frequency of seizures is higher among individuals with BRAF-related CFC syndrome ... BRAF and MEK mutations in cardio-facio-cutaneous syndrome ...Diagnosis · Clinical Characteristics · Differential Diagnosis · Management
  59. [59]
    Review Article Noonan syndrome-causing genes: Molecular update ...
    RAF1 ([OMIM 164760 ]) mutations cause 3–17% of Noonan syndrome cases. RAF1 consists of 17 exons coding for multidomain protein that act as a serine–threonine ...
  60. [60]
    Sorafenib inhibits LPS-induced inflammation by regulating Lyn ...
    Jun 9, 2022 · Lyn kinase controls TLR4-dependent IKK and MAPK activation ... Role of Raf kinase in cancer: therapeutic potential of targeting the Raf ...
  61. [61]
    p38γ and p38δ kinases regulate the Toll-like receptor 4 (TLR4) - PNAS
    TLR4 stimulation of myeloid cells by LPS activates all three major MAPK pathways, c-Jun N-terminal kinase (JNK), p38α, and ERK1/2, as well as the canonical NF- ...
  62. [62]
    Activation of Ras-ERK Signaling and GSK-3 by Amyloid Precursor ...
    Mar 27, 2017 · In addition to enhanced Ras-ERK signaling and GSK-3 activation, neurons showed enhanced phosphorylation of APP and tau upon Aβ treatment, ...
  63. [63]
    Sorafenib | Raf inhibitor | Mechanism - Selleck Chemicals
    Sorafenib is a multikinase inhibitor of Raf-1 and B-Raf with IC50 of 6 nM and 22 nM in cell-free assays, respectively. Sorafenib inhibits VEGFR-2, VEGFR-3, ...
  64. [64]
    [PDF] Pharmacology Review(s) - accessdata.fda.gov
    Oct 28, 2005 · Sorafenib was initially developed as CRAF kinase inhibitor. However, it was later shown that sorafenib could inhibit other kinases as well (15).
  65. [65]
    PLX4032, a selective BRAFV600E kinase inhibitor, activates the ...
    PLX4032 is a BRAF V600K kinase inhibitor that has demonstrated encouraging responses in current Phase I/II clinical trials.
  66. [66]
    BI-3406, a Potent and Selective SOS1-KRAS Interaction ... - PubMed
    Aug 19, 2020 · BI-3406 inhibits SOS1, preventing its interaction with KRAS, reducing GTP-loaded RAS, and limiting cancer cell proliferation. It also enhances ...Missing: allosteric | Show results with:allosteric
  67. [67]
    BI-3406, a potent and selective SOS1::KRAS interaction inhibitor, is ...
    The previously described catalytic RAS interaction site (dark red; PDB: 1NVU) and the allosteric site (green) are highlighted. The enlarged area depicts the key ...
  68. [68]
    BRD4 PROTAC degrader ARV-825 inhibits T-cell acute ... - PubMed
    Apr 22, 2021 · The BRD4 degrader ARV-825 can effectively suppress the proliferation and promote apoptosis of T-ALL cells via BET protein depletion and c-Myc inhibition.Missing: Raf 2016
  69. [69]
  70. [70]
    Sorafenib in Advanced Hepatocellular Carcinoma
    Jul 24, 2008 · The estimated rate of progression-free survival at 4 months was 62% in the sorafenib group and 42% in the placebo group. Response Rates and ...Missing: FDA | Show results with:FDA
  71. [71]
    Sorafenib for the Treatment of Advanced Renal Cell Carcinoma
    Dec 22, 2006 · On December 20, 2005, the Food and Drug Administration (FDA) granted regular marketing approval for sorafenib tosylate 200-mg tablets (Nexavar, ...Missing: IC50 | Show results with:IC50
  72. [72]
    Preclinical Evaluation of a Radiolabeled Pan-RAF Inhibitor for RAF ...
    Sep 20, 2024 · We sought to develop an 18F-labeled molecular probe from LXH-254 for PET imaging of tumors overexpressing RAF to noninvasively screen patients ...
  73. [73]
    Differential interaction of the ras family GTP-binding proteins H-Ras ...
    Interestingly, whereas H-Ras binds with high affinity (KD = 20 nM) to Raf ... Proto-Oncogene Proteins c-raf; ral GTP-Binding Proteins; rap GTP-Binding ...
  74. [74]
    Interaction of the Protein Kinase Raf-1 with 14-3-3 Proteins - Science
    Optimal complex formation required the amino-terminal regulatory domain of Raf-1. The association of 14-3-3 proteins and Raf-1 was not substantially affected by ...Missing: seminal 1990s
  75. [75]
    Ras binding opens c‐Raf to expose the docking site for mitogen ...
    Introduction. c‐Raf serine‐threonine kinase regulates various aspects of cellular function, including the proliferation, differentiation and oncogenic ...
  76. [76]
    p50(cdc37) acting in concert with Hsp90 is required for Raf-1 function
    In this study, we examined the role of p50(cdc37) and its Hsp90 chaperone partner in Raf/Mek/MAPK signaling biochemically.
  77. [77]
    Regulation of RAF family kinases: new insights
    May 2, 2024 · This review will focus on recent structural and biochemical studies that have provided 'snapshots' into the RAF regulatory cycle.
  78. [78]
    Exploring the Binding Interaction of Raf Kinase Inhibitory Protein ...
    RKIP interferes with the RAF/MEK/ERK signaling cascade by endogenously binding with C-Raf (Raf-1 kinase) and preventing its activation.
  79. [79]
    “RAF” neighborhood: Protein–protein interaction in the Raf/Mek/Erk ...
    Jun 14, 2014 · This specific role of C-Raf is not mediated through Erk phosphorylation, which is similar in B- or C-Raf-deficient epidermis and is necessary, ...