BRAF (gene)

Last updated
BRAF
Protein BRAF PDB 1uwh.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases BRAF , B-RAF1, BRAF1, NS7, RAFB1, B-Raf, B-Raf proto-oncogene, serine/threonine kinase
External IDs OMIM: 164757 MGI: 88190 HomoloGene: 3197 GeneCards: BRAF
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_004333
NM_001354609
NM_001374244
NM_001374258

NM_139294

RefSeq (protein)

NP_647455

Location (UCSC) Chr 7: 140.72 – 140.92 Mb Chr 6: 39.58 – 39.7 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

BRAF is a human gene that encodes a protein called B-Raf. The gene is also referred to as proto-oncogene B-Raf and v-Raf murine sarcoma viral oncogene homolog B, while the protein is more formally known as serine/threonine-protein kinase B-Raf. [5] [6]

The B-Raf protein is involved in sending signals inside cells which are involved in directing cell growth. In 2002, it was shown to be mutated in some human cancers. [7]

Certain other inherited BRAF mutations cause birth defects.

Drugs that treat cancers driven by BRAF mutations have been developed. Two of these drugs, vemurafenib [8] and dabrafenib are approved by FDA for treatment of late-stage melanoma. Vemurafenib was the first approved drug to come out of fragment-based drug discovery. [9]

Function

Overview of signal transduction pathways involved in apoptosis. The role of Raf proteins like B-Raf is indicated in the center. Signal transduction pathways.svg
Overview of signal transduction pathways involved in apoptosis. The role of Raf proteins like B-Raf is indicated in the center.

B-Raf is a member of the Raf kinase family of growth signal transduction protein kinases. This protein plays a role in regulating the MAP kinase/ERKs signaling pathway, which affects cell division, differentiation, and secretion. [10]

Structure

B-Raf is a 766-amino acid, regulated signal transduction serine/threonine-specific protein kinase. Broadly speaking, it is composed of three conserved domains characteristic of the Raf kinase family: conserved region 1 (CR1), a Ras-GTP-binding [11] self-regulatory domain, conserved region 2 (CR2), a serine-rich hinge region, and conserved region 3 (CR3), a catalytic protein kinase domain that phosphorylates a consensus sequence on protein substrates. [12] In its active conformation, B-Raf forms dimers via hydrogen-bonding and electrostatic interactions of its kinase domains. [13]

CR1

Conserved region 1 (CR1) autoinhibits B-Raf's kinase domain (CR3) so that B-Raf signaling is regulated rather than constitutive. [12] Residues 155–227 [14] make up the Ras-binding domain (RBD), which binds to Ras-GTP's effector domain to release CR1 and halt kinase inhibition. Residues 234–280 comprise a phorbol ester/DAG-binding zinc finger motif that participates in B-Raf membrane docking after Ras-binding. [14] [15]

CR2

Conserved region 2 (CR2) provides a flexible linker that connects CR1 and CR3 and acts as a hinge.[ citation needed ]

CR3

Figure 1: Inactive conformation of B-Raf kinase (CR3) domain. P-Loop (orange) hydrophobic interactions with activation loop (gray) residues that stabilize the inactive kinase conformation are shown with sticks. F595 (red) blocks the hydrophobic pocket where the ATP adenine binds (yellow). D576 (orange) is shown as part of the catalytic loop (magenta). Figure modified from PDB id 1UWH. BRAF Kinase Inactive.png
Figure 1: Inactive conformation of B-Raf kinase (CR3) domain. P-Loop (orange) hydrophobic interactions with activation loop (gray) residues that stabilize the inactive kinase conformation are shown with sticks. F595 (red) blocks the hydrophobic pocket where the ATP adenine binds (yellow). D576 (orange) is shown as part of the catalytic loop (magenta). Figure modified from PDB id 1UWH.

Conserved region 3 (CR3), residues 457–717, [14] makes up B-Raf's enzymatic kinase domain. This largely conserved structure [16] is bi-lobal, connected by a short hinge region. [17] The smaller N-lobe (residues 457–530) is primarily responsible for ATP binding while the larger C-lobe (residues 535–717) binds substrate proteins. [16] The active site is the cleft between the two lobes, and the catalytic Asp576 residue is located on the C-lobe, facing the inside of this cleft. [14] [16]

Subregions

P-Loop

The P-loop of B-Raf (residues 464–471) stabilizes the non-transferable phosphate groups of ATP during enzyme ATP-binding. Specifically, S467, F468, and G469 backbone amides hydrogen-bond to the β-phosphate of ATP to anchor the molecule. B-Raf functional motifs have been determined by analyzing the homology of PKA analyzed by Hanks and Hunter to the B-Raf kinase domain. [16]

Nucleotide-binding pocket

V471, C532, W531, T529, L514, and A481 form a hydrophobic pocket within which the adenine of ATP is anchored through Van der Waals attractions upon ATP binding. [16] [18]

Catalytic loop

Residues 574–581 compose a section of the kinase domain responsible for supporting the transfer of the γ-phosphate of ATP to B-Raf's protein substrate. In particular, D576 acts as a proton acceptor to activate the nucleophilic hydroxyl oxygen on substrate serine or threonine residues, allowing the phosphate transfer reaction to occur mediated by base-catalysis. [16]

DFG motif

D594, F595, and G596 compose a motif central to B-Raf's function in both its inactive and active state. In the inactive state, F595 occupies the nucleotide-binding pocket, prohibiting ATP from entering and decreasing the likelihood of enzyme catalysis. [13] [18] [19] In the active state, D594 chelates the divalent magnesium cation that stabilizes the β- and γ-phosphate groups of ATP, orienting the γ-phosphate for transfer. [16]

Activation loop

Residues 596–600 form strong hydrophobic interactions with the P-loop in the inactive conformation of the kinase, locking the kinase in its inactive state until the activation loop is phosphorylated, destabilizing these interactions with the presence of negative charge. This triggers the shift to the active state of the kinase. Specifically, L597 and V600 of the activation loop interact with G466, F468, and V471 of the P-loop to keep the kinase domain inactive until it is phosphorylated. [17]

Enzymology

B-Raf is a serine/threonine-specific protein kinase. As such, it catalyzes the phosphorylation of serine and threonine residues in a consensus sequence on target proteins by ATP, yielding ADP and a phosphorylated protein as products. [16] Since it is a highly regulated signal transduction kinase, B-Raf must first bind Ras-GTP before becoming active as an enzyme. [15] Once B-Raf is activated, a conserved protein kinase catalytic core phosphorylates protein substrates by promoting the nucleophilic attack of the activated substrate serine or threonine hydroxyl oxygen atom on the γ-phosphate group of ATP through bimolecular nucleophilic substitution. [16] [20] [21] [22]

Activation

Relieving CR1 autoinhibition

The kinase (CR3) domain of human Raf kinases is inhibited by two mechanisms: autoinhibition by its own regulatory Ras-GTP-binding CR1 domain and a lack of post-translational phosphorylation of key serine and tyrosine residues (S338 and Y341 for c-Raf) in the CR2 hinge region. During B-Raf activation, the protein's autoinhibitory CR1 domain first binds Ras-GTP's effector domain to the CR1 Ras-binding domain (RBD) to release the kinase CR3 domain like other members of the human Raf kinase family. The CR1-Ras interaction is later strengthened through the binding of the cysteine-rich subdomain (CRD) of CR1 to Ras and membrane phospholipids. [12] Unlike A-Raf and C-Raf, which must be phosphorylated on hydroxyl-containing CR2 residues before fully releasing CR1 to become active, B-Raf is constituitively phosphorylated on CR2 S445. [23] This allows the negatively charged phosphoserine to immediately repel CR1 through steric and electrostatic interactions once the regulatory domain is unbound, freeing the CR3 kinase domain to interact with substrate proteins.

CR3 domain activation

After the autoinhibitory CR1 regulatory domain is released, B-Raf's CR3 kinase domain must change to its ATP-binding active conformer before it can catalyze protein phosphorylation. In the inactive conformation, F595 of the DFG motif blocks the hydrophobic adenine binding pocket while activation loop residues form hydrophobic interactions with the P-loop, stopping ATP from accessing its binding site. When the activation loop is phosphorylated, the negative charge of the phosphate is unstable in the hydrophobic environment of the P-loop. As a result, the activation loop changes conformation, stretching out across the C-lobe of the kinase domain. In this process, it forms stabilizing β-sheet interactions with the β6 strand. Meanwhile, the phosphorylated residue approaches K507, forming a stabilizing salt bridge to lock the activation loop into place. The DFG motif changes conformation with the activation loop, causing F595 to move out of the adenine nucleotide binding site and into a hydrophobic pocket bordered by the αC and αE helices. Together, DFG and activation loop movement upon phosphorylation open the ATP binding site. Since all other substrate-binding and catalytic domains are already in place, phosphorylation of the activation loop alone activates B-Raf's kinase domain through a chain reaction that essentially removes a lid from an otherwise-prepared active site. [17]

Mechanism of catalysis

Figure 2: Base-catalyzed nucleophilic attack of a serine/threonine substrate residue on the g-phosphate group of ATP. Step 1: chelation of secondary magnesium ion by N581 and deprotonation of substrate Ser/Thr by D576. Step 2: nucleophilic attack of activated substrate hydroxyl on ATP g-phosphate. Step 3: magnesium complex breaks down and D576 deprotonates. Step 4: release of products. B-Raf Phosphorylation Mechanism.png
Figure 2: Base-catalyzed nucleophilic attack of a serine/threonine substrate residue on the γ-phosphate group of ATP. Step 1: chelation of secondary magnesium ion by N581 and deprotonation of substrate Ser/Thr by D576. Step 2: nucleophilic attack of activated substrate hydroxyl on ATP γ-phosphate. Step 3: magnesium complex breaks down and D576 deprotonates. Step 4: release of products.

To effectively catalyze protein phosphorylation via the bimolecular substitution of serine and threonine residues with ADP as a leaving group, B-Raf must first bind ATP and then stabilize the transition state as the γ-phosphate of ATP is transferred. [16]

ATP binding

B-Raf binds ATP by anchoring the adenine nucleotide in a nonpolar pocket (yellow, Figure 1) and orienting the molecule through hydrogen-bonding and electrostatic interactions with phosphate groups. In addition to the P-loop and DFG motif phosphate binding described above, K483 and E501 play key roles in stabilizing non-transferable phosphate groups. The positive charge on the primary amine of K483 allows it to stabilize the negative charge on ATP α- and β-phosphate groups when ATP binds. When ATP is not present, the negative charge of the E501 carboxyl group balances this charge. [16] [17]

Phosphorylation

Once ATP is bound to the B-Raf kinase domain, D576 of the catalytic loop activates a substrate hydroxyl group, increasing its nucleophilicity to kinetically drive the phosphorylation reaction while other catalytic loop residues stabilize the transition state.(Figure 2). N581 chelates the divalent magnesium cation associated with ATP to help orient the molecule for optimal substitution. K578 neutralizes the negative charge on the γ-phosphate group of ATP so that the activated ser/thr substrate residue won't experience as much electron-electron repulsion when attacking the phosphate. After the phosphate group is transferred, ADP and the new phosphoprotein are released. [16]

Inhibitors

Since constitutively active B-Raf mutants commonly cause cancer (see Clinical Significance) by excessively signaling cells to grow, inhibitors of B-Raf have been developed for both the inactive and active conformations of the kinase domain as cancer therapeutic candidates. [17] [18] [19]

Sorafenib

Figure 3: B-Raf kinase domain locked in the inactive conformation by bound BAY43-9006. Hydrophobic interactions anchor BAY43-9006 in the ATP binding site while urea group hydrogen-bonding traps D594 of the DFG motif. The BAY43-9006 trifluoromethyl phenyl ring further prohibits DFG motif and activation loop movement to the active confermer via steric blockage. Sorafenib BRAF Labeled.png
Figure 3: B-Raf kinase domain locked in the inactive conformation by bound BAY43-9006. Hydrophobic interactions anchor BAY43-9006 in the ATP binding site while urea group hydrogen-bonding traps D594 of the DFG motif. The BAY43-9006 trifluoromethyl phenyl ring further prohibits DFG motif and activation loop movement to the active confermer via steric blockage.

BAY43-9006 (Sorafenib, Nexavar) is a V600E mutant B-Raf and C-Raf inhibitor approved by the FDA for the treatment of primary liver and kidney cancer. Bay43-9006 disables the B-Raf kinase domain by locking the enzyme in its inactive form. The inhibitor accomplishes this by blocking the ATP binding pocket through high-affinity for the kinase domain. It then binds key activation loop and DFG motif residues to stop the movement of the activation loop and DFG motif to the active conformation. Finally, a trifluoromethyl phenyl moiety sterically blocks the DFG motif and activation loop active conformation site, making it impossible for the kinase domain to shift conformation to become active. [17]

The distal pyridyl ring of BAY43-9006 anchors in the hydrophobic nucleotide-binding pocket of the kinase N-lobe, interacting with W531, F583, and F595. The hydrophobic interactions with catalytic loop F583 and DFG motif F595 stabilize the inactive conformation of these structures, decreasing the likelihood of enzyme activation. Further hydrophobic interaction of K483, L514, and T529 with the center phenyl ring increase the affinity of the kinase domain for the inhibitor. Hydrophobic interaction of F595 with the center ring as well decreases the energetic favorability of a DFG conformation switch further. Finally, polar interactions of BAY43-9006 with the kinase domain continue this trend of increasing enzyme affinity for the inhibitor and stabilizing DFG residues in the inactive conformation. E501 and C532 hydrogen bond the urea and pyridyl groups of the inhibitor respectively while the urea carbonyl accepts a hydrogen bond from D594's backbone amide nitrogen to lock the DFG motif in place. [17]

The trifluoromethyl phenyl moiety cements the thermodynamic favorability of the inactive conformation when the kinase domain is bound to BAY43-9006 by sterically blocking the hydrophobic pocket between the αC and αE helices that the DFG motif and activation loop would inhabit upon shifting to their locations in the active conformation of the protein. [17]

Vemurafenib

Figure 4: Structures of Vemurafenib (right) and its precursor, PLX 4720 (left), two inhibitors of the active conformation of the B-Raf kinase domain Vemurafenib.png
Figure 4: Structures of Vemurafenib (right) and its precursor, PLX 4720 (left), two inhibitors of the active conformation of the B-Raf kinase domain

PLX4032 (Vemurafenib) is a V600 mutant B-Raf inhibitor approved by the FDA for the treatment of late-stage melanoma. [13] Unlike BAY43-9006, which inhibits the inactive form of the kinase domain, Vemurafenib inhibits the active "DFG-in" form of the kinase, [18] [19] firmly anchoring itself in the ATP-binding site. By inhibiting only the active form of the kinase, Vemurafenib selectively inhibits the proliferation of cells with unregulated B-Raf, normally those that cause cancer.

Since Vemurafenib only differs from its precursor, PLX4720, in a phenyl ring added for pharmacokinetic reasons, [19] PLX4720's mode of action is equivalent to Vemurafenib's. PLX4720 has good affinity for the ATP binding site partially because its anchor region, a 7-azaindole bicyclic, only differs from the natural adenine that occupies the site in two places where nitrogen atoms have been replaced by carbon. This enables strong intermolecular interactions like N7 hydrogen bonding to C532 and N1 hydrogen bonding to Q530 to be preserved. Excellent fit within the ATP-binding hydrophobic pocket (C532, W531, T529, L514, A481) increases binding affinity as well. Ketone linker hydrogen bonding to water and difluoro-phenyl fit in a second hydrophobic pocket (A481, V482, K483, V471, I527, T529, L514, and F583) contribute to the exceptionally high binding affinity overall. Selective binding to active Raf is accomplished by the terminal propyl group that binds to a Raf-selective pocket created by a shift of the αC helix. Selectivity for the active conformation of the kinase is further increased by a pH-sensitive deprotonated sulfonamide group that is stabilized by hydrogen bonding with the backbone peptide NH of D594 in the active state. In the inactive state, the inhibitor's sulfonamide group interacts with the backbone carbonyl of that residue instead, creating repulsion. Thus, Vemurafenib binds preferentially to the active state of B-Raf's kinase domain. [18] [19]

Clinical significance

Mutations in the BRAF gene can cause disease in two ways. First, mutations can be inherited and cause birth defects. Second, mutations can appear later in life and cause cancer, as an oncogene.

Inherited mutations in this gene cause cardiofaciocutaneous syndrome, a disease characterized by heart defects, mental retardation and a distinctive facial appearance. [24]

Mutations in this gene have been found in cancers, including non-Hodgkin lymphoma, colorectal cancer, malignant melanoma, papillary thyroid carcinoma, non-small-cell lung carcinoma, adenocarcinoma of the lung, brain tumors including glioblastoma and pleomorphic xanthoastrocytoma as well as inflammatory diseases like Erdheim-Chester disease. [10]

The V600E mutation of the BRAF gene has been associated with hairy cell leukemia in numerous studies and has been suggested for use in screening for Lynch syndrome to reduce the number of patients undergoing unnecessary MLH1 sequencing. [25] [26]

Mutants

More than 30 mutations of the BRAF gene associated with human cancers have been identified. The frequency of BRAF mutations varies widely in human cancers, from more than 80% in melanomas and nevi, to as little as 0–18% in other tumors, such as 1–3% in lung cancers and 5% in colorectal cancer. [27] In 90% of the cases, thymine is substituted with adenine at nucleotide 1799. This leads to valine (V) being substituted for by glutamate (E) at codon 600 (now referred to as V600E) in the activation segment that has been found in human cancers. [28] This mutation has been widely observed in papillary thyroid carcinoma, colorectal cancer, melanoma and non-small-cell lung cancer. [29] [30] [31] [32] [33] [34] [35] BRAF-V600E mutation are present in 57% of Langerhans cell histiocytosis patients. [36] The V600E mutation is a likely driver mutation in 100% of cases of hairy cell leukaemia. [37] High frequency of BRAF V600E mutations have been detected in ameloblastoma, a benign but locally infiltrative odontogenic neoplasm. [38] The V600E mutation may also be linked, as a single-driver mutation (a genetic 'smoking gun') to certain cases of papillary craniopharyngioma development. [39]

Other mutations which have been found are R461I, I462S, G463E, G463V, G465A, G465E, G465V, G468A, G468E, G469R, N580S, E585K, D593V, F594L, G595R, L596V, T598I, V599D, V599E, V599K, V599R, V600K, A727V, etc. and most of these mutations are clustered to two regions: the glycine-rich P loop of the N lobe and the activation segment and flanking regions. [17] These mutations change the activation segment from inactive state to active state, for example in the previous cited paper it has been reported that the aliphatic side chain of Val599 interacts with the phenyl ring of Phe467 in the P loop. Replacing the medium-sized hydrophobic Val side chain with a larger and charged residue as found in human cancer(Glu, Asp, Lys, or Arg) would be expected to destabilize the interactions that maintain the DFG motif in an inactive conformation, so flipping the activation segment into the active position. Depending on the type of mutation the kinase activity towards MEK may also vary. Most of the mutants stimulate enhanced B-Raf kinase activity toward MEK. However, a few mutants act through a different mechanism because although their activity toward MEK is reduced, they adopt a conformation that activates wild-type C-RAF, which then signals to ERK.

BRAF-V600E

  • BRAFV600E mutation confers a poor prognosis in metastatic colorectal cancer. Upon targeted inhibition of BRAF and/or EGFR in BRAF V600E colorectal cancer, SRC kinases become activated in a systematic manner. Remarkably, concurrent targeting of SRC alongside BRAF and EGFR has demonstrated increased treatment effectiveness. The compensatory activation of SRC kinases is mediated by an autocrine prostaglandin E2 loop, which can be blocked using cyclooxygenase-2 (COX2) inhibitors. Notably, the simultaneous targeting of COX2 with BRAF and EGFR has shown significant potential in preclinical models, leading to a sustained suppression of tumor growth. [40]
  • BRAF V600E is a determinant of sensitivity to proteasome inhibitors. Vulnerability to proteasome inhibitors is dependent on persistent BRAF signaling, because BRAF-V600E blockade by PLX4720 reversed sensitivity to carfilzomib in BRAF-mutant colorectal cancer cells. Proteasome inhibition might represent a valuable targeting strategy in BRAF V600E-mutant colorectal tumors. [41]

BRAF inhibitors

As mentioned above, some pharmaceutical firms are developing specific inhibitors of mutated B-raf protein for anticancer use because BRAF is a well-understood, high yield target. [18] [42] Vemurafenib (RG7204 or PLX4032) was licensed by the US Food and Drug Administration as Zelboraf for the treatment of metastatic melanoma in August 2011 based on Phase III clinical data. Improved survival was seen, as well as a response rate to treatment of 53%, compared to from 7-12% with the former best chemotherapeutic treatment, dacarbazine. [43] In clinical trials, B-Raf increased metastatic melanoma patient chance of survival. In spite of the drug's high efficacy, 20% of tumors still develop resistance to the treatment. In mice, 20% of tumors become resistant after 56 days. [44] While the mechanisms of this resistance are still disputed, some hypotheses include the overexpression of B-Raf to compensate for high concentrations of Vemurafenib [44] and upstream upregulation of growth signaling. [45]

More general B-Raf inhibitors include GDC-0879, PLX-4720, Sorafenib, dabrafenib and encorafenib.

panRAF inhibitors

Belvarafenib is classified as a panRAF inhibitor. A panRAF inhibitor blocks the catalytic function of both proteins in the dimer. [46]

Interactions

BRAF (gene) has been shown to interact with:

Related Research Articles

<span class="mw-page-title-main">Protein kinase</span> Enzyme that adds phosphate groups to other proteins

A protein kinase is a kinase which selectively modifies other proteins by covalently adding phosphates to them (phosphorylation) as opposed to kinases which modify lipids, carbohydrates, or other molecules. Phosphorylation usually results in a functional change of the target protein (substrate) by changing enzyme activity, cellular location, or association with other proteins. The human genome contains about 500 protein kinase genes and they constitute about 2% of all human genes. There are two main types of protein kinase. The great majority are serine/threonine kinases, which phosphorylate the hydroxyl groups of serines and threonines in their targets. Most of the others are tyrosine kinases, although additional types exist. Protein kinases are also found in bacteria and plants. Up to 30% of all human proteins may be modified by kinase activity, and kinases are known to regulate the majority of cellular pathways, especially those involved in signal transduction.

<span class="mw-page-title-main">Kinase</span> Enzyme catalyzing transfer of phosphate groups onto specific substrates

In biochemistry, a kinase is an enzyme that catalyzes the transfer of phosphate groups from high-energy, phosphate-donating molecules to specific substrates. This process is known as phosphorylation, where the high-energy ATP molecule donates a phosphate group to the substrate molecule. As a result, kinase produces a phosphorylated substrate and ADP. Conversely, it is referred to as dephosphorylation when the phosphorylated substrate donates a phosphate group and ADP gains a phosphate group. These two processes, phosphorylation and dephosphorylation, occur four times during glycolysis.

<span class="mw-page-title-main">Tyrosine kinase</span> Class hi residues

A tyrosine kinase is an enzyme that can transfer a phosphate group from ATP to the tyrosine residues of specific proteins inside a cell. It functions as an "on" or "off" switch in many cellular functions.

<span class="mw-page-title-main">Cyclin-dependent kinase</span> Class of enzymes

Cyclin-dependent kinases (CDKs) are a predominant group of serine/threonine protein kinases involved in the regulation of the cell cycle and its progression, ensuring the integrity and functionality of cellular machinery. These regulatory enzymes play a crucial role in the regulation of eukaryotic cell cycle and transcription, as well as DNA repair, metabolism, and epigenetic regulation, in response to several extracellular and intracellular signals. They are present in all known eukaryotes, and their regulatory function in the cell cycle has been evolutionarily conserved. The catalytic activities of CDKs are regulated by interactions with CDK inhibitors (CKIs) and regulatory subunits known as cyclins. Cyclins have no enzymatic activity themselves, but they become active once they bind to CDKs. Without cyclin, CDK is less active than in the cyclin-CDK heterodimer complex. CDKs phosphorylate proteins on serine (S) or threonine (T) residues. The specificity of CDKs for their substrates is defined by the S/T-P-X-K/R sequence, where S/T is the phosphorylation site, P is proline, X is any amino acid, and the sequence ends with lysine (K) or arginine (R). This motif ensures CDKs accurately target and modify proteins, crucial for regulating cell cycle and other functions. Deregulation of the CDK activity is linked to various pathologies, including cancer, neurodegenerative diseases, and stroke.

c-Raf Mammalian protein found in Homo sapiens

RAF proto-oncogene serine/threonine-protein kinase, also known as proto-oncogene c-RAF or simply c-Raf or even Raf-1, is an enzyme that in humans is encoded by the RAF1 gene. The c-Raf protein is part of the ERK1/2 pathway as a MAP kinase (MAP3K) that functions downstream of the Ras subfamily of membrane associated GTPases. C-Raf is a member of the Raf kinase family of serine/threonine-specific protein kinases, from the TKL (Tyrosine-kinase-like) group of kinases.

<span class="mw-page-title-main">Serine/threonine-specific protein kinase</span> Class of protein kinase enzymes

A serine/threonine protein kinase is a kinase enzyme, in particular a protein kinase, that phosphorylates the OH group of the amino-acid residues serine or threonine, which have similar side chains. At least 350 of the 500+ human protein kinases are serine/threonine kinases (STK).

<span class="mw-page-title-main">Cyclin-dependent kinase inhibitor protein</span> Protein which inhibits cyclin-dependent kinase

A cyclin-dependent kinase inhibitor protein(also known as CKIs, CDIs, or CDKIs) is a protein which inhibits the enzyme cyclin-dependent kinase (CDK) and Cyclin activity by stopping the cell cycle if there are unfavorable conditions, therefore, acting as tumor suppressors. Cell cycle progression is stopped by Cyclin-dependent kinase inhibitor protein at the G1 phase. CKIs are vital proteins within the control system that point out whether the process of DNA synthesis, mitosis, and cytokines control one another. If a malfunction prevents the successful completion of DNA synthesis during the G1 phase, a signal is sent to delay or stop the progression to the S phase. Cyclin-dependent kinase inhibitor proteins are essential in the regulation of the cell cycle. If cell mutations surpass the cell cycle checkpoints during cell cycle regulation, it can result in various types of cancer.

The ErbB family of proteins contains four receptor tyrosine kinases, structurally related to the epidermal growth factor receptor (EGFR), its first discovered member. In humans, the family includes Her1, Her2 (ErbB2), Her3 (ErbB3), and Her4 (ErbB4). The gene symbol, ErbB, is derived from the name of a viral oncogene to which these receptors are homologous: erythroblastic leukemia viral oncogene. Insufficient ErbB signaling in humans is associated with the development of neurodegenerative diseases, such as multiple sclerosis and Alzheimer's disease, while excessive ErbB signaling is associated with the development of a wide variety of types of solid tumor.

The IκB kinase is an enzyme complex that is involved in propagating the cellular response to inflammation, specifically the regulation of lymphocytes.

<span class="mw-page-title-main">Protein kinase domain</span>

The protein kinase domain is a structurally conserved protein domain containing the catalytic function of protein kinases. Protein kinases are a group of enzymes that move a phosphate group onto proteins, in a process called phosphorylation. This functions as an on/off switch for many cellular processes, including metabolism, transcription, cell cycle progression, cytoskeletal rearrangement and cell movement, apoptosis, and differentiation. They also function in embryonic development, physiological responses, and in the nervous and immune system. Abnormal phosphorylation causes many human diseases, including cancer, and drugs that affect phosphorylation can treat those diseases.

A non-receptor tyrosine kinase (nRTK) is a cytosolic enzyme that is responsible for catalysing the transfer of a phosphate group from a nucleoside triphosphate donor, such as ATP, to tyrosine residues in proteins. Non-receptor tyrosine kinases are a subgroup of protein family tyrosine kinases, enzymes that can transfer the phosphate group from ATP to a tyrosine residue of a protein (phosphorylation). These enzymes regulate many cellular functions by switching on or switching off other enzymes in a cell.

The Walker A and Walker B motifs are protein sequence motifs, known to have highly conserved three-dimensional structures. These were first reported in ATP-binding proteins by Walker and co-workers in 1982.

<span class="mw-page-title-main">Vemurafenib</span> Targeted cancer drug

Vemurafenib (INN), sold under the brand name Zelboraf, is a medication used for the treatment of late-stage melanoma. It is an inhibitor of the B-Raf enzyme and was developed by Plexxikon.

Bcr-Abl tyrosine-kinase inhibitors (TKI) are the first-line therapy for most patients with chronic myelogenous leukemia (CML). More than 90% of CML cases are caused by a chromosomal abnormality that results in the formation of a so-called Philadelphia chromosome. This abnormality was discovered by Peter Nowell in 1960 and is a consequence of fusion between the Abelson (Abl) tyrosine kinase gene at chromosome 9 and the break point cluster (Bcr) gene at chromosome 22, resulting in a chimeric oncogene (Bcr-Abl) and a constitutively active Bcr-Abl tyrosine kinase that has been implicated in the pathogenesis of CML. Compounds have been developed to selectively inhibit the tyrosine kinase.

<span class="mw-page-title-main">Balanol</span> Fungal metabolite

Balanol is a fungal metabolite produced by the fungus Verticillium balanoides. It is a potent inhibitor of the serine/threonine kinases protein kinase A (PKA) and protein kinase C (PKC), binding in a similar manner with that of ATP. Balanol was discovered in 1993 in the search for novel inhibitors of PKC, a member of a family of serine/threonine kinases whose overactivation is associated with numerous human diseases of signal transduction including cancer. However, much of the research on balanol focuses on how chemical modifications of the molecular structure affect binding to PKA. Indeed, balanol, its chemically altered analogs, and their interactions with PKA in particular are used to illuminate the roles of selectivity and protein flexibility in the inhibition of kinases. For instance, the X-ray crystal structure of balanol in complex with PKA was used in order to confer selectivity and to improve pharmacological efficacy of inhibitors of the H. sapiens Akt (PKB), another serine/threonine protein kinase implicated in the proper functioning of many cellular processes.

<span class="mw-page-title-main">Autophosphorylation</span>

Autophosphorylation is a type of post-translational modification of proteins. It is generally defined as the phosphorylation of the kinase by itself. In eukaryotes, this process occurs by the addition of a phosphate group to serine, threonine or tyrosine residues within protein kinases, normally to regulate the catalytic activity. Autophosphorylation may occur when a kinases' own active site catalyzes the phosphorylation reaction, or when another kinase of the same type provides the active site that carries out the chemistry. The latter often occurs when kinase molecules dimerize. In general, the phosphate groups introduced are gamma phosphates from nucleoside triphosphates, most commonly ATP.

A MEK inhibitor is a chemical or drug that inhibits the mitogen-activated protein kinase kinase enzymes MEK1 and/or MEK2. They can be used to affect the MAPK/ERK pathway which is often overactive in some cancers.

<span class="mw-page-title-main">Encorafenib</span> Chemical compound

Encorafenib, sold under the brand name Braftovi, is a medication for the treatment of certain melanoma cancers. It is a small molecule BRAF inhibitor that targets key enzymes in the MAPK signaling pathway. This pathway occurs in many different cancers including melanoma and colorectal cancers. The substance was being developed by Novartis and then by Array BioPharma. In June 2018, it was approved by the FDA in combination with binimetinib for the treatment of patients with unresectable or metastatic BRAF V600E or V600K mutation-positive melanoma.

<span class="mw-page-title-main">Binimetinib</span> Chemical compound

Binimetinib, sold under the brand name Mektovi, is an anti-cancer medication used to treat various cancers. Binimetinib is a selective inhibitor of MEK, a central kinase in the tumor-promoting MAPK pathway. Inappropriate activation of the pathway has been shown to occur in many cancers. In June 2018 it was approved by the FDA in combination with encorafenib for the treatment of patients with unresectable or metastatic BRAF V600E or V600K mutation-positive melanoma. In October 2023, it was approved by the FDA for treatment of NSCLC with a BRAF V600E mutation in combination with encorafenib. It was developed by Array Biopharma.

V600E is a mutation of the BRAF gene in which valine (V) is substituted by glutamic acid (E) at amino acid 600. It is a driver mutation in a proportion of certain diagnoses, including melanoma, hairy cell leukemia, papillary thyroid carcinoma, colorectal cancer, non-small-cell lung cancer, Langerhans cell histiocytosis, Erdheim–Chester disease and ameloblastoma.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000157764 Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000002413 Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Sithanandam G, Kolch W, Duh FM, Rapp UR (December 1990). "Complete coding sequence of a human B-raf cDNA and detection of B-raf protein kinase with isozyme specific antibodies". Oncogene. 5 (12): 1775–1780. PMID   2284096.
  6. Sithanandam G, Druck T, Cannizzaro LA, Leuzzi G, Huebner K, Rapp UR (April 1992). "B-raf and a B-raf pseudogene are located on 7q in man". Oncogene. 7 (4): 795–799. PMID   1565476.
  7. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. (June 2002). "Mutations of the BRAF gene in human cancer" (PDF). Nature. 417 (6892): 949–954. Bibcode:2002Natur.417..949D. doi:10.1038/nature00766. PMID   12068308. S2CID   3071547.
  8. "FDA Approves Zelboraf (Vemurafenib) and Companion Diagnostic for BRAF Mutation-Positive Metastatic Melanoma, a Deadly Form of Skin Cancer" (Press release). Genentech. Retrieved 2011-08-17.
  9. Erlanson DA, Fesik SW, Hubbard RE, Jahnke W, Jhoti H (September 2016). "Twenty years on: the impact of fragments on drug discovery". Nature Reviews. Drug Discovery. 15 (9): 605–619. doi:10.1038/nrd.2016.109. PMID   27417849. S2CID   19634793.
  10. 1 2 "Entrez Gene: BRAF".
  11. Daum G, Eisenmann-Tappe I, Fries HW, Troppmair J, Rapp UR (November 1994). "The ins and outs of Raf kinases". Trends in Biochemical Sciences. 19 (11): 474–480. doi:10.1016/0968-0004(94)90133-3. PMID   7855890.
  12. 1 2 3 Cutler RE, Stephens RM, Saracino MR, Morrison DK (August 1998). "Autoregulation of the Raf-1 serine/threonine kinase". Proceedings of the National Academy of Sciences of the United States of America. 95 (16): 9214–9219. Bibcode:1998PNAS...95.9214C. doi: 10.1073/pnas.95.16.9214 . PMC   21318 . PMID   9689060.
  13. 1 2 3 Bollag G, Tsai J, Zhang J, Zhang C, Ibrahim P, Nolop K, et al. (November 2012). "Vemurafenib: the first drug approved for BRAF-mutant cancer". Nature Reviews. Drug Discovery. 11 (11): 873–886. doi:10.1038/nrd3847. PMID   23060265. S2CID   9337155.
  14. 1 2 3 4 "Serine/threonine protein kinase B-rAF" . Retrieved 4 Mar 2013.
  15. 1 2 Morrison DK, Cutler RE (April 1997). "The complexity of Raf-1 regulation". Current Opinion in Cell Biology. 9 (2): 174–179. doi:10.1016/S0955-0674(97)80060-9. PMID   9069260.
  16. 1 2 3 4 5 6 7 8 9 10 11 12 Hanks SK, Hunter T (May 1995). "Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification". FASEB Journal. 9 (8): 576–596. doi: 10.1096/fasebj.9.8.7768349 . PMID   7768349. S2CID   21377422.
  17. 1 2 3 4 5 6 7 8 9 Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. (March 2004). "Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF". Cell. 116 (6). Cancer Genome Project: 855–867. doi: 10.1016/S0092-8674(04)00215-6 . PMID   15035987. S2CID   126161.
  18. 1 2 3 4 5 6 Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, et al. (February 2008). "Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity". Proceedings of the National Academy of Sciences of the United States of America. 105 (8): 3041–3046. Bibcode:2008PNAS..105.3041T. doi: 10.1073/pnas.0711741105 . PMC   2268581 . PMID   18287029.
  19. 1 2 3 4 5 Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, et al. (September 2010). "Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma". Nature. 467 (7315): 596–599. Bibcode:2010Natur.467..596B. doi:10.1038/nature09454. PMC   2948082 . PMID   20823850.
  20. Hanks SK, Quinn AM, Hunter T (July 1988). "The protein kinase family: conserved features and deduced phylogeny of the catalytic domains". Science. 241 (4861): 42–52. Bibcode:1988Sci...241...42H. doi:10.1126/science.3291115. PMID   3291115.
  21. Hanks SK (June 1991). "Eukaryotic protein kinases". Curr. Opin. Struct. Biol. 1 (3): 369–383. doi:10.1016/0959-440X(91)90035-R.
  22. Hanks SK, Quinn AM (1991). "[2] Protein kinase catalytic domain sequence database: Identification of conserved features of primary structure and classification of family members". Protein kinase catalytic domain sequence database: identification of conserved features of primary structure and classification of family members. Methods in Enzymology. Vol. 200. pp. 38–62. doi:10.1016/0076-6879(91)00126-H. ISBN   978-0-12-182101-2. PMID   1956325.
  23. Mason CS, Springer CJ, Cooper RG, Superti-Furga G, Marshall CJ, Marais R (April 1999). "Serine and tyrosine phosphorylations cooperate in Raf-1, but not B-Raf activation". The EMBO Journal. 18 (8): 2137–2148. doi:10.1093/emboj/18.8.2137. PMC   1171298 . PMID   10205168.
  24. Roberts A, Allanson J, Jadico SK, Kavamura MI, Noonan J, Opitz JM, et al. (November 2006). "The cardiofaciocutaneous syndrome". Journal of Medical Genetics. 43 (11): 833–842. doi:10.1136/jmg.2006.042796. PMC   2563180 . PMID   16825433.
  25. Ewalt M, Nandula S, Phillips A, Alobeid B, Murty VV, Mansukhani MM, et al. (December 2012). "Real-time PCR-based analysis of BRAF V600E mutation in low and intermediate grade lymphomas confirms frequent occurrence in hairy cell leukaemia". Hematological Oncology. 30 (4): 190–193. doi:10.1002/hon.1023. PMID   22246856. S2CID   204843221.
  26. Palomaki GE, McClain MR, Melillo S, Hampel HL, Thibodeau SN (January 2009). "EGAPP supplementary evidence review: DNA testing strategies aimed at reducing morbidity and mortality from Lynch syndrome". Genetics in Medicine. 11 (1): 42–65. doi:10.1097/GIM.0b013e31818fa2db. PMC   2743613 . PMID   19125127.
  27. Namba H, Nakashima M, Hayashi T, Hayashida N, Maeda S, Rogounovitch TI, et al. (September 2003). "Clinical implication of hot spot BRAF mutation, V599E, in papillary thyroid cancers". The Journal of Clinical Endocrinology and Metabolism. 88 (9): 4393–4397. doi: 10.1210/jc.2003-030305 . PMID   12970315.
  28. Tan YH, Liu Y, Eu KW, Ang PW, Li WQ, Salto-Tellez M, et al. (April 2008). "Detection of BRAF V600E mutation by pyrosequencing". Pathology. 40 (3): 295–298. doi:10.1080/00313020801911512. PMID   18428050. S2CID   32051681.
  29. Li WQ, Kawakami K, Ruszkiewicz A, Bennett G, Moore J, Iacopetta B (January 2006). "BRAF mutations are associated with distinctive clinical, pathological and molecular features of colorectal cancer independently of microsatellite instability status". Molecular Cancer. 5 (1): 2. doi: 10.1186/1476-4598-5-2 . PMC   1360090 . PMID   16403224.
  30. Benlloch S, Payá A, Alenda C, Bessa X, Andreu M, Jover R, et al. (November 2006). "Detection of BRAF V600E mutation in colorectal cancer: comparison of automatic sequencing and real-time chemistry methodology". The Journal of Molecular Diagnostics. 8 (5): 540–543. doi:10.2353/jmoldx.2006.060070. PMC   1876165 . PMID   17065421.
  31. Deng G, Bell I, Crawley S, Gum J, Terdiman JP, Allen BA, et al. (January 2004). "BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer". Clinical Cancer Research. 10 (1 Pt 1): 191–195. doi: 10.1158/1078-0432.CCR-1118-3 . PMID   14734469.
  32. Gear H, Williams H, Kemp EG, Roberts F (August 2004). "BRAF mutations in conjunctival melanoma". Investigative Ophthalmology & Visual Science. 45 (8): 2484–2488. doi: 10.1167/iovs.04-0093 . PMID   15277467.
  33. Maldonado JL, Fridlyand J, Patel H, Jain AN, Busam K, Kageshita T, et al. (December 2003). "Determinants of BRAF mutations in primary melanomas". Journal of the National Cancer Institute. 95 (24): 1878–1890. doi: 10.1093/jnci/djg123 . PMID   14679157.
  34. Puxeddu E, Moretti S, Elisei R, Romei C, Pascucci R, Martinelli M, et al. (May 2004). "BRAF(V599E) mutation is the leading genetic event in adult sporadic papillary thyroid carcinomas". The Journal of Clinical Endocrinology and Metabolism. 89 (5): 2414–2420. doi: 10.1210/jc.2003-031425 . PMID   15126572.
  35. Elisei R, Ugolini C, Viola D, Lupi C, Biagini A, Giannini R, et al. (October 2008). "BRAF(V600E) mutation and outcome of patients with papillary thyroid carcinoma: a 15-year median follow-up study". The Journal of Clinical Endocrinology and Metabolism. 93 (10): 3943–3949. doi: 10.1210/jc.2008-0607 . PMID   18682506.
  36. Badalian-Very G, Vergilio JA, Degar BA, Rodriguez-Galindo C, Rollins BJ (January 2012). "Recent advances in the understanding of Langerhans cell histiocytosis". British Journal of Haematology. 156 (2): 163–172. doi: 10.1111/j.1365-2141.2011.08915.x . PMID   22017623. S2CID   34922416.
  37. Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, et al. (June 2011). "BRAF mutations in hairy-cell leukemia". The New England Journal of Medicine. 364 (24): 2305–2315. doi:10.1056/NEJMoa1014209. PMC   3689585 . PMID   21663470. *Lay summary in: "Research on hairy-cell leukaemia shows the promise of new DNA-scanning technologies". Cancer Research UK. June 11, 2011.
  38. Kurppa KJ, Catón J, Morgan PR, Ristimäki A, Ruhin B, Kellokoski J, et al. (April 2014). "High frequency of BRAF V600E mutations in ameloblastoma". The Journal of Pathology. 232 (5): 492–498. doi:10.1002/path.4317. PMC   4255689 . PMID   24374844.
  39. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-Santagata D, Thorner AR, et al. (February 2014). "Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas". Nature Genetics. 46 (2): 161–165. doi:10.1038/ng.2868. PMC   3982316 . PMID   24413733.*Lay summary in: Leah Eisenstadt (30 January 2014). "Single driver mutation found in rare brain tumor". BROAD Institute.
  40. Ruiz-Saenz A, Atreya CE, Wang C, Pan B, Dreyer CA, Brunen D, et al. (February 2023). "A reversible SRC-relayed COX2 inflammatory program drives resistance to BRAF and EGFR inhibition in BRAFV600E colorectal tumors". Nature Cancer. 4 (2): 240–256. doi:10.1038/s43018-022-00508-5. PMC   9970872 . PMID   36759733.
  41. Zecchin D, Boscaro V, Medico E, Barault L, Martini M, Arena S, et al. (December 2013). "BRAF V600E is a determinant of sensitivity to proteasome inhibitors". Molecular Cancer Therapeutics. 12 (12): 2950–2961. doi: 10.1158/1535-7163.MCT-13-0243 . hdl: 2318/140935 . PMID   24107445. S2CID   17012966.
  42. King AJ, Patrick DR, Batorsky RS, Ho ML, Do HT, Zhang SY, et al. (December 2006). "Demonstration of a genetic therapeutic index for tumors expressing oncogenic BRAF by the kinase inhibitor SB-590885". Cancer Research. 66 (23): 11100–11105. doi: 10.1158/0008-5472.CAN-06-2554 . PMID   17145850.
  43. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. (June 2011). "Improved survival with vemurafenib in melanoma with BRAF V600E mutation". The New England Journal of Medicine. 364 (26). BRIM-3 Study Group: 2507–2516. doi:10.1056/NEJMoa1103782. PMC   3549296 . PMID   21639808.
  44. 1 2 Das Thakur M, Salangsang F, Landman AS, Sellers WR, Pryer NK, Levesque MP, et al. (February 2013). "Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance". Nature. 494 (7436): 251–255. Bibcode:2013Natur.494..251D. doi:10.1038/nature11814. PMC   3930354 . PMID   23302800.
  45. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, et al. (December 2010). "Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation". Nature. 468 (7326): 973–977. Bibcode:2010Natur.468..973N. doi:10.1038/nature09626. PMC   3143360 . PMID   21107323.
  46. Degirmenci U, Yap J, Sim YR, Qin S, Hu J (2021). "Drug resistance in targeted cancer therapies with RAF inhibitors". Cancer Drug Resistance. 4 (3): 665–683. doi: 10.20517/cdr.2021.36 . PMC   9094075 . PMID   35582307.
  47. Guan KL, Figueroa C, Brtva TR, Zhu T, Taylor J, Barber TD, et al. (September 2000). "Negative regulation of the serine/threonine kinase B-Raf by Akt". The Journal of Biological Chemistry. 275 (35): 27354–27359. doi: 10.1074/jbc.M004371200 . PMID   10869359.
  48. Weber CK, Slupsky JR, Kalmes HA, Rapp UR (May 2001). "Active Ras induces heterodimerization of cRaf and BRaf". Cancer Research. 61 (9): 3595–3598. PMID   11325826.
  49. Stang S, Bottorff D, Stone JC (June 1997). "Interaction of activated Ras with Raf-1 alone may be sufficient for transformation of rat2 cells". Molecular and Cellular Biology. 17 (6): 3047–3055. doi:10.1128/MCB.17.6.3047. PMC   232157 . PMID   9154803.
  50. Reuter CW, Catling AD, Jelinek T, Weber MJ (March 1995). "Biochemical analysis of MEK activation in NIH3T3 fibroblasts. Identification of B-Raf and other activators". The Journal of Biological Chemistry. 270 (13): 7644–7655. doi: 10.1074/jbc.270.13.7644 . PMID   7706312.
  51. Ewing RM, Chu P, Elisma F, Li H, Taylor P, Climie S, et al. (2007). "Large-scale mapping of human protein-protein interactions by mass spectrometry". Molecular Systems Biology. 3 (1): 89. doi:10.1038/msb4100134. PMC   1847948 . PMID   17353931.
  52. Qiu W, Zhuang S, von Lintig FC, Boss GR, Pilz RB (October 2000). "Cell type-specific regulation of B-Raf kinase by cAMP and 14-3-3 proteins". The Journal of Biological Chemistry. 275 (41): 31921–31929. doi: 10.1074/jbc.M003327200 . PMID   10931830.

Further reading

PD-icon.svg This article incorporates public domain material from Dictionary of Cancer Terms. U.S. National Cancer Institute.This article incorporates text from the United States National Library of Medicine, which is in the public domain.