![Open Access](/resources/images/iconopenaccess.png)
Evaluation of co‑inhibition of ErbB family kinases and PI3K for HPV‑negative head and neck squamous cell carcinoma
- Authors:
- Published online on: January 27, 2025 https://doi.org/10.3892/or.2025.8871
- Article Number: 38
-
Copyright: © Geng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Head and neck squamous cell carcinoma (HNSCC) is the sixth most prevalent type of cancer worldwide with 890,000 new cases and 450,000 deaths reported in 2018 (1). Based on etiological factors, HNSCC is classified into two types of disease: HPV-positive or HPV-negative. HPV-positive HNSCC occurs mainly in oropharyngeal tissues, whereas HPV-negative HNSCC is primarily found in the oral cavity and larynx (2). The occurrence of HPV-negative HNSCC is associated with the use of tobacco and excessive consumption of alcohol, and is characterized by mutations in diverse oncogenic driver genes. By contrast, HPV-positive HNSCC is related to human papillomavirus (HPV) infection, whereby viral proteins E6 and E7 cause oncogenic transformation of host cells (3–6). Compared with HPV-negative HNSCC, HPV-positive HNSCC expresses viral proteins and other neoantigens, as well as mutagenesis caused by viral infection, which may be advantageous to immunotherapy (2). In addition, HPV-positive HNSCC has better clinical outcomes with standard therapy in comparison to HPV-negative HNSCC (7). Reports have indicated that patients with HPV-positive HNSCC have a higher 5-year survival rate (~80%) than HPV-negative patients (~50%) (8); therefore, there is an urgent need for new effective therapeutics to treat HPV-negative HNSCC (9,10).
Classical therapies for patients with HNSCC without distant metastasis typically include surgical resection, radiation therapy, chemotherapy or a combination of these regimens. The specific therapeutic approach depends on various factors, such as pre-existing clinical conditions, cancer location and the Tumor-Node-Metastasis stage of the tumor (4,11). The combination of these treatments could reduce the rate of recurrence and distant metastasis for patients with metastatic disease (12). However, chemotherapy remains the primary option for patients with recurrent and distant metastatic HNSCC (13). Cisplatin is the most commonly used anticancer drug to treat advanced HNSCC; however, while a number of newly diagnosed patients with advanced HNSCC initially respond well to cisplatin-based chemotherapies, most patients either have intrinsic resistance or will eventually develop acquired resistance to cisplatin, which often leads to death within 1 year (14). Immunotherapy has been introduced to treat refractory HNSCC, but its impact has been limited (14,15). Therefore, it remains of the utmost importance to identify new therapeutic alternatives to improve the efficacy of cisplatin-based therapies, or to replace them, for the treatment of patients with advanced HNSCC.
Epidermal growth factor receptor (EGFR), a member of the ErbB kinase family, is reported to be upregulated in 90% of HNSCC cases and cell lines, according to a study that included 24 patients with SCCHN and 10 cell lines, as opposed to seven healthy control individuals (16) and serves a crucial role in the pathogenesis and clinical course of cancer (13,17,18). EGFR controls the activation of several essential pathways, such as PI3K/Akt/mTOR and RAS-RAF-MEK-ERK, which regulate cell proliferation, survival and migration (19,20). In 2006, the monoclonal EGFR antibody cetuximab was approved by the Food and Drug Administration (FDA) to treat HNSCC in combination with the standard therapy (21–23). However, the use of cetuximab has been reported to result in very limited improvement in survival rates for patients undergoing cisplatin-based therapy (24). In addition, small molecule kinase inhibitors, such as gefitinib and erlotinib, while effective in targeted therapies for non-small cell lung cancer, have not demonstrated any benefits for patients with HNSCC (25,26). Increasing evidence has demonstrated the importance of the ErbB family, which contains EGFR, HER2, HER3 and HER4, in the carcinogenesis of HNSCC and its response to therapies (27). HER2 and HER3 form heterodimers with EGFR and play a role in PI3K/Akt activation. In addition, HER2 and HER3 have been shown to be associated with resistance to EGFR and PI3K inhibitors in cancer (28,29). These results indicated that targeting the ErbB family kinases could more effectively suppress HNSCC compared with using EGFR inhibitors alone (25,30). Notably, the FDA-approved ErbB family inhibitor afatinib has shown positive results in HNSCC clinical trials and is now listed in the National Comprehensive Cancer Network (NCCN) guidelines as a third-line single agent for HNSCC treatment (25,26,31–33). Understanding the mechanisms behind resistance to afatinib and exploring methods to avoid that resistance would be beneficial.
PI3K is one of the most important downstream effectors of the EGFR/ErbB receptor family. The genes PIK3CA, PIK3CB and PIK3CD encode three highly homologous catalytic isoforms of class IA PI3K, p110α, p110β and p110δ, respectively. These isoforms associate with any of five regulatory isoforms: p85α, its splicing variants p55α and p50α, p85β and p55γ (34). The most important PI3K-p85 complex is PI3Kα/p85α. The PI3K pathway has been reported to be the most frequently mutated oncogenic pathway, detected in 30.5% of tumors, according to whole-exome sequencing of the genes in the JAK/STAT, MAPK and PI3K pathways in 151 HNSCC tumors (35). Previous studies have also shown that PIK3CA, the gene that codes for PI3K p110α, is one of the most frequently mutated genes in HNSCC (35–37), and PIK3CA amplification and overexpression have also been identified in HNSCC (38). According to The Cancer Genome Atlas (TCGA), profiling of 279 HNSCC tumors identified amplification or mutation of PIK3CA in 34% of HPV-negative HNSCC tumors and 56% of HPV-positive tumors (39,40). PI3K activation in turn activates Akt, which then phosphorylates its substrates, such as TSC2, PRAS40, GSK3β and FOXO, to regulate multiple cellular functions that consequently control cell proliferation, survival and response to therapy. The tumor suppressor gene PTEN encodes the PTEN protein that dephosphorylates PIP3 to inhibit the PI3K pathway. Mutations that result in PTEN loss or deceased PTEN expression have been frequently observed in HPV-positive and negative HNSCC (41,42). These alterations further result in PI3K/Akt activation (43). Therefore, PI3K is an attractive target for the treatment of HNSCC (40,44,45).
