Materials and Methods
Rabbit Antibodies and Human Sera
Anti-EGF VacAbs were obtained by immunizing rabbits with four injections of a human recombinant EGF (Scotia Biologics Ltd., Aberdeen, United Kingdom) combined with Montanide adjuvant (Seppic, Paris, France). Pre-immunization sera from the same animals were collected and purified to be used as control antibodies (C-Abs). Sera were purified by Melon gel and treated by caprylic acid to remove contaminants. Because the purification process dilutes the antibodies, the final preparation was re-analyzed by enzyme-linked immunosorbent assay (ELISA) revealing a titer of approximately 1:1,000; significantly below the 1:10,000 to 1:64,000 titers usually achieved in patients vaccinated with EGF. Sera from patients enrolled in the BV-NSCLC-001 clinical trial (NCT01444118) were kindly provided by Bioven Europe Ltd. The BV-NSCLC-001 is a randomized trial to study the safety and efficacy of EGF cancer vaccination in late-stage (IIIB/IV) NSCLC patients who are immunized with cyclophosphamide and the human rEGF-P64K/Montanide ISA 51 vaccine. All patients provided written, informed consent.
Materials and Cell Lines
TKIs were purchased from Selleck Chemicals (Houston, Texas), human EGF from Cell Signaling Technologies (Beverly, Massachusetts), and antibodies for Western blotting from Cell Signaling Technology or Sigma-Aldrich (St. Louis, Missouri). All tissue culture materials were obtained from Invitrogen (Paisley, Scotland, United Kingdom).
Six
EGFR-mut cell lines with different molecular alterations were used in the study (
Supplementary Table 1). NCI-H1975 cells were purchased from the American Type Culture Collection (ATCC, Manassas, Virginia), whereas PC9 cells were provided by F. Hoffman-La Roche Ltd (Basel, Switzerland) with the authorization of Dr. Mayumi Ono (Kyushu University, Fukuoka, Japan). PC9-GR2 and GR4 are gefitinib-resistant whereas PC9-OR4 and PC9-GR4AZD1 are osimertinib-resistant cells; the four lines were generated in our laboratory by exposing PC9 cells to increasing concentrations of the drug (
Supplementary Fig. 1A).
13- Jacobsen K.
- Bertran-Alamillo J.
- Molina M.A.
- et al.
Convergent Akt activation drives acquired EGFR inhibitor resistance in lung cancer.
The four PC9-derived cells over-express AXL, PC9-GR2 cells also present hepatocyte growth factor receptor (MET) activation. Cell lines were maintained in Roswell Park Memorial Institute (RPMI) medium supplemented with 10% fetal bovine serum (FBS), 50 μg/mL penicillin-streptomycin, 2 mmol/L L-glutamine, and 2.5 μmol/L gefitinib (PC9-GR2, PC9-GR4) or osimertinib (PC9-OR4, PC9-GR4AZD1) in a humidified atmosphere with 5% CO
2 at 37ºC. Cells were weekly tested for mycoplasmas and authenticated by monthly genotyping for
EGFR and tumor protein p53 (
TP53) mutations and a panel of four polymorphisms. After no more than 15 passages, cells were discarded and a new, low-passage vial was thawed.
Cell Growth, Viability, and Emergence of Resistant Assays
Cell viability was assessed by the thiazolyl blue tetrazolium bromide (MTT; Sigma) assay. Cells were seeded at 2000 (PC9), 4000 (PC9-GR4, PC9-OR4), or 6000 (H1975) per well in 96-well plates, allowed to attach for 24 hours in RPMI + 10% FBS, washed twice with phosphate-buffered saline (PBS), and treated with EGF (10 ng/mL), antibodies, EGFR TKIs, or combinations for 72 hours in RPMI + 0.5% human serum (HS). After treatment, cells were incubated with medium containing MTT (0.75 mg/mL) for 1 to 2 hours at 37ºC. Culture medium was removed; formazan crystals reabsorbed in DMSO (Merck, Darmstadt, Germany) and cell numbers were estimated using an Anthos 2020 microplate reader (Biochrom Ltd., Cambridge, United Kingdom). Data were derived from at least three independent experiments. To study the acquisition of resistance to TKIs, we seeded 300 PC9 cells per well in 96-well plates, using two plates per treatment (120 wells). Cells were allowed to attach and the treatments were started after 24 hours in RPMI + 10% FBS. Media were changed every week, plates were inspected thrice a week under the microscope, and wells >50% confluent were scored as positive.
14- Tricker E.M.
- Xu C.
- Uddin S.
- et al.
Combined EGFR/MEK inhibition prevents the emergence of resistance in EGFR-mutant lung cancer.
Western Blot Analysis
Cells were treated with EGF (10 ng/mL), C-Abs, anti-EGF VacAbs, EGFR TKIs, or combinations, for 2 to 24 hours in RPMI + 0.5% HS. After washing twice with cold PBS, cultures were scraped into radioimmunoprecipitation assay buffer (20 mmol/L Tris-HCl pH 7.5, 150 mmol/L NaCl, 1 mmol/L Na2EDTA, 1 mmol/L EGTA, 1% NP-40, 1% sodium deoxycholate, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L β-glycerophosphate, 1 mmol/L Na3VO4, 1 μg/mL leupeptin (Cell Signaling Technologies), 2 mmol/L phenylmethylsulfonyl fluoride (PMSF) and protease inhibitor cocktail (Roche Diagnostics, Mannheim, Germany) and passed through an insulin syringe. Lysates were incubated on ice for 15 minutes, centrifuged for 10 minutes at 14,000 rpm, and immediately analyzed or frozen at –80 ºC. Protein extracts (25 μg) were boiled in Laemmli buffer (NuPAGE- LDS sample buffer 4X; Invitrogen. Carlsbad, California), resolved in sodium dodecyl sulfate –polyacrylamide gels and transferred to polyvinylidene difluoride membranes (Bio-Rad, Hercules, California). Membranes were incubated for 1 hour in Odyssey blocking buffer (Li-Cor Biosciences, Lincoln, Nebraska) or Phosphoblocker reagent (Cell Biolabs Inc., San Diego, California), cut, incubated with primary antibodies o/n at 4 ºC, washed three times with PBS-TWEEN 0.1% and incubated for 2 hours with a secondary antibody (anti-rabbit or anti-mouse immunoglobulin G [IgG] horseradish peroxidase-conjugated secondary antibody; GE Healthcare, New York, New York). Finally, membranes were washed with PBS-TWEEN 0.1% and revealed with Supersignal Chemiluminescence substrate (Thermofisher, Waltham, Massachusetts).
Flow Cytometry
Cells were treated under the same conditions described for Western blotting, trypsinized and centrifuged. For cycle analyses, cells were resuspended in PBS, fixed in 70% ethanol, incubated o/n at –20 ºC, centrifuged at 1200 rpm for 5 minutes at 4 ºC, incubated for 1 hour at 37 ºC in 250 μL of PBS with 50 μg/mL RNAse A (Sigma-Aldrich) and stained with propidium iodide (PI) (Roche Diagnostics) for 30 minutes at room temperature in the dark. For cell death analyses, the Annexin-V-FLUOS staining kit (Roche Diagnostics) was used according to the manufacturer’s instructions. Stained cells were analyzed with a FACSCanto II cytometer (BD Biosciences, Franklin Lakes, New Jersey) using the FACSDiva software version 6.1.2. In the case of cell death analyses, the combination of annexin V and PI was used to differentiate four cell populations; namely, viable cells (An -/PI-), early apoptotic (An+/PI-), necrotic (An-/PI+), and later apoptotic/necrotic (An+/PI+).
mRNA Analysis
RNA was isolated from cell lines as previously described.
15- Chaib I.
- Karachaliou N.
- Pilotto S.
- et al.
Co-activation of STAT3 and YES-associated protein 1 (YAP1) pathway in EGFR-mutant NSCLC.
Primer and probe sets were designed using Primer Express 3.0 Software (Applied Biosystems, Foster City, California) according to their reference sequences (http://
www.ncbi.nlm.nih.gov/gene). Quantification of gene expression was performed using the ABI Prism 7900HT Sequence Detection System (Applied Biosystems). Expression levels were calculated according to the comparative ΔΔCt method. Commercial RNA controls were used as calibrators (Liver and Lung; Stratagene, La Jolla, California). For each cell line, three independent experiments were performed.
Discussion
NSCLC patients with
EGFR mutations are less likely to derive benefit from immune checkpoint inhibitors. In three recent clinical trials of second-line docetaxel versus nivolumab, pembrolizumab, and atezolizumab, the inhibitors failed to improve the overall survival of
EGFR-mut patients progressing to cytotoxic chemotherapy or EGFR TKIs.
16- Borghaei H.
- Paz-Ares L.
- Horn L.
- et al.
Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer.
, 17- Herbst R.S.
- Baas P.
- Kim D.W.
- et al.
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non–small-cell lung cancer (KEYNOTE-010): a randomised controlled trial.
, 18- Rittmeyer A.
- Barlesi F.
- Waterkamp D.
- et al.
Atezolizumab versus docetaxel in patients with previously treated non–small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial.
, 19- Lee C.K.
- Man J.
- Lord S.
- et al.
Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer—a meta-analysis.
This inferior response can be a consequence of the un-inflamed microenvironment present in
EGFR-mut tumors, with low programmed death ligand 1 expression and lack of T cell infiltration.
20- Dong Z.Y.
- Zhang J.T.
- Liu S.Y.
- et al.
EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non–small cell lung cancer.
In consequence, the development of new immunotherapeutic approaches for
EGFR-mut NSCLC patients can be of particular interest.
Vaccination against EGF constitutes a novel strategy that, contrary to programmed death 1 blockade, is not intended at reversing tumor-induced immunosuppression by activating the T cells. Instead, it aims to stimulate B cells to produce neutralizing antibodies that sequester circulating EGF, thus preventing its binding to EGFR. Vaccination against EGF, also referred to EGF–pathway targeted immunization, is well tolerated, generates few cases of severe adverse effects, and has shown promising results in two trials enrolling unselected advanced NSCLC patients.
11- Rodriguez P.C.
- Popa X.
- Martinez O.
- et al.
A phase III clinical trial of the epidermal growth factor vaccine CIMAvax-EGF as switch maintenance therapy in advanced non-small cell lung cancer patients.
, 12- Rosell R.
- Neninger E.
- Nicolson M.
- et al.
Pathway targeted immunotherapy: rationale and evidence of durable clinical responses with a novel, EGF-directed agent for advanced NSCLC.
, 21- Neninger Vinageras E.
- de la Torre A.
- Osorio Rodriguez M.
- et al.
Phase II randomized controlled trial of an epidermal growth factor vaccine in advanced non–small-cell lung cancer.
However, little was known about the molecular and cellular mechanisms involved in the effects of anti-EGF antibodies besides their capability to block ligand binding and phosphorylation of EGFR, or about their differential activity in tumors with
EGFR mutations or other genetic alterations.
22- Garcia B.
- Neninger E.
- de la Torre A.
- et al.
Effective inhibition of the epidermal growth factor/epidermal growth factor receptor binding by anti-epidermal growth factor antibodies is related to better survival in advanced non-small-cell lung cancer patients treated with the epidermal growth factor cancer vaccine.
Our study shows that anti-EGF VacAbs raised in rabbits suppress the effects of EGF on cell proliferation, cell cycle, and signal transduction pathways in
EGFR-mut NSCLC cell lines, particularly in those derived from untreated patients. The concentrations of EGF used in our experiments (10 ng/mL) are close to those reported in HS, which have a median around 1 ng/mL and show a significant inter-individual variability.
11- Rodriguez P.C.
- Popa X.
- Martinez O.
- et al.
A phase III clinical trial of the epidermal growth factor vaccine CIMAvax-EGF as switch maintenance therapy in advanced non-small cell lung cancer patients.
, 23- Joh T.
- Itoh M.
- Katsumi K.
- et al.
Physiological concentrations of human epidermal growth factor in biological fluids: use of a sensitive enzyme immunoassay.
Remarkably, the anti-EGF VacAbs were also found to consistently reduce the levels of pErk1/2 in absence of exogenous EGF not only in PC9 cells (
Fig. 3A and
B, compare lanes “C” and “Ab”), but also in PC9-GR4 cells, where the growth factor did not show significant effects (
Fig. 3C, compare lanes “C” and “Ab”). One of the possible explanations for this observation could be the existence of receptor/ligand feedback loops in the cell lines used. Sera from patients immunized with an anti-EGF vaccine were also shown to efficiently block the activation of pErk1/2 by EGF. Control sera from nonimmunized patients had little effect on Erk1/2 but strongly activated Akt, indicating that the blood of healthy individuals contains growth factors that specifically trigger pAkt in
EGFR-mut cells. In contrast, the sera from the four immunized patients analyzed were less active in inducing Akt phosphorylation (
Fig. 4D). Significant differences were observed in the potency of the sera from vaccinated individuals to block Erk1/2 phosphorylation and, to a lesser extent, to activate Akt (
Fig. 4D, compare patients #2 and #7). Being a new therapeutic approach with only a phase III trial completed, the availability of samples from patients vaccinated against EGF is limited, and we could not gain access to additional sera to further explore this variability or to correlate it with clinical outcomes.
Next, we discovered that EGF significantly reduced the antiproliferative effects of gefitinib, erlotinib, afatinib, and osimertinib in several
EGFR-mut NSCLC cells, both sensitive and resistant to EGFR TKIs. This finding correlated with the results of Western blotting experiments where the levels of pErk1/2 in cells treated with EGFR TKIs were significantly higher if EGF was present (
Fig. 3A–D, compare lanes “TKI” with “TKI+EGF”). To the best of our knowledge, this is the first time that the effects of EGF on the sensitivity of
EGFR-mut cells to TKIs are investigated. In the initial report describing the development of gefitinib, EGF was found to have the opposite effect in the two
EGFR-wild-type (wt) cell line models used, the human umbilical vein endothelial cell endothelial and the KB squamous carcinoma cells, where the concentration that inhibits 50% of the drug decreased approximately 100 times in the presence of EGF.
24- Wakeling A.E.
- Guy S.P.
- Woodburn J.R.
- et al.
ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy.
Our finding suggests that
EGFR-mut patients with high EGF levels might have worse outcomes to EGFR TKIs, and additional studies are warranted to validate this hypothesis. Increased serum levels of two EGFR ligands, transforming growth factor alpha and amphiregulin, have been reported to correlate with worse responses to EGFR TKIs in unselected NSCLC patients.
25- Vollebergh M.A.
- Kappers I.
- Klomp H.M.
- et al.
Ligands of epidermal growth factor receptor and the insulin-like growth factor family as serum biomarkers for response to epidermal growth factor receptor inhibitors in patients with advanced non–small cell lung cancer.
, 26- Ishikawa N.
- Daigo Y.
- Takano A.
- et al.
Increases of amphiregulin and transforming growth factor-alpha in serum as predictors of poor response to gefitinib among patients with advanced non–small cell lung cancers.
Regarding EGF, in the only study published so far, which included 11
EGFR-mut and 21
EGFR-wt NSCLC patients treated with erlotinib, those with high concentration of EGF in serum had shorter progression-free survival, although the differences did not reach statistical significance.
27- Romero-Ventosa E.Y.
- Blanco-Prieto S.
- Gonzalez-Pineiro A.L.
- et al.
Pretreatment levels of the serum biomarkers CEA, CYFRA 21-1, SCC and the soluble EGFR and its ligands EGF, TGF-alpha, HB-EGF in the prediction of outcome in erlotinib treated non–small-cell lung cancer patients.
The reduction in the efficacy of EGFR TKIs induced by EGF provided us with a rationale to test the effects of the addition of anti-EGF VacAbs. The combination of gefitinib, erlotinib, afatinib, and osimertinib with anti-EGF VacAbs showed a stronger antiproliferative effect than the EGFR TKIs alone in the
EGFR-mut cell lines tested (
Fig. 2), a finding that correlated with a consistent decrease in pErk1/2 (
Fig. 3 and
Supplementary Fig. 2D, compare lanes TKI + EGF with TKI + Ab + EGF). The combination was also superior in blocking the anti-apoptotic and G2/M stimulating effects of EGF.
The anti-EGFR monoclonal antibody cetuximab has also been tested in
EGFR-mut cell line models. Similarly to anti-EGF VacAbs, cetuximab blocks ligand binding in vitro and has been shown to prevent ligand-induced EGFR, Erk1/2, and Akt phosphorylation in PC9 and H1975 cells.
28- Perez-Torres M.
- Guix M.
- Gonzalez A.
- et al.
Epidermal growth factor receptor (EGFR) antibody down-regulates mutant receptors and inhibits tumors expressing EGFR mutations.
However, it showed a relatively little effect on EGFR downstream signaling in other
EGFR-mut lines such as H3255 or DFCILU-011.
29- Mukohara T.
- Engelman J.A.
- Hanna N.H.
- et al.
Differential effects of gefitinib and cetuximab on non–small-cell lung cancers bearing epidermal growth factor receptor mutations.
Regarding receptor down-regulation, there seems to be significant differences between the two antibodies. Cetuximab has been shown to markedly decrease the levels of total EGFR after 1 to 2 hours of incubation in
EGFR-mut cells such as PC9, H1975 or H3255, whereas anti-EGF VacAbs did not induce any significant down-regulation of the receptor after 24 hours of incubation (
Fig. 5).
28- Perez-Torres M.
- Guix M.
- Gonzalez A.
- et al.
Epidermal growth factor receptor (EGFR) antibody down-regulates mutant receptors and inhibits tumors expressing EGFR mutations.
Finally, although cetuximab has been reported to amplify the induction of apoptosis and tumor regression in
EGFR-wt, head and neck cancer cell lines, and subcutaneous tumors, it failed to enhance the effects of gefitinib in PC9 xenografts.
28- Perez-Torres M.
- Guix M.
- Gonzalez A.
- et al.
Epidermal growth factor receptor (EGFR) antibody down-regulates mutant receptors and inhibits tumors expressing EGFR mutations.
, 30- Huang S.
- Armstrong E.A.
- Benavente S.
- et al.
Dual-agent molecular targeting of the epidermal growth factor receptor (EGFR): combining anti-EGFR antibody with tyrosine kinase inhibitor.
In contrast, we found that the anti-EGF VacAbs potentiated the antiproliferative activity of EGFR TKIs in PC9 and the rest of
EGFR-mut cell lines tested. This potentiating effect reached statistical significance in all cases, with the only exception being PC9-GR4 cells (
Fig. 2). The fact that the anti-EGF VacAbs target the ligand instead of the receptor and do not induce EGFR down-regulation might offer a possible explanation for the differences found between the effects of cetuximab and anti-EGF antibodies. One of the limitations of our work was that we could not perform xenograft studies, which would have offered valuable information about the effects of the anti-EGF VacAbs in vivo. Xenografts models involve athymic mice, which cannot be easily vaccinated against EGF due to their defective immune system.
Finally, we examined the effects of prolonged exposure of
EGFR-mut cells to TKIs and anti-EGF VacAbs. We found that the addition of the anti-EGF VacAbs significantly delayed the appearance of clones resistant to gefitinib and afatinib in the PC9 cell line. Cetuximab has also been reported to delay the emergence of resistance to erlotinib and afatinib in animal models of
EGFR-mut lung adenocarcinoma but, again, we found some differences between the mechanisms of action of the two antibodies.
31- Pirazzoli V.
- Ayeni D.
- Meador C.B.
- et al.
Afatinib plus cetuximab delays resistance compared to single-agent erlotinib or afatinib in mouse models of TKI-naive EGFR L858R-induced lung adenocarcinoma.
As we have previously reported, we observed that gefitinib, erlotinib, and afatinib induced phosphorylation of STAT3 after 24 hours.
15- Chaib I.
- Karachaliou N.
- Pilotto S.
- et al.
Co-activation of STAT3 and YES-associated protein 1 (YAP1) pathway in EGFR-mutant NSCLC.
The addition of anti-EGF VacAbs consistently suppressed this TKI-induced STAT3 activation. In contrast, pSTAT3 has been shown to be elevated in head and neck human tumors progressing to cetuximab, suggesting that STAT3 activation is involved in resistance to this drug.
32- Sen M.
- Joyce S.
- Panahandeh M.
- et al.
Targeting Stat3 abrogates EGFR inhibitor resistance in cancer.
We also found that the anti-EGF VacAbs reduced the total levels of other proteins associated with the emergence of resistance to EGFR TKIs, such as AXL, BMI1, HES1, or NOTCH-3 cleaved in PC9 cells.
33- Zhang Z.
- Lee J.C.
- Lin L.
- et al.
Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer.
, 34- Schulz D.
- Wirth M.
- Piontek G.
- et al.
HNSCC cells resistant to EGFR pathway inhibitors are hypermutated and sensitive to DNA damaging substances.
, 35- Codony-Servat C.
- Codony-Servat J.
- Karachaliou N.
- et al.
Activation of signal transducer and activator of transcription 3 (STAT3) signaling in EGFR mutant non–small-cell lung cancer (NSCLC).
, 36- Hashida S.
- Yamamoto H.
- Shien K.
- et al.
Acquisition of cancer stem cell-like properties in non–small cell lung cancer with acquired resistance to afatinib.
Cetuximab resistance in NSCLC is characterized by upregulation of AXL and EGFR ligands. In cetuximab-resistant cells, increased EGFR ligand production leads to AXL and EGFR heterodimerization which, in turn, induces the transcription of the
AXL gene via Erk1/2.
37- Vouri M.
- Croucher D.R.
- Kennedy S.P.
- et al.
Axl-EGFR receptor tyrosine kinase hetero-interaction provides EGFR with access to pro-invasive signalling in cancer cells.
, 38- Brand T.M.
- Iida M.
- Stein A.P.
- et al.
AXL mediates resistance to cetuximab therapy.
In our case, because anti-EGF VacAbs capture EGF, preventing its binding to EGFR, we can hypothesize that they also block the formation of
EGFR-AXL heterodimers and, consequently, the expression of AXL (
Fig. 6C). Regarding HES1 and NOTCH-3 cleaved, they have been described to be elevated in
EGFR-mut cells after EGFR TKI treatment and this event has been related to an increase in stem-like cells and emergence of resistance.
35- Codony-Servat C.
- Codony-Servat J.
- Karachaliou N.
- et al.
Activation of signal transducer and activator of transcription 3 (STAT3) signaling in EGFR mutant non–small-cell lung cancer (NSCLC).
, 39- Arasada R.R.
- Amann J.M.
- Rahman M.A.
- et al.
EGFR blockade enriches for lung cancer stem-like cells through notch3-dependent signaling.
In summary, we have shown that anti-EGF VacAbs suppress the effects of EGF and significantly enhance the antitumor activity of EGFR TKIs in EGFR-mutated NSCLC cell lines. They also block STAT3 activation, reduce AXL expression, and delay the acquisition of resistance. Based on these findings, a phase I trial of an anti-EGF vaccine in combination with afatinib as a first-line therapy in EGFR-mut NSCLC patients has been initiated (EPICAL study).