Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 1.
Published in final edited form as: Cancer J. 2022 Sep-Oct;28(5):354–362. doi: 10.1097/PPO.0000000000000615

Therapeutic Targeting of FGFR Signaling in Head and Neck Cancer

Zechen Wang 1, Karen S Anderson 1,2,*
PMCID: PMC9523489  NIHMSID: NIHMS1821642  PMID: 36165723

Abstract

Squamous cell carcinoma of the head and neck (HNSCC) is the sixth most prevalent cancer worldwide, with an annual incidence of 600,000 new cases. Despite of advances in surgery, chemotherapy, and radiotherapy, the overall survival for HNSCC patients hasn’t been significantly improved over the past several decades. FGF/FGFR genomic alterations are frequently detected in HNSCC, including amplification, activating mutation, and chromosomal rearrangement. Among them, FGFR1 amplification, FGF amplifications, and FGFR3 mutations are the most prevalent. In addition, FGF/FGFR expression has also been observed in the majority of HNSCC. However, the prognostic value of FGF/FGFR aberrations remains unclear, especially for gene amplification and overexpression. Nonetheless, FGF/FGFR has been a promising target for HNSCC treatment, and recent preclinical studies demonstrate the potential of the combination treatment regimens involving FGFR inhibitors on HNSCC. Therefore, there are a number of FGFR inhibitors currently in clinical trials for the treatment of head and neck cancers.

Keywords: Head and neck cancer, FGF, FGFR, genomic alterations, overexpression, FGFR inhibitors, targeted therapy, combination treatment


Squamous cell carcinoma of the head and neck (HNSCC) is the sixth most prevalent cancer worldwide, with a global incidence of over 600,000 new cases every year1. HNSCC commonly arises from the squamous cells of the mucosal lining of oral cavity, oropharynx, larynx, and hypopharynx, accounting for 90% of the head and neck cancers (HNC)2. HNC can also originate from the salivary glands, sinuses, and muscles or nerves in the head and neck region, but they are much less common than HNSCC3. The two classical risk factors for HNSCC are alcohol consumption and tobacco use, and in recent years, human papillomavirus (HPV) has also been recognized as a high-risk factor, especially for oropharyngeal SCCs (OPSCC)2. In spite of the progress and advances in surgery, radiotherapy, and chemotherapy over the past several decades for the treatment of HNSCC, the overall survival rate for HNSCC hasn’t been significantly improved, with the five-year survival rate around 50% and over 300,000 deaths annually1,4. Therefore, there is still a significant unmet need for the development of new therapies, including targeted therapies, immunotherapies, and combination therapies, for the treatment of HNSCC patients.

Fibroblast growth factor receptors (FGFRs) belong to the receptor tyrosine kinase (RTK) superfamily5 and consist of four family members (FGFR1–4)6. All FGFRs share a common construct composed of one extracellular domain, one single-pass transmembrane helix, and one intracellular tyrosine kinase domain7. The extracellular region of FGFRs contains three immunoglobulin-like domains responsible for the ligand binding7. FGFs, the corresponding growth factor ligands, can mediate a variety of cellular responses by binding to FGFRs with the cofactors, heparin sulfate or Klotho. FGF binding can cause the dimerization and autophosphorylation of FGFRs, which can lead to the activation of FGFR kinase, followed by the recruitment and activation of downstream cellular signaling pathways7. The four major signaling pathways controlled by FGFRs are RAS/RAF/ERK, PI3K/Akt, STATs, and PLCγ1 (Fig. 1). Therefore, FGFR signaling plays a critical role in various biological processes, including embryogenesis, organogenesis, tissue repair and wound healing, and tumor angiogenesis, etc7.

Figure 1. FGF/FGFR intracellular signaling transduction network.

Figure 1.

FGF ligands and the cofactors, heparin sulfate (HS) or Klotho, will first bind to the immunoglobulin-like domains in the extracellular region of FGFRs, which will lead to FGFR dimerization and autophosphorylation. Tyrosine autophosphorylation sites of FGFR are indicated in red dots. FGFR autophosphorylation can activate FGFR kinase activity and recruit downstream signaling molecules. The four major signaling pathways that will be recruited and activated by FGFR are RAS/RAF/ERK (red), PI3K/AKT (light green), STAT1/3/5 (gray), and PLCγ1 (blue). The activation of PLCγ1 can cleave phospholipid phosphatidylinositol 4,5-biphosphate (PIP2) into diacyl glycerol (DAG) and inositol 1,4,5-trisphosphote (IP3), which can further activate PKC and intracellular Ca2+ signaling, respectively. I/II/III, immunoglobulin-like domain I/II/III.

FGFR genomic alterations have been implicated in ~7% of all human malignancies, including urothelial, breast, endometrial, lung, and ovarian cancer8. In particular, FGFR aberrations are one of the most frequent RTK genomic alterations in HNSCC9, making FGF/FGFR axis a promising target for the development of new treatment options for HNSCC patients. FGF/FGFR genomic alterations can be divided into ligand-dependent aberrations, which involves FGF genomic alterations, and ligand-independent aberrations that consist of FGFR aberrations10 (Fig. 2). FGFR gene amplifications, gain-of-function activating mutations, and chromosomal rearrangements/gene fusions are the three major classes of FGFR genomic alterations11. In this review, we will summarize the recent studies of FGFR genomic alterations in HNSCC and the current progress in terms of the development of FGFR-oriented therapeutic strategies for HNSCC patients, including FGFR targeted therapies as monotherapies and in combination with other chemo-, radio-, target-, and immuno-therapies.

Figure 2. FGF/FGFR genomic alterations in HNSCC.

Figure 2.

Ligand-dependent aberrations include FGF amplifications and mutations. Wild-type FGF ligands are indicated in blue spheres and FGFs with mutations are in dark red spheres. Ligand-independent aberrations consist of FGFR amplifications, mutations, and chromosomal rearrangements/gene fusions. Wild-type FGFRs are in red, FGFR mutations are indicated by orange stars, and gene fusion partners are in green and blue. The figure was created with BioRender.com.

FGFR genomic alterations in HNSCC

FGF/FGFR gene deregulation, including gene amplification, mutation, and chromosomal rearrangement, has been detected in ~30–50% HNSCC patients based on the genomic analysis of a The Cancer Genome Atlas (TCGA) HNSCC cohort12,13. Among them, FGFR1 gene amplification, FGF3/4/19 gene amplifications, and FGFR3 mutations are the most frequent FGF/FGFR genomic alterations9,12,13 (Table 1 and Fig. 2).

Table 1.

FGF/FGFR genomic alterations in HNSCC

Genes Genomic alterations Description Frequency in HNSCC
FGF Amplifications FGF3, FGF4, FGF6, FGF19, FGF23 ~20–25%
FGFR1 Amplification Predominantly detected in HPV-negative HNSCC ~5–30%
Mutations ~1.4%
FGFR2 Mutations FGFR2 N549D, N549K;
Predominantly detected in HPV-positive HNSCC
~4.3%-17.6%
FGFR3 Mutations FGFR3 V50I, S131L, E157K, R248C, S249C, A393E, K413N, E629K, K650Q, R671G, G697C, D764H, D788N;
Predominantly detected in HPV-positive HNSCC
~5.8–62%
Fusions FGFR3-TACC3;
Predominantly detected in HPV-positive HNSCC
~2.5–3.7%
FGFR4 Mutations ~8%
SNP Can give rise to FGFR4 G388R ~33.8%

SNP, single nucleotide polymorphism.

FGF ligand amplification and overexpression

FGF2 is the most well-studied FGF ligand in HNSCC. FGF2 has been reported to be highly expressed in up to 60% of HNSCC tumor14. Multiple studies observed that FGF2 is over-expressed in HNSCC tumor and cell lines15,16, and FGF2 overexpression is related to tumor aggressiveness and unfavorable prognosis17,18, while other reports suggest that the a statistically significant enhancement of FGF2 expression in their studies is less clear19. Marshall and colleagues demonstrated that FGF2 are frequently co-expressed with FGFRs in the majority of HNSCC cell lines they tested, which can form an autocrine loop to drive oncogenesis20. FGF2 and FGFRs are more highly expressed in early stages of HNSCC and well differentiated tumors1517, which indicates that FGFR signaling may play a role in early-stage tumor initiation and development. This is in consistent with the observation that most of the potentially malignant oral lesions that transformed into oral squamous cell carcinoma express FGF2 and FGFR216. In addition to FGF2 overexpression, FGF2 single nucleotide polymorphism is also reported to be associated with overall survival of the HNSCC patients treated by chemoradiotherapy18. Other than FGF2, gene amplifications of FGF3, FGF4, and FGF19 have also been detected in more than 20% of HNSCC patients in one TCGA cohort12. FGF19 mainly signals through FGFR4 and FGF19 amplification and overexpression is related to poor prognosis for HNSCC patients21.

FGFR1 amplification and overexpression

FGFR1 gene amplification is one of the most frequent FGF/FGFR genomic alterations in HNSCC, which has been reported in ~5–30% HNSCC patients13,22,23. FGFR1 gene amplification is predominantly detected in HPV-negative HNSCC patients22,24,25 and is more prevalent in laryngeal (LPSCC) and hypopharyngeal squamous cell carcinoma (HPSCC)26,27, which is consistent with the fact that HPV mainly affect oropharyngeal squamous cell carcinoma (OPSCC), rather than LPSCC and HPSCC4. In addition, FGFR1 and FGFR3 amplifications have also been observed in some rare HNSCC, such as external auditory canal SCC28. The prognostic value of FGFR1 amplification in HNSCC is debatable, and recently, more studies reported that FGFR1 amplification is not related to patient overall survival29,30. One of the plausible explanations is that FGFR1 amplification is not associated with FGFR1 expression level25,30. On the other hand, FGFR1 overexpression has been observed in ~10–82% of HNSCC patients26,31,32 and there is no difference in FGFR1 expression between HPV-positive and HPV-negative HNSCC patients25. However, at present, conflicting results in terms of FGFR1 expression as a prognosis biomarker have been reported. Multiple studies demonstrated that FGFR1 overexpression is related to poor prognosis in HPV-negative HNSCC patients31,33,34, whereas others suggest the contrary26,29,35. The perceived discrepancy may be attributed to a number of factors including: the difference in the cutoff value for FGFR1 expression, various anatomic positions of HNSCC, different sample size, and distinct patient ethnicities from different studies. In line with this suggestion, Fisher and colleagues reported that the sensitivity of AZD4547, a selective FGFR inhibitor, is not associated with FGFR expression level in a panel of ten HNSCC cell lines they examined, including both HPV-positive and HPV-negative cells32. Therefore, the prognostic value of FGFR1 amplification and overexpression should be further examined in a larger patient cohort and stratified based on HPV status and anatomic positions, etc. Additionally, other than gene amplification, FGFR1 mutations are also detected in ~1.4% HNSCC patients36.

FGFR2 mutations and overexpression

Genomic alterations in FGFR2 are less prevalent than FGFR1 and FGFR3 in HNSCC. FGFR2 mutations, such as FGFR2 N549D and N549K, have been detected in ~4.3%-17.6% HNSCC patients13,36,37 (Table 1 and Fig. 2). Unlike FGFR1 gene amplification, FGFR2 mutations are mainly enriched in HPV-positive HNSCC patients37. In addition, FGFR2 is more expressed in HNSCC tumor tissue than normal epithelium16,19 and the majority of HNSCC expresses FGFR232.

FGFR3 mutations and fusions

FGFR3 mutations have been implicated in ~5.8–24% of HNSCC patients36,38,39 (Table 1 and Fig. 2), and FGFR3 S249C is the most prevalent gain-of-function mutation37,40. Of note, FGFR3 G649C, which is a constitutively activating mutation in FGFR3 kinase domain, was also reported in 62% (44/71) of one Japanese HNSCC cohort41. In addition, FGFR3-TACC3 fusion is also reported in ~2.5–3.7% of HNSCC patients24,42. Intriguingly, different HNSCC patients may have distinct breakpoints for FGFR3-TACC3 fusion42, which may affect the tumorigenic abilities of the FGFR3-TACC3 fusions43. FGFR3-TACC3 is considered as a constitutively activated oncogene and has also been detected in other cancers, including bladder cancer44 and glioblastoma45. Similar to FGFR2 mutations, FGFR3 mutations and fusions are also primarily detected in HPV-positive HNSCC25 and FGFR3 mutations are usually associated with worse 3-year disease-free survival39. Furthermore, a number of studies reported that FGFR3 is over-expressed in HNSCC tumor tissue as compared with the non-cancerous control tissue16,19. However, contrary to these results, other studies by Mässenhausen and colleagues suggest that FGFR3 is more highly expressed in normal mucosa, compared to the HNSCC primary tumor, and FGFR3 expression further decreased as tumor progressed46. And in accordance with FGFR1 overexpression, the prognostic value of FGFR3 overexpression is also contradictive and there has been accumulating evidence showing that FGFR3 expression is not related to patient overall survival32,33,47.

FGFR4 genomic alterations and overexpression

FGFR4 is much less studied than other FGFRs in HNSCC. FGFR4 is highly expressed in 16–39% of HNSCC patients35, and FGFR4 overexpression has been reported to be associated with poorer overall survival of HNSCC patients in several studies48,49. In addition, one of the FGFR4 single nucleotide polymorphisms that gives rise to the FGFR4 G388R mutation is demonstrated to be associated with advanced HNSCC tumor stages and reduced disease-free survival48. Furthermore, FGFR4 mutations are also observed in ~8% of HNSCC patients based on the analysis from one of the TCGA HNSCC cohorts13.

Current progress in the development of FGF/FGFR-oriented therapies for HNSCC

Due to the prevalence of FGF/FGFR genomic alterations and overexpression in HNSCC, FGF/FGFR axis has been a promising target for the treatment of HNSCC patients. Constitutively activated aberrations, such as gain-of-function mutations and chromosomal rearrangements, are usually regarded as driver mutations for oncogenesis, and the effects of FGFR inhibitors on the tumors with those constitutively activated FGFR genomic aberrations have been extensively demonstrated both preclinically and clinically5052. However, the sensitivity of FGFR inhibitors varies dramatically among the HNSCC with wild-type FGF/FGFR overexpression and/or FGFR amplifications32,53. Therefore, recent studies have been primarily focusing on the development of new combination treatments that concatenate FGFR inhibitors with chemo-, radio-, immuno-, and/or other targeted therapies.

FGFR inhibitors and chemotherapies

Surgery, chemotherapy, and radiation are the three typical standard-of-care strategies for treatment of HNSCC patients. Aytatli and colleagues indicated that the activation of FGFR/Akt/SOX2 signaling pathway is upregulated in taxol-resistant HNSCC cell lines and AZD4547 can potentiate the anti-tumor effects of taxol and cisplatin in those cell lines54. SOX2 is SRY-Box transcription factor 2, which is essential for the maintenance of the stem cell features in tumors55. Consistent with this finding, McDermott and colleagues found FGF2 expression is increased in head and neck cancer stem cells after cisplatin treatment56, suggesting that FGF/FGFR signaling may play a role in the resistance of HNSCC to cisplatin. Likewise, Holzhauser and colleagues revealed that Erdafitinib (JNJ-42756493), another selective FGFR inhibitor, can be synergistic or additive with cisplatin and docetaxel in most of the tonsillar (TSCC) and base of tongue (BOTSCC) squamous cell carcinoma cell lines they tested, regardless of FGFR mutation status57.

FGFR inhibitors and radiotherapies

Ishigami and colleagues performed microarray analysis and found that FGFR3 is overexpressed in oral squamous cell carcinoma (OSCC) cell lines that are resistant to radiation therapy, compared to radiosensitive OSCC cell lines58. When FGFR3 is knocked down by siRNA or inhibited by selective FGFR inhibitors, radiosensitivity can be induced in the radioresistant cell lines59. And consistent with that observation, multiple studies have reported that AZD4547 can also augment the response to radiotherapy of HNSCC both in vitro and in vivo, by inducing apoptosis and potentially inhibiting mTOR signaling pathway32,53. However, this AZD4547-mediated radio-sensitization is limited to only the HNSCC cell lines that are sensitive to AZD4547 monotherapy, and the cell lines that are not responsive to AZD4547 cannot exhibit radio-sensitization32.

FGFR inhibitors and other targeted therapies

There has been accumulating evidence indicating that FGFR expression level is not related to FGFR inhibitor sensitivity in HNSCC with wild-type FGFR overexpression and/or FGFR amplifications32,53. One of the possible explanations is that wild-type FGFR overexpression is not the only driver factor for HNSCC oncogenesis and the activation of other alternative signaling pathways can lead to the resistance to FGFR inhibition, which is a common mechanism for the resistance of targeted therapies in cancer60. This idea is verified by the study showing that some HNSCC cell lines can rely on multiple RTKs for full growth and survival, including FGFRs, ERBB family, and MET61. And inhibiting all of those RTKs simultaneously can achieve synergistic effects in those cell lines61. Likewise, FGF/FGFR genomic alterations and overexpression may also cause the resistance to other targeted therapies. Gyanchandani and colleagues revealed that FGF2 and FGFR3 expression can be upregulated after long-term treatment of bevacizumab, a VEGF-A monoclonal antibody, which can mediate the resistance to bevacizumab62. Combination treatment of bevacizumab with a selective FGFR inhibitor, PD-173074, can sensitize the HNSCC tumor to bevacizumab in vivo. Similarly, it has also been reported that upregulation of FGF2 expression and FGFR3 phosphorylation after selumetinib treatment, a MEK inhibitor, can mediate the resistance to selumetinib63. In addition, high throughput screening identified that FGFR inhibitors and EGFR inhibitors can achieve synergistic effects both in HNSCC cell lines and xenografts64. This is further validated by Koole and colleagues who demonstrated that EGFR signaling can mediate the AZD4547 resistance in some HNSCC cell lines and AZD4547 and gefitinib can synergistically inhibit the proliferation of those cell lines31. Furthermore, Kumar and colleagues confirmed that the secretion of HGF and FGF2 by cancer-associated fibroblast (CAF) and HNSCC tumor, respectively, can mediate the interaction between HNSCC tumor and CAF65. In line with this finding, FGFR inhibitors and MET inhibitors can achieve additive inhibitory effects on HNSCC growth both in vitro and in vivo.

In addition to co-targeting FGFR with other RTKs, the combination between FGFR inhibitors and inhibitors for other kinases has also been explored. Singleton and colleagues performed a kinome RNAi screen and identified that FGFR inhibitors and mTOR inhibitors can achieve synergism in HNSCC cell lines regardless of FGFR mutations66. Similar to this, PI3K inhibitors have also been reported to be synergistic or additive with FGFR inhibitors in TSCC/BOTSCC cell lines disregarding FGFR and PI3K mutation status57.

FGFR inhibitors and immunotherapies

Immunotherapy has opened a new era for cancer treatment over the past decades. FDA approved two PD-1 monoclonal antibodies, pembrolizumab and nivolumab, for the treatment of HNSCC67. However, only a small portion of HNSCC patients (~10–20%) can benefit from immune checkpoint inhibitors68. The combinatorial effects between FGFR inhibitors and immunotherapies have been more extensively investigated in other cancers, including breast69, lung70, and bladder cancers71, whereas studies of such combination are still very limited in HNSCC. Kono and colleagues demonstrated that PD-173074 and anti-PD-1 antibodies showed synergistic anti-tumor effects in HNSCC xenografts, and FGFR inhibitors can recruit CD4+ and CD8+ T cells to the tumor environment72. Furthermore, they also revealed that FGFR inhibitors can upregulate the expression of MHC class I and MHC class II both in vitro and in vivo by inhibiting MAPK signaling pathway72.

FGFR inhibitors and tumor microenvironment

Tumor microenvironment is essential for the regulation of tumor initiation, progression, metastasis, and drug sensitivity73. It has been reported that FGF2 and FGFR1 expression in the fibroblasts of OSCC is related to tumor metastasis and poor prognosis74. And consistent with that observation, in vitro studies showed that co-culturing with fibroblasts can enhance the proliferation of HNSCC cell lines75. Furthermore, this fibroblast-enhanced tumor growth can also be suppressed by PD-174074 in vivo through FGFR inhibition, which can lead to stromal component shrinkage, decreased proliferation, and increased apoptosis75.

Current FGFR inhibitors in clinical trials

Based on the progress and results from preclinical studies, FGFR inhibitors show substantial potential for treatment of HNSCC patients. In fact, one HNSCC patient with gene amplifications of multiple FGFs, including FGF3, FGF4, FGF6, FGF19, and FGF23, achieved a complete response to a FGFR inhibitor76. In addition, another head and neck cancer patient with FGF3 and FGF19 amplifications achieved stable disease for 4 months after ponatinib treatment, a multi-target FGFR inhibitor77. In two phase I clinical trials of rogaratinib, out of 10 HNSCC patients who received rogaratinib, two patients achieved partial response and four patients showed stable disease51,52, indicating the efficacy of FGFR inhibitors in HNSCC patients. Furthermore, 1 out of 9 head and neck cancer patients achieved partial response in a phase I clinical trial of pemigatinib78. However, the patient with the partial response did not possess any known FGF/FGFR aberrations78 and the underlying mechanism(s) of the response still needs to be further explored. In addition, anlotinib is another multi-target FGFR inhibitor that targets at FGFR, VEGFR, PDGFR, and c-Kit. One phase II clinical trial of anlotinib on recurrent or metastatic nasopharyngeal carcinoma patients revealed that out of 39 enrolled patients, 8 patients achieved partial response and 20 patients reached stable disease79.

Therefore, there are several FGFR inhibitors currently in clinical trials for treatment of head and neck cancers (Table 2), including [225Ac]-FPI-1966, nintedanib, dovitinib, anlotinib, ICP-192, and AZD4547. [225Ac]-FPI-1966 is a FGFR targeted alpha therapeutic that conjugate vofatamab, a FGFR3 monoclonal antibody, with actinium-225, an alpha particle emitting radionuclide. Nintedanib and dovitinib are both multi-target FGFR inhibitors. Nintedanib can target at FGFR, VEGFR, and PDGFR, and dovitinib can inhibit FGFR, VEGFR, c-Kit, and Flt3. ICP-192 is an irreversible selective FGFR inhibitor that can covalently bind to FGFR. Anlotinib is now currently examined in phase II clinical trials as a part of combination therapies. Anlotinib and chemoradiation are currently tested on locally advanced nasopharyngeal carcinoma patients, and the combination of anlotinib and PD-1 monoclonal antibody are investigated on advanced head and neck cancer. In addition, lenvatinib, a multi-target FGFR inhibitor that targets at FGFR, VEGFR, PDGFR, c-Kit, and RET, is currently in clinical trials for the treatment of adenoid cystic carcinomas of the salivary glands80 (Table 2).

Table 2.

Representative clinical trials of FGFR inhibitors for head and neck cancers

Inhibitors Inhibitor types ClinicalTrials.gov
ID
Phase Conditions or diseases Status
Lenvatinib Multi-kinase FGFR inhibitor (FGFR, VEGFR, PDGFR, c-Kit, RET) NCT02860936 2 Adenoid Cystic Carcinomas of the Salivary Glands Completed
Nintedanib Multi-kinase FGFR inhibitor (FGFR, VEGFR, PDGFR) NCT03292250 2 HNSCC Completed
Dovitinib Multi-kinase FGFR inhibitor (FGFR, VEGFR, c-Kit, Flt3) NCT01831726 2 Hematologic malignancies and/or solid tumors, including head and neck cancers Completed
ICP-192 Selective FGFR inhibitor NCT03758664 1/2 Solid tumors, including head and neck cancers Recruiting
AZD4547 Selective FGFR inhibitor NCT02465060 2 Lymphomas, multiple myeloma, and advanced malignant solid neoplasm, including head and neck carcinoma Recruiting
Rogaratinib Selective FGFR inhibitor NCT01976741 1 Refractory, locally advanced or metastatic solid tumors, including head and neck cancer Completed
[225Ac]-FPI-1966 FGFR targeted alpha therapeutics NCT05363605 1/2 Advanced solid tumors, including head and neck squamous cell carcinoma Recruiting
Anlotinib + Chemoradiation Anlotinib (FGFR, VEGFR, PDGFR, c-Kit inhibitor) NCT05232552 2 Locally advanced nasopharyngeal carcinoma Recruiting
Anlotinib + Penpulimab Anlotinib (FGFR, VEGFR, PDGFR, c-Kit inhibitor), Penpulimab (PD-1 monoclonal antibody) NCT04203719 2 Advanced head, neck and chest cancers Unknown

Besides the clinical trials that include HNC patients, there are a number of clinical trials of FGFR inhibitors on solid tumors (Table 3), which do not necessarily include HNC patients but can be applied for the treatment of HNC in future studies. These clinical trials include multi-target FGFR inhibitors (lucitanib, 3D185, sulfatinib, MAX-40279–01, TT-00420, and regorafenib), selective FGFR inhibitors (erdafitinib, HMPL-453, futibatinib, KIN-3248, infigratinib, ASP-5878, LY2874455, CPL304110, pemigatinib, and E7090), monoclonal antibodies (Bemarituzumab and BAY1179470), antibody-drug conjugates (LY3076226), FGF ligand traps (GSK3052230), and combination treatments (FGFR inhibitors with PI3K inhibitors, mTOR inhibitors, chemotherapy, or immunotherapy).

Table 3.

Representative clinical trials of FGFR inhibitors for solid tumors

Inhibitors Inhibitor types ClinicalTrials.gov
ID
Phase Conditions or diseases Status
Lucitanib Multi-kinase FGFR inhibitor (FGFR, VEGFR, PDGFR) NCT01283945 1/2 Solid Tumors Completed
3D185 Multi-kinase FGFR inhibitor (FGFR, CSF1R) NCT04221204 1 Solid tumors Recruiting
Sulfatinib Multi-kinase FGFR inhibitor (FGFR, VEGFR, CSF1R) NCT02133157 1 Tumors Completed
MAX-40279–01 Multi-kinase FGFR inhibitor (FGFR, Flt3) NCT05369286 1 Solid tumors Recruiting
TT-00420 Multi-kinase FGFR inhibitor (FGFR, VEGFR, Aurora Kinases, JAK) NCT04742959 1/2 Advanced solid tumors Recruiting
Regorafenib Multi-kinase FGFR inhibitor (RET, VEGFR, c-KIT, PDGFR, FGFR, TIE2, DDR2, TrkA, Eph2A, RAF-1, BRAF, SAPK2, PTK5, Abl) NCT04116541 2 Malignant solid tumors Recruiting
Erdafitinib Selective FGFR inhibitor NCT04083976 2 Advanced solid tumors Recruiting
HMPL-453 Selective FGFR inhibitor NCT05173142 1/2 Solid tumors Recruiting
Pemigatinib Selective FGFR inhibitor NCT04591431 2 Progressive diseases of breast cancer, metastatic gastrointestinal tumors, non-small cell lung cancer or others Recruiting
Futibatinib Selective FGFR inhibitor NCT04189445 2 Advanced or metastatic solid tumors Recruiting
KIN-3248 Selective FGFR inhibitor NCT05242822 1 Solid tumors Recruiting
Infigratinib Selective FGFR inhibitor NCT04233567 2 Advanced or metastatic solid tumors Recruiting
ASP-5878 Selective FGFR inhibitor NCT02038673 1 Solid tumors Completed
LY2874455 Selective FGFR inhibitor NCT01212107 1 Advanced cancer Completed
CPL304110 Selective FGFR inhibitor NCT04149691 1 Advanced solid malignancies Recruiting
E7090 Selective FGFR inhibitor NCT04962867 2 Advanced or recurrent solid tumors Recruiting
Bemarituzumab Monoclonal antibody NCT05325866 1 Solid tumors Not yet recruiting
BAY1179470 Monoclonal antibody NCT01881217 1 Advanced, refractory solid tumors Completed
LY3076226 Antibody-drug conjugate NCT02529553 1 Advanced or metastatic cancer Completed
GSK3052230 FGF ligand trap NCT01868022 1 Neoplasms Completed
EVER4010001 + Pembrolizuman EVER4010001 (FGFR4 inhibitor), Pembrolizuman (PD-1 inhibitor) NCT04699643 1/2 Advanced solid tumors Recruiting
Infigratinib + BYL719 Infigratinib (selective FGFR inhibitor), BYL719 (PIK3K inhibitor) NCT01928459 1 Advanced or metastatic solid tumors Completed
Nintedanib + Pembrolizumab Nintedanib (FGFR, VEGFR, PDGFR inhibitor), Pembrolizumab (PD-1 inhibitor) NCT02856425 1 Advanced solid tumors Recruiting
Surufatinib + Gemcitabine Surufatinib (FGFR, VEGFR, CSF1R inhibitor), Gemcitabine (Chemotherapy) NCT05093322 1/2 Recurrent or refractory solid tumors or lymphoma Recruiting
Nintedanib + Everolimus Nintedanib (FGFR, VEGFR, PDGFR inhibitor), Everolimus (mTOR inhibitor) NCT01349296 1 Solid tumors Completed

Future perspective

Due to the prevalence of FGF/FGFR genomic alterations and overexpression in HNSCC, FGF/FGFR has been a potential target for the treatment of HNSCC. However, the prognostic value of FGF/FGFR aberrations, especially gene amplification and overexpression, are still unclear. The prognosis of HNSCC patients with FGF/FGFR alterations should be examined in a larger sample size, and patients might need to be stratified based on HPV status, patient ethnicity, genomic alterations, and HNSCC anatomic positions. In addition, even though FGF/FGFR overexpression has been detected in the majority of the HNSCC patients, the sensitivity to FGFR inhibitor varies dramatically in the HNSCC with wild-type FGF/FGFR overexpression. Therefore, the underlying molecular mechanism(s) of this variation needs further study. This mechanistic information will provide a better understanding of the molecular features of HNSCC expressing FGF/FGFR and can also contribute to the future development of better FGFR targeted therapies. Other than developing more potent FGFR inhibitors with less side effects, combination treatment is another promising strategy for the future development of FGFR-oriented therapies. In particular, combining FGFR inhibitors with chemo-, radio-, immuno-, and/or other targeted therapies has already demonstrated considerable potential in preclinical studies, which can be further explored and tested in clinical trials.

Summary

FGF/FGFR genomic alterations, including gene amplification, gain-of-function mutation, and chromosomal rearrangement, are one of the most prevalent RTK aberrations in HNSCC. FGFR1 gene amplification, FGF gene amplifications, and FGFR3 mutations are among the most frequent aberrations in HNSCC patients. In addition, FGF/FGFR expression has also been observed in the majority of HNSCC. However, the prognostic value of FGF/FGFR aberrations remains obscure, especially for gene amplification and overexpression. Thus, proper biomarkers which can distinguish HNSCC patients that may benefit from FGFR inhibitors are still urgently needed. Nevertheless, FGF/FGFR demonstrates substantial potential as a target for the treatment of HNSCC. Recent studies in terms of the development of FGFR-oriented therapies for HNSCC have been focusing on combination strategies. Particularly, FGFR inhibitors combined with chemo-, radio-, immuno-, and/or other targeted therapies have already shown considerable efficacy and potency for the treatment of HNSCC in preclinical studies. Therefore, a number of FGFR inhibitors are currently in clinical trials, including multi-target FGFR inhibitors, selective FGFR inhibitors, FGFR targeted alpha therapeutics, monoclonal antibodies, antibody-drug conjugates, FGF ligand traps, and several combination treatments involving FGFR inhibitors.

Acknowledgement:

Support of NIH/NIDCR R01 DE027942 and NIH/NIDCR P50DE030707 to K.S.A. and China Scholarship Council–Yale World Scholars Fellowship to Z.W. is gratefully acknowledged.

Footnotes

Conflicts of Interest and Source of Funding: None

References

  • 1.Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209–49. [DOI] [PubMed] [Google Scholar]
  • 2.Gougis P, Moreau Bachelard C, Kamal M, et al. Clinical Development of Molecular Targeted Therapy in Head and Neck Squamous Cell Carcinoma. JNCI Cancer Spectr 2019;3:pkz055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Son E, Panwar A, Mosher CH, Lydiatt D. Cancers of the Major Salivary Gland. J Oncol Pract 2018;14:99–108. [DOI] [PubMed] [Google Scholar]
  • 4.Leemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer 2018;18:269–82. [DOI] [PubMed] [Google Scholar]
  • 5.Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010;141:1117–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev 2005;16:139–49. [DOI] [PubMed] [Google Scholar]
  • 7.Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip Rev Dev Biol 2015;4:215–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Grünewald S, Politz O, Bender S, et al. Rogaratinib: A potent and selective pan-FGFR inhibitor with broad antitumor activity in FGFR-overexpressing preclinical cancer models. Int J Cancer 2019;145:1346–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bao Y, Gabrielpillai J, Dietrich J, et al. Fibroblast growth factor (FGF), FGF receptor (FGFR), and cyclin D1 (CCND1) DNA methylation in head and neck squamous cell carcinomas is associated with transcriptional activity, gene amplification, human papillomavirus (HPV) status, and sensitivity to tyrosine kinase inhibitors. Clin Epigenetics 2021;13:228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Porta R, Borea R, Coelho A, et al. FGFR a promising druggable target in cancer: Molecular biology and new drugs. Crit Rev Oncol Hematol 2017;113:256–67. [DOI] [PubMed] [Google Scholar]
  • 11.Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 2010;10:116–29. [DOI] [PubMed] [Google Scholar]
  • 12.Tillman BN, Yanik M, Birkeland AC, et al. Fibroblast growth factor family aberrations as a putative driver of head and neck squamous cell carcinoma in an epidemiologically low-risk patient as defined by targeted sequencing. Head Neck 2016;38 Suppl 1:E1646–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brands RC, Knierim LM, De Donno F, et al. Targeting VEGFR and FGFR in head and neck squamous cell carcinoma in vitro. Oncol Rep 2017;38:1877–85. [DOI] [PubMed] [Google Scholar]
  • 14.Shareef KN, Majeed AH. Immunohistochemical Expression of Basic Fibroblast Growth Factor - 2 and Heparanase in Oral Squamous Cell Carcinoma. Journal of Baghdad College of Dentistry 2013;2013 Vol.25 Issue 1, pp.94–98:1–5. [Google Scholar]
  • 15.Dellacono FR, Spiro J, Eisma R, Kreutzer D. Expression of basic fibroblast growth factor and its receptors by head and neck squamous carcinoma tumor and vascular endothelial cells. Am J Surg 1997;174:540–4. [DOI] [PubMed] [Google Scholar]
  • 16.Nayak S, Goel MM, Makker A, et al. Fibroblast Growth Factor (FGF-2) and Its Receptors FGFR-2 and FGFR-3 May Be Putative Biomarkers of Malignant Transformation of Potentially Malignant Oral Lesions into Oral Squamous Cell Carcinoma. PLoS One 2015;10:e0138801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mariz B, Soares CD, de Carvalho MGF, Jorge-Júnior J. FGF-2 and FGFR-1 might be independent prognostic factors in oral tongue squamous cell carcinoma. Histopathology 2019;74:311–20. [DOI] [PubMed] [Google Scholar]
  • 18.Butkiewicz D, Gdowicz-Kłosok A, Krześniak M, Rutkowski T, Łasut-Szyszka B, Składowski K. Germline Variants in Angiogenesis-Related Genes Contribute to Clinical Outcome in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wakulich C, Jackson-Boeters L, Daley TD, Wysocki GP. Immunohistochemical localization of growth factors fibroblast growth factor-1 and fibroblast growth factor-2 and receptors fibroblast growth factor receptor-2 and fibroblast growth factor receptor-3 in normal oral epithelium, epithelial dysplasias, and squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2002;93:573–9. [DOI] [PubMed] [Google Scholar]
  • 20.Marshall ME, Hinz TK, Kono SA, et al. Fibroblast growth factor receptors are components of autocrine signaling networks in head and neck squamous cell carcinoma cells. Clin Cancer Res 2011;17:5016–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gao L, Lang L, Zhao X, Shay C, Shull AY, Teng Y. FGF19 amplification reveals an oncogenic dependency upon autocrine FGF19/FGFR4 signaling in head and neck squamous cell carcinoma. Oncogene 2019;38:2394–404. [DOI] [PubMed] [Google Scholar]
  • 22.Dubot C, Bernard V, Sablin MP, et al. Comprehensive genomic profiling of head and neck squamous cell carcinoma reveals FGFR1 amplifications and tumour genomic alterations burden as prognostic biomarkers of survival. Eur J Cancer 2018;91:47–55. [DOI] [PubMed] [Google Scholar]
  • 23.von Mässenhausen A, Franzen A, Heasley L, Perner S. FGFR1 as a novel prognostic and predictive biomarker in squamous cell cancers of the lung and the head and neck area. Ann Transl Med 2013;1:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015;517:576–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Göke F, Franzen A, Hinz TK, et al. FGFR1 Expression Levels Predict BGJ398 Sensitivity of FGFR1-Dependent Head and Neck Squamous Cell Cancers. Clin Cancer Res 2015;21:4356–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kim EK, Cho YA, Koh YW, Shin HA, Cho BC, Yoon SO. Prognostic implications of Fibroblast growth factor receptor 1 (FGFR1) gene amplification and protein overexpression in hypopharyngeal and laryngeal squamous cell carcinoma. BMC Cancer 2020;20:348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Göke F, Bode M, Franzen A, et al. Fibroblast growth factor receptor 1 amplification is a common event in squamous cell carcinoma of the head and neck. Mod Pathol 2013;26:1298–306. [DOI] [PubMed] [Google Scholar]
  • 28.Basura GJ, Smith JD, Ellsperman S, Bhangale A, Brenner JC. Targeted molecular characterization of external auditory canal squamous cell carcinomas. Laryngoscope Investig Otolaryngol 2021;6:1151–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Freier K, Schwaenen C, Sticht C, et al. Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral Oncol 2007;43:60–6. [DOI] [PubMed] [Google Scholar]
  • 30.Park S, Lee M, Cho KJ, et al. Association Between Fibroblast Growth Factor Receptor 1 Gene Amplification and Human Papillomavirus Prevalence in Tonsillar Squamous Cell Carcinoma With Clinicopathologic Analysis. J Histochem Cytochem 2018;66:511–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Koole K, Brunen D, van Kempen PM, et al. FGFR1 Is a Potential Prognostic Biomarker and Therapeutic Target in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2016;22:3884–93. [DOI] [PubMed] [Google Scholar]
  • 32.Fisher MM, SenthilKumar G, Hu R, et al. Fibroblast Growth Factor Receptors as Targets for Radiosensitization in Head and Neck Squamous Cell Carcinomas. Int J Radiat Oncol Biol Phys 2020;107:793–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Starska K, Forma E, Lewy-Trenda I, Stasikowska-Kanicka O, Skóra M, Bryś M. Fibroblast growth factor receptor 1 and 3 expression is associated with regulatory PI3K/AKT kinase activity, as well as invasion and prognosis, in human laryngeal cancer. Cell Oncol (Dordr) 2018;41:253–68. [DOI] [PubMed] [Google Scholar]
  • 34.Jiao J, Zhao X, Liang Y, Tang D, Pan C. FGF1-FGFR1 axis promotes tongue squamous cell carcinoma (TSCC) metastasis through epithelial-mesenchymal transition (EMT). Biochem Biophys Res Commun 2015;466:327–32. [DOI] [PubMed] [Google Scholar]
  • 35.Koole K, Clausen MJ, van Es RJ, et al. FGFR Family Members Protein Expression as Prognostic Markers in Oral Cavity and Oropharyngeal Squamous Cell Carcinoma. Mol Diagn Ther 2016;20:363–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chen SJ, Liu H, Liao CT, et al. Ultra-deep targeted sequencing of advanced oral squamous cell carcinoma identifies a mutation-based prognostic gene signature. Oncotarget 2015;6:18066–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Seiwert TY, Zuo Z, Keck MK, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res 2015;21:632–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gillison ML, Akagi K, Xiao W, et al. Human papillomavirus and the landscape of secondary genetic alterations in oral cancers. Genome Res 2019;29:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bersani C, Sivars L, Haeggblom L, et al. Targeted sequencing of tonsillar and base of tongue cancer and human papillomavirus positive unknown primary of the head and neck reveals prognostic effects of mutated FGFR3. Oncotarget 2017;8:35339–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bersani C, Haeggblom L, Ursu RG, et al. Overexpression of FGFR3 in HPV-positive Tonsillar and Base of Tongue Cancer Is Correlated to Outcome. Anticancer Res 2018;38:4683–90. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang Y, Hiraishi Y, Wang H, et al. Constitutive activating mutation of the FGFR3b in oral squamous cell carcinomas. Int J Cancer 2005;117:166–8. [DOI] [PubMed] [Google Scholar]
  • 42.Yuan L, Liu ZH, Lin ZR, Xu LH, Zhong Q, Zeng MS. Recurrent FGFR3-TACC3 fusion gene in nasopharyngeal carcinoma. Cancer Biol Ther 2014;15:1613–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nelson KN, Meyer AN, Wang CG, Donoghue DJ. Oncogenic driver FGFR3-TACC3 is dependent on membrane trafficking and ERK signaling. Oncotarget 2018;9:34306–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kikuchi A, Suzuki T, Nakazawa T, et al. ASP5878, a selective FGFR inhibitor, to treat FGFR3-dependent urothelial cancer with or without chemoresistance. Cancer Sci 2017;108:236–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Singh D, Chan JM, Zoppoli P, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 2012;337:1231–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.von Mässenhausen A, Deng M, Billig H, et al. Evaluation of FGFR3 as a Therapeutic Target in Head and Neck Squamous Cell Carcinoma. Target Oncol 2016;11:631–42. [DOI] [PubMed] [Google Scholar]
  • 47.Koole K, van Kempen PM, Swartz JE, et al. Fibroblast growth factor receptor 3 protein is overexpressed in oral and oropharyngeal squamous cell carcinoma. Cancer Med 2016;5:275–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wimmer E, Ihrler S, Gires O, Streit S, Issing W, Bergmann C. Fibroblast growth factor receptor 4 single nucleotide polymorphism Gly388Arg in head and neck carcinomas. World J Clin Oncol 2019;10:136–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shi S, Li X, You B, Shan Y, Cao X, You Y. High Expression of FGFR4 Enhances Tumor Growth and Metastasis in Nasopharyngeal Carcinoma. J Cancer 2015;6:1245–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Collin MP, Lobell M, Hübsch W, et al. Discovery of Rogaratinib (BAY 1163877): a pan-FGFR Inhibitor. ChemMedChem 2018;13:437–45. [DOI] [PubMed] [Google Scholar]
  • 51.Schuler M, Cho BC, Sayehli CM, et al. Rogaratinib in patients with advanced cancers selected by FGFR mRNA expression: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol 2019;20:1454–66. [DOI] [PubMed] [Google Scholar]
  • 52.Joerger M, Takahashi S, Sayehli CM, et al. 1056PD - Phase I experience with rogaratinib in patients with head and neck cancer selected based on FGFR mRNA overexpression. Annals of Oncology 2018;29:viii377. [Google Scholar]
  • 53.Baschnagel AM, Fisher M, Miller M, et al. Molecular Profiling of Head and Neck Cancer Patient Derived Xenografts Identifies FGFR As a Target for Radiosensitization. International Journal of Radiation Oncology*Biology*Physics 2018;102:S188. [Google Scholar]
  • 54.Aytatli A, Barlak N, Sanli F, et al. AZD4547 targets the FGFR/Akt/SOX2 axis to overcome paclitaxel resistance in head and neck cancer. Cell Oncol (Dordr) 2022;45:41–56. [DOI] [PubMed] [Google Scholar]
  • 55.Schaefer T, Lengerke C. SOX2 protein biochemistry in stemness, reprogramming, and cancer: the PI3K/AKT/SOX2 axis and beyond. Oncogene 2020;39:278–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.McDermott SC, Rodriguez-Ramirez C, McDermott SP, Wicha MS, Nör JE. FGFR signaling regulates resistance of head and neck cancer stem cells to cisplatin. Oncotarget 2018;9:25148–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Holzhauser S, Wild N, Zupancic M, et al. Targeted Therapy With PI3K and FGFR Inhibitors on Human Papillomavirus Positive and Negative Tonsillar and Base of Tongue Cancer Lines With and Without Corresponding Mutations. Front Oncol 2021;11:640490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ishigami T, Uzawa K, Higo M, et al. Genes and molecular pathways related to radioresistance of oral squamous cell carcinoma cells. Int J Cancer 2007;120:2262–70. [DOI] [PubMed] [Google Scholar]
  • 59.Uzawa K, Ishigami T, Fushimi K, et al. Targeting fibroblast growth factor receptor 3 enhances radiosensitivity in human squamous cancer cells. Oncogene 2011;30:4447–52. [DOI] [PubMed] [Google Scholar]
  • 60.Niederst MJ, Engelman JA. Bypass mechanisms of resistance to receptor tyrosine kinase inhibition in lung cancer. Sci Signal 2013;6:re6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Singleton KR, Kim J, Hinz TK, et al. A receptor tyrosine kinase network composed of fibroblast growth factor receptors, epidermal growth factor receptor, v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, and hepatocyte growth factor receptor drives growth and survival of head and neck squamous carcinoma cell lines. Mol Pharmacol 2013;83:882–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gyanchandani R, Ortega Alves MV, Myers JN, Kim S. A proangiogenic signature is revealed in FGF-mediated bevacizumab-resistant head and neck squamous cell carcinoma. Mol Cancer Res 2013;11:1585–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ban MJ, Byeon HK, Yang YJ, et al. Fibroblast growth factor receptor 3-mediated reactivation of ERK signaling promotes head and neck squamous cancer cell insensitivity to MEK inhibition. Cancer Sci 2018;109:3816–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Michmerhuizen NL, Ludwig ML, Birkeland AC, et al. Small molecule profiling to define synergistic EGFR inhibitor combinations in head and neck squamous cell carcinoma. Head Neck 2022;44:1192–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kumar D, New J, Vishwakarma V, et al. Cancer-Associated Fibroblasts Drive Glycolysis in a Targetable Signaling Loop Implicated in Head and Neck Squamous Cell Carcinoma Progression. Cancer Res 2018;78:3769–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Singleton KR, Hinz TK, Kleczko EK, et al. Kinome RNAi Screens Reveal Synergistic Targeting of MTOR and FGFR1 Pathways for Treatment of Lung Cancer and HNSCC. Cancer Res 2015;75:4398–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ngan HL, Law CH, Choi YCY, Chan JY, Lui VWY. Precision drugging of the MAPK pathway in head and neck cancer. NPJ Genom Med 2022;7:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ferris RL, Blumenschein G Jr., Fayette J, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 2016;375:1856–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Akhand SS, Liu Z, Purdy SC, et al. Pharmacologic Inhibition of FGFR Modulates the Metastatic Immune Microenvironment and Promotes Response to Immune Checkpoint Blockade. Cancer Immunol Res 2020;8:1542–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Palakurthi S, Kuraguchi M, Zacharek SJ, et al. The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity. Cancer Immunol Res 2019;7:1457–71. [DOI] [PubMed] [Google Scholar]
  • 71.Kacew A, Sweis RF. FGFR3 Alterations in the Era of Immunotherapy for Urothelial Bladder Cancer. Front Immunol 2020;11:575258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kono M, Komatsuda H, Yamaki H, et al. Immunomodulation via FGFR inhibition augments FGFR1 targeting T-cell based antitumor immunotherapy for head and neck squamous cell carcinoma. Oncoimmunology 2022;11:2021619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther 2020;5:166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hase T, Kawashiri S, Tanaka A, et al. Correlation of basic fibroblast growth factor expression with the invasion and the prognosis of oral squamous cell carcinoma. J Oral Pathol Med 2006;35:136–9. [DOI] [PubMed] [Google Scholar]
  • 75.Sweeny L, Liu Z, Lancaster W, Hart J, Hartman YE, Rosenthal EL. Inhibition of fibroblasts reduced head and neck cancer growth by targeting fibroblast growth factor receptor. Laryngoscope 2012;122:1539–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dumbrava EI, Alfattal R, Miller VA, Tsimberidou AM. Complete Response to a Fibroblast Growth Factor Receptor Inhibitor in a Patient With Head and Neck Squamous Cell Carcinoma Harboring FGF Amplifications. JCO Precis Oncol 2018;2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Taghizadeh H, Mader RM, Müllauer L, Fuereder T, Kautzky-Willer A, Prager GW. Outcome of Targeted Therapy Recommendations for Metastatic and Recurrent Head and Neck Cancers. Cancers (Basel) 2020;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Subbiah V, Iannotti NO, Gutierrez M, et al. FIGHT-101, a first-in-human study of potent and selective FGFR 1–3 inhibitor pemigatinib in pan-cancer patients with FGF/FGFR alterations and advanced malignancies. Ann Oncol 2022;33:522–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cai Q, SU N, Fang Y, et al. Anlotinib for patients with recurrent or metastatic nasopharyngeal carcinoma: A phase II study. Journal of Clinical Oncology 2022;40:e18020-e. [Google Scholar]
  • 80.Locati LD, Galbiati D, Calareso G, et al. Patients with adenoid cystic carcinomas of the salivary glands treated with lenvatinib: Activity and quality of life. Cancer 2020;126:1888–94. [DOI] [PubMed] [Google Scholar]

RESOURCES