Our previous study reported that co-targeting the ErbB family and PI3K, through a combination of afatnib and copanlisib, suppressed the growth of HPV-positive HNSCC (46). The present study further explored whether this combination was also effective at suppressing the growth of HPV-negative HNSCC. The present findings indicated that the combination of afatnib and copanlisib may be a promising treatment option for HPV-negative HNSCC.
Materials and methods
Cell culture
HNSCC cell lines, Cal27 and FaDu, were purchased from American Type Culture Collection, authenticated by short tandem repeat analysis and tested for Mycoplasma contamination at the Translational Core Facility at Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland (Baltimore, MD, USA). All cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 2 mM glutamine, and 100 U/ml penicillin and streptomycin (all from Gibco; Thermo Fisher Scientific) at 37°C in an incubator containing 5% CO2 for 24 h before each experiment was performed.
Antibodies and inhibitors
The following antibodies were purchased from Cell Signaling Technology, Inc.: Phosphorylated (P)-Akt-S473 (cat. no. 4058), P-Akt-T308 (cat. no. 9275), Akt (cat. no. 2938), P-HER2-Y1248 (cat. no. 2247), HER2 (cat. no. 4290), P-HER3-Y1289 (cat. no. 2842), HER3 (cat. no. 12708), caspase-3 (cat. no. 9665), cleaved-caspase-3 (cat. no. 9664) and β-actin (cat. no. 4967). HRP-labeled anti-mouse (cat. no. W4028) and anti-rabbit (cat. no. W4018) secondary antibodies were purchased from Promega Corporation. Gefitinib, erlotinib and afatinib, as well as all PI3K inhibitors (copanlisib, BKM120, GDC-0941, BYL179, AZD8186 and CAL-101) were purchased from Selleck Chemicals. Copanlisib was dissolved in 10% trifluoroacetic acid (TFA), while all other drugs were dissolved in 100% DMSO. The final concentrations of TFA and DMSO in cell culture media were 0.01% and 0.1%. For single EGFR/ErbB inhibitor treatment, cells were treated with DMSO or increasing concentrations of gefitinib (3 nM-30 µM), erlotinib (3 nM-30 µM) and afatinib (1 nM-10 µM) at 37°C for 96 h. For single PI3K inhibitor treatment, cells were treated with TFA, DMSO or increasing concentrations of copanlisib (0.01 nM-10 µM), GDC-0941 (1 nM-10 nM) and other PI3K inhibitors (3 nM-30 µM) at 37°C for 96 h. For the combination treatment, Cal27 cells were treated with the combination of various concentrations of copanlisib (2.875–92 nM) and afatinib (0.2875–9.2 nM), and FaDu cells were treated with the combination of various concentrations of copanlisib (3.375–118 nM) and afatinib (0.3125–11.6 nM) at 37°C for 96 h.
Cell lysis and western blot analysis
Cell lysis and western blot analysis were performed as previously described (46,47). Briefly, cells grown on 100-mm dishes were rinsed twice with 1X cold PBS, then lysed on ice for 30 min in 1 ml lysis buffer (M-PER™ Mammalian Protein Extraction Reagent; cat. no. 78503; Pierce; Thermo Fisher Scientific, Inc.) with phosphatase inhibitor and EDTA-free protease inhibitors (Roche Diagnostics). After centrifugation at 13,000 × g for 20 min, lysates that contained 30 µg protein were separated by SDS-PAGE on 4–12% gels, and the proteins were transferred to Pure Nitrocellulose Membranes (Bio-Rad Laboratories, Inc.), which were blocked in 5% nonfat milk at room temperature for 1 h. The membranes were then incubated with the indicated primary antibodies (1:1,000 dilutions) overnight at 4°C, and with HRP-conjugated secondary antibodies (1:4,000 dilutions) at room temperature for 1 h. Proteins were finally visualized by using Clarity™ Western ECL Substrate (cat. no. 1705060; Bio-Rad Laboratories, Inc.).
Analyzing apoptosis by Annexin V/propidium iodide (PI) staining
Apoptosis analysis was performed using the FITC Annexin V Apoptosis Detection Kit II (cat. no. 556570; BD Biosciences) combined with PI (cat. no. P1304MP; Invitrogen; Thermo Fisher Scientific, Inc.) staining as previously described (46,47). Cells were plated into 6-well plates at a density of 4.5×105/well overnight, before being treated with vehicle control, copanlisib (30 nM in Cal27 and 125 nM in FaDu), afatinib (0.5 µM in Cal27 and 1 µM in FaDu), or a combination of copanlisib and afatinib at 37°C for 48 h. Both floating and adherent cells were collected and stained with Annexin V and PI, according to the manufacturers' instructions. Samples and single stained controls were subsequently analyzed using the BD FACSCanto II flow cytometer (BD Biosciences). Data were analyzed using FCS Express 7 (Research Edition; De Novo Software). Cells stained solely with Annexin V (early apoptotic cells) or double stained with Annexin V and PI (late apoptotic cells) were considered apoptotic cells.
Cell viability assay
Cell viability was assessed using sulforhodamine B (SRB) staining as described previously (48). Briefly, cells were plated in 96-well plates at a density of 3,500 cells/well overnight before being subjected to treatment with a serial concentration of the indicated drugs or vehicle control for 96 h at 37°C. Following an overnight fixation in methanol, the cells were stained with SRB for 1 h and then dissolved with 10 mmol/l Tris base for 1 h while being shaken. The aforementioned procedures were all carried out at room temperature. Subsequently, the absorbance was read at 492 nm. The half maximal effective concentration (EC50) values were calculated using nonlinear regression analysis with variable slope in Graph-Pad Prism v8.0 (Dotmatics). The effect of the inhibitors on cell viability are represented as percentages compared with vehicle control-treated cells. Each experiment was performed in triplicate and repeated at least twice. The vehicle controls were treated with the reagents that were used to dissolve the chemicals (0.01% TFA for copanlisib and 0.1% DMSO for the other drugs). To determine synergy of the drug combination, the combination index (CI) values were determined according to the Chou-Talalay method (49) using CalcuSyn software (version 2.0; BIOSOFT).
Tumor xenograft formation in mice
In vivo experiments were conducted with the approval of the Institutional Animal Care and Use Committee (IACUC) of University of Maryland (IACUC #1021001) and within an Association for Assessment and Accreditation of Laboratory Animal Care International-accredited vivarium. Female 6-week-old nude mice (n=45), purchased from Inotiv, Inc., were used for the animal studies. All mice were housed under the following conditions: Ambient temperature (21–22°C), regulated humidity (40–50%) ad libitum access to food and water, 12-h light/dark cycle. The animals were treated and handled in accordance with the guidelines set by the IACUC of the University of Maryland. A total of 45 nude mice received injection of 0.5×106 FaDu cells into the right flank. The mice were split into four groups (n=7/group) 13 days after cell injection, so that the mean tumor volume was similar (~150 mm3). Notably, 45 mice were injected but only 28 mice entered the study; mice were excluded from entering the study based on irregular shape of the tumors (difficult to measure). One mouse per group was excluded from analysis due to necrosis at termination of the study; mice were sacrificed as soon as necrosis was observed. The day of sorting occurred on the first day of dosing and was defined as Day 1. The four treatment groups (n=6/group at termination) were: Vehicle control, copanlisib (6 mg/kg, intraperitoneal, 5 times/week, Monday to Friday), afatinib (6 mg/kg, oral, 5 times/week, Monday to Friday), and a combination of copanlisib and afatinib. Tumor volume was measured twice per week using electronic calipers and the animals were weighed five times per week. Tumor volume was calculated as (L × W2)/2, where W is the smaller dimension and L is tumor length. At the end of the study (32 days), mice were euthanized by CO2 asphyxiation (30% vol/min) followed by cervical dislocation as noted in the IACUC-approved protocol, and the tumors were excised, weighed and cut in half. Half of the tumors were fixed in formalin (4%) at room temperature for 24 h and then transferred to 70% ethanol (half), whereas the other half were frozen at −80°C. Tumors were preserved by fixation and freezing for future evaluation. All mice were euthanized 32 days after the start of dosing. No animal who entered the study was found dead; however, as aforementioned, one mouse from each group was removed from the study and euthanized due to necrosis. The study was terminated based on tumor volume (~2,000 mm3) as listed in the IACUC-approved protocol as a humane endpoint. No analgesics were administered during the study since flank tumor studies are considered painless. Mice were briefly anesthetized with 2.5% isoflurane during the subcutaneous injection of cells.
Statistical analysis
In vitro data were repeated at least three times unless specifically described in figure legends. The data were presented as the mean ± SD and animal data are shown as the mean ± SEM. Statistical analysis was performed using GraphPad Prism version 10.3.1 (Dotmatics). One-way ANOVA was used to statistically analyze multiple groups, followed by Tukey's post hoc analysis. P<0.05 was considered to indicate a statistically significantly difference.
Results
HPV-negative HNSCC cell lines are sensitive to afatinib
The EC50 values of EGFR/ErbB family inhibitors, gefitinib, erlotinib and afatinib, were determined in two HPV-negative HNSCC cell lines, Cal27 and FaDu. Both cell lines were relatively resistant to erlotinib and sensitive to gefitinib (Fig. 1A and B). However, they were revealed to be very sensitive to afatinib, although Cal27 cells were more sensitive to afatinib in comparison to FaDu cells. These results suggested that afatinib was the more effective small molecule inhibitor for the treatment of HPV-negative HNSCC in comparison to other EGFR inhibitors.
Copanlisib is the most effective PI3K inhibitor to suppress HPV-negative HNSCC viability
To identify the most effective PI3K inhibitor, the EC50 values of six PI3K inhibitors were determined, including three pan-PI3K inhibitors, copanlisib, BKM120 and GDC-09410; the PI3Kα inhibitor Byl719; the PI3Kβ inhibitor AZD8186; and the PI3Kδ inhibitor Cal-101 (Fig. 2). Cal27 cells were strongly resistant to the PI3Kδ inhibitor, and relatively resistant to PI3Kα and PI3Kβ inhibitors. However, they were much more sensitive to the pan-PI3K inhibitors copanlisib, GDC-0941 and BKM120. Moreover, the EC50 value of copanlisib was much lower in comparison to those of BKM120 and GDC-0941. Similar results were found in FaDu cells (Fig. S1). These results suggested that copanlisib may be the most effective small molecule PI3K inhibitor to treat HPV-negative HNSCC.
Synergistic inhibition of cell viability using a combination of afatinib and copanlisib
The present study aimed to assess whether simultaneous inhibition of ErbB family and PI3K pathways could more effectively inhibit HPV-negative HNSCC cell viability. Based on the aforementioned data, afatinib and copanlisib were selected for the combination therapy. Similar to the results shown in Figs. 1 and 2, afatinib or copanlisib alone inhibited cell viability; however, the combination caused enhanced inhibition of viability in both Cal27 (Fig. 3A) and FaDu cells (Fig. 3B). Furthermore, the related CI value for each combination was calculated according to the Chou-Talalay method (49) using CalcuSyn software. The CI values for all combinations were <1.0, which indicated a synergistic effect in the combination of afatinib and copanlisib (Fig. 3C and D). These data indicated that afatinib and copanlisib may synergistically inhibit HPV-negative HNSCC cell viability.
Synergistic inhibition of xenograft tumor growth via the combination of afatinib and copanlisib in mice
The present study also evaluated the antitumor activity of the afatinib and copanlisib combination in vivo using a mouse xenograft model. FaDu cells were inoculated into the mice, and when the tumors reached ~200 mm3, the mice were randomly split into four groups, which were treated with a vehicle control, copanlisib, afatinib or their combination. The original aim was to treat the mice for more than 6–8 weeks; however, the experiment was terminated on day 32 when tumor necrosis was observed in the majority of the mice in the control group. The average tumor volume in groups treated with either copanlisib or afatinib was lower than that of the control group, but there were no significant differences in tumor volume among these three groups, which may be due to tumor necrosis-induced decreases in tumor volumes in the control group (Fig. 4A). However, tumor volumes in the combination treatment group were significantly lower than in the single treatment groups, and they were more significantly lower than those in the control group by the end of the study.
Similarly, after the tumors were excised and weighed at the end of the study, there was no significant difference in tumor weight (mg) between the groups treated with either copanlisib or afatinib (Fig. 4B). In addition, there were no significant differences in tumor weight (mg) between the control group and the single agent treatment groups due to individual differences. However, tumor weight in the combination treatment group was significantly lower compared with that in the control agent treated group. In summary, while afatinib or copanlisib alone had modest inhibitory effects on tumor growth, they did not reach statistical significance, whereas the combination of the two drugs significantly inhibited tumor growth.
Notably, all doses of copanlisib and afatinib in single and combination treatments were well-tolerated, as indicated by no significant weight loss observed during the study (Fig. 4C). These results indicated the feasibility of treatment with a combination of afatinib and copanlisib to suppress HPV-negative HNSCC.
A combination of afatinib and copanlisib induces apoptosis
The present study assessed whether a combination of afatinib and copanlisib could induce more apoptosis compared with either single treatment. Cal27 cells were treated with afatinib (0.5 µM), copanlisib (30 nM) or their combination for 48 h before an apoptosis assay was performed. Afatinib, but not copanlisib alone, induced modest cell apoptosis, whereas their combination significantly increased cell apoptosis (Figs. 5A and S2). Since FaDu cells exhibited higher EC50 to afatinib and copanlisib in comparison to Cal27 cells (Figs. 1, 2 and S1), higher concentrations of afatinib and copanlisib were chosen for the treatments. Treatment with copanlisib (125 nM) significantly enhanced apoptosis, whereas treatment with afatinib (1.0 µM) did not significantly increase apoptosis. However, a combination of copanlisib (125 nM) and afatinib (1.0 µM) led to significantly increased apoptosis compared with either single treatment (Figs. 5B and S3). These data indicated that afatinib and copanlisib may cooperate to induce apoptosis in HPV-negative HNSCC.
Combination of afatinib and copanlisib completely inhibits ErbB and PI3K pathways, which results in induction of caspase-3 cleavage
Previous studies have demonstrated that PI3K inhibitors can induce HER2 and HER3 phosphorylation, thus conferring resistance to PI3K inhibitors (50,51). Our recent study showed that copanlisib induced an increase in P-HER2 Y1248 and P-HER3 Y1289 in HPV-positive HNSCC cells, whereas a combination of afatinib and copanlisib blocked the phosphorylation of HER2 and HER3 (46). The present study further tested whether copanlisib induced P-HER2 Y1248 and P-HER3 Y1289 in HPV-negative Cal27 (Fig. 6A) and FaDu cells (Fig. 6B). Similarly, copanlisib markedly inhibited the phosphorylation of Akt and induced phosphorylation of HER2 Y1248 and HER3 Y1289, whereas the combination of copanlisib and afatinib completely blocked phosphorylation of Akt, HER2 and HER3. In addition, increased caspase-3 cleavage was induced by the combination compared with the single treatments.
Discussion
Afatinib has shown positive results in HNSCC clinical trials and is currently listed in the NCCN guidelines as a third-line single agent for HNSCC treatment (25,26,31–33). It is important to determine the mechanisms by which patients with HNSCC develop resistance to afatinib in order to identify new afatinib-based combination therapies for patients with advanced HNSCC. The present study assessed the efficacy of co-inhibiting the ErbB family and PI3K through a combination of afatinib and copanlisib in the treatment of HPV-negative HNSCC. The results showed that the combination of afatinib and copanlisib induced a marked inhibition of cell viability and suppressed cell survival in vitro in comparison to either treatment alone. Notably, the combination also led to a significant inhibition of xenograft tumor growth without affecting the body weight of the mice. These results suggested that the combination of afatinib and copanlisib may have clinical potential for the treatment of HPV-negative HNSCC.
HNSCC is a heterogenous disease, and its gene mutations/alterations are associated with EGFR inhibitor resistance (5,40,52). In a previous study by Cheng et al (53), the genomic and transcriptomic changes for 15 HPV-negative and 11 HPV-positive HNSCC cell lines were characterized and compared with data from 279 tumors from TCGA. This study identified a number of genetic alterations, including in TP53, PTEN, PIK3CA and FAT1, in HPV-negative and HPV-positive HNSCC cells. The EGFR/ErbB and PI3K/Akt/mTOR pathways are considered the most attractive pathways to target for treatment of HNSCC due to overexpression or activating mutation of PIK3CA, and loss of function mutations of PTEN (5,54–57). It has been reported that constitutive activation of the PI3K/Akt/mTOR pathway due to the alterations in PIK3CA, PTEN, Akt or mTOR may be associated with resistance to EGFR inhibitors (5). The present study used two cell lines: Cal27 and FaDu cells. Both cell lines have TP53 mutations, whereas FaDu cells also have PIK3CA amplification, but Cal27 cells do not (58,59). The present study showed that treating both Cal27 and FaDu cells with afatinib alone blocked Akt phosphorylation at Thr308, but only partially blocked the phosphorylation of Akt at Ser473. Furthermore, it has been reported that PI3K inhibition can lead to increased phosphorylation and total levels of HER3, which confer resistance to PI3K inhibitors (50,51,60–62). The present data showed that copanlisib increased P-HER3 (Y1289), which was counteracted by the addition of afatinib. Notably, the combination of copanlisib and afatinib induced caspase-3 cleavage in addition to the complete inhibition of ErbB and PI3K/Akt pathways. These results provide a rationale for the co-inhibition of ErbB and PI3K as a method to treat HNSCC with or without PIK3CA amplification. However, it is important to test the efficacy of this combination in more HNSCC cell lines with more genetic alterations.
Our previous study reported that the combination of afatinib and copanlisib effectively suppressed HPV-positive HNSCC. The combination therapy blocked both ErbB and PI3K/Akt pathways, which was accompanied by deceased E6 and E7, and the induction of apoptosis, which indicated the increased efficacy of this combination in HPV-positive HNSCC (46). Milewska et al (63) reported that cell lines from multiple types of cancers, including HNSCC with PIK3CA mutations, are sensitive to the combination of afatinib and copanlisib. As the basal level of PI3K/Akt is also high in HPV-negative HNSCC and serves essential roles in the regulation of growth, metastasis and sensitivity to chemotherapy and targeted therapies (5,37,40,64), it would be reasonable to predict that this combination would also be beneficial in HPV-negative HNSCC with upregulated PI3K/Akt signaling.
One of the challenges in treating patients with PI3K pathway inhibitors is the associated toxicity from on-target and off-target effects (65). The most common side-effects observed with PI3K pathway inhibitors in patients are hyperglycemia, rash, stomatitis, diarrhea, nausea and fatigue (65,66). Due to the limitation of the xenograft model, this information is lacking in this study. Moreover, it is important to analyze the toxicity of the combination of the two drugs to the liver and kidney of mice to illustrate the feasibility of the combination; the lack of this analysis is another limitation of the present study. Thus, to accelerate its transition to treatment for patients with HNSCC, it is important to further determine the drug dosage and administration method of the combination in additional preclinical models.
Cisplatin-based chemotherapy is the primary option for the treatment of advanced HNSCC. It is interesting to compare the efficacy and side effects of co-targeting ErbB and PI3K, or in combination with cisplatin in preclinical models. We are currently performing these experiments using HPV-negative HNSCC cell lines with different PIK3CA and PTEN alterations.
Immunotherapy, which includes immune checkpoint blockade that targets PD-L1/PD-1 using PD-L1 or PD-1 inhibitors, has been an important advancement in the treatment of advanced HNSCC. Afatinib modulates PD-L1 expression in multiple types of cancer, including gastric cancer (67). In addition, it has been reported that PI3K inhibitors, such as BKM120, can decrease the expression of PD-L1 in HNSCC cells (68). It would be interesting to determine the effects of afatinib, copanlisib and their combination on the expression of PD-L1 in HNSCC cells, in order to determine the impact of the combination of afatinib and copanlisib on immune checkpoints.
In conclusion, the present results suggested that co-targeting the ErbB family kinases and PI3K using a combination of afatinib and copanlisib may have clinical potential for the treatment of patients with HNSCC.
Supplementary Material
Supporting Data
Acknowledgements
Not applicable.
Funding
This research was supported, in part, by grants from the National Cancer Institute and National Institute of Dental and Craniofacial Research to HD (grant nos. R00CA149178, R01CA212094 and R56DE030423) and the Orakowa Foundation. This research was also supported by funds through the National Cancer Institute-Cancer Center Support Grant (grant no. P30CA134274) and the Maryland Department of Health's Cigarette Restitution Fund Program (grant no. CH-649-CRF).
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
XG contributed to the study conception and design, performed the experiments and data analysis, wrote the original draft, and reviewed and edited the manuscript. SA performed the experiments and conducted data analysis. ZY, RGL and XF contributed to data analysis. YT and RM were involved in data analysis, and reviewing and editing the manuscript. KJC contributed to conceptualization, data analysis, supervision, review, editing the manuscript and funding acquisition. HD contributed to conceptualization, data analysis, supervision, writing, review, editing the manuscript and funding acquisition. XG, SA and HD confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
In vivo experiments were conducted with the approval of the Institutional Animal Care and Use Committee of University of Maryland.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Powell SF, Vu L, Spanos WC and Pyeon D: The key differences between human papillomavirus-positive and -negative head and neck cancers: Biological and clinical implications. Cancers (Basel). 13:52062021. View Article : Google Scholar : PubMed/NCBI | |
Hashibe M, Boffetta P, Zaridze D, Shangina O, Szeszenia-Dabrowska N, Mates D, Fabiánová E, Rudnai P and Brennan P: Contribution of tobacco and alcohol to the high rates of squamous cell carcinoma of the supraglottis and glottis in Central Europe. Am J Epidemiol. 165:814–820. 2007. View Article : Google Scholar : PubMed/NCBI | |
Hashibe M, Brennan P, Benhamou S, Castellsague X, Chen C, Curado MP, Dal Maso LD, Daudt AW, Fabianova E, Fernandez L, et al: Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: Pooled analysis in the international head and neck cancer epidemiology consortium. J Natl Cancer Inst. 99:777–789. 2007. View Article : Google Scholar : PubMed/NCBI | |
Zaryouh H, De Pauw I, Baysal H, Peeters M, Vermorken JB, Lardon F and Wouters A: Recent insights in the PI3K/Akt pathway as a promising therapeutic target in combination with EGFR-targeting agents to treat head and neck squamous cell carcinoma. Med Res Rev. 42:112–155. 2022. View Article : Google Scholar : PubMed/NCBI | |
Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 6:922020. View Article : Google Scholar : PubMed/NCBI | |
Fakhry C, Westra WH, Li S, Cmelak A, Ridge JA, Pinto H, Forastiere A and Gillison ML: Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. 100:261–269. 2008. View Article : Google Scholar : PubMed/NCBI | |
Qin T, Li S, Henry LE, Liu S and Sartor MA: Molecular tumor subtypes of HPV-positive head and neck cancers: Biological characteristics and implications for clinical outcomes. Cancers (Basel). 13:27212021. View Article : Google Scholar : PubMed/NCBI | |
Hennessey PT, Westra WH and Califano JA: Human papillomavirus and head and neck squamous cell carcinoma: Recent evidence and clinical implications. J Dent Res. 88:300–306. 2009. View Article : Google Scholar : PubMed/NCBI | |
Ferreira CC: The relation between human papillomavirus (HPV) and oropharyngeal cancer: A review. PeerJ. 11:e155682023. View Article : Google Scholar : PubMed/NCBI | |
Tran NH, Sais D and Tran N: Advances in human papillomavirus detection and molecular understanding in head and neck cancers: Implications for clinical management. J Med Virol. 96:e297462024. View Article : Google Scholar : PubMed/NCBI | |
Zumsteg ZS, Luu M, Yoshida EJ, Kim S, Tighiouart M, David JM, Shiao SL, Mita AC, Scher KS, Sherman EJ, et al: Combined high-intensity local treatment and systemic therapy in metastatic head and neck squamous cell carcinoma: An analysis of the national cancer data base. Cancer. 123:4583–4593. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wen Y and Grandis JR: Emerging drugs for head and neck cancer. Expert Opin Emerg Drugs. 20:313–329. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lee YS, Johnson DE and Grandis JR: An update: Emerging drugs to treat squamous cell carcinomas of the head and neck. Expert Opin Emerg Drugs. 23:283–299. 2018. View Article : Google Scholar : PubMed/NCBI | |
Perri F, Ionna F, Longo F, Della Vittoria Scarpati G, De Angelis C, Ottaiano A, Botti G and Caponigro F: Immune response against head and neck cancer: Biological mechanisms and implication on therapy. Transl Oncol. 13:262–274. 2020. View Article : Google Scholar : PubMed/NCBI | |
Grandis JR and Tweardy DJ: Elevated levels of transforming growth factor alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer. Cancer Res. 53:3579–3584. 1993.PubMed/NCBI | |
Park BJ, Chiosea SI and Grandis JR: Molecular changes in the multistage pathogenesis of head and neck cancer. Cancer Biomark. 9:325–339. 2010. View Article : Google Scholar : PubMed/NCBI | |
Sharafinski ME, Ferris RL, Ferrone S and Grandis JR: Epidermal growth factor receptor targeted therapy of squamous cell carcinoma of the head and neck. Head Neck. 32:1412–1421. 2010. View Article : Google Scholar : PubMed/NCBI | |
Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wee P and Wang Z: Epidermal growth factor receptor cell proliferation signaling pathways. Cancers (Basel). 9:522017. View Article : Google Scholar : PubMed/NCBI | |
Alorabi M, Shonka NA and Ganti AK: EGFR monoclonal antibodies in locally advanced head and neck squamous cell carcinoma: What is their current role? Crit Rev Oncol Hematol. 99:170–179. 2016. View Article : Google Scholar : PubMed/NCBI | |
Blaszczak W, Barczak W, Wegner A, Golusinski W and Suchorska WM: Clinical value of monoclonal antibodies and tyrosine kinase inhibitors in the treatment of head and neck squamous cell carcinoma. Med Oncol. 34:602017. View Article : Google Scholar : PubMed/NCBI | |
Mehra R, Cohen RB and Burtness BA: The role of cetuximab for the treatment of squamous cell carcinoma of the head and neck. Clin Adv Hematol Oncol. 6:742–750. 2008.PubMed/NCBI | |
Argiris A, Heron DE, Smith RP, Kim S, Gibson MK, Lai SY, Branstetter BF, Posluszny DM, Wang L, Seethala RR, et al: Induction docetaxel, cisplatin, and cetuximab followed by concurrent radiotherapy, cisplatin, and cetuximab and maintenance cetuximab in patients with locally advanced head and neck cancer. J Clin Oncol. 28:5294–5300. 2010. View Article : Google Scholar : PubMed/NCBI | |
Specenier P and Vermorken J: Afatinib in squamous cell carcinoma of the head and neck. Expert Opin Pharmacother. 17:1295–1301. 2016. View Article : Google Scholar : PubMed/NCBI | |
Machiels JPH, Haddad RI, Fayette J, Licitra LF, Tahara M, Vermorken JB, Clement PM, Gauler T, Cupissol D, Grau JJ, et al: Afatinib versus methotrexate as second-line treatment in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 1): An open-label, randomised phase 3 trial. Lancet Oncol. 16:583–594. 2015. View Article : Google Scholar : PubMed/NCBI | |
Saddawi-Konefka R, Schokrpur S, Lui AJ and Gutkind JS: HER2 and HER3 as therapeutic targets in head and neck cancer. Cancer J. 28:339–345. 2022. View Article : Google Scholar : PubMed/NCBI | |
Sacco AG and Worden FP: Molecularly targeted therapy for the treatment of head and neck cancer: A review of the ErbB family inhibitors. Onco Targets Ther. 9:1927–1943. 2016.PubMed/NCBI | |
Rysman B, Mouawad F, Gros A, Lansiaux A, Chevalier D and Meignan S: Human epidermal growth factor receptor 3 in head and neck squamous cell carcinomas. Head Neck. 38 (Suppl 1):E2412–E2418. 2016. View Article : Google Scholar : PubMed/NCBI | |
Palumbo C, Benvenuto M, Focaccetti C, Albonici L, Cifaldi L, Rufini A, Nardozi D, Angiolini V, Bei A, Masuelli L and Bei R: Recent findings on the impact of ErbB receptors status on prognosis and therapy of head and neck squamous cell carcinoma. Front Med (Lausanne). 10:10660212023. View Article : Google Scholar : PubMed/NCBI | |
Kao HF, Liao BC, Huang YL, Huang HC, Chen CN, Chen TC, Hong YJ, Chan CY, Chia JS and Hong RL: Afatinib and pembrolizumab for recurrent or metastatic head and neck squamous cell carcinoma (ALPHA Study): A phase II study with biomarker analysis. Clin Cancer Res. 28:1560–1571. 2022. View Article : Google Scholar : PubMed/NCBI | |
Guo Y, Ahn MJ, Chan A, Wang CH, Kang JH, Kim SB, Bello M, Arora RS, Zhang Q, He X, et al: Afatinib versus methotrexate as second-line treatment in Asian patients with recurrent or metastatic squamous cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 3): An open-label, randomised phase III trial. Ann Oncol. 30:1831–1839. 2019. View Article : Google Scholar : PubMed/NCBI | |
Haddad R, Guigay J, Keilholz U, Clement PM, Fayette J, de Souza Viana L, Rolland F, Cupissol D, Geoffrois L, Kornek G, et al: Afatinib as second-line treatment in patients with recurrent/metastatic squamous cell carcinoma of the head and neck: Subgroup analyses of treatment adherence, safety and mode of afatinib administration in the LUX-Head and Neck 1 trial. Oral Oncol. 97:82–91. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mellor P, Furber LA, Nyarko JNK and Anderson DH: Multiple roles for the p85α isoform in the regulation and function of PI3K signalling and receptor trafficking. Biochem J. 441:23–37. 2012. View Article : Google Scholar : PubMed/NCBI | |
Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, et al: Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 3:761–769. 2013. View Article : Google Scholar : PubMed/NCBI | |
Cochicho D, Esteves S, Rito M, Silva F, Martins L, Montalvão P, Cunha M, Magalhães M, da Costa RMG and Felix A: PIK3CA gene mutations in HNSCC: Systematic review and correlations with HPV status and patient survival. Cancers (Basel). 14:12862022. View Article : Google Scholar : PubMed/NCBI | |
Jung K, Kang H and Mehra R: Targeting phosphoinositide 3-kinase (PI3K) in head and neck squamous cell carcinoma (HNSCC). Cancers Head Neck. 3:32018. View Article : Google Scholar : PubMed/NCBI | |
García-Escudero R, Segrelles C, Dueñas M, Pombo M, Ballestín C, Alonso-Riaño M, Nenclares P, Álvarez-Rodríguez R, Sánchez-Aniceto G, Ruíz-Alonso A, et al: Overexpression of PIK3CA in head and neck squamous cell carcinoma is associated with poor outcome and activation of the YAP pathway. Oral Oncol. 79:55–63. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Network, . Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI | |
Marquard FE and Jücker M: PI3K/AKT/mTOR signaling as a molecular target in head and neck cancer. Biochem Pharmacol. 172:1137292020. View Article : Google Scholar : PubMed/NCBI | |
Poetsch M, Lorenz G and Kleist B: Detection of new PTEN/MMAC1 mutations in head and neck squamous cell carcinomas with loss of chromosome 10. Cancer Genet Cytogenet. 132:20–24. 2002. View Article : Google Scholar : PubMed/NCBI | |
Sangale Z, Prass C, Carlson A, Tikishvili E, Degrado J, Lanchbury J and Stone S: A robust immunohistochemical assay for detecting PTEN expression in human tumors. Appl Immunohistochem Mol Morphol. 19:173–183. 2011. View Article : Google Scholar : PubMed/NCBI | |
Squarize CH, Castilho RM, Abrahao AC, Molinolo A, Lingen MW and Gutkind JS: PTEN deficiency contributes to the development and progression of head and neck cancer. Neoplasia. 15:461–471. 2013. View Article : Google Scholar : PubMed/NCBI | |
Psyrri A, Seiwert TY and Jimeno A: Molecular pathways in head and neck cancer: EGFR, PI3K, and more. Am Soc Clin Oncol Educ Book. 246–255. 2013. View Article : Google Scholar : PubMed/NCBI | |
Pezzuto F, Buonaguro L, Caponigro F, Ionna F, Starita N, Annunziata C, Buonaguro FM and Tornesello ML: Update on head and neck cancer: Current knowledge on epidemiology, risk factors, molecular features and novel therapies. Oncology. 89:125–136. 2015. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Liao J, Schumaker L, Carter-Cooper B, Lapidus RG, Fan X, Gaykalova DA, Mehra R, Cullen KJ and Dan H: Simultaneously targeting ErbB family kinases and PI3K in HPV-positive head and neck squamous cell carcinoma. Oral Oncol. 131:1059392022. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Liao J, Carter-Cooper BA, Lapidus RG, Cullen KJ and Dan H: Regulation of cisplatin-resistant head and neck squamous cell carcinoma by the SRC/ETS-1 signaling pathway. BMC Cancer. 19:4852019. View Article : Google Scholar : PubMed/NCBI | |
Packer LM, Geng X, Bonazzi VF, Ju RJ, Mahon CE, Cummings MC, Stephenson SA and Pollock PM: PI3K inhibitors synergize with FGFR inhibitors to enhance antitumor responses in FGFR2mutant endometrial cancers. Mol Cancer Ther. 16:637–648. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chou TC: Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 70:440–446. 2010. View Article : Google Scholar : PubMed/NCBI | |
Chakrabarty A, Sánchez V, Kuba MG, Rinehart C and Arteaga CL: Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc Natl Acad Sci USA. 109:2718–2723. 2012. View Article : Google Scholar : PubMed/NCBI | |
Meister KS, Godse NR, Khan NI, Hedberg ML, Kemp C, Kulkarni S, Alvarado D, LaVallee T, Kim S, Grandis JR and Duvvuri U: HER3 targeting potentiates growth suppressive effects of the PI3K inhibitor BYL719 in pre-clinical models of head and neck squamous cell carcinoma. Sci Rep. 9:91302019. View Article : Google Scholar : PubMed/NCBI | |
Young NR, Liu J, Pierce C, Wei TF, Grushko T, Olopade OI, Liu W, Shen C, Seiwert TY and Cohen EE: Molecular phenotype predicts sensitivity of squamous cell carcinoma of the head and neck to epidermal growth factor receptor inhibition. Mol Oncol. 7:359–368. 2013. View Article : Google Scholar : PubMed/NCBI | |
Cheng H, Yang X, Si H, Saleh AD, Xiao W, Coupar J, Gollin SM, Ferris RL, Issaeva N, Yarbrough WG, et al: Genomic and transcriptomic characterization links cell lines with aggressive head and neck cancers. Cell Rep. 25:1332–1345.e5. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zaryouh H, Van Loenhout J, Peeters M, Vermorken JB, Lardon F and Wouters A: Co-targeting the EGFR and PI3K/Akt pathway to overcome therapeutic resistance in head and neck squamous cell carcinoma: What about autophagy? Cancers (Basel). 14:61282022. View Article : Google Scholar : PubMed/NCBI | |
Zaryouh H, De Pauw I, Baysal H, Pauwels P, Peeters M, Vermorken JB, Lardon F and Wouters A: The role of Akt in acquired cetuximab resistant head and neck squamous cell carcinoma: An in vitro study on a novel combination strategy. Front Oncol. 11:6979672021. View Article : Google Scholar : PubMed/NCBI | |
Mock A, Plath M, Moratin J, Tapken MJ, Jäger D, Krauss J, Fröhling S, Hess J and Zaoui K: EGFR and PI3K pathway activities might guide drug repurposing in HPV-negative head and neck cancers. Front Oncol. 11:6789662021. View Article : Google Scholar : PubMed/NCBI | |
Izumi H, Wang Z, Goto Y, Ando T, Wu X, Zhang X, Li H, Johnson DE, Grandis JR and Gutkind JS: Pathway-specific genome editing of PI3K/mTOR tumor suppressor genes reveals that PTEN loss contributes to cetuximab resistance in head and neck cancer. Mol Cancer Ther. 19:1562–1571. 2020. View Article : Google Scholar : PubMed/NCBI | |
Martin D, Abba MC, Molinolo AA, Vitale-Cross L, Wang Z, Zaida M, Delic NC, Samuels Y, Lyons JG and Gutkind JS: The head and neck cancer cell oncogenome: A platform for the development of precision molecular therapies. Oncotarget. 5:8906–8923. 2014. View Article : Google Scholar : PubMed/NCBI | |
van Harten AM, Poell JB, Buijze M, Brink A, Wells SI, René Leemans C, Wolthuis RMF and Brakenhoff RH: Characterization of a head and neck cancer-derived cell line panel confirms the distinct TP53-proficient copy number-silent subclass. Oral Oncol. 98:53–61. 2019. View Article : Google Scholar : PubMed/NCBI | |
Cook RS, Garrett JT, Sánchez V, Stanford JC, Young C, Chakrabarty A, Rinehart C, Zhang Y, Wu Y, Greenberger L, et al: ErbB3 ablation impairs PI3K/Akt-dependent mammary tumorigenesis. Cancer Res. 71:3941–3951. 2011. View Article : Google Scholar : PubMed/NCBI | |
Mishra R, Patel H, Alanazi S, Yuan L and Garrett JT: HER3 signaling and targeted therapy in cancer. Oncol Rev. 12:3552018.PubMed/NCBI | |
Garrett JT, Sutton CR, Kurupi R, Bialucha CU, Ettenberg SA, Collins SD, Sheng Q, Wallweber J, Defazio-Eli L and Arteaga CL: Combination of antibody that inhibits ligand-independent HER3 dimerization and a p110α inhibitor potently blocks PI3K signaling and growth of HER2+ breast cancers. Cancer Res. 73:6013–6023. 2013. View Article : Google Scholar : PubMed/NCBI | |
Milewska M, Cremona M, Morgan C, O'Shea J, Carr A, Vellanki SH, Hopkins AM, Toomey S, Madden SF, Hennessy BT and Eustace AJ: Development of a personalized therapeutic strategy for ERBB-gene-mutated cancers. Ther Adv Med Oncol. 10:17588340177460402018. View Article : Google Scholar : PubMed/NCBI | |
Akbari Dilmaghani N, Safaroghli-Azar A, Pourbagheri-Sigaroodi A and Bashash D: The PI3K/Akt/mTORC signaling axis in head and neck squamous cell carcinoma: Possibilities for therapeutic interventions either as single agents or in combination with conventional therapies. IUBMB Life. 73:618–642. 2021. View Article : Google Scholar : PubMed/NCBI | |
Nunnery SE and Mayer IA: Management of toxicity to isoform α-specific PI3K inhibitors. Ann Oncol. 30 (Suppl 10):x21–x26. 2019. View Article : Google Scholar | |
Chia S, Gandhi S, Joy AA, Edwards S, Gorr M, Hopkins S, Kondejewski J, Ayoub JP, Califaretti N, Rayson D and Dent SF: Novel agents and associated toxicities of inhibitors of the pi3k/Akt/mtor pathway for the treatment of breast cancer. Curr Oncol. 22:33–48. 2015. View Article : Google Scholar : PubMed/NCBI | |
Suh KJ, Sung JH, Kim JW, Han SH, Lee HS, Min A, Kang MH, Kim JE, Kim JW, Kim SH, et al: EGFR or HER2 inhibition modulates the tumor microenvironment by suppression of PD-L1 and cytokines release. Oncotarget. 8:63901–63910. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fiedler M, Schulz D, Piendl G, Brockhoff G, Eichberger J, Menevse AN, Beckhove P, Hautmann M, Reichert TE, Ettl T and Bauer RJ: Buparlisib modulates PD-L1 expression in head and neck squamous cell carcinoma cell lines. Exp Cell Res. 396:1122592020. View Article : Google Scholar : PubMed/NCBI |