Abstract
The epidermal growth factor receptor (EGFR) is amplified or mutated in various human epithelial tumors. Its expression and activation leads to cell proliferation, differentiation, and survival. Consistently, EGFR amplification or expression of EGFR variant 3 (EGFRvIII) is associated with resistance to conventional cancer therapy through activation of pro-survival signaling and DNA-repair mechanisms. EGFR targeting has successfully been exploited as strategy to increase treatment efficacy. Nevertheless, these targeting strategies have only been proven effective in a limited percentage of human tumors.
Recent knowledge indicates that EGFR deregulated tumors display differences in autophagy and dependence on autophagy for growth and survival and the use of autophagy to increase resistance to EGFR-targeting drugs. In this review the dependency on autophagy and its role in mediating resistance to EGFR-targeting agents will be discussed. Considering the current knowledge, autophagy inhibition could provide a novel strategy to enhance therapy efficacy in treatment of EGFR deregulated tumors.
Keywords: EGFR, EGFRvIII, mutations, autophagy, cancer treatment, treatment resistance, hypoxia, starvation, metabolic stress
Introduction
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase within the ErbB family consisting of 4 members; EGFR (ErbB1, HER1), ErbB2 (HER2), ErbB3 (HER3), and ErbB4 (HER4). ErbBs are typical receptor tyrosine kinases that were implicated in cancer in the early 1980s, when the avian erythroblastosis tumor virus was found to encode an aberrant form of the human epidermal growth factor receptor.1 In many different cancer cell types, the ErbB pathway becomes hyperactivated by a range of mechanisms, including overproduction of ligands, overproduction of receptors, or constitutive activation of receptors.2 In general, EGFR signaling is triggered by ligand binding to the extracellular ligand binding domain. This initiates receptor homo-/hetero-dimerization and autophosphorylation through the intracellular kinase domain, resulting in receptor activation. Following activation, cytoplasmic substrates are phosphorylated and initiate a signaling cascade that drives multiple cellular responses, including changes in gene expression, cytoskeletal rearrangement, apoptosis inhibition, and increased cell proliferation.3,4 In cancer, EGFR signaling is often deregulated, leading to treatment resistance of the tumor and poor survival of patients. This deregulation is often mediated by overexpression (e.g., through gene amplification) and numerous mutations that lead to uncontrolled and sustained EGFR-signaling. Several EGFR targeting therapies have been developed (e.g., tyrosine kinase inhibitors (TKI) that inhibit EGFR signaling and antibodies that prevent EGFR expression and dimerization). Unfortunately, these therapies have only been proven effective in a limited percentage of cancer patients despite the presence of EGFR in many of the targeted tumors.5 Novel strategies that, potentially combined with earlier EGFR-targeting agents, lead to enhanced cell killing are therefore still desired.
Current research has indicated that EGFR-deregulated cells and tumors display alterations in their autophagic response, a pro-survival mechanism that allows cells to recycle nutrients for energy- and macromolecule production.6 Importantly: (1) EGFR-deregulated cells seem to be more dependent on autophagy for growth and survival; and (2) resistance to EGFR-targeting agents can be reduced through autophagy inhibition, providing a potential novel modality to target these tumors. In this review we highlight current knowledge that may provide insights as to why EGFR-deregulated cells display differences in autophagic responses and dependency on autophagy for survival and provide rationale for combining autophagy inhibition with conventional cancer therapy.
Gene Amplification and Overexpression
One of the most investigated alterations in the EGFR function is activation of signaling through increased gene copy number arising from amplification or polysomy.7-9 Elevated EGFR expression is a strong prognostic indicator in head and neck, ovarian, cervical, bladder, and esophageal cancer. In gastric, breast endometrial, and colorectal cancers (CRC) EGFR expression is a modest predictor. This in contrast to non-small cell lung carcinoma (NSCLC), where increased EGFR expression rarely has a prognostic value.10
EGFR mutations often determine the responsiveness of tumors to EGFR inhibitors; this is often related to the dependency of cancer on continued oncogenic signaling (oncogene addiction). For a number of different oncogenes, data supporting addiction in tumors have been gathered.11,12 For EGFR in particular, positive results in clinical trials with different antagonists have been considered as clinical evidence of oncogene addiction, even though the clinical benefits from the use of either monoclonal antibodies (mAbs) or TKIs have been limited.5 Only a small portion (9–20%) of tumors with hyperactive EGFR signaling is exquisitely sensitive to such specific inhibitors.13-15 This percentage is much higher (88–94.1%) when sensitizing mutations (e.g., L858R) in the EGFR gene are present.16,17
In NSCLC and CRC, increased EGFR gene copy number has been associated with increased clinical efficacy of EGFR antagonists erlotinib and cetuximab.18 Both drugs have shown clinical promise, and the anti-EGFR antibody cetuximab is used in treatment of head and neck squamous cell cancer (HNSCC) and CRC. Despite clinical gain, both intrinsic resistance and the development of acquired resistance have been observed.19
The Tyrosine Kinase Domain
Both mutations associated with drug resistance and sensitivity have been described within the tyrosine kinase (TK) domain of EGFR in subsets of NSCLC, rare cases in HNSCC, CRC, small cell lung carcinomas (SCLC), ovarian, esophageal, and pancreatic cancers.20 Distribution of mutations is not random and may be related to cancer etiology. For instance, in NSCLC the incidence of EGFR mutations among clinical responders to gefitinib or erlotinib is 77%, compared with only 7% in NSCLC cases that are refractory to tyrosine kinase inhibitor (TKI) treatment.20
Multiple studies have shown differences in treatment outcome associated with EGFR mutations. For example, mutations in exon 18 (nucleotide-binding loop), accounting for 5% of the mutations, are usually amino acid substitutions that contribute to drug sensitivity. Mutations in exon 19 are characterized by small in-frame deletions and account for 45% of EGFR mutations, making it the most prominent EGFR kinase domain mutation in NSCLC. These tumors are, in general, sensitive to TKIs like gefitinib and erlotinib.20 The L858R substitution in exon 21, within the activation loop of EGFR, comprises approximately 40–45% of EGFR mutations. Tumors harboring the L858R mutation are, in general, sensitive to TKIs, although some clinical studies have shown that these tumors are not as responsive in comparison to tumors with deletion mutations in exon 19.20
EGFR exon 20 mutations, typically located after the C-helix of the tyrosine kinase domain, may account for up to 4% of all EGFR mutations, with the T790M substitution as the most prominent one (up to 50% of all mutations in exon 20). This T790M mutation is considered an acquired mutation and converts TKI-sensitive tumors into (reversible) TKI-resistant tumors.21 Like the T790M mutation, other exon 20 mutated proteins are resistant to clinically achievable doses of reversible (gefitinib, erlotinib) and irreversible (neratinib, afatinib, PF00299804) TKIs in preclinical models.22 Growing clinical experience with tumors harboring EGFR exon 20 insertions correspond with the preclinical data; only few patients have shown responsiveness to EGFR TKIs.22
EGFRvIII
In a significant proportion of tumors, amplification of the EGFR gene is accompanied by rearrangements, although amplification is not mandatory for gene rearrangement.23 The most abundant rearrangement is a deletion variant that lacks exon 2–7 of the extracellular domain, yielding a constitutively active receptor, EGFRvIII or Δ2–7.24-26 This mutation is most prevalent in malignant gliomas (20–30% in unselected patients with a glioblastoma multiforme [GBM] and 50–60% in patients whose tumors show amplification of wild-type EGFR).27 Recent studies identified EGFRvIII in head and neck squamous cell carcinomas (~21%),28 squamous cell carcinomas of the lung (~5%),29,30 and breast (~5%),31 suggesting broader implications to human cancer.32
EGFRvIII is known to contribute to radio resistance of tumor cells33 at least in part through enhanced repair of DNA double-strand breaks.34 Additionally, EGFRvIII expression is associated with resistance to gefitinib and leads to sustained EGFR signaling and AKT activity.35 Furthermore, the tumor microenvironment, and in particular tumor hypoxia, significantly contributes to therapy resistance.36-38 Expression of EGFRvIII provides cells with a survival advantage when exposed to stresses such as hypoxia and nutrient starvation.39
Although EGFRvIII expression is frequently observed in GBM, culturing GBM cells in vitro will lead to a rapid loss of EGFRvIII expression,40,41 and thus complicates assessment of EGFRvIII-targeting strategies in GBM. Researchers therefore often use cell lines that artificially express EGFRvIII. Although informative, these cell lines have their limitations as, unlike in primary GBM (a range of 1–100% EGFRvIII positive cells in GBM is observed),23 all cells will express EGFRvIII. Furthermore, the heterogeneous expression levels observed in GBM are difficult to mimic due to the use of artificial promoters; in addition, the cells were established without EGFRvIII and are thus not dependent on EGFRvIII for growth and survival. To maintain EGFRvIII expression in vitro, cells could be cultured under stem cell culture conditions.42 Alternatively, EGFRvIII expression is also maintained when primary tumors are xenografted subcutaneously on mice42 and should be considered in validating results obtained in transgenic models.
For GBM patients, EGFR overexpression is a significant prognostic value for predicting survival, and the expression of EGFRvIII with EGFR amplification plays an important role in enhanced tumorigenicity. EGFRvIII overexpression in the presence of EGFR amplification is the strongest indicator for poor survival prognosis in 2 large cohorts of patients. Shinojima and colleagues found in a cohort of 87 patients that EGFRvIII expression, assessed by immunohistochemistry (IHC), was not a predictor for overall survival (OS). However, in patients with EGFR amplification, multivariate analysis revealed that EGFRvIII expression was an independent, significant, poor prognostic factor for OS (P = 0.0044, HR = 2.71).23 These findings were endorsed by Pelloski et al.,43 who observed that the median survival of a patient group with EGFRvIII expression (n = 36, assessed by IHC) was reduced from 85 to 47 wk compared with EGFRvIII-negative patient group (n = 81). In contrast, Montano et al.44 showed, in a cohort of 73 patients, that EGFRvIII (assessed by reverse transcription-PCR) is a molecular predictor of improved overall survival (P = 0.0023, HR = 2.59) in GBM patients treated with surgery followed by adjuvant radiotherapy and temozolomide (TMZ). This discrepancy could potentially be explained by the EGFRvIII detection method. Montano used the more sensitive RT-PCR, whereas Pelloski and Shinojima used IHC and may have missed very low levels of EGFRvIII expression. Another possible explanation for the differences could be the uniformness of the patient group. Montano used patients that all underwent surgery, radiotherapy, and TMZ treatment, whereas the other cohorts were treated more heterogeneously. Furthermore, all patients in Pelloski’s study were wild-type for YKL-40 (a Ras activator), were Montano does not discriminate between Ras activator status, and the Karnofsky performance status (KPS score) of the patients in Pelloski’s and Shinojima’s cohort was much higher.23,43,44 Taken together, more and lager cohorts with uniform treatment are required to gain additional insight in the clinical relevance of EGFRvIII.
Cancer Stem Cells
Recent data showed that EGFR and EGFRvIII signaling are involved in maintaining a cancer stem cell (CSC) phenotype. In glioblastoma, both the EGFRpos and EGFRneg tumor-initiating cells (TICs) derived from primary GBM can give rise to experimental tumors. However, the EGFRpos TICs displayed enhanced tumorigenic potential and highly invasive behavior.45 Conversely, EGFRneg TICs formed tumors with low efficiency and needed to re-upregulate their EGFR expression to become tumorigenic. These “potential” CSCs might be kept in a dormancy-like state by EGFR downregulation and be reactivated when exposed to stimuli of the in vivo tumor microenvironment.45 Indeed, GBMs that were EGFRneg in origin expressed EGFR on recurrence.46 The idea of EGFRpos and EGFRneg CSC is further supported by the finding that CSC propagation is possible in the absence of exogenous growth factors (like EGF), suggesting that EGF signaling is not critical for GBM CSC maintenance.47 In contrast, EGFR signaling is required for GMB CSC proliferation,48,49 and gefitinib treatment decreases CSC number in nasopharyngeal carcinoma models.50 In this study, cisplatin-treated tumor cells regrew rapidly upon re-implantation, whereas regrowth of gefitinib-treated tumor cells was severely diminished.50 Furthermore, Clark et al.51 showed that GBM CSC lines displayed tumor-initiating capacity after EGF withdrawal or cetuximab treatment by compensatory activation of ErbB2 and ErbB3, suggesting a resistance mechanism for EGFR-targeted therapy. Lapatinib, a dual EGFR/ErbB2 inhibitor, treatment inhibited CSCs proliferation, indicating that a simultaneous blockade of multiple ErbB family members could be needed for more efficient GBM treatment.
In relation to EGFRvIII in CSC, a population of the cells derived from pediatric diffuse intrinsic pontine gliomas (DIPG) neurospheres displayed co-expression of the CSC marker CD133 and EGFRvIII.52 In another study, EGFRvIII expression on invasive breast cancer carcinomas resulted in increased expression of genes related to self-renewal and epithelial–mesenchymal transition, along with a higher percentage of CSC-like cells.31 Furthermore, Liu et al.53 showed that the CD133+ fraction of GBM exclusively expressed EGFRvIII, whereas wild-type EGFR was not detected. These data indicate a role for EGFRvIII in the propagation of CSC that could explain the relative therapy resistance of EGFRvIII tumors.
EGFR Signaling Pathways Implicated in Autophagy
After ligand binding by EGFR or constitutive signaling by EGFRvIII the activation of several parallel pathways has been described. These include: (1) activation of the PI3K-AKT-mTOR pathway; (2) increased Ras and (3) STAT3 signaling; and (4) Beclin1 (Fig. 1).54 All pathways involved in autophagy regulation.
Figure 1. EGFR- and EGFRvIII-signaling pathways associated with autophagy regulation. Both receptors signal through all 4 pathways; nevertheless, EGFR preferentially signals via the RAS pathway, whereas EGFRvIII predominantly uses mTOR signaling.
Autophagy is a catabolic process that allows cells to recycle cellular components through degradation by the lysosomal machinery.55,56 Autophagy is an evolutionarily conserved process that results in the targeting of cellular proteins and organelles to lysosomes for degradation. Autophagy serves to regulate normal organelle turnover and the removal of those with compromised function to maintain cellular homeostasis. Additionally, autophagy is a survival mechanism during periods of metabolic stress, where self-digestion provides an alternative energy source and facilitates the disposal of unfolded proteins.57-60
Previously, we and others showed that cells with deregulated EGFR signaling display differences in autophagic response.61-63 Interestingly, EGFR expression represses autophagy activity. For example, EGFR reduction by siRNA treatment leads to an induction of autophagy activity in prostate cancer cells.63 Furthermore, induction in autophagy was observed after targeting with TKIs or cetuximab.64 Recently, in a panel of HNSSC xenografts, we observed a correlation between EGFR and expression of the autophagy marker Lc3b, suggesting a close interplay between EGFR signaling and autophagy. This correlation is most likely mediated through controlling Lc3b protein production, as this correlation was also observed on the mRNA level.61 This was further confirmed in a panel of cell lines, where EGFR expression negatively correlated with autophagic flux, as determined by Lc3b-turnover. Interestingly, the suppressive activity of EGFR in these cells can be independent of its kinase activity65 and mediated through maintaining high glucose levels through association with sodium/glucose cotransporter 1 (SGLT1).63 In addition, EGFR can suppress autophagy dependent on its kinase domain through maintaining high activation of the PI3K/Akt/mTOR pathway.66 Furthermore, EGFR activity results in inhibition of autophagy through inhibition of beclin1,62 a potent inducer of autophagy. Together these data indicate that the expression of EGFR is closely related to expression of autophagic markers and autophagic activity of cells.
Although the effect of EGFR seems to be mostly autophagy-suppressive, in constitutive EGFR-signaling cells the effect on autophagy activity is less pronounced during normal conditions and seems to be stimulatory during metabolic stresses. For example, in stably transduced glioblastoma cell lines and prostate cancer cells that express EGFRvIII, a faster and more pronounced autophagic response during starvation or severe hypoxia is observed (unpublished data). The enhanced autophagic response provides these cells with survival and growth advantage.
The suppressive action of EGFR on autophagy activity and the opposing action of EGFRvIII during stressful conditions could result from signaling via different signal-transduction pathways. For example, Wolf-Yadlin et al.67 showed that EGFR predominantly signals via Erk1, Erk2, and STAT3, whereas EGFRvIII favors signaling via the PI3K and STAT3 pathway.68,69 This difference in signaling preference of these pathways associated with autophagy activity is likely to result in differences between EGFR and EGFRvIII.
EGFR–PI3K–AKT–mTOR Pathway
Activated EGFR binds GRB2-associated binding protein 1 (GAB1) together with growth factor receptor-bound protein 2 (GRB2) to recruit phosphoinositide-3-kinase (PI3K). PI3K phosphorylates PI(4,5)P2 (phosphatidylinositol) into PI(3,4,5)P3. This process is negatively regulated by phosphatase and tensin homolog (PTEN). 3-phosphoinositide dependent protein kinase-1 (PDK1) brings v-akt murine thymoma viral oncogene homolog 1 (AKT) to the plasma membrane, where PIP3 is located, to phosphorylate and activate AKT. AKT subsequently activates mTOR (mammalian target of Rapamycin).54
mTOR, a central growth regulator downstream of oxygen, energy, nutrient, and growth factor signaling, inhibits autophagy. Hence, insufficiency in either results in mTOR inhibition and rapid induction of autophagy in most systems. In conditions of nutrient sufficiency, high mTOR activity prevents Unc-51-like kinase (ULK1) activation by phosphorylating ULK1 Ser757 and disrupting the interaction between ULK1 and 5′ AMP-activated protein kinase (AMPK), thereby preventing ULK1 to initiate an autophagy activating complex with FIP200 and ATG13.70,71 During periods of starvation, mTOR dissociates from the ULK1 complex, leading to less ULK1 phosphorylation, and increases ULK1 kinase activity.72,73 Recently, a role for ULK1 activation for survival of hypoxic cells was identified.74,75
EGFR–RAS Signaling Pathway
The RAS oncogene is a member of small GTPase family involved in the regulation of cell survival and growth and is frequently activated in cancer.76 Next to frequently detected activating mutations in RAS, growth factor signaling, e.g., through EGFR, can lead to uncontrolled RAS signaling. After auto-phosphorylation, the adaptor protein growth factor receptor-bound protein 2 (GRB2) binds EGFR at the phosphorylated sites and activates Son of sevenless (SOS), a GTP-exchange factor for RAS. SOS then converts RAS-GDP into active RAS-GTP.
Several studies have implicated RAS activity in the induction of autophagy, as displayed by a high autophagic flux after oncogenic RAS transformation.77 Increased autophagy in these cells is required to sustain a high metabolic rate, to prevent accumulation of damaged mitochondria, reduce oxygen consumption, and to prevent metabolic substrate depletion.77-79 In relation, autophagy inhibition in RAS transformed cells leads to enhanced cell killing during nutrient deprivation.77 Furthermore, it has been shown that RAS plays a role in regulating the redox state of the cell, and that constitutive production of ROS correlates with RAS-induced cell transformation80,81 and mediates autophagy induction through activation of protein kinase 8 (JNK) and subsequent upregulation of ATG5 and ATG7.80
EGFR–STAT3 Signaling Pathway
The third main signaling mediator downstream of activated EGFR is the signal transducer and activator of transcription (STAT3) protein. STAT3 belongs to a family of at least 7 transcription factors that share conservation in coiled-coil, SRC homology (SH2), and DNA-binding domains.82 STAT3 is a latent transcription factor present in the cytoplasm of cells. Phosphorylation at Y705, is mediated through activation of several transmembrane receptors, such as EGFR,83 and is required for transcriptional activity or transactivation of members of the Janus kinase (JAK) protein family.84 Phosphorylation leads to dimerization, nuclear translocation, DNA binding, and gene activation.85 Recently, STAT3 has been recognized as a new autophagy regulator through suppression of PKR.86 Shen et al.86 proposed that in normal conditions, latent cytoplasmic STAT3 binds to protein kinase R (PKR), inhibiting its activity, and reduces autophagy levels through eIF2α inhibition, a signaling cascade involved in both transcriptional and translational regulation of Lc3b and ATG5 production.60 Hence, STAT3 phosphorylation leads to homodimerization and enables the free PKR to phosphorylate eIF2α via direct interaction between STAT3 and PKR.87 Furthermore, STAT3 controls the expression of several autophagy-associated proteins, including BCL-2, Bcl-XL, and MCL-1,88,89 which inhibit autophagy through sequestration of Beclin 1.60
EGFR-Beclin 1
Beclin 1 is a coiled-coil protein involved in the regulation of autophagy in mammalian cells and is a component of the class III phosphatidylinositol-3-kinase (PI3K) complex.90 Beclin 1 promotes autophagy, and cells with reduced Beclin 1 expression exhibit reduced autophagic activity.91 Beclin 1 is an essential gene for early embryonic development and is a haploinsufficient tumor suppressor.92 Intriguingly, Beclin 1 is tumor suppressive in breast cancer cells; mice that have only one functional allele of Beclin 1 display higher incidence of spontaneous tumors, and mono-allelical deletions of Beclin 1 have been described for 40–75% of human ovarian, breast, and prostate cancers.91,93-95 Beclin 1 may also promote survival as an interacting partner of an anti-apoptotic protein Bcl-2.96 Binding of Bcl-2 to Beclin 1 inhibits Beclin 1-dependent autophagy and Beclin 1-dependent autophagic cell death.91,97 Recently, it was shown that EGFR phosphorylates Beclin 1 at 3 different tyrosine residues, Y229, Y233, and Y352, after activation by EGF. This tyrosine phosphorylation favors the formation of Beclin 1 dimers, which are incapable of VPS34 binding, and results in reduced autophagy activation (Fig. 1).62
EGFRvIII Tumors Require Increased Metabolism
Why EGFRvIII-expressing tumors require higher activation of autophagy during metabolic stress remains unclear, but could be related to the higher proliferation rate and associated nutritional demand. For example, Guo et al.98 showed that EGFRvIII expression induces major shifts in GBM cell metabolism. Uptake of 18FDG in EGFRvIII-expressing U87 xenografts was doubled compared with volume matched control xenografts. In relation, gene expression arrays showed upregulation of genes involved in regulation of the cell metabolism, e.g., glucose transporter 1 (GLUT1) and GLUT3, Hexokinase2 (HK2), and pyruvate dehydrogenase kinase (PDK1).99
In general, EGFRvIII-expressing tumors require upregulation of cell metabolism proteins and require increased glucose uptake to maintain their elevated growth rate. This might explain why these tumors may display increased dependence on autophagy for their energy supply in a tumor microenvironment that is low in glucose or deprived of oxygen.
EGFR Mediates Mitochondrial Homeostasis
In relation to the involvement of EGFR in cell metabolism, Rasbach et al. showed the involvement of EGFR in mitochondrial biogenesis after oxidant injury via EGFR-dependent p38 MAPK activation of the mitochondrial biogenesis regulator PPAR-γ cofactor-1α (PGC-1),100 allowing the cells to maintain high metabolism and their increased proliferation rate.
Additionally, EGFR is involved in stabilizing mitochondria and preventing apoptosis. Synergistic interaction between EGFR and c-Src via phosphorylation of EGFR at Y845 causes translocation to the mitochondria. There, it interacts with cytochrome c oxidase subunit II (COX II) and prevents apoptosis. This seems independent of EGFR kinase activity but is enhanced by EGF treatment.101,102 Although cells did not undergo apoptosis, ATP production was drastically reduced by binding of EGFR to COX II.102 Similar mechanisms and translocation to the mitochondria to antagonize apoptosis have been observed for EGFRvIII.103,104
COX II inhibition by EGFR leaves the cell with reduced ATP production after insults such as chemo- and radiotherapy or starvation and must revert to other sources for their ATP production. Autophagy may provide an alternative source for energy production, and could be exploited therapeutically in stressed cells with EGFR overexpression. This could also explain why EGFR-expressing cells are more dependent on autophagy for their survival.61
EGFR, Treatment Resistance, and Therapeutic Potential of Autophagy Inhibition
EGFR expression or mutations contribute to tumor treatment resistance. For instance, acquired mutations in the kinase domain of EGFR (like the T790M) can abrogate the susceptibility to TKIs like gefitinib or erlotinib.21 Furthermore, EGFR contributes to radiotherapy resistance either through activation of the pro-survival pathway PLCγ-PKC-RAF105 or through activation of DNA repair via DNA-PK.106 We have also shown that expression of EGFRvIII contributes to stress resistance typical for the tumor microenvironment, including nutrient deprivation and hypoxia.39 Hypoxia is a common feature of tumors and an important contributor to malignancy and treatment resistance,36,37,107 and in HNSCC, the degree of hypoxia is the most significant factor explaining variability in survival.37 Targeting hypoxia in pre-clinical models has been shown to sensitize tumors to therapy through various modalities.60,108,109 Importantly, a meta-analysis in HNSCC demonstrated therapeutic benefit of hypoxia modification.110
Tumor cells adapt to hypoxia through multiple mechanisms, including activation of autophagy.6,60,111-115 Genetic and pharmacological inhibition of autophagy sensitizes human tumor cells to hypoxia, reduces the fraction of viable hypoxic tumor cells, and sensitizes human tumors xenografts to irradiation (Fig. 2A).60
Figure 2. (A) In EGFR-deregulated tumors, inhibition of autophagy leads to increased cell killing of metabolic stressed cells. (B) Resistance of tumor cells with active EGFR signaling to monoclonal antibodies (mAbs) or tyrosine kinase inhibitors (TKIs) can be reduced by autophagy inhibition.
In relation to EGFR expression, although we showed reduced autophagic flux in cells expressing EGFR, these cells were already under normal conditions dependent on autophagy for proliferation and survival.61 In general, EGFR-expressing tumors are considered highly radioresistant;116 also in our setting, a large dose irradiation had only a minor effect on tumor delay. Interestingly, chloroquine administration to inhibit autophagy led to a large delay in tumor growth that exceeded the effect of irradiation and, in addition, sensitized tumors to irradiation.61
Recent data showed that EGFR and EGFRvIII signaling is involved in maintaining a CSC phenotype, and recently it was shown that autophagy is important for CSC self-renewal and tumorigenic potential in breast cancer stem cells,117 and for regulation of energy metabolism and migration and invasion of GBM-derived stem cells.118 Taken together, these data suggest that autophagy and EGFR or EGFRvIII signaling are very important in CSC and could therefore be considered for dual targeted therapy for treatment of CSCs in patients. Why EGFR- and EGFRvIII-expressing cells and tumors are more sensitive to chloroquine treatment remains matter of investigation.
Clinical efficacy of anti EGFR drugs to date has been limited by both acquired and intrinsic resistance. Cancer cells adapt rapidly to EGFR inhibition treatment, resulting in only a small success rate for EGFR inhibition as mono therapy in cancer treatment119,120 (Fig. 2B). Nevertheless, inhibition of EGFR signaling in combination with autophagy inhibition looks promising in vitro. In NSCLC cell lines with activating EGFR mutation (exon 19 deletion) erlotinib induces both apoptosis and autophagy. Inhibition of autophagy can further enhance sensitivity to erlotinib in these NSCLC cells, suggesting that autophagy serves as a protective mechanism.121 Moreover, wild-type EGFR-expressing NSCLC cells’ resistance to erlotinib can be abrogated through autophagy inhibition.122 Furthermore, ZD6474, a small molecule inhibitor of VEGFR, EGFR, and RET induces apoptosis in 2 glioblastoma cell lines, which can be enhanced by the inhibition of autophagy.123 These findings suggest a therapeutic opportunity for the inhibition of autophagy in combination with conventional cancer therapies.
Conclusion
Over the last decades EGFR has evolved as highly investigated target in the field of anti-cancer treatment. This has led to the development of EGFR-targeting antibodies like cetuximab or panitumumab and TKIs like gefitinib, erlotinib, and lapatinib. More recently, the potential of autophagy inhibition as therapy in cancer is being evaluated. Several reports indicate that cells and tumors with amplified or overactivated EGFR are particularly sensitive to autophagy inhibition for growth, survival, and resistance to conventional therapies. Additionally, resistance to EGFR-targeting therapies can also be reduced by autophagy-inhibition. Inhibition of autophagy may therefore provide a novel treatment opportunity for EGFR-overexpressing tumors and should be pursued clinically.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Acknowledgments
This work was financially supported by the Dutch Cancer Society (KWF Grants UM 2010-4714 and 2012-5506), foundations “STOPhersentumoren” and “Zeldzame Ziekten Fonds”.
Footnotes
Previously published online: www.landesbioscience.com/journals/cc/article/27518
References
- 1.Downward J, Yarden Y, Mayes E, Scrace G, Totty N, Stockwell P, Ullrich A, Schlessinger J, Waterfield MD. Close similarity of epidermal growth factor receptor and v-erb-B oncogene protein sequences. Nature. 1984;307:521–7. doi: 10.1038/307521a0. [DOI] [PubMed] [Google Scholar]
- 2.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–37. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
- 3.Lemmon MA, Bu Z, Ladbury JE, Zhou M, Pinchasi D, Lax I, Engelman DM, Schlessinger J. Two EGF molecules contribute additively to stabilization of the EGFR dimer. EMBO J. 1997;16:281–94. doi: 10.1093/emboj/16.2.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Linggi B, Carpenter G. ErbB receptors: new insights on mechanisms and biology. Trends Cell Biol. 2006;16:649–56. doi: 10.1016/j.tcb.2006.10.008. [DOI] [PubMed] [Google Scholar]
- 5.Perez R, Crombet T, de Leon J, Moreno E. A view on EGFR-targeted therapies from the oncogene-addiction perspective. Front Pharmacol. 2013;4:53. doi: 10.3389/fphar.2013.00053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mizushima N. The pleiotropic role of autophagy: from protein metabolism to bactericide. Cell Death Differ. 2005;12(Suppl 2):1535–41. doi: 10.1038/sj.cdd.4401728. [DOI] [PubMed] [Google Scholar]
- 7.King CR, Kraus MH, Aaronson SA. Amplification of a novel v-erbB-related gene in a human mammary carcinoma. Science. 1985;229:974–6. doi: 10.1126/science.2992089. [DOI] [PubMed] [Google Scholar]
- 8.King CR, Kraus MH, Williams LT, Merlino GT, Pastan IH, Aaronson SA. Human tumor cell lines with EGF receptor gene amplification in the absence of aberrant sized mRNAs. Nucleic Acids Res. 1985;13:8477–86. doi: 10.1093/nar/13.23.8477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Libermann TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H, Whittle N, Waterfield MD, Ullrich A, Schlessinger J. Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature. 1985;313:144–7. doi: 10.1038/313144a0. [DOI] [PubMed] [Google Scholar]
- 10.Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J Cancer. 2001;37(Suppl 4):S9–15. doi: 10.1016/S0959-8049(01)00231-3. [DOI] [PubMed] [Google Scholar]
- 11.Weinstein IB, Joe A. Oncogene addiction. Cancer Res. 2008;68:3077–80, discussion 3080. doi: 10.1158/0008-5472.CAN-07-3293. [DOI] [PubMed] [Google Scholar]
- 12.Weinstein IB, Joe AK. Mechanisms of disease: Oncogene addiction--a rationale for molecular targeting in cancer therapy. Nat Clin Pract Oncol. 2006;3:448–57. doi: 10.1038/ncponc0558. [DOI] [PubMed] [Google Scholar]
- 13.Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Verslype C, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45. doi: 10.1056/NEJMoa033025. [DOI] [PubMed] [Google Scholar]
- 14.Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins R, et al. National Cancer Institute of Canada Clinical Trials Group Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353:123–32. doi: 10.1056/NEJMoa050753. [DOI] [PubMed] [Google Scholar]
- 15.Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, Lorimer I, Zhang T, Liu N, Daneshmand M, et al. Erlotinib in lung cancer - molecular and clinical predictors of outcome. N Engl J Med. 2005;353:133–44. doi: 10.1056/NEJMoa050736. [DOI] [PubMed] [Google Scholar]
- 16.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–39. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
- 17.Taron M, Ichinose Y, Rosell R, Mok T, Massuti B, Zamora L, Mate JL, Manegold C, Ono M, Queralt C, et al. Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas. Clin Cancer Res. 2005;11:5878–85. doi: 10.1158/1078-0432.CCR-04-2618. [DOI] [PubMed] [Google Scholar]
- 18.Hirsch FR, Varella-Garcia M, Bunn PA, Jr., Di Maria MV, Veve R, Bremmes RM, Barón AE, Zeng C, Franklin WA. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol. 2003;21:3798–807. doi: 10.1200/JCO.2003.11.069. [DOI] [PubMed] [Google Scholar]
- 19.Brand TM, Iida M, Wheeler DL. Molecular mechanisms of resistance to the EGFR monoclonal antibody cetuximab. Cancer Biol Ther. 2011;11:777–92. doi: 10.4161/cbt.11.9.15050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–81. doi: 10.1038/nrc2088. [DOI] [PubMed] [Google Scholar]
- 21.Yun CH, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong KK, Meyerson M, Eck MJ. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A. 2008;105:2070–5. doi: 10.1073/pnas.0709662105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yasuda H, Kobayashi S, Costa DB. EGFR exon 20 insertion mutations in non-small-cell lung cancer: preclinical data and clinical implications. Lancet Oncol. 2012;13:e23–31. doi: 10.1016/S1470-2045(11)70129-2. [DOI] [PubMed] [Google Scholar]
- 23.Shinojima N, Tada K, Shiraishi S, Kamiryo T, Kochi M, Nakamura H, Makino K, Saya H, Hirano H, Kuratsu J, et al. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res. 2003;63:6962–70. [PubMed] [Google Scholar]
- 24.Wong AJ, Ruppert JM, Bigner SH, Grzeschik CH, Humphrey PA, Bigner DS, Vogelstein B. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc Natl Acad Sci U S A. 1992;89:2965–9. doi: 10.1073/pnas.89.7.2965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sugawa N, Ekstrand AJ, James CD, Collins VP. Identical splicing of aberrant epidermal growth factor receptor transcripts from amplified rearranged genes in human glioblastomas. Proc Natl Acad Sci U S A. 1990;87:8602–6. doi: 10.1073/pnas.87.21.8602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yamazaki H, Fukui Y, Ueyama Y, Tamaoki N, Kawamoto T, Taniguchi S, Shibuya M. Amplification of the structurally and functionally altered epidermal growth factor receptor gene (c-erbB) in human brain tumors. Mol Cell Biol. 1988;8:1816–20. doi: 10.1128/mcb.8.4.1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gan HK, Kaye AH, Luwor RB. The EGFRvIII variant in glioblastoma multiforme. J Clin Neurosci. 2009;16:748–54. doi: 10.1016/j.jocn.2008.12.005. [DOI] [PubMed] [Google Scholar]
- 28.Smilek P, Neuwirthova J, Jarkovsky J, Dusek L, Rottenberg J, Kostrica R, Srovnal J, Hajduch M, Drabek J, Klozar J. Epidermal growth factor receptor (EGFR) expression and mutations in the EGFR signaling pathway in correlation with anti-EGFR therapy in head and neck squamous cell carcinomas. Neoplasma. 2012;59:508–15. doi: 10.4149/neo_2012_065. [DOI] [PubMed] [Google Scholar]
- 29.Sasaki H, Kawano O, Endo K, Yukiue H, Yano M, Fujii Y. EGFRvIII mutation in lung cancer correlates with increased EGFR copy number. Oncol Rep. 2007;17:319–23. [PubMed] [Google Scholar]
- 30.Ji H, Zhao X, Yuza Y, Shimamura T, Li D, Protopopov A, Jung BL, McNamara K, Xia H, Glatt KA, et al. Epidermal growth factor receptor variant III mutations in lung tumorigenesis and sensitivity to tyrosine kinase inhibitors. Proc Natl Acad Sci U S A. 2006;103:7817–22. doi: 10.1073/pnas.0510284103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Del Vecchio CA, Jensen KC, Nitta RT, Shain AH, Giacomini CP, Wong AJ. Epidermal growth factor receptor variant III contributes to cancer stem cell phenotypes in invasive breast carcinoma. Cancer Res. 2012;72:2657–71. doi: 10.1158/0008-5472.CAN-11-2656. [DOI] [PubMed] [Google Scholar]
- 32.Moscatello DK, Holgado-Madruga M, Godwin AK, Ramirez G, Gunn G, Zoltick PW, Biegel JA, Hayes RL, Wong AJ. Frequent expression of a mutant epidermal growth factor receptor in multiple human tumors. Cancer Res. 1995;55:5536–9. [PubMed] [Google Scholar]
- 33.Weppler SA, Li Y, Dubois L, Lieuwes N, Jutten B, Lambin P, Wouters BG, Lammering G. Expression of EGFR variant vIII promotes both radiation resistance and hypoxia tolerance. Radiother Oncol. 2007;83:333–9. doi: 10.1016/j.radonc.2007.04.025. [DOI] [PubMed] [Google Scholar]
- 34.Golding SE, Morgan RN, Adams BR, Hawkins AJ, Povirk LF, Valerie K. Pro-survival AKT and ERK signaling from EGFR and mutant EGFRvIII enhances DNA double-strand break repair in human glioma cells. Cancer Biol Ther. 2009;8:730–8. doi: 10.4161/cbt.8.8.7927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Learn CA, Hartzell TL, Wikstrand CJ, Archer GE, Rich JN, Friedman AH, Friedman HS, Bigner DD, Sampson JH. Resistance to tyrosine kinase inhibition by mutant epidermal growth factor receptor variant III contributes to the neoplastic phenotype of glioblastoma multiforme. Clin Cancer Res. 2004;10:3216–24. doi: 10.1158/1078-0432.CCR-03-0521. [DOI] [PubMed] [Google Scholar]
- 36.Hockel M, Schlenger K, Aral B, Mitze M, Schaffer U, Vaupel P. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996;56:4509–15. [PubMed] [Google Scholar]
- 37.Nordsmark M, Bentzen SM, Rudat V, Brizel D, Lartigau E, Stadler P, Becker A, Adam M, Molls M, Dunst J, et al. Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiother Oncol. 2005;77:18–24. doi: 10.1016/j.radonc.2005.06.038. [DOI] [PubMed] [Google Scholar]
- 38.Wouters BG, van den Beucken T, Magagnin MG, Lambin P, Koumenis C. Targeting hypoxia tolerance in cancer. Drug Resist Updat. 2004;7:25–40. doi: 10.1016/j.drup.2003.12.004. [DOI] [PubMed] [Google Scholar]
- 39.Theys J, Jutten B, Dubois L, Rouschop KM, Chiu RK, Li Y, Paesmans K, Lambin P, Lammering G, Wouters BG. The deletion mutant EGFRvIII significantly contributes to stress resistance typical for the tumour microenvironment. Radiother Oncol. 2009;92:399–404. doi: 10.1016/j.radonc.2009.06.017. [DOI] [PubMed] [Google Scholar]
- 40.Bigner SH, Humphrey PA, Wong AJ, Vogelstein B, Mark J, Friedman HS, Bigner DD. Characterization of the epidermal growth factor receptor in human glioma cell lines and xenografts. Cancer Res. 1990;50:8017–22. [PubMed] [Google Scholar]
- 41.Pandita A, Aldape KD, Zadeh G, Guha A, James CD. Contrasting in vivo and in vitro fates of glioblastoma cell subpopulations with amplified EGFR. Genes Chromosomes Cancer. 2004;39:29–36. doi: 10.1002/gcc.10300. [DOI] [PubMed] [Google Scholar]
- 42.Stockhausen MT, Broholm H, Villingshøj M, Kirchhoff M, Gerdes T, Kristoffersen K, Kosteljanetz M, Spang-Thomsen M, Poulsen HS. Maintenance of EGFR and EGFRvIII expressions in an in vivo and in vitro model of human glioblastoma multiforme. Exp Cell Res. 2011;317:1513–26. doi: 10.1016/j.yexcr.2011.04.001. [DOI] [PubMed] [Google Scholar]
- 43.Pelloski CE, Ballman KV, Furth AF, Zhang L, Lin E, Sulman EP, Bhat K, McDonald JM, Yung WK, Colman H, et al. Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. J Clin Oncol. 2007;25:2288–94. doi: 10.1200/JCO.2006.08.0705. [DOI] [PubMed] [Google Scholar]
- 44.Montano N, Cenci T, Martini M, D’Alessandris QG, Pelacchi F, Ricci-Vitiani L, Maira G, De Maria R, Larocca LM, Pallini R. Expression of EGFRvIII in glioblastoma: prognostic significance revisited. Neoplasia. 2011;13:1113–21. doi: 10.1593/neo.111338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mazzoleni S, Politi LS, Pala M, Cominelli M, Franzin A, Sergi Sergi L, Falini A, De Palma M, Bulfone A, Poliani PL, et al. Epidermal growth factor receptor expression identifies functionally and molecularly distinct tumor-initiating cells in human glioblastoma multiforme and is required for gliomagenesis. Cancer Res. 2010;70:7500–13. doi: 10.1158/0008-5472.CAN-10-2353. [DOI] [PubMed] [Google Scholar]
- 46.Martinez R, Rohde V, Schackert G. Different molecular patterns in glioblastoma multiforme subtypes upon recurrence. J Neurooncol. 2010;96:321–9. doi: 10.1007/s11060-009-9967-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kelly JJ, Stechishin O, Chojnacki A, Lun X, Sun B, Senger DL, Forsyth P, Auer RN, Dunn JF, Cairncross JG, et al. Proliferation of human glioblastoma stem cells occurs independently of exogenous mitogens. Stem Cells. 2009;27:1722–33. doi: 10.1002/stem.98. [DOI] [PubMed] [Google Scholar]
- 48.Griffero F, Daga A, Marubbi D, Capra MC, Melotti A, Pattarozzi A, Gatti M, Bajetto A, Porcile C, Barbieri F, et al. Different response of human glioma tumor-initiating cells to epidermal growth factor receptor kinase inhibitors. J Biol Chem. 2009;284:7138–48. doi: 10.1074/jbc.M807111200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Soeda A, Inagaki A, Oka N, Ikegame Y, Aoki H, Yoshimura S, Nakashima S, Kunisada T, Iwama T. Epidermal growth factor plays a crucial role in mitogenic regulation of human brain tumor stem cells. J Biol Chem. 2008;283:10958–66. doi: 10.1074/jbc.M704205200. [DOI] [PubMed] [Google Scholar]
- 50.Ma L, Zhang G, Miao XB, Deng XB, Wu Y, Liu Y, Jin ZR, Li XQ, Liu QZ, Sun DX, et al. Cancer stem-like cell properties are regulated by EGFR/AKT/β-catenin signaling and preferentially inhibited by gefitinib in nasopharyngeal carcinoma. FEBS J. 2013;280:2027–41. doi: 10.1111/febs.12226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Clark PA, Iida M, Treisman DM, Kalluri H, Ezhilan S, Zorniak M, Wheeler DL, Kuo JS. Activation of multiple ERBB family receptors mediates glioblastoma cancer stem-like cell resistance to EGFR-targeted inhibition. Neoplasia. 2012;14:420–8. doi: 10.1596/neo.12432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Li G, Mitra SS, Monje M, Henrich KN, Bangs CD, Nitta RT, Wong AJ. Expression of epidermal growth factor variant III (EGFRvIII) in pediatric diffuse intrinsic pontine gliomas. J Neurooncol. 2012;108:395–402. doi: 10.1007/s11060-012-0842-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liu XJ, Wu WT, Wu WH, Yin F, Ma SH, Qin JZ, Liu XX, Liu YN, Zhang XY, Li P, et al. A minority subpopulation of CD133(+) /EGFRvIII(+) /EGFR(-) cells acquires stemness and contributes to gefitinib resistance. CNS Neurosci Ther. 2013;19:494–502. doi: 10.1111/cns.12092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kolch W, Pitt A. Functional proteomics to dissect tyrosine kinase signalling pathways in cancer. Nat Rev Cancer. 2010;10:618–29. doi: 10.1038/nrc2900. [DOI] [PubMed] [Google Scholar]
- 55.Edinger AL, Thompson CB. Defective autophagy leads to cancer. Cancer Cell. 2003;4:422–4. doi: 10.1016/S1535-6108(03)00306-4. [DOI] [PubMed] [Google Scholar]
- 56.Levine B. Eating oneself and uninvited guests: autophagy-related pathways in cellular defense. Cell. 2005;120:159–62. doi: 10.1016/j.cell.2005.01.005. [DOI] [PubMed] [Google Scholar]
- 57.Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol. 2005;169:425–34. doi: 10.1083/jcb.200412022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature. 2004;432:1032–6. doi: 10.1038/nature03029. [DOI] [PubMed] [Google Scholar]
- 59.Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gélinas C, Fan Y, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10:51–64. doi: 10.1016/j.ccr.2006.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rouschop KM, van den Beucken T, Dubois L, Niessen H, Bussink J, Savelkouls K, Keulers T, Mujcic H, Landuyt W, Voncken JW, et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Invest. 2010;120:127–41. doi: 10.1172/JCI40027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Jutten B, Keulers TG, Schaaf MB, Savelkouls K, Theys J, Span PN, Vooijs MA, Bussink J, Rouschop KM. EGFR overexpressing cells and tumors are dependent on autophagy for growth and survival. Radiother Oncol. 2013;108:479–83. doi: 10.1016/j.radonc.2013.06.033. [DOI] [PubMed] [Google Scholar]
- 62.Wei Y, Zou Z, Becker N, Anderson M, Sumpter R, Xiao G, Kinch L, Koduru P, Christudass CS, Veltri RW, et al. EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell. 2013;154:1269–84. doi: 10.1016/j.cell.2013.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Weihua Z, Tsan R, Huang WC, Wu Q, Chiu CH, Fidler IJ, Hung MC. Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell. 2008;13:385–93. doi: 10.1016/j.ccr.2008.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li X, Fan Z. The epidermal growth factor receptor antibody cetuximab induces autophagy in cancer cells by downregulating HIF-1alpha and Bcl-2 and activating the beclin 1/hVps34 complex. Cancer Res. 2010;70:5942–52. doi: 10.1158/0008-5472.CAN-10-0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Han W, Pan H, Chen Y, Sun J, Wang Y, Li J, Ge W, Feng L, Lin X, Wang X, et al. EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells. PLoS One. 2011;6:e18691. doi: 10.1371/journal.pone.0018691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Li X, Lu Y, Pan T, Fan Z. Roles of autophagy in cetuximab-mediated cancer therapy against EGFR. Autophagy. 2010;6:1066–77. doi: 10.4161/auto.6.8.13366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wolf-Yadlin A, Hautaniemi S, Lauffenburger DA, White FM. Multiple reaction monitoring for robust quantitative proteomic analysis of cellular signaling networks. Proc Natl Acad Sci U S A. 2007;104:5860–5. doi: 10.1073/pnas.0608638104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Huang PH, Mukasa A, Bonavia R, Flynn RA, Brewer ZE, Cavenee WK, Furnari FB, White FM. Quantitative analysis of EGFRvIII cellular signaling networks reveals a combinatorial therapeutic strategy for glioblastoma. Proc Natl Acad Sci U S A. 2007;104:12867–72. doi: 10.1073/pnas.0705158104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Moscatello DK, Holgado-Madruga M, Emlet DR, Montgomery RB, Wong AJ. Constitutive activation of phosphatidylinositol 3-kinase by a naturally occurring mutant epidermal growth factor receptor. J Biol Chem. 1998;273:200–6. doi: 10.1074/jbc.273.1.200. [DOI] [PubMed] [Google Scholar]
- 70.Alers S, Löffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32:2–11. doi: 10.1128/MCB.06159-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132–41. doi: 10.1038/ncb2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y, Iemura S, Natsume T, Takehana K, Yamada N, et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20:1981–91. doi: 10.1091/mbc.E08-12-1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jung CH, Seo M, Otto NM, Kim DH. ULK1 inhibits the kinase activity of mTORC1 and cell proliferation. Autophagy. 2011;7:1212–21. doi: 10.4161/auto.7.10.16660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Pike LR, Singleton DC, Buffa F, Abramczyk O, Phadwal K, Li JL, Simon AK, Murray JT, Harris AL. Transcriptional up-regulation of ULK1 by ATF4 contributes to cancer cell survival. Biochem J. 2013;449:389–400. doi: 10.1042/BJ20120972. [DOI] [PubMed] [Google Scholar]
- 75.Schaaf MB, Cojocari D, Keulers TG, Jutten B, Starmans MH, de Jong MC, Begg AC, Savelkouls KG, Bussink J, Vooijs M, et al. The autophagy associated gene, ULK1, promotes tolerance to chronic and acute hypoxia. Radiother Oncol. 2013;108:529–34. doi: 10.1016/j.radonc.2013.06.015. [DOI] [PubMed] [Google Scholar]
- 76.Schubbert S, Shannon K, Bollag G. Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer. 2007;7:295–308. doi: 10.1038/nrc2109. [DOI] [PubMed] [Google Scholar]
- 77.Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzunbas G, Kamphorst JJ, Chen G, Lemons JM, Karantza V, et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011;25:460–70. doi: 10.1101/gad.2016311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kim MJ, Woo SJ, Yoon CH, Lee JS, An S, Choi YH, Hwang SG, Yoon G, Lee SJ. Involvement of autophagy in oncogenic K-Ras-induced malignant cell transformation. J Biol Chem. 2011;286:12924–32. doi: 10.1074/jbc.M110.138958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lock R, Roy S, Kenific CM, Su JS, Salas E, Ronen SM, Debnath J. Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation. Mol Biol Cell. 2011;22:165–78. doi: 10.1091/mbc.E10-06-0500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Alexandrova AY, Kopnin PB, Vasiliev JM, Kopnin BP. ROS up-regulation mediates Ras-induced changes of cell morphology and motility. Exp Cell Res. 2006;312:2066–73. doi: 10.1016/j.yexcr.2006.03.004. [DOI] [PubMed] [Google Scholar]
- 81.Kopnin PB, Agapova LS, Kopnin BP, Chumakov PM. Repression of sestrin family genes contributes to oncogenic Ras-induced reactive oxygen species up-regulation and genetic instability. Cancer Res. 2007;67:4671–8. doi: 10.1158/0008-5472.CAN-06-2466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ren Z, Mao X, Mertens C, Krishnaraj R, Qin J, Mandal PK, Romanowski MJ, McMurray JS, Chen X. Crystal structure of unphosphorylated STAT3 core fragment. Biochem Biophys Res Commun. 2008;374:1–5. doi: 10.1016/j.bbrc.2008.04.049. [DOI] [PubMed] [Google Scholar]
- 83.Zhong Z, Wen Z, Darnell JE., Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264:95–8. doi: 10.1126/science.8140422. [DOI] [PubMed] [Google Scholar]
- 84.Boulton TG, Zhong Z, Wen Z, Darnell JE, Jr., Stahl N, Yancopoulos GD. STAT3 activation by cytokines utilizing gp130 and related transducers involves a secondary modification requiring an H7-sensitive kinase. Proc Natl Acad Sci U S A. 1995;92:6915–9. doi: 10.1073/pnas.92.15.6915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Levy DE, Darnell JE., Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–62. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
- 86.Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H, Souquere S, Mariño G, Lachkar S, Senovilla L, et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol Cell. 2012;48:667–80. doi: 10.1016/j.molcel.2012.09.013. [DOI] [PubMed] [Google Scholar]
- 87.Niso-Santano M, Shen S, Adjemian S, Malik SA, Mariño G, Lachkar S, Senovilla L, Kepp O, Galluzzi L, Maiuri MC, et al. Direct interaction between STAT3 and EIF2AK2 controls fatty acid-induced autophagy. Autophagy. 2013;9:415–7. doi: 10.4161/auto.22910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Schwarze MM, Hawley RG. Prevention of myeloma cell apoptosis by ectopic bcl-2 expression or interleukin 6-mediated up-regulation of bcl-xL. Cancer Res. 1995;55:2262–5. [PubMed] [Google Scholar]
- 89.Tai WT, Shiau CW, Chen HL, Liu CY, Lin CS, Cheng AL, Chen PJ, Chen KF. Mcl-1-dependent activation of Beclin 1 mediates autophagic cell death induced by sorafenib and SC-59 in hepatocellular carcinoma cells. Cell Death Dis. 2013;4:e485. doi: 10.1038/cddis.2013.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N, Maejima I, Shirahama-Noda K, Ichimura T, Isobe T, et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 2009;11:385–96. doi: 10.1038/ncb1846. [DOI] [PubMed] [Google Scholar]
- 91.Erlich S, Mizrachy L, Segev O, Lindenboim L, Zmira O, Adi-Harel S, Hirsch JA, Stein R, Pinkas-Kramarski R. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy. 2007;3:561–8. doi: 10.4161/auto.4713. [DOI] [PubMed] [Google Scholar]
- 92.Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci U S A. 2003;100:15077–82. doi: 10.1073/pnas.2436255100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Aita VM, Liang XH, Murty VV, Pincus DL, Yu W, Cayanis E, Kalachikov S, Gilliam TC, Levine B. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics. 1999;59:59–65. doi: 10.1006/geno.1999.5851. [DOI] [PubMed] [Google Scholar]
- 94.Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 1999;402:672–6. doi: 10.1038/45257. [DOI] [PubMed] [Google Scholar]
- 95.Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 2003;112:1809–20. doi: 10.1172/JCI20039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Itakura E, Kishi C, Inoue K, Mizushima N. Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol Biol Cell. 2008;19:5360–72. doi: 10.1091/mbc.E08-01-0080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–39. doi: 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
- 98.Guo D, Hildebrandt IJ, Prins RM, Soto H, Mazzotta MM, Dang J, Czernin J, Shyy JY, Watson AD, Phelps M, et al. The AMPK agonist AICAR inhibits the growth of EGFRvIII-expressing glioblastomas by inhibiting lipogenesis. Proc Natl Acad Sci U S A. 2009;106:12932–7. doi: 10.1073/pnas.0906606106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Babic I, Anderson ES, Tanaka K, Guo D, Masui K, Li B, Zhu S, Gu Y, Villa GR, Akhavan D, et al. EGFR mutation-induced alternative splicing of Max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 2013;17:1000–8. doi: 10.1016/j.cmet.2013.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Rasbach KA, Schnellmann RG. Signaling of mitochondrial biogenesis following oxidant injury. J Biol Chem. 2007;282:2355–62. doi: 10.1128/MCB.24.16.7059-7071.2004. [DOI] [PubMed] [Google Scholar]
- 101.Boerner JL, Demory ML, Silva C, Parsons SJ. Phosphorylation of Y845 on the epidermal growth factor receptor mediates binding to the mitochondrial protein cytochrome c oxidase subunit II. Mol Cell Biol. 2004;24:7059–71. doi: 10.1128/MCB.24.16.7059-7071.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Demory ML, Boerner JL, Davidson R, Faust W, Miyake T, Lee I, Hüttemann M, Douglas R, Haddad G, Parsons SJ. Epidermal growth factor receptor translocation to the mitochondria: regulation and effect. J Biol Chem. 2009;284:36592–604. doi: 10.1074/jbc.M109.000760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Cao X, Zhu H, Ali-Osman F, Lo HW. EGFR and EGFRvIII undergo stress- and EGFR kinase inhibitor-induced mitochondrial translocalization: a potential mechanism of EGFR-driven antagonism of apoptosis. Mol Cancer. 2011;10:26. doi: 10.1186/1476-4598-10-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Cvrljevic AN, Akhavan D, Wu M, Martinello P, Furnari FB, Johnston AJ, Guo D, Pike L, Cavenee WK, Scott AM, et al. Activation of Src induces mitochondrial localisation of de2-7EGFR (EGFRvIII) in glioma cells: implications for glucose metabolism. J Cell Sci. 2011;124:2938–50. doi: 10.1242/jcs.083295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Schmidt-Ullrich RK, Mikkelsen RB, Dent P, Todd DG, Valerie K, Kavanagh BD, Contessa JN, Rorrer WK, Chen PB. Radiation-induced proliferation of the human A431 squamous carcinoma cells is dependent on EGFR tyrosine phosphorylation. Oncogene. 1997;15:1191–7. doi: 10.1038/sj.onc.1201275. [DOI] [PubMed] [Google Scholar]
- 106.Dittmann K, Mayer C, Fehrenbacher B, Schaller M, Kehlbach R, Rodemann HP. Nuclear EGFR shuttling induced by ionizing radiation is regulated by phosphorylation at residue Thr654. FEBS Lett. 2010;584:3878–84. doi: 10.1016/j.febslet.2010.08.005. [DOI] [PubMed] [Google Scholar]
- 107.Harris AL. Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2:38–47. doi: 10.1038/nrc704. [DOI] [PubMed] [Google Scholar]
- 108.Rouschop KM, Dubois L, Schaaf MB, van den Beucken T, Lieuwes N, Keulers TG, Savelkouls KG, Bussink J, van der Kogel AJ, Koritzinsky M, et al. Deregulation of cap-dependent mRNA translation increases tumour radiosensitivity through reduction of the hypoxic fraction. Radiother Oncol. 2011;99:385–91. doi: 10.1016/j.radonc.2011.05.047. [DOI] [PubMed] [Google Scholar]
- 109.Rouschop KM, Dubois LJ, Keulers TG, van den Beucken T, Lambin P, Bussink J, van der Kogel AJ, Koritzinsky M, Wouters BG. PERK/eIF2α signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS. Proc Natl Acad Sci U S A. 2013;110:4622–7. doi: 10.1073/pnas.1210633110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck–a systematic review and meta-analysis. Radiother Oncol. 2011;100:22–32. doi: 10.1016/j.radonc.2011.03.004. [DOI] [PubMed] [Google Scholar]
- 111.Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouysségur J, Mazure NM. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol. 2009;29:2570–81. doi: 10.1128/MCB.00166-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290:1717–21. doi: 10.1126/science.290.5497.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, Gonzalez FJ, Semenza GL. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem. 2008;283:10892–903. doi: 10.1074/jbc.M800102200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 114.Papandreou I, Lim AL, Laderoute K, Denko NC. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L. Cell Death Differ. 2008;15:1572–81. doi: 10.1038/cdd.2008.84. [DOI] [PubMed] [Google Scholar]
- 115.Rouschop KM, Ramaekers CH, Schaaf MB, Keulers TG, Savelkouls KG, Lambin P, Koritzinsky M, Wouters BG. Autophagy is required during cycling hypoxia to lower production of reactive oxygen species. Radiother Oncol. 2009;92:411–6. doi: 10.1016/j.radonc.2009.06.029. [DOI] [PubMed] [Google Scholar]
- 116.Stegeman H, Kaanders JH, van der Kogel AJ, Iida M, Wheeler DL, Span PN, Bussink J. Predictive value of hypoxia, proliferation and tyrosine kinase receptors for EGFR-inhibition and radiotherapy sensitivity in head and neck cancer models. Radiother Oncol. 2013;106:383–9. doi: 10.1016/j.radonc.2013.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gong C, Bauvy C, Tonelli G, Yue W, Deloménie C, Nicolas V, Zhu Y, Domergue V, Marin-Esteban V, Tharinger H, et al. Beclin 1 and autophagy are required for the tumorigenicity of breast cancer stem-like/progenitor cells. Oncogene. 2013;32:2261–72, 1-11. doi: 10.1038/onc.2012.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Galavotti S, Bartesaghi S, Faccenda D, Shaked-Rabi M, Sanzone S, McEvoy A, Dinsdale D, Condorelli F, Brandner S, Campanella M, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32:699–712. doi: 10.1038/onc.2012.111. [DOI] [PubMed] [Google Scholar]
- 119.Camp ER, Summy J, Bauer TW, Liu W, Gallick GE, Ellis LM. Molecular mechanisms of resistance to therapies targeting the epidermal growth factor receptor. Clin Cancer Res. 2005;11:397–405. [PubMed] [Google Scholar]
- 120.Sequist LV, Lynch TJ. EGFR tyrosine kinase inhibitors in lung cancer: an evolving story. Annu Rev Med. 2008;59:429–42. doi: 10.1146/annurev.med.59.090506.202405. [DOI] [PubMed] [Google Scholar]
- 121.Li YY, Lam SK, Mak JC, Zheng CY, Ho JC. Erlotinib-induced autophagy in epidermal growth factor receptor mutated non-small cell lung cancer. Lung Cancer. 2013;81:354–61. doi: 10.1016/j.lungcan.2013.05.012. [DOI] [PubMed] [Google Scholar]
- 122.Zou Y, Ling YH, Sironi J, Schwartz EL, Perez-Soler R, Piperdi B. The autophagy inhibitor chloroquine overcomes the innate resistance of wild-type EGFR non-small-cell lung cancer cells to erlotinib. J Thorac Oncol. 2013;8:693–702. doi: 10.1097/JTO.0b013e31828c7210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Shen J, Zheng H, Ruan J, Fang W, Li A, Tian G, Niu X, Luo S, Zhao P. Autophagy inhibition induces enhanced proapoptotic effects of ZD6474 in glioblastoma. Br J Cancer. 2013;109:164–71. doi: 10.1038/bjc.2013.306. [DOI] [PMC free article] [PubMed] [Google Scholar]