Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Biochim Biophys Acta Rev Cancer. 2020 Sep 18;1874(2):188432. doi: 10.1016/j.bbcan.2020.188432

Targeting Protein Tyrosine Kinase 6 in Cancer

Milica B Gilic 1, Angela L Tyner 1,2,*
PMCID: PMC7716696  NIHMSID: NIHMS1630316  PMID: 32956764

Abstract

Protein tyrosine kinase 6 (PTK6) is the most well studied member of the PTK6 family of intracellular tyrosine kinases. While it is expressed at highest levels in differentiated cells in the regenerating epithelial linings of the gastrointestinal tract and skin, induction and activation of PTK6 is detected in several cancers, including breast and prostate cancer where high PTK6 expression correlates with worse outcome. PTK6 expression is regulated by hypoxia and cell stress, and its kinase activity is induced by several growth factor receptors implicated in cancer including members of the ERBB family, IGFR1 and MET. Activation of PTK6 at the plasma membrane has been associated with the epithelial mesenchymal transition and tumor metastasis. Several lines of evidence indicate that PTK6 has context dependent functions that depend on cell type, intracellular localization and kinase activation. Systemic disruption of PTK6 has been shown to reduce tumorigenesis in mouse models of breast and prostate cancer, and more recently small molecule inhibitors of PTK6 have exhibited efficacy in inhibiting tumor growth in animal models. Here we review data that suggest targeting PTK6 may have beneficial therapeutic outcomes in some cancers.

INTRODUCTION

Protein tyrosine kinase 6 (PTK6), also referred to as BRK (Breast tumor kinase) and SIK (Src-Related Intestinal Kinase), is a nonreceptor tyrosine kinase distantly related to the SRC family, and its family members include FRK (Fyn-related kinase) and SRMS (SRC-related kinase lacking C-terminal regulatory tyrosine and N-terminal myristoylation sites). PTK6 was first identified in cultured human melanocytes (PTK6) [1], and subsequently cloned from human breast cancer cells (BRK) [2] and normal mouse intestinal epithelial cells (SIK) [3, 4]. FRK (formerly called RAK, BSK, GTK, and IYK) was also initially cloned from breast cancer cells [5, 6] and the HEP3B hepatoma cell line [7], and SRMS was cloned from embryonic mouse neural precursor cells [8]. Members of the PTK6 family generally exhibit a more restricted pattern of expression than SRC family kinases and are largely expressed in epithelial tissues and a variety of solid tumors. Although cloned more than 25 years ago, functions of the PTK6 family are still being elucidated. Here we focus on summarizing roles of PTK6 and its potential as a therapeutic target in cancers.

The PTK6 Gene

The genes encoding PTK6 and SRC family members share a common ancestral gene [9], but PTK6, FRK, and SRMS genes share the same gene structure that is different than the structure of genes encoding SRC family members, and this differentiates them as a separate family [10]. The PTK6 gene maps to human chromosome 20q13.3 [11] and is very tightly linked to SRMS [10] (Figure 1A). Because of this tight linkage, any copy number alterations would be expected to impact both PTK6 and SRMS. Both PTK6 and SRMS are deleted in 20q13.33 deletion syndrome, associated with a rare developmental psychiatric disorder [12]. Early studies using comparative genomic hybridization analysis revealed amplification of chromosome 20q13 in breast cancers [13] and increased expression of PTK6 [14]. PTK6 has been reported to be amplified in ovarian tumors [15] and ERBB2 positive breast cancers [16]. Copy number gains of PTK6 were recently reported in breast cancer metastases compared with primary tumors and correlated with significantly shorter disease specific survival [17].

Figure 1. The PTK6 gene and PTK6 proteins.

Figure 1.

A) Organization of PTK6 and SRMS genes on human chromosome 20q13.33. Exons are represented by rectangles, with protein coding in blue, while introns are represented by a line. The distance between PTK6 and SRMS genes is 3440 bp. Information on genes was retrieved from Ensembl [166]. B) Structure and key amino acid residues in PTK6. Full length PTK6 contains conserved phosphorylation sites that correspond with its activation (Y342) and inhibition (Y447). In addition, sequences have been identified in the SH2 domain that mediate phospho-tyrosine (R85, H126) and lipid binding (R131, R136) [28]. Mutation of K219 in the ATP binding site abolishes kinase activity [32]. The schematic protein diagram was drawn using DOG 2.0 software [167]. C and D) Sequences surrounding the activating (Y342) (C) and inhibitory (Y447) (D) regulatory phosphorylation sites in human PTK6 are not well conserved, allowing for the production and use of phospho-specific antibodies with specificity for PTK6 PY342 or PTK6 PY447 (see Figure 2). However, specificity of commercially available anti-PY342 and PY447 PTK6 polyclonal antibodies may vary from lot to lot and each lot should be validated. Primary sequences were aligned using Clustal Omega [168] and visualized in Jalview [169].

PTK6 encodes a 451 amino acid protein that consists of SRC homology 3 (SH3), SRC homology 2 (SH2) and tyrosine kinase catalytic domains [2], as well as an alternatively spliced transcript that encodes a 15 kDa protein termed ALT-PTK6 that lacks kinase activity, and consists of a SH3 domain and unique proline rich C-terminus[18](Figure 1B). ALT-PTK6 was originally detected in the T47D human breast cell line and named λm5 [19], but later found in a variety of human cell lines [18]. ALT-PTK6 expression was detected in normal prostate and prostate tumors, with the PTK6/ALT-PTK6 ratio being higher in the cancer tissues [18]. Normal physiological functions for ALT-PTK6 have not been well defined. However, ALT-PTK6 can compete with full length PTK6 for substrate binding, through their shared SH3 domain sequence, and it can act as a PTK6 inhibitor [18, 20].

PTK6 Protein

PTK6 shares structural features with the SRC family kinases and has a short unique amino terminus followed by a SH3, SH2, and kinase domain. While SRC family members share from 54 – 75% amino acid sequence identity, the three PTK6 family members share only 44 – 45% identity with one another and with members of the SRC family. None of the studied PTK6 family members contains a signal for lipid modification such as myristoylation or palmitoylation, found in SRC family members, and all of them have a conserved tyrosine in the kinase catalytic domain that corresponds to tyrosine residue 419 in SRC that is involved in autophosphorylation and activation (Figure 1C). Both PTK6 and FRK have a conserved carboxy-terminal tyrosine that can be phosphorylated to promote intramolecular interactions that negatively regulate kinase activity. SRMS lacks a C-terminal regulatory tyrosine, as does C-terminal SRC kinase CSK [8] (Figure 1D) and it has been suggested that SRMS may be a CSK-like kinase ([21],reviewed in [22]).

No regulatory role for the unique amino terminus of PTK6 that consists of the first 7 amino acids has been demonstrated [23]. The SH3 domain of PTK6 is important for substrate recognition but is also involved in inhibition of its activity through intramolecular interactions between a hydrophobic patch of the SH3 domain and the linker region located between SH2 and catalytic domains [2325]. Unique features of the PTK6 SH2 domain are present in the loop regions; the CD loop of PTK6 is shorter while AB loop is longer than those for canonical SH2 domains, which could influence the rearrangement of the central β-barrel and influence ligand specificity [26, 27]. Mutation of R85 and H126, which are conserved in other kinases, is sufficient to disrupt interactions between the SH2 domain and phosphotyrosine containing peptides, thereby mediating SH2 domain-phosphotyrosine binding [27] (Figure 1B).

The PTK6 SH2 domain can bind phospholipids via a distinct lipid binding groove exposed on the surface of the protein that interacts with phosphoinositols with low specificity for different headgroups. Arginine residues 131 and 136 (Figure 1B) are critical for interaction with lipids and mutating them leads to interruption of protein-lipid association [28]. This interaction may be a regulatory step for PTK6 signaling, since it could promote recruitment of PTK6 to the plasma membrane and prime the protein for interaction with binding partners that contain phosphotyrosine binding sites. It could also lead to recruitment of PTK6 to specific membrane locations and compartmentalize the protein. Lipids could also behave like allosteric regulators of the protein [28]. Protein-protein and protein-lipid interactions may cooperate to regulate PTK6 relocalization in prostate cancer.

The structure of the PTK6 kinase domain resembles other tyrosine kinases. It consists of two lobes, a β-strand rich N-lobe and an α-helix rich C-lobe. The two lobes are connected by a 9 amino acid long hinge region that binds adenine nucleotide with T264 as the gatekeeper [29]. The gatekeeper threonine residue controls the access of small molecules to the hydrophobic pocket deep in the active site and its mutation can stabilize the active kinase conformation [30]. All other kinase structure motifs are present in the PTK6 kinase domain, including the Pi-loop, αC-helix, hinge, HRD-loop and DFG-loop. The catalytic triad consists of K219, D330 and E235 [29]. Solved crystal structures are in DFG-in and αC-helix-out conformation without activating phosphorylation [29, 31]. Mutation of lysine residue 219 that is involved in ATP binding renders the kinase inactive [32].

PTK6 undergoes activating autophosphorylation at tyrosine residues 342 and 351; tyrosine residue 342 corresponds to the activating phosphorylation site conserved in SRC kinases [33] (Figure 1B). Autophosphorylation was also detected at tyrosine residues 13, 61, 66, and 114, although the MS peaks for peptides around these residues were smaller than those around tyrosine residue 342, which is a major autophosphorylation site. A later study indicated that a peptide corresponding to the carboxy terminus of PTK6 containing the terminal regulatory tyrosine residue at position 447 was also a PTK6 substrate [34]. Using phospho-specific antibodies, we found that expression of full length PTK6 in bacteria results in autophosphorylation at tyrosine residues 342 and 447, indicating that PTK6 may undergo both autoactivation and autoinhibition (Figure 2A). Transient transfection of Myc-tagged wild type PTK6 into LNCaP prostate cancer cell line results in PTK6 phosphorylation at both Y342 and Y447, with peak activating phosphorylation at the earlier 24 hour time point (Figure 2B).

Figure 2. Autophosphorylation of PTK6 at the regulatory tyrosine residues Y342 and Y447.

Figure 2.

A) The cDNAs encoding wild type, kinase dead (KM) and active (YF) PTK6 were cloned into the pGEX-KG vector and expressed as GST fusion proteins in the Novagen Rosetta(DE3)pLysS bacterial strain (Millipore Sigma, Burlington, MA) at 16°C in the presence of 1 mM IPTG for 16 hours. Proteins were purified using glutathione sepharose beads and subjected to immunoblotting as previously described [69]. Anti-GST antibodies (#2622) were purchased from Cell Signaling Technology (Danvers, MA). Mouse monoclonal anti-human PTK6 (G6, sc-166171) was purchased from Santa Cruz Biotechnology (Dallas, TX). Rabbit polyclonal antibodies against the PTK6 activating phosphorylation site PY342 (09–144) were purchased from Millipore Sigma. Rabbit polyclonal antibodies against PTK6 phospho-Y447 (ab138368) were purchased from abcam (Cambridge, MA). Expression of wild type PTK6 (WT) in bacteria leads to autophosphorylation of PTK6 at residues Y342 and Y447. The constitutively active PTK6 mutant PTK6 YF contains a mutation of the negative regulatory tyrosine at amino acid position 447 to phenylalanine and we detect phosphorylation only at Y342. Catalytically dead PTK6 KM lacks kinase activity and no tyrosine phosphorylation can be detected, underscoring the requirement for kinase active PTK6 for PTK6 Y342 and Y447 phosphorylation. B) Transfection of Myc-tagged wild type PTK6 into the LNCaP prostate cancer cell line (ATCC CRL-1740) leads to peak activation (PY342) of ectopic PTK6 at 24 hours post transfection. While expression of Myc-tagged wild type PTK6 increases at 48 hours post transfection, activating phosphorylation (PY342) decreases and inhibitory phosphorylation (PY447) increases or is maintained. Cells were harvested 24 and 48 hours post transfection, and cells were stimulated with fresh media 10 minutes prior to lysis. GAPDH (14C10, #2118), and Myc-tag (9B11, #2276) antibodies were purchased from Cell Signaling Technology.

Intramolecular SH2 domain binding with phosphorylated Y447 (PY447) in the C-terminal tail leads to inactivation of PTK6 [27, 33]. The SH3 domain also controls the activity of PTK6 through intramolecular interaction with proline rich linker region. It was demonstrated that amino acids R22, W44, N65, and Y66 bind the linker region that forms a compact hairpin structure. Mutation of any of these residues results in increase kinase activity when compared to wild type protein [24]. The long noncoding RNA (lncRNA) LINK-A (long intergenic noncoding RNA for kinase activation) binds the PTK6 SH3 and kinase domains and may cause a conformational change that promotes its recruitment to the membrane [35] (Figure 3). The PTK6 SH2 domain binds the consensus sequence phospho-Y-(D/E)-(D/E)-Y [25]. The consensus substrate sequence phosphorylated by PTK6 X-(E/I/L/N)-Y-(D/E)-(D/E) is also rich in acidic amino acid residues [36, 37].

Figure 3. PTK6 contributes to several kinase pathways that can promote cancer.

Figure 3.

PTK6 activation occurs in response to activation of different members of the ERBB family including EGFR and ERBB2. Feedback between PTK6 and EGFR also exists, as PTK6 has been reported to phosphorylate EGFR at Y845, promoting its activation. PTK6 is also downstream of the IGF1R and MET receptors, targeting PAK4 led to increased PTK6 expression. While integrins may activate both PTK6 and FAK, FAK is a direct substrate of PTK6. PTK6 has also been shown to regulate activation of RHO, RAC, ERK5, AKT, p38 MAPK, and CDK/CYCLIN complexes (see text for references). In response to HB-EGF, PTK6 is recruited to EGFR and GPNMB by the lncRNA LINK-A leading to PTK6 activation and its phosphorylation of HIF-1α resulting in the normoxic stabilization of HIF-1α [35]. Targeting PTK6 in addition to other kinases in combination therapies may have enhanced benefits in cancers with high PTK6 activation and expression.

Cancer-Associated Mutations in PTK6

Several cancer-associated somatic mutations that may impact PTK6 activity have been identified, and Tsui et al examined effects of some of these on PTK6 activation and downstream substrate phosphorylation [38]. They found mutations that increase PTK6 activity, include L16F found in clear cell renal cell carcinoma, R131L found in gastric cancer, L343F in cutaneous squamous cell carcinoma, and P450L detected in pancreatic cancer. PTK6 mutations V253M found in head and neck squamous carcinoma and N317S in ovarian carcinoma lead to decreased PTK6 activation [38]. The cBioportal TCGA PanCancer Atlas includes 32 studies with 10,967 samples and PTK6 is mutated in 37 patients [39, 40], while the COSMIC database that includes 38256 unique samples lists 246 unique samples with PTK6 mutations [41].

Regulation of PTK6 Expression

The PTK6 promoter region overlaps with the tightly linked SRMS gene and factors contributing to its epithelial specific expression have not been identified. Because of their tight linkage, we examined possible coregulation of Ptk6 and Srms in the mouse gastrointestinal tract and skin, and found that Srms is also expressed in skin, but not in the gastrointestinal tract, and Srms expression was not induced in intestines of Ptk6−/− mice (W. Bie and A. L. Tyner, unpublished data). Early studies discovered binding sites for NFkB and SP1 in the promoter sequence [42]. The ribosomal S6 protein kinase RSK2 has also been shown to upregulate PTK6 mRNA expression through activation of the CREB transcription factor [43]. The bioactive lipid sphingosine 1-phosphate induced PTK6 mRNA expression in thyroid cancer cells [44].

In triple negative breast cancers, PTK6 transcription is induced by the hypoxia inducible factors HIF-1α and HIF-2α [45]. A glucocorticoid response element was identified near a hypoxia response element and glucocorticoid receptor (GR) binding was demonstrated by chromatin immunoprecipitation. While HIFs can regulate PTK6 independently of GR, GR requires cooperation with HIFs to upregulate PTK6. [46].PTK6 may be regulated by estrogen receptor-α (ERα) through estradiol signaling [47], and ERα positive cell lines and tissues express higher protein levels of PTK6 [47, 48].

PTK6 expression is induced by a variety of cellular stresses including DNA damage [49, 50], hypoxia, and reactive oxygen species [45]. PTK6 expression is induced in the proliferating progenitor cell compartments of the intestine and skin after DNA-damage generated by different mechanisms, including gamma [49, 50] and UV [51] irradiation and DNA-damaging drugs [50, 52]. In cancer cells, PTK6 expression increased following treatment with radiation and chemotherapeutic agents, including doxorubicin [50], Taxol, 5’ fluorouracil [46], and gemcitabine [53]. Treatment of bladder cancer cells with a PAK4 inhibitor also led to induction of PTK6 expression [54]. Induction of PTK6 in colon cancer cells following irradiation or treatment with doxorubicin was at least partially dependent upon p53 [50].

In breast cancer cells, ERBB receptor ligand mediated activation of PTK6 leads to ERK5 and p38 MAPK activation [55]. Regan Anderson et al. showed that PTK6 activates p38 when cellular stress is present, leading to a positive feedback loop where GR is phosphorylated by p38 and induces PTK6 expression, resulting in even stronger activation of p38 [46].

Expression of PTK6 is modulated by several microRNAs, including miR-214 [56], miR-93 [57], miR-187 [58], and miR-17 [44]. MicroRNA-214 is located in the intron of Dynamin-3 gene on chromosome 1q24.3 and is involved in regulation of signaling networks in normal and cancer tissues in context dependent manner [59]. miR-214 is deregulated in osteosarcoma, melanoma, breast, ovarian, cervical, prostate, gastric, and hepatocellular cancers [56, 59, 60]. It was shown that miR-214 directly targets the PTK6-3’UTR by binding to potential binding sites located at positions 28–35 and 201–207 downstream of the stop codon, and this leads to decreased PTK6 expression in several prostate cancer cell lines [56].

miR-93 targets PTK6 directly and was able to improve barrier disfunction that was caused by the proinflammatory cytokines TNFα/INFγ in mouse colonic epithelial cells [57]. The miR-133a cluster is shown to be downregulated in colorectal cancer [61], and one member of the cluster, miR-187, may target PTK6 directly. Introduction of miR-187 into colon cancer cells led to decreased PTK6 protein levels [58]. miR-17 binds the PTK6-3’UTR and reduces PTK6 levels and inhibits the migration of thyroid follicular carcinoma cells. The expression of miR-17 can be regulated by sphingosine-1-phosphate levels [44].

PTK6 expression is also regulated at the level of protein stability. PTK6 binds the molecular chaperone HSP90 in a kinase-independent manner. Treatment of breast cancer cells with the HSP90 inhibitor geldanamycin led to proteosomal degradation of PTK6 but not the related kinase SRC [62]. PTK6 was shown to be ubiquitylated in an oxygen-dependent manner in breast cancer cells, and hypoxia promoted rapid PTK6 protein stabilization prior to induction of PTK6 mRNA expression by HIF-1α [63]. PTK6 can also be targeted for degradation by calpain-1, a calcium regulated cysteine protease that is negatively regulated by ERBB2 [64].

Regulation of PTK6 Activity

PTK6 activity is regulated by phosphorylation on tyrosine residues, and the protein tyrosine phosphatase 1B (PTP1B) negatively regulates PTK6 activity in ovarian cancer cells [65]. PTP1B directly dephosphorylates tyrosine residue 342 in PTK6 inhibiting its kinase activity, while MSI-1436, a small molecule inhibitor of PTP1B promoted PTK6 activation [21]. The tumor suppressor protein PTEN, which has both lipid and protein phosphatase activities, also directly dephosphorylates PTK6 at tyrosine residue 342 to inhibit its oncogenic activities [66]. Loss of PTEN in the mouse prostate leads to activation of PTK6 at the plasma membrane which promotes the epithelial mesenchymal transition (EMT) [67] and prostate tumorigenesis in vivo [66].

CSK phosphorylates the carboxy terminal tyrosine in SRC family kinases to induce an inactive conformation, but it does not target PTK6 [33]. The PTK6 family member SRMS shares structural similarity to CSK and different substrate specificity than PTK6 and has been proposed to be the carboxy-terminal PTK6 kinase [21]. However, no in vivo studies examining the impact of endogenous SRMS on PTK6 Y447 phosphorylation and kinase activity have been performed, and most SRMS functional studies have relied on tagged overexpressed SRMS protein. PTK6 may also autoregulate itself, as PY447 was detected in bacterially expressed PTK6, and PY447 levels increase with time in mammalian cells ectopically expressing PTK6 (Figure 2).

Ligands for the ERBB family of growth factor receptors activate PTK6. EGF promoted activation of ectopically expressed PTK6 in cell lines, leading to paxillin phosphorylation and Rac1 activation, thereby promoting cell migration and invasion [68]. The ERBB ligand heregulin induced PTK6 activation to positively regulate Rac1, p38 MAPK and ERK5 and cell migration in breast cancer cells [55]. PTK6 promoted activation of AKT induced by physiological levels of EGF in mouse embryonic fibroblasts lacking the SRC-family kinases SRC, YES and FYN [69]. PTK6 kinase activation was promoted by calcium in a keratinocyte differentiation model [70], and osteopontin in breast cancer cells [71].

Intracellular localization plays an important role in regulating PTK6 activities, but mechanisms regulating its intracellular localization are not fully understood. While PTK6 can be found in all cellular compartments, it lacks both membrane and nuclear targeting signals. Targeting active PTK6 YF to either the membrane or nucleus had opposite effects in prostate [67, 72, 73] and colon cancer [74] cells, with membrane-targeted PTK6 being oncogenic and nuclear PTK6 growth suppressive. The SH2 domain of PTK6 may bind to phosphorylated tyrosine residues in membrane associated proteins such as activated growth factor receptors, as well as to membrane lipids, to facilitate its membrane association [28]. In triple negative breast cancer cells, the lncRNA LINK-A was found to mediate recruitment of PTK6 to EGFR and transmembrane glycoprotein NMB (GPNMB) in response to HB-EGF stimulation [35].

Nuclear localization of PTK6 is lost in prostate cancer [75], and targeting PTK6 to the nucleus by addition of a nuclear localization signal is growth inhibitory [72, 76]. Inhibition of Crm1/exportin-1 mediated nuclear export did not restore PTK6 nuclear localization [72]. In addition, overexpression of the nuclear RNA-binding proteins SAM68, which interacts with the SH3 and SH2 domains of PTK6 [25], was not sufficient to promote PTK6 nuclear localization [72]. Recently, the nuclear paraspeckle protein PSPC1 was reported to be able sequester PTK6 in the nucleus of liver cancer cells, to inhibit its cancer promoting activities [77, 78].

PTK6 Substrates and Interacting Proteins

At the plasma membrane, PTK6 acts downstream of several growth factor receptors including members of the ERBB family [16, 32, 7981], IGFR1 [82], and MET [83, 84] (Figure 3). PTK6 enhanced ERBB3 tyrosine phosphorylation and activation of PI3K and AKT in the presence of EGF [80]. PTK6 can promote epidermal growth factor receptor (EGFR) activation by directly phosphorylating it and inhibiting its downregulation upon activation [81]. Membrane targeted PTK6 also phosphorylates the EGFR pathway substrate Eps8, contributing to oncogenic signaling in breast cancer cells [85].

PTK6 binds and phosphorylates RNA binding proteins SAM68 [25, 79, 86], SLM1 and SLM2 [87]. PTK6-mediated phosphorylation of these proteins inhibits RNA binding, which can affect stability, translation and transport of RNAs [25, 87, 88]. The RNA splicing factor PSF is a substrate of PTK6, and its phosphorylation leads to its relocalization to the cytoplasm and cell cycle arrest [89]. PTK6 also disrupts nuclear localization of kinesin associated protein 3A (KAP3A) through its phosphorylation of tyrosine residues in the KAP3A carboxy terminus [90].

PTK6 phosphorylates several transcription factors including STAT3 [91], STAT5b [92], β-catenin [74], HIF-1α [35] and parafibromin [93]. PTK6 phosphorylates STAT3 and STAT5b to promote their activation and proliferation of breast cancer cells [91, 92]. In Ptk6 knockout mice, STAT3 activation is impaired in colon [52] and breast cancer [94] models. PTK6 phosphorylates β-catenin at several different tyrosine residues, but the effect it has is dependent on PTK6 intracellular localization [74]. Nuclear targeted PTK6 inhibits β-catenin/TCF transcriptional activity, while PTK6 phosphorylation of β-catenin at the plasma membrane enhances Wnt signaling [74]. HB-EGF promotes PTK6 activation and phosphorylation of HIF-1α on conserved tyrosine residue 565, which inhibits proline hydroxylation and stabilizes HIF-1α under normoxia [35]. Parafibromin, encoded by the HRPT2 gene and part of the RNA-polymerase-associated factor complex, is phosphorylated by PTK6, which blocks its scaffolding functions and interactions with β-catenin and Gli-1 [95]. PTK6-mediated phosphorylation of parafibromin leads to selective activation of YAP and transcription of TEAD target genes [93].

PTK6 binds and phosphorylates several adaptor proteins including STAP2 [96, 97], paxillin [68], BCAR1 [98], and Dok1 [34, 99]. STAP-2 interacts with STAT3 and STAT5b and modulates their activity through its PH domain. Knockdown of STAP-2 disrupts PTK6 activation of STAT3 and STAT5 [97, 100, 101]. Ectopically expressed PTK6 promoted phosphorylation of paxillin, a molecular scaffold protein localized at the plasma membrane leading to activation of Rac1 to promote cell migration and activation [68].

The Cdk-Cyclin assembly factor/inhibitor p27Kip1 is a protein substrate of PTK6. The SH3 domain of PTK6 binds to p27 and PTK6 phosphorylates p27 at tyrosine residue 88, thereby promoting activation of the CDK4-Cyclin complex [20]. PTK6-mediated phosphorylation of p27 can be inhibited by ectopic expression of the alternative PTK6 splice product ALT-PTK6, which competes for p27 binding with the full-length PTK6 [18, 20]. It has been suggested that inhibition of PTK6-mediated p27 tyrosine phosphorylation may have advantages in breast cancer patients sensitive to CDK4/6 inhibition [102].

Membrane targeted active PTK6 promotes ERK1/2 activation [103]. This could occur through growth factor receptor activation (i.e. EGFR) [81]. Alternatively, PTK6 binds and phosphorylates p190RhoGAP to promote inactivation of RHO and activation of RAS (Figure 3) contributing to proliferation and cell migration [104]. Cisplatin treatment of testicular cancer was reported to epigenetically impact the “PTK6 -> RHO GTPase ->RAS GTPase -> MAPK” reactome [105].

In several instances, activities of PTK6 are kinase-independent and may be attributed to adaptor/scaffolding functions mediated by its SH3 and SH2 domains. Association with certain substrates such as SAM68 and ERBB3 does not require kinase activity [25, 80, 106]. While the PTK6 SH2 and SH3 domains regulate its association with IRS-4, a member of the insulin receptor substrate family, IRS-4 did not appear to be a PTK6 substrate [107]. Kinase-dead PTK6 was shown to promote proliferation of the T47D breast cancer cell line [108]. PTK6 kinase activity was not required for PTK6-dependent HGF induced cell migration or for PTK6-ERK5 interaction and ERK5 activation [83]. PTK6 was able to promote epithelial characteristics in colon cancer cell lines independent of kinase activity [109]. These data suggest that PTK6 has both kinase-dependent and -independent functions in cancer.

PTK6 in Normal Tissues

PTK6 is expressed in differentiated epithelial cells of the gastrointestinal tract [3, 4, 110112], skin [51, 70, 113], and prostate [66, 75, 114]. PTK6 was detected in nuclei of normal prostate epithelial cells [75], and in T cells upon activation [115]. PTK6 expression was not detected in the normal mouse mammary gland [110, 116], but was detected in noncancerous human breast biopsy tissues [117]. Expression of an inducible PTK6 transgene in mammary glands of mice forced to wean litters led to delayed involution and activation of p38 MAPK signaling [116].

Expression of Ptk6 is developmentally regulated and was first detected late in gestation at mouse embryonic day 15.5 in the differentiating suprabasal layers of the skin, and later in the small intestine as the crypt-villus axis develops near the end of gestation [4]. PTK6 was activated in a calcium induced mouse embryonic keratinocyte differentiation model, and its expression promoted keratinocyte differentiation marker expression [70].

PTK6 promotes epithelial cell differentiation during normal turnover of the intestinal epithelium. Systemic disruption of the Ptk6 gene led to increased epithelial proliferation and impaired differentiation in the mouse small intestine [112]. This was correlated with increased β-catenin activation and MYC expression in intestinal crypts. Later studies demonstrated that disruption of Ptk6 led to increased expression of a β-catenin regulated LacZ reporter transgene in the mouse intestine [74], suggesting that PTK6 inhibits β-catenin transcription in the intestine during normal homeostasis. In addition, studies with cultured Ptk6 null young adult mouse colon cells suggested that PTK6 may play roles in regulating epithelial barrier function [57, 118].

Although primarily studied in epithelial cells, PTK6 expression has been reported in normal activated T cells [115] and endothelial cells [119]. PTK6 expression was not detected in normal resting peripheral blood mononuclear cells (PBMCs). Stimulation of PBMCs with the mitogen phytohemagglutinin or CD3/CD28, which promote T-cell activation, induced expression and activation of PTK6 [115]. PTK6 was detected in normal primary mouse microvascular endothelial cell cultures, where it was shown to contribute to TNFα induced barrier dysfunction [119].

PTK6 sensitized nontransformed Rat1a fibroblasts to inducers of apoptosis such as starvation and UV irradiation [120]. In the mouse, DNA-damage induced by total body irradiation led to induction of PTK6 expression in proliferating progenitor cells of intestinal crypts. Systemic disruption of Ptk6 impaired DNA-damage induced apoptosis in the small intestine [49]. PTK6 expression was also induced in the normal colon by the carcinogen azoxymethane [52].

PTK6 in Cancer

Breast Cancer:

PTK6 has been most well studied in breast cancer and increased expression has been identified in the majority of breast cancers including ER positive, ERBB2 positive and triple negative breast cancers [16, 48, 55, 82, 121123] reviewed in [124, 125]. PTK6 expression has been shown to contribute to breast cancer cell migration, invasion and metastases [45, 63, 83, 126]. Several studies indicate PTK6 promotes breast cancer cell survival. PTK6 regulates IGF-1 mediated anoikis resistance in breast and ovarian cancer cell lines [82]. Knockdown of PTK6 promoted apoptosis of Lapatinib-resistant ERBB2 positive breast cancer cells [127]. PTK6 may also protect breast cancer cells from autophagic cell death [128].

While PTK6 can be detected at low levels in normal human breast tissue, no signal for active PTK6 phosphorylated at tyrosine residue 342 (PY342) could be detected in the normal mammary gland [117]. The pool of active PTK6 is at the plasma membrane in breast cancer cells, but a small percentage of the active protein is also found in the cytoplasm [117]. PTK6 expression was induced in all mammary gland tumors originating in mice, including spontaneous tumors and those promoted by activated ERBB2 and H-Ras [129].

Mammary gland specific transgenic expression of PTK6 promoted tumorigenesis in transgenic mice [116, 129]. PTK6 expression also enhanced MET driven mammary gland tumorigenesis in transgenic mice [45]. Systemic disruption of Ptk6 in mice expressing activated ERBB2 in the mammary gland prolonged survival and delayed tumor formation and reduced lung metastases [94]. PTK6 can promote mammary gland tumorigenesis through activation of STAT3 [91] and STAT5b [92]. Reduced STAT3 activation was detected in Ptk6−/− ERBB2 positive mouse mammary glands [94].

Prostate Cancer:

High PTK6 mRNA levels correlate with poor prognosis in prostate cancer, and recurrent and androgen independent prostate cancers have higher levels of PTK6 mRNA when compared to normal or adjacent tissue [67, 130]. PTK6 is expressed in normal prostate tissue and its localization is generally nuclear [75]. In prostate cancers, PTK6 is localized in the cytoplasm [75], and active PTK6 is found at the membrane [66, 67, 75]. Targeted expression of PTK6 to the nucleus inhibited cell proliferation [72]. Translocation of PTK6 from the nuclear to cytoplasmic compartment allows it to phosphorylate substrates involved in promoting cell proliferation, survival and migration [37]. Membrane localized active PTK6 promoted the EMT and prostate tumor cell metastasis in xenograft models [67].

Systemic disruption of Ptk6 in mice with conditional disruption of Pten in the prostate led to decreased development of invasive prostate adenocarcinomas and urethral carcinomas [66]. An inverse correlation between PTEN expression and PTK6 activation and expression was demonstrated in patient samples in a prostate tumor tissue microarray. In addition, higher PTK6expression coincided with decreased PTEN expression in different prostate cancer datasets including TCGA.

Loss of the tumor suppressor PTEN in prostate cancers leads to PTK6 activation, since PTEN negatively regulates PTK6 activity by dephosphorylating it at Y342 [66]. PTK6 can then directly phosphorylate BCAR1 (p130CAS) [98] as well as FAK [73] and AKT [69], thereby promoting their activation in prostate cancer cells [73]. Membrane associated PTK6 promoted resistance to anoikis and cell survival through enhanced activation of AKT in prostate cancer cells [73].

Colon Cancer:

PTK6 is expressed throughout the normal human gastrointestinal tract. Early work showed that PTK6 RNA expression was slightly increased in a small sample of colon tumors compared with adjacent normal tissue when normalized to keratin expression [110]. However, later studies which included tumor tissue microarray staining, showed that PTK6 expression is highest in normal differentiated colon epithelial cells and decreases during colon tumor progression [109]. In addition, analysis of several datasets including TCGA Colorectal Cancer do not reveal increased PTK6 expression in colorectal cancers [109].

Active PTK6 PY342 is not readily detected in human colon cancer cell lines, where PTK6 appears to function in maintaining the epithelial phenotype independent of PTK6 kinase activity, similar to its role the normal intestinal tract. PTK6 was shown to suppress tumorigenesis by opposing the EMT in colon cancer cell line models [109]. While PTK6 is most often reported as being oncogenic, it has been shown to have tumor suppressive roles in a few cancers including esophageal cancer [131, 132] and laryngeal squamous cell carcinomas [133, 134].

Systemic disruption of the Ptk6 gene did not lead to spontaneous tumor development in the gastrointestinal tract [112]. However, impaired tumorigenesis was detected in Ptk6−/− mice in an azoxymethane/dextran sodium sulfate mouse model of colon cancer. This was initially surprising since PTK6 has a growth inhibitory role during normal homeostasis in the normal intestine, and we had predicted that disruption of Ptk6 might lead to increased tumor formation [112]. However, in the azoxymethane model, carcinogen driven induction of PTK6 promoted STAT3 activation in the crypts to promote survival and proliferation of damaged cells and colon tumorigenesis [52]. PTK6 promoted resistance to DNA-damage induced apoptosis through activating STAT3 in mice and in human colon cancer cell lines [50, 52].

Other Cancers:

PTK6 is not expressed in normal ovarian tissue but is detected in ovarian cancer tissues and cell lines [15, 65]. mRNA and protein levels of PTK6 are higher in nonsmall cell lung cancers (NSCLC) than in normal tissues [135, 136], and increased PTK6 mRNA levels are associated with decreased survival [136]. PTK6 is expressed in a number of other tumor types including skin [51], head and neck [111, 137], thyroid [44, 138], pancreatic [139], hepatocellular [77, 140], cervical [141] and bladder [142] [54] cancers. Systemic disruption of Ptk6 reduced skin tumor formation caused by UV-radiation in mice [51]. One study reported PTK6 expression in cutaneous T-cell lymphomas and transformed B and T cells, and knockdown of PTK6 led to increased apoptosis and reduced proliferation in the murine BaF3 lymphocyte cell line [115]. Ectopic expression of wild type but not kinase dead PTK6 restored proliferation of BaF3 cells following withdrawal of IL-3 [115]. Several earlier comprehensive reviews go into depth about the roles of PTK6 in signaling in cancer [37, 84, 106, 114, 124, 125].

Targeting PTK6 in Cancer

Increased PTK6 expression has been reported in several different types of cancer, and often patients with high levels of PTK6 expression exhibit reduced periods of disease remission and survival. No PTK6 specific inhibitors are currently used in the clinic. However, multiple compounds have been identified that display activity against PTK6 and these are summarized in Table 1.

Table 1.

Compounds that Target PTK6 Activity.

Inhibitor Targeted PTK6 In Vivo References Notes
Vemurafenib (PLX4032) and PLX4720 Yes [103, 145] Related B-RAF(V600E) inhibitors; PTK6 is a selective off-target
Ibrutinib No [56] Bruton’s tyrosine kinase (BTK) inhibitor that also inhibits BLK and PTK6
21a, 21c No [147] Type I PTK6 inhibitors
PF-6683324, PF-6689840 No [147] Type II PTK6 inhibitors
Erlotinib No [164] EGFR inhibitor; promoted PTK6 Y447 inhibitory phosphorylation
Pyrazolopyrimidines PP1 and PP2 No [157] SRC family kinase inhibitors
XMU-MP-2 Yes [146] Developed as a specific PTK6 inhibitor
Oleanolic acid derivatives No [163] Analogues of triterpene oleanolic acid extracted from Terminalia bentzoe L.
Phenylmethylene hydantoins Yes [162] Derivatives of Z-4-hydroxyphenylmethylene hydantoin (PMH)
Marine triterpene sipholenols derivatives No [161] Derivatives of sipholenol A, a sipholane triterpene isolated from the Red Sea sponge Callyspongia siphonella
Dasatinib No [29, 150, 151] Dual inhibitor of SRC/ABL and c-KIT

The ability of vemurafenib and the structurally related compound PLX4720 to inhibit PTK6 has been demonstrated in vitro and in vivo [103]. Both vemurafenib and PLX4720 were developed as small molecule inhibitors of BRAF V600E, but they also inhibit PTK6 as an off-target [143, 144]. Using a quantitative competitive binding assay, Vin et. al. calculated IC50s for a panel of kinases and showed that PTK6 is inhibited by vemurafenib and PLX4720 with efficacy comparable to inhibition of BRAF V600E [145]. Calculated IC50s for vemurafenib were 64.78 nM for BRAF(V600E) and 68.04 nM PTK6. The calculated IC50s for PLX4720 were 32.04 nM for BRAF(V600E) and 30.38 nM for PTK6. Vemurafenib inhibited PTK6 activity and downstream signaling in prostate cancer cell lines, and treatment with PLX4720 reduced xenograft prostate tumor growth in mice [103]. Vemurafenib targets the PTK6 catalytic domain and interacts with PTK6 through sulfonamide and propyl groups with minimal interaction with azaindole part of the molecule [103].

Jiang et al. performed high-throughput screening of a compound library designed to target ATP binding sites of kinases and identified and further developed a novel small molecule XMU-MP-2 that acts as a potent inhibitor of PTK6 with low toxicity [146]. XMu-MP-2 exhibited an IC50 of 29.7 nmol/L in BRK transformed Ba/F3 cells and was shown to bind PTK6 and inhibit its activity. The drug suppressed growth of tumors induced by PTK6 transformed cells in xenograft models and showed synergistic effects with a type I ERBB2 inhibitor and tamoxifen [146].

Qiu et al. identified novel small molecule Type I and Type II PTK6 inhibitors that were used to inhibit PTK6 in breast cancer cell lines with endogenous PTK6 or engineered to overexpress wild type PTK6, and it was concluded that PTK6 inhibition did not significantly impact tumor cell growth [147]. A caveat with the cell culture experiments is that only cell lines with very low endogenous PTK6 activity were targeted in vitro; PTK6 activation was not detected by immunoblotting and cells were not stimulated to promote kinase activation prior to the treatment with inhibitors. In addition, the influence of possible inhibitory phosphorylation at the terminal carboxy tyrosine residue was not taken into account in the experiments with ectopic overexpression of wild type PTK6 (Figure 2B). PTK6 expression does not correlate with its activation, and it is important to take contributions of environmental cues and signaling pathways that activate PTK6 into consideration when designing assays to test the efficacy of PTK6 inhibitors.

Dasatinib is a small molecule inhibitor of BCR-ABL and SRC family tyrosine kinases used for treatment of several types of leukemias [148, 149]. PTK6 is one of the off-target tyrosine kinases inhibited by dasatinib [150]. Using in vitro kinase assays, Anastassiadis et al. demonstrated that dasatinib reduces PTK6 activity to 4.7% when normalized to the uninhibited reaction [151]. Dasatinib binds to the PTK6 ATP binding pocket [31] in manner similar to how it interacts with ABL [152], BMX [153], and BTK [154]. Lee and colleagues performed chemical library screens to identify potential PTK6 inhibitors [155, 156] and determined that the SRC-family inhibitors PP1 and PP2, and a LCK inhibitor have some selectivity against PTK6 [157].

Several natural products and their derivatives have been described in literature as potential PTK6 inhibitors, but mechanisms of action and efficacy in vivo have not been extensively studied. It was shown that treating breast cancer cell line MDA-MB-231 with Hwanggeumchal sorghum leads to downregulation of PTK6 expression, although the mechanism was not elucidated [158].

Novel synthesized 4-anilino-α-carboline derivatives were screened in silico as potential PTK6 inhibitors. Molecular docking was performed with PTK6 homology model and IC50 were determined for all the analogues [159], and two derivatives, MK138 and MK150 were used for further in vitro studies. Two derivatives were shown to target PTK6 and reduce activation of STAT3 in cell lines [160]. Bioactive sipholane triterpene from Red Sea sponge Callyspongia siphonellai was used as a starting point for development of novel molecules that could potentially exibit inhibitory effect on some enzymes. Ester derivatives showed the best results in vitro so they were used in KINOMEscan where PTK6 was detected as a major target among 451 human protein kinases. Derivatives of this molecule reduce invasiveness and migration of MDA-MB-231 cells [161].

Some compounds have been shown to promote a decrease in activating Y342 phosphorylation or an increase in inhibitory Y447 phosphorylation. Marine natural products were used as a starting material for synthesis of phenylmethylene hydantoins which inhibited PTK6 phosphorylation in MDA-MB-231 cell lines and in a murine xenograft model [162]. Oleanolic acid derivatives that inhibited breast cancer cell line migration and invasion also inhibited PTK6 phosphorylation [163]. These last compounds require further characterization to understand their value because specific phosphorylation sites of PTK6 that were affected were not identified and PTK6 kinase activity was not measured. Using a deep phosphotyrosine proteomics approach Abe et. al. reported increase in Y447 phosphorylation upon treatment with erlotinib [164].

PTK6 expression is induced by chemotherapeutic agents and radiation, and it may be particularly useful to target PTK6 when administering combination chemotherapy. Induction of PTK6 was important for survival of breast cancer cells following Taxol treatment [46]. Treatment of pancreatic cancer cell lines with gemcitabine led to induction of PTK6 [53]. The PTK6 inhibitor XMU-MP-2 synergized with tamoxifen inhibiting growth of ER positive MCF7 cells grown in the absence of estrogen [146]. PTK6 expression/activation was found to be part of the kinome reprogramming that occurred with acquisition of lapatinib resistance in ERBB2 positive breast cancer cells [165]. PTK6 expression increased in bladder cancer cells that were treated with a PAK4 inhibitor and targeting both PTK6 and PAK4 reduced viability of the bladder cancer cells better than targeting either kinase alone [54].

SUMMARY

PTK6 has been described as an “effective collaborative oncogene” [82]. It is capable of promoting oncogenic signaling from a variety of growth factor receptors including members of the ERBB family, IGF1R, and MET. In addition, PTK6 regulates a number of intracellular signaling pathways and substrates (see Figure 3), which can be differentially impacted by cell type specific factors and intracellular localization of the PTK6.

A variety of data indicates that targeting PTK6 may have therapeutic benefits in specific cancers, particularly those that express high levels of PTK6 and/or maintain PTK6 activation at the plasma membrane. These would include cancers with amplification of specific growth factor receptors or loss of inhibitors such as the tumor suppressor PTEN. In addition, PTK6 expression and activity may be induced by cancer treatment regimens. Multiple genetic studies have demonstrated instrumental roles for PTK6 in breast and prostate cancer. Genetic ablation of Ptk6 has been shown to impact breast, prostate, skin, and colon tumorigenesis in vivo. In normal tissues where PTK6 is expressed at highest levels, such as the intestine and skin, PTK6 induction in progenitor cells following DNA-damage may also promote tumorigenesis by enhancing cell survival. PTK6 activation can be regulated by environmental signals, intracellular substrates and associated proteins, including other kinases and phosphatases, and intracellular localization. It is important to take into account the activation status, localization and accessibility of PTK6 when evaluating the functionality of novel PTK6 inhibitors in cancer cells and tumor tissues.

ACKNOWLEDGEMENTS

We thank Wanian M. Alwanian and Katarina Vlajic for careful reading of the manuscript and helpful comments. Studies were supported by NIH grant RO1 CA188427.

Footnotes

Disclosure of Potential Conflicts of Interest: The authors have no conflicts of interest to report.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • [1].Lee ST, Strunk KM, Spritz RA, A survey of protein tyrosine kinase mRNAs expressed in normal human melanocytes, Oncogene 8(12) (1993) 3403–10. [PubMed] [Google Scholar]
  • [2].Mitchell PJ, Barker KT, Martindale JE, Kamalati T, Lowe PN, Page MJ, Gusterson BA, Crompton MR, Cloning and characterisation of cDNAs encoding a novel non-receptor tyrosine kinase, brk, expressed in human breast tumours., Oncogene 9 (1994) 2383–2390. [PubMed] [Google Scholar]
  • [3].Siyanova EY, Serfas MS, Mazo IA, Tyner AL, Tyrosine kinase gene expression in the mouse small intestine, Oncogene 9(7) (1994) 2053–7. [PubMed] [Google Scholar]
  • [4].Vasioukhin V, Serfas MS, Siyanova EY, Polonskaia M, Costigan VJ, Liu B, Thomason A, Tyner AL, A novel intracellular epithelial cell tyrosine kinase is expressed in the skin and gastrointestinal tract, Oncogene 10(2) (1995) 349–57. [PubMed] [Google Scholar]
  • [5].Cance WG, Craven RJ, Weiner TM, Liu ET, Novel protein kinases expresed in human breast cancer, Int. J. Cancer 54 (1993) 571–577. [DOI] [PubMed] [Google Scholar]
  • [6].Cance WG, Craven RJ, Bergman M, Xu L, Aitalo K, Liu ET, Rak, a novel nuclear tyrosine kinase expressed in epithelial cells, Cell Growth Diff. 5 (1994) 1347–1355. [PubMed] [Google Scholar]
  • [7].Lee J, Wang Z, Luoh S-M, Wood WL, Scadden DT, Cloning of FRK, a novel human intracellular SRC-like tyrosine kinase-encoding gene, Gene 138 (1994) 247–251. [DOI] [PubMed] [Google Scholar]
  • [8].Kohmura N, Yagi T, Tomooka Y, Oyanagi M, Kominami R, Takeda N, Chiba J, Ikawa Y, Aizawa S, A novel nonreceptor tyrosine kinase, Srm: cloning and targeted disruption, Mol Cell Biol 14(10) (1994) 6915–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].D’Aniello S, Irimia M, Maeso I, Pascual-Anaya J, Jimenez-Delgado S, Bertrand S, Garcia-Fernandez J, Gene expansion and retention leads to a diverse tyrosine kinase superfamily in amphioxus, Molecular biology and evolution 25(9) (2008) 1841–54. [DOI] [PubMed] [Google Scholar]
  • [10].Serfas MS, Tyner AL, Brk, Srm, Frk, and Src42A form a distinct family of intracellular Src-like tyrosine kinases, Oncology research 13(6–10) (2003) 409–19. [DOI] [PubMed] [Google Scholar]
  • [11].Park SH, Lee KH, Kim H, Lee ST, Assignment of the human PTK6 gene encoding a non-receptor protein tyrosine kinase to 20q13.3 by fluorescence in situ hybridization, Cytogenet Cell Genet 77(3–4) (1997) 271–2. [DOI] [PubMed] [Google Scholar]
  • [12].Kolaitis G, Bouwkamp CG, Papakonstantinou A, Otheiti I, Belivanaki M, Haritaki S, Korpa T, Albani Z, Terzioglou E, Apostola P, Skamnaki A, Xaidara A, Kosma K, Kitsiou-Tzeli S, Tzetis M, A boy with conduct disorder (CD), attention deficit hyperactivity disorder (ADHD), borderline intellectual disability, and 47,XXY syndrome in combination with a 7q11.23 duplication, 11p15.5 deletion, and 20q13.33 deletion, Child Adolesc Psychiatry Ment Health 10 (2016) 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Kallioniemi A, Kallioniemi OP, Piper J, Tanner M, Stokke T, Chen L, Smith HS, Pinkel D, Gray JW, Waldman FM, Detection and mapping of amplified DNA sequences in breast cancer by comparative genomic hybridization, Proc Natl Acad Sci U S A 91(6) (1994) 2156–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Zhao C, Yasui K, Lee CJ, Kurioka H, Hosokawa Y, Oka T, Inazawa J, Elevated expression levels of NCOA3, TOP1, and TFAP2C in breast tumors as predictors of poor prognosis, Cancer 98(1) (2003) 18–23. [DOI] [PubMed] [Google Scholar]
  • [15].Schmandt RE, Bennett M, Clifford S, Thornton A, Jiang F, Broaddus RR, Sun CC, Lu KH, Sood AK, Gershenson DM, The BRK tyrosine kinase is expressed in high-grade serous carcinoma of the ovary, Cancer Biol Ther 5(9) (2006) 1136–41. [DOI] [PubMed] [Google Scholar]
  • [16].Xiang B, Chatti K, Qiu H, Lakshmi B, Krasnitz A, Hicks J, Yu M, Miller WT, Muthuswamy SK, Brk is coamplified with ErbB2 to promote proliferation in breast cancer, Proc Natl Acad Sci U S A 105(34) (2008) 12463–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Paul MR, Pan TC, Pant DK, Shih NN, Chen Y, Harvey KL, Solomon A, Lieberman D, Morrissette JJ, Soucier-Ernst D, Goodman NG, Stavropoulos SW, Maxwell KN, Clark C, Belka GK, Feldman M, DeMichele A, Chodosh LA, Genomic landscape of metastatic breast cancer identifies preferentially dysregulated pathways and targets, J Clin Invest (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Brauer PM, Zheng Y, Evans MD, Dominguez-Brauer C, Peehl DM, Tyner AL, The alternative splice variant of protein tyrosine kinase 6 negatively regulates growth and enhances PTK6-mediated inhibition of beta-catenin, PLoS ONE 6(3) (2011) e14789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Mitchell PJ, Barker KT, Shipley J, Crompton MR, Characterisation and chromosome mapping of the human non receptor tyrosine kinase gene, brk, Oncogene 15(12) (1997) 1497–502. [DOI] [PubMed] [Google Scholar]
  • [20].Patel P, Asbach B, Shteyn E, Gomez C, Coltoff A, Bhuyan S, Tyner AL, Wagner R, Blain SW, Brk/Protein tyrosine kinase 6 phosphorylates p27KIP1, regulating the activity of cyclin D-cyclin-dependent kinase 4, Mol Cell Biol 35(9) (2015) 1506–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Fan G, Aleem S, Yang M, Miller WT, Tonks NK, Protein-tyrosine Phosphatase and Kinase Specificity in Regulation of SRC and Breast Tumor Kinase, J Biol Chem 290(26) (2015) 15934–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].McClendon CJ, Miller WT, Structure, Function, and Regulation of the SRMS Tyrosine Kinase, Int J Mol Sci 21(12) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Qiu H, Miller WT, Role of the Brk SH3 domain in substrate recognition, Oncogene 23(12) (2004) 2216–23. [DOI] [PubMed] [Google Scholar]
  • [24].Ko S, Ahn K-E, Lee Y-M, Ahn H-C, Lee W, Structural basis of the auto-inhibition mechanism of nonreceptor tyrosine kinase PTK6, Biochemical and Biophysical Research Communications 384(2) (2009) 236–242. [DOI] [PubMed] [Google Scholar]
  • [25].Derry JJ, Richard S, Valderrama Carvajal H, Ye X, Vasioukhin V, Cochrane AW, Chen T, Tyner AL, Sik (BRK) phosphorylates Sam68 in the nucleus and negatively regulates its RNA binding ability, Mol Cell Biol 20(16) (2000) 6114–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Hong E, Shin J, Bang E, Kim MH, Lee ST, Lee W, Complete sequence-specific 1H, 13C and 15N resonance assignments of the human PTK6 SH2 domain, J Biomol NMR 19(3) (2001) 291–2. [DOI] [PubMed] [Google Scholar]
  • [27].Hong E, Shin J, Kim HI, Lee ST, Lee W, Solution structure and backbone dynamics of the non-receptor protein-tyrosine kinase-6 Src homology 2 domain, J Biol Chem 279(28) (2004) 29700–8. [DOI] [PubMed] [Google Scholar]
  • [28].Park M-J, Sheng R, Silkov A, Jung D-J, Wang Z-G, Xin Y, Kim H, Thiagarajan-Rosenkranz P, Song S, Yoon Y, Nam W, Kim I, Kim E, Lee D-G, Chen Y, Singaram I, Wang L, Jang MH, Hwang C-S, Honig B, Ryu S, Lorieau J, Kim Y-M, Cho W, SH2 Domains Serve as Lipid-Binding Modules for pTyr-Signaling Proteins, Molecular cell 62(1) (2016) 7–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Thakur MK, Kumar A, Birudukota S, Swaminathan S, Tyagi R, Gosu R, Crystal structure of the kinase domain of human protein tyrosine kinase 6 (PTK6) at 2.33 Å resolution, Biochemical and Biophysical Research Communications 478(2) (2016) 637–642. [DOI] [PubMed] [Google Scholar]
  • [30].Azam M, Seeliger MA, Gray NS, Kuriyan J, Daley GQ, Activation of tyrosine kinases by mutation of the gatekeeper threonine, Nat Struct Mol Biol 15(10) (2008) 1109–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Thakur MK, Birudukota S, Swaminathan S, Battula SK, Vadivelu S, Tyagi R, Gosu R, Co-crystal structures of PTK6: With Dasatinib at 2.24 Å, with novel imidazo[1,2-a]pyrazin-8-amine derivative inhibitor at 1.70 Å resolution, Biochemical and Biophysical Research Communications 482(4) (2017) 1289–1295. [DOI] [PubMed] [Google Scholar]
  • [32].Kamalati T, Jolin HE, Mitchell PJ, Barker KT, Jackson LE, Dean CJ, Page MJ, Gusterson BA, Crompton MR, Brk, a breast tumor-derived non-receptor protein-tyrosine kinase, sensitizes mammary epithelial cells to epidermal growth factor, J Biol Chem 271(48) (1996) 30956–63. [DOI] [PubMed] [Google Scholar]
  • [33].Qiu H, Miller WT, Regulation of the nonreceptor tyrosine kinase Brk by autophosphorylation and by autoinhibition, J Biol Chem 277(37) (2002) 34634–41. [DOI] [PubMed] [Google Scholar]
  • [34].Takeda H, Kawamura Y, Miura A, Mori M, Wakamatsu A, Yamamoto J, Isogai T, Matsumoto M, Nakayama KI, Natsume T, Nomura N, Goshima N, Comparative analysis of human SRC-family kinase substrate specificity in vitro, J Proteome Res 9(11) (2010) 5982–93. [DOI] [PubMed] [Google Scholar]
  • [35].Lin A, Li C, Xing Z, Hu Q, Liang K, Han L, Wang C, Hawke DH, Wang S, Zhang Y, Wei Y, Ma G, Park PK, Zhou J, Zhou Y, Hu Z, Marks JR, Liang H, Hung MC, Lin C, Yang L, The LINK-A lncRNA activates normoxic HIF1alpha signalling in triple-negative breast cancer, Nature cell biology 18(2) (2016) 213–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Shin DS, Kim YG, Kim EM, Kim M, Park HY, Kim JH, Lee BS, Kim BG, Lee YS, Solid-phase peptide library synthesis on HiCore resin for screening substrate specificity of Brk protein tyrosine kinase, Journal of combinatorial chemistry 10(1) (2008) 20–3. [DOI] [PubMed] [Google Scholar]
  • [37].Brauer PM, Tyner AL, Building a better understanding of the intracellular tyrosine kinase PTK6 - BRK by BRK, Biochim Biophys Acta 1806(1) (2010) 66–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Tsui T, Miller WT, Cancer-Associated Mutations in Breast Tumor Kinase/PTK6 Differentially Affect Enzyme Activity and Substrate Recognition, Biochemistry 54(20) (2015) 3173–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal, Sci Signal 6(269) (2013) pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data, Cancer Discov 2(5) (2012) 401–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, Boutselakis H, Cole CG, Creatore C, Dawson E, Fish P, Harsha B, Hathaway C, Jupe SC, Kok CY, Noble K, Ponting L, Ramshaw CC, Rye CE, Speedy HE, Stefancsik R, Thompson SL, Wang S, Ward S, Campbell PJ, Forbes SA, COSMIC: the Catalogue Of Somatic Mutations In Cancer, Nucleic Acids Res 47(D1) (2019) D941–D947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Kang KN, Kim M, Pae KM, Lee ST, Characterization of the 5’-flanking region of the human PTK6 gene, Biochim Biophys Acta 1574(3) (2002) 365–9. [DOI] [PubMed] [Google Scholar]
  • [43].Yu G, Lee YC, Cheng CJ, Wu CF, Song JH, Gallick GE, Yu-Lee LY, Kuang J, Lin SH, RSK promotes prostate cancer progression in bone through ING3, CKAP2, and PTK6-mediated cell survival, Mol Cancer Res 13(2) (2015) 348–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Zhao S, Li J, Sphingosine-1-phosphate induces the migration of thyroid follicular carcinoma cells through the microRNA-17/PTK6/ERK1/2 pathway, PLoS ONE 10(3) (2015) e0119148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Regan Anderson TM, Peacock DL, Daniel AR, Hubbard GK, Lofgren KA, Girard BJ, Schorg A, Hoogewijs D, Wenger RH, Seagroves TN, Lange CA, Breast tumor kinase (Brk/PTK6) is a mediator of hypoxia-associated breast cancer progression, Cancer Res 73(18) (2013) 5810–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Regan Anderson TM, Ma S, Perez Kerkvliet C, Peng Y, Helle TM, Krutilina RI, Raj GV, Cidlowski JA, Ostrander JH, Schwertfeger KL, Seagroves TN, Lange CA, Taxol Induces Brk-dependent Prosurvival Phenotypes in TNBC Cells through an AhR/GR/HIF-driven Signaling Axis, Mol Cancer Res 16(11) (2018) 1761–1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Miah S, Bagu E, Goel R, Ogunbolude Y, Dai C, Ward A, Vizeacoumar FS, Davies G, Vizeacoumar FJ, Anderson D, Lukong KE, Estrogen receptor signaling regulates the expression of the breast tumor kinase in breast cancer cells, BMC Cancer 19(1) (2019) 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Ito K, Park SH, Katsyv I, Zhang W, De Angelis C, Schiff R, Irie HY, PTK6 regulates growth and survival of endocrine therapy-resistant ER+ breast cancer cells, NPJ Breast Cancer 3 (2017) 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Haegebarth A, Perekatt AO, Bie W, Gierut JJ, Tyner AL, Induction of protein tyrosine kinase 6 in mouse intestinal crypt epithelial cells promotes DNA damage-induced apoptosis, Gastroenterology 137(3) (2009) 945–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Gierut JJ, Mathur PS, Bie W, Han J, Tyner AL, Targeting protein tyrosine kinase 6 enhances apoptosis of colon cancer cells following DNA damage, Molecular Cancer Therapeutics 11(11) (2012) 2311–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Chastkofsky MI, Bie W, Ball-Kell SM, He YY, Tyner AL, Protein Tyrosine Kinase 6 Regulates UVB-Induced Signaling and Tumorigenesis in Mouse Skin, The Journal of investigative dermatology 135(10) (2015) 2492–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Gierut J, Zheng Y, Bie W, Carroll RE, Ball-Kell S, Haegebarth A, Tyner AL, Disruption of the mouse protein tyrosine kinase 6 gene prevents STAT3 activation and confers resistance to azoxymethane, Gastroenterology 141(4) (2011) 1371–80, 1380 e1–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Ono H, Basson MD, Ito H, PTK6 Potentiates Gemcitabine-Induced Apoptosis by Prolonging S-phase and Enhancing DNA Damage in Pancreatic Cancer, Molecular cancer research : MCR 13(8) (2015) 1174–84. [DOI] [PubMed] [Google Scholar]
  • [54].Chandrashekar DS, Chakravarthi BVSK, Robinson AD, Anderson JC, Agarwal S, Balasubramanya SAH, Eich M-L, Bajpai AK, Davuluri S, Guru MS, Guru AS, Naik G, Della Manna DL, Acharya KK, Carskadon S, Manne U, Crossman DK, Ferguson JE, Grizzle WE, Palanisamy N, Willey CD, Crowley MR, Netto GJ, Yang ES, Varambally S, Sonpavde G, Therapeutically actionable PAK4 is amplified, overexpressed, and involved in bladder cancer progression, Oncogene (2020). [DOI] [PubMed] [Google Scholar]
  • [55].Ostrander JH, Daniel AR, Lofgren K, Kleer CG, Lange CA, Breast tumor kinase (protein tyrosine kinase 6) regulates heregulin-induced activation of ERK5 and p38 MAP kinases in breast cancer cells, Cancer Res 67(9) (2007) 4199–209. [DOI] [PubMed] [Google Scholar]
  • [56].Cagle P, Niture S, Srivastava A, Ramalinga M, Aqeel R, Rios-Colon L, Chimeh U, Suy S, Collins SP, Dahiya R, Kumar D, MicroRNA-214 targets PTK6 to inhibit tumorigenic potential and increase drug sensitivity of prostate cancer cells, Sci Rep 9(1) (2019) 9776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Haines RJ, Beard RS Jr., Eitner RA, Chen L, Wu MH, TNFα/IFNγ Mediated Intestinal Epithelial Barrier Dysfunction Is Attenuated by MicroRNA-93 Downregulation of PTK6 in Mouse Colonic Epithelial Cells, PLoS One 11(4) (2016) e0154351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Zhang F, Luo Y, Shao Z, Xu L, Liu X, Niu Y, Shi J, Sun X, Liu Y, Ding Y, Zhao L, MicroRNA-187, a downstream effector of TGFbeta pathway, suppresses Smad-mediated epithelial-mesenchymal transition in colorectal cancer, Cancer Lett 373(2) (2016) 203–13. [DOI] [PubMed] [Google Scholar]
  • [59].Penna E, Orso F, Taverna D, miR-214 as a key hub that controls cancer networks: small player, multiple functions, J Invest Dermatol 135(4) (2015) 960–969. [DOI] [PubMed] [Google Scholar]
  • [60].Zhu XB, Zhang ZC, Han GS, Han JZ, Qiu DP, Overexpression of miR214 promotes the progression of human osteosarcoma by regulating the Wnt/betacatenin signaling pathway, Mol Med Rep 15(4) (2017) 1884–1892. [DOI] [PubMed] [Google Scholar]
  • [61].Wang H, An H, Wang B, Liao Q, Li W, Jin X, Cui S, Zhang Y, Ding Y, Zhao L, miR-133a represses tumour growth and metastasis in colorectal cancer by targeting LIM and SH3 protein 1 and inhibiting the MAPK pathway, Eur J Cancer 49(18) (2013) 3924–35. [DOI] [PubMed] [Google Scholar]
  • [62].Kang SA, Cho HS, Yoon JB, Chung IK, Lee ST, Hsp90 rescues PTK6 from proteasomal degradation in breast cancer cells, Biochem J 447(2) (2012) 313–20. [DOI] [PubMed] [Google Scholar]
  • [63].Pires IM, Blokland NJ, Broos AW, Poujade FA, Senra JM, Eccles SA, Span PN, Harvey AJ, Hammond EM, HIF-1alpha-independent hypoxia-induced rapid PTK6 stabilization is associated with increased motility and invasion, Cancer Biol Ther 15(10) (2014) 1350–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Ai M, Qiu S, Lu Y, Fan Z, HER2 regulates Brk/PTK6 stability via upregulating calpastatin, an inhibitor of calpain, Cell Signal 25(9) (2013) 1754–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Fan G, Lin G, Lucito R, Tonks NK, Protein-tyrosine phosphatase 1B antagonized signaling by insulin-like growth factor-1 receptor and kinase BRK/PTK6 in ovarian cancer cells, J Biol Chem 288(34) (2013) 24923–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Wozniak DJ, Kajdacsy-Balla A, Macias V, Ball-Kell S, Zenner ML, Bie W, Tyner AL, PTEN is a protein phosphatase that targets active PTK6 and inhibits PTK6 oncogenic signaling in prostate cancer, Nat Commun 8(1) (2017) 1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Zheng Y, Wang Z, Bie W, Brauer PM, Perez White BE, Li J, Nogueira V, Raychaudhuri P, Hay N, Tonetti DA, Macias V, Kajdacsy-Balla A, Tyner AL, PTK6 activation at the membrane regulates epithelial-mesenchymal transition in prostate cancer, Cancer Research 73(17) (2013) 5426–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Chen HY, Shen CH, Tsai YT, Lin FC, Huang YP, Chen RH, Brk activates rac1 and promotes cell migration and invasion by phosphorylating paxillin, Mol Cell Biol 24(24) (2004) 10558–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Zheng Y, Peng M, Wang Z, Asara JM, Tyner AL, Protein tyrosine kinase 6 directly phosphorylates AKT and promotes AKT activation in response to epidermal growth factor, Molecular and Cellular Biology 30(17) (2010) 4280–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Vasioukhin V, Tyner AL, A role for the epithelial-cell-specific tyrosine kinase Sik during keratinocyte differentiation, Proc Natl Acad Sci U S A 94(26) (1997) 14477–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Chakraborty G, Jain S, Kundu GC, Osteopontin promotes vascular endothelial growth factor-dependent breast tumor growth and angiogenesis via autocrine and paracrine mechanisms, Cancer Res 68(1) (2008) 152–61. [DOI] [PubMed] [Google Scholar]
  • [72].Brauer PM, Zheng Y, Wang L, Tyner AL, Cytoplasmic retention of protein tyrosine kinase 6 promotes growth of prostate tumor cells, Cell Cycle 9(20) (2010) 4190–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Zheng Y, Gierut J, Wang Z, Miao J, Asara JM, Tyner AL, Protein tyrosine kinase 6 protects cells from anoikis by directly phosphorylating focal adhesion kinase and activating AKT, Oncogene 32(36) (2013) 4304–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Palka-Hamblin HL, Gierut JJ, Bie W, Brauer PM, Zheng Y, Asara JM, Tyner AL, Identification of beta-catenin as a target of the intracellular tyrosine kinase PTK6, J Cell Sci 123(Pt 2) (2010) 236–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Derry JJ, Prins GS, Ray V, Tyner AL, Altered localization and activity of the intracellular tyrosine kinase BRK/Sik in prostate tumor cells, Oncogene 22(27) (2003) 4212–20. [DOI] [PubMed] [Google Scholar]
  • [76].Ie Kim H, Lee ST, Oncogenic functions of PTK6 are enhanced by its targeting to plasma membrane but abolished by its targeting to nucleus, Journal of biochemistry 146(1) (2009) 133–9. [DOI] [PubMed] [Google Scholar]
  • [77].Lang YD, Chen HY, Ho CM, Shih JH, Hsu EC, Shen R, Lee YC, Chen JW, Wu CY, Yeh HW, Chen RH, Jou YS, PSPC1-interchanged interactions with PTK6 and beta-catenin synergize oncogenic subcellular translocations and tumor progression, Nat Commun 10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Lang YD, Jou YS, PSPC1: a contextual determinant of tumor progression, Mol Cell Oncol 7(2) (2020) 1721253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Aubele M, Walch AK, Ludyga N, Braselmann H, Atkinson MJ, Luber B, Auer G, Tapio S, Cooke T, Bartlett JMS, Prognostic value of protein tyrosine kinase 6 (PTK6) for lon-gterm survival of breast cancer patients, British journal of cancer 99(7) (2008) 1089–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Kamalati T, Jolin HE, Fry MJ, Crompton MR, Expression of the BRK tyrosine kinase in mammary epithelial cells enhances the coupling of EGF signalling to PI 3-kinase and Akt, via erbB3 phosphorylation, Oncogene 19(48) (2000) 5471–6. [DOI] [PubMed] [Google Scholar]
  • [81].Li X, Lu Y, Liang K, Hsu JM, Albarracin C, Mills GB, Hung MC, Fan Z, Brk/PTK6 sustains activated EGFR signaling through inhibiting EGFR degradation and transactivating EGFR, Oncogene 31(40) (2012) 4372–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Irie HY, Shrestha Y, Selfors LM, Frye F, Iida N, Wang Z, Zou L, Yao J, Lu Y, Epstein CB, Natesan S, Richardson AL, Polyak K, Mills GB, Hahn WC, Brugge JS, PTK6 regulates IGF-1-induced anchorage-independent survival, PLoS ONE 5(7) (2010) e11729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Castro NE, Lange CA, Breast tumor kinase and extracellular signal-regulated kinase 5 mediate Met receptor signaling to cell migration in breast cancer cells, Breast Cancer Res 12(4) (2010) R60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Locatelli A, Lofgren KA, Daniel AR, Castro NE, Lange CA, Mechanisms of HGF/Met signaling to Brk and Sam68 in breast cancer progression, Horm Cancer 3(1–2) (2012) 14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Shin WS, Shim HJ, Lee YH, Pyo M, Park JS, Ahn SY, Lee ST, PTK6 Localized at the Plasma Membrane Promotes Cell Proliferation and MigratiOn Through Phosphorylation of Eps8, J Cell Biochem 118(9) (2017) 2887–2895. [DOI] [PubMed] [Google Scholar]
  • [86].Lukong KE, Larocque D, Tyner AL, Richard S, Tyrosine phosphorylation of sam68 by breast tumor kinase regulates intranuclear localization and cell cycle progression, J Biol Chem 280(46) (2005) 38639–47. [DOI] [PubMed] [Google Scholar]
  • [87].Haegebarth A, Heap D, Bie W, Derry JJ, Richard S, Tyner AL, The nuclear tyrosine kinase BRK/Sik phosphorylates and inhibits the RNA-binding activities of the Sam68-like mammalian proteins SLM-1 and SLM-2, J Biol Chem 279(52) (2004) 54398–404. [DOI] [PubMed] [Google Scholar]
  • [88].Coyle JH, Guzik BW, Bor Y-C, Jin L, Eisner-Smerage L, Taylor SJ, Rekosh D, Hammarskjöld M-L, Sam68 enhances the cytoplasmic utilization of intron-containing RNA and is functionally regulated by the nuclear kinase Sik/BRK, Molecular and cellular biology 23(1) (2003) 92–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Lukong KE, Huot ME, Richard S, BRK phosphorylates PSF promoting its cytoplasmic localization and cell cycle arrest, Cell Signal 21(9) (2009) 1415–22. [DOI] [PubMed] [Google Scholar]
  • [90].Lukong KE, Richard S, Breast tumor kinase BRK requires kinesin-2 subunit KAP3A in modulation of cell migration, Cell Signal 20(2) (2008) 432–42. [DOI] [PubMed] [Google Scholar]
  • [91].Liu L, Gao Y, Qiu H, Miller WT, Poli V, Reich NC, Identification of STAT3 as a specific substrate of breast tumor kinase, Oncogene 25(35) (2006) 4904–12. [DOI] [PubMed] [Google Scholar]
  • [92].Weaver AM, Silva CM, Signal transducer and activator of transcription 5b: a new target of breast tumor kinase/protein tyrosine kinase 6, Breast Cancer Res 9(6) (2007) R79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Tang C, Takahashi-Kanemitsu A, Kikuchi I, Ben C, Hatakeyama M, Transcriptional Coactivator Functions of YAP and TAZ Are Inversely Regulated by Tyrosine Phosphorylation Status of Parafibromin, iScience 2 (2018) 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Peng M, Ball-Kell SM, Tyner AL, Protein tyrosine kinase 6 promotes ERBB2-induced mammary gland tumorigenesis in the mouse, Cell Death & Disease 6 (2015) e1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Kikuchi I, Takahashi-Kanemitsu A, Sakiyama N, Tang C, Tang P-J, Noda S, Nakao K, Kassai H, Sato T, Aiba A, Hatakeyama M, Dephosphorylated parafibromin is a transcriptional coactivator of the Wnt/Hedgehog/Notch pathways, Nature communications 7 (2016) 12887–12887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Mitchell PJ, Sara EA, Crompton MR, A novel adaptor-like protein which is a substrate for the non-receptor tyrosine kinase, BRK, Oncogene 19(37) (2000) 4273–82. [DOI] [PubMed] [Google Scholar]
  • [97].Ikeda O, Miyasaka Y, Sekine Y, Mizushima A, Muromoto R, Nanbo A, Yoshimura A, Matsuda T, STAP-2 is phosphorylated at tyrosine-250 by Brk and modulates Brk-mediated STAT3 activation, Biochem Biophys Res Commun 384(1) (2009) 71–5. [DOI] [PubMed] [Google Scholar]
  • [98].Zheng Y, Asara JM, Tyner AL, Protein-tyrosine Kinase 6 Promotes Peripheral Adhesion Complex Formation and Cell Migration by Phosphorylating p130 CRK-associated Substrate, The Journal of Biological Chemistry 287(1) (2012) 148–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Miah S, Goel RK, Dai C, Kalra N, Beaton-Brown E, Bagu ET, Bonham K, Lukong KE, BRK targets Dok1 for ubiquitin-mediated proteasomal degradation to promote cell proliferation and migration, PLoS ONE 9(2) (2014) e87684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Ikeda O, Mizushima A, Sekine Y, Yamamoto C, Muromoto R, Nanbo A, Oritani K, Yoshimura A, Matsuda T, Involvement of STAP-2 in Brk-mediated phosphorylation and activation of STAT5 in breast cancer cells, Cancer Sci 102(4) (2011) 756–61. [DOI] [PubMed] [Google Scholar]
  • [101].Ikeda O, Sekine Y, Mizushima A, Nakasuji M, Miyasaka Y, Yamamoto C, Muromoto R, Nanbo A, Oritani K, Yoshimura A, Matsuda T, Interactions of STAP-2 with Brk and STAT3 participate in cell growth of human breast cancer cells, J Biol Chem 285(49) (2010) 38093–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Patel P, Tsiperson V, Gottesman SRS, Somma J, Blain SW, Dual Inhibition of CDK4 and CDK2 via Targeting p27 Tyrosine Phosphorylation Induces a Potent and Durable Response in Breast Cancer Cells, Mol Cancer Res 16(3) (2018) 361–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Wozniak DJ, Hitchinson B, Gilic MB, Bie W, Gaponenko V, Tyner AL, Vemurafenib Inhibits Active PTK6 in PTEN-null Prostate Tumor Cells, Mol Cancer Ther 18(5) (2019) 937–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Shen CH, Chen HY, Lin MS, Li FY, Chang CC, Kuo ML, Settleman J, Chen RH, Breast tumor kinase phosphorylates p190RhoGAP to regulate rho and ras and promote breast carcinoma growth, migration, and invasion, Cancer Res 68(19) (2008) 7779–87. [DOI] [PubMed] [Google Scholar]
  • [105].Bucher-Johannessen C, Page CM, Haugen TB, Wojewodzic MW, Fossa SD, Grotmol T, Haugnes HS, Rounge TB, Cisplatin treatment of testicular cancer patients introduces long-term changes in the epigenome, Clin Epigenetics 11(1) (2019) 179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Harvey AJ, Crompton MR, The Brk protein tyrosine kinase as a therapeutic target in cancer: opportunities and challenges, Anticancer Drugs 15(2) (2004) 107–11. [DOI] [PubMed] [Google Scholar]
  • [107].Qiu H, Zappacosta F, Su W, Annan RS, Miller WT, Interaction between Brk kinase and insulin receptor substrate-4, Oncogene 24(36) (2005) 5656–64. [DOI] [PubMed] [Google Scholar]
  • [108].Harvey AJ, Crompton MR, Use of RNA interference to validate Brk as a novel therapeutic target in breast cancer: Brk promotes breast carcinoma cell proliferation, Oncogene 22(32) (2003) 5006–10. [DOI] [PubMed] [Google Scholar]
  • [109].Mathur PS, Gierut JJ, Guzman G, Xie H, Xicola RM, Llor X, Chastkofsky MI, Perekatt AO, Tyner AL, Kinase-Dependent and -Independent Roles for PTK6 in Colon Cancer, Molecular cancer research : MCR 14(6) (2016) 563–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Llor X, Serfas MS, Bie W, Vasioukhin V, Polonskaia M, Derry J, Abbott CM, Tyner AL, BRK/Sik expression in the gastrointestinal tract and in colon tumors, Clin Cancer Res 5(7) (1999) 1767–77. [PubMed] [Google Scholar]
  • [111].Petro BJ, Tan RC, Tyner AL, Lingen MW, Watanabe K, Differential expression of the non-receptor tyrosine kinase BRK in oral squamous cell carcinoma and normal oral epithelium, Oral Oncol 40(10) (2004) 1040–7. [DOI] [PubMed] [Google Scholar]
  • [112].Haegebarth A, Bie W, Yang R, Crawford SE, Vasioukhin V, Fuchs E, Tyner AL, Protein tyrosine kinase 6 negatively regulates growth and promotes enterocyte differentiation in the small intestine, Molecular and Cellular Biology 26(13) (2006) 4949–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Wang TC, Jee SH, Tsai TF, Huang YL, Tsai WL, Chen RH, Role of breast tumour kinase in the in vitro differentiation of HaCaT cells, Br J Dermatol 153(2) (2005) 282–9. [DOI] [PubMed] [Google Scholar]
  • [114].Zheng Y, Tyner AL, Context-specific protein tyrosine kinase 6 (PTK6) signalling in prostate cancer, Eur J Clin Invest 43(4) (2013) 397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Kasprzycka M, Majewski M, Wang ZJ, Ptasznik A, Wysocka M, Zhang Q, Marzec M, Gimotty P, Crompton MR, Wasik MA, Expression and oncogenic role of Brk (PTK6/Sik) protein tyrosine kinase in lymphocytes, Am J Pathol 168(5) (2006) 1631–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Lofgren KA, Ostrander JH, Housa D, Hubbard GK, Locatelli A, Bliss RL, Schwertfeger KL, Lange CA, Mammary gland specific expression of Brk/PTK6 promotes delayed involution and tumor formation associated with activation of p38 MAPK, Breast cancer research : BCR 13(5) (2011) R89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Peng M, Emmadi R, Wang Z, Wiley EL, Gann PH, Khan SA, Banerji N, McDonald W, Asztalos S, Pham TN, Tonetti DA, Tyner AL, PTK6/BRK is expressed in the normal mammary gland and activated at the plasma membrane in breast tumors, Oncotarget 5(15) (2014) 6038–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Whitehead RH, Robinson PS, Williams JA, Bie W, Tyner AL, Franklin JL, Conditionally immortalized colonic epithelial cell line from a Ptk6 null mouse that polarizes and differentiates in vitro, J Gastroenterol Hepatol 23(7 Pt 1) (2008) 1119–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Haines RJ, Beard RS Jr., Wu MH, Protein tyrosine kinase 6 mediates TNFalpha-induced endothelial barrier dysfunction, Biochem Biophys Res Commun 456(1) (2015) 190–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Haegebarth A, Nunez R, Tyner AL, The intracellular tyrosine kinase Brk sensitizes non-transformed cells to inducers of apoptosis, Cell Cycle 4(9) (2005) 1239–46. [DOI] [PubMed] [Google Scholar]
  • [121].Barker K, Jackson L, Crompton M, BRK tyrosine kinase expression in a high proportion of human breast carcinomas, Oncogene 15(7) (1997) 799. [DOI] [PubMed] [Google Scholar]
  • [122].Born M, Quintanilla-Fend L, Braselmann H, Reich U, Richter M, Hutzler P, Aubele M, Simultaneous over-expression of the Her2/neu and PTK6 tyrosine kinases in archival invasive ductal breast carcinomas, J Pathol 205(5) (2005) 592–6. [DOI] [PubMed] [Google Scholar]
  • [123].Regan Anderson TM, Ma SH, Raj GV, Cidlowski JA, Helle TM, Knutson TP, Krutilina RI, Seagroves TN, Lange CA, Breast Tumor Kinase (Brk/PTK6) Is Induced by HIF, Glucocorticoid Receptor, and PELP1-Mediated Stress Signaling in Triple-Negative Breast Cancer, Cancer Res 76(6) (2016) 1653–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Goel RK, Lukong KE, Tracing the footprints of the breast cancer oncogene BRK - Past till present, Biochim Biophys Acta 1856(1) (2015) 39–54. [DOI] [PubMed] [Google Scholar]
  • [125].Ostrander JH, Daniel AR, Lange CA, Brk/PTK6 signaling in normal and cancer cell models, Current opinion in pharmacology 10(6) (2010) 662–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Ito K, Park SH, Nayak A, Byerly JH, Irie HY, PTK6 inhibition suppresses metastases of triple negative breast cancer via Snail-dependent E-cadherin regulation, Cancer research (2016). [DOI] [PubMed] [Google Scholar]
  • [127].Park SH, Ito K, Olcott W, Katsyv I, Halstead-Nussloch G, Irie HY, PTK6 inhibition promotes apoptosis of Lapatinib-resistant Her2(+) breast cancer cells by inducing Bim, Breast Cancer Res 17 (2015) 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Harvey AJ, Pennington CJ, Porter S, Burmi RS, Edwards DR, Court W, Eccles SA, Crompton MR, Brk protects breast cancer cells from autophagic cell death induced by loss of anchorage, Am J Pathol 175(3) (2009) 1226–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Peng M, Ball-Kell SM, Franks RR, Xie H, Tyner AL, Protein tyrosine kinase 6 regulates mammary gland tumorigenesis in mouse models, Oncogenesis 2 (2013) e81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Alwanian WM, Tyner AL, Protein tyrosine kinase 6 signaling in prostate cancer, Am J Clin Exp Urol 8(1) (2020) 1–8. [PMC free article] [PubMed] [Google Scholar]
  • [131].Chen YF, Ma G, Cao X, Huang ZL, Zeng MS, Wen ZS, Downregulated expression of PTK6 is correlated with poor survival in esophageal squamous cell carcinoma, Med Oncol 31(12) (2014) 317. [DOI] [PubMed] [Google Scholar]
  • [132].Ma S, Bao JYJ, Kwan PS, Chan YP, Tong CM, Fu L, Zhang N, Tong AHY, Qin YR, Tsao SW, Chan KW, Lok S, Guan XY, Identification of PTK6, via RNA sequencing analysis, as a suppressor of esophageal squamous cell carcinoma, Gastroenterology 143(3) (2012) 675–686 e12. [DOI] [PubMed] [Google Scholar]
  • [133].Liu XK, Zhang XR, Zhong Q, Li MZ, Liu ZM, Lin ZR, Wu D, Zeng MS, Low expression of PTK6/Brk predicts poor prognosis in patients with laryngeal squamous cell carcinoma, J Transl Med 11 (2013) 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Wu D, Zhang X, Liu Z, Yan H, Mai J, Zhao Z, Zhong Q, Liu X, Decreased expression of protein tyrosine kinase 6 contributes to tumor progression and metastasis in laryngeal squamous cell carcinoma, Biochem Biophys Res Commun 503(3) (2018) 1378–1384. [DOI] [PubMed] [Google Scholar]
  • [135].Fan C, Zhao Y, Liu D, Zhang X, Wang E, Detection of Brk expression in non-small cell lung cancer: clinicopathological relevance, Tumour Biol 32(5) (2011) 873–80. [DOI] [PubMed] [Google Scholar]
  • [136].Zhao C, Chen Y, Zhang W, Zhang J, Xu Y, Li W, Chen S, Deng A, Expression of protein tyrosine kinase 6 (PTK6) in nonsmall cell lung cancer and their clinical and prognostic significance, Onco Targets Ther 6 (2013) 183–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Lin HS, Berry GJ, Fee WE Jr., Terris DJ, Sun Z, Identification of tyrosine kinases overexpressed in head and neck cancer, Arch Otolaryngol Head Neck Surg 130(3) (2004) 311–6. [DOI] [PubMed] [Google Scholar]
  • [138].Cho NL, Lin CI, Du J, Whang EE, Ito H, Moore FD Jr., Ruan DT, Global tyrosine kinome profiling of human thyroid tumors identifies Src as a promising target for invasive cancers, Biochem Biophys Res Commun 421(3) (2012) 508–13. [DOI] [PubMed] [Google Scholar]
  • [139].Ono H, Basson MD, Ito H, PTK6 promotes cancer migration and invasion in pancreatic cancer cells dependent on ERK signaling, PLoS ONE 9(5) (2014) e96060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Chen XH, Song B, Lin YL, Cao LJ, Feng SY, Zhang L, Wang FX, PTK6 promotes hepatocellular carcinoma cell proliferation and invasion, Am J Transl Res 8(10) (2016) 4354–4361. [PMC free article] [PubMed] [Google Scholar]
  • [141].Wang XJ, Xiong Y, Ma ZB, Xia JC, Li YF, The expression and prognostic value of protein tyrosine kinase 6 in early-stage cervical squamous cell cancer, Chin J Cancer 35(1) (2016) 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Xu XL, Ye YL, Wu ZM, He QM, Tan L, Xiao KH, Wu RY, Yu Y, Mai J, Li ZL, Peng XD, Huang Y, Li X, Zhang HL, Zhu XF, Qin ZK, Overexpression of PTK6 predicts poor prognosis in bladder cancer patients, J Cancer 8(17) (2017) 3464–3473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Sala E, Mologni L, Truffa S, Gaetano C, Bollag GE, Gambacorti-Passerini C, BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells, Molecular cancer research : MCR 6(5) (2008) 751–9. [DOI] [PubMed] [Google Scholar]
  • [144].Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KSM, Fong D, Zhu Y-L, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim S-H, Schlessinger J, Zhang KYJ, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G, Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity, Proceedings of the National Academy of Sciences 105(8) (2008) 3041–3046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Vin H, Ojeda SS, Ching G, Leung ML, Chitsazzadeh V, Dwyer DW, Adelmann CH, Restrepo M, Richards KN, Stewart LR, Du L, Ferguson SB, Chakravarti D, Ehrenreiter K, Baccarini M, Ruggieri R, Curry JL, Kim KB, Ciurea AM, Duvic M, Prieto VG, Ullrich SE, Dalby KN, Flores ER, Tsai KY, BRAF inhibitors suppress apoptosis through off-target inhibition of JNK signaling, Elife 2 (2013) e00969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Jiang J, Gui F, He Z, Li L, Li Y, Li S, Wu X, Deng Z, Sun X, Huang X, Huang W, Han S, Zhang T, Wang Z, Jiao B, Song S, Wang H, Chen L, Zhou D, Liu Q, Ren R, Zhang J, Deng X, Targeting BRK-Positive Breast Cancers with Small-Molecule Kinase Inhibitors, Cancer Res 77(1) (2017) 175–186. [DOI] [PubMed] [Google Scholar]
  • [147].Qiu L, Levine K, Gajiwala KS, Cronin CN, Nagata A, Johnson E, Kraus M, Tatlock J, Kania R, Foley T, Sun S, Small molecule inhibitors reveal PTK6 kinase is not an oncogenic driver in breast cancers, PLoS ONE 13(6) (2018) e0198374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Rix U, Hantschel O, Durnberger G, Remsing Rix LL, Planyavsky M, Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J, Kocher T, Superti-Furga G, Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets, Blood 110(12) (2007) 4055–63. [DOI] [PubMed] [Google Scholar]
  • [149].Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cornelius LA, Das J, Doweyko AM, Fairchild C, Hunt JT, Inigo I, Johnston K, Kamath A, Kan D, Klei H, Marathe P, Pang S, Peterson R, Pitt S, Schieven GL, Schmidt RJ, Tokarski J, Wen ML, Wityak J, Borzilleri RM, Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays, J Med Chem 47(27) (2004) 6658–61. [DOI] [PubMed] [Google Scholar]
  • [150].Li J, Rix U, Fang B, Bai Y, Edwards A, Colinge J, Bennett KL, Gao J, Song L, Eschrich S, Superti-Furga G, Koomen J, Haura EB, A chemical and phosphoproteomic characterization of dasatinib action in lung cancer, Nat Chem Biol 6(4) (2010) 291–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Anastassiadis T, Deacon SW, Devarajan K, Ma H, Peterson JR, Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity, Nat Biotechnol 29(11) (2011) 1039–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Tokarski JS, Newitt JA, Chang CY, Cheng JD, Wittekind M, Kiefer SE, Kish K, Lee FY, Borzillerri R, Lombardo LJ, Xie D, Zhang Y, Klei HE, The structure of Dasatinib (BMS-354825) bound to activated ABL kinase domain elucidates its inhibitory activity against imatinib-resistant ABL mutants, Cancer Res 66(11) (2006) 5790–7. [DOI] [PubMed] [Google Scholar]
  • [153].Muckelbauer J, Sack JS, Ahmed N, Burke J, Chang CY, Gao M, Tino J, Xie D, Tebben AJ, X-ray crystal structure of bone marrow kinase in the x chromosome: a Tec family kinase, Chem Biol Drug Des 78(5) (2011) 739–48. [DOI] [PubMed] [Google Scholar]
  • [154].Marcotte DJ, Liu YT, Arduini RM, Hession CA, Miatkowski K, Wildes CP, Cullen PF, Hong V, Hopkins BT, Mertsching E, Jenkins TJ, Romanowski MJ, Baker DP, Silvian LF, Structures of human Bruton’s tyrosine kinase in active and inactive conformations suggest a mechanism of activation for TEC family kinases, Protein Sci 19(3) (2010) 429–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Zeng H, Belanger DB, Curran PJ, Shipps GW Jr., Miao H, Bracken JB, Arshad Siddiqui M, Malkowski M, Wang Y, Discovery of novel imidazo[1,2-a]pyrazin-8-amines as Brk/PTK6 inhibitors, Bioorg Med Chem Lett 21(19) (2011) 5870–5. [DOI] [PubMed] [Google Scholar]
  • [156].Shim HJ, Yang HR, Kim HI, Kang SA, No KT, Jung YH, Lee ST, Discovery of (E)-5-(benzylideneamino)-1H-benzo[d]imidazol-2(3H)-one derivatives as inhibitors for PTK6, Bioorg Med Chem Lett 24(19) (2014) 4659–4663. [DOI] [PubMed] [Google Scholar]
  • [157].Shim HJ, Kim HI, Lee ST, The associated pyrazolopyrimidines PP1 and PP2 inhibit protein tyrosine kinase 6 activity and suppress breast cancer cell proliferation, Oncol Lett 13(3) (2017) 1463–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Park JH, Darvin P, Lim EJ, Joung YH, Hong DY, Park EU, Park SH, Choi SK, Moon ES, Cho BW, Park KD, Lee HK, Kim MJ, Park DS, Chung IM, Yang YM, Hwanggeumchal sorghum induces cell cycle arrest, and suppresses tumor growth and metastasis through Jak2/STAT pathways in breast cancer xenografts, PLoS ONE 7(7) (2012) e40531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Mahmoud KA, Krug M, Wersig T, Slynko I, Schachtele C, Totzke F, Sippl W, Hilgeroth A, Discovery of 4-anilino alpha-carbolines as novel Brk inhibitors, Bioorg Med Chem Lett 24(8) (2014) 1948–51. [DOI] [PubMed] [Google Scholar]
  • [160].Oelze M, Mahmoud KA, Sippl W, Wersig T, Hilgeroth A, Ritter CA, Novel 4-anilino-alpha-carboline derivatives induce cell death in nonadhesive breast cancer cells through inhibition of Brk activity, Int J Clin Pharmacol Ther 53(12) (2015) 1052–5. [DOI] [PubMed] [Google Scholar]
  • [161].Foudah AI, Jain S, Busnena BA, El Sayed KA, Optimization of marine triterpene sipholenols as inhibitors of breast cancer migration and invasion, ChemMedChem 8(3) (2013) 497–510. [DOI] [PubMed] [Google Scholar]
  • [162].Sallam AA, Mohyeldin MM, Foudah AI, Akl MR, Nazzal S, Meyer SA, Liu YY, El Sayed KA, Marine natural products-inspired phenylmethylene hydantoins with potent in vitro and in vivo antitumor activities via suppression of Brk and FAK signaling, Org Biomol Chem 12(28) (2014) 5295–303. [DOI] [PubMed] [Google Scholar]
  • [163].Elsayed HE, Akl MR, Ebrahim HY, Sallam AA, Haggag EG, Kamal AM, El Sayed KA, Discovery, optimization, and pharmacophore modeling of oleanolic acid and analogues as breast cancer cell migration and invasion inhibitors through targeting Brk/Paxillin/Rac1 axis, Chemical biology & drug design 85(2) (2015) 231–43. [DOI] [PubMed] [Google Scholar]
  • [164].Abe Y, Nagano M, Tada A, Adachi J, Tomonaga T, Deep Phosphotyrosine Proteomics by Optimization of Phosphotyrosine Enrichment and MS/MS Parameters, J Proteome Res 16(2) (2017) 1077–1086. [DOI] [PubMed] [Google Scholar]
  • [165].Stuhlmiller TJ, Miller SM, Zawistowski JS, Nakamura K, Beltran AS, Duncan JS, Angus SP, Collins KA, Granger DA, Reuther RA, Graves LM, Gomez SM, Kuan PF, Parker JS, Chen X, Sciaky N, Carey LA, Earp HS, Jin J, Johnson GL, Inhibition of Lapatinib-Induced Kinome Reprogramming in ERBB2-Positive Breast Cancer by Targeting BET Family Bromodomains, Cell Rep 11(3) (2015) 390–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Hunt SE, McLaren W, Gil L, Thormann A, Schuilenburg H, Sheppard D, Parton A, Armean IM, Trevanion SJ, Flicek P, Cunningham F, Ensembl variation resources, Database (Oxford) 2018 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Ren J, Wen L, Gao X, Jin C, Xue Y, Yao X, DOG 1.0: illustrator of protein domain structures, Cell research 19(2) (2009) 271–3. [DOI] [PubMed] [Google Scholar]
  • [168].Sievers F, Higgins DG, Clustal Omega for making accurate alignments of many protein sequences, Protein Sci 27(1) (2018) 135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Waterhouse AM, Procter JB, Martin DM, Clamp M, Barton GJ, Jalview Version 2--a multiple sequence alignment editor and analysis workbench, Bioinformatics 25(9) (2009) 1189–91. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES