Journal list menu

Volume 10, Issue 4 p. 601-609
Article
Open Access

EMT is associated with, but does not drive resistance to ALK inhibitors among EML4-ALK non-small cell lung cancer

Arjan Gower

Arjan Gower

Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA

Search for more papers by this author
Wei-Hsun Hsu

Wei-Hsun Hsu

Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA

Search for more papers by this author
Shuo-Tse Hsu

Shuo-Tse Hsu

Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA

Search for more papers by this author
Yisong Wang

Yisong Wang

Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA

Search for more papers by this author
Giuseppe Giaccone

Corresponding Author

Giuseppe Giaccone

Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA

Corresponding author. Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20007, USA. Tel.: +1 202 687 7072; fax: +1 202 687 0313.Search for more papers by this author
First published: 24 November 2015
Citations: 26

Abstract

ALK gene fusion occurs in approximately 3–7% of non-small cell lung cancer (NSCLC). For patients with ALK positive NCSLC, crizotinib and ceritinib are FDA approved ALK inhibitors, however, patients inevitably acquire resistance to such therapies typically within one to two years. Interrogation of in vitro ALK-positive NSCLC cell line models of acquired resistance to first and second-generation ALK inhibitors revealed acquired epithelial-to-mesenchymal transition (EMT) mechanisms. Here we demonstrated that knockdown of upregulated mesenchymal markers in acquired resistant lines decreased the invasive and migratory capabilities of the cells, however, it did not restore sensitivity to ALK inhibitors. Removing drug for 5 weeks from H3122 cell line that acquired resistance to ceritinib restored its sensitivity to ceritinib. In addition, HSP90 inhibitors ganetespib and 17-AAG were potent in inducing cell death in cell lines resistant to crizotinib and ceritinib. Taken together, EMT does not drive resistance to ALK inhibitors and HSP90 inhibition demonstrates more efficacy when further ALK inhibition may not. This study warrants more exploration of HSP90 inhibitors for ALK-positive patients who progress on 1st and 2nd line ALK inhibitor therapy.

1 Introduction

Lung cancer is the leading cause of cancer-related mortality worldwide (Jemal et al., 2011). NSCLC accounts for approximately 85% of cases, most frequently presented as adenocarcinoma of the lung. In the past decade, the molecular classification of adenocarcinoma has evolved based on the elucidation of oncogenic drivers (Gower et al., 2014). Oncogenic gene rearrangements of anaplastic lymphoma kinase (ALK) occur in 3–7% of NSCLC; this genetic abnormality that defines the disease subset causes constitutive activation of ALK and its downstream signaling pathways including MAPK, JAK/STAT, and PI3K/AKT (Soda et al., 2007; Li et al., 2011). ALK has several fusion partners, the most common being echinoderm microtubule-associated protein-like 4 (EML4) (Hallberg and Palmer, 2013). ALK gene rearrangements are generated by a small inversion within the short arm of chromosome 2, and typically occur in young, never-smokers and are mutually exclusive of other oncogenic drivers such as EGFR and KRAS mutations (Gainor et al., 2013).

For NSCLC patients harboring ALK gene rearrangements, ALK inhibitors have shown to be clinically efficacious. Crizotinib was FDA approved in 2013 based on a superior response rate and progression-free survival (PFS) compared to standard chemotherapy (Kwak et al., 2010; Shaw et al., 2013). However, despite the clinical success of crizotinib, patients inevitably progress while on therapy usually within one to two years. Acquired resistance to crizotinib can occur via secondary mutations in ALK protein or ALK gene amplification, both of which render crizotinib ineffective and are observed in about one-third of crizotinib-resistant patients (Katayama et al., 2012; Doebele et al., 2012). Other mechanisms of resistance include activation of bypass signaling pathways such as KIT amplification, increased phospho-EGFR levels due to elevated ligand levels, activating mutations in EGFR, mutations in KRAS oncogene, and increased NRG1 levels activating HER2/3 kinases (Katayama et al., 2012; Doebele et al., 2012; Wilson et al., 2015). In addition, multiple resistance mechanisms within the same patient have been observed in the clinic: a crizotinib-resistant patient had both aberrant activation of KIT/SCF and a secondary mutation in ALK, confirming the notion that multiple mechanisms of resistance can coexist in the same patient (Katayama et al., 2012).

Patients who acquire resistance to crizotinib can be treated with a second-generation ALK inhibitor ceritinib, which has been shown to overcome several of the secondary mutations that confer resistance to crizotinib (Friboulet et al., 2014; Shaw et al., 2014). Ceritinib treatment in a subset of 80 crizotinib-resistant ALK-positive NSCLC patients resulted in a 56% ORR (Shaw et al., 2014). Some of these patients showed no detectable ALK secondary mutation or gene amplification. This suggests that crizotinib-resistant patients may still be ALK-dependent and just necessitate a more potent inhibitor than crizotinib, which is administered at sub-therapeutic level. Another explanation could be that ceritinib has off-target effects that inhibit the function of a protein that plays an essential role in tumor progression (Shaw et al., 2014). Other second-generation ALK inhibitors, such as alectinib, are under clinical investigation for ALK-positive NSCLC with promising preliminary data in both ALK-inhibitor-naïve patients and crizotinib-resistant patients (Gower et al., 2014).

It is noteworthy that acquired resistance in cell line models recapitulates the mechanisms of resistance to targeted therapies observed in the clinic (Engelman et al., 2007). In a recent study, MEK activation is shown to drive resistance to ceritinib. In a patient-derived, ceritinib-resistant cell line, MEK inhibitor treatment resensitized the cells to ceritinib (Crystal et al., 2014). Additionally, purinergic P2Y receptors have been shown to drive resistance to crizotinib and ceritinib in H3122 cells in vitro via a protein kinase C-dependent mechanism (Wilson et al., 2015).

Epithelial-mesenchymal transition (EMT) is the process in which cells undergo morphological and phenotypic changes. Epithelial cells lose their polarity and cell-to-cell junction while becoming morphologically more fibroblastic. These characteristics accompany enhanced cell motility and invasiveness in addition to conferring drug resistance (Lamouille et al., 2014). EMT has been shown to confer resistance to crizotinib in NSCLC (Kim et al., 2013). In this study, we investigate how EMT contributes increased invasiveness and metastatic potential to ALK inhibitor resistance in cell lines that harbor EML4-ALK translocation and clarify that EMT is only associated with resistance, and is not a driver of acquired resistance to first and second-generation ALK inhibitors. As an alternative therapeutic option, we show that HSP90 inhibitors can induce cell death in 1st and 2nd generation resistant cell lines that offer some translational significance.

2 Materials and methods

2.1 Cell culture

The H2228 cell line was obtained from American Type Culture Collection. The H3122 cell line was a gift from Dr. John Minna. H2228 and H3122 cells were grown in RPMI 1640. Media was supplemented with 10% fetal bovine serum (FBS) and 1× antibiotics (Gibco Life Technologies), maintained at 37 °C in 5% CO2/95% air, and passaged when cells reached 80–90% confluency.

2.2 Drugs

Alectinib (CH5424802) and ceritinib (LDK378) were purchased from Selleck Chemicals. Crizotinib (PF-2341066) was purchased from ChemieTek. NMS-E628 (NMS; RXDX-101) was obtained from Nerviano Medical Sciences (NCI MTA#31471-11). HSP90 inhibitors 17-AAG (17-allylamino-17demothoxygeldanamycin) and ganetespib were purchased Synta Pharmaceuticals. Drugs were dissolved in DMSO, aliquoted, and stored at −80 °C, and diluted in fresh medium before use. The final DMSO concentration in medium in all experiments was <0.1%.

2.3 Generation of ALK inhibitor resistance in H2228 and H3122 cells

H2228 and H3122 resistant lines were generated by continuous exposure to increasing concentrations of specific ALK inhibitor. H2228 cells and H3122 cells were exposed up to 2 μM and 3 μM NMS, respectively, and designated H2228 NMS_R(2) and H3122 NMS_R. H3122 was exposed up to 3 μM and 4 μM of crizotinib and ceritinib and designated H3122 Criz_R and H3122 LDK_R. Established resistant cell lines were maintained in medium containing maximal dose of ALK inhibitor to maintain selective pressure for ALK inhibitor resistance. In all experiments involving resistant cell lines, cells were seeded without relevant drug to eliminate the effect of drug interaction.

2.4 Western blot analysis

Cells were seeded in 6-well plates at a density of 5 × 105 cells/well in drug-free media. The next morning, cell lysates were extracted using RIPA buffer (Sigma–Aldrich) containing Halt protease and phosphatase inhibitor cocktail (Thermo-Scientific) and clarified by centrifugation. Protein concentrations were quantified using Pierce BCA protein assay (Thermo-Scientific), and equal amounts of protein was separated by 4–20% SDS-PAGE, transferred onto PVDF membrane (Bio-Rad) for protein blot analysis, blocked with 5% non-fat milk in Tris-buffered saline with 0.1% Tween-20, and incubated with primary antibody overnight at 4 °C. Following incubation with appropriate horseradish peroxidase-conjugated secondary antibody (Bio-Rad) in blocking buffer, protein expression was detected using SuperSignal West Pico Chemiluminescent Substrate (Thermo-Scientific). Primary antibodies to phospho-ALK (Y1078), total ALK, E-cadherin, phospho-AXL (Y779), total AXL, phospho-ERK (T202/Y204), total ERK, and vimentin were purchased from Cell Signaling Technology. N-cadherin was purchased from BD Pharmingen. Primary antibody to twist was purchased from Santa Cruz Biotechnologies. β-actin or α-tubulin was used as a loading control.

2.5 Cell viability assays

Cells were seeded overnight at a density of 3000 cells per well in 96-well plates in RPMI 1640 containing 10% FBS. The next day, cells were treated with relevant kinase inhibitors for 72 h. Viable cell numbers were determined using CellTiterGlo (Promega) according to manufacturer's protocol. Each assay consisted of triplicates and was repeated at least twice. Data was expressed as percentages of control cells, calculated from the absorbance corrected for media-only background.

2.6 Cell migration and invasion assay

Migration and invasion of cells were assessed using transwells (12-well inserts; pore size 8uM; Millipore). 2 × 105 cells in serum-free medium were plated in the top chamber of the transwell and the lower chamber was filled with medium supplemented with 10% FBS as a chemoattractant. After 24 h of incubation at 37 °C, cells were fixed with 4% paraformaldehyde and stained with 0.2% crystal violet. Non-migrating cells were removed using cotton swabs, and cells that migrated to the lower surface of the filter were counted using a light microscope. The invasion assay was conducted as described above except transwell inserts were coated with Matrigel and incubated at room temperature for 30 min prior to seeding cells.

2.7 Transfection of siRNA

Small interfering RNA (siRNA) oligonucleotides targeting AXL, Twist1, and vimentin were obtained from Dharmacon. Transfection of siRNA was performed using Lipofectamine RNAiMAX (Life Technologies) in accordance with manufacturer's protocol. Target gene expression was analyzed using western blot after 48 or 72 h of incubation. For cell viability assays, 3000 cells/well were seeded onto 96 well plate on day 0, transfected with siRNA on day 1, and treated with respective drug on day 2 for 72 h.

3 Results

3.1 Generation of H3122 and H2228 cell lines resistant to ALK inhibitors

To investigate the mechanisms of acquired resistance to ALK inhibitors, H3122 and H2228 cell lines which harbor EML4-ALK rearrangement (variant 1 and 3, respectively) were selected and shown to be sensitive to 1st generation ALK inhibitors, crizotinib and NMS (Figure 1A). The 2nd generation ALK inhibitors, ceritinib and alectinib, were superior to the first 1st generation inhibitors in inducing cell death in H3122 cells. Therefore, these cell lines have clinical relevance for determining the mechanisms of resistance. Of note, NMS (renamed RXDX-101) was initially developed as an ALK inhibitor, but is also highly potent to Trk and ROS1 rearrangements, which make up two other molecularly based subtypes of NSCLC (Gower et al., 2014; De Braud et al., 2014; Vaishnavi et al., 2013). RXDX-101 is currently under clinical investigation in patients with solid tumors with relevant genetic alterations (NCT #02097810).

Details are in the caption following the image
Generation of acquired resistance to ALK inhibitors in EML4-ALK positive cell lines. (A) H2228 and H3122 cell lines are sensitive to ALK inhibition, as measured by CellTiterGlo. The 2nd generation ALK inhibitors (right) are more potent than the 1st generation inhibitors (left) in H3122. H3122 NMS_R and H2228 NMS_R are (B) resistant to NMS with IC50>2 μM and (C) exhibit fibroblastic morphology consistent with mesenchymal characteristics. Scale bar is 400 μM.

H3122 and H2228 cells were treated with continuous, increasing concentration of ALK inhibitors until they were growing at a concentration significantly above their IC50. H3122 NMS_R and H2228 NMS_R cell lines both had IC50 values greater than 2 μM (Figure 1B). These resistant cell lines showed dramatic changes to their morphology compared to parental H3122 and H2228 cell lines (Figure 1C). Both resistant cell lines underwent a morphological change consistent with epithelial-to-mesenchymal transition (EMT), a developmental process in which cells change polarity, reorganize their cytoskeleton, and acquire migratory and invasive properties to contribute towards cancer metastasis (Lamouille et al., 2014). H2228 NMS_R cells changed from a polarized shape to a fibroblastic morphology, consistent with EMT. On the other hand, the selective pressure of NMS drug induced a portion, but not all, of H3122 NMS_R cells to change morphologically. Of note, H3122 NMS_R and H2228 NMS_R did not reveal any secondary ALK mutations that can confer resistance to ALK inhibitors (data not shown). Additionally, H3122 NMS_R was cross-resistant to ceritinib and alectinib (Supplementary Figure S1). It is noteworthy that acquired resistance to crizotinib in H3122 (H3122 Criz_R) and to NMS in H2228 (H2228 NMS_R2) showed upregulation of heregulin (Supplementary Figure S2), a ligand that activates the HER family of receptor tyrosine kinase. The resistant lines can acquire resistance through various bypass resistance mechanisms and this data corroborates a previously published paper (Wilson et al., 2015).

3.2 Mechanisms of acquired resistance to 1st generation ALK inhibitors

EMT-associated invasive and migratory potential of H2228 NMS_R and H3122 NMS_R cell lines were assessed. Both resistant lines proved to have significantly increased invasive and migratory capabilities compared to parental control cells (Figure 2A,B). To reveal the potential mechanisms of acquired resistance to 1st generation ALK inhibitors, a series of EMT-related proteins such as E-cadherin, vimentin, twist, and AXL were examined (Figure 2C). Both resistant cell lines had downregulation of E-cadherin compared to parental lines, with H2228 NMS_R completely losing expression of E-cadherin, consistent with the extensive mesenchymal morphology. H2228 NMS_R also exhibited dramatic upregulation of vimentin, an important marker of EMT, whereas H3122 NMS_R has moderate increase of vimentin expression. H3122 NMS_R had significant upregulation of AXL kinase, and pAXL indicating activation of AXL. Additionally, H3122 NMS_R had upregulation of master regulating transcription factor, twist, which likely contributed towards the changes in gene expression that repress the epithelial phenotype and activate the mesenchymal phenotype (Lamouille et al., 2014). It is noteworthy that ZEB, Slug, and Snail transcription factors were examined, however, there was no detectable levels of protein. Collectively, these findings indicated that the mechanisms of resistance to 1st generation ALK inhibitors were associated with EMT, however, the mechanism through which EMT occurs is different and is cell-line dependent.

Details are in the caption following the image
Characterization of ALK-positive NSCLC cell models with acquired resistance to 1st generation ALK inhibitor NMS. H3122 NMS_R and H2228 NMS_R have (A,B) increased invasive and migratory capabilities (*p < 0.05 using student's t-test) and (C) show downregulation of E-cadherin expression and upregulation of mesenchymal markers including vimentin in resistant lines compared to parental lines. It is noteworthy that only H3122 NMS_R has upregulation of twist and N-cadherin, and activation of AXL kinase compared to parental line. Red arrow represent a single migrated or invaded cell.

3.3 Knockdown of mesenchymal markers reduces invasive and migratory capabilities, but does not revert ALK inhibitor resistance

AXL is a receptor tyrosine kinase (RTK) that has previously been implicated in EMT, and activation of AXL can confer resistance to EGFR-targeted therapies in NSCLC (Kim et al., 2013; Vuoriluoto et al., 2011; Zhang et al., 2012). Twist transcription factor has been known to have a key role in EMT progression, specifically its ability to repress E-cadherin in cancer cells (Lamouille et al., 2014). E-cadherin is critical in maintaining the cell-to-cell contact in epithelial cells, and decreased expression of E-cadherin is necessary in the context of EMT. In H3122 NMS_R, AXL kinase was activated and twist was upregulated. To determine if modulation of these mesenchymal markers could restore sensitivity to ALK inhibitors, H3122 NMS_R was treated with siRNA specific for AXL or twist. Genetic knockdown of AXL using two different targeted siRNA sequences at 10 nM caused a loss of N-cadherin, which was acquired during EMT (Figure 3A). In addition, reduction in MAPK p38 was observed, a kinase that plays a role in invasion and migration. Although there were no changes to vimentin or twist following AXL siRNA treatment, the invasive and migratory capabilities of H3122 NMS_R were significantly reduced (Figure 3B,C). Knockdown of twist in H3122 NMS_R restored E-cadherin expression (Figure 3D), and also decreased its invasive and migratory capabilities (Figure 3E,F). Similarly to the knockdown of AXL, twist knockdown did not affect AXL or vimentin protein expression. Knockdown of AXL and twist in H3122 NMS_R (Figure 3G,H; Supplementary Figure S3), and vimentin in H3122 LDK_R (Supplementary Figure S5) did not restore sensitivity to ALK inhibitors. Thus, attempting to revert the resistant cell phenotype back to its epithelial state by genetic knockdown of upregulated mesenchymal markers suggest that EMT was involved in the physiological process of invasion and migration, but was not driving the acquired resistance.

Details are in the caption following the image
Genetic knockdown of mesenchymal markers decreases migratory and invasive capabilities in H3122 NMS_R, but does not restore ALK inhibitor sensitivity. (A) Knockdown of AXL using 2 different siRNA at 10 nM in H3122 NMS_R reduces N-cadherin and P38 expression compared to non-targeting control (NTC). (B,C) AXL knockdown decreases migration and invasion (*p < 0.05 using student's t-test). (D) Knockdown of twist at 20 nM upregulates E-cadherin and (E,F) reduces migratory and invasive capabilities. (G) First generation ALKi NMS did not significantly induce cell death in AXL knockdown H3122 NMS_R, (H) nor did either 2nd generation ALKi ceritinib or alectinib. Genetic knockdown of twist using siRNA1 did not restore sensitivity to ALKi (data not shown).

3.4 Interrogation of EMT-associated acquired resistance to ceritinib in H3122

We generated ceritinib resistant H3122 cells by continuous exposure to increasing concentrations of the drug. When the cells acquired resistance to ceritinib, the cells showed a morphological change consistent with EMT (Supplementary Figure S4). H3122 LDK_R had a significantly higher IC50 to ceritinib than parental cells (Supplementary Figure S5) and were resistant to crizotinib as well (Supplementary Figure S6). ALK gene sequencing of H3122 LDK_R detected no mutations that confer resistance to ALK inhibitors (data not shown). H3122 LDK_R had increased invasive and migratory capabilities, and this was confirmed by blotting for EMT markers (Figure 4A,B). H3122 LDK_R expressed high levels of AXL and vimentin. Treatment with vimentin siRNA caused reduction in AXL levels in H3122 LDK_R, whereas knockdown of AXL or twist had no effect on vimentin levels in H3122 NMS_R. This suggests that AXL is functioning downstream of vimentin in mediating EMT. Similar to the genetic knockdown of AXL and twist in H3122 NMS_R cells, vimentin knockdown in H3122 LDK_R did not restore sensitivity to ceritinib, suggesting that EMT is associated with, but does not drive, the resistance to ALK inhibitors (Supplementary Figure S5B). HSP90 inhibitors have previously been shown to be potent against EML4-ALK-positive NSCLC (Normant et al., 2011; Sang et al., 2013). H3122 cell lines that were grown to be resistant to 1st and 2nd generation ALK inhibitors and had morphological and functional changes consistent with EMT were sensitive to both 17-AAG and ganetespib (Figure 4C, Supplementary Figure S7). Ganetespib was shown to be more potent than 17-AAG in inducing cell death. When H3122 LDK_R cells were grown without the presence of ceritinib for 5 weeks, H3122 LDK_R (5wks), sensitivity to ALK inhibition was restored at 100 nM (Figure 4D). It is noteworthy that H3122 LDK_R cells grown without drug for up to 3 weeks did not restore sensitivity (Supplementary Figure S8).

Details are in the caption following the image
Acquired resistance to ceritinib in H3122 and treatments to induce cell death in H3122 LDK_R. (A) H3122 LDK_R show increased invasive and migratory capabilities compared to parental H3122, consistent with (B) loss of E-cadherin and increased mesenchymal markers including vimentin and AXL. Knockdown of vimentin decreased AXL expression in H3122 LDK_R. (C) Resistant cell lines are sensitive to the HSP90i ganetespib, and (D) restore sensitivity to ceritinib after passaging cells in drug-free media for 5 weeks (*p < 0.05 using student's t-test). Experiments were performed in triplicate with average and SD shown.

4 Discussion

Despite the recent improvements in the treatment of adenocarcinoma of the lung due to the advent of molecularly targeted therapies, resistance to such therapies remains a grave clinical problem. Some ALK-positive patients exhibit de novo resistance to ALK inhibitors that are expected to achieve clinical responses based on their genetics, while other patients acquire resistance after initially responding to therapy. Acquired mechanisms of resistance to ALK inhibitors typically occur through one of two ways: genetic alteration of ALK or activation of bypass signaling pathways that can allow for tumor cells to evade cell death (Engelman et al., 2007). Induction of EMT traditionally falls into the latter category, where the transformations of morphological and phenotypic characteristics of the cancer cells induce drug resistance, invasion, and metastasis. Recently, Kim et al. (2013) have shown that EMT drives acquired resistance to crizotinib in H2228 cell line, and that transiently knocking down vimentin can restore sensitivity to ALK inhibitors. However, here we demonstrate that EMT is merely associated with resistance to ALK inhibitors, and this phenotypic change is not driving the drug resistance.

The initiation and progression of EMT involves various signaling mechanisms, which can activate EMT master regulating transcription factors (Lamouille et al., 2014). Although EMT is associated with increased migration, invasiveness, and resistance to targeted therapies (Scheel and Weinberg, 2012), this process is reversible (known as mesenchymal–epithelial transition or MET). Understanding the precise mechanism through which EMT is being initiated will help to provide a strategy to reverse the mesenchymal phenotype, and ultimately lessen the metastatic potential and invasiveness of a tumor.

Here, we describe three ALK-positive NSCLC cell lines that underwent EMT through activation of different EMT regulators. All of the resistant lines increase their invasive and migratory potential and upregulate vimentin, however only H3122 NMS_R shows upregulation of EMT markers N-cadherin and transcription factor twist. H2228 NMS_R has a loss of E-cadherin without AXL activation or twist upregulation. This demonstrates that during the process of resistance acquisition, initiation of EMT can occur through different mechanisms, which may be cell line dependent. Furthermore, a tumor cell population can acquire multiple new physiological mechanisms in response to drug pressure, as mentioned earlier in the crizotinib-resistant patient. The partial EMT phenotype of H3122 NMS_R suggests that the epithelial-like cells acquire an unknown bypass mechanism that confers resistance to ALK-targeted therapies, while some of the cells additionally acquire EMT. It is unknown whether the bypass mechanism, which confers resistance to the epithelial-like tumor cell population in H3122 NMS_R, is the same bypass mechanism as that in the mesenchymal cells. Our data here suggests that EMT is not driving the acquired resistance based on the inability to restore sensitivity to ALK inhibitors after genetic knockdown of upregulated mesenchymal markers AXL, twist, and vimentin, and that these proteins may have more of a role in the physiological process of resistance acquisition than a pathological one.

Inhibition of molecular chaperone protein HSP90 can also induce cell death in ALK-positive NSCLC models (Normant et al., 2011), and has been proven to overcome acquired resistance to ALK inhibitor crizotinib (Sang et al., 2013). Sang et al. (2013) did not explain the rationale behind why HSP90 inhibition may be effective in inducing cell death in crizotinib-resistant cells that have acquired EMT, but their results run concordant with our data. We demonstrate that HSP90 inhibitors ganetespib and 17-AAG could overcome crizotinib and ceritinib resistance in ALK-positive NSCLC, suggesting that regardless of how the resistance is occurring, whether it be through genetic alteration of ALK (Sang et al., 2013) or through bypass mechanisms including EMT in combination with other unidentified factors, HSP90 inhibitors can be a potent therapeutic alternative following acquired resistance to both 1st and 2nd generation ALK inhibitors. Our study also demonstrates that additional treatment with ALK inhibitor may overcome acquired resistance to the more potent ceritinib therapy if the patient has been off therapy and other options are exhausted. This suggests that the resistance mechanism is partially reversible and that ALK is still the oncogenic driver behind this subset of disease. This concept of a drug holiday should be further explored in a clinical setting.

Though EMT is not required for resistance, this transition may predispose patients to a more aggressive tumor phenotype following acquired resistance to ALK inhibitors. Although it is critical to understand the mechanisms of resistance to targeted therapies on a molecular level so clinicians can effectively strategize subsequent therapeutic regimens, it is also important to evade the possibility of invasion and metastasis which may occur following acquired resistance to current ALK inhibitors. Although clinically efficacious, caution should be taken when treating with ALK-targeted therapies because resistant cells may tend to become more invasive and prone to metastasis. The development of new potent drugs that will not induce EMT is ideal to evade this grave clinical issue. Finally, our data suggests that further investigation into HSP90 inhibitors in the setting of ceritinib-resistant patients is warranted.

Acknowledgments

This work was supported by the Lombardi Comprehensive Cancer Center at Georgetown University.

    Supplementary data A

    Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.molonc.2015.11.007.