1. Introduction
Head and neck squamous cell carcinoma (HNSCC) is the sixth most prevalent cancer globally, with 890,000 new cases and 450,000 deaths each year [
1,
2,
3]. Based on etiological factors, HNSCC is classified into two types of disease: either HPV-positive or HPV-negative. The occurrence of HPV-negative HNSCC is associated with the use of tobacco and excessive consumption of alcohol, while HPV-positive HNSCC is related to human papillomavirus (HPV) infection [
4,
5,
6]. Reports indict that patients with HPV-positive HNSCC have a higher 5-year survival rate (~80%) than HPV-negative patients (~50%) [
7]. However, since HPV-positive HNSCC occurs mainly in oral and oropharyngeal tissues rather than other regions of head and neck, and may also progress to recurrent/metastatic disease in a significant portion of patients [
8], there is an urgent need for new therapeutics to treat both HPV-positive and HPV-negative HNSCC [
9,
10].
Classical therapies for HNSCC patients 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, location of the cancer, and the TNM stages of the tumor. A combination of these treatments could reduce the rate of recurrence and distant metastasis for patients with local-regional disease. However, chemotherapy remains the primary option for patients with recurrent and distant metastatic HNSCC [
11]. Cisplatin has been the most commonly used anticancer drug for treatment of advanced HNSCC, but, while many 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, leading to death within one year [
12]. Immunotherapy has been recently introduced for refractory HNSCC, but its impact has been limited [
12,
13]. Therefore, it remains of the utmost importance to find new therapeutic alternatives
The epidermal growth factor receptor (EGFR), a member of the ErbB kinases family, is notably overexpressed in 90-95% of HNSCC, and plays a crucial role in the cancer’s pathogenesis and clinical course [
14,
15,
16]. EGFR controls the activation of several essential pathways such as PI3K/Akt/mTOR and RAS-RAF-MAPK (MEK)-ERK, which regulate cell proliferation, survival, and migration [
17,
18]. In 2006, the monoclonal EGFR antibody cetuximab was approved by FDA for treatment of HNSCC in combination with the standard therapy [
19,
20,
21]. However, the use of cetuximab resulted in very limited improvement in survival rates for patients undergoing cisplatin-based therapy [
22]. In addition, small molecule kinase inhibitors like Gefitinib and Erlotinib, while effective in targeted therapies for Non-Small Cell Lung Cancer (NSCLC), have not demonstrated any benefits for HNSCC patients [
23,
24].
Increasing evidence demonstrates the importance of the ErbB family, which contains EGFR, HER2, HER3, and HER4, in the carcinogenesis of HNSCC and its response to therapies. HER2 and HER3 form heterodimers with EGFR and play a role in PI3K/Akt activation. In addition, HER2 and HER3 are also associated with resistance to EGFR and PI3K inhibitors in cancer [
25,
26]. These results indicate that targeting the ErbB family kinases could more effectively suppress HNSCC compared to solely using EGFR inhibitors [
27,
28]. In fact, FDA-approved ErbB family inhibitor, Afatinib, has shown positive results in HNSCC clinical trials and is now listed on the National Comprehensive Cancer Network (NCCN) guidelines as a third-line single agent for HNSCC treatment [
28,
29,
30,
31,
32]. Understanding the mechanisms behind resistance to Afatinib and exploring methods to avoid that resistance would be beneficial.
Phosphatidylinositol 3-Kinase (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 [
33]. The most important PI3K-p85 complex is PI3Kα/p85α. Recent studies demonstrate that mutations of PIK3CA, which codes for PI3Kα, are one of the most frequent mutations in HNSCC. In addition, PIK3CA/PI3Kα amplification or overexpression were also identified in HNSCC. Furthermore, PI3K/Akt signaling is activated in 34% of HPV-negative HNSCC tumors and 56% of HPV-positive tumors, which describe the prognosis of HNSCC [
34,
35,
36,
37]. 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 therapies. The tumor suppressor gene
PTEN encodes the PTEN protein that dephosphorylates PIP3 to inhibit the PI3K pathway. Mutations that resulted in PTEN loss or deceased PTEN expression were frequently observed in HPV-positive and negative HNSCC [
38,
39]. These alterations further result in the activation of PI3K/Akt [
40]. Therefore, PI3Ks is one of the most attractive targets for the treatment of HNSCC [
41,
42,
43].
We previously reported that co-targeting the ErbB family and PI3K, through a combination of Afatnib and Copanlisib, suppressed growth of HPV-positive HNSCC [
44]. In this study, we further explored whether this combination is also effective at suppressing the growth of HPV-negative HNSCC. We found that the combination of Afatinib and Copanlisib led to significant inhibition of cell proliferation and induction of apoptosis in HPV-negative HNSCC. Furthermore, this combination suppressed tumor growth in xenograft models, while having no obvious effect on body weight loss in mice. These results highlight the feasibility of this combination for the treatment of HPV-negative HNSCC.
2. Materials and Methods
2.1. Cell Culture
HNSCC cell lines, Cal27, and FaDu were purchased from ATCC and were authenticated by short tandem repeat analysis (STR) and tested for mycoplasma contamination in the Translational Core Facility of the University of Maryland Marlene and Stewart Greenebaum Cancer Center. All cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 2 mM glutamine, and 100 U/mL penicillin and streptomycin (Gibco).
2.2. Antibodies and Inhibitors
The following antibodies were purchased from Cell Signaling: phospho-Akt-S473 (CST-4508), phospho-Akt-T308 (CST-9275), Akt (CST-2938), phospho-S6K-T389 (CST-9205), S6K (CST-9202), phospho-HER2-Y1248 (CST-2247), HER2 (CST-4290), phospho-HER3-Y1289 (CST-2842), HER3 (CST-12708), C-caspase-3 (CST-xxx), and β-actin (CST-4967). Gefitinib, Erlotinib and Afatinib and all PI3K inhibitors were purchased from Selleck Chemicals.
2.3. Cell Lysis and Western Blot Analysis
Cell lysis and Western blot analysis were performed as previously described [
44,
45].
2.4. Analyzing apoptosis by Annexin V/propidium iodide staining
Apoptosis analysis by Annexin V/propidium iodide staining was performed as previously described [
44,
45].
2.5. Cell Viability Assay
Cell viability was assessed by sulforhodamine B (SRB) staining as described previously [
46]. Each experiment was performed in triplicate. To determine synergy of drug combination, the combination index values were determined according to the Chou–Talalay method [
47] using CalcuSyn software.
2.6. Tumor Xenograft Formation in Mice
FaDu cells were subcutaneously injected on the right flank Nu/nu mice (Envigo, Frederick MD) at a density of 0.5 x106 cells/ml in the presence of 33% MatrigelTM (Fisher Scientific). When tumors reached approximately 200 mm3, mice were randomized to one of four treatment groups (7 mice/group): vehicle control, Copanlisib (6 mg/kg, IP), Afatinib (6 mg/kg, PO), or a combination of Copanlisib and Afatinib. Tumor volume was measured twice per week using electronic calipers and animals were weighed 5 days per week. Tumor volume was calculated as (L x W2)/2, where W is the smaller dimension and L is tumor length. Mice were euthanized on Day 32 of the treatment, and the tumors were excised, weighed, fixed, and frozen.
2.7. Statistical Analysis
All in vitro data are shown as mean ± SD and animal data was shown as mean ± SEM. Statistical analysis was performed using GraphPad Prism version 7.04 (GraphPad Software Inc.).
4. Discussion
In this study, we tested the efficacy of co-inhibiting the ErbB family and PI3K through the combination of Afatinib and Copanlisib to inhibit HPV-negative HNSCC. Our results showed that the combination of Afatinib and Copanlisib caused dramatic inhibition of cell proliferation and suppressed cell survival in vitro in comparison to treatment with either Afatinib or Copanlisib alone. Notably, the combination led to significant inhibition of xenograft tumor growth without affecting the body weight of the mice. These results suggest that the combination of Afatinib and Copanlisib may have clinical potential for the treatment of HPV-negative HNSCC.
The EGFR/ErbB and PI3K/Akt/mTOR pathways have been the most attractive pathways to target for treatment of HNSCC due to over-expression or activating mutation of PIK3CA and loss function mutations of PTEN [
6,
50,
51,
52,
53]. It has been reported that constitutive activation of PI3K/Akt/mTOR pathway due to the alterations in PIK3CA, PTEN, Akt or mTOR is associated with resistance to EGFR inhibitor [
6]. Our data showed that treatment with Afatinib alone cannot completely block the phosphorylation of Akt (
Figure 6). Furthermore, it has been reported that PI3K inhibition led to increased phosphorylation and total levels of HER3, which confer resistance to PI3K inhibitors [
48,
49,
54,
55,
56]. Our data also showed that Copanlisib increased phospho-HER3 (Y1289), which was counteracted by the addition of Afatinib (
Figure 6). Notably, the combination of Copanlisib and Afatinib induced significant caspase-3 cleavage in addition to the complete inhibition of ErbB and PI3K/Akt pathways (
Figure 6). These results provide a rationale for the co-inhibition of ErbB and PI3K as a method to treat HNSCC.
We recently 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, indicating increased efficacy of this combination in HPV+ HNSCC [
44]. A publication by Milewska et al, reported that cell lines from multiple cancers, including HNSCC with PIK3CA mutations, are sensitive to the combination of Afatinib and Copanlisib [
57]. As the basal level of PI3K/Akt is also high in HPV-negative HNSCC and plays essential roles in the regulation of growth, metastasis, and sensitivity to chemo- and targeted therapies [
6,
42,
58,
59], it would be reasonable to predict that this combination would also be beneficial in HPV-negative HNSCC with upregulated PI3K/Akt signaling. Afatinib has shown positive results in HNSCC clinical trials and is now listed on the National Comprehensive Cancer Network (NCCN) guidelines as a third-line single agent for HNSCC treatment[
28,
29,
30,
31,
32]. Our results indicate that the combination of Afatinib with Copanlisib would more effectively suppress HNSCC in patients with refractory disease.
Immunotherapy, including immune checkpoint blockade (ICB) targeting PD-L1/PD-1 using PD-1 inhibitor, Nivolumab or Pembrolizumab, was another important advancement in the treatment of advanced HNSCC. Afatinib modulates PD-L1 expression in multiple cancers, including gastric cancer [
60]. In addition, it has been reported that PI3K inhibitors such as BKM120 deceased the expression of PD-L1 in HNSCC cells [
61]. It would be interesting to determine the effects of Afatinib, Copanlisib, and their combination on the expression of PD-L1 in HNSCC and immune cells, such as T-cells, and the impact of the combination of Afatinib and Copanlisib on immunotherapy.
Author Contributions
Xinyan Geng: Data curation, Formal analysis, Writing – original draft. Shirin Azarbarzin: Data curation, Formal analysis. Zejia Yang: Data curation, Formal analysis. Rena G. Lapidus: Data curation, Formal analysis. Xiaoxuan Fan: Data curation, Formal analysis. Yong Teng: Formal analysis, Writing – review & editing. Ranee Mehra: Formal analysis, Writing – review & editing. Kevin J. Cullen: Conceptualization, Data curation, Formal analysis, Supervision, Writing – review & editing, Funding acquisition. Hancai Dan: Conceptualization, Data curation, Formal analysis, Supervision, Writing – review & editing, Funding acquisition.
Figure 1.
Afatinib more effectively inhibited cell proliferation in HPV-negative HNSCC compared to Gefitinib and Erlotinib. A and B. Cal27 (A) and FaDu (B) cells were treated with DMSO or increasing concentrations of Gefitinib, Erlotinib, and Afatinib for 96 hours and cell proliferation was measured by SRB assay and EC50 were determined by GraphPad Prism version 7.04.
Figure 1.
Afatinib more effectively inhibited cell proliferation in HPV-negative HNSCC compared to Gefitinib and Erlotinib. A and B. Cal27 (A) and FaDu (B) cells were treated with DMSO or increasing concentrations of Gefitinib, Erlotinib, and Afatinib for 96 hours and cell proliferation was measured by SRB assay and EC50 were determined by GraphPad Prism version 7.04.
Figure 2.
Copanlisib more effectively inhibited cell proliferation compared to other PI3K inhibitors in Cal27 cells. A. Cal27 cells were treated with DMSO or increasing concentrations of Copanlisib and other PI3K inhibitors for 96 hours and cell proliferation was measured by SRB assay. The growth curves are shown. The experiments were performed in triplicate. The associated EC50 to the 6 inhibitors were determined by GraphPad Prism version 7.04. B. EC50 to six PI3k inhibitors in Cal27 and FaDu cells were listed.
Figure 2.
Copanlisib more effectively inhibited cell proliferation compared to other PI3K inhibitors in Cal27 cells. A. Cal27 cells were treated with DMSO or increasing concentrations of Copanlisib and other PI3K inhibitors for 96 hours and cell proliferation was measured by SRB assay. The growth curves are shown. The experiments were performed in triplicate. The associated EC50 to the 6 inhibitors were determined by GraphPad Prism version 7.04. B. EC50 to six PI3k inhibitors in Cal27 and FaDu cells were listed.
Figure 3.
Synergistic inhibition of cell proliferation by combination of Afatinib and Copanlisib in vitro. A and C. Cal27 (A) and FaDu (C) cells were treated with different concentrations of Afatinib, Copanlisib, or their combinations for 96 hours and cell proliferation was measured by SRB assay. The experiments were performed in triplicate. B and D. The combination index values (CI values) for different combinations were determined using CalcuSyn, Version 2.0 (C and D).
Figure 3.
Synergistic inhibition of cell proliferation by combination of Afatinib and Copanlisib in vitro. A and C. Cal27 (A) and FaDu (C) cells were treated with different concentrations of Afatinib, Copanlisib, or their combinations for 96 hours and cell proliferation was measured by SRB assay. The experiments were performed in triplicate. B and D. The combination index values (CI values) for different combinations were determined using CalcuSyn, Version 2.0 (C and D).
Figure 4.
Inhibition of HNSCC growth by combination of Afatinib and Copanlisib in vivo. A, B, and C. FaDu cells were inoculated into mice. When tumors reached approximately 200 mm3, mice were randomized to one of four treatment groups (7 mice/group): vehicle control, Copanlisib (6 mg/kg, IP), Afatinib (6 mg/kg, PO), or a combination of Copanlisib and Afatinib. The treatments were performed for 32 days. The xenograft tumor volumes (A), final average weights of tumors (B), and average body weights of mice (C) in each group were compared. (*P<0.05, **P<0.01).
Figure 4.
Inhibition of HNSCC growth by combination of Afatinib and Copanlisib in vivo. A, B, and C. FaDu cells were inoculated into mice. When tumors reached approximately 200 mm3, mice were randomized to one of four treatment groups (7 mice/group): vehicle control, Copanlisib (6 mg/kg, IP), Afatinib (6 mg/kg, PO), or a combination of Copanlisib and Afatinib. The treatments were performed for 32 days. The xenograft tumor volumes (A), final average weights of tumors (B), and average body weights of mice (C) in each group were compared. (*P<0.05, **P<0.01).
Figure 5.
A combination of Afatinib and Copanlisib increased cell apoptosis compared to Afatinib or Copanlisib treatment alone. A and B. Cal27CP (A) and FaDu (B) cells were treated with vehicle control, Copanlisib, Afatinib, or a combination for 48 hours, and cell apoptosis was analyzed by Annexin V/propidium iodide staining. The experiments were performed in triplicate, early and late-stage apoptotic, and dead cells were counted, and statistical analysis was performed. P values < 0.05 were considered to be statistically significant. Note: in A, (*P<0.05, **P<0.01, ***P<0.0001). .
Figure 5.
A combination of Afatinib and Copanlisib increased cell apoptosis compared to Afatinib or Copanlisib treatment alone. A and B. Cal27CP (A) and FaDu (B) cells were treated with vehicle control, Copanlisib, Afatinib, or a combination for 48 hours, and cell apoptosis was analyzed by Annexin V/propidium iodide staining. The experiments were performed in triplicate, early and late-stage apoptotic, and dead cells were counted, and statistical analysis was performed. P values < 0.05 were considered to be statistically significant. Note: in A, (*P<0.05, **P<0.01, ***P<0.0001). .
Figure 6.
Inhibition of both ErbB and PI3K/Akt pathways and induced caspase cleavage by combination of Afatinib and Copanlisib. A-B. Combination of Copanlisb and Afatinib effectively blocks the phosphorylation of HER2, HER3 and Akt and induces more caspase 3 cleavage. Cal27 (A), and FaDu (B) cells were treated with increasing concentrations of Copanlisib, Afatinib (0.5 μM), or their combination for 24 hours before lysed. The indicated proteins were detected by Western blot analysis.
Figure 6.
Inhibition of both ErbB and PI3K/Akt pathways and induced caspase cleavage by combination of Afatinib and Copanlisib. A-B. Combination of Copanlisb and Afatinib effectively blocks the phosphorylation of HER2, HER3 and Akt and induces more caspase 3 cleavage. Cal27 (A), and FaDu (B) cells were treated with increasing concentrations of Copanlisib, Afatinib (0.5 μM), or their combination for 24 hours before lysed. The indicated proteins were detected by Western blot analysis.