Preprint
Review

This version is not peer-reviewed.

Tyrosine Kinases: Structural Insights and Mechanistic Roles in Cancer Progression and Therapeutics

A peer-reviewed article of this preprint also exists.

Submitted:

01 May 2025

Posted:

05 May 2025

You are already at the latest version

Abstract
Protein tyrosine kinases (PTKs) are key enzymes of cellular signaling, regulating key processes such as proliferation, differentiation, migration, metabolism, and apoptosis. TKs modulate protein functions in normal and disease states by phosphorylation of tyrosine residues on target proteins. On this critical role, dysregulation of TKs is directly linked with disease progression, particularly in cancer, therefore making TKs an attractive target for therapeutic intervention. The PTK family is broadly classified into receptor tyrosine kinases (RTKs) and non-receptor tyrosine kinases (NRTKs), having variation at both structural and functional levels. RTKs are membrane-bound kinases that initiate intracellular signaling when they react with extracellular ligands, whereas NRTKs within the cytoplasm or nucleus convey intracellular signaling upon receptor activation. This review aims at the organization, mechanistic activity, and therapeutic potential of PTKs, with a particular focus on epidermal growth factor receptor (EGFR) and Src kinase as representative of RTK and NRTK, respectively. Additionally, this review also focuses on addressing emerging strategies to enhance TKI efficacy and overcome acquired resistance in cancer therapy.
Keywords: 
;  ;  ;  ;  ;  ;  

1. Introduction

Tyrosine kinases are crucial enzymes involved in signal transduction, which regulate key cellular processes such as proliferation, differentiation, migration, metabolism, and apoptosis[1,2,3,4,5]. By catalyzing the phosphorylation of tyrosine residues in target proteins, kinases mediate vital cellular communication and maintain homeostasis[6,7]. Hence, phosphorylation acts as a post-translational modification that plays a central role in normal cellular functions, but its dysregulation can lead to pathological conditions, including cancer [8,9,10,11,12,13]. Unusual activation of protein tyrosine kinases (PTKs) is usually associated with disease progression and therapy resistance, while making them critical targets for therapeutic interventions, particularly in cancer treatment[14,15,16,17,18,19,20].

1.1. Broad Classification of Tyrosine Kinases

The PTK family is diverse, with members varying in structure and function[21]. These kinases are classified into two major subgroups: receptor tyrosine kinases (RTKs) and non-receptor tyrosine kinases (NRTKs)[22,23]. RTKs are membrane-bound enzymes that transmit extracellular signals such as growth factors, cytokines, and hormones to the cytoplasm and nucleus, initiating a cascade of cellular responses[7,24,25]. The key function of RTKs is to rapidly and reversibly phosphorylate protein substrates, which leads to alterations in protein conformation and interaction, driving various cellular processes such as growth and survival[26]. On the other hand, NRTKs lack extracellular and transmembrane domains and are found in the cytoplasm or nucleus. These kinases are involved in mediating intracellular signals, often in response to receptor-dependent activation at the cell membrane[27,28,29]. While RTKs and NRTKs function similarly by regulating crucial cellular processes, including cell division, growth, and immune responses, their structures are strikingly distinct [30,31]. Due to their essential roles in cellular signaling, both RTKs and NRTKs are critical in the regulation of various physiological functions and are often implicated in the progression of cancers when their activation becomes dysregulated [32]. The discovery of the Src oncogene and the identification of the epidermal growth factor receptor (EGFR) as the first RTK laid the foundation for understanding the role of tyrosine kinases in cancer[33,34]. So far, over 90 tyrosine kinases have been identified, and these enzymes are now recognized as pivotal players in cellular signaling circuits that contribute to cancer development[35]. Hence, tyrosine kinases represent a significant portion of oncoproteins, and targeting these for therapeutic development is a promising strategy in the treatment of cancers associated with their dysregulation[14,15,16].
As described above PTKs are primarily classified as RTKs and NRTKs[22,23]. Based on the composition of the extracellular regions, the 58 identified RTKs in humans are further categorized into 20 distinct families (Table 1).

1.2. Cancer-Associated Receptor Tyrosine Kinases

The epidermal growth factor family (EGF) includes EGFR (HER1), HER2, HER3, and HER4[37,38]. These receptors are often overexpressed in epithelial tumors, such as colorectal, head and neck, non-small cell lung, breast, pancreatic, and renal cell cancers[39,40]. The insulin growth factor (IGF) and insulin receptor (InsR) family consist of the IGF1R and InsR receptors. Both IGF1 and IGF2 are capable of binding to and activating the IGF1R transmembrane receptor kinase. However, when IGF2 binds, it does not activate any downstream signaling pathways because the IGF2R lacks the kinase structural domain necessary for this activation[41]. Platelet-derived growth factor receptor (PDGFR), colony-stimulating factor 1 receptor (CSF1R), KIT proto-oncogene receptor (KIT), and FMS-like tyrosine kinase 3 (FLT3) receptors are critical for various cellular processes[42]. PDGF is essential for tissue growth, division, and blood vessel formation. CSF1R, secreted by cancer cells to evade immune detection, promotes the growth and recruitment of tumor-associated myeloid cells, contributing to poorer survival in many cancers[43]. The vascular endothelial growth factor (VEGF) receptor family—VEGFR-1, VEGFR-2, and VEGFR-3—regulates processes like cell migration, angiogenesis, and metabolic homeostasis[44,45,46]. Likewise, the fibroblast growth factor (FGF) receptor family, including FGFR1-4, plays a role in tissue repair, regeneration, and the growth and differentiation of cells during development and organ formation[47,48]. Protein tyrosine kinase-like 7 (PTK7) and colon carcinoma kinase 4 (CCK4) receptors are involved in epithelial cell polarization and brain structure formation[49]. These receptors are catalytically active protein kinases and play roles in the Wnt and VEGF signaling pathways[50]. The neurotrophin receptor (NTRK) family includes TRKA, TRKB, and TRKC receptors, which are vital for the proliferation and migration of the nervous system[51,52]. TRKA responds to nerve growth factor (NGF), TRKB to brain-derived neurotrophic factor (BDNF), and TRKC to neurotrophin-3[53]. The RTK-like orphan receptor (ROR) family includes ROR1 and ROR2 receptors. ROR1 acts as a substitute receptor and co-receptor for Wnt signaling, regulating cell division, polarity, and tissue maintenance[54]. In contrast, ROR2's role in tumor development varies depending on the tumor type or stage; it can either repress or activate tumor growth through atypical Wnt signaling[55]. The muscle-specific kinase (MuSK) receptor is essential for the formation and organization of neuromuscular junctions in skeletal muscle[56,57]. The hepatocyte growth factor (HGF) receptor family includes MET (c-Met) and RON receptors. When HGF binds to MET, it activates the proliferation, migration, and morphogenesis of epithelial cells[58,59]. The TAM receptors (TYRO3, AXL, MER) are activated by the vitamin K-dependent proteins Gas6 and protein S, regulating cell proliferation, survival, adhesion, and migration[60,61]. They also have anti-inflammatory properties and are implicated in carcinogenesis in various malignancies[62,63,64]. The TIE receptor family, consisting of TIE1 and TIE2, regulates angiogenesis and lymph angiogenesis[65,66,67]. The Eph receptor family (EphA1–A10, EphB1–B6) controls angiogenesis, cell migration, patterning, and neuronal formation[68,69,70]. The RET receptor, activated by glial cell-derived neurotrophic factor ligands, is crucial for cell proliferation, neuronal navigation, migration, and differentiation[71,72]. The receptor tyrosine kinase (RYK) is characterized by extracellular Wnt-binding domains and is closely associated with Wnt signaling[73,74]. The discoidin domain receptor (DDR) family, which includes DDR1 and DDR2, regulates cell adhesion, proliferation, and metalloproteinase expression[75,76,77]. DDR1 also promotes tumor cell invasion and enhances the survival of tumor stem cells in collagen-rich environments[78,79]. The reactive oxygen species (ROS) receptor family is present in various malignant tumors, making it a promising target for anticancer drugs[79,80,81]. Lemur receptor kinases (LMR/LMTK) are linked to cancer and influence multiple signaling pathways involved in cell proliferation, migration, and invasiveness[82,83,84]. The anaplastic lymphoma kinase (ALK) receptor family includes ALK and leukocyte tyrosine kinase (LTK)[85,86]. ALK gene fusion is linked to the formation of various tumors[87,88]. Additionally, the serine/threonine/tyrosine kinase (STYK) receptor plays a role in cellular processes such as proliferation, differentiation, and survival[30,89,90].

1.3. Cancer-Associated Non-Receptor Tyrosine Kinases

Non-receptor tyrosine kinases (NRTKs) include Ack, Jak, Fes, Fak, Tec, Src, Csk, Abl, and Syk kinases[91,92]. These NRTKs typically consist of the N-terminal kinase domain, which is around 300 residues long, and the C-terminal region, which contains several functional domains[92]. NRTKs share significant sequence similarity within their kinase domains, and their catalytic domains are like those of Ser/Thr protein kinases[93,94]. In addition to their catalytic domains, NRTKs also feature non-catalytic domains that regulate their activity[95,96]. The classification of NRTKs into distinct families is based on molecular analysis of their domain structures, variations in amino acid sequences, and genomic organization of the kinase domains[36,97,98,99]. Below is a brief overview of the most common NRTK families.
The Ack is a large protein of 120 kDa whose kinase activity can be mediated by the phosphorylation of its tyrosine residues[100,101,102]. Ack1 is a non-receptor tyrosine kinase with a unique multidomain structure, including an SH3 and CRIB domain, which regulates cellular functions like migration and adhesion and plays a critical role in cancer progression[103,104,105]. Furthermore, Ack1 promotes tumor growth, resistance to chemotherapy, and recurrence through gene amplification, mutations, and epigenetic regulation[106,107]. The Jak/Janus family consists of four kinases (JAK1, JAK2, JAK3, and TYK2), each with two kinase domains, one functional and one pseudo-kinase[108,109]. These kinases are activated by cytokine receptor ligation, leading to transphosphorylation and downstream signaling[110,111,112]. JAKs play crucial roles in immune cell regulation and tumor development through the JAK-STAT pathway[113,114]. JAK3 is primarily found in hematopoietic cells, while other JAKs are involved in diverse cytokine signaling processes[115,116,117]. Feline sarcoma (Fes) and Fes-related (Fer) kinases are a subgroup of NRTKs with similarities to viral oncogenes from feline sarcoma virus and avian Fujinami poultry sarcoma virus[91,118,119]. Fes kinases have a unique FCH domain, coiled-coil motifs, an SH2 domain, and a C-terminal kinase domain[120,121]. Fes and Fer kinases are implicated in cancer progression, with Fes playing a role in cell signaling pathways that influence cell migration, proliferation, and survival, contributing to tumorigenesis[122,123,124]. The Fak family includes Fak, Pyk2, Cak-beta, Cadtk, Raftk, and Fak2, with varying expression in organs like the brain, liver, and hematopoietic cells[125,126]. Fak family kinases feature a FERM domain that mediates interactions with integrins and RTKs and a C-terminal FAT region involved in focal adhesion targeting[127,128,129,130,131]. Fak plays a crucial role in tumor cell signaling, including transcriptional regulation within the tumor microenvironment[132,133]. Overexpression of Fak is linked to aggressive cancers, including breast, colon, ovarian, and pancreatic and is commonly associated with metastasis and poor prognosis [134,135,136,137,138,139]. The Tec family consists of five NRTK members, including Tec, Itk, Btk, Txk, and Bmx, characterized by domains such as PH, TH, SH3, SH2, and a kinase domain[140,141].Tec kinases are involved in immune cell signaling, with specific expression in T, B, and NK cells[142,143]. The Src family is one of the largest NRTK family that includes eight members, such as Fyn, Yes, Fgr, and Lyn, divided into two subfamilies: Src-A (Fgr, Fyn, Src, Yes) and Src-B (Blk, Hck, Lck, Lyn)[144,145,146]. These kinases share a similar structure with SH4, SH3, SH2, and kinase domains, but differ in their C-terminal regulatory regions[147,148,149]. Src family kinases are involved in diverse cellular processes, with distinct expression patterns in hematopoietic and other tissues[146,150,151]. FRK (Fyn-related kinase) is a member of the breast tumor kinase (BRK) family, closely related to Src family kinases[152,153,154]. FRN kinases feature an SH3, SH2, and kinase domain, but lack the N-myristoylation site, which prevents membrane localization and allows nuclear localization[155,156]. Unique to FRK and IYK kinases is the presence of a nuclear localization signal (NLS) within the SH2 domain[157,158,159,160]. The NLS is a bipartite motif that enables nuclear targeting and functional regulation in the cell[161]. The Abl family includes Abl and Arg kinases, which are widely expressed, with high levels in the thymus, spleen, and brain[162,163]. Both kinases have structures similar to Src family members but feature a unique C-terminal actin-binding domain and nuclear localization signals[164,165,166,167]. Abl activation, through mutation or phosphorylation, is linked to leukemia and solid tumors like brain, lung, and prostate cancers[168].[169,170,171] The Syk family includes Syk and Zap70 kinases, which share a similar structure containing two SH2 domains followed by a catalytic kinase domain[172,173,174]. These kinases are cytosolic proteins lacking fatty acid modification sites, and upon cell stimulation, Syk and Zap70 translocate to immune receptor complexes at the membrane to trigger downstream signaling[175,176,177]. A tabular representation of kinases that play a significant role in various cancer types is presented in Table 2.

3. Structural and Regulatory Mechanism of Tyrosine Kinases

PTKs play a critical role in cellular signaling pathways, and their catalytic activity is tightly regulated. Numerous atomic structures of PTKs reported in the literature have provided structural and mechanistic insights into the regulation of both receptor and nonreceptor PTKs[145,178,179,180]. As several PTKs are available in the PDB, the current review will focus on EGFR kinase as a representative of receptor PTK and Src for nonreceptor PTKs.

3.1. PTK Domain Architecture

RTKs are composed of three main regions: a large extracellular region, which binds to polypeptide ligands, a transmembrane helix, and a cytoplasmic region, which possesses tyrosine kinase activity. The extracellular region of RTKs is classically composed of a diverse array of distinct globular domains, including immunoglobulin (Ig)-like domains (domain-1), fibronectin type-III-like domains (domain-2), cysteine-rich domains (domain-3), and EGF like domains (domain-4). In the case of EGFR kinase, the extracellular region includes amino acids 1-165 (domain-1), 166-310 (domain-2), 311-480 (domain-3), 481-621 (domain-4). However, the cytosolic region of RTKs domain organization is simple, consisting of the juxtamembrane region (amino acids 643-685), immediately followed by the transmembrane helix, a tyrosine kinase domain (amino acids 686-952), and a carboxy region (amino acids 953-1186) (Fig. 1A, B, C). Unlike RTKs, the extracellular and transmembrane regions in NRTKs are absent, and most of the NRTKs are present in the cytosol. The NRTKs comprise intrinsically disordered regions (IDR) and folded domains. At the N-terminus IDR region, unique myristoylated Src homology 4 (SH4) fragments, a smaller SH3 domain (∼60 residues), a short Src homology 2 (SH2 ∼100 residues), SH2 kinase linker, catalytic tyrosine-protein kinase domain (SH1), and a short intrinsically disordered C-terminal tail. While the kinase domain (KD) has a catalytic function, the SH2 and SH3 domains are commonly involved in non-catalytic regulatory properties. However, all these three domains are essential in signal transduction[21,181,182,183,184,185] (Fig. 1D, E).
Figure 1. A. The domain architecture of EGFR. B. The Extracellular region of EGFR is composed of 4 domains I-IV, domain I (red), domain II (cyan), domain III (green), and domain IV (orange). C. The EGFR kinase domain is displayed in medium purple D. The domain architecture of Src. The boundaries of domains are based on the chicken numbering system. E. Ribbon diagram displaying the overall structure of Src (PDBID: 2SRC). The SH3 (pale yellow) and SH2 (green) domains coordinate the linker and C-terminal tail regions, respectively. The kinase domain is colored in blue.
Figure 1. A. The domain architecture of EGFR. B. The Extracellular region of EGFR is composed of 4 domains I-IV, domain I (red), domain II (cyan), domain III (green), and domain IV (orange). C. The EGFR kinase domain is displayed in medium purple D. The domain architecture of Src. The boundaries of domains are based on the chicken numbering system. E. Ribbon diagram displaying the overall structure of Src (PDBID: 2SRC). The SH3 (pale yellow) and SH2 (green) domains coordinate the linker and C-terminal tail regions, respectively. The kinase domain is colored in blue.
Preprints 158087 g001

3.2. Src Structure and Regulatory Mechanism

The primary function of the Src is to transmit the external signal to the cell interior by phosphorylating tyrosine residues on substrates, mainly downstream of RTKs and integrins[186]. Src kinases are crucial in various cellular processes, such as cell proliferation, adhesion, migration, and more[91]. The Src kinase’s complicated regulation is due to its complex structure. The structures of SH3, SH2, and SH1 kinase domains of Src kinases have been extensively studied and reviewed elsewhere[33,96,99,185,187]. The Src kinase domain features a characteristic bilobed architecture comprising a small N-terminal lobe and a large C-terminal lobe. The residues 267-337 and 341-520 make up these lobes, respectively[188,189,190,191]. The N-lobe predominantly anchors and orients ATP, featuring a G-rich loop, which is a part of the nucleotide-phosphate binding site. The N-lobe is mostly composed of antiparallel β sheet structures[192]. The C-lobe is predominantly composed of ⍺ helix, responsible for binding the protein substrates and contributing to the ATP-binding site. The catalytic site of Src is situated in a cleft between these two lobes; they open and close during ATP hydrolysis[187]. The dynamic conformational switch regulates ATP binding and ADP release; the open form is required to allow ATP to bind to its catalytic pocket and release ADP; the closed form is important to bring residues into the catalytically active form. The Src kinase regulation is precisely involved in the coordination of non-regulatory SH2 and SH3 domains and a regulatory kinase domain[145,187]. In the autoinhibitory conformation, the SH2 domain binds to phosphotyrosine-containing motifs, precisely phosphorylated Tyr527 in the C-terminal tail of Src, and stabilizes the conformation[145,187]. The SH3 domain interacts with a polyproline-rich motif situated between the SH2 and Kinase domains. This interaction positions SH2-SH3 domains as a compact structural unit, which further prevents the movement of the kinase domain and, consequently, locks the Src in its inactive state. The activation loop (residues 404-418) conformations in the kinase domain dictate the active and inactive state of the Src kinase. In the inactive Src kinase, the activation loop forms a short ⍺-helix between the N and C-lobes known as the A-loop helix[145,187]. As a result, the Tyr416 residue side chain is buried between the N and C-lobes; this conformational switch leads to the prevention of the formation of a salt bridge between Lys295 and Glu310 required for enzyme activity. The autophosphorylation of Tyr416 disrupts the autoinhibitory state of the Src kinase, leading to an extended conformational switch in the activation loop and alignment of catalytic residues such as Asp386 and Asp404. Asp386 residue acts as a catalytic base for the tyrosine substrate, whereas Asp404 interacts with magnesium ions that stabilize ATP. Numerous studies on Src have revealed that SH2 and SH3 domains are critical for maintaining the autoinhibited state of Src. However, the kinase domain is involved in severe conformational changes to switch between active and inactive states. This structural equilibrium is disrupted when C-terminal Tyr527 is mutated. In the case of v-Src, a mutation at Tyr527 impaired the SH2-SH3 interaction between the kinase domain and resulted in constitutive kinase activity[7,145,186,187].
The Src protein-tyrosine phosphorylation levels are balanced by counteraction between C-terminal Src kinase (CSK) and protein-tyrosine phosphatases (PTPs). Okada and Nakagawa[193] were the first to demonstrate that CSK, a cytoplasmic PTK, controls the regulatory tyrosine phosphorylation in rat brains. They also highlighted its efficiency in phosphorylating Src at Tyr527, a key regulatory site for its activation. In contrast, PTPs such as PTP⍺ and PTPε facilitate the dephosphorylation of phosphotyrosine 527 in the Src kinase domain, thereby displacing it, leading to Src kinase activation (Fig. 2C, D). Structural studies have revealed that the substrate recognition mechanism between Src and PTPs relies on the cysteine-dependent active site of PTPs and the phosphorylated tyrosine side chain of Src (Fig. 2C, D)[194]. Recent findings have identified two additional key charge-charge interactions between rPTPε and phospho-Src beyond the active site interactions [195]. These biochemical and structural insights are extremely important for the development of novel therapeutic strategies for targeting kinases, particularly in cancer treatment.

3.3. EGFR Structure and Regulatory Mechanism

EGFR regulates multiple functions involved in developmental, metabolic, and physiological processes[196]. When exposed to ligands like EGF, the receptor EGFR binds to EGF, undergoing a conformational switch from an inactive monomer to an active dimer (Fig. 2A). This conformational change leads to autophosphorylation of the receptor, which sequentially activates downstream signaling pathways to control cell proliferation and differentiation. EGFR, along with growth factor-α, amphiregulin, and other ligands, promotes either homodimerization of two EGFRs or heterodimerization of EGFR with other family members[197]. Upon activation of receptor TKs, there is a subsequent activation of downstream Ras/mitogen-activated protein kinase pathway, the pI3K/Akt pathway, and activation of transcription pathways[198].
Figure 2. A. A schematic diagram of inactive EGFR, ligand-bound active dimeric extracellular EGFR, and an asymmetric dimer of kinase domain B. Structure of TGF⍺ dimer of sEGFR (PDB ID: 1MOX). An extracellular structure bound to EGF, a transmembrane helix dimer. C. Phosphotyrosine displacement by PTP⍺ and activation mechanism of Src kinase. D. Haddock model of PTP⍺ and Src kinase complex, displaying the phosphatase and tyrosine kinase interaction.
Figure 2. A. A schematic diagram of inactive EGFR, ligand-bound active dimeric extracellular EGFR, and an asymmetric dimer of kinase domain B. Structure of TGF⍺ dimer of sEGFR (PDB ID: 1MOX). An extracellular structure bound to EGF, a transmembrane helix dimer. C. Phosphotyrosine displacement by PTP⍺ and activation mechanism of Src kinase. D. Haddock model of PTP⍺ and Src kinase complex, displaying the phosphatase and tyrosine kinase interaction.
Preprints 158087 g002

3.4. Extracellular Structure of EGFR

The extracellular structural modules of all four EGFR members have been thoroughly studied both in the presence and absence of their respective ligands, as well as in complexes with antibodies[199,200]. Atomic structures reveal two key conformations that are important in the extracellular modules. One is an extended form that facilitates the conformation of one protomer in the active dimer, while the other is folded over or tethered conformation where dimerization elements are buried. Upon ligand binding, the extracellular domains display significant conformational change, transitioning the module from tethered to the extended state, resulting in dimerization and activation of the EGFR (Fig. 2B). This extended conformation is represented as a back-back dimer configuration, with the ligand positioning between domains I and III of each receptor subunit. The glycosylation of the EGFR extracellular region is critical for its activation; the sugar moiety, around 40 kDa is known to play a role in EGFR maturation and cell surface translocation. Mutation studies have identified that Asn579 is crucial for regulating receptor conformation and ligand binding affinity. Another mutation at Asn 579, located on a specific glycosylation site, influences the structural conformation of EGFR and ligand binding. Furthermore, a mutation N420D in EGFR was shown to display ligand-independent activation through spontaneous oligomer formation [200]. Together, the biochemical and structural details underscore the complexity of these receptors’ regulation and offer a base for therapeutic strategies targeting EGFR family members.

3.5. EGFR Intracellular Kinase Structure Activation

The intracellular region of EGFR is mostly comprised of its kinase domain (KD), the KD adopts a canonical kinase fold that exists as both active and inactive conformations. EGFR (Unphosphorylated) structure in the presence of erlotinib from Genentech is the first atomic structure of the EGFR kinase domain; this structure provides its unique structural features and activation mechanism[178,198]. The structure features the conserved Asp-Phe-Gly (DFG) motif at the base of the activation loop, which is a key activation/regulatory motif. In inactive conformation, the aspartic acid residue flips out of the catalytic center, making the kinase inactive and preventing the entry of ATP. This is observed in several kinases[201]. In the EGFR: erlotinib complex, the DFG motif is found in the ‘in’ conformation; in this, the activation loop is open and properly configured to bind its ligands. Additionally, the active site element ⍺C in the N-lobe switches inward and facilitates the ion pair interaction between Glu 738 and Lys, which is critical for catalytic activity[200]. In contrast to other kinases, EGFR does not require phosphorylation of its activation loop to transition to the active state. The atomic structure of EGFR in complex with lapatinib captured in its inactive state, surprisingly, this structure resembles the inactive states of Src-family kinases[200]. In the structure, the ⍺C in the N-lobe switched outwards, and the activation loop formed a short helix, blocking its ATP binding. Mutations on the activation loop phosphorylation sites revealed that the phosphorylation is not an absolute requirement for EGFR’s activation. Overall, the atomic details of these structures detailed the understanding of EGFR’s regulatory flexibility and underlined its divergence from other RTKs, which rely on autoinhibitory interactions and activation loop phosphorylation for regulation[200].
The activation mechanism of EGFR was revealed through the determination of the homodimer KD structure. In this structure, one kinase domain (activator) allosterically interacts with its partner (receiver) to activate the EGFR. This dimerization interaction occurs at the N-lobe of the receiver and the C-lobe of the activator, resembling the cyclin-mediated CDK type of activation. Unlike other RTKs, the EGFR activation mechanism is driven by protein-protein interactions at the dimerization interface. This mechanism is also observed in other members such as Her2, Her3, and Her4 (Fig. 2A, B)[202,203,204,205,206]. Further MD simulation studies demonstrated how EGFR transitions between active and inactive conformations through local unfolding at the hinge region between N- and C-lobes. Together, these structural insights have significant clinical implications, helping in developing novel targeted antibodies like erlotinib and lapatinib, which exploit EGFR’s conformational flexibility. For example, EGFR inhibitor Mig6 is known to block the asymmetric dimer interface and inhibit activation[207,208,209,210,211,212,213,214,215,216]. This understanding highlights the unique regulatory mechanism of EGFR and its critical role in cancer biology.

4. The Role of Tyrosine Kinases in Cancers

Tyrosine kinases, a large family of kinases that include both RTKs and NRTKs, serve as critical molecular switches in regulating various cellular processes such as growth, survival, development, and differentiation[108,217,218,219]. Several studies have highlighted the role of PTKs in various cancers and their potential for drug discovery. The current review focuses on EGFR and Src’s role as therapeutic targets for developing treatments against cancer cell-specific pathways.

4.1. Role of EFGR-Tyrosine Kinase in Cancers

The EGFR family regulates developmental, metabolic, and physiological processes[196]. A key aspect of EGFR-driven cancers involves mutations in the tyrosine kinase domain of the EGFR gene (exon 18-21), categorized into three classes: class-I (In-frame deletions in exon 19), class II (single-nucleotide substitutions), class III (In-frame duplications and or insertions in exon 20)[220,221]. Class I accounts for approximately 44% of the activating EGFR-TK domain mutations, including deletion at LRE (Leu-747 to Glu-749, while class II mutations contribute ~41%, often affecting the kinase domain C-helix. Class III mutations, constituting ~5%, are less frequent but still play a role in tumor progression[198,220,222,223,224]. Lemmon and Schlessinger, in 2010, best characterized the function of EGFR in ligand- and kinase-dependent activation, also known as the canonical EGFR signaling pathway[225]. Several of these stress pathways are activated in cancer cells to induce survival advantage as well as resistance to cancer therapy[226,227]. Casanova et al. (2002) demonstrated that EGFR signaling is responsible for the Ha-ras-dependent activation in epidermal tumor cells[228]. Recent publications support the activation of EGFR signaling pathways in epithelial cancers, including breast, ovarian, prostate, and NSCLC[229,230,231].

4.2. Role of Src-Tyrosine Kinase in Cancers

SFKs play a crucial role in various cellular processes, such as cell proliferation, adhesion, migration etc[91]. Their dysregulation is frequently implicated in tumors, where they are often overexpressed due to their role in cell-cell adhesion[232,233]. Particularly, Src is involved in activating STAT transcription factors, promoting tumorigenesis, and influencing cytokine signaling in hematopoietic cells[234]. It also plays a significant role in regulating the RAS/RAF/MEK/ERK MAPK and VEGF pathways in various tumors[235,236,237,238]. Additionally, Src plays a vital role in facilitating tumor cell invasion by phosphorylating target substrates, aiding in the translocation of tumor cells through matrix barriers and tissue compartments. Invasion is a complex process, and tumor Src activation leads to the phosphorylation of targeted substrates, influencing the activity of cellular proteins to carry out this entire cellular process[239,240,241]. SFKs are activated in tumors through mutations of the Src allele, leading to a disorganized negative regulatory pathway or by binding to activating partners such as growth factors (Her 2/Neu, PDGF, EGFR). Oncogenic Src (v- Src) can activate Ras by recruiting the Grd 2/Sos complex, thereby stimulating Ras-mediated tumorigenic signals[242,243]. Furthermore, p120RasGAP-mediated activation of c-Src is important for Ras-induced tumor invasion[244]. The tumor microenvironment plays a crucial role in Src upregulation, leading to enhanced Src activity during cancer progression[245]. Additionally, inhibitory phosphorylation of Tyr 530 is mediated by the kinase Csk, which acts as a crucial regulator of Src activity[246]. Given the importance of Src/EGFR in tumor progression, the review will explore tyrosine kinase therapeutic targets and also provide insights into potential strategies for overcoming therapeutic resistance.

5. Tyrosine Kinases as Therapeutic Targets

5.1. Development of TKIs

Cancer cell survival in the tumor microenvironment (TME) is challenging and highly influenced by external factors. Cancer treatment has advanced in developing tyrosine kinase inhibitors (TKIs). Discovery and development of imatinib (Gleevec, Inc) as the first effective TKI to treat chronic myeloid leukemia (CML), established as tumor-targeted therapy that acts specifically against Bcr-Abl fusion protein. Inhibitors such as Sorafenib and sunitinib served as early examples of TKIs approved for solid tumors and renal cell carcinoma[247,248,249,250,251,252]. Over the past 20 years, robust and specific TKIs with single or multiple targets have been identified, which include EGFR, ROS1, VEGFR, MEK, FGFR, and PDGFR[253,254]. The known FDA-approved TKI is listed in Table 3. IRIS trials (2000-2001) confirmed the long-term survival benefit of treating Imatinib[255]. However, there has been concern over the emergence of resistance to imatinib. Nilotinib and dasatinib, are two of the TKIs (second generation) approved worldwide for the treatment of chronic myeloid leukemia after imatinib failure[255]. Development of TKIs always challenging because of the resistance over a while, most patients developed acquired resistance against TKIs upon a median period of 10-15 months[256,257].
Two main approaches to therapeutically targeting EGFR rely on using mAbs and small molecules of EGFR-tyrosine kinase Inhibitors (EGFR-TKIs). mAbs specific to EGFR target the extracellular domain, whereas EGFR-TKIs block the binding of ATP to the intracellular catalytic domain of EGFR[273]. For example, Panitumumab and cetuximab are the two approved mAbs widely used in the treatment of colorectal cancer where EGFR with KRAS expressions[274,275,276]. Erlotinib and gefitinib are two selective TKIs used in combination with mAbs in the treatment of NSCLC. Several preclinical and clinical studies were conducted to study the effect of these EGFR inhibitors alone and in combination with mAbs/chemotherapies[277,278,279]. Cetuximab and panitumumab have been studied in combination with anthracycline/texane-based chemotherapy through pilot studies of multicentric neoadjuvant TNBC[280,281,282]. Studies reported that using cetuximab in combination with either gefitinib or erlotinib has proven to enhance apoptosis and growth inhibition of neck cancer cell lines over using them alone in the treatment[283]. Additionally, it is suggested that cetuximab and gefitinib showed a synergistic effect on EGFR downstream signaling pathways[284,285]. Trastuzumab, in combination with lapatinib, is used to treat HER2-overexpressed breast cancer, these two develop resistance in patients when treated alone[286]. One of the strong reasons to use combinational therapy on mAbs and selective EGFR-TKIs was to target different molecular domains of the EGFR. However, selective targeting of EGFR was limited to EGFR-driven cancers, in the case of EGFR-and KRAS or STKs driven cancers, one needs to be more selective in choosing combinational therapies.

5.2. Resistance to TKIs and Strategies to Overcome Resistance

TKIs are the most common and successful strategies for targeting cancer cells[287,288,289]. However, eventually, cancer cells develop resistance to these drugs. Multi-drug resistance (MDR) in cancer arises when tumors become nonresponsive to chemotherapeutic agents. Many factors contribute to MDR, including enhanced drug efflux caused by overexpressed ABC transporters[290], genetic mutations, the activation of specific signaling pathways, and intracellular-extracellular ATP, which also promotes drug resistance[291,292,293]. Mutations in the EGFR and Src also contribute to drug resistance in cancers. To overcome MDR, researchers have developed strategies emphasizing the use of monoclonal antibodies (mAbs) that target specific receptors or signaling components of the pathway, or any protein that specifically promotes tumor oncogenesis. Here, we highlight the use of mAbs alone and in combination to achieve effective treatment against cancers.
Resistance to TKIs in EGFR-mutants NSCLC remains a challenge in cancer therapy. Studies have identified that, on average, 50% of resistance to first- and second-generation EGFR-TKIs is due to EGFR T 790 M mutation. This amino acid substitution in EGFR leads to an increased affinity to ATP caused by a conformational change, resulting in steric hindrance and reducing drug efficacy[294,295]. Osimertinib, a third-generation EGFR-TKI, inhibits both EGFR T 790 M and EGFR-sensitizing mutations, demonstrating increased efficiency over gefitinib and erlotinib[296,297,298]. However, patients developed resistance to long-term usage of third-generation EGFR- TKIs, particularly EGFRC 797 S on exon 20, as the main cause for this acquired resistance[299,300]. Patients responded to a combination of first- and third-generation EGFR- TKIs when harboring C 797 S in trans with T 790 M, whereas those with C 797 S in cis with T 790 M did not respond to this combination[301,302]. EGFR T790 and Src-mediated resistance are two distinct mechanisms where tumor cells develop resistance to therapies, especially EGFR-targeted therapies. Most of the TKIs that target EGFR were less sensitive because of the specific mutation in the EGFR gene. Whereas Src-mediated resistance, on the other hand, involves the activation of Src kinase, which can also bypass the effects of EGFR inhibitors and drugs that target NTKIs[303,304]. To overcome this evolving resistance, researchers are developing fourth-generation EGFR-TKIs and also exploring combination therapies. For instance, EGFR-TKIs combined with programmed death ligand 1 (PD-L1) antibodies with chemotherapy have shown significant survival benefits to patients suffering from EGFR mutation-driven drug resistance in cancers[305]. The FDA-approved TKI and NTKI inhibitors used in cancer therapy are listed in Table 4 and Table 5.

5.3. Resistance and Mechanism of Developing Resistance to Therapy

Trastuzumab (Herceptin), a drug used to treat breast cancer, is resistant in patients. Trastuzumab binds to an epitope in the juxtamembrane region of the HER2 RTKs. Upon binding, trastuzumab induces uncoupling of ligand-independent HER2-HER3 heterodimers and inhibits downstream signaling as well as antibody-dependent cell cytotoxicity[329]. The main reasons reported for resistance of the trastuzumab in patients were as explained in decreased interactions with HER2 due to blockage by cell surface proteins like mucin-4 (MUC4)[330]. Consistent treatment with trastuzumab leads to decreased expression of the tumor suppressor PTEN gene and activation of the Akt signaling pathway. Another main reason for developing resistance was the activation of the phosphatidylinositol 3-kinase/Akt pathway, which can lead to decreased sensitivity to trastuzumab[331]. Another potential explanation for developing trastuzumab resistance is its ability to bind to hyaluronan and CD44 a transmembrane receptor that can hinder trastuzumab access to HER2[332]. A clinical study was conducted to analyze sensitivity to trastuzumab treatment and reported in the study on 46 patients with breast cancer, 11.1% of patients responded to trastuzumab (expressing p95HER2), 51.4% with expressing p185HER2 achieved clinical response[333]. Lapatinib, a small molecule that can inhibit both HER2 and EGFR kinase, was tested in p95HER2 preclinical studies to prevent Her2 signaling loss of the trastuzumab binding site[333]. When lapatinib is coupled with trastuzumab, clinical studies have shown it to be progressive in patients with stage IV HER-overexpressing breast cancer[334].
Cetuximab is a mAb that treats metastatic colorectal cancer and squamous cell cancer (head and neck squamous cell cancer-HNSCC). The use of cetuximab and panitumumab in colorectal cancer patients is successful[335,336]. However, treatment with cetuximab and panitumumab as single agents was only 10% effective in clinical significance. This clearly explains the development of resistance to the therapy. Most patients develop resistance within 3-12 months of starting therapy[337]. The most probable explanation for developing resistance is but not limited to RAS mutations (these mutations prevent patients from having a response with therapy). Acquired resistance is another important reason when using EGFR-targeted mAbs. Preclinical and molecular profiling of clinical specimens developed resistance to EGFR-targeted mAbs are having genetic alterations of genes in EGFR-RAS-RAF-MEK signaling pathway and of RTKs are the mechanism of acquired resistance to anti-EGFR mAbs[338,339,340]. Mutations in the codons 12 and 13 of KRAS were the first identified mechanism of primary resistance to anti-EGFR therapy; later, patients screened for KRAS mutations prior to mAbs. Researchers also reported that oncogenic Ras and wildtype p53 stimulate STAT non-cell autonomously and promote tumor radioresistance[238]. However, in some instances, RAS wild-type patients can be non-responders to anti-EGFR therapy, as it was well understood that additional mechanisms of intrinsic resistance have been attributed to mutations in PI3KCA/BRAF[341,342]. The above genetic mutations leading to acquired resistance and escape from anti-EGFR blockade appear to converge on the activation of MEK-ERK/AKT signaling pathways. Considering each mAbs has its advantages over disadvantages in therapy.
Pertuzumab (Omnitarg, 2C4) is an anti-HER2 mAb that binds to the domain II epitope of Her2 and is able to block a binding pocket essential for receptor dimerization and signaling. Pertuzumab is suggested to be a potential synergism with trastuzumab in HER2 overexpression cell lines[343]. Phase II clinical trials of pertuzumab in combination with trastuzumab have shown disease progression over trastuzumab in patients with HER2-overexpressing metastatic breast cancer[344]. Currently, clinical trials in different stages testing pertuzumab in combination with trastuzumab in different settings and as well as pertuzumab with chemotherapy are ongoing[345]. Toxicity profiles of these new antibodies (mAbs) are comparable to that of Cetuximab, even though they are associated with less hypersensitive reactions. Mostly, mAbs administrations needed frequent clinical visits due to their mode of administration (intravenous infusions). Also, the proposed resistance to cetuximab can be applied to most EGFR-targeted mAbs. From these studies, it is well understood that mAbs targeting specific signaling molecules or receptors have shown progress in combination with other mAb or chemotherapy to overcome resistance in cancers.

5.4. Combined Targeting EGFR and Src as a Potential Therapeutic Approach

Triple-negative breast cancers (TNBC) are an aggressive subtype of breast cancer with limited therapeutic options. It is characterized by the absence of estrogen and progesterone receptors and lack of EGFR2 (HER2) gene amplification and protein expression[346]. Notably, overexpression of EGFR is highlighted in TNBC, attracting significant research interest in evaluating EGFR-TKIs as potential treatments[347]. Despite overexpression of EGFR in TNBC, the EGFR-specific TKIs have shown limited efficacy due to their intrinsic or acquired resistance mechanisms[348]. Studies identified a key factor that contributes to EGFR resistance is the association of Src family kinases, which have been shown to increase HER-family receptor expression[349,350]. The overexpression of Src enhances HER2/HER3 dimerization, consequently delaying receptor internalization and hence prolonging its downstream oncogenic signaling[351,352,353]. This crosstalk between EGFR and Src kinases suggests that targeting EGFR alone may not be sufficient, suggesting a dual-targeted approach that can inhibit Src signaling.
Src inhibitor dasatinib exhibits initial sensitivity in TNBC cells; however, later, it develops resistance[354,355]. However, combining both EGFR and Src inhibitors has shown promising results[356]. For instance, an irreversible pan-HER inhibitor, afatinib, and Src inhibitors have shown synergistic effects in MDA-MB-468, TNBC cell lines. Additionally, the combination of afatinib and dasatinib has also been shown to enhance apoptosis and growth suppression of NSCLC in vitro and in vivo [357,358].
These preclinical research studies have progressed to phase-I clinical trials evaluating the efficacy of these combination therapies (ClinicalTrials.gov identifier: NCT01999985). The cooperative interactions between these Src-tyrosine kinases and HER-family members in acquired resistance reveal the significance of developing novel combination drug therapies targeting both pathways. This combination of therapies may hold significant potential in overcoming MDR, improving treatment response, and increasing clinical benefits in TNBC and other EGFR-driven cancers.

5.5. Therapeutic Challenges and Limitations

The development of TKIs against cancer has significantly advanced in recent years. However, their clinical utility is often diminished by adverse effects related to the heart due to toxicity. It is well documented that older generations of TKIs can cause a wide range of cardiovascular issues such as hypertension, atrial fibrillation, and heart failure. These adverse effects highlight a critical therapeutic challenge: the necessity to design novel TKIs that maintain therapeutic efficacy while reducing off-target toxicities. In addition to TKI toxicity, another limitation is the development of drug resistance, which can cause postmenopausal symptoms, muscle and joint pains, and osteoporosis—common issues with prolonged use of TKI therapy[359,360]. Both drug toxicity and the emergence of long-term treatment drug resistance necessitate the development of novel TKIs that balance specific target inhibition with favorable safety profiles. In general, the therapeutic design must prioritize both efficacy and toxicity reduction to improve patient outcomes and ensure long-term treatment sustainability.

6. Summary and Conclusion

This review highlights the crucial role of PTKs, with special emphasis on EGFR and Src, in regulating important cellular functions like growth, differentiation, survival, and dysregulation that often lead to cancer. Furthermore, this review addresses the structural mechanism of EGFR and Src kinases that provides valuable insights into designing novel cancer therapies. Besides that, this review emphasizes the development of tyrosine kinase inhibitors (TKIs), including gefitinib and erlotinib, and the challenges posed by resistance in cancer treatment. We also outline and evaluate the existing clinician trials of combination therapy targeting EGFR and Src kinases, particularly in aggressive cancers like triple-negative breast cancer (TNBC). In conclusion, EGFR and Src kinases are significant players in tumor development and therapeutic resistance. Hence, the development of inhibitors/combination treatment is extremely promising in overcoming multi-drug resistance (MDR) and in augmenting therapeutic response in a broad spectrum of cancers.

Literature search and Methodology

For structural analysis, we used the AlphaFold models, the protein data bank (PDB) to retrieve structures, and used Chimera to analyze and generate figures. For protein-protein interaction studies, we used the HADDOCK online portal (https://rascar.science.uu.nl/haddock2.4/). For the chemo and immunotherapy drug search, we used already published literature, Drugs.com, antibodysocity.org, and cancerresearch.org.

Author Contributions

Conceptualization: E.K.N. and R.S.K.N.; writing—original draft preparation: E.K.N., G.P.V., and R.S.K.N.; writing—review and editing: E.K.N. and R.S.K.N. and All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The HADDOCK generated Src: PTP⍺ complex model used in the current study is available from the corresponding authors on request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EGFR Epidermal growth factor receptor
Src Sarcoma (proto-oncogene tyrosine-protein kinase Src)
TKI Tyrosine kinase inhibitors
TNBC Triple-negative breast cancer
MDR Multi-drug resistance
PTK Protein tyrosine kinase
FDA Food and Drug Administration
RTK Receptor tyrosine kinase
NRTK Non-receptor tyrosine kinase
KD Kinase domain
IDR Intrinsically disordered regions

References

  1. Blume-Jensen, P.; Hunter, T. Oncogenic kinase signalling. Nature 2001, 411, 355–365. [Google Scholar] [CrossRef]
  2. Díaz Galicia, M.E.; Aldehaiman, A.; Hong, S.; Arold, S.T.; Grünberg, R. Chapter Eight - Methods for the recombinant expression of active tyrosine kinase domains: Guidelines and pitfalls. In Methods in Enzymology, Shukla, A.K., Ed.; Academic Press: 2019; Volume 621, pp. 131-152.
  3. Hunter, T. Signaling--2000 and beyond. Cell 2000, 100, 113–127. [Google Scholar] [CrossRef] [PubMed]
  4. K. Bhanumathy, K.; Balagopal, A.; Vizeacoumar, F.S.; Vizeacoumar, F.J.; Freywald, A.; Giambra, V. Protein Tyrosine Kinases: Their Roles and Their Targeting in Leukemia. Cancers 2021, 13, 184. [Google Scholar] [CrossRef]
  5. Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2000, 103, 211–225. [Google Scholar] [CrossRef]
  6. De Meyts, P. Receptor Tyrosine Kinase Signal Transduction and the Molecular Basis of Signalling Specificity. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease, Wheeler, D.L., Yarden, Y., Eds.; Springer New York: New York, NY, 2015; pp. 51–76. [Google Scholar]
  7. Zhao, M.; Jung, Y.; Jiang, Z.; Svensson, K.J. Regulation of Energy Metabolism by Receptor Tyrosine Kinase Ligands. Front Physiol 2020, 11, 354. [Google Scholar] [CrossRef] [PubMed]
  8. Mihwa, K.; Minwoo, B.; Dae Joon, K. Protein Tyrosine Signaling and its Potential Therapeutic Implications in Carcinogenesis. Current Pharmaceutical Design 2017, 23, 4226–4246. [Google Scholar] [CrossRef]
  9. Dutta, H.; Jain, N. Post-translational modifications and their implications in cancer. Front Oncol 2023, 13, 1240115. [Google Scholar] [CrossRef] [PubMed]
  10. Geffen, Y.; Anand, S.; Akiyama, Y.; Yaron, T.M.; Song, Y.; Johnson, J.L.; Govindan, A.; Babur, Ö.; Li, Y.; Huntsman, E.; et al. Pan-cancer analysis of post-translational modifications reveals shared patterns of protein regulation. Cell 2023, 186, 3945–3967.e3926. [Google Scholar] [CrossRef]
  11. Chen, L.; Liu, S.; Tao, Y. Regulating tumor suppressor genes: post-translational modifications. Signal Transduction and Targeted Therapy 2020, 5, 90. [Google Scholar] [CrossRef]
  12. Ardito, F.; Giuliani, M.; Perrone, D.; Troiano, G.; Lo Muzio, L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (Review). Int J Mol Med 2017, 40, 271–280. [Google Scholar] [CrossRef]
  13. Singh, V.; Ram, M.; Kumar, R.; Prasad, R.; Roy, B.K.; Singh, K.K. Phosphorylation: Implications in Cancer. Protein J 2017, 36, 1–6. [Google Scholar] [CrossRef]
  14. Yoshida, K.; Yokoi, A.; Yamamoto, T.; Hayashi, Y.; Nakayama, J.; Yokoi, T.; Yoshida, H.; Kato, T.; Kajiyama, H.; Yamamoto, Y. Aberrant Activation of Cell-Cycle-Related Kinases and the Potential Therapeutic Impact of PLK1 or CHEK1 Inhibition in Uterine Leiomyosarcoma. Clin Cancer Res 2022, 28, 2147–2159. [Google Scholar] [CrossRef]
  15. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Molecular Cancer 2018, 17, 58. [Google Scholar] [CrossRef] [PubMed]
  16. Turdo, A.; D'Accardo, C.; Glaviano, A.; Porcelli, G.; Colarossi, C.; Colarossi, L.; Mare, M.; Faldetta, N.; Modica, C.; Pistone, G.; et al. Targeting Phosphatases and Kinases: How to Checkmate Cancer. Front Cell Dev Biol 2021, 9, 690306. [Google Scholar] [CrossRef]
  17. Yang, Y.; Li, S.; Wang, Y.; Zhao, Y.; Li, Q. Protein tyrosine kinase inhibitor resistance in malignant tumors: molecular mechanisms and future perspective. Signal Transduction and Targeted Therapy 2022, 7, 329. [Google Scholar] [CrossRef] [PubMed]
  18. Shyam Sunder, S.; Sharma, U.C.; Pokharel, S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduction and Targeted Therapy 2023, 8, 262. [Google Scholar] [CrossRef] [PubMed]
  19. Cohen, P. Immune diseases caused by mutations in kinases and components of the ubiquitin system. Nat Immunol 2014, 15, 521–529. [Google Scholar] [CrossRef]
  20. Stenberg, K.A.; Riikonen, P.T.; Vihinen, M. KinMutBase, a database of human disease-causing protein kinase mutations. Nucleic Acids Res 2000, 28, 369–371. [Google Scholar] [CrossRef]
  21. Wilks, A.F. Structure and function of the protein tyrosine kinases. Progress in Growth Factor Research 1990, 2, 97–111. [Google Scholar] [CrossRef]
  22. Aschner, Y.; Downey, G.P. The Importance of Tyrosine Phosphorylation Control of Cellular Signaling Pathways in Respiratory Disease: pY and pY Not. Am J Respir Cell Mol Biol 2018, 59, 535–547. [Google Scholar] [CrossRef]
  23. Tautz, L.; Critton, D.A.; Grotegut, S. Protein tyrosine phosphatases: structure, function, and implication in human disease. Methods Mol Biol 2013, 1053, 179–221. [Google Scholar] [CrossRef] [PubMed]
  24. Vaparanta, K.; Jokilammi, A.; Tamirat, M.; Merilahti, J.A.M.; Salokas, K.; Varjosalo, M.; Ivaska, J.; Johnson, M.S.; Elenius, K. An extracellular receptor tyrosine kinase motif orchestrating intracellular STAT activation. Nat Commun 2022, 13, 6953. [Google Scholar] [CrossRef]
  25. Gonzalez-Magaldi, M.; McCabe, J.M.; Cartwright, H.N.; Sun, N.; Leahy, D.J. Conserved roles for receptor tyrosine kinase extracellular regions in regulating receptor and pathway activity. Biochem J 2020, 477, 4207–4220. [Google Scholar] [CrossRef] [PubMed]
  26. Yao, Z.; Stagljar, I. Multiple functions of protein phosphatases in receptor tyrosine kinase signaling revealed by interactome analysis. Mol Cell Oncol 2017, 4, e1297101. [Google Scholar] [CrossRef] [PubMed]
  27. Solouki, S.; August, A.; Huang, W. Non-receptor tyrosine kinase signaling in autoimmunity and therapeutic implications. Pharmacology & Therapeutics 2019, 201, 39–50. [Google Scholar] [CrossRef]
  28. Gocek, E.; Moulas, A.N.; Studzinski, G.P. Non-receptor protein tyrosine kinases signaling pathways in normal and cancer cells. Crit Rev Clin Lab Sci 2014, 51, 125–137. [Google Scholar] [CrossRef]
  29. Siveen, K.S.; Prabhu, K.S.; Achkar, I.W.; Kuttikrishnan, S.; Shyam, S.; Khan, A.Q.; Merhi, M.; Dermime, S.; Uddin, S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Molecular Cancer 2018, 17, 31. [Google Scholar] [CrossRef]
  30. Tomuleasa, C.; Tigu, A.-B.; Munteanu, R.; Moldovan, C.-S.; Kegyes, D.; Onaciu, A.; Gulei, D.; Ghiaur, G.; Einsele, H.; Croce, C.M. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduction and Targeted Therapy 2024, 9, 201. [Google Scholar] [CrossRef]
  31. Zhou, B.; Lin, W.; Long, Y.; Yang, Y.; Zhang, H.; Wu, K.; Chu, Q. Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduction and Targeted Therapy 2022, 7, 95. [Google Scholar] [CrossRef]
  32. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduction and Targeted Therapy 2021, 6, 218. [Google Scholar] [CrossRef]
  33. Hunter, T.; Cooper, J.A. Protein-tyrosine kinases. Annu Rev Biochem 1985, 54, 897–930. [Google Scholar] [CrossRef]
  34. Carpenter, G.; King, L., Jr.; Cohen, S. Epidermal growth factor stimulates phosphorylation in membrane preparations in vitro. Nature 1978, 276, 409–410. [Google Scholar] [CrossRef] [PubMed]
  35. Sawyers, C.L. Rational therapeutic intervention in cancer: kinases as drug targets. Curr Opin Genet Dev 2002, 12, 111–115. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, N.; Li, Y. Receptor tyrosine kinases: biological functions and anticancer targeted therapy. MedComm (2020) 2023, 4, e446. [Google Scholar] [CrossRef]
  37. Yu, J.; Fang, T.; Yun, C.; Liu, X.; Cai, X. Antibody-Drug Conjugates Targeting the Human Epidermal Growth Factor Receptor Family in Cancers. Front Mol Biosci 2022, 9, 847835. [Google Scholar] [CrossRef] [PubMed]
  38. Ye, P.; Wang, Y.; Li, R.; Chen, W.; Wan, L.; Cai, P. The HER family as therapeutic targets in colorectal cancer. Crit Rev Oncol Hematol 2022, 174, 103681. [Google Scholar] [CrossRef]
  39. Nair, S.; Bonner, J.A.; Bredel, M. EGFR Mutations in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2022, 23. [Google Scholar] [CrossRef]
  40. Harrison, P.T.; Vyse, S.; Huang, P.H. Rare epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer. Semin Cancer Biol 2020, 61, 167–179. [Google Scholar] [CrossRef]
  41. Vigneri, P.G.; Tirrò, E.; Pennisi, M.S.; Massimino, M.; Stella, S.; Romano, C.; Manzella, L. The Insulin/IGF System in Colorectal Cancer Development and Resistance to Therapy. Front Oncol 2015, 5, 230. [Google Scholar] [CrossRef]
  42. Chen, P.H.; Chen, X.; He, X. Platelet-derived growth factors and their receptors: structural and functional perspectives. Biochim Biophys Acta 2013, 1834, 2176–2186. [Google Scholar] [CrossRef]
  43. Tomassetti, C.; Insinga, G.; Gimigliano, F.; Morrione, A.; Giordano, A.; Giurisato, E. Insights into CSF-1R Expression in the Tumor Microenvironment. Biomedicines 2024, 12, 2381. [Google Scholar] [CrossRef]
  44. Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
  45. Cébe-Suarez, S.; Zehnder-Fjällman, A.; Ballmer-Hofer, K. The role of VEGF receptors in angiogenesis; complex partnerships. Cell Mol Life Sci 2006, 63, 601–615. [Google Scholar] [CrossRef]
  46. Liu, Z.-L.; Chen, H.-H.; Zheng, L.-L.; Sun, L.-P.; Shi, L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduction and Targeted Therapy 2023, 8, 198. [Google Scholar] [CrossRef] [PubMed]
  47. Farooq, M.; Khan, A.W.; Kim, M.S.; Choi, S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021, 10. [Google Scholar] [CrossRef]
  48. Xie, Y.; Su, N.; Yang, J.; Tan, Q.; Huang, S.; Jin, M.; Ni, Z.; Zhang, B.; Zhang, D.; Luo, F.; et al. FGF/FGFR signaling in health and disease. Signal Transduction and Targeted Therapy 2020, 5, 181. [Google Scholar] [CrossRef] [PubMed]
  49. Berger, H.; Wodarz, A.; Borchers, A. PTK7 Faces the Wnt in Development and Disease. Front Cell Dev Biol 2017, 5, 31. [Google Scholar] [CrossRef]
  50. Ji, J.; Qian, Q.; Cheng, W.; Ye, X.; Jing, A.; Ma, S.; Ding, Y.; Ma, X.; Wang, Y.; Sun, Q.; et al. FOXP4-mediated induction of PTK7 activates the Wnt/β-catenin pathway and promotes ovarian cancer development. Cell Death & Disease 2024, 15, 332. [Google Scholar] [CrossRef]
  51. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018, 15, 731–747. [Google Scholar] [CrossRef]
  52. Hechtman, J.F. NTRK insights: best practices for pathologists. Modern Pathology 2022, 35, 298–305. [Google Scholar] [CrossRef]
  53. Belliveau, D.J.; Krivko, I.; Kohn, J.; Lachance, C.; Pozniak, C.; Rusakov, D.; Kaplan, D.; Miller, F.D. NGF and neurotrophin-3 both activate TrkA on sympathetic neurons but differentially regulate survival and neuritogenesis. J Cell Biol 1997, 136, 375–388. [Google Scholar] [CrossRef] [PubMed]
  54. Menck, K.; Heinrichs, S.; Baden, C.; Bleckmann, A. The WNT/ROR Pathway in Cancer: From Signaling to Therapeutic Intervention. Cells 2021, 10. [Google Scholar] [CrossRef]
  55. Song, P.; Gao, Z.; Bao, Y.; Chen, L.; Huang, Y.; Liu, Y.; Dong, Q.; Wei, X. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. Journal of Hematology & Oncology 2024, 17, 46. [Google Scholar] [CrossRef]
  56. Hubbard, S.R.; Gnanasambandan, K. Structure and activation of MuSK, a receptor tyrosine kinase central to neuromuscular junction formation. Biochim Biophys Acta 2013, 1834, 2166–2169. [Google Scholar] [CrossRef]
  57. Cao, M.; Koneczny, I.; Vincent, A. Myasthenia Gravis With Antibodies Against Muscle Specific Kinase: An Update on Clinical Features, Pathophysiology and Treatment. Front Mol Neurosci 2020, 13, 159. [Google Scholar] [CrossRef]
  58. Gao, C.F.; Woude, G.F.V. HGF/SF-Met signaling in tumor progression. Cell Research 2005, 15, 49–51. [Google Scholar] [CrossRef] [PubMed]
  59. Raj, S.; Kesari, K.K.; Kumar, A.; Rathi, B.; Sharma, A.; Gupta, P.K.; Jha, S.K.; Jha, N.K.; Slama, P.; Roychoudhury, S.; et al. Molecular mechanism(s) of regulation(s) of c-MET/HGF signaling in head and neck cancer. Molecular Cancer 2022, 21, 31. [Google Scholar] [CrossRef]
  60. Linger, R.M.; Keating, A.K.; Earp, H.S.; Graham, D.K. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv Cancer Res 2008, 100, 35–83. [Google Scholar] [CrossRef] [PubMed]
  61. Tsou, W.I.; Nguyen, K.Q.; Calarese, D.A.; Garforth, S.J.; Antes, A.L.; Smirnov, S.V.; Almo, S.C.; Birge, R.B.; Kotenko, S.V. Receptor tyrosine kinases, TYRO3, AXL, and MER, demonstrate distinct patterns and complex regulation of ligand-induced activation. J Biol Chem 2014, 289, 25750–25763. [Google Scholar] [CrossRef]
  62. Aehnlich, P.; Powell, R.M.; Peeters, M.J.W.; Rahbech, A.; Thor Straten, P. TAM Receptor Inhibition-Implications for Cancer and the Immune System. Cancers (Basel) 2021, 13. [Google Scholar] [CrossRef]
  63. Graham, D.K.; DeRyckere, D.; Davies, K.D.; Earp, H.S. The TAM family: phosphatidylserine-sensing receptor tyrosine kinases gone awry in cancer. Nature Reviews Cancer 2014, 14, 769–785. [Google Scholar] [CrossRef] [PubMed]
  64. Vázquez-Bellón, N.; Martínez-Bosch, N.; García de Frutos, P.; Navarro, P. Hallmarks of pancreatic cancer: spotlight on TAM receptors. eBioMedicine 2024, 107. [Google Scholar] [CrossRef] [PubMed]
  65. Korhonen, E.A.; Lampinen, A.; Giri, H.; Anisimov, A.; Kim, M.; Allen, B.; Fang, S.; D'Amico, G.; Sipilä, T.J.; Lohela, M.; et al. Tie1 controls angiopoietin function in vascular remodeling and inflammation. J Clin Invest 2016, 126, 3495–3510. [Google Scholar] [CrossRef]
  66. Saharinen, P.; Eklund, L.; Alitalo, K. Therapeutic targeting of the angiopoietin–TIE pathway. Nature Reviews Drug Discovery 2017, 16, 635–661. [Google Scholar] [CrossRef]
  67. Leppänen, V.M.; Saharinen, P.; Alitalo, K. Structural basis of Tie2 activation and Tie2/Tie1 heterodimerization. Proc Natl Acad Sci U S A 2017, 114, 4376–4381. [Google Scholar] [CrossRef]
  68. Zhang, J.; Hughes, S. Role of the ephrin and Eph receptor tyrosine kinase families in angiogenesis and development of the cardiovascular system. J Pathol 2006, 208, 453–461. [Google Scholar] [CrossRef] [PubMed]
  69. Liang, L.-Y.; Patel, O.; Janes, P.W.; Murphy, J.M.; Lucet, I.S. Eph receptor signalling: from catalytic to non-catalytic functions. Oncogene 2019, 38, 6567–6584. [Google Scholar] [CrossRef]
  70. Darling, T.K.; Lamb, T.J. Emerging Roles for Eph Receptors and Ephrin Ligands in Immunity. Front Immunol 2019, 10, 1473. [Google Scholar] [CrossRef]
  71. Mahato, A.K.; Sidorova, Y.A. RET Receptor Tyrosine Kinase: Role in Neurodegeneration, Obesity, and Cancer. Int J Mol Sci 2020, 21. [Google Scholar] [CrossRef]
  72. Melillo, R.M.; Santoro, M. The RET Receptor Family. In Receptor Tyrosine Kinases: Family. In Receptor Tyrosine Kinases: Family and Subfamilies, Wheeler, D.L., Yarden, Y., Eds.; Springer International Publishing: Cham, 2015; pp. 559–591. [Google Scholar]
  73. Green, J.; Nusse, R.; van Amerongen, R. The role of Ryk and Ror receptor tyrosine kinases in Wnt signal transduction. Cold Spring Harb Perspect Biol 2014, 6. [Google Scholar] [CrossRef]
  74. Shi, F.; Mendrola, J.M.; Sheetz, J.B.; Wu, N.; Sommer, A.; Speer, K.F.; Noordermeer, J.N.; Kan, Z.-Y.; Perry, K.; Englander, S.W.; et al. ROR and RYK extracellular region structures suggest that receptor tyrosine kinases have distinct WNT-recognition modes. Cell Reports 2021, 37, 109834. [Google Scholar] [CrossRef] [PubMed]
  75. Leitinger, B. Discoidin domain receptor functions in physiological and pathological conditions. Int Rev Cell Mol Biol 2014, 310, 39–87. [Google Scholar] [CrossRef]
  76. Chen, L.; Kong, X.; Fang, Y.; Paunikar, S.; Wang, X.; Brown, J.A.L.; Bourke, E.; Li, X.; Wang, J. Recent Advances in the Role of Discoidin Domain Receptor Tyrosine Kinase 1 and Discoidin Domain Receptor Tyrosine Kinase 2 in Breast and Ovarian Cancer. Front Cell Dev Biol 2021, 9, 747314. [Google Scholar] [CrossRef] [PubMed]
  77. Toy, K.A.; Valiathan, R.R.; Núñez, F.; Kidwell, K.M.; Gonzalez, M.E.; Fridman, R.; Kleer, C.G. Tyrosine kinase discoidin domain receptors DDR1 and DDR2 are coordinately deregulated in triple-negative breast cancer. Breast Cancer Res Treat 2015, 150, 9–18. [Google Scholar] [CrossRef] [PubMed]
  78. Gao, Y.; Zhou, J.; Li, J. Discoidin domain receptors orchestrate cancer progression: A focus on cancer therapies. Cancer Sci 2021, 112, 962–969. [Google Scholar] [CrossRef]
  79. Shenoy, G.P.; Pal, R.; Purwarga Matada, G.S.; Singh, E.; Raghavendra, N.M.; Dhiwar, P.S. Discoidin domain receptor inhibitors as anticancer agents: A systematic review on recent development of DDRs inhibitors, their resistance and structure activity relationship. Bioorganic Chemistry 2023, 130, 106215. [Google Scholar] [CrossRef]
  80. Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9. [Google Scholar] [CrossRef]
  81. An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death & Disease 2024, 15, 556. [Google Scholar] [CrossRef]
  82. Ditsiou, A.; Gagliano, T.; Samuels, M.; Vella, V.; Tolias, C.; Giamas, G. The multifaceted role of lemur tyrosine kinase 3 in health and disease. Open Biol 2021, 11, 210218. [Google Scholar] [CrossRef]
  83. Mórotz, G.M.; Bradbury, N.A.; Caluseriu, O.; Hisanaga, S.-i.; Miller, C.C.J.; Swiatecka-Urban, A.; Lenz, H.-J.; Moss, S.J.; Giamas, G. A revised nomenclature for the lemur family of protein kinases. Communications Biology 2024, 7, 57. [Google Scholar] [CrossRef]
  84. Bencze, J.; Szarka, M.; Bencs, V.; Szabó, R.N.; Smajda, M.; Aarsland, D.; Hortobágyi, T. Neuropathological characterization of Lemur tyrosine kinase 2 (LMTK2) in Alzheimer’s disease and neocortical Lewy body disease. Scientific Reports 2019, 9, 17222. [Google Scholar] [CrossRef]
  85. Della Corte, C.M.; Viscardi, G.; Di Liello, R.; Fasano, M.; Martinelli, E.; Troiani, T.; Ciardiello, F.; Morgillo, F. Role and targeting of anaplastic lymphoma kinase in cancer. Molecular Cancer 2018, 17, 30. [Google Scholar] [CrossRef]
  86. Huang, H. Anaplastic Lymphoma Kinase (ALK) Receptor Tyrosine Kinase: A Catalytic Receptor with Many Faces. International Journal of Molecular Sciences 2018, 19, 3448. [Google Scholar] [CrossRef]
  87. Webb, T.R.; Slavish, J.; George, R.E.; Look, A.T.; Xue, L.; Jiang, Q.; Cui, X.; Rentrop, W.B.; Morris, S.W. Anaplastic lymphoma kinase: role in cancer pathogenesis and small-molecule inhibitor development for therapy. Expert Rev Anticancer Ther 2009, 9, 331–356. [Google Scholar] [CrossRef] [PubMed]
  88. Hallberg, B.; Palmer, R.H. The role of the ALK receptor in cancer biology. Annals of Oncology 2016, 27, iii4–iii15. [Google Scholar] [CrossRef] [PubMed]
  89. Hu, L.; Chen, H.Y.; Cai, J.; Zhang, Y.; Qi, C.Y.; Gong, H.; Zhai, Y.X.; Fu, H.; Yang, G.Z.; Gao, C.F. Serine threonine tyrosine kinase 1 is a potential prognostic marker in colorectal cancer. BMC Cancer 2015, 15, 246. [Google Scholar] [CrossRef]
  90. Rajpurohit, Y.S.; Sharma, D.K.; Misra, H.S. Involvement of serine / threonine protein kinases in DNA damage response and cell division in bacteria. Research in Microbiology 2022, 173, 103883. [Google Scholar] [CrossRef] [PubMed]
  91. Siveen, K.S.; Prabhu, K.S.; Achkar, I.W.; Kuttikrishnan, S.; Shyam, S.; Khan, A.Q.; Merhi, M.; Dermime, S.; Uddin, S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol Cancer 2018, 17, 31. [Google Scholar] [CrossRef]
  92. Hubbard, S.R.; Miller, W.T. Receptor tyrosine kinases: mechanisms of activation and signaling. Curr Opin Cell Biol 2007, 19, 117–123. [Google Scholar] [CrossRef]
  93. Kan, Y.; Paung, Y.; Seeliger, M.A.; Miller, W.T. Domain Architecture of the Nonreceptor Tyrosine Kinase Ack1. Cells 2023, 12. [Google Scholar] [CrossRef]
  94. Seok, S.H. Structural Insights into Protein Regulation by Phosphorylation and Substrate Recognition of Protein Kinases/Phosphatases. Life (Basel) 2021, 11. [Google Scholar] [CrossRef] [PubMed]
  95. Prieto-Echagüe, V.; Gucwa, A.; Craddock, B.P.; Brown, D.A.; Miller, W.T. Cancer-associated mutations activate the nonreceptor tyrosine kinase Ack1. J Biol Chem 2010, 285, 10605–10615. [Google Scholar] [CrossRef] [PubMed]
  96. Hubbard, S.R.; Till, J.H. Protein tyrosine kinase structure and function. Annu Rev Biochem 2000, 69, 373–398. [Google Scholar] [CrossRef] [PubMed]
  97. Sun, G.; Ayrapetov, M.K. Dissection of the catalytic and regulatory structure-function relationships of Csk protein tyrosine kinase. Front Cell Dev Biol 2023, 11, 1148352. [Google Scholar] [CrossRef]
  98. Gan, W.; Roux, B. Binding specificity of SH2 domains: insight from free energy simulations. Proteins 2009, 74, 996–1007. [Google Scholar] [CrossRef]
  99. Pawson, T.; Gish, G.D.; Nash, P. SH2 domains, interaction modules and cellular wiring. Trends Cell Biol 2001, 11, 504–511. [Google Scholar] [CrossRef]
  100. Mahajan, K.; Mahajan, N.P. ACK1/TNK2 tyrosine kinase: molecular signaling and evolving role in cancers. Oncogene 2015, 34, 4162–4167. [Google Scholar] [CrossRef]
  101. Yokoyama, N.; Miller, W.T. Biochemical properties of the Cdc42-associated tyrosine kinase ACK1. Substrate specificity, authphosphorylation, and interaction with Hck. J Biol Chem 2003, 278, 47713–47723. [Google Scholar] [CrossRef]
  102. Yang, W.; Cerione, R.A. Cloning and Characterization of a Novel Cdc42-associated Tyrosine Kinase, ACK-2, from Bovine Brain*. Journal of Biological Chemistry 1997, 272, 24819–24824. [Google Scholar] [CrossRef]
  103. Prieto-Echagüe, V.; Miller, W.T. Regulation of ack-family nonreceptor tyrosine kinases. J Signal Transduct 2011, 2011, 742372. [Google Scholar] [CrossRef]
  104. Gajiwala, K.S.; Maegley, K.; Ferre, R.; He, Y.A.; Yu, X. Ack1: activation and regulation by allostery. PLoS One 2013, 8, e53994. [Google Scholar] [CrossRef] [PubMed]
  105. Ahmed, S.; Miller, W.T. The noncatalytic regions of the tyrosine kinase Tnk1 are important for activity and substrate specificity. Journal of Biological Chemistry 2022, 298, 102664. [Google Scholar] [CrossRef] [PubMed]
  106. Sawant, M.; Wilson, A.; Sridaran, D.; Mahajan, K.; O'Conor, C.J.; Hagemann, I.S.; Luo, J.; Weimholt, C.; Li, T.; Roa, J.C.; et al. Epigenetic reprogramming of cell cycle genes by ACK1 promotes breast cancer resistance to CDK4/6 inhibitor. Oncogene 2023, 42, 2263–2277. [Google Scholar] [CrossRef]
  107. Sridaran, D.; Chouhan, S.; Mahajan, K.; Renganathan, A.; Weimholt, C.; Bhagwat, S.; Reimers, M.; Kim, E.H.; Thakur, M.K.; Saeed, M.A.; et al. Inhibiting ACK1-mediated phosphorylation of C-terminal Src kinase counteracts prostate cancer immune checkpoint blockade resistance. Nature Communications 2022, 13, 6929. [Google Scholar] [CrossRef] [PubMed]
  108. Yamaoka, K.; Saharinen, P.; Pesu, M.; Holt, V.E., 3rd; Silvennoinen, O.; O'Shea, J.J. The Janus kinases (Jaks). Genome Biol 2004, 5, 253. [Google Scholar] [CrossRef]
  109. Lupardus, P.J.; Ultsch, M.; Wallweber, H.; Bir Kohli, P.; Johnson, A.R.; Eigenbrot, C. Structure of the pseudokinase-kinase domains from protein kinase TYK2 reveals a mechanism for Janus kinase (JAK) autoinhibition. Proc Natl Acad Sci U S A 2014, 111, 8025–8030. [Google Scholar] [CrossRef]
  110. Ghoreschi, K.; Laurence, A.; O'Shea, J.J. Janus kinases in immune cell signaling. Immunol Rev 2009, 228, 273–287. [Google Scholar] [CrossRef]
  111. Caveney, N.A.; Saxton, R.A.; Waghray, D.; Glassman, C.R.; Tsutsumi, N.; Hubbard, S.R.; Garcia, K.C. Structural basis of Janus kinase trans-activation. Cell Rep 2023, 42, 112201. [Google Scholar] [CrossRef]
  112. Garrido-Trigo, A.; Salas, A. Molecular Structure and Function of Janus Kinases: Implications for the Development of Inhibitors. Journal of Crohn's and Colitis 2019, 14, S713–S724. [Google Scholar] [CrossRef]
  113. Xue, C.; Yao, Q.; Gu, X.; Shi, Q.; Yuan, X.; Chu, Q.; Bao, Z.; Lu, J.; Li, L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduction and Targeted Therapy 2023, 8, 204. [Google Scholar] [CrossRef]
  114. Villarino, A.V.; Kanno, Y.; Ferdinand, J.R.; O'Shea, J.J. Mechanisms of Jak/STAT signaling in immunity and disease. J Immunol 2015, 194, 21–27. [Google Scholar] [CrossRef] [PubMed]
  115. Hu, X.; li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduction and Targeted Therapy 2021, 6, 402. [Google Scholar] [CrossRef]
  116. Lv, Y.; Qi, J.; Babon, J.J.; Cao, L.; Fan, G.; Lang, J.; Zhang, J.; Mi, P.; Kobe, B.; Wang, F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduction and Targeted Therapy 2024, 9, 221. [Google Scholar] [CrossRef] [PubMed]
  117. Rah, B.; Rather, R.A.; Bhat, G.R.; Baba, A.B.; Mushtaq, I.; Farooq, M.; Yousuf, T.; Dar, S.B.; Parveen, S.; Hassan, R.; et al. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022, 13, 821344. [Google Scholar] [CrossRef]
  118. Groffen, J.; Heisterkamp, N.; Shibuya, M.; Hanafusa, H.; Stephenson, J.R. Transforming genes of avian (v-ƒps) and mammalian (v-ƒes) retroviruses correspond to a common cellular locus. Virology 1983, 125, 480–486. [Google Scholar] [CrossRef] [PubMed]
  119. Craig, A.W.B. FES/FER kinase signaling in hematopoietic cells and leukemias. FBL 2012, 17, 861–875. [Google Scholar] [CrossRef]
  120. Laurent, C.E.; Delfino, F.J.; Cheng, H.Y.; Smithgall, T.E. The human c-Fes tyrosine kinase binds tubulin and microtubules through separate domains and promotes microtubule assembly. Mol Cell Biol 2004, 24, 9351–9358. [Google Scholar] [CrossRef]
  121. Hellwig, S.; Miduturu, C.V.; Kanda, S.; Zhang, J.; Filippakopoulos, P.; Salah, E.; Deng, X.; Choi, H.G.; Zhou, W.; Hur, W.; et al. Small-molecule inhibitors of the c-Fes protein-tyrosine kinase. Chem Biol 2012, 19, 529–540. [Google Scholar] [CrossRef]
  122. Zhang, S.; Chitu, V.; Stanley, E.R.; Elliott, B.E.; Greer, P.A. Fes tyrosine kinase expression in the tumor niche correlates with enhanced tumor growth, angiogenesis, circulating tumor cells, metastasis, and infiltrating macrophages. Cancer Res 2011, 71, 1465–1473. [Google Scholar] [CrossRef]
  123. Ivanova, I.A.; Vermeulen, J.F.; Ercan, C.; Houthuijzen, J.M.; Saig, F.A.; Vlug, E.J.; van der Wall, E.; van Diest, P.J.; Vooijs, M.; Derksen, P.W.B. FER kinase promotes breast cancer metastasis by regulating α6- and β1-integrin-dependent cell adhesion and anoikis resistance. Oncogene 2013, 32, 5582–5592. [Google Scholar] [CrossRef]
  124. Ivanova, I.A.; Arulanantham, S.; Barr, K.; Cepeda, M.; Parkins, K.M.; Hamilton, A.M.; Johnston, D.; Penuela, S.; Hess, D.A.; Ronald, J.A.; et al. Targeting FER Kinase Inhibits Melanoma Growth and Metastasis. Cancers 2019, 11, 419. [Google Scholar] [CrossRef]
  125. Rogers, J.A.; Read, R.D.; Li, J.; Peters, K.L.; Smithgall, T.E. Autophosphorylation of the Fes Tyrosine Kinase: EVIDENCE FOR AN INTERMOLECULAR MECHANISM INVOLVING TWO KINASE DOMAIN TYROSINE RESIDUES*. Journal of Biological Chemistry 1996, 271, 17519–17525. [Google Scholar] [CrossRef]
  126. Menegon, A.; Burgaya, F.; Baudot, P.; Dunlap, D.D.; Girault, J.A.; Valtorta, F. FAK+ and PYK2/CAKbeta, two related tyrosine kinases highly expressed in the central nervous system: similarities and differences in the expression pattern. Eur J Neurosci 1999, 11, 3777–3788. [Google Scholar] [CrossRef]
  127. Tapial Martínez, P.; López Navajas, P.; Lietha, D. FAK Structure and Regulation by Membrane Interactions and Force in Focal Adhesions. Biomolecules 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  128. Cooper, L.A.; Shen, T.L.; Guan, J.L. Regulation of focal adhesion kinase by its amino-terminal domain through an autoinhibitory interaction. Mol Cell Biol 2003, 23, 8030–8041. [Google Scholar] [CrossRef] [PubMed]
  129. Prutzman, K.C.; Gao, G.; King, M.L.; Iyer, V.V.; Mueller, G.A.; Schaller, M.D.; Campbell, S.L. The focal adhesion targeting domain of focal adhesion kinase contains a hinge region that modulates tyrosine 926 phosphorylation. Structure 2004, 12, 881–891. [Google Scholar] [CrossRef]
  130. Kadaré, G.; Gervasi, N.; Brami-Cherrier, K.; Blockus, H.; El Messari, S.; Arold, S.T.; Girault, J.A. Conformational dynamics of the focal adhesion targeting domain control specific functions of focal adhesion kinase in cells. J Biol Chem 2015, 290, 478–491. [Google Scholar] [CrossRef] [PubMed]
  131. Shen, Y.; Schaller, M.D. Focal adhesion targeting: the critical determinant of FAK regulation and substrate phosphorylation. Mol Biol Cell 1999, 10, 2507–2518. [Google Scholar] [CrossRef]
  132. Birge, R.B.; Kalodimos, C.; Inagaki, F.; Tanaka, S. Crk and CrkL adaptor proteins: networks for physiological and pathological signaling. Cell Communication and Signaling 2009, 7, 13. [Google Scholar] [CrossRef]
  133. Schlaepfer, D.D.; Hauck, C.R.; Sieg, D.J. Signaling through focal adhesion kinase. Prog Biophys Mol Biol 1999, 71, 435–478. [Google Scholar] [CrossRef]
  134. Rigiracciolo, D.C.; Cirillo, F.; Talia, M.; Muglia, L.; Gutkind, J.S.; Maggiolini, M.; Lappano, R. Focal Adhesion Kinase Fine Tunes Multifaced Signals toward Breast Cancer Progression. Cancers (Basel) 2021, 13. [Google Scholar] [CrossRef] [PubMed]
  135. Golubovskaya, V.M. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci (Landmark Ed) 2014, 19, 687–706. [Google Scholar] [CrossRef]
  136. Yoon, H.; Dehart, J.P.; Murphy, J.M.; Lim, S.T. Understanding the roles of FAK in cancer: inhibitors, genetic models, and new insights. J Histochem Cytochem 2015, 63, 114–128. [Google Scholar] [CrossRef] [PubMed]
  137. Murphy, J.M.; Rodriguez, Y.A.R.; Jeong, K.; Ahn, E.-Y.E.; Lim, S.-T.S. Targeting focal adhesion kinase in cancer cells and the tumor microenvironment. Experimental & Molecular Medicine 2020, 52, 877–886. [Google Scholar] [CrossRef]
  138. Kanteti, R.; Batra, S.K.; Lennon, F.E.; Salgia, R. FAK and paxillin, two potential targets in pancreatic cancer. Oncotarget 2016, 7, 31586–31601. [Google Scholar] [CrossRef]
  139. Davidson, C.; Taggart, D.; Sims, A.H.; Lonergan, D.W.; Canel, M.; Serrels, A. FAK promotes stromal PD-L2 expression associated with poor survival in pancreatic cancer. British Journal of Cancer 2022, 127, 1893–1905. [Google Scholar] [CrossRef]
  140. Roberts, J.M.; Tarafdar, S.; Joseph, R.E.; Andreotti, A.H.; Smithgall, T.E.; Engen, J.R.; Wales, T.E. Dynamics of the Tec-family tyrosine kinase SH3 domains. Protein Sci 2016, 25, 852–864. [Google Scholar] [CrossRef]
  141. Yin, Z.; Zou, Y.; Wang, D.; Huang, X.; Xiong, S.; Cao, L.; Zhang, Y.; Sun, Y.; Zhang, N. Regulation of the Tec family of non-receptor tyrosine kinases in cardiovascular disease. Cell Death Discovery 2022, 8, 119. [Google Scholar] [CrossRef]
  142. Yang, W.-C.; Ghiotto, M.; Barbarat, B.; Olive, D. The Role of Tec Protein-tyrosine Kinase in T Cell Signaling *. Journal of Biological Chemistry 1999, 274, 607–617. [Google Scholar] [CrossRef]
  143. Hussain, A.; Yu, L.; Faryal, R.; Mohammad, D.K.; Mohamed, A.J.; Smith, C.I. TEC family kinases in health and disease--loss-of-function of BTK and ITK and the gain-of-function fusions ITK-SYK and BTK-SYK. Febs j 2011, 278, 2001–2010. [Google Scholar] [CrossRef]
  144. Boggon, T.J.; Eck, M.J. Structure and regulation of Src family kinases. Oncogene 2004, 23, 7918–7927. [Google Scholar] [CrossRef]
  145. Engen, J.R.; Wales, T.E.; Hochrein, J.M.; Meyn, M.A., 3rd; Banu Ozkan, S.; Bahar, I.; Smithgall, T.E. Structure and dynamic regulation of Src-family kinases. Cell Mol Life Sci 2008, 65, 3058–3073. [Google Scholar] [CrossRef] [PubMed]
  146. Ortiz, M.A.; Mikhailova, T.; Li, X.; Porter, B.A.; Bah, A.; Kotula, L. Src family kinases, adaptor proteins and the actin cytoskeleton in epithelial-to-mesenchymal transition. Cell Communication and Signaling 2021, 19, 67. [Google Scholar] [CrossRef]
  147. Kovács, M.; Németh, T.; Jakus, Z.; Sitaru, C.; Simon, E.; Futosi, K.; Botz, B.; Helyes, Z.; Lowell, C.A.; Mócsai, A. The Src family kinases Hck, Fgr, and Lyn are critical for the generation of the in vivo inflammatory environment without a direct role in leukocyte recruitment. J Exp Med 2014, 211, 1993–2011. [Google Scholar] [CrossRef] [PubMed]
  148. Marhäll, A.; Kazi, J.U.; Rönnstrand, L. The Src family kinase LCK cooperates with oncogenic FLT3/ITD in cellular transformation. Scientific Reports 2017, 7, 13734. [Google Scholar] [CrossRef]
  149. Pestina, T.I.; Stenberg, P.E.; Druker, B.J.; Steward, S.A.; Hutson, N.K.; Barrie, R.J.; Jackson, C.W. Identification of the Src family kinases, Lck and Fgr in platelets. Their tyrosine phosphorylation status and subcellular distribution compared with other Src family members. Arterioscler Thromb Vasc Biol 1997, 17, 3278–3285. [Google Scholar] [CrossRef] [PubMed]
  150. Abram, C.L.; Lowell, C.A. The diverse functions of Src family kinases in macrophages. Front Biosci 2008, 13, 4426–4450. [Google Scholar] [CrossRef]
  151. Pelaz, S.G.; Tabernero, A. Src: coordinating metabolism in cancer. Oncogene 2022, 41, 4917–4928. [Google Scholar] [CrossRef]
  152. Goel, R.K.; Lukong, K.E. Understanding the cellular roles of Fyn-related kinase (FRK): implications in cancer biology. Cancer Metastasis Rev 2016, 35, 179–199. [Google Scholar] [CrossRef]
  153. Goel, R.K.; Lukong, K.E. Tracing the footprints of the breast cancer oncogene BRK — Past till present. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 2015, 1856, 39–54. [Google Scholar] [CrossRef]
  154. Goel, R.K.; Kim, N.; Lukong, K.E. Seeking a better understanding of the non-receptor tyrosine kinase, SRMS. Heliyon 2023, 9, e16421. [Google Scholar] [CrossRef] [PubMed]
  155. Liang, X.; Lu, Y.; Wilkes, M.; Neubert, T.A.; Resh, M.D. The N-terminal SH4 region of the Src family kinase Fyn is modified by methylation and heterogeneous fatty acylation: role in membrane targeting, cell adhesion, and spreading. J Biol Chem 2004, 279, 8133–8139. [Google Scholar] [CrossRef] [PubMed]
  156. Fhu, C.W.; Ali, A. Protein Lipidation by Palmitoylation and Myristoylation in Cancer. Front Cell Dev Biol 2021, 9, 673647. [Google Scholar] [CrossRef] [PubMed]
  157. Bagnato, G.; Leopizzi, M.; Urciuoli, E.; Peruzzi, B. Nuclear Functions of the Tyrosine Kinase Src. International Journal of Molecular Sciences 2020, 21, 2675. [Google Scholar] [CrossRef]
  158. Goel, R.K.; Miah, S.; Black, K.; Kalra, N.; Dai, C.; Lukong, K.E. The unique N-terminal region of SRMS regulates enzymatic activity and phosphorylation of its novel substrate docking protein 1. Febs j 2013, 280, 4539–4559. [Google Scholar] [CrossRef]
  159. Sen, B.; Johnson, F.M. Regulation of SRC family kinases in human cancers. J Signal Transduct 2011, 2011, 865819. [Google Scholar] [CrossRef]
  160. Kinoshita-Kikuta, E.; Utsumi, T.; Miyazaki, A.; Tokumoto, C.; Doi, K.; Harada, H.; Kinoshita, E.; Koike, T. Protein-N-myristoylation-dependent phosphorylation of serine 13 of tyrosine kinase Lyn by casein kinase 1γ at the Golgi during intracellular protein traffic. Sci Rep 2020, 10, 16273. [Google Scholar] [CrossRef]
  161. Berclaz, G.; Altermatt, H.J.; Rohrbach, V.; Dreher, E.; Ziemiecki, A.; Andres, A.C. Hormone-dependent nuclear localization of the tyrosine kinase iyk in the normal human breast epithelium and loss of expression during carcinogenesis. Int J Cancer 2000, 85, 889–894. [Google Scholar] [CrossRef]
  162. Annerén, C.; Welsh, M.; Jansson, L. Glucose intolerance and reduced islet blood flow in transgenic mice expressing the FRK tyrosine kinase under the control of the rat insulin promoter. Am J Physiol Endocrinol Metab 2007, 292, E1183–1190. [Google Scholar] [CrossRef]
  163. Gu, J.J.; Ryu, J.R.; Pendergast, A.M. Abl tyrosine kinases in T-cell signaling. Immunol Rev 2009, 228, 170–183. [Google Scholar] [CrossRef]
  164. Colicelli, J. ABL tyrosine kinases: evolution of function, regulation, and specificity. Sci Signal 2010, 3, re6. [Google Scholar] [CrossRef] [PubMed]
  165. Panjarian, S.; Iacob, R.E.; Chen, S.; Engen, J.R.; Smithgall, T.E. Structure and Dynamic Regulation of Abl Kinases*. Journal of Biological Chemistry 2013, 288, 5443–5450. [Google Scholar] [CrossRef]
  166. Preyer, M.; Vigneri, P.; Wang, J.Y. Interplay between kinase domain autophosphorylation and F-actin binding domain in regulating imatinib sensitivity and nuclear import of BCR-ABL. PLoS One 2011, 6, e17020. [Google Scholar] [CrossRef]
  167. Underhill-Day, N.; Pierce, A.; Thompson, S.E.; Xenaki, D.; Whetton, A.D.; Owen-Lynch, P.J. Role of the C-terminal actin binding domain in BCR/ABL-mediated survival and drug resistance. Br J Haematol 2006, 132, 774–783. [Google Scholar] [CrossRef] [PubMed]
  168. Smith, K.M.; Van Etten, R.A. Activation of c-Abl kinase activity and transformation by a chemical inducer of dimerization. J Biol Chem 2001, 276, 24372–24379. [Google Scholar] [CrossRef]
  169. Greuber, E.K.; Smith-Pearson, P.; Wang, J.; Pendergast, A.M. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nature Reviews Cancer 2013, 13, 559–571. [Google Scholar] [CrossRef]
  170. Ganguly, S.S.; Fiore, L.S.; Sims, J.T.; Friend, J.W.; Srinivasan, D.; Thacker, M.A.; Cibull, M.L.; Wang, C.; Novak, M.; Kaetzel, D.M.; et al. c-Abl and Arg are activated in human primary melanomas, promote melanoma cell invasion via distinct pathways, and drive metastatic progression. Oncogene 2012, 31, 1804–1816. [Google Scholar] [CrossRef] [PubMed]
  171. Ganguly, S.S.; Plattner, R. Activation of abl family kinases in solid tumors. Genes Cancer 2012, 3, 414–425. [Google Scholar] [CrossRef]
  172. Woodside, D.G.; Obergfell, A.; Talapatra, A.; Calderwood, D.A.; Shattil, S.J.; Ginsberg, M.H. The N-terminal SH2 domains of Syk and ZAP-70 mediate phosphotyrosine-independent binding to integrin beta cytoplasmic domains. J Biol Chem 2002, 277, 39401–39408. [Google Scholar] [CrossRef]
  173. Isakov, N.; Wange, R.L.; Burgess, W.H.; Watts, J.D.; Aebersold, R.; Samelson, L.E. ZAP-70 binding specificity to T cell receptor tyrosine-based activation motifs: the tandem SH2 domains of ZAP-70 bind distinct tyrosine-based activation motifs with varying affinity. J Exp Med 1995, 181, 375–380. [Google Scholar] [CrossRef]
  174. Hobbs, H.T.; Shah, N.H.; Badroos, J.M.; Gee, C.L.; Marqusee, S.; Kuriyan, J. Differences in the dynamics of the tandem-SH2 modules of the Syk and ZAP-70 tyrosine kinases. Protein Sci 2021, 30, 2373–2384. [Google Scholar] [CrossRef]
  175. Mócsai, A.; Ruland, J.; Tybulewicz, V.L.J. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nature Reviews Immunology 2010, 10, 387–402. [Google Scholar] [CrossRef]
  176. Qu, C.; Zheng, D.; Li, S.; Liu, Y.; Lidofsky, A.; Holmes, J.A.; Chen, J.; He, L.; Wei, L.; Liao, Y.; et al. Tyrosine kinase SYK is a potential therapeutic target for liver fibrosis. Hepatology 2018, 68, 1125–1139. [Google Scholar] [CrossRef] [PubMed]
  177. Zhou, F.; Hu, J.; Ma, H.; Harrison, M.L.; Geahlen, R.L. Nucleocytoplasmic trafficking of the Syk protein tyrosine kinase. Mol Cell Biol 2006, 26, 3478–3491. [Google Scholar] [CrossRef]
  178. Stamos, J.; Sliwkowski, M.X.; Eigenbrot, C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J Biol Chem 2002, 277, 46265–46272. [Google Scholar] [CrossRef] [PubMed]
  179. Eigenbrot, C. Structure-function of EGFR kinase domain and its inhibitors. In EGFR Signaling Networks in Cancer Therapy, Haley, J.D., Gullick, W.J., Eds.; Humana Press: Totowa, NJ, 2008; pp. 30–44. [Google Scholar]
  180. Roskoski, R. Src protein–tyrosine kinase structure and regulation. Biochemical and Biophysical Research Communications 2004, 324, 1155–1164. [Google Scholar] [CrossRef] [PubMed]
  181. Hubbard, S.R. Structural analysis of receptor tyrosine kinases. Progress in Biophysics and Molecular Biology 1999, 71, 343–358. [Google Scholar] [CrossRef]
  182. Lawrence, M.C.; Ward, C.W. Structural Features of the Receptor Tyrosine Kinase Ectodomains. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease, Wheeler, D.L., Yarden, Y., Eds.; Springer New York: New York, NY, 2015; pp. 163–193. [Google Scholar]
  183. Süveges, D.; Jura, N. Structural Features of the Kinase Domain. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease, Wheeler, D.L., Yarden, Y., Eds.; Springer New York: New York, NY, 2015; pp. 195–223. [Google Scholar]
  184. Eshaq, A.M.; Flanagan, T.W.; Hassan, S.-Y.; Al Asheikh, S.A.; Al-Amoudi, W.A.; Santourlidis, S.; Hassan, S.-L.; Alamodi, M.O.; Bendhack, M.L.; Alamodi, M.O.; et al. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers 2024, 16, 2754. [Google Scholar] [CrossRef] [PubMed]
  185. Brown, M.T.; Cooper, J.A. Regulation, substrates and functions of src. Biochim Biophys Acta 1996, 1287, 121–149. [Google Scholar] [CrossRef]
  186. Abram, C.L.; Courtneidge, S.A. Src family tyrosine kinases and growth factor signaling. Exp Cell Res 2000, 254, 1–13. [Google Scholar] [CrossRef]
  187. Roskoski, R., Jr. Src protein-tyrosine kinase structure and regulation. Biochem Biophys Res Commun 2004, 324, 1155–1164. [Google Scholar] [CrossRef]
  188. Xu, W.; Doshi, A.; Lei, M.; Eck, M.J.; Harrison, S.C. Crystal structures of c-Src reveal features of its autoinhibitory mechanism. Mol Cell 1999, 3, 629–638. [Google Scholar] [CrossRef] [PubMed]
  189. Williams, J.C.; Weijland, A.; Gonfloni, S.; Thompson, A.; Courtneidge, S.A.; Superti-Furga, G.; Wierenga, R.K. The 2.35 A crystal structure of the inactivated form of chicken Src: a dynamic molecule with multiple regulatory interactions. J Mol Biol 1997, 274, 757–775. [Google Scholar] [CrossRef] [PubMed]
  190. Sicheri, F.; Moarefi, I.; Kuriyan, J. Crystal structure of the Src family tyrosine kinase Hck. Nature 1997, 385, 602–609. [Google Scholar] [CrossRef]
  191. Xu, W.; Harrison, S.C.; Eck, M.J. Three-dimensional structure of the tyrosine kinase c-Src. Nature 1997, 385, 595–602. [Google Scholar] [CrossRef] [PubMed]
  192. Knighton, D.R.; Zheng, J.H.; Ten Eyck, L.F.; Ashford, V.A.; Xuong, N.H.; Taylor, S.S.; Sowadski, J.M. Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science 1991, 253, 407–414. [Google Scholar] [CrossRef]
  193. Okada, M.; Nakagawa, H. A protein tyrosine kinase involved in regulation of pp60c-src function. J Biol Chem 1989, 264, 20886–20893. [Google Scholar] [CrossRef]
  194. Zheng, X.M.; Resnick, R.J.; Shalloway, D. A phosphotyrosine displacement mechanism for activation of Src by PTPalpha. Embo j 2000, 19, 964–978. [Google Scholar] [CrossRef]
  195. EswarKumar, N.; Yang, C.-H.; Tewary, S.; Peng, W.-H.; Chen, G.-C.; Yeh, Y.-Q.; Yang, H.-C.; Ho, M.-C. An integrative approach unveils a distal encounter site for rPTPε and phospho-Src complex formation. Structure 2023, 31, 1567–1577.e1565. [Google Scholar] [CrossRef]
  196. Gazdar, A.F. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 2009, 28 Suppl 1, S24–31. [Google Scholar] [CrossRef]
  197. Bazley, L.A.; Gullick, W.J. The epidermal growth factor receptor family. Endocr Relat Cancer 2005, 12 Suppl 1, S17–27. [Google Scholar] [CrossRef]
  198. Kumar, A.; Petri, E.T.; Halmos, B.; Boggon, T.J. Structure and clinical relevance of the epidermal growth factor receptor in human cancer. J Clin Oncol 2008, 26, 1742–1751. [Google Scholar] [CrossRef]
  199. Schmitz, K.R.; Bagchi, A.; Roovers, R.C.; van Bergen en Henegouwen, P.M.; Ferguson, K.M. Structural evaluation of EGFR inhibition mechanisms for nanobodies/VHH domains. Structure 2013, 21, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  200. Ferguson, K.M. Structure-based view of epidermal growth factor receptor regulation. Annu Rev Biophys 2008, 37, 353–373. [Google Scholar] [CrossRef] [PubMed]
  201. Stamos, J.; Sliwkowski, M.X.; Eigenbrot, C. Structure of the Epidermal Growth Factor Receptor Kinase Domain Alone and in Complex with a 4-Anilinoquinazoline Inhibitor *. Journal of Biological Chemistry 2002, 277, 46265–46272. [Google Scholar] [CrossRef]
  202. Monsey, J.; Shen, W.; Schlesinger, P.; Bose, R. Her4 and Her2/neu Tyrosine Kinase Domains Dimerize and Activate in a Reconstituted <em>in Vitro</em> System *<sup> </sup>. Journal of Biological Chemistry 2010, 285, 7035–7044. [Google Scholar] [CrossRef] [PubMed]
  203. Jura, N.; Endres, N.F.; Engel, K.; Deindl, S.; Das, R.; Lamers, M.H.; Wemmer, D.E.; Zhang, X.; Kuriyan, J. Mechanism for Activation of the EGF Receptor Catalytic Domain by the Juxtamembrane Segment. Cell 2009, 137, 1293–1307. [Google Scholar] [CrossRef]
  204. Arkhipov, A.; Shan, Y.; Kim, E.T.; Dror, R.O.; Shaw, D.E. Her2 activation mechanism reflects evolutionary preservation of asymmetric ectodomain dimers in the human EGFR family. eLife 2013, 2, e00708. [Google Scholar] [CrossRef]
  205. Tsai, C.-J.; Nussinov, R. Emerging Allosteric Mechanism of EGFR Activation in Physiological and Pathological Contexts. Biophysical Journal 2019, 117, 5–13. [Google Scholar] [CrossRef]
  206. Schultz, D.F.; Billadeau, D.D.; Jois, S.D. EGFR trafficking: effect of dimerization, dynamics, and mutation. Frontiers in Oncology 2023, 13. [Google Scholar] [CrossRef]
  207. Jorissen, R.N.; Walker, F.; Pouliot, N.; Garrett, T.P.J.; Ward, C.W.; Burgess, A.W. Epidermal growth factor receptor: mechanisms of activation and signalling. Experimental Cell Research 2003, 284, 31–53. [Google Scholar] [CrossRef]
  208. Ono, M.; Kuwano, M. Molecular Mechanisms of Epidermal Growth Factor Receptor (EGFR) Activation and Response to Gefitinib and Other EGFR-Targeting Drugs. Clinical Cancer Research 2006, 12, 7242–7251. [Google Scholar] [CrossRef]
  209. Burgess, A.W.; Garrett, T.P.J. EGFR Receptor Family Extracellular Domain Structures and Functions. In EGFR Signaling Networks in Cancer Therapy, Haley, J.D., Gullick, W.J., Eds.; Humana Press: Totowa, NJ, 2008; pp. 2–13. [Google Scholar]
  210. Wan, S.; Wright, D.W.; Coveney, P.V. Mechanism of Drug Efficacy Within the EGF Receptor Revealed by Microsecond Molecular Dynamics Simulation. Molecular Cancer Therapeutics 2012, 11, 2394–2400. [Google Scholar] [CrossRef] [PubMed]
  211. Arkhipov, A.; Shan, Y.; Das, R.; Endres, Nicholas F. ; Eastwood, Michael P.; Wemmer, David E.; Kuriyan, J.; Shaw, David E. Architecture and Membrane Interactions of the EGF Receptor. Cell 2013, 152, 557–569. [Google Scholar] [CrossRef]
  212. Koland, J.G. Coarse-Grained Molecular Simulation of Epidermal Growth Factor Receptor Protein Tyrosine Kinase Multi-Site Self-Phosphorylation. PLOS Computational Biology 2014, 10, e1003435. [Google Scholar] [CrossRef] [PubMed]
  213. Songtawee, N.; Bevan, D.R.; Choowongkomon, K. Molecular dynamics of the asymmetric dimers of EGFR: Simulations on the active and inactive conformations of the kinase domain. Journal of Molecular Graphics and Modelling 2015, 58, 16–29. [Google Scholar] [CrossRef]
  214. Kaplan, M.; Narasimhan, S.; de Heus, C.; Mance, D.; van Doorn, S.; Houben, K.; Popov-Čeleketić, D.; Damman, R.; Katrukha, E.A.; Jain, P.; et al. EGFR Dynamics Change during Activation in Native Membranes as Revealed by NMR. Cell 2016, 167, 1241–1251.e1211. [Google Scholar] [CrossRef] [PubMed]
  215. EGFR Undergoes Ligand-Induced Conformational Changes during Activation. Cancer Discovery 2017, 7, 9–9. [CrossRef]
  216. Martin-Fernandez, M.L.; Clarke, D.T.; Roberts, S.K.; Zanetti-Domingues, L.C.; Gervasio, F.L. Structure and Dynamics of the EGF Receptor as Revealed by Experiments and Simulations and Its Relevance to Non-Small Cell Lung Cancer. Cells 2019, 8. [Google Scholar] [CrossRef]
  217. Levy, D.E.; Darnell, J.E., Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol 2002, 3, 651–662. [Google Scholar] [CrossRef]
  218. Darnell, J.E., Jr.; Kerr, I.M.; Stark, G.R. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994, 264, 1415–1421. [Google Scholar] [CrossRef]
  219. Rane, S.G.; Reddy, E.P. JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene 1994, 9, 2415–2423. [Google Scholar] [PubMed]
  220. Shigematsu, H.; Gazdar, A.F. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. Int J Cancer 2006, 118, 257–262. [Google Scholar] [CrossRef]
  221. Tatematsu, A.; Shimizu, J.; Murakami, Y.; Horio, Y.; Nakamura, S.; Hida, T.; Mitsudomi, T.; Yatabe, Y. Epidermal growth factor receptor mutations in small cell lung cancer. Clin Cancer Res 2008, 14, 6092–6096. [Google Scholar] [CrossRef] [PubMed]
  222. Yun, C.H.; Boggon, T.J.; Li, Y.; Woo, M.S.; Greulich, H.; Meyerson, M.; Eck, M.J. Structures of lung cancer-derived EGFR mutants and inhibitor complexes: mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell 2007, 11, 217–227. [Google Scholar] [CrossRef] [PubMed]
  223. Yoshikawa, S.; Kukimoto-Niino, M.; Parker, L.; Handa, N.; Terada, T.; Fujimoto, T.; Terazawa, Y.; Wakiyama, M.; Sato, M.; Sano, S.; et al. Structural basis for the altered drug sensitivities of non-small cell lung cancer-associated mutants of human epidermal growth factor receptor. Oncogene 2013, 32, 27–38. [Google Scholar] [CrossRef]
  224. Doss, G.P.; Rajith, B.; Chakraborty, C.; NagaSundaram, N.; Ali, S.K.; Zhu, H. Structural signature of the G719S-T790M double mutation in the EGFR kinase domain and its response to inhibitors. Sci Rep 2014, 4, 5868. [Google Scholar] [CrossRef]
  225. Sigismund, S.; Avanzato, D.; Lanzetti, L. Emerging functions of the EGFR in cancer. Mol Oncol 2018, 12, 3–20. [Google Scholar] [CrossRef]
  226. Tan, X.; Lambert, P.F.; Rapraeger, A.C.; Anderson, R.A. Stress-Induced EGFR Trafficking: Mechanisms, Functions, and Therapeutic Implications. Trends Cell Biol 2016, 26, 352–366. [Google Scholar] [CrossRef]
  227. Jutten, B.; Keulers, T.G.; Schaaf, M.B.; Savelkouls, K.; Theys, J.; Span, P.N.; Vooijs, M.A.; Bussink, J.; Rouschop, K.M. EGFR overexpressing cells and tumors are dependent on autophagy for growth and survival. Radiother Oncol 2013, 108, 479–483. [Google Scholar] [CrossRef]
  228. Casanova, M.L.; Larcher, F.; Casanova, B.; Murillas, R.; Fernández-Aceñero, M.J.; Villanueva, C.; Martínez-Palacio, J.; Ullrich, A.; Conti, C.J.; Jorcano, J.L. A critical role for ras-mediated, epidermal growth factor receptor-dependent angiogenesis in mouse skin carcinogenesis. Cancer Res 2002, 62, 3402–3407. [Google Scholar] [PubMed]
  229. Ekstrand, A.J.; Sugawa, N.; James, C.D.; Collins, V.P. Amplified and rearranged epidermal growth factor receptor genes in human glioblastomas reveal deletions of sequences encoding portions of the N- and/or C-terminal tails. Proc Natl Acad Sci U S A 1992, 89, 4309–4313. [Google Scholar] [CrossRef]
  230. Wong, A.J.; Ruppert, J.M.; Bigner, S.H.; Grzeschik, C.H.; Humphrey, P.A.; Bigner, D.S.; Vogelstein, B. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc Natl Acad Sci U S A 1992, 89, 2965–2969. [Google Scholar] [CrossRef]
  231. Sugawa, N.; Ekstrand, A.J.; James, C.D.; Collins, V.P. Identical splicing of aberrant epidermal growth factor receptor transcripts from amplified rearranged genes in human glioblastomas. Proc Natl Acad Sci U S A 1990, 87, 8602–8606. [Google Scholar] [CrossRef]
  232. Reynolds, A.B.; Roczniak-Ferguson, A. Emerging roles for p120-catenin in cell adhesion and cancer. Oncogene 2004, 23, 7947–7956. [Google Scholar] [CrossRef]
  233. Summy, J.M.; Gallick, G.E. Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev 2003, 22, 337–358. [Google Scholar] [CrossRef] [PubMed]
  234. Silva, C.M. Role of STATs as downstream signal transducers in Src family kinase-mediated tumorigenesis. Oncogene 2004, 23, 8017–8023. [Google Scholar] [CrossRef]
  235. Miranda, M.B.; Johnson, D.E. Signal transduction pathways that contribute to myeloid differentiation. Leukemia 2007, 21, 1363–1377. [Google Scholar] [CrossRef] [PubMed]
  236. Bai, L.; Lehnert, B.P.; Liu, J.; Neubarth, N.L.; Dickendesher, T.L.; Nwe, P.H.; Cassidy, C.; Woodbury, C.J.; Ginty, D.D. Genetic Identification of an Expansive Mechanoreceptor Sensitive to Skin Stroking. Cell 2015, 163, 1783–1795. [Google Scholar] [CrossRef]
  237. Ahmad, V.; Vadla, G.P.; Chabu, C.Y. Syd/JIP3 controls tissue size by regulating Diap1 protein turnover downstream of Yorkie/YAP. Dev Biol 2021, 469, 37–45. [Google Scholar] [CrossRef]
  238. Dong, Y.L.; Vadla, G.P.; Lu, J.J.; Ahmad, V.; Klein, T.J.; Liu, L.F.; Glazer, P.M.; Xu, T.; Chabu, C.Y. Cooperation between oncogenic Ras and wild-type p53 stimulates STAT non-cell autonomously to promote tumor radioresistance. Commun Biol 2021, 4, 374. [Google Scholar] [CrossRef] [PubMed]
  239. Guarino, M.; Rubino, B.; Ballabio, G. The role of epithelial-mesenchymal transition in cancer pathology. Pathology 2007, 39, 305–318. [Google Scholar] [CrossRef]
  240. Friedl, P.; Wolf, K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer 2003, 3, 362–374. [Google Scholar] [CrossRef] [PubMed]
  241. Delgado, L.; Monteiro, L.; Silva, P.; Bousbaa, H.; Garcez, F.; Silva, J.; Brilhante-Simões, P.; Pires, I.; Prada, J. BUBR1 as a Prognostic Biomarker in Canine Oral Squamous Cell Carcinoma. Animals (Basel) 2022, 12. [Google Scholar] [CrossRef]
  242. van der Geer, P.; Wiley, S.; Gish, G.D.; Pawson, T. The Shc adaptor protein is highly phosphorylated at conserved, twin tyrosine residues (Y239/240) that mediate protein-protein interactions. Curr Biol 1996, 6, 1435–1444. [Google Scholar] [CrossRef] [PubMed]
  243. Tokumitsu, Y.; Nakano, S.; Ueno, H.; Niho, Y. Suppression of malignant growth potentials of v-Src-transformed human gallbladder epithelial cells by adenovirus-mediated dominant negative H-Ras. J Cell Physiol 2000, 183, 221–227. [Google Scholar] [CrossRef]
  244. Jaber Chehayeb, R.; Stiegler, A.L.; Boggon, T.J. Crystal structures of p120RasGAP N-terminal SH2 domain in its apo form and in complex with a p190RhoGAP phosphotyrosine peptide. PLOS ONE 2020, 14, e0226113. [Google Scholar] [CrossRef]
  245. Biscardi, J.S.; Tice, D.A.; Parsons, S.J. c-Src, receptor tyrosine kinases, and human cancer. Adv Cancer Res 1999, 76, 61–119. [Google Scholar] [CrossRef]
  246. Ingley, E. Src family kinases: regulation of their activities, levels and identification of new pathways. Biochim Biophys Acta 2008, 1784, 56–65. [Google Scholar] [CrossRef]
  247. Cohen, P. Protein kinases--the major drug targets of the twenty-first century? Nat Rev Drug Discov 2002, 1, 309–315. [Google Scholar] [CrossRef]
  248. Yu, H.A.; Riely, G.J.; Lovly, C.M. Therapeutic strategies utilized in the setting of acquired resistance to EGFR tyrosine kinase inhibitors. Clin Cancer Res 2014, 20, 5898–5907. [Google Scholar] [CrossRef] [PubMed]
  249. Zeng, C.; Nie, D.; Wang, X.; Zhong, S.; Zeng, X.; Liu, X.; Qiu, K.; Peng, X.; Zhang, W.; Chen, S.; et al. Combined targeting of GPX4 and BCR-ABL tyrosine kinase selectively compromises BCR-ABL+ leukemia stem cells. Mol Cancer 2024, 23, 240. [Google Scholar] [CrossRef]
  250. Zhang, H.; To, K.K.W. Serum creatine kinase elevation following tyrosine kinase inhibitor treatment in cancer patients: Symptoms, mechanism, and clinical management. Clin Transl Sci 2024, 17, e70053. [Google Scholar] [CrossRef] [PubMed]
  251. Zhang, X.; Maity, T.; Kashyap, M.K.; Bansal, M.; Venugopalan, A.; Singh, S.; Awasthi, S.; Marimuthu, A.; Charles Jacob, H.K.; Belkina, N.; et al. Quantitative Tyrosine Phosphoproteomics of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor-treated Lung Adenocarcinoma Cells Reveals Potential Novel Biomarkers of Therapeutic Response. Mol Cell Proteomics 2017, 16, 891–910. [Google Scholar] [CrossRef] [PubMed]
  252. Zhong, L.; Zhao, Z.; Peng, X.; Zou, J.; Yang, S. Recent advances in small-molecular therapeutics for COVID-19. Precis Clin Med 2022, 5, pbac024. [Google Scholar] [CrossRef]
  253. Broekman, F.; Giovannetti, E.; Peters, G.J. Tyrosine kinase inhibitors: Multi-targeted or single-targeted? World J Clin Oncol 2011, 2, 80–93. [Google Scholar] [CrossRef]
  254. Krug, M.; Hilgeroth, A. Recent advances in the development of multi-kinase inhibitors. Mini Rev Med Chem 2008, 8, 1312–1327. [Google Scholar] [CrossRef]
  255. Agrawal, M.; Garg, R.J.; Cortes, J.; Quintás-Cardama, A. Tyrosine kinase inhibitors: the first decade. Curr Hematol Malig Rep 2010, 5, 70–80. [Google Scholar] [CrossRef]
  256. Yang, J.C.; Shih, J.Y.; Su, W.C.; Hsia, T.C.; Tsai, C.M.; Ou, S.H.; Yu, C.J.; Chang, G.C.; Ho, C.L.; Sequist, L.V.; et al. Afatinib for patients with lung adenocarcinoma and epidermal growth factor receptor mutations (LUX-Lung 2): a phase 2 trial. Lancet Oncol 2012, 13, 539–548. [Google Scholar] [CrossRef]
  257. Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011, 3, 75ra26. [Google Scholar] [CrossRef]
  258. Sequist, L.V.; Besse, B.; Lynch, T.J.; Miller, V.A.; Wong, K.K.; Gitlitz, B.; Eaton, K.; Zacharchuk, C.; Freyman, A.; Powell, C.; et al. Neratinib, an irreversible pan-ErbB receptor tyrosine kinase inhibitor: results of a phase II trial in patients with advanced non-small-cell lung cancer. J Clin Oncol 2010, 28, 3076–3083. [Google Scholar] [CrossRef] [PubMed]
  259. Pietanza, M.C.; Lynch, T.J., Jr.; Lara, P.N., Jr.; Cho, J.; Yanagihara, R.H.; Vrindavanam, N.; Chowhan, N.M.; Gadgeel, S.M.; Pennell, N.A.; Funke, R.; et al. XL647--a multitargeted tyrosine kinase inhibitor: results of a phase II study in subjects with non-small cell lung cancer who have progressed after responding to treatment with either gefitinib or erlotinib. J Thorac Oncol 2012, 7, 219–226. [Google Scholar] [CrossRef] [PubMed]
  260. Miller, V.A.; Hirsh, V.; Cadranel, J.; Chen, Y.M.; Park, K.; Kim, S.W.; Zhou, C.; Su, W.C.; Wang, M.; Sun, Y.; et al. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol 2012, 13, 528–538. [Google Scholar] [CrossRef]
  261. Katakami, N.; Atagi, S.; Goto, K.; Hida, T.; Horai, T.; Inoue, A.; Ichinose, Y.; Koboyashi, K.; Takeda, K.; Kiura, K.; et al. LUX-Lung 4: a phase II trial of afatinib in patients with advanced non-small-cell lung cancer who progressed during prior treatment with erlotinib, gefitinib, or both. J Clin Oncol 2013, 31, 3335–3341. [Google Scholar] [CrossRef] [PubMed]
  262. Reckamp, K.L.; Giaccone, G.; Camidge, D.R.; Gadgeel, S.M.; Khuri, F.R.; Engelman, J.A.; Koczywas, M.; Rajan, A.; Campbell, A.K.; Gernhardt, D.; et al. A phase 2 trial of dacomitinib (PF-00299804), an oral, irreversible pan-HER (human epidermal growth factor receptor) inhibitor, in patients with advanced non-small cell lung cancer after failure of prior chemotherapy and erlotinib. Cancer 2014, 120, 1145–1154. [Google Scholar] [CrossRef]
  263. Pollak, M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 2008, 8, 915–928. [Google Scholar] [CrossRef]
  264. Camidge, D.R.; Bazhenova, L.; Salgia, R.; Weiss, G.J.; Langer, C.J.; Shaw, A.T.; Narasimhan, N.I.; Dorer, D.J.; Rivera, V.M.; Zhang, J.; et al. First-in-human dose-finding study of the ALK/EGFR inhibitor AP26113 in patients with advanced malignancies: Updated results. Journal of Clinical Oncology 31, 8031. [Google Scholar] [CrossRef]
  265. Sequist, L.V.; Soria, J.-C.; Gadgeel, S.M.; Wakelee, H.A.; Camidge, D.R.; Varga, A.; Solomon, B.J.; Papadimitrakopoulou, V.; Jaw-Tsai, S.S.; Caunt, L.; et al. First-in-human evaluation of CO-1686, an irreversible, highly selective tyrosine kinase inhibitor of mutations of EGFR (activating and T790M). Journal of Clinical Oncology 32, 8010. [Google Scholar] [CrossRef]
  266. Jiang, T.; Zhou, C. Clinical activity of the mutant-selective EGFR inhibitor AZD9291 in patients with EGFR inhibitor-resistant non-small cell lung cancer. Transl Lung Cancer Res 2014, 3, 370–372. [Google Scholar] [CrossRef]
  267. Kim, D.-W.; Lee, D.H.; Kang, J.H.; Park, K.; Han, J.-Y.; Lee, J.-S.; Jang, I.-J.; Kim, H.-Y.; Son, J.; Kim, J.-H. Clinical activity and safety of HM61713, an EGFR-mutant selective inhibitor, in advanced non-small cell lung cancer (NSCLC) patients (pts) with EGFR mutations who had received EGFR tyrosine kinase inhibitors (TKIs). Journal of Clinical Oncology 32, 8011. [Google Scholar] [CrossRef]
  268. Janjigian, Y.Y.; Azzoli, C.G.; Krug, L.M.; Pereira, L.K.; Rizvi, N.A.; Pietanza, M.C.; Kris, M.G.; Ginsberg, M.S.; Pao, W.; Miller, V.A.; et al. Phase I/II trial of cetuximab and erlotinib in patients with lung adenocarcinoma and acquired resistance to erlotinib. Clin Cancer Res 2011, 17, 2521–2527. [Google Scholar] [CrossRef]
  269. Janjigian, Y.Y.; Smit, E.F.; Groen, H.J.; Horn, L.; Gettinger, S.; Camidge, D.R.; Riely, G.J.; Wang, B.; Fu, Y.; Chand, V.K.; et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov 2014, 4, 1036–1045. [Google Scholar] [CrossRef]
  270. Maemondo, M.; Inoue, A.; Kobayashi, K.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010, 362, 2380–2388. [Google Scholar] [CrossRef]
  271. Goldberg, S.B.; Oxnard, G.R.; Digumarthy, S.; Muzikansky, A.; Jackman, D.M.; Lennes, I.T.; Sequist, L.V. Chemotherapy with Erlotinib or chemotherapy alone in advanced non-small cell lung cancer with acquired resistance to EGFR tyrosine kinase inhibitors. Oncologist 2013, 18, 1214–1220. [Google Scholar] [CrossRef]
  272. Kelly, M.P.; Nikolaev, V.O.; Gobejishvili, L.; Lugnier, C.; Hesslinger, C.; Nickolaus, P.; Kass, D.A.; Pereira de Vasconcelos, W.; Fischmeister, R.; Brocke, S.; et al. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025, 77, 100042. [Google Scholar] [CrossRef] [PubMed]
  273. Ciardiello, F.; Tortora, G. A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin Cancer Res 2001, 7, 2958–2970. [Google Scholar] [PubMed]
  274. Pointreau, Y.; Azzopardi, N.; Ternant, D.; Calais, G.; Paintaud, G. Cetuximab Pharmacokinetics Influences Overall Survival in Patients With Head and Neck Cancer. Ther Drug Monit 2016, 38, 567–572. [Google Scholar] [CrossRef] [PubMed]
  275. Douillard, J.Y.; Siena, S.; Cassidy, J.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol 2010, 28, 4697–4705. [Google Scholar] [CrossRef]
  276. Van Cutsem, E.; Köhne, C.H.; Láng, I.; Folprecht, G.; Nowacki, M.P.; Cascinu, S.; Shchepotin, I.; Maurel, J.; Cunningham, D.; Tejpar, S.; et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011, 29, 2011–2019. [Google Scholar] [CrossRef]
  277. Corkery, B.; Crown, J.; Clynes, M.; O'Donovan, N. Epidermal growth factor receptor as a potential therapeutic target in triple-negative breast cancer. Ann Oncol 2009, 20, 862–867. [Google Scholar] [CrossRef]
  278. Carey, L.A.; Rugo, H.S.; Marcom, P.K.; Mayer, E.L.; Esteva, F.J.; Ma, C.X.; Liu, M.C.; Storniolo, A.M.; Rimawi, M.F.; Forero-Torres, A.; et al. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 2012, 30, 2615–2623. [Google Scholar] [CrossRef]
  279. Baselga, J.; Albanell, J.; Ruiz, A.; Lluch, A.; Gascón, P.; Guillém, V.; González, S.; Sauleda, S.; Marimón, I.; Tabernero, J.M.; et al. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol 2005, 23, 5323–5333. [Google Scholar] [CrossRef]
  280. Baselga, J.; Gómez, P.; Greil, R.; Braga, S.; Climent, M.A.; Wardley, A.M.; Kaufman, B.; Stemmer, S.M.; Pêgo, A.; Chan, A.; et al. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol 2013, 31, 2586–2592. [Google Scholar] [CrossRef]
  281. Nabholtz, J.M.; Chalabi, N.; Radosevic-Robin, N.; Dauplat, M.M.; Mouret-Reynier, M.A.; Van Praagh, I.; Servent, V.; Jacquin, J.P.; Benmammar, K.E.; Kullab, S.; et al. Multicentric neoadjuvant pilot Phase II study of cetuximab combined with docetaxel in operable triple negative breast cancer. Int J Cancer 2016, 138, 2274–2280. [Google Scholar] [CrossRef]
  282. Nabholtz, J.M.; Abrial, C.; Mouret-Reynier, M.A.; Dauplat, M.M.; Weber, B.; Gligorov, J.; Forest, A.M.; Tredan, O.; Vanlemmens, L.; Petit, T.; et al. Multicentric neoadjuvant phase II study of panitumumab combined with an anthracycline/taxane-based chemotherapy in operable triple-negative breast cancer: identification of biologically defined signatures predicting treatment impact. Ann Oncol 2014, 25, 1570–1577. [Google Scholar] [CrossRef] [PubMed]
  283. Huang, S.; Armstrong, E.A.; Benavente, S.; Chinnaiyan, P.; Harari, P.M. Dual-agent molecular targeting of the epidermal growth factor receptor (EGFR): combining anti-EGFR antibody with tyrosine kinase inhibitor. Cancer Res 2004, 64, 5355–5362. [Google Scholar] [CrossRef] [PubMed]
  284. Ferraro, D.A.; Gaborit, N.; Maron, R.; Cohen-Dvashi, H.; Porat, Z.; Pareja, F.; Lavi, S.; Lindzen, M.; Ben-Chetrit, N.; Sela, M.; et al. Inhibition of triple-negative breast cancer models by combinations of antibodies to EGFR. Proc Natl Acad Sci U S A 2013, 110, 1815–1820. [Google Scholar] [CrossRef]
  285. Matar, P.; Rojo, F.; Cassia, R.; Moreno-Bueno, G.; Di Cosimo, S.; Tabernero, J.; Guzmán, M.; Rodriguez, S.; Arribas, J.; Palacios, J.; et al. Combined epidermal growth factor receptor targeting with the tyrosine kinase inhibitor gefitinib (ZD1839) and the monoclonal antibody cetuximab (IMC-C225): superiority over single-agent receptor targeting. Clin Cancer Res 2004, 10, 6487–6501. [Google Scholar] [CrossRef] [PubMed]
  286. Garrett, J.T.; Arteaga, C.L. Resistance to HER2-directed antibodies and tyrosine kinase inhibitors: mechanisms and clinical implications. Cancer Biol Ther 2011, 11, 793–800. [Google Scholar] [CrossRef]
  287. Huang, L.; Jiang, S.; Shi, Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). Journal of Hematology & Oncology 2020, 13, 143. [Google Scholar] [CrossRef]
  288. Kumar, R.; Goel, H.; Solanki, R.; Rawat, L.; Tabasum, S.; Tanwar, P.; Pal, S.; Sabarwal, A. Recent developments in receptor tyrosine kinase inhibitors: A promising mainstay in targeted cancer therapy. Medicine in Drug Discovery 2024, 23, 100195. [Google Scholar] [CrossRef]
  289. Sankarapandian, V.; Rajendran, R.L.; Miruka, C.O.; Sivamani, P.; Maran, B.A.V.; Krishnamoorthy, R.; Gangadaran, P.; Ahn, B.-C. A review on tyrosine kinase inhibitors for targeted breast cancer therapy. Pathology - Research and Practice 2024, 263, 155607. [Google Scholar] [CrossRef]
  290. Nagampalli, R.S.; Vadla, G.P.; Nadendla, E.K. Emerging Strategies to Overcome Chemoresistance: Structural Insights and Therapeutic Targeting of Multidrug Resistance-Linked ATP-Binding Cassette Transporters. International Journal of Translational Medicine 2025, 5. [Google Scholar] [CrossRef]
  291. Juliano, R.L.; Ling, V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta 1976, 455, 152–162. [Google Scholar] [CrossRef]
  292. Pamphlett, R.; Bishop, D.P. Elemental biomapping of human tissues suggests toxic metals such as mercury play a role in the pathogenesis of cancer. Front Oncol 2024, 14, 1420451. [Google Scholar] [CrossRef] [PubMed]
  293. Hu, X.; Zhang, Z. Understanding the Genetic Mechanisms of Cancer Drug Resistance Using Genomic Approaches. Trends Genet 2016, 32, 127–137. [Google Scholar] [CrossRef]
  294. Ou, S.H.; Soo, R.A. Dacomitinib in lung cancer: a "lost generation" EGFR tyrosine-kinase inhibitor from a bygone era? Drug Des Devel Ther 2015, 9, 5641–5653. [Google Scholar] [CrossRef]
  295. Lim, S.M.; Syn, N.L.; Cho, B.C.; Soo, R.A. Acquired resistance to EGFR targeted therapy in non-small cell lung cancer: Mechanisms and therapeutic strategies. Cancer Treat Rev 2018, 65, 1–10. [Google Scholar] [CrossRef]
  296. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  297. Soria, J.C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018, 378, 113–125. [Google Scholar] [CrossRef]
  298. Mok, T.S.; Wu, Y.L.; Ahn, M.J.; Garassino, M.C.; Kim, H.R.; Ramalingam, S.S.; Shepherd, F.A.; He, Y.; Akamatsu, H.; Theelen, W.S.; et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med 2017, 376, 629–640. [Google Scholar] [CrossRef] [PubMed]
  299. Arulananda, S.; Do, H.; Musafer, A.; Mitchell, P.; Dobrovic, A.; John, T. Combination Osimertinib and Gefitinib in C797S and T790M EGFR-Mutated Non-Small Cell Lung Cancer. J Thorac Oncol 2017, 12, 1728–1732. [Google Scholar] [CrossRef]
  300. Wang, Z.; Yang, J.J.; Huang, J.; Ye, J.Y.; Zhang, X.C.; Tu, H.Y.; Han-Zhang, H.; Wu, Y.L. Lung Adenocarcinoma Harboring EGFR T790M and In Trans C797S Responds to Combination Therapy of First- and Third-Generation EGFR TKIs and Shifts Allelic Configuration at Resistance. J Thorac Oncol 2017, 12, 1723–1727. [Google Scholar] [CrossRef]
  301. Okura, N.; Nishioka, N.; Yamada, T.; Taniguchi, H.; Tanimura, K.; Katayama, Y.; Yoshimura, A.; Watanabe, S.; Kikuchi, T.; Shiotsu, S.; et al. ONO-7475, a Novel AXL Inhibitor, Suppresses the Adaptive Resistance to Initial EGFR-TKI Treatment in EGFR-Mutated Non-Small Cell Lung Cancer. Clin Cancer Res 2020, 26, 2244–2256. [Google Scholar] [CrossRef] [PubMed]
  302. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  303. Bertoli, E.; De Carlo, E.; Del Conte, A.; Stanzione, B.; Revelant, A.; Fassetta, K.; Spina, M.; Bearz, A. Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It? Int J Mol Sci 2022, 23. [Google Scholar] [CrossRef]
  304. Choudhury, N.J.; Marra, A.; Sui, J.S.Y.; Flynn, J.; Yang, S.R.; Falcon, C.J.; Selenica, P.; Schoenfeld, A.J.; Rekhtman, N.; Gomez, D.; et al. Molecular Biomarkers of Disease Outcomes and Mechanisms of Acquired Resistance to First-Line Osimertinib in Advanced EGFR-Mutant Lung Cancers. J Thorac Oncol 2023, 18, 463–475. [Google Scholar] [CrossRef] [PubMed]
  305. Meng, Y.; Bai, R.; Cui, J. Precision targeted therapy for EGFR mutation-positive NSCLC: Dilemmas and coping strategies. Thorac Cancer 2023, 14, 1121–1134. [Google Scholar] [CrossRef] [PubMed]
  306. Park, K.; Tan, E.H.; O'Byrne, K.; Zhang, L.; Boyer, M.; Mok, T.; Hirsh, V.; Yang, J.C.; Lee, K.H.; Lu, S.; et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 2016, 17, 577–589. [Google Scholar] [CrossRef]
  307. Shi, Y.; Zhang, L.; Liu, X.; Zhou, C.; Zhang, L.; Zhang, S.; Wang, D.; Li, Q.; Qin, S.; Hu, C.; et al. Icotinib versus gefitinib in previously treated advanced non-small-cell lung cancer (ICOGEN): a randomised, double-blind phase 3 non-inferiority trial. Lancet Oncol 2013, 14, 953–961. [Google Scholar] [CrossRef]
  308. Shepherd, F.A.; Rodrigues Pereira, J.; Ciuleanu, T.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 2005, 353, 123–132. [Google Scholar] [CrossRef]
  309. Johnson, J.R.; Cohen, M.; Sridhara, R.; Chen, Y.F.; Williams, G.M.; Duan, J.; Gobburu, J.; Booth, B.; Benson, K.; Leighton, J.; et al. Approval summary for erlotinib for treatment of patients with locally advanced or metastatic non-small cell lung cancer after failure of at least one prior chemotherapy regimen. Clin Cancer Res 2005, 11, 6414–6421. [Google Scholar] [CrossRef]
  310. Solca, F.; Dahl, G.; Zoephel, A.; Bader, G.; Sanderson, M.; Klein, C.; Kraemer, O.; Himmelsbach, F.; Haaksma, E.; Adolf, G.R. Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker. J Pharmacol Exp Ther 2012, 343, 342–350. [Google Scholar] [CrossRef]
  311. Karachaliou, N.; Fernandez-Bruno, M.; Bracht, J.W.P.; Rosell, R. EGFR first- and second-generation TKIs-there is still place for them in EGFR-mutant NSCLC patients. Transl Cancer Res 2019, 8, S23–s47. [Google Scholar] [CrossRef] [PubMed]
  312. Wissner, A.; Mansour, T.S. The development of HKI-272 and related compounds for the treatment of cancer. Arch Pharm (Weinheim) 2008, 341, 465–477. [Google Scholar] [CrossRef] [PubMed]
  313. Xuhong, J.C.; Qi, X.W.; Zhang, Y.; Jiang, J. Mechanism, safety and efficacy of three tyrosine kinase inhibitors lapatinib, neratinib and pyrotinib in HER2-positive breast cancer. Am J Cancer Res 2019, 9, 2103–2119. [Google Scholar] [PubMed]
  314. Ellis, P.M.; Shepherd, F.A.; Millward, M.; Perrone, F.; Seymour, L.; Liu, G.; Sun, S.; Cho, B.C.; Morabito, A.; Leighl, N.B.; et al. Dacomitinib compared with placebo in pretreated patients with advanced or metastatic non-small-cell lung cancer (NCIC CTG BR.26): a double-blind, randomised, phase 3 trial. Lancet Oncol 2014, 15, 1379–1388. [Google Scholar] [CrossRef]
  315. Engelman, J.A.; Zejnullahu, K.; Gale, C.M.; Lifshits, E.; Gonzales, A.J.; Shimamura, T.; Zhao, F.; Vincent, P.W.; Naumov, G.N.; Bradner, J.E.; et al. PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib. Cancer Res 2007, 67, 11924–11932. [Google Scholar] [CrossRef]
  316. Takeda, M.; Nakagawa, K. First- and Second-Generation EGFR-TKIs Are All Replaced to Osimertinib in Chemo-Naive EGFR Mutation-Positive Non-Small Cell Lung Cancer? Int J Mol Sci 2019, 20. [Google Scholar] [CrossRef]
  317. Ma, F.; Ouyang, Q.; Li, W.; Jiang, Z.; Tong, Z.; Liu, Y.; Li, H.; Yu, S.; Feng, J.; Wang, S.; et al. Pyrotinib or Lapatinib Combined With Capecitabine in HER2-Positive Metastatic Breast Cancer With Prior Taxanes, Anthracyclines, and/or Trastuzumab: A Randomized, Phase II Study. J Clin Oncol 2019, 37, 2610–2619. [Google Scholar] [CrossRef]
  318. Lin, Y.; Lin, M.; Zhang, J.; Wang, B.; Tao, Z.; Du, Y.; Zhang, S.; Cao, J.; Wang, L.; Hu, X. Real-World Data of Pyrotinib-Based Therapy in Metastatic HER2-Positive Breast Cancer: Promising Efficacy in Lapatinib-Treated Patients and in Brain Metastasis. Cancer Res Treat 2020, 52, 1059–1066. [Google Scholar] [CrossRef]
  319. Kian, W.; Christopoulos, P.; Remilah, A.A.; Levison, E.; Dudnik, E.; Shalata, W.; Krayim, B.; Marei, R.; Yakobson, A.; Faehling, M.; et al. Real-world efficacy and safety of mobocertinib in EGFR exon 20 insertion-mutated lung cancer. Front Oncol 2022, 12, 1010311. [Google Scholar] [CrossRef]
  320. Vasconcelos, P.; Kobayashi, I.S.; Kobayashi, S.S.; Costa, D.B. Preclinical characterization of mobocertinib highlights the putative therapeutic window of this novel EGFR inhibitor to EGFR exon 20 insertion mutations. JTO Clin Res Rep 2021, 2. [Google Scholar] [CrossRef]
  321. Takeda, M.; Okamoto, I.; Nishimura, Y.; Nakagawa, K. Nimotuzumab, a novel monoclonal antibody to the epidermal growth factor receptor, in the treatment of non-small cell lung cancer. Lung Cancer (Auckl) 2011, 2, 59–67. [Google Scholar] [CrossRef] [PubMed]
  322. Mazorra, Z.; Chao, L.; Lavastida, A.; Sanchez, B.; Ramos, M.; Iznaga, N.; Crombet, T. Nimotuzumab: beyond the EGFR signaling cascade inhibition. Semin Oncol 2018, 45, 18–26. [Google Scholar] [CrossRef]
  323. Garnock-Jones, K.P.; Keating, G.M.; Scott, L.J. Trastuzumab: A review of its use as adjuvant treatment in human epidermal growth factor receptor 2 (HER2)-positive early breast cancer. Drugs 2010, 70, 215–239. [Google Scholar] [CrossRef] [PubMed]
  324. Boekhout, A.H.; Beijnen, J.H.; Schellens, J.H. Trastuzumab. Oncologist 2011, 16, 800–810. [Google Scholar] [CrossRef]
  325. Kim, H.Y.; Choi, J.H.; Haque, M.M.; Park, J.H.; Kim, I.H.; Choi, B.K.; Lee, A.; Park, S. Combined treatment with anti-HER2/neu and anti-4-1BB monoclonal antibodies induces a synergistic antitumor effect but requires dose optimization to maintain immune memory for protection from lethal rechallenge. Cancer Immunol Immunother 2022, 71, 967–978. [Google Scholar] [CrossRef] [PubMed]
  326. Gallois, C.; Bergen, E.S.; Auclin, É.; Pernot, S.; Higué, J.; Trouilloud, I.; Touchefeu, Y.; Turpin, A.; Mazard, T.; Sartore-Bianchi, A.; et al. Efficacy and safety of the combination of encorafenib/cetuximab with or without binimetinib in patients with BRAF V600E-mutated metastatic colorectal cancer: an AGEO real-world multicenter study. ESMO Open 2024, 9, 103696. [Google Scholar] [CrossRef]
  327. Zwart, K.; van Nassau, S.; van der Baan, F.H.; Koopman, M.; Snaebjornsson, P.; van Gestel, A.J.; Vink, G.R.; Roodhart, J.M.L. Efficacy-effectiveness analysis on survival in a population-based real-world study of BRAF-mutated metastatic colorectal cancer patients treated with encorafenib-cetuximab. Br J Cancer 2024, 131, 110–116. [Google Scholar] [CrossRef]
  328. Mendelsohn, J. Jeremiah Metzger Lecture. Targeted cancer therapy. Trans Am Clin Climatol Assoc 2000, 111, 95–110. [Google Scholar]
  329. Park, S.; Jiang, Z.; Mortenson, E.D.; Deng, L.; Radkevich-Brown, O.; Yang, X.; Sattar, H.; Wang, Y.; Brown, N.K.; Greene, M.; et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 2010, 18, 160–170. [Google Scholar] [CrossRef]
  330. Nahta, R.; Yu, D.; Hung, M.C.; Hortobagyi, G.N.; Esteva, F.J. Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer. Nat Clin Pract Oncol 2006, 3, 269–280. [Google Scholar] [CrossRef]
  331. Tseng, P.H.; Wang, Y.C.; Weng, S.C.; Weng, J.R.; Chen, C.S.; Brueggemeier, R.W.; Shapiro, C.L.; Chen, C.Y.; Dunn, S.E.; Pollak, M.; et al. Overcoming trastuzumab resistance in HER2-overexpressing breast cancer cells by using a novel celecoxib-derived phosphoinositide-dependent kinase-1 inhibitor. Mol Pharmacol 2006, 70, 1534–1541. [Google Scholar] [CrossRef] [PubMed]
  332. Molina, M.A.; Codony-Servat, J.; Albanell, J.; Rojo, F.; Arribas, J.; Baselga, J. Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res 2001, 61, 4744–4749. [Google Scholar] [PubMed]
  333. Scaltriti, M.; Rojo, F.; Ocaña, A.; Anido, J.; Guzman, M.; Cortes, J.; Di Cosimo, S.; Matias-Guiu, X.; Ramon y Cajal, S.; Arribas, J.; et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst 2007, 99, 628–638. [Google Scholar] [CrossRef]
  334. Geyer, C.E.; Forster, J.; Lindquist, D.; Chan, S.; Romieu, C.G.; Pienkowski, T.; Jagiello-Gruszfeld, A.; Crown, J.; Chan, A.; Kaufman, B.; et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006, 355, 2733–2743. [Google Scholar] [CrossRef]
  335. Van Cutsem, E.; Köhne, C.H.; Hitre, E.; Zaluski, J.; Chang Chien, C.R.; Makhson, A.; D'Haens, G.; Pintér, T.; Lim, R.; Bodoky, G.; et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 2009, 360, 1408–1417. [Google Scholar] [CrossRef] [PubMed]
  336. Cunningham, D.; Humblet, Y.; Siena, S.; Khayat, D.; Bleiberg, H.; Santoro, A.; Bets, D.; Mueser, M.; Harstrick, A.; Verslype, C.; et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 2004, 351, 337–345. [Google Scholar] [CrossRef]
  337. Van Emburgh, B.O.; Sartore-Bianchi, A.; Di Nicolantonio, F.; Siena, S.; Bardelli, A. Acquired resistance to EGFR-targeted therapies in colorectal cancer. Mol Oncol 2014, 8, 1084–1094. [Google Scholar] [CrossRef]
  338. Karapetis, C.S.; Khambata-Ford, S.; Jonker, D.J.; O'Callaghan, C.J.; Tu, D.; Tebbutt, N.C.; Simes, R.J.; Chalchal, H.; Shapiro, J.D.; Robitaille, S.; et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 2008, 359, 1757–1765. [Google Scholar] [CrossRef]
  339. Benvenuti, S.; Sartore-Bianchi, A.; Di Nicolantonio, F.; Zanon, C.; Moroni, M.; Veronese, S.; Siena, S.; Bardelli, A. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res 2007, 67, 2643–2648. [Google Scholar] [CrossRef]
  340. Amado, R.G.; Wolf, M.; Peeters, M.; Van Cutsem, E.; Siena, S.; Freeman, D.J.; Juan, T.; Sikorski, R.; Suggs, S.; Radinsky, R.; et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 2008, 26, 1626–1634. [Google Scholar] [CrossRef]
  341. De Roock, W.; Claes, B.; Bernasconi, D.; De Schutter, J.; Biesmans, B.; Fountzilas, G.; Kalogeras, K.T.; Kotoula, V.; Papamichael, D.; Laurent-Puig, P.; et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 2010, 11, 753–762. [Google Scholar] [CrossRef] [PubMed]
  342. Sartore-Bianchi, A.; Martini, M.; Molinari, F.; Veronese, S.; Nichelatti, M.; Artale, S.; Di Nicolantonio, F.; Saletti, P.; De Dosso, S.; Mazzucchelli, L.; et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 2009, 69, 1851–1857. [Google Scholar] [CrossRef] [PubMed]
  343. Nahta, R.; Hung, M.C.; Esteva, F.J. The HER-2-targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res 2004, 64, 2343–2346. [Google Scholar] [CrossRef]
  344. Baselga, J.; Cameron, D.; Miles, D.; Verma, S.; Climent, M.; Ross, G.; Gimenez, V.; Gelmon, K. Objective response rate in a phase II multicenter trial of pertuzumab (P), a HER2 dimerization inhibiting monoclonal antibody, in combination with trastuzumab (T) in patients (pts) with HER2-positive metastatic breast cancer (MBC) which has progressed during treatment with T. Journal of Clinical Oncology 25, 1004. [Google Scholar] [CrossRef]
  345. Baselga, J.; Swain, S.M. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 2009, 9, 463–475. [Google Scholar] [CrossRef] [PubMed]
  346. Desmonts, G.; Daffos, F.; Forestier, F.; Capella-Pavlovsky, M.; Thulliez, P.; Chartier, M. Prenatal diagnosis of congenital toxoplasmosis. Lancet 1985, 1, 500–504. [Google Scholar] [CrossRef]
  347. Masuda, H.; Zhang, D.; Bartholomeusz, C.; Doihara, H.; Hortobagyi, G.N.; Ueno, N.T. Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res Treat 2012, 136, 331–345. [Google Scholar] [CrossRef]
  348. Viale, G.; Rotmensz, N.; Maisonneuve, P.; Bottiglieri, L.; Montagna, E.; Luini, A.; Veronesi, P.; Intra, M.; Torrisi, R.; Cardillo, A.; et al. Invasive ductal carcinoma of the breast with the "triple-negative" phenotype: prognostic implications of EGFR immunoreactivity. Breast Cancer Res Treat 2009, 116, 317–328. [Google Scholar] [CrossRef]
  349. Finn, R.S. Targeting Src in breast cancer. Ann Oncol 2008, 19, 1379–1386. [Google Scholar] [CrossRef]
  350. Zheng, R.; Gagan, J.R.; Botten, G.A.; Koduru, P.; Weinberg, O.K.; Chen, M.; Cantu, M.D.; Jaso, J.; Germans, S.; Luu, H.S.; et al. Genomic Landscape of Mixed Phenotype Acute Leukemia Associated With Immunophenotypic Lineage Predominance: Impact on Diagnosis and Treatment. Eur J Haematol 2025. [Google Scholar] [CrossRef]
  351. Irby, R.B.; Yeatman, T.J. Role of Src expression and activation in human cancer. Oncogene 2000, 19, 5636–5642. [Google Scholar] [CrossRef]
  352. Zhang, J.; Kalyankrishna, S.; Wislez, M.; Thilaganathan, N.; Saigal, B.; Wei, W.; Ma, L.; Wistuba, II; Johnson, F. M.; Kurie, J.M. SRC-family kinases are activated in non-small cell lung cancer and promote the survival of epidermal growth factor receptor-dependent cell lines. Am J Pathol 2007, 170, 366–376. [Google Scholar] [CrossRef] [PubMed]
  353. Ishizawar, R.C.; Miyake, T.; Parsons, S.J. c-Src modulates ErbB2 and ErbB3 heterocomplex formation and function. Oncogene 2007, 26, 3503–3510. [Google Scholar] [CrossRef] [PubMed]
  354. Tryfonopoulos, D.; Walsh, S.; Collins, D.M.; Flanagan, L.; Quinn, C.; Corkery, B.; McDermott, E.W.; Evoy, D.; Pierce, A.; O'Donovan, N.; et al. Src: a potential target for the treatment of triple-negative breast cancer. Ann Oncol 2011, 22, 2234–2240. [Google Scholar] [CrossRef] [PubMed]
  355. Finn, R.S.; Dering, J.; Ginther, C.; Wilson, C.A.; Glaspy, P.; Tchekmedyian, N.; Slamon, D.J. Dasatinib, an orally active small molecule inhibitor of both the src and abl kinases, selectively inhibits growth of basal-type/"triple-negative" breast cancer cell lines growing in vitro. Breast Cancer Res Treat 2007, 105, 319–326. [Google Scholar] [CrossRef]
  356. Canonici, A.; Browne, A.L.; Ibrahim, M.F.K.; Fanning, K.P.; Roche, S.; Conlon, N.T.; O'Neill, F.; Meiller, J.; Cremona, M.; Morgan, C.; et al. Combined targeting EGFR and SRC as a potential novel therapeutic approach for the treatment of triple negative breast cancer. Ther Adv Med Oncol 2020, 12, 1758835919897546. [Google Scholar] [CrossRef]
  357. Belli, S.; Esposito, D.; Servetto, A.; Pesapane, A.; Formisano, L.; Bianco, R. c-Src and EGFR Inhibition in Molecular Cancer Therapy: What Else Can We Improve? Cancers (Basel) 2020, 12. [Google Scholar] [CrossRef]
  358. Yoshida, T.; Zhang, G.; Smith, M.A.; Lopez, A.S.; Bai, Y.; Li, J.; Fang, B.; Koomen, J.; Rawal, B.; Fisher, K.J.; et al. Tyrosine phosphoproteomics identifies both codrivers and cotargeting strategies for T790M-related EGFR-TKI resistance in non-small cell lung cancer. Clin Cancer Res 2014, 20, 4059–4074. [Google Scholar] [CrossRef]
  359. Shyam Sunder, S.; Sharma, U.C.; Pokharel, S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther 2023, 8, 262. [Google Scholar] [CrossRef]
  360. Zhou, Y.; Yao, Z.; Lin, Y.; Zhang, H. From Tyrosine Kinases to Tyrosine Phosphatases: New Therapeutic Targets in Cancers and Beyond. Pharmaceutics 2024, 16. [Google Scholar] [CrossRef]
Table 1. RTKs classification is based on the sequence of the kinase domain. Table adapted and modified from[36].
Table 1. RTKs classification is based on the sequence of the kinase domain. Table adapted and modified from[36].
Class Family Receptors
I EGF/ErbB EGFR, ErbB2/HER2, ErbB3/HER3, ErbB4/HER3
II Ins InsR, IGF1R, InsRR
III PDGF PDGFRα, PDGFRβ, CSF1R, KIT, FLT3
IV VEGF VEGFR1/Flt1, VEGFR2/KDR, VEGFR3/Flt4
V FGF FGFR1, FGFR2, FGFR3, FGER4
VI PKT7 PKT7/CCK4
VII TRK TRKA, TRKB, TRKC
VIII ROR ROR1, ROR2
IX MuSK MuSK
X HGF MET, MST1R(RON)
XI TAM AXL, MER, TYRO3
XII TIIE TIE1, TEK(TIE2)
XIII Eph EphA1-8, EphA10, EphB1-4, EphB6
XIV RET RET
XV RYK RYK
XVI DDR DDR1, DDR2
XVII ROS ROS
XVIII LMR LMR1, LMR2, LMR3
XIX ALK LTK, ALK
XX STYK1 STYK1
Table 2. Summary of cancer-associated tyrosine kinases.
Table 2. Summary of cancer-associated tyrosine kinases.
Class of Tyrosine Kinase Cancer type
EGFR (HER1, HER2, HER3, HER4) Epithelial tumors-Lung, Breast and Colon[39,40].
VEGFR-1-3 Regulates angiogenesis and cell migration in tumors[44,45,46]
FGFR-1-4 Tissue cancer [47]
TRKA, TRKB, TRKC (NTRK family) Neuronal cancer[51,52]
RET Implicated in multiple cancers[71,72]
RYK Contributes to tumorigenesis[73,74]
DDR1, DDR2 Regulate adhesion, invasion, survival in collagen-rich tumors[75,76,77]
ROS Present in many cancers types[79]
LMTK/LMR Cancer-linked; influence proliferation, migration [84,85]
ALK, LTK Fusion-driven cancers (e.g., ALK fusions in lymphoma, lung cancer[86,88,89]
STYK Involved in proliferation, survival; emerging cancer target [90]
Ack1 Promotes tumor growth, chemoresistance, gene amplification[94,95]
JAK1, JAK2, JAK3, TYK2 Crucial for immune modulation in cancers [108,109]
Fes, Fer Signal for migration, survival; linked to oncogenesis[122,123,125]
FAK family Adhesion, motility; high expression in aggressive tumors[135,136]
Src family Major signaling mediators; upregulated in various tumors[151]
Abl, Arg Leukemia[169,170,171]
Syk, Zap70 Hematologic cancers[176]
Table 3. TKI inhibitors are used in research studies.
Table 3. TKI inhibitors are used in research studies.
TKIs from clinical studies, research studies result from patients with EGFR-mutated lung cancer drug target TKI Clinical phase #pts (%EGFRm+) RR% Reference
1st/2nd-generation EGFR TKI Neratinib II 91 (100) 3 [258]
XL647 II 33 (53) 3 [259]
Afatinib (A) vs. placebo (P) IIB/III) 585 (16) 7 (A)<1 (P) [260]
Afatinib II 62 (73) 8 [261]
Dacomitinib II 62 (73) 8 [262]
MM-121 + erlotinib II 50 (48) 9 [263]
AP26113 I 32 (35) 3a [264]
Mutant-specific TKI CO-1686 I 40 T790M+ (100) 58 [265]
AZD9291 I 107 T790M+ (100) 64 [266]
HM61713 I 48 T790M+ (100) 29 [267]
EGFR antibodies Cetuximab + erlotinib II 19 (84) 0 [268]
Cetuximab + afatinib IB 126 (98) 29 [269]
Chemotherapy Carboplatin/paclitaxel III 52 (100) 28.8 [270]
Chemo/erlotinib (CE) vs. chemo (C) Retro 78 (100) 41 (CE); 18 (C) [271]
Pemetrexed + gefitinib or erlotinib II 27 (100) 25.9 [272]
RR: Response rate, #pts: Patients.
Table 4. US FDA approved TKI and NTKi inhibitors for use in cancer therapy.
Table 4. US FDA approved TKI and NTKi inhibitors for use in cancer therapy.
TKI Family targeted Inhibitor name Application Adverse effects (Cardio related) Extra-cardio adverse effects
TKI-first generation EGFR/ERBB family Gefitinib[306] NSCLC MI Skin rashes, nausea, diarrhea, anorexia, stomatitis, nausea,
TKI-first generation EGFR/ERBB family Icotinib[307] NSCLC HTN Diarrhea, nausea, skin rashes, loss of appetite
TKI-first generation EGFR/ERBB family Lapatinib[308,309] Breast cancer HF, LVD Skin rashes, diarrhea, nausea
TKI-first generation EGFR/ERBB family Erlotinib[308,309] NSCLC and prostate cancer Edema Skin rashes, diarrhea, nausea, loss of appetite, fatigue, neuropathy, alopecia
TKI-second generation EGFR/ERBB family Afatinib[310,311] NSCLC HTN Severe diarrhea, loss of appetite, paronmychia, dry skin, rashes
TKI-second generation EGFR/ERBB family Neratinib[312,313] Breast cancer Low rates and decline in LVEF and QT prolongation GI related disorders, headache, fatigue, diarrhea
TKI-second generation EGFR/ERBB family Dacomitinib[314,315] EGFR-mutated NSCLC HTN Dry Skin, appetite loss, diarrhea, Weight Loss, Alopecia, Cough, Hemorrhoids, Wound, Back pain, Headache
TKI-third generation EGFR/ERBB family Osimertinib[316] NSCLC MI, pericardial effusion, LVD, HF Diarrhea, nausea, fatigue, stomatitis
TKI-third generation EGFR/ERBB family Pyrotinib[317,318] HER2-postive Diarrhea, Hand-foot syndrome, Leukopenia, Neutropenia, GI disorders, Increased ALT, Anemia, Asthenia
TKI-third generation EGFR/ERBB family mobocertinib[319,320] EGFR-mutation driven NSCLC Dermatitis acneiform, GI Disorders, Rash, Dry skin, Stomatitis, Fatigue, Rash, Paronychia, Anemia
Table 5. List of approved mAbs targeting EGFR.
Table 5. List of approved mAbs targeting EGFR.
mAbs Nature of molecule Binds to Antibody dependent cell mediated cytotoxicity
(ADCC)
Type of
cancer tested
Mechanism Side effects Clinical approved
Nimotuzumab[321] Humanized, mouse mAb Extracellular domain of EGFR, - Squamous cell carcinoma of head and neck (SCCHN), glioma and nasopharyngeal cancer Prevents binding of EGF Reported Yes, III
(approved for treating HNSCC in non-USA countries)
Zalutumumab[322] Humanized IgG1 Extracellular domain of EGFR Squamous cell carcinoma of head and neck (SCCHN) preventing the binding of ligands like EGF and TGF-alpha, thereby inhibiting EGFR signaling YES, III
Trastuzumab[323,324] Humanized IgG1 Juxtamembrane domain IV Yes Several HER2-postive cancers including Breast and gastric cancer Inhibits HER2 homodimers and ligand-independent HER2–HER3 dimers Reported YES
Pertuzumab[325] Humanized IgG1 Heterodimerization domain II Yes HER2-postive cancer such as breast cancer Inhibits ligand-induced HER2-containing heterodimers Reported YES
Cetuximab[326,327] Humanized IgG1 Extracellular domain of EGFR preventing the binding of ligands like EGF and TGF-alpha, thereby inhibiting EGFR signaling Certain advanced colorectal, head and neck cancer EGFR Reported Approved
Panitumumab[328] Humanized IgG1 Extracellular domain of EGFR preventing the binding of ligands like EGF and TGF-alpha, thereby inhibiting EGFR signaling Certain types of metastatic colorectal cancer (mCRC) EGFR Reported Yes, III
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated