Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 8.
Published in final edited form as: Cancer Lett. 2010 Dec 8;298(2):139–149. doi: 10.1016/j.canlet.2010.08.014

Targeting SRC in glioblastoma tumors and brain metastases: rationale and preclinical studies

Manmeet Ahluwalia 1, John de Groot 2, Wei (Michael) Liu 3, Candece L Gladson 4,
PMCID: PMC3212431  NIHMSID: NIHMS245529  PMID: 20947248

Abstract

Glioblastoma (GBM) is an extremely aggressive, infiltrative tumor with a poor prognosis. The regulatory approval of bevacizumab for recurrent GBM has confirmed that molecularly targeted agents have potential for GBM treatment. Preclinical data showing that SRC and SRC-family kinases (SFKs) mediate intracellular signaling pathways controlling key biologic/oncogenic processes provide a strong rationale for investigating SRC/SFK inhibitors, eg, dasatinib, in GBM and clinical studies are underway. The activity of these agents against solid tumors suggests that they may also be useful in treating brain metastases. This article reviews the potential for using SRC/SFK inhibitors to treat GBM and brain metastases. Word count =99/100

Indexing/key words: SRC family kinase, targeted therapy, glioma, glioblastoma, metastasis

1. Introduction

WHO grade III gliomas (anaplastic astrocytoma, anaplastic oligodendroglioma and anaplastic oligoastrocytoma) and grade IV gliomas (glioblastoma or GBM and gliosarcoma) are collectively referred to as malignant gliomas. GBM is the most common primary CNS tumor, accounting for 50% of the 17000 primary brain tumors diagnosed annually in the US [[1, 2]]. GBM occurs in all age groups with two discrete peaks of incidence in patients aged 0-8 years [[3]] and 45-70 years [[4]]. GBMs are infiltrative tumors, able to migrate to and invade areas of normal brain, and regardless of treatment, GBM almost always recurs, highlighting the need for novel therapies to treat this tumor.

Histologically, GBM is characterized by hypercellularity, atypical nuclei, increased proliferation, increased microvascularity and dilated vessels (angiogenesis), invasion of adjacent brain, and evidence of necrosis (particularly in large tumors that likely have hypoxic cores) [[1]]. Until recently, GBMs were thought to be derived from astrocytes based on their frequent expression of glial fibrillary acidic protein (GFAP), an intermediate filament and marker of astrocytes; however, GBMs have been shown to contain a proliferative tumorinitiating population of cells termed glioma stem cells [[5, 6]].

GBMs are extremely aggressive tumors and are associated with a dismal prognosis. Untreated patients typically die within 3 months and progression usually occurs within 6-9 months from initial diagnosis, even among those who receive the most current treatment. Approximately 25% of patients survive for 2 years and fewer than 10% survive for 5 years [[1]]. The current standard of care for newly diagnosed GBM is surgical resection, followed by concomitant radiotherapy and temozolomide (TMZ) chemotherapy for 6 weeks, then adjuvant TMZ for 6-12 months. The efficacy of this approach was demonstrated in a phase III trial (n = 573) conducted by the European Organisation for the Research and Treatment of Cancer and the National Cancer Institute of Canada [[7]]. Patients who received concurrent radiotherapy and TMZ followed by adjuvant TMZ had a higher median survival than those who received radiotherapy alone (14.6 and 12.1 months, respectively). The 2-year overall survival (OS) rate was more than double with combination therapy compared with radiotherapy alone (26.5% and 10.4%, respectively).

Recently, single-agent bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF) with antiangiogenic activity, was approved by the FDA for treatment of recurrent GBM. Bevacizumab with or without irinotecan was well tolerated and increased the estimated historical 6-month progression-free survival (PFS) rate from 15% to 43% and 50%, respectively [[8, 9]]. To date, bevacizumab has not been shown to improve OS. Two ongoing randomized phase III studies in patients with newly diagnosed GBM are investigating the efficacy of first-line bevacizumab combined with TMZ and radiotherapy (NCT00884741; NCT00943826) (see ClinicalTrials.gov). Even if ongoing trials demonstrate prolonged OS with bevacizumab, novel therapies are still needed to inhibit GBM invasion, extend duration of treatment response, or provide additional options to prevent disease recurrence.

Although bevacizumab prolonged PFS in patients with recurrent GBM, resistance to antiangiogenic (or anti-VEGF) therapy is expected. Two main mechanisms of resistance to anti-VEGF therapy are thought to exist: (i) during prolonged VEGF sequestration, tumor vasculature continues to grow via VEGF-independent neoangiogenesis (e.g., via basic fibroblast growth factor release); and (ii) glioma cells co-opt the host vasculature to invade normal brain without promoting angiogenesis. Preclinical models suggest that anti-VEGF therapy can induce a previously noninvasive glioma tumor to invade normal brain [[10-12]]. In these examples, bevacizumab does not block tumor invasion but rather promotes it underscoring the need for combination therapies that block GBM invasion.

Although GBM is the most common primary CNS tumor, metastatic tumors to the CNS also have a poor prognosis and limited treatment options. Brain metastases are a frequent complication of many solid tumor types, particularly breast cancer, lung cancer, and melanoma [[13]]. Although brain metastases have diverse tumors of origin, standard treatment involves surgical resection of accessible tumors and/or whole-brain radiotherapy, which can be combined with stereotactic radiosurgery [[14]]. Because whole-brain irradiation can negatively affect normal brain function, including cognition, additional therapeutic options are needed.

Further work is needed to develop novel targeted agents for patients with CNS tumors. Specifically, there is a critical need to develop therapies that target invading GBM cells. In this review, evidence supporting the evaluation of agents targeting SRC kinase or SRC-family kinase (SFK) members in GBM is discussed. The potential for using SRC inhibitors in the treatment of brain metastases is also considered.

2. SRC and SFKs as anticancer targets

Targeted therapies block growth, invasion or progression of tumor cells by interfering with signaling molecules or pathways important for the malignant phenotype. Identification of proteins with GBM-specific upregulation/activation or defects in expression may reveal potential novel targets. Based on this hypothesis, phase I and II trials are underway to investigate targeted agents that inhibit a range of targets, including inhibitors of integrins (cilengitide), tenascin (131I-81C6), phosphatidylinositol-3-kinase (PI3K; XL765), tyrosine kinases (sorafenib), and matriolytic enzymes (prinomostat) [[15]].

Accumulating data demonstrate that SRC kinase and SFKs are promising targets for anticancer therapy. The viral homolog of the SRC gene, v-SRC, was discovered as an extension of studies to identify a transmissible noncellular agent that promoted formation of chicken sarcomas [[16]]. The Rous sarcoma virus, which encoded v-SRC was responsible for malignant transformation [[17]]. Subsequent research found a gene in normal avian DNA closely related to v-SRC that was named cellular Src (represented as c-Src in early publications), which was the first proto-oncogene to be identified [[18]]. Structurally, v-SRC protein lacks the negative regulatory C-terminal domain of SRC and has several point mutations, resulting in constitutive activation of v-SRC and the potential to induce malignant transformation [[19-21]].

SFKs are a group of homologous nonreceptor tyrosine kinases with highly conserved structures consisting of four SRC-homology domains (SH1-4), a tyrosine kinase domain, and a C-terminal negative regulatory domain [[22]]. SFKs mediate intracellular signaling from multiple cell surface proteins, including growth factor receptors, integrins, and the EPH family of kinases. SFKs also bind to key intracellular signaling proteins, notably focal adhesion kinase (FAK) and the related proline-rich protein tyrosine kinase (PYK2/PTK2B) [[23, 24]]. Through these interactions and others, SFKs perform essential roles in regulating normal cellular processes, including cytoskeletal organization and remodeling that accompanies cell adhesion, motility, and cell division.

3. Activation and regulation of SRC

Much of what has been learned regarding SFK activation and function was derived from studies of c-SRC protein. SRC is maintained in an inactive state by C-terminal SRC kinase (CSK)-mediated phosphorylation of a negative-regulatory tyrosine residue (Y530). When phosphorylated, Y530 binds to the SH2 domain, folding SRC into an inactive configuration [[22]]. Negative regulation of SRC by CSK can have powerful antimetastatic and antiinvasive effects, as demonstrated by CSK overexpression in metastatic mouse colon carcinoma cell lines [[25]]. For SRC activation, the Y530 residue is dephosphorylated by protein tyrosine phosphatase-α [[26]], allowing the SRC structure to unfold into an active configuration that permits access of substrates to the kinase domain [[22]]. SRC can also be activated by direct binding of its SH2 and SH3 domains to intracellular proteins (e.g., FAK, the adaptor molecule p130CAS, or the actin filament-associated protein AFAP-110), or to activated tyrosine kinase growth factor receptors (e.g., epidermal growth factor receptor [EGFR] family members EGFR1 and human epidermal growth factor receptor-2 [HER2], platelet-derived growth factor receptor [PDGFR], VEGF receptor [VEGFR], and fibroblast growth factor receptor). Interaction between SRC and these proteins disrupts intramolecular inhibitory interactions within SRC, resulting in its activation [[21, 27, 28]].

Cell adhesion promotes association of FAK with SFKs [[29]]. FAK was first identified as a SRC-associated tyrosine-phosphorylated protein in v-SRC-transformed cells [[30]]. FAK colocalizes with integrins at focal adhesions and is rapidly activated by autophosphorylation during integrin-mediated cell adhesion. The autophosphorylation site (Y397) and two proline-rich regions in FAK provide high-affinity binding sites for the SRC SH2 and SH3 domains, respectively [[31]]. Once bound, SRC phosphorylates two tyrosine residues in the FAK kinase domain to increase kinase activity. SRC also phosphorylates tyrosine residues in the C-terminal half of FAK that act as docking sites for other signaling molecules [[32]]. The absence of FAK or SRC impairs the function of the partner molecule. For example, fibronectin-induced cell migration is significantly reduced in cells lacking FAK, corresponding with reduced SRC activation [[33]]. Similarly, fibronectin-induced migration and FAK activation are reduced in cells with triple null mutations of c-SRC, YES, and FYN [[34]]. These findings show the importance of SFK-FAK interactions for normal intracellular signaling in response to extracellular matrix (ECM)-dependent stimulation.

4. Expression and function of individual SFKs within the CNS

Nine SFKs have been identified, eight of which are expressed in humans. Several SFKs have a distinct tissue distribution pattern. Those expressed in neural tissue are LYN, FYN, YRK (expressed in chickens only), LCK [[35]], and the ubiquitous SFKs c-SRC and YES [[23, 36]]. LYN appears to have a functional role in both embryonic and adult brain. In embryonic brain, oligodendrocyte progenitor proliferation requires LYN activation by integrin αvβ3 [[37]], LYN activity is also detected in normal adult brain [[38]]. Examination of LYN mRNA distribution during mouse brain development using in situ hybridization showed lower LYN expression in embryonic mouse brain relative to adult mouse brain, in which high levels of mRNA were detected in discrete regions [[39]]. LYN also plays an antiapoptotic role in the brain, protecting neurons from glutamate-related neurotoxicity through integrin signaling [[40, 41]]. FYN plays a role in brain development, demonstrated by studies showing that expression of FYN is necessary for oligodendrocyte maturation [[37, 42]] and synapse formation [[43]]. FYN is involved in promoting myelination [[44]] and may also play a role in Alzheimer's disease progression by disrupting tau protein-mediated microtubule stabilization [[45]]. In mouse studies, LCK expression has been detected in distinct brain regions including the hippocampus and cerebellum [[35]]. The above data suggest that individual SFKs likely have distinct functions within the CNS. This may depend in part on individual SFK expression levels and on whether there is co-temporal expression of proteins that activate individual SFK members (such as integrins or growth factor receptors).

5. Increased SRC activity in glioblastoma

Investigators have found elevated SRC activity in GBM compared with normal brain samples using both Western blotting for phosphorylated SRC and SRC kinase activity assays [[38]]. The increased SFK activity in GBM samples is largely (>90%) due to elevated LYN activity [[38]]. More recently, investigators have shown that SRC activity/phosphorylation was elevated in GBM samples and cancer cell lines, including T98G and U87, compared with normal tissue [[46]]. The increased SRC activity in GBM is not due to amplification or mutation of SFK genes, because the Cancer Genome Atlas Research Network reported no change in SRC mRNA expression, gene amplification, or mutation in GBM tumors [[47]]. This is consistent with several reports studying colorectal and rectal cancer, where no activating mutations, deletions or amplifications of SRC were found [[48-50]]. In a very small subset of colon cancers an activating mutation of c-SRC at codon 531 has been described; however, the presence of this mutation in the population studied (a Chinese population) did not correlate with an increased risk of developing colon cancer [[51, 52]]. Also, in certain leukemias there may be loss of SRC due to deletion of chromosome 20q [[53, 54]]. Thus, the increased SRC activity in GBMs is likely due to increased activation of cell surface growth factor receptors and integrins that activate SFKs.

6. Mechanisms of action of SFKs in tumor biology: evidence of a role for SRC in glioblastoma

Tumor growth is typically associated with one or more of the following: increased proliferation, reduced apoptosis, inhibition of autophagy, and increased angiogenesis. Also, invasion of primary tumors and metastasis involves altered cell adhesion processes, increased motility and increased protease expression. In addition, metastasis requires intravasation/extravasation of tumor cells and colonization of a site distant to the primary tumor [[55]]. Preclinical studies of cancer cells, including GBM cell lines, have shown that dysregulated SFK signaling plays a role in the above processes [[56-61]]. The role for SRC in GBM tumor development is supported by a study in transgenic mice expressing v-SRC. These mice spontaneously developed low-grade tumors that progressed to a morphology resembling human GBM (14% of mice at 65 weeks) [[62]].

6.1. Proliferation

SRC plays an important role in promoting tumor cell proliferation, as demonstrated by several breast cancer, prostate cancer, and GBM tumor models in which blockade of SRC activity inhibited proliferation [[63-67]]. In tumors, SRC mediates signaling from extracellular proteins known to promote proliferation, including growth factor receptors [[68, 69]] and integrins [[70]]. SRC activation leads to downstream signaling through the RAS/mitogen-activated protein kinase (MAPK) and PI3K pathways, which have known roles in promoting tumor proliferation, survival, and invasion (Fig. 1) [[71],[72, 73]]. In addition, v-SRC has been shown to stimulate proliferation by inducing cyclin expression [[74]]. SRC/FAK complexes can also activate RAS, as shown by promotion of anchorage-independent proliferation of FAK-overexpressing GBM cells and by the growth of GBM xenografts in the mouse brain [[75]]. This proliferation appears to be mediated by a combination of FAK-induced KLF8 expression, cyclin D1 activation, and reduced expression of the cyclin-dependent kinase inhibitor p27 [[76]]. These data suggest that SRC-induced effects may involve FAK and that inhibition of SRC/FAK signaling may be effective in inhibiting tumor growth.

Fig. 1.

Fig. 1

Contribution of SRC-mediated pathways to tumor progression. In tumor cells, interaction of SRC-family kinases (SFKs) with receptor tyrosine kinases (RTK), integrins, or focal adhesion kinase (FAK) activates the kinase activity of SFKs. SFKs activate downstream signaling pathways that mediate proliferation (green), angiogenesis (red), survival (blue), and motility/migration (orange). Cyclin D1 can be activated by v-SRC. RTK, receptor tyrosine kinase; FAK, focal adhesion kinase; MAPK, mitogen activated protein kinase; VEGF, vascular endothelial growth factor; JAK, Janus kinase; STAT, signal transducers and activators of transcription; IL, interleukin; PI3K, phosphatidylinositol 3-kinase; NFKB, nuclear factor kappa B; IKK, inhibitor of kappa-B kinase; Shc, SRC transforming protein; GRB2, growth factor receptor-bound protein 2; SOS, son-of-sevenless; MEK, mitogen-activated protein kinase kinase; ERK, extracellular signal-regulated kinase; MLCK, myosin light chain kinase; CAS, Crk-associated substrate; JNK, Jun N-terminal Kinase; RhoGAP, Rho GTPase activating protein.

6.2. Apoptosis

Anoikis is a form of programmed cell death induced by detachment of adherent cells from the ECM [[77]]. For solid tumor cells to detach from the primary tumor and metastasize to a distant site, they must acquire anoikis resistance. There is evidence that several SFKs (c-SRC, LYN, FYN, YES, and LCK) activate antiapoptotic or prosurvival signal transduction pathways, including those mediated by PI3K and AKT signaling [[78-80]]. Dasatinib, a potent SFK inhibitor, induces apoptosis of certain adherent cancer cells, such as bone sarcoma cells, potentially by inhibiting the FAK/p130CAS pro-survival pathway [[81]]. Although further preclinical studies are needed to clarify the role of SFKs in anoikis and apoptosis resistance, it is feasible that SFK inhibitors may potentially promote anoikis or apoptosis, thereby protecting against metastasis.

6.3. Autophagy

Recently, Milano and coworkers [[82]] showed that dasatinib inhibited in vitro GBM cell growth by inducing autophagic cell death. This was enhanced in cells expressing functional phosphatase and tensin homolog deleted on chromosome 10 (PTEN) gene and when dasatinib was combined with TMZ. Combining dasatinib with carboplatin or irinotecan (agents thought to be pro-apoptotic) did not significantly increase autophagic cell death in most GBM cell lines [[82]]. Because autophagic cell death results in caspase activation, this type of cell death may have contributed to inhibition of cell growth or induction of apoptosis observed in earlier studies of SFK inhibitors in cancer cells.

6.4. Altered cell adhesion and motility/migration

During solid tumor development, reduced cell-cell adhesion and increased turnover of cell–ECM contacts can contribute to tumor cell migration and metastasis. SFK signaling is necessary for cell adhesion, but SFKs can also disrupt cell-cell adhesion, thereby promoting tumor cell invasion. SRC-mediated decreases in cellular adhesiveness result from multiple mechanisms, including redistribution of focal adhesion component proteins [[83, 84]], altered regulation of integrin signaling, and decreased deposition of ECM proteins [[85, 86]]. For example, in epidermoid squamous carcinomas, FYN activation was essential for EGF-dependent hemidesmosome disassembly and tumor invasion [[87]]. Hemidesmosome formation required integrin α6β4. In U87 GBM cells stimulated with PDGF-BB, LYN and FYN were differentially activated and had different intracellular associations with integrins. LYN, but not FYN, was specifically activated when integrin αvβ3 was engaged and was required for cell migration on vitronectin [[88]]. Thus, context-dependent growth factor receptor signaling (dependent on the integrin engaged and the SFK expressed) can determine whether there is cell-ECM or cell-cell adhesion.

In tumor cell models, including GBM, treatment with a SRC/SFK inhibitor or short interfering RNA blocked tumor cell invasion and migration [[46, 59, 67, 82, 88-90]]. A recent study has examined the mechanisms behind GBM cell migration induced by mutation or loss of PTEN protein, which occurs in 60-70% of GBMs. Stable overexpression of CSK or treatment with the laboratory SFK inhibitor PP1 inhibited vitronectin-stimulated transwell migration of U87MG and U373MG GBM cells. FYN was specifically shown to promote vitronectin-stimulated migration induced by loss of PTEN expression [[91]]. Interestingly, in a separate study, human GBM cells with functional PTEN showed increased sensitivity to growth inhibition by dasatinib compared with cells with low PTEN expression levels [[82]]. Inhibition of in vitro invasion was also observed.

6.5. Intravasation, extravasation, and invasion

GBM tumors rarely metastasize outside of the CNS, possibly due to a combination of the blood-brain barrier's (BBB) resistance to intravasation and a lack of suitable cell adhesion molecules on the surface of GBM cells to facilitate homing to target tissues [[92]]. However, GBM tumors are characterized by diffuse local invasion, which involves processes shared with metastasis, including increased motility, altered adhesion, and remodeling of ECM. Invasive tumor cells are probably one of the major factors in GBM recurrence following surgery, radiotherapy, and chemotherapy.

The importance of SFKs in local invasion has been demonstrated in mouse intracerebral xenograft models. GBM invasion was significantly decreased in SRC knockout mice compared with control mice, suggesting that SRC expression by normal brain tissue is required for invasion/infiltration of GBM cells [[61]]. It has been suggested that VEGF may increase vascular permeability in normal mice by increasing SRC activity; therefore VEGF inhibitors should reduce invasiveness. However, VEGF inhibitors have been shown to cause changes in tumors that are associated with increased infiltrative activity [[93]]. This indicates that direct inhibition of SRC activity may be a more direct method of blocking GBM invasion than VEGF inhibition. In a primary human GBM xenograft model, increased SRC- and PI3 kinase-dependent invasiveness in response to bevacizumab has been observed, which was prevented by dasatinib [[94]]. SRC inhibition may also inhibit invasion by disrupting crosstalk between SRC and FAK, because FAK activation promotes GBM invasion through downstream signaling molecules such as human enhancer of filamentation 1 (HEF1), a p130CAS family member [[95]].

In vitro studies have shown that SRC regulates formation of invadopodia -specialized actin-rich membrane protrusions that degrade ECM [[63, 96, 97]]. Laboratory SFK inhibitors PP2 and SU6656, dominant-negative SRC, and CSK all significantly inhibit GBM cell invasion in vitro, with a corresponding loss of actin in invadopodia [[98]].

Matrix metallo-proteases (MMPs) have a well-established role in tumor invasion [[99]]. SRC-dependent expression of membrane-bound and secreted MMPs has been demonstrated in models of cancer cell invasion including GBM tumors [[60, 100-102]]. In T98G and NCH125 GBM cells stimulated by CD95 activation, YES signaling mediated upregulated MMP expression and increased invasion [[58]].

Intravasation and extravasation enable systemic cancers to metastasize to the CNS. SRC-mediated extravasation may be particularly important for tumors that metastasize to the brain. In vitro studies of gastric and colorectal cancer cells have shown that SRC signaling is involved in intravasation through its role in the upregulated expression of chemokine receptors (CXCR1 and CXCR 2) and the urokinase receptor [[103, 104]]. SRC activity can also affect extravasation through its downstream role in VEGF signaling pathways. Disruption of vascular endothelial cell-cell junctions following VEGF-activated SRC signaling increases vascular permeability, permitting tumor cell extravasation [[105, 106]]. At the BBB, angiogenesis and tight junction formation are regulated by SRC-suppressed C-kinase substrate (SSeCKS), a protein that decreases expression of VEGF and increases expression of angiopoietin-1, an antipermeability factor [[107]]. Decreased SSeCKS activity caused by increased SRC activity could potentially enhance vascular permeability facilitating metastasis to the brain.

6.6. Angiogenesis

Angiogenesis is critical for establishment and growth of solid tumors and is a key feature of GBM. As discussed previously, the antiangiogenic agent bevacizumab increases short-term PFS in patients with recurrent GBM. Because SRC can induce VEGF expression, SFKs have a potential role in the angiogenic process [[108]]. This is supported by studies of GBM-like tumors that develop in transgenic v-SRC-expressing mice, in which expression of VEGF and other proangiogenic factors was observed at early stages of tumor development [[109]]. In addition, in human solid tumor cell lines, SRC inhibition reduces VEGF expression, thereby decreasing proangiogenic activity [[110]].

Because of their role in mediating VEGF receptor signaling, c-SRC and YES are required for VEGF-induced angiogenesis seen in mouse studies [[111]]. The potential additive effects of combining SFK inhibitors with antiangiogenic agents are being explored in clinical trials.

6.7. SFKs may be activated by irradiation

An in vitro study in PTEN-mutated glioma cells found that exposure to ionizing radiation activated SRC, in turn activating EGFR, p38 MAP kinase/AKT, and PI3K/AKT signaling pathways, and increasing MMP2 expression and cell invasiveness [[60]]. Therefore, SRC inhibitors should inhibit SRC-dependent increases in invasion following radiotherapy. In preclinical studies, saracatinib, an inhibitor of SFKs, BCR-ABL, KIT, PDGFRα/β, and VEGFR2 kinases, increased radiosensitivity of lung cancer cells [[112, 113]]. In addition, SU6656 in combination with radiotherapy decreased human umbilical vein endothelial cell survival in vitro compared with radiation alone. These observations were corroborated by an in vivo study showing that SU6656 administered prior to irradiation significantly enhanced radiation-induced destruction of tumor-associated blood vessels, and delayed tumor growth of Lewis lung carcinoma models [[114]].

7. SRC activation in brain metastases from solid tumors

Brain metastases are a frequent complication of solid tumors and may arise several years after removal of the primary tumor. As discussed, SRC has a key role in metastasis, suggesting that increased SRC activity could increase the likelihood of brain metastasis. SRC inhibitors may potentially prevent implantation of cancer cells from outside the brain or prevent growth of dormant brain metastases arising from primary tumors with elevated SFK activity.

Breast cancer is one of the most common causes of brain metastases. A study of gene expression signatures found increased expression of components of the SRC signaling pathway in 60% of breast cancer metastases to bone and in 25% of breast cancer metastases to the brain [[115]]. Patients with HER2-positive breast cancer have an increased risk of brain metastases [[116, 117]]. Although no direct role for SRC in brain targeting by HER2-positive metastatic breast cancer cells has been demonstrated, HER2 could potentially promote metastasis through direct phosphorylation of SRC [[118]] or other processes involving SRC, such as disruption of vascular endothelial integrity [[119]] or upregulation of urokinase-type plasminogen activator receptor, a proinvasive factor [[120, 121]].

Studies of melanoma and melanocyte cell lines have shown that neurotrophin and nerve growth factor (NGF) signaling elevated protein levels and activity of YES, with no effect on SRC activity. The degree of YES activation correlated with the invasive potential of melanoma cell lines, suggesting that upregulated YES expression or activity may contribute to the brain-metastatic phenotype [[122]].

In addition to a potential role in metastasis to brain, the antiproliferative activity of SRC inhibitors against primary tumors, including breast cancer, lung cancer, and melanoma, suggests that these agents may also have some activity against brain metastases derived from these tumors [[123-125]].

8. Preclinical studies of SRC inhibitors in glioblastoma

Dasatinib

Dasatinib is a potent inhibitor of c-SRC and other SFKs such as LYN and FYN, and also has activity against BCR-ABL, KIT, PDGFRα/β, and EPHA2 [[126, 127]]. The effects of dasatinib on GBM have been investigated in several preclinical studies. In a recent study, dasatinib decreased viability in 13/22 GBM cell lines (effector concentration for halfmaximum response (EC50) of <1 μM). Dasatinib also induced apoptosis in three GBM lines and inhibited cell migration (EC50 <500 nM) in 14/22 GBM cell lines. Site-directed mutation of potential kinase targets for dasatinib confirmed that SRC inhibition mediated the effects of dasatinib in GBM cells [[46]]. In addition, dasatinib was more effective in blocking in vitro glioma cell proliferation combined with TMZ [[82]].

PP2

Inhibition of proliferation by PP2, a laboratory SFK inhibitor, has been demonstrated in the U373MG human GBM cell line [[128]]. PP2 also inhibited GBM cell migration stimulated by phorbol 12-myristate 13-acetate-induced cytoskeletal reorganization, and decreased formation of CAS/CRK/RAC complexes that associate with focal adhesions to promote cell migration [[59]].

SU6656

SU6656 is a small-molecule inhibitor of SRC, FYN, YES, and LYN that inhibits PDGF-stimulated DNA synthesis and cell growth in U251 glioma cells and invasion of glioma cell line spheroids implanted in collagen type I matrices [[98]]. Effects on invasion appeared to be mediated by a rapid change in actin dynamics, similar to effects seen in PP2-treated cells. SU6656 also enhanced in vivo radiation-induced apoptosis and destruction of vascular endothelium, suggesting potential use of SFK inhibitors as radiosensitizers [[129]].

Bosutinib and saracatinib

Although other SFK inhibitors, including bosutinib (SKI-606) [[130]] and saracatinib (AZD0530) [[131]] have been investigated in solid tumors, data have not been published using GBM cells or in vivo models.

9. Potential for resistance to SRC inhibitors

Using a targeted therapy to inhibit a heterogenous population of tumor cells applies a selective pressure that could lead to development of treatment resistance. Although studies have not been performed in GBM, a recently published breast cancer xenograft study reported that estrogen receptor-positive tumors developed resistance to saracatinib monotherapy by activating alternative signaling pathways, including MEK and PI3K/AKT/mammalian target of rapamycin signaling [[132]]. In this study, resistance was overcome by combining saracatinib with the aromatase inhibitor letrozole, thus providing another reason to investigate combination therapy with SRC inhibitors and other classes of drugs.

In addition to potently inhibiting SFKs, dasatinib also inhibits BCR-ABL, a chimeric kinase formed during the development of chronic myeloid leukemia (CML). During dasatinib treatment of patients with CML, BCR-ABL point mutations have been associated with dasatinib resistance [[133]] and specific mutations detected in clinical samples from resistant patients correlated with preclinical mutagenesis studies [[134]]. The potential of BCR-ABL to develop resistance mutations is thought to be due predominantly to genomic instability resulting from BCR-ABL kinase activity [[135]]. No preclinical studies have been performed to determine if SFKs have the potential to develop mutations during SRC inhibitor treatment, although this would be an interesting area for future investigation.

10. Clinical trials of SRC inhibitors in glioblastoma

Despite decades of research, no therapy has been shown to block the invasive tumor phenotype that makes glioblastoma so difficult to treat. The preclinical data discussed above provide strong support for the hypothesis that SRC/SFK inhibition may be a promising strategy to inhibit GBM invasion into normal brain. Among the SRC inhibitors that are in clinical trials for solid tumors, only dasatinib is currently being assessed as a therapy for GBM.

Five ongoing trials are investigating dasatinib as a treatment for newly diagnosed and recurrent GBM in adults, either as a monotherapy or in combination with other agents/radiotherapy. In a current phase II clinical study, patients with recurrent GBM or gliosarcoma (NCT00423735) received dasatinib monotherapy and preliminary evidence of activity was reported in a small number of patients; further optimization of dasatinib dosing is ongoing via intra-patient dose escalation [[136]]. Additional studies are evaluating dasatinibbased combination therapy for recurrent GBM, including a phase I trial evaluating dasatinib plus the EGFR inhibitor erlotinib (NCT00609999) and a randomized phase II trial of lomustine alone versus lomustine plus dasatinib (NCT00948389). A phase I/II study in patients with newly diagnosed glioblastoma is evaluating the efficacy of dasatinib combined with radiotherapy and concomitant TMZ, followed by adjuvant treatment with dasatinib plus TMZ (NCT00895960). All of these studies are currently recruiting patients and have yet to publish data in peer-reviewed journals (Table 1).

Table 1. Ongoing clinical trials of dasatinib in glioblastoma.

ClinicalTrials.gov identifier Agent(s) Phase Disease stage Primary endpoints Estimated enrolment Sponsor
NCT00895960 Dasatinib + RT/TMZ 1/2 Newly diagnosed • Maximum tolerated dose 72 MDACC

NCT00609999 Dasatinib + erlotinib 1 Recurrent • Maximum tolerated dose 48 Duke University
• Dose-limiting toxicity

NCT00423735 Dasatinib 2 Recurrent • Progression-free survival at 6 months 113 RTOG

NCT00948389 Dasatinib + lomustine 2 Recurrent • Hematologic and nonhematologic toxicity 108 Bristol-Myers Squibb European Organization for Research and Treatment of Cancer
• Proportion of patients disease free by MRI

RT, radiotherapy; TMZ, temozolomide; MDACC, MD Anderson Cancer Center; NCCTG, North Central Cancer Treatment Group; RTOG, Radiation Therapy Oncology Group.

When investigating novel agents for treating CNS tumors, the issue of BBB penetration must be considered. A preclinical study showed that dasatinib crossed the BBB and reduced intracranial leukemia xenograft volume [[137]]. These investigators also reported clinical activity of dasatinib in 11 patients with CNS metastases of Philadelphia chromosome-positive leukemia, suggesting that dasatinib and perhaps other small-molecule SFK inhibitors can cross the BBB to exert a clinical effect.

11. Unanswered questions

In vitro data have shown that SFKs and FAK have a close relationship during several normal and oncogenic cellular activities, which may be of increased relevance in GBM. This raises the question of what happens to FAK activity when SFKs are inhibited, and whether the antitumor activity seen with SFK inhibitors is partly due to a decrease in FAK activity. Also, the effect of SFK inhibitors on the interactions between SFKs and the FAK family member PYK2 requires further investigation.

As discussed above, SFK activity is significantly elevated in de novo GBM tumors [[38]]; however, it is not known if SFK activity is elevated in secondary GBM tumors arising from lower-grade gliomas. This is an important consideration because different genetic alterations and tyrosine kinase activities are detected in these two types of GBM. The development of molecular probes to quantify different SFK activities in complex tissue sections would be beneficial to further studies in this area.

Finally, the concept that anti-VEGF therapy-induced invasion may be blocked by SRC inhibition raises several unanswered questions. First, it is not known if increased invasion during antiangiogenic therapy is due to overexpression or activation of SRC/SFKs. Secondly, if SRC/SFKs mediate this invasive phenotype, it is not known if delivery of small molecule targeted agents will be reduced following the reduction in vascular permeability seen with antiangiogenic therapy. Ongoing preclinical and clinical studies will attempt to answer these important questions.

12. Conclusions

Because of the key role of SFKs in tumor development and oncogenic signal transduction, SFKs are clearly a rational target for GBM treatment. This hypothesis is supported by preclinical evidence demonstrating that increased SRC or SFK activity is associated with GBM proliferation and invasion. Preclinical data show that SFK-targeting agents have single-agent activity and that combining dasatinib with TMZ has increased activity compared with TMZ alone [[82]]. In addition, preliminary evidence suggests that SFK inhibition may increase tumor cell sensitivity to radiation, which could have direct relevance for radiotherapy of both GBM and brain metastases. Preliminary results from clinical trials of dasatinib will provide the first indication of whether SFK inhibitors have the potential to become a new class of therapy for GBM. In addition to a possible role in GBM there is considerable evidence of the role of SFKs during metastasis. Therefore, solid tumors with aberrant SFK expression or activity may have an increased potential for migration into the brain from primary sites outside the CNS or growth of a previously undetectable dormant micrometastasis. This provides a further area of investigation for SFK inhibitors that is also relevant to CNS tumors.

Acknowledgments

The authors take full responsibility for the content of this publication, and confirm that it reflects their viewpoint and medical expertise. StemScientific, funded by Bristol-Myers Squibb, assisted with the writing and editing support. Bristol-Myers Squibb did not influence the content of the manuscript, nor did the authors receive financial compensation for authoring the manuscript.

Footnotes

Conflicts of interest: Dr. John de Groot was a paid consultant for Bristol-Myers Squibb last year, and that relationship has ended. None of the other authors have a financial or personal relationship with other people or organizations that could inappropriately influence this manuscript.

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 citable 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.

Contributor Information

Dr Manmeet Ahluwalia, Email: ahluwam@ccf.org, Cleveland Clinic Main Campus, Mail Code ND40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-444-6145.

Dr John de Groot, Email: jdegroot@mdanderson.org, The Brain Tumor Center, The University of Texas, M.D. Anderson Cancer Center, 1515, Holcombe Blvd., Unit 431, Houston, TX 77030, Phone: 713-792-7255.

Dr Wei (Michael) Liu, Email: liuw@ccf.org, Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9494.

Dr Candece L Gladson, Email: gladsoc@ccf.org, Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9493, Fax: 216-445-6269.

Reference List

  • 1.Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med. 2008;359:492–507. doi: 10.1056/NEJMra0708126. [DOI] [PubMed] [Google Scholar]
  • 2.Hess KR, Broglio KR, Bondy ML. Adult glioma incidence trends in the United States, 1977-2000. Cancer. 2004;101:2293–2299. doi: 10.1002/cncr.20621. [DOI] [PubMed] [Google Scholar]
  • 3.Pollack IF. Brain tumors in children. N Engl J Med. 1994;331:1500–1507. doi: 10.1056/NEJM199412013312207. [DOI] [PubMed] [Google Scholar]
  • 4.Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 2005;64:479–489. doi: 10.1093/jnen/64.6.479. [DOI] [PubMed] [Google Scholar]
  • 5.Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66:7843–7848. doi: 10.1158/0008-5472.CAN-06-1010. [DOI] [PubMed] [Google Scholar]
  • 6.Gilbertson RJ, Rich JN. Making a tumour's bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer. 2007;7:733–736. doi: 10.1038/nrc2246. [DOI] [PubMed] [Google Scholar]
  • 7.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
  • 8.Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA, Yung WK. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17:2572–2578. doi: 10.1200/JCO.1999.17.8.2572. [DOI] [PubMed] [Google Scholar]
  • 9.Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27:4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 10.Lucio-Eterovic AK, Piao Y, de Groot JF. Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res. 2009;15:4589–4599. doi: 10.1158/1078-0432.CCR-09-0575. [DOI] [PubMed] [Google Scholar]
  • 11.Rubenstein JL, Kim J, Ozawa T, Zhang M, Westphal M, Deen DF, Shuman MA. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2000;2:306–314. doi: 10.1038/sj.neo.7900102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lamszus K, Kunkel P, Westphal M. Invasion as limitation to anti-angiogenic glioma therapy. Acta Neurochir Suppl. 2003;88:169–177. doi: 10.1007/978-3-7091-6090-9_23. [DOI] [PubMed] [Google Scholar]
  • 13.Eichler AF, Loeffler JS. Multidisciplinary management of brain metastases. Oncologist. 2007;12:884–898. doi: 10.1634/theoncologist.12-7-884. [DOI] [PubMed] [Google Scholar]
  • 14.Richards GM, Khuntia D, Mehta MP. Therapeutic management of metastatic brain tumors. Crit Rev Oncol Hematol. 2007;61:70–78. doi: 10.1016/j.critrevonc.2006.06.012. [DOI] [PubMed] [Google Scholar]
  • 15.Gladson CL, Prayson RA, Liu WM. The Pathobiology of Glioma Tumors. Annu Rev Pathol. 2010;5:33–50. doi: 10.1146/annurev-pathol-121808-102109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rous P. A sarcoma of the fowl transmissible by an agent separable for the tumor cells. J Exp Med. 1911;13:397–411. doi: 10.1084/jem.13.4.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Martin GS. Rous sarcoma virus: a function required for the maintenance of the transformed state. Nature. 1970;227:1021–1023. doi: 10.1038/2271021a0. [DOI] [PubMed] [Google Scholar]
  • 18.Stehelin D, Varmus HE, Bishop JM, Vogt PK. DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature. 1976;260:170–173. doi: 10.1038/260170a0. [DOI] [PubMed] [Google Scholar]
  • 19.Jove R, Hanafusa H. Cell transformation by the viral src oncogene. Annu Rev Cell Biol. 1987;3:31–56. doi: 10.1146/annurev.cb.03.110187.000335. [DOI] [PubMed] [Google Scholar]
  • 20.Takeya T, Hanafusa H. DNA sequence of the viral and cellular src gene of chickens. II. Comparison of the src genes of two strains of avian sarcoma virus and of the cellular homolog. J Virol. 1982;44:12–18. doi: 10.1128/jvi.44.1.12-18.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yeatman TJ. A renaissance for SRC. Nat Rev Cancer. 2004;4:470–480. doi: 10.1038/nrc1366. [DOI] [PubMed] [Google Scholar]
  • 22.Ly QP, Yeatman TJ. Clinical relevance of targeted interference with Src-mediated signal transduction events. Recent Results Cancer Res. 2007;172:169–188. doi: 10.1007/978-3-540-31209-3_10. [DOI] [PubMed] [Google Scholar]
  • 23.Thomas SM, Brugge JS. Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol. 1997;13:513–609. doi: 10.1146/annurev.cellbio.13.1.513. [DOI] [PubMed] [Google Scholar]
  • 24.Girault JA, Costa A, Derkinderen P, Studler JM, Toutant M. FAK and PYK2/CAKbeta in the nervous system: a link between neuronal activity, plasticity and survival? Trends Neurosci. 1999;22:257–263. doi: 10.1016/s0166-2236(98)01358-7. [DOI] [PubMed] [Google Scholar]
  • 25.Nakagawa T, Tanaka S, Suzuki H, Takayanagi H, Miyazaki T, Nakamura K, Tsuruo T. Overexpression of the csk gene suppresses tumor metastasis in vivo. Int J Cancer. 2000;88:384–391. [PubMed] [Google Scholar]
  • 26.Egan C, Pang A, Durda D, Cheng HC, Wang JH, Fujita DJ. Activation of Src in human breast tumor cell lines: elevated levels of phosphotyrosine phosphatase activity that preferentially recognizes the Src carboxy terminal negative regulatory tyrosine 530. Oncogene. 1999;18:1227–1237. doi: 10.1038/sj.onc.1202233. [DOI] [PubMed] [Google Scholar]
  • 27.Playford MP, Schaller MD. The interplay between Src and integrins in normal and tumor biology. Oncogene. 2004;23:7928–7946. doi: 10.1038/sj.onc.1208080. [DOI] [PubMed] [Google Scholar]
  • 28.Gatesman A, Walker VG, Baisden JM, Weed SA, Flynn DC. Protein kinase Calpha activates c-Src and induces podosome formation via AFAP-110. Mol Cell Biol. 2004;24:7578–7597. doi: 10.1128/MCB.24.17.7578-7597.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Schlaepfer DD, Hunter T. Evidence for in vivo phosphorylation of the Grb2 SH2-domain binding site on focal adhesion kinase by Src-family protein-tyrosine kinases. Mol Cell Biol. 1996;16:5623–5633. doi: 10.1128/mcb.16.10.5623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kanner SB, Reynolds AB, Vines RR, Parsons JT. Monoclonal antibodies to individual tyrosine-phosphorylated protein substrates of oncogene-encoded tyrosine kinases. Proc Natl Acad Sci U S A. 1990;87:3328–3332. doi: 10.1073/pnas.87.9.3328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Thomas JW, Ellis B, Boerner RJ, Knight WB, White GC, Schaller MD. SH2- and SH3-mediated interactions between focal adhesion kinase and Src. J Biol Chem. 1998;273:577–583. doi: 10.1074/jbc.273.1.577. [DOI] [PubMed] [Google Scholar]
  • 32.Brunton VG, Frame MC. Src and focal adhesion kinase as therapeutic targets in cancer. Curr Opin Pharmacol. 2008;8:427–432. doi: 10.1016/j.coph.2008.06.012. [DOI] [PubMed] [Google Scholar]
  • 33.Sieg DJ, Hauck CR, Schlaepfer DD. Required role of focal adhesion kinase (FAK) for integrin-stimulated cell migration. J Cell Sci. 1999;112(Pt 16):2677–2691. doi: 10.1242/jcs.112.16.2677. [DOI] [PubMed] [Google Scholar]
  • 34.Klinghoffer RA, Sachsenmaier C, Cooper JA, Soriano P. Src family kinases are required for integrin but not PDGFR signal transduction. EMBO J. 1999;18:2459–2471. doi: 10.1093/emboj/18.9.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Omri B, Crisanti P, Marty MC, Alliot F, Fagard R, Molina T, Pessac B. The Lck tyrosine kinase is expressed in brain neurons. J Neurochem. 1996;67:1360–1364. doi: 10.1046/j.1471-4159.1996.67041360.x. [DOI] [PubMed] [Google Scholar]
  • 36.Jacobs C, Rubsamen H. Expression of pp60c-src protein kinase in adult and fetal human tissue: high activities in some sarcomas and mammary carcinomas. Cancer Res. 1983;43:1696–1702. [PubMed] [Google Scholar]
  • 37.Colognato H, Ramachandrappa S, Olsen IM, Ffrench-Constant C. Integrins direct Src family kinases to regulate distinct phases of oligodendrocyte development. J Cell Biol. 2004;167:365–375. doi: 10.1083/jcb.200404076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Stettner MR, Wang W, Nabors LB, Bharara S, Flynn DC, Grammer JR, Gillespie GY, Gladson CL. Lyn kinase activity is the predominant cellular SRC kinase activity in glioblastoma tumor cells. Cancer Res. 2005;65:5535–5543. doi: 10.1158/0008-5472.CAN-04-3688. [DOI] [PubMed] [Google Scholar]
  • 39.Umemori H, Wanaka A, Kato H, Takeuchi M, Tohyama M, Yamamoto T. Specific expressions of Fyn and Lyn, lymphocyte antigen receptor-associated tyrosine kinases, in the central nervous system. Brain Res Mol Brain Res. 1992;16:303–310. doi: 10.1016/0169-328x(92)90239-8. [DOI] [PubMed] [Google Scholar]
  • 40.Sharp F, Liu DZ, Zhan X, Ander BP. Intracerebral hemorrhage injury mechanisms: glutamate neurotoxicity, thrombin, and Src. Acta Neurochir Suppl. 2008;105:43–46. doi: 10.1007/978-3-211-09469-3_9. [DOI] [PubMed] [Google Scholar]
  • 41.Chudakova DA, Zeidan YH, Wheeler BW, Yu J, Novgorodov SA, Kindy MS, Hannun YA, Gudz TI. Integrin-associated Lyn kinase promotes cell survival by suppressing acid sphingomyelinase activity. J Biol Chem. 2008;283:28806–28816. doi: 10.1074/jbc.M803301200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wang PS, Wang J, Xiao ZC, Pallen CJ. Protein-tyrosine phosphatase alpha acts as an upstream regulator of Fyn signaling to promote oligodendrocyte differentiation and myelination. J Biol Chem. 2009;284:33692–33702. doi: 10.1074/jbc.M109.061770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lim SH, Kwon SK, Lee MK, Moon J, Jeong DG, Park E, Kim SJ, Park BC, Lee SC, Ryu SE, Yu DY, Chung BH, Kim E, Myung PK, Lee JR. Synapse formation regulated by protein tyrosine phosphatase receptor T through interaction with cell adhesion molecules and Fyn. EMBO J. 2009;28:3564–3578. doi: 10.1038/emboj.2009.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lu Z, Ku L, Chen Y, Feng Y. Developmental abnormalities of myelin basic protein expression in fyn knock-out brain reveal a role of Fyn in posttranscriptional regulation. J Biol Chem. 2005;280:389–395. doi: 10.1074/jbc.M405973200. [DOI] [PubMed] [Google Scholar]
  • 45.Lee G, Thangavel R, Sharma VM, Litersky JM, Bhaskar K, Fang SM, Do LH, Andreadis A, Van HG, Ksiezak-Reding H. Phosphorylation of tau by fyn: implications for Alzheimer's disease. J Neurosci. 2004;24:2304–2312. doi: 10.1523/JNEUROSCI.4162-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Du J, Bernasconi P, Clauser KR, Mani DR, Finn SP, Beroukhim R, Burns M, Julian B, Peng XP, Hieronymus H, Maglathlin RL, Lewis TA, Liau LM, Nghiemphu P, Mellinghoff IK, Louis DN, Loda M, Carr SA, Kung AL, Golub TR. Bead-based profiling of tyrosine kinase phosphorylation identifies SRC as a potential target for glioblastoma therapy. Nat Biotechnol. 2009;27:77–83. doi: 10.1038/nbt.1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.The Cancer Genome Atlas Research Network. Comprehensive gnomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Daigo Y, Furukawa Y, Kawasoe T, Ishiguro H, Fujita M, Sugai S, Nakamori S, Liefers GJ, Tollenaar RA, d van V, Nakamura Y. Absence of genetic alteration at codon 531 of the human c-src gene in 479 advanced colorectal cancers from Japanese and Caucasian patients. Cancer Res. 1999;59:4222–4224. [PubMed] [Google Scholar]
  • 49.Nilbert M, Fernebro E. Lack of activating c-SRC mutations at codon 531 in rectal cancer. Cancer Genet Cytogenet. 2000;121:94–95. doi: 10.1016/s0165-4608(00)00226-0. [DOI] [PubMed] [Google Scholar]
  • 50.Laghi L, Bianchi P, Orbetegli O, Gennari L, Roncalli M, Malesci A. Lack of mutation at codon 531 of SRC in advanced colorectal cancers from Italian patients. Br J Cancer. 2001;84:196–198. doi: 10.1054/bjoc.2000.1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Irby RB, Mao W, Coppola D, Kang J, Loubeau JM, Trudeau W, Karl R, Fujita DJ, Jove R, Yeatman TJ. Activating SRC mutation in a subset of advanced human colon cancers. Nat Genet. 1999;21:187–190. doi: 10.1038/5971. [DOI] [PubMed] [Google Scholar]
  • 52.Wang NM, Yeh KT, Tsai CH, Chen SJ, Chang JG. No evidence of correlation between mutation at codon 531 of src and the risk of colon cancer in Chinese. Cancer Lett. 2000;150:201–204. doi: 10.1016/s0304-3835(99)00398-5. [DOI] [PubMed] [Google Scholar]
  • 53.Le Beau MM, Westbrook CA, Diaz MO, Rowley JD. c-src is consistently conserved in the chromosomal deletion (20q) observed in myeloid disorders. Proc Natl Acad Sci U S A. 1985;82:6692–6696. doi: 10.1073/pnas.82.19.6692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Morris CM, Honeybone LM, Hollings PE, Fitzgerald PH. Localization of the SRC oncogene to chromosome band 20q11.2 and loss of this gene with deletion (20q) in two leukemic patients. Blood. 1989;74:1768–1773. [PubMed] [Google Scholar]
  • 55.Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563–572. doi: 10.1038/nrc865. [DOI] [PubMed] [Google Scholar]
  • 56.Beretta F, Bassani S, Binda E, Verpelli C, Bello L, Galli R, Passafaro M. The GluR2 subunit inhibits proliferation by inactivating Src-MAPK signalling and induces apoptosis by means of caspase 3/6-dependent activation in glioma cells. Eur J Neurosci. 2009;30:25–34. doi: 10.1111/j.1460-9568.2009.06804.x. [DOI] [PubMed] [Google Scholar]
  • 57.Oliveira-Ferrer L, Hauschild J, Fiedler W, Bokemeyer C, Nippgen J, Celik I, Schuch G. Cilengitide induces cellular detachment and apoptosis in endothelial and glioma cells mediated by inhibition of FAK/src/AKT pathway. J Exp Clin Cancer Res. 2008;27:86. doi: 10.1186/1756-9966-27-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kleber S, Sancho-Martinez I, Wiestler B, Beisel A, Gieffers C, Hill O, Thiemann M, Mueller W, Sykora J, Kuhn A, Schreglmann N, Letellier E, Zuliani C, Klussmann S, Teodorczyk M, Grone HJ, Ganten TM, Sultmann H, Tuttenberg J, von Deimling A, Regnier-Vigouroux A, Herold-Mende C, Martin-Villalba A. Yes and PI3K bind CD95 to signal invasion of glioblastoma. Cancer Cell. 2008;13:235–248. doi: 10.1016/j.ccr.2008.02.003. [DOI] [PubMed] [Google Scholar]
  • 59.Nomura N, Nomura M, Sugiyama K, Hamada J. Src regulates phorbol 12-myristate 13-acetate-activated PKC-induced migration via Cas/Crk/Rac1 signaling pathway in glioblastoma cells. Int J Mol Med. 2007;20:511–519. [PubMed] [Google Scholar]
  • 60.Park CM, Park MJ, Kwak HJ, Lee HC, Kim MS, Lee SH, Park IC, Rhee CH, Hong SI. Ionizing radiation enhances matrix metalloproteinase-2 secretion and invasion of glioma cells through Src/epidermal growth factor receptor-mediated p38/Akt and phosphatidylinositol 3-kinase/Akt signaling pathways. Cancer Res. 2006;66:8511–8519. doi: 10.1158/0008-5472.CAN-05-4340. [DOI] [PubMed] [Google Scholar]
  • 61.Lund CV, Nguyen MT, Owens GC, Pakchoian AJ, Shaterian A, Kruse CA, Eliceiri BP. Reduced glioma infiltration in Src-deficient mice. J Neurooncol. 2006;78:19–29. doi: 10.1007/s11060-005-9068-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Weissenberger J, Steinbach JP, Malin G, Spada S, Rulicke T, Aguzzi A. Development and malignant progression of astrocytomas in GFAP-v-src transgenic mice. Oncogene. 1997;14:2005–2013. doi: 10.1038/sj.onc.1201168. [DOI] [PubMed] [Google Scholar]
  • 63.Pichot CS, Hartig SM, Xia L, Arvanitis C, Monisvais D, Lee FY, Frost JA, Corey SJ. Dasatinib synergizes with doxorubicin to block growth, migration, and invasion of breast cancer cells. Br J Cancer. 2009;101:38–47. doi: 10.1038/sj.bjc.6605101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yamaguchi K, Kugimiya T, Miyazaki T. Substance P receptor in U373 MG human astrocytoma cells activates mitogen-activated protein kinases ERK1/2 through Src. Brain Tumor Pathol. 2005;22:1–8. doi: 10.1007/s10014-005-0178-1. [DOI] [PubMed] [Google Scholar]
  • 65.Chang YM, Bai L, Liu S, Yang JC, Kung HJ, Evans CP. Src family kinase oncogenic potential and pathways in prostate cancer as revealed by AZD0530. Oncogene. 2008;27:6365–6375. doi: 10.1038/onc.2008.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Nam S, Kim D, Cheng JQ, Zhang S, Lee JH, Buettner R, Mirosevich J, Lee F, Jove R. Dasatinib (BMS-354825), a novel multi-targeted kinase inhibitor that blocks tumor cell migration and invasion, is a promising therapeutic agent for metastatic prostate cancer. Proc Amer Assoc Cancer Res. 2006;47:3797. [Google Scholar]
  • 67.Jallal H, Valentino ML, Chen G, Boschelli F, Ali S, Rabbani SA. A Src/Abl kinase inhibitor, SKI-606, blocks breast cancer invasion, growth, and metastasis in vitro and in vivo. Cancer Res. 2007;67:1580–1588. doi: 10.1158/0008-5472.CAN-06-2027. [DOI] [PubMed] [Google Scholar]
  • 68.Amos S, Martin PM, Polar GA, Parsons SJ, Hussaini IM. Phorbol 12-myristate 13-acetate induces epidermal growth factor receptor transactivation via protein kinase Cdelta/c-Src pathways in glioblastoma cells. J Biol Chem. 2005;280:7729–7738. doi: 10.1074/jbc.M409056200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lin CC, Lee CW, Chu TH, Cheng CY, Luo SF, Hsiao LD, Yang CM. Transactivation of Src, PDGF receptor, and Akt is involved in IL-1 beta-induced ICAM-1 expression in A549 cells. J Cell Physiol. 2007;211:771–780. doi: 10.1002/jcp.20987. [DOI] [PubMed] [Google Scholar]
  • 70.Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18:516–523. doi: 10.1016/j.ceb.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 71.Wong ML, Kaye AH, Hovens CM. Targeting malignant glioma survival signalling to improve clinical outcomes. J Clin Neurosci. 2007;14:301–308. doi: 10.1016/j.jocn.2006.11.005. [DOI] [PubMed] [Google Scholar]
  • 72.Schlaepfer DD, Hauck CR, Sieg DJ. Signaling through focal adhesion kinase. Prog Biophys Mol Biol. 1999;71:435–478. doi: 10.1016/s0079-6107(98)00052-2. [DOI] [PubMed] [Google Scholar]
  • 73.Levy JB, Iba H, Hanafusa H. Activation of the transforming potential of p60c-src by a single amino acid change. Proc Natl Acad Sci U S A. 1986;83:4228–4232. doi: 10.1073/pnas.83.12.4228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Riley D, Carragher NO, Frame MC, Wyke JA. The mechanism of cell cycle regulation by v-Src. Oncogene. 2001;20:5941–5950. doi: 10.1038/sj.onc.1204826. [DOI] [PubMed] [Google Scholar]
  • 75.Hecker TP, Ding Q, Rege TA, Hanks SK, Gladson CL. Overexpression of FAK promotes Ras activity through the formation of a FAK/p120RasGAP complex in malignant astrocytoma cells. Oncogene. 2004;23:3962–3971. doi: 10.1038/sj.onc.1207541. [DOI] [PubMed] [Google Scholar]
  • 76.Ding Q, Grammer JR, Nelson MA, Guan JL, Stewart JE, Jr, Gladson CL. p27Kip1 and cyclin D1 are necessary for focal adhesion kinase regulation of cell cycle progression in glioblastoma cells propagated in vitro and in vivo in the scid mouse brain. J Biol Chem. 2005;280:6802–6815. doi: 10.1074/jbc.M409180200. [DOI] [PubMed] [Google Scholar]
  • 77.Frisch SM, Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol. 1994;124:619–626. doi: 10.1083/jcb.124.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Laplante P, Raymond MA, Labelle A, Abe J, Iozzo RV, Hebert MJ. Perlecan proteolysis induces an alpha2beta1 integrin- and Src family kinase-dependent anti-apoptotic pathway in fibroblasts in the absence of focal adhesion kinase activation. J Biol Chem. 2006;281:30383–30392. doi: 10.1074/jbc.M606412200. [DOI] [PubMed] [Google Scholar]
  • 79.Lue H, Thiele M, Franz J, Dahl E, Speckgens S, Leng L, Fingerle-Rowson G, Bucala R, Luscher B, Bernhagen J. Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene. 2007;26:5046–5059. doi: 10.1038/sj.onc.1210318. [DOI] [PubMed] [Google Scholar]
  • 80.Sade H, Krishna S, Sarin A. The anti-apoptotic effect of Notch-1 requires Cp56lck-dependent, Akt/PKB-mediated signaling in T cells. J Biol Chem. 2004;279:2937–2944. doi: 10.1074/jbc.M309924200. [DOI] [PubMed] [Google Scholar]
  • 81.Shor AC, Keschman EA, Lee FY, Muro-Cacho C, Letson GD, Trent JC, Pledger WJ, Jove R. Dasatinib inhibits migration and invasion in diverse human sarcoma cell lines and induces apoptosis in bone sarcoma cells dependent on SRC kinase for survival. Cancer Res. 2007;67:2800–2808. doi: 10.1158/0008-5472.CAN-06-3469. [DOI] [PubMed] [Google Scholar]
  • 82.Milano V, Piao Y, LaFortune T, de G J. Dasatinib-induced autophagy is enhanced in combination with temozolomide in glioma. Mol Cancer Ther. 2009;8:394–406. doi: 10.1158/1535-7163.MCT-08-0669. [DOI] [PubMed] [Google Scholar]
  • 83.Fincham VJ, Frame MC. The catalytic activity of Src is dispensable for translocation to focal adhesions but controls the turnover of these structures during cell motility. EMBO J. 1998;17:81–92. doi: 10.1093/emboj/17.1.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992;69:11–25. doi: 10.1016/0092-8674(92)90115-s. [DOI] [PubMed] [Google Scholar]
  • 85.Datta A, Huber F, Boettiger D. Phosphorylation of beta3 integrin controls ligand binding strength. J Biol Chem. 2002;277:3943–3949. doi: 10.1074/jbc.M109536200. [DOI] [PubMed] [Google Scholar]
  • 86.Johansson MW, Larsson E, Luning B, Pasquale EB, Ruoslahti E. Altered localization and cytoplasmic domain-binding properties of tyrosine-phosphorylated beta 1 integrin. J Cell Biol. 1994;126:1299–1309. doi: 10.1083/jcb.126.5.1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mariotti A, Kedeshian PA, Dans M, Curatola AM, Gagnoux-Palacios L, Giancotti FG. EGF-R signaling through Fyn kinase disrupts the function of integrin alpha6beta4 at hemidesmosomes: role in epithelial cell migration and carcinoma invasion. J Cell Biol. 2001;155:447–458. doi: 10.1083/jcb.200105017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ding Q, Stewart J, Jr, Olman MA, Klobe MR, Gladson CL. The pattern of enhancement of Src kinase activity on platelet-derived growth factor stimulation of glioblastoma cells is affected by the integrin engaged. J Biol Chem. 2003;278:39882–39891. doi: 10.1074/jbc.M304685200. [DOI] [PubMed] [Google Scholar]
  • 89.Nam S, Kim D, Cheng JQ, Zhang S, Lee JH, Buettner R, Mirosevich J, Lee FY, Jove R. Action of the Src family kinase inhibitor, dasatinib (BMS-354825), on human prostate cancer cells. Cancer Res. 2005;65:9185–9189. doi: 10.1158/0008-5472.CAN-05-1731. [DOI] [PubMed] [Google Scholar]
  • 90.Buettner R, Mesa T, Vultur A, Lee F, Jove R. Inhibition of Src family kinases with dasatinib blocks migration and invasion of human melanoma cells. Mol Cancer Res. 2008;6:1766–1774. doi: 10.1158/1541-7786.MCR-08-0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dey N, Crosswell HE, De P, Parsons R, Peng Q, Su JD, Durden DL. The protein phosphatase activity of PTEN regulates SRC family kinases and controls glioma migration. Cancer Res. 2008;68:1862–1871. doi: 10.1158/0008-5472.CAN-07-1182. [DOI] [PubMed] [Google Scholar]
  • 92.Pilkington GJ. The paradox of neoplastic glial cell invasion of the brain and apparent metastatic failure. Anticancer Res. 1997;17:4103–4105. [PubMed] [Google Scholar]
  • 93.de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010;12:233–242. doi: 10.1093/neuonc/nop027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lewis-Tuffin L, Scheithauer B, Giannini C, Jaeckle KA, Buckner JC, Galanis E, Sarkaria JN, Anastasiadis PZ. Combining anti-invasive and anti-angiogenic therapies for the treatment of GBM. 2009 http://ncitranslates.nci.nih.gov/docs/TSM2%20Abstract%20Book%20FINAL.pdf (abstract 368)
  • 95.Natarajan M, Stewart JE, Golemis EA, Pugacheva EN, Alexandropoulos K, Cox BD, Wang W, Grammer JR, Gladson CL. HEF1 is a necessary and specific downstream effector of FAK that promotes the migration of glioblastoma cells. Oncogene. 2006;25:1721–1732. doi: 10.1038/sj.onc.1209199. [DOI] [PubMed] [Google Scholar]
  • 96.Webb BA, Jia L, Eves R, Mak AS. Dissecting the functional domain requirements of cortactin in invadopodia formation. Eur J Cell Biol. 2007;86:189–206. doi: 10.1016/j.ejcb.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 97.Chan KT, Cortesio CL, Huttenlocher A. FAK alters invadopodia and focal adhesion composition and dynamics to regulate breast cancer invasion. J Cell Biol. 2009;185:357–370. doi: 10.1083/jcb.200809110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Angers-Loustau A, Hering R, Werbowetski TE, Kaplan DR, Del Maestro RF. SRC regulates actin dynamics and invasion of malignant glial cells in three dimensions. Mol Cancer Res. 2004;2:595–605. [PubMed] [Google Scholar]
  • 99.Fillmore HL, VanMeter TE, Broaddus WC. Membrane-type matrix metalloproteinases (MT-MMPs): expression and function during glioma invasion. J Neurooncol. 2001;53:187–202. doi: 10.1023/a:1012213604731. [DOI] [PubMed] [Google Scholar]
  • 100.Shih WL, Liao MH, Yu FL, Lin PY, Hsu HY, Chiu SJ. AMF/PGI transactivates the MMP-3 gene through the activation of Src-RhoA-phosphatidylinositol 3-kinase signaling to induce hepatoma cell migration. Cancer Lett. 2008;270:202–217. doi: 10.1016/j.canlet.2008.05.005. [DOI] [PubMed] [Google Scholar]
  • 101.Nyalendo C, Michaud M, Beaulieu E, Roghi C, Murphy G, Gingras D, Beliveau R. Src-dependent phosphorylation of membrane type I matrix metalloproteinase on cytoplasmic tyrosine 573: role in endothelial and tumor cell migration. J Biol Chem. 2007;282:15690–15699. doi: 10.1074/jbc.M608045200. [DOI] [PubMed] [Google Scholar]
  • 102.Shin EY, Ma EK, Kim CK, Kwak SJ, Kim EG. Src/ERK but not phospholipase D is involved in keratinocyte growth factor-stimulated secretion of matrix metalloprotease-9 and urokinase-type plasminogen activator in SNU-16 human stomach cancer cell. J Cancer Res Clin Oncol. 2002;128:596–602. doi: 10.1007/s00432-002-0388-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Leupold JH, Asangani I, Maurer GD, Lengyel E, Post S, Allgayer H. Src induces urokinase receptor gene expression and invasion/intravasation via activator protein-1/p-c-Jun in colorectal cancer. Mol Cancer Res. 2007;5:485–496. doi: 10.1158/1541-7786.MCR-06-0211. [DOI] [PubMed] [Google Scholar]
  • 104.Lin BR, Chang CC, Chen LR, Wu MH, Wang MY, Kuo IH, Chu CY, Chang KJ, Lee PH, Chen WJ, Kuo ML, Lin MT. Cysteine-rich 61 (CCN1) enhances chemotactic migration, transendothelial cell migration, and intravasation by concomitantly up-regulating chemokine receptor 1 and 2. Mol Cancer Res. 2007;5:1111–1123. doi: 10.1158/1541-7786.MCR-06-0289. [DOI] [PubMed] [Google Scholar]
  • 105.Criscuoli ML, Nguyen M, Eliceiri BP. Tumor metastasis but not tumor growth is dependent on Src-mediated vascular permeability. Blood. 2005;105:1508–1514. doi: 10.1182/blood-2004-06-2246. [DOI] [PubMed] [Google Scholar]
  • 106.Weis S, Cui J, Barnes L, Cheresh D. Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J Cell Biol. 2004;167:223–229. doi: 10.1083/jcb.200408130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lee SW, Kim WJ, Choi YK, Song HS, Son MJ, Gelman IH, Kim YJ, Kim KW. SSeCKS regulates angiogenesis and tight junction formation in blood-brain barrier. Nat Med. 2003;9:900–906. doi: 10.1038/nm889. [DOI] [PubMed] [Google Scholar]
  • 108.Mukhopadhyay D, Tsiokas L, Zhou XM, Foster D, Brugge JS, Sukhatme VP. Hypoxic induction of human vascular endothelial growth factor expression through c-Src activation. Nature. 1995;375:577–581. doi: 10.1038/375577a0. [DOI] [PubMed] [Google Scholar]
  • 109.Theurillat JP, Hainfellner J, Maddalena A, Weissenberger J, Aguzzi A. Early induction of angiogenetic signals in gliomas of GFAP-v-src transgenic mice. Am J Pathol. 1999;154:581–590. doi: 10.1016/S0002-9440(10)65303-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Summy JM, Trevino JG, Lesslie DP, Baker CH, Shakespeare WC, Wang Y, Sundaramoorthi R, Metcalf CA, III, Keats JA, Sawyer TK, Gallick GE. AP23846, a novel and highly potent Src family kinase inhibitor, reduces vascular endothelial growth factor and interleukin-8 expression in human solid tumor cell lines and abrogates downstream angiogenic processes. Mol Cancer Ther. 2005;4:1900–1911. doi: 10.1158/1535-7163.MCT-05-0171. [DOI] [PubMed] [Google Scholar]
  • 111.Eliceiri BP, Paul R, Schwartzberg PL, Hood JD, Leng J, Cheresh DA. Selective requirement for Src kinases during VEGF-induced angiogenesis and vascular permeability. Mol Cell. 1999;4:915–924. doi: 10.1016/s1097-2765(00)80221-x. [DOI] [PubMed] [Google Scholar]
  • 112.Hennequin LF, Allen J, Breed J, Curwen J, Fennell M, Green TP, Lambertvan der BC, Morgentin R, Norman RA, Olivier A, Otterbein L, Ple PA, Warin N, Costello G. N-(5-chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine, a novel, highly selective, orally available, dual-specific c-Src/Abl kinase inhibitor. J Med Chem. 2006;49:6465–6488. doi: 10.1021/jm060434q. [DOI] [PubMed] [Google Scholar]
  • 113.Purnell PR, Mack PC, Tepper CG, Evans CP, Green TP, Gumerlock PH, Lara PN, Gandara DR, Kung HJ, Gautschi O. The Src inhibitor AZD0530 blocks invasion and may act as a radiosensitizer in lung cancer cells. J Thorac Oncol. 2009;4:448–454. doi: 10.1097/JTO.0b013e31819c78fb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cuneo KC, Geng L, Tan J, Brousal J, Shinohara ET, Osusky K, Fu A, Shyr Y, Wu H, Hallahan DE. SRC family kinase inhibitor SU6656 enhances antiangiogenic effect of irradiation. Int J Radiat Oncol Biol Phys. 2006;64:1197–1203. doi: 10.1016/j.ijrobp.2005.11.014. [DOI] [PubMed] [Google Scholar]
  • 115.Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M, Foekens JA, Massague J. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell. 2009;16:67–78. doi: 10.1016/j.ccr.2009.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Altaha R, Crowell E, Hobbs G, Higa G, Abraham J. Increased risk of brain metastases in patients with HER-2/neu-positive breast carcinoma. Cancer. 2005;103:442–443. doi: 10.1002/cncr.20813. [DOI] [PubMed] [Google Scholar]
  • 117.Leyland-Jones B. Human epidermal growth factor receptor 2-positive breast cancer and central nervous system metastases. J Clin Oncol. 2009;27:5278–5286. doi: 10.1200/JCO.2008.19.8481. [DOI] [PubMed] [Google Scholar]
  • 118.Vadlamudi RK, Sahin AA, Adam L, Wang RA, Kumar R. Heregulin and HER2 signaling selectively activates c-Src phosphorylation at tyrosine 215. FEBS Lett. 2003;543:76–80. doi: 10.1016/s0014-5793(03)00404-6. [DOI] [PubMed] [Google Scholar]
  • 119.Carter WB, Niu G, Ward MD, Small G, Hahn JE, Muffly BJ. Mechanisms of HER2-induced endothelial cell retraction. Ann Surg Oncol. 2007;14:2971–2978. doi: 10.1245/s10434-007-9442-4. [DOI] [PubMed] [Google Scholar]
  • 120.Rucci N, Recchia I, Angelucci A, Alamanou M, Del FA, Fortunati D, Susa M, Fabbro D, Bologna M, Teti A. Inhibition of protein kinase c-Src reduces the incidence of breast cancer metastases and increases survival in mice: implications for therapy. J Pharmacol Exp Ther. 2006;318:161–172. doi: 10.1124/jpet.106.102004. [DOI] [PubMed] [Google Scholar]
  • 121.Hashimoto S, Hirose M, Hashimoto A, Morishige M, Yamada A, Hosaka H, Akagi K, Ogawa E, Oneyama C, Agatsuma T, Okada M, Kobayashi H, Wada H, Nakano H, Ikegami T, Nakagawa A, Sabe H. Targeting AMAP1 and cortactin binding bearing an atypical src homology 3/proline interface for prevention of breast cancer invasion and metastasis. Proc Natl Acad Sci U S A. 2006;103:7036–7041. doi: 10.1073/pnas.0509166103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Marchetti D, Parikh N, Sudol M, Gallick GE. Stimulation of the protein tyrosine kinase c-Yes but not c-Src by neurotrophins in human brain-metastatic melanoma cells. Oncogene. 1998;16:3253–3260. doi: 10.1038/sj.onc.1201877. [DOI] [PubMed] [Google Scholar]
  • 123.Song L, Morris M, Bagui T, Lee FY, Jove R, Haura EB. Dasatinib (BMS-354825) selectively induces apoptosis in lung cancer cells dependent on epidermal growth factor receptor signaling for survival. Cancer Res. 2006;66:5542–5548. doi: 10.1158/0008-5472.CAN-05-4620. [DOI] [PubMed] [Google Scholar]
  • 124.Nautiyal J, Majumder P, Patel BB, Lee FY, Majumdar AP. Src inhibitor dasatinib inhibits growth of breast cancer cells by modulating EGFR signaling. Cancer Lett. 2009;283:143–151. doi: 10.1016/j.canlet.2009.03.035. [DOI] [PubMed] [Google Scholar]
  • 125.Homsi J, Cubitt CL, Zhang S, Munster PN, Yu H, Sullivan DM, Jove R, Messina JL, Daud AI. Src activation in melanoma and Src inhibitors as therapeutic agents in melanoma. Melanoma Res. 2009;19:167–175. doi: 10.1097/CMR.0b013e328304974c. [DOI] [PubMed] [Google Scholar]
  • 126.Chang Q, Jorgensen C, Pawson T, Hedley DW. Effects of dasatinib on EphA2 receptor tyrosine kinase activity and downstream signalling in pancreatic cancer. Br J Cancer. 2008;99:1074–1082. doi: 10.1038/sj.bjc.6604676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.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. 2004;47:6658–6661. doi: 10.1021/jm049486a. [DOI] [PubMed] [Google Scholar]
  • 128.Hanke JH, Gardner JP, Dow RL, Changelian PS, Brissette WH, Weringer EJ, Pollok BA, Connelly PA. Discovery of a novel, potent, and Src family-selective tyrosine kinase inhibitor. Study of Lck- and FynT-dependent T cell activation. J Biol Chem. 1996;271:695–701. doi: 10.1074/jbc.271.2.695. [DOI] [PubMed] [Google Scholar]
  • 129.Cuneo KC, Geng L, Tan J, Brousal J, Shinohara ET, Osusky K, Fu A, Shyr Y, Wu H, Hallahan DE. SRC family kinase inhibitor SU6656 enhances Oantiangiogenic effect of irradiation. Int J Radiat Oncol Biol Phys. 2006;64:1197–1203. doi: 10.1016/j.ijrobp.2005.11.014. [DOI] [PubMed] [Google Scholar]
  • 130.Puttini M, Coluccia AM, Boschelli F, Cleris L, Marchesi E, Donella-Deana A, Ahmed S, Redaelli S, Piazza R, Magistroni V, Andreoni F, Scapozza L, Formelli F, Gambacorti-Passerini C. In vitro and in vivo activity of SKI-606, a novel Src-Abl inhibitor, against imatinib-resistant Bcr-Abl+ neoplastic cells. Cancer Res. 2006;66:11314–11322. doi: 10.1158/0008-5472.CAN-06-1199. [DOI] [PubMed] [Google Scholar]
  • 131.Green TP, Fennell M, Whittaker R, Curwen J, Jacobs V, Allen J, Logie A, Hargreaves J, Hickinson DM, Wilkinson RW, Elvin P, Boyer B, Carragher N, Ple PA, Bermingham A, Holdgate GA, Ward WH, Hennequin LF, Davies BR, Costello GF. Preclinical anticancer activity of the potent, oral Src inhibitor AZD0530. Mol Oncol. 2009;3:248–261. doi: 10.1016/j.molonc.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Chen Y, Guggisberg N, Jorda M, Gonzalez-Angulo A, Hennessy B, Mills GB, Tan CK, Slingerland JM. Combined Src and aromatase inhibition impairs human breast cancer growth in vivo and bypass pathways are activated in AZD0530-resistant tumors. Clin Cancer Res. 2009;15:3396–3405. doi: 10.1158/1078-0432.CCR-08-3127. [DOI] [PubMed] [Google Scholar]
  • 133.Soverini S, Gnani A, Colarossi S, Castagnetti F, Abruzzese E, Paolini S, Merante S, Orlandi E, De MS, Gozzini A, Iacobucci I, Palandri F, Gugliotta G, Papayannidis C, Poerio A, Amabile M, Cilloni D, Rosti G, Baccarani M, Martinelli G. Philadelphia-positive patients who already harbor imatinib-resistant Bcr-Abl kinase domain mutations have a higher likelihood of developing additional mutations associated with resistance to second- or third-line tyrosine kinase inhibitors. Blood. 2009;114:2168–2171. doi: 10.1182/blood-2009-01-197186. [DOI] [PubMed] [Google Scholar]
  • 134.Bradeen HA, Eide CA, O'Hare T, Johnson KJ, Willis SG, Lee FY, Druker BJ, Deininger MW. Comparison of imatinib mesylate, dasatinib (BMS-354825), and nilotinib (AMN107) in an N-ethyl-N-nitrosourea (ENU)-based mutagenesis screen: high efficacy of drug combinations. Blood. 2006;108:2332–2338. doi: 10.1182/blood-2006-02-004580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Koptyra M, Falinski R, Nowicki MO, Stoklosa T, Majsterek I, Nieborowska-Skorska M, Blasiak J, Skorski T. BCR/ABL kinase induces self-mutagenesis via reactive oxygen species to encode imatinib resistance. Blood. 2006;108:319–327. doi: 10.1182/blood-2005-07-2815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lassman AB, Wang M, Gilbert MW, Adalpe K, Wright J, Brachman DG, Malkin MG, Mehta M. Phase 2 trial of dasatinib in patients with recurrent glioblastoma (RTOG 0627) Neuro-Oncology. 2008;10 doi: 10.1093/neuonc/nov011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Porkka K, Koskenvesa P, Lundan T, Rimpilainen J, Mustjoki S, Smykla R, Wild R, Luo R, Arnan M, Brethon B, Eccersley L, Hjorth-Hansen H, Hoglund M, Klamova H, Knutsen H, Parikh S, Raffoux E, Gruber F, Brito-Babapulle F, Dombret H, Duarte RF, Elonen E, Paquette R, Zwaan CM, Lee FY. Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia. Blood. 2008;112:1005–1012. doi: 10.1182/blood-2008-02-140665. [DOI] [PubMed] [Google Scholar]
  • 138.Summy JM, Gallick GE. Treatment for advanced tumors: SRC reclaims center stage. Clin Cancer Res. 2006;12:1398–1401. doi: 10.1158/1078-0432.CCR-05-2692. [DOI] [PubMed] [Google Scholar]

RESOURCES