103
Views
18
CrossRef citations to date
0
Altmetric
Review

Novel genetic alterations and their impact on target therapy response in head and neck squamous cell carcinoma

, , , &
Pages 1321-1336 | Published online: 08 Feb 2019

Abstract

Head and neck squamous cell carcinoma (HNSCC) is highly variable by tumor site, histologic type, molecular characteristics, and clinical outcome. During recent years, emerging targeted therapies have been focused on driver genes. HNSCC involves several genetic alterations, such as co-occurrence, multiple feedback loops, and cross-talk communications. These different kinds of genetic alterations interact with each other and mediate targeted therapy response. In the current review, it is emphasized that future treatment strategy in HNSCC will not solely be based on “synthetic lethality” approaches directed against overactivated genes. More importantly, biologic, genetic, and epigenetic alterations of HNSCC will be taken into consideration to guide the therapy. The emerging genetic alterations in HNSCC and its effect on targeted therapy response are discussed in detail. Hopefully, novel combination regimens for the treatment of HNSCC can be developed.

Introduction

Head and neck squamous cell carcinoma (HNSCC) causes more than 300,000 deaths worldwide each year and its incidence is still growing.Citation1 HNSCC is a fatal heterogeneous disease, with highly variable tumor sites and biologic behaviors.Citation2 The Cancer Genome Atlas (TCGA) has profiled 279 of HNSCC, from the oral cavity (n=172/279, 62%), oropharynx (n=33/279, 12%), and laryngeal sites (n=72/279, 26%).Citation3,Citation4 They generate comprehensive lists of genomic and epigenomic alterations present in diverse tumor samples. The MSK-IMPACT (Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets) assay is a targeted next-generation sequencing (NGS) assay approved through Clinical Laboratory Improvement Amendments.Citation5 There were 151 patients with advanced, treatment-resistant head and neck tumors, including HNSCC, adenoid cystic carcinoma, and other salivary and cutaneous cancers, whose tumors were sequenced.Citation6 Thus, it provided a comprehensive understanding of the molecular pathogenesis of HNSCC progression and treatment response.Citation7

Comprehensive understanding of the biologic, genetic, and epigenetic alterations of HNSCC is necessary to guide the therapy.Citation8 Drug responses are determined largely by pharmacogenetics, cancer microenvironment, and genetic aberrations.Citation9 Identifying molecular aberrations that link to drug sensitivity helps to avoid unnecessary and unhelpful treatment.Citation10 Although associations between genomic alterations and drug response were previously observed in HNSCC, no therapeutically targetable genomic subtypes have been identified. Therefore, surgery, radiation, and chemotherapy still remain to be the standard therapy for the disease.

We reviewed the emerging genetic alterations that represent a novel therapeutic target in HNSCC. These include frequent mutations of several genes (TP53, PIK3CA, CDKN2A, NOTCH1, and MET) as well as copy number gain in EGFR, CCND1, and PIK3CA. Those genomic alterations have been analyzed in relevance of sensitivity to chemotherapy, targeted therapy, and ionizing radiation that could potentially guide the therapy (). It is essential to determine whether proteins or pathways are involved in the targeted therapy of HNSCCs, aside from gene mutation.Citation11 These genetic alterations may serve well as novel therapeutic candidates and predictive biomarkers for prognosis in HNSCC.

Table 1 Genomic expression and alterations associated with drug resistance in HNSCC studies

Based on the recently identified mutations in HNSCCs, the major pathologic pathways implicated in the tumorigenesis of HNSCC include dysregulation of four processes: 1) cellular survival and proliferation (eg, TP53, EGFR, MET, and PIK3CA); 2) cell-cycle control (eg, CDKN2A and CCND1); 3) cellular differentiation (eg, NOTCH1); and 4) adhesion and invasion signaling (eg, FAT1).Citation7 TP53, EGFR, PIK3CA, CDKN2A, CCND1, and MET participate in several common signaling pathways (). Alterations of these genes are most frequently seen in alcohol and tobacco-related HNSCC.Citation12 Genetic aberration interactions involve pathway reactivation (downstream re-engagement of original effectors), pathway bypass (recruitment of a parallel pathway converging on the same downstream output), and pathway indifference (development of a cellular state independent of the initial therapeutic target). Gene–drug interactions are analyzed in cBioPortal. Data were from TCGA Head and Neck and Recurrent and Metastatic Head & Neck Cancer ().Citation3

Figure 1 Schematic diagram of signaling pathways that emerging biomarkers TP53, EGFR, PIK3CA, CDKN2A, CCND1, and MET participated in.

Notes: EGFR is a member of the HER family of cell-surface receptor tyrosine kinases. Ligand binding triggers EGFR and activates downstream effectors, thus promoting cell proliferation. Hypoxia induces the expression of EGFR and enhances the phosphorylation of EGFR thereby regulating intrinsic DNA-repair mechanisms, meanwhile EGFR can stabilize HIF-1a. MET is a receptor tyrosine kinase associated with enhanced migration, invasion, and angiogenesis when overexpressed in cancer. PIK3CA encodes p110α, a catalytic subunit of PI3K, which is a heterodimeric kinase with enzymatic activity on lipid and protein substrates. The TP53 has a vital role in the regulation of genes responsible for cell-cycle arrest, senescence, and apoptosis. MDM2 promotes the rapid degradation of the TP53 protein. TP53 missense mutations cause single-amino-acid substitutions that lead to loss of DNA-binding capability. Cyclin D1 is a cell-cycle protein that regulates the key G1-to-S phase transition through formation of complexes with CDKs, such as CDK4 and CDK6. Upon ligand binding, NOTCH receptors undergo a conformational change enabling cleavage and nuclear translocation of the intracellular domain to release the transcriptional repression of downstream target genes.

Abbreviations: CDKs, cyclin-dependent kinases; HIF-1a, hypoxia-inducible factor 1a; PI3K, phosphoinositide 3 kinase.

Figure 1 Schematic diagram of signaling pathways that emerging biomarkers TP53, EGFR, PIK3CA, CDKN2A, CCND1, and MET participated in.Notes: EGFR is a member of the HER family of cell-surface receptor tyrosine kinases. Ligand binding triggers EGFR and activates downstream effectors, thus promoting cell proliferation. Hypoxia induces the expression of EGFR and enhances the phosphorylation of EGFR thereby regulating intrinsic DNA-repair mechanisms, meanwhile EGFR can stabilize HIF-1a. MET is a receptor tyrosine kinase associated with enhanced migration, invasion, and angiogenesis when overexpressed in cancer. PIK3CA encodes p110α, a catalytic subunit of PI3K, which is a heterodimeric kinase with enzymatic activity on lipid and protein substrates. The TP53 has a vital role in the regulation of genes responsible for cell-cycle arrest, senescence, and apoptosis. MDM2 promotes the rapid degradation of the TP53 protein. TP53 missense mutations cause single-amino-acid substitutions that lead to loss of DNA-binding capability. Cyclin D1 is a cell-cycle protein that regulates the key G1-to-S phase transition through formation of complexes with CDKs, such as CDK4 and CDK6. Upon ligand binding, NOTCH receptors undergo a conformational change enabling cleavage and nuclear translocation of the intracellular domain to release the transcriptional repression of downstream target genes.Abbreviations: CDKs, cyclin-dependent kinases; HIF-1a, hypoxia-inducible factor 1a; PI3K, phosphoinositide 3 kinase.

Figure 2 General interactions of genomic mutations (TP53, CDKN2A, CCND1, NOTCH1, PIK3CA, EGFR, and MET) and current target drugs.

Notes: Primary data are from tumor samples with sequencing and CNA data (132 patients/samples) and could be accessed in cBioPortal.Citation6 Detailed description of data mining is (filter neighbors by max 28.8% alteration) found in http://www.cbioportal.org/results/network?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016&case_set_id=hnc_mskcc_2016_cnaseq&data_priority=0&gene_list=TP53%2520CDKN2A%2520CCND1%2520NOTCH1%2520PIK3CA%2520EGFR%2520MET&geneset_list=%20&genetic_profile_ids_PROFILE_COPY_NUMBER_ALTERATION=hnc_mskcc_2016_gistic&genetic_profile_ids_PROFILE_MUTATION_EXTENDED=hnc_mskcc_2016_mutations&tab_index=tab_visualize.Citation134,Citation135

Abbreviations: US FDA, US Food and Drug Administration.

Figure 2 General interactions of genomic mutations (TP53, CDKN2A, CCND1, NOTCH1, PIK3CA, EGFR, and MET) and current target drugs.Notes: Primary data are from tumor samples with sequencing and CNA data (132 patients/samples) and could be accessed in cBioPortal.Citation6 Detailed description of data mining is (filter neighbors by max 28.8% alteration) found in http://www.cbioportal.org/results/network?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016&case_set_id=hnc_mskcc_2016_cnaseq&data_priority=0&gene_list=TP53%2520CDKN2A%2520CCND1%2520NOTCH1%2520PIK3CA%2520EGFR%2520MET&geneset_list=%20&genetic_profile_ids_PROFILE_COPY_NUMBER_ALTERATION=hnc_mskcc_2016_gistic&genetic_profile_ids_PROFILE_MUTATION_EXTENDED=hnc_mskcc_2016_mutations&tab_index=tab_visualize.Citation134,Citation135Abbreviations: US FDA, US Food and Drug Administration.

TP53

TP53 mutation

Mutations in the tumor suppressor P53(TP53) gene are present in about 70% of HNSCC.Citation13 Missense mutations in TP53, including those at codons R248, R273, G245, R175, R282, and H179, are the most frequent hotspot mutations in HNSCC.Citation7 Two thousand four hundred ninety-eight samples in seven studies (Head and Neck Squamous Cell Carcinoma [Broad, Science 2011]; Head and Neck Squamous Cell Carcinoma [Johns Hopkins, Science 2011]; Head and Neck Squamous Cell Carcinoma [TCGA, Nature 2015]; Head and Neck Squamous Cell Carcinoma [TCGA, PanCancer Atlas]; Head and Neck Squamous Cell Carcinoma [TCGA, Provisional]; Oral Squamous Cell Carcinoma [MD Anderson, Cancer Discov 2013])Citation4,Citation14,Citation15 showed 62.7% of somatic mutation and 45.2% of missense mutations (http://www.cbioportal.org/). TP53 mutation was markedly higher in metastatic HNSCC.Citation6 TP53 mutation involves varieties of proteins that contribute to tumorigenesis and tumor progression.Citation4,Citation16 It occurs early in carcinoma progression and more frequently in those with greater histologic severity.Citation17,Citation18

Data TCGA Head and Neck and Recurrent and Metastatic Head & Neck Cancer (MSKCC, JAMA Oncol 2016) analyzed by cBioPortal (detailed description of data mining could be found in the figure legends) demonstrated that only TP53 mutation is a predictor for overall survival (OS) rate and disease-free survival rate (). Moreover, tumors of the larynx and hypopharynx have the highest TP53 mutation rate (83.5%). Tumors of the tongue and oral cavity have a TP53 mutation rate of 75.6%, and those of the oropharynx (including the tonsils), and base of the tongue have the lowest TP53 mutation rate (28.6%).Citation13 Previous studies have shown that TP53 mutation correlated with resistance to chemotherapy drugs such as cisplatin, doxorubicin, and paclitaxel.Citation19Citation22 It has been recently further demonstrated that cisplatin resistance was associated with aneuploidy of chromosome 17, increased TP53 copy numbers, and overexpression of mutant variant R248L.Citation21

TP53 mutation has also been indicated for assessment of postoperative radiotherapy.Citation23 If there are no histologically detectable tumors and no TP53 mutations in the surgical margin, patients can be spared postoperative radiotherapy. It is supported by the studies that show the absence of TP53-mutated DNA in surgical margins was significantly associated with local recurrence-free survival.Citation24,Citation25

Classification of TP53 mutation

The value of TP53 mutation in diagnosis is different between subtypes. Some are associated with more aggressive HNSCC phenotypes, whereas others are linked with a more indolent pattern of tumor progression.Citation26

Perrone et al categorized TP53 mutation significance based on the transactivation activity as functional, partially functional, or nonfunctional.Citation27 Loss of function of TP53 (transactivation activities) predicts a significantly low rate of pathologic complete remission and suboptimal response to cisplatin-based neoadjuvant chemotherapy in patients with oral squamous cell carcinoma (OSCC).Citation28 Accumulating evidence suggests that gain of function (GOF) of TP53 mutants as well mediates drug resistance. The underlying mechanisms include apoptotic proteins inhibition and gene regulations.Citation29,Citation30

Another large study classified TP53 mutation as disruptive and nondisruptive, based on alteration of DNA binding.Citation31 The disruptive is defined as any mutation in L2 or L3 loop of the DNA-binding domain or stop codon, resulting in a polarity change within the protein. Disruptive TP53 mutation strongly predicted locoregional recurrence driven by tumor cell radioresistance. The radioresistance is measured by SA-β-gal staining, p21 expression, and release of ROS.Citation31

Evolutionary action (EATP53), a novel computational approach, has been applied to stratify tumor patients with TP53 mutation as high- or low risk. This system was validated both in vivo and in vitroCitation32 (available at http://mammoth.bcm.tmc.edu/EATP53). High-risk mutations promote invasion, metastasis, as well as cisplatin resistance in head and neck cancer cell lines, as they acquired oncogenic GOF properties, while low-risk mutations retained wild-type (WT) TP53 activity.Citation32,Citation33 Different effect of TP53 mutation on cisplatin response has been observed in vitro and in vivo. Mice with HNSCC harboring WT or low-risk mutations responded well to cisplatin treatment. Quite the opposite, TP53 null type or high-risk TP53 mutations failed to show any growth inhibition with cisplatin therapy.Citation34 Similar results were seen in patients with those characters. The high-risk TP53 mutations were associated with decreased OS.Citation32

Targeting TP53 mutation and other coexisting alterations to induce synthetic lethality

Researchers have explored introduction of exogenous WT TP53 into HNSCC cells, or reactivation of some level of WT function in mutant p53-bearing cells, otherwise promotion of mutant TP53 degradation. Of all the compounds that restore WT activity, the only drug directly targeting mutant TP53 that has reached the clinical stage is PRIMA-1(met)/APR-246.Citation35Citation37 It also induced apoptosis and enhanced the cytotoxicity of standard chemotherapy in HNSCC cells.

In addition to targeting mutant p53 directly, investigators have used strategies targeting mutant TP53-regulated downstream targets and signaling pathways. Mutant TP53 proteins are believed to achieve GOF activity by interacting with other molecular alterations that coexist in HNSCC. Understanding the impact of TP53 mutation on cellular growth and survival signaling pathways can help design more effective therapeutic strategies that target TP53 mutation-bearing HNSCC.

CDKN2A deletions

TP53 and CDKN2A are involved in cell cycle.Citation33 A genomic analysis of human HNSCCs detected that CDKN2A suppresses the oncogenic function of TP53 mutation that promotes malignant progression, and the prognostic value of mutant p53 needs to be considered in the context of CDKN2A.Citation26 The survival of patients with HNSCCs bearing co-occurring high-risk TP53 mutation and CDKN2A homozygous deletions was extremely lower than that of patients with tumors in which high-risk TP53 mutation did not contain CDKN2A homozygous deletions, or that of patients with tumors in which homozygous CDKN2A deletions coexisted with either low-risk TP53 mutation or potential loss-of-function mutations in TP53 (high- and low risk according to the EATP53 classification system).Citation32

PIK3CA activation

Preliminary findings from TCGA Head and Neck Cancer have detected amplifications or putative activating mutations in PIK3CA in 30% of 279 HNSCC tumors, which overlap with WT and mutant TP53 subsets.Citation38 The aforementioned studies indicate that increase in phosphoinositide 3-kinase (PI3K)/Akt activation has been linked to frequent alterations in TP53 via multiple mechanisms.Citation10,Citation38Citation40 Comparing half-maximal inhibitory concentration in a wider panel of HNSCC cell lines suggests that TP53 status contributes to the sensitivity of PI3K/mTOR inhibitor.

In human xenograft, PI3K/mTOR inhibition has been proven to enhance WT TP53 expression, enhance the TP53 DNA-damage response, and further delay tumor regrowth with radiation;Citation38 thus, the combination of PI3K/TP53 events seems to be necessary for PI3K inhibitor sensitivity. The dual PI3K/PLK inhibitor rigosertib has such potent antiproliferative effects on patient tumor xenografts, which carried combination of a PIK3CA-activating event driven by amplification or mutation and a TP53-inactivating event driven by either human papillomavirus (HPV)-negative or nonsense TP53 mutation.Citation41

CDKN2A

CDKN2A mutations

Global genomic analyses identified the tumor suppressor CDKN2A as the second most commonly altered gene in HNSCC.Citation13 CDKN2A (also known as P16, INK4, p16INK4A, and MTS1) is allelic to chromosome 9p21 and encodes a CDK4/CDK6 kinase inhibitor that constrains cells from progressing through the G1 restriction point. It is thought to be involved in early stages of HNSCC development. It is affected in up to 80% of HNSCC – often deleted, hyper-methylated, or, much rarely, mutated.Citation42 CDKN2A mutation is considered as “noncoding mutations”, “inactivation”, or “loss of function”.Citation43 It is associated with worse oversurvival in patients with recurrent and metastatic HNSCC (MSKCC, JAMA Oncol 2016) ().Citation13 Most of the mutations in CDKN2A were found in exon 2 of the gene.Citation42 However, these are likely insufficient to drive tumorigenesis by CDKN2A mutations themselves.Citation44 For example, CDKN2A mutations in benign epithelial lesions have low potential to transform into malignancy.Citation44 Therapeutic targeting of CDKN2A presents the challenge of restoring tumor suppressor activity or inhibiting downstream targets that have been rendered overactive.

CDKN2A mutation reflects p16INK4a function

p16INK4a immunohistochemical expression has been proposed as a surrogate marker for viral oncogene activity in HNSCC.Citation45 The overexpression of p16INK4a is an outstanding surrogate for HPV positivity extensively documented in OSCC.Citation46,Citation47 Numerous studies have reported a considerably improved survival (death risk reduced by 40%–80%) or lower locoregional recurrences (reduced by 60%–70%) and better response to chemo- and radiotherapy in HPV-positive patients with p16INK4a expression compared to HPV-negative patients, when treated with standard multimodality treatments.Citation48Citation53

HPV-positive HNSCC rarely has CDKN2A mutation and thus has the p16INK4a function;Citation26,Citation54 those patients are significantly less likely to develop a locoregional recurrence.Citation43 On the other hand, up to 90% of HPV-negative HNSCCs have loss of p16INK4a function as a result of CDKN2A mutation, promoter methylation, or gene/chromosome 9p21 deletion.Citation45 But the recent study found that CDKN2A mutation still reflects the p16INK4a expression in HPV-negative HNSCC. Loss of CDKN2A function as a result of CDKN2A mutation induced p16INK4a mRNA transcription strongly.Citation45 Hence, p16INK4a overexpression is associated with CDKN2A mutation in a subset of cases. In those cases, the value of p16INK4a for inducing HPV infection was not reliable.

CCND1

CCND1 amplification

CCND1 encodes cyclin D1 on chromosome 11q13, and regulates the key G1-to-S phase transition through formation of complexes with cyclin-dependent kinases (CDKs), such as CDK4 and CDK6.Citation55 Mutations, amplification, and overexpression of this gene, which alters cell-cycle progression, are observed frequently in a variety of tumors and may contribute to tumorigenesis. CCND1 is amplified in as many as 30%–40% of head and neck cancers with cyclin D1 overexpression.Citation56,Citation57 Comparative studies have either shown no prognostic advantage of protein expression over gene amplification or have favored cyclin D1 gene amplification as a better marker of poor prognosis. CCND1 amplification had a statistically significant association with recurrence, distant metastasis, platinum and cisplatin resistance, and EGFR-inhibitor resistance.Citation58Citation61 Allele A may be a risk factor for upper aerodigestive tract cancer, especially in nonalcoholics. Patients of recurrent and metastatic head and neck cancer with CCND1 alterations have poor OS rate (). CCND1 may be a valuable marker for predicting regional response to radiotherapy in metastatic HNSCC and might assist when deciding on appropriate primary therapy.Citation62 Kinetin riboside, an inhibitor of CCND2 transactivation, rapidly suppressed cyclin D1,Citation63 which is a promising CCND1 inhibitor.

CCND1 amplification confers to EGFR inhibitor resistance

Maintenance of cyclin D1 levels is critical to the resistance HNSCC cells display to gefitinib.Citation54 Sensitivity of the six HNSCC cell lines (FaDu, Detroit 562, SCC 9, SCC 15, SCC 25, and CAL 27) to gefitinib was related to cyclin D1 overexpression. These cell lines that overexpressed cyclin D1 continued to proliferate when treated with up to lethal level of gefitinib, but cell numbers remained static or decreased after gefitinib treatment in the non-overexpressed cyclin D1 cell lines.Citation65 Furthermore, the most resistant cell line displayed the highest level of CCND1 amplification and cyclin D1 overexpression. Cyclin D1 upregulation also combines with cortactin overexpression to promote resistance to gefitinib.Citation66 Cortactin overexpression attenuated ligand-induced down-regulation of EGFR, leading to sustained signaling.Citation66

A study in vitro explored the relationship between deregulated cyclin D1 expression and sensitivity to gefitinib to determine whether this frequently occurring oncogenic change affected the cellular response to gefitinib. Three of six cell lines displayed cyclin D1 amplification and/or overexpression, and these cell lines were resistant to gefi-tinib.Citation65 No preclinical studies have yet addressed whether cyclin D1 expression is maintained in CCND1-amplified cancers after EGFR inhibition, and this is an issue warranting investigation.

NOTCH1

NOTCH1 mutations

The NOTCH pathway consists of four receptors (NOTCH1–4) and two families of ligands: the Jagged (JAG1 and JAG2) and the Delta-like (DLL1, DLL3, and DLL4) proteins. NOTCH1 is a transmembrane receptor and controls cell differentiation and embryonic development. Upon ligand binding, functions of NOTCH1 in oral SCC are considered tumor-suppressive. In tongue cancer xenografts, the presence of NOTCH1 caused a dramatic tumor reduction compared to tumors in which this gene was absent.Citation67 Higher expression and activity of NOTCH1 have previously been associated with radio- and chemoresistance.Citation68

Point mutations affecting this gene occurred in 11% of the HNSCC tumors.Citation13 It is considered to represent “loss of function”.Citation13,Citation69,Citation70 Missense mutations in NOTCH1 are considered tumor-suppressive in OSCC. Notably, OSCC patients with NOTCH pathway mutations (AR, ARNT, EP300, CREBBP, JAK2, JAK, NOTCH1, NOTCH2, NOTCH3, and PARP1) were 3.3 times more likely to die with recurrent disease compared to those who did not have these alterations.Citation71 The NOTCH1 mutation status recently served as biomarker for the identification of HNSCC with higher sensitivity to radio- and chemotherapy, as significant association of NOTCH1 mutations with improved localregional control and increased overall survival was observed after chemora-diotherapy.Citation70 Using of gamma-secretase inhibitors supposed to be an attractive strategy for improvement of chemoradiotherapy in malignancies harboring wild-type NOTCH1. For HNSCC it is should be carefully evaluated given evidence of an increased frequency of squamous cell carcinoma in vivo with genetic reductional γ-secretase activity.Citation133

Analysis of recurrent and metastatic head and neck cancer suggests a similar trend toward better 5-year (60 months) OS in patients with alterations in NOTCH1, but after 5 years the OS in patients with alterations was worse; however, this was not statistically significant ().

Activated NOTCH1 contributes to resistance of PI3K inhibitors

Active domain of NOTCH1 conferred resistance to the dual PI3K/mTOR inhibitor BEZ-235 in breast adenocarcinoma-like cell line MCF7 and the ductal carcinoma-like cell lines BT474, HCC70, and BT549.Citation10 They showed resistance to BEZ-235 treatment upon expression of ICN1 (intracellular active domain of NOTCH1).Citation72 Furthermore, in NOTCH1 mutant breast cells that lack the extracellular domain, BEZ-235 sensitivity could also be restored by inhibiting γ-secretase, indicating that naturally cleaved NOTCH1 also confers resistance to PI3K inhibition. A significant correlation was detected between low expression of negative regulator of NOTCH (NUMB) and resistance to PI3K/mTOR inhibition in cell lines derived from various tumor types, including melanoma and hepatocellular carcinoma. These results suggest that uncoupling proliferation from the PI3K pathway via NOTCH1 activation may be a general phenomenon across cancer cell lines.

PIK3CA

PIK3CA mutations

The PI3K pathway (PIK3CA, PTEN, AKT, and STK11) was the most frequently somatically mutated oncogenic pathway in HNSCC tumors.Citation73Citation75

PIK3CA alterations encoding the key catalytic subunit of PI3K pathway were subsequently found to be prevalent. Data from the querying 2,498 samples showed PIK3CA alterations around 29%, including copy number gain (high polysomy or amplification) or mutationsCitation13 ().Citation4,Citation14,Citation15,Citation67 However, PIK3CA alteration has no significant prognoses value for recurrent and metastatic head and neck cancer analyzed in cBioPortal ().

Figure 3 Alteration profiles of the emerging biomarkers from querying 1,627 patients/1,629 samples in 7 studies analyzed in cBioPortal.

Notes: Data from 11,000 cases and all TCGA tumor types from TCGA. Detailed description for data mining could be found in http://www.cbioportal.org/results/cancerTypesSummary?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016%2Chnsc_broad%2Chnsc_jhu%2Chnsc_tcga_pub%2Chnsc_tcga_pan_can_atlas_2018%2Chnsc_tcga%2Chnsc_mdanderson_2013&case_set_id=all&data_priority=0&gene_list=TP53%2520CDKN2A%2520CCND1%2520NOTCH1%2520PIK3CA%2520EGFR%2520MET&geneset_list=%20&tab_index=tab_visualize.Citation134,Citation135

Abbreviation: TCGA, The Cancer Genome Atlas.

Figure 3 Alteration profiles of the emerging biomarkers from querying 1,627 patients/1,629 samples in 7 studies analyzed in cBioPortal.Notes: Data from 11,000 cases and all TCGA tumor types from TCGA. Detailed description for data mining could be found in http://www.cbioportal.org/results/cancerTypesSummary?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016%2Chnsc_broad%2Chnsc_jhu%2Chnsc_tcga_pub%2Chnsc_tcga_pan_can_atlas_2018%2Chnsc_tcga%2Chnsc_mdanderson_2013&case_set_id=all&data_priority=0&gene_list=TP53%2520CDKN2A%2520CCND1%2520NOTCH1%2520PIK3CA%2520EGFR%2520MET&geneset_list=%20&tab_index=tab_visualize.Citation134,Citation135Abbreviation: TCGA, The Cancer Genome Atlas.

Figure 4 Recurrent and metastatic head and neck cancer samples with sequencing and CNA data (132 patients/samples) analyzed in cBioPortal.

Notes: OS/recurrence-free survival, for cases with/without alteration(s) in query gene. Survival probabilities were calculated with the Kaplan–Meier method, according to the original article.Citation6 Detailed description of data mining could be found in http://www.cbioportal.org/results/survival?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016&case_set_id=hnc_mskcc_2016_cnaseq&data_priority=0&gene_list=TP53&geneset_list=%20&genetic_profile_ids_PROFILE_COPY_NUMBER_ALTERATION=hnc_mskcc_2016_gistic&genetic_profile_ids_PROFILE_MUTATION_EXTENDED=hnc_mskcc_2016_mutations&tab_index=tab_visualize.Citation134,Citation135

Abbreviation: OS, overall survival.

Figure 4 Recurrent and metastatic head and neck cancer samples with sequencing and CNA data (132 patients/samples) analyzed in cBioPortal.Notes: OS/recurrence-free survival, for cases with/without alteration(s) in query gene. Survival probabilities were calculated with the Kaplan–Meier method, according to the original article.Citation6 Detailed description of data mining could be found in http://www.cbioportal.org/results/survival?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016&case_set_id=hnc_mskcc_2016_cnaseq&data_priority=0&gene_list=TP53&geneset_list=%20&genetic_profile_ids_PROFILE_COPY_NUMBER_ALTERATION=hnc_mskcc_2016_gistic&genetic_profile_ids_PROFILE_MUTATION_EXTENDED=hnc_mskcc_2016_mutations&tab_index=tab_visualize.Citation134,Citation135Abbreviation: OS, overall survival.
Figure 4 Recurrent and metastatic head and neck cancer samples with sequencing and CNA data (132 patients/samples) analyzed in cBioPortal.Notes: OS/recurrence-free survival, for cases with/without alteration(s) in query gene. Survival probabilities were calculated with the Kaplan–Meier method, according to the original article.Citation6 Detailed description of data mining could be found in http://www.cbioportal.org/results/survival?Action=Submit&RPPA_SCORE_THRESHOLD=2&Z_SCORE_THRESHOLD=2&cancer_study_list=hnc_mskcc_2016&case_set_id=hnc_mskcc_2016_cnaseq&data_priority=0&gene_list=TP53&geneset_list=%20&genetic_profile_ids_PROFILE_COPY_NUMBER_ALTERATION=hnc_mskcc_2016_gistic&genetic_profile_ids_PROFILE_MUTATION_EXTENDED=hnc_mskcc_2016_mutations&tab_index=tab_visualize.Citation134,Citation135Abbreviation: OS, overall survival.

PIK3CA mutations seem to favor advanced HNSCC, which harbor multiple PI3K-pathway mutations.Citation38 Lui et al detected that 100% HNSCC (10/10 cases) with concurrent PI3K mutations were advanced (Stage IV).Citation75 PIK3CA copy number gain (high polysomy or amplification) but not PIK3CA mutation was detected to be associated with significantly lower disease-free survival, resulting from an NGS.Citation77 The most frequent mutations occur in exons 9 and 20, with hotspots at H1047R (eight mutations total), E542K (three mutations), and E545K/G (four mutations).Citation78 H1047R was more potent than E545K at inducing resistance in PI3K pathway activation.Citation10 PIK3CA canonical and novel mutations increase survival. These canonical hotspot mutations showed significantly enhanced HNSCC growth compared to overexpression of WT PIK3CA.Citation74 This observation implies that the PI3K pathway-mutated HNSCC tumors have “onco-genic” advantage even with genomic instability, which can be partly explained by PIK3CA “driver” mutations’ growth-promoting activity.

Although there is not enough evidence associated with oversurvival and recurrence rates, PIK3CA alterations are providing a potential approach to treat a substantial subset of patients in advanced stage.

Targeted PIK3CA activation

There are currently a variety of different inhibitors that are being studied for targeting the PI3K pathway.Citation74,Citation79 PIK3CA activation either caused by mutation or copy number gain is oncogenic and is thus a commonly activating point mutation with targeted agents.Citation73,Citation74 Targeting the PIK3CA activation has emerged as one of the most promising therapeutic targets in HNSCC.Citation75 They are already in clinical development for other cancer types.Citation10

HNSCC cell lines containing hotspots endogenous PIK3CA (H1047R) mutations demonstrated increased sensitivity to PI3K pathway inhibition by the mTOR/PI3K inhibitor BEZ-235 compared to the representative HNSCC cells with WT PIK3CA.Citation80 Furthermore, they were more sensitive to the combination of BEZ-235 plus cetuximab compared to cetuximab alone.Citation80 PIK3CA mutations are more common in HPV-positive HNSCC and are associated with activation of mTOR.Citation4,Citation74,Citation81 Patient-derived HPV-positive OPSCC tumor-grafts in vivo with PIK3CA (E542K) mutation were also sensitive to a dual mTOR/PI3K inhibitor (BEZ-235), in contrast to PIK3CA WT tumorgrafts.Citation76,Citation80 These results suggest that mTOR/PI3K inhibitors may have activity against PIK3CA mutant HNSCC, particularly in HPV-positive HNSCC.

Activated PI3K/Akt confers resistance to MET inhibitors

PIK3CA and MET may play an important role in oncogenesis of certain specific subtypes of head and neck cancer. In several cancers, gene copy numbers of MET and PIK3CA have been found to be prognostic and predictive for therapy response.Citation82 The MET receptor tyrosine kinase represents a promising target in cancer. PIK3CA activating mutations are common in several tumor types and can potentially confer resistance to anti-receptor tyrosine kinase therapy.Citation78 Hotspots E545K and H1047R confer resistance to MET inhibition in MET-driven models. Hence, resistance to MET inhibition could be synergistically overcome by co-targeting PI3K. Combination of PI3K/MET inhibitors led to more-than-additive effects.

Combined MET/PI3K inhibition leads to enhanced antitumor activity in MET-expressing HNSCC harboring endogenous PIK3CA (H1047R).Citation78 PIK3CA mutations can lead to resistance to MET inhibition, supporting future clinical evaluation of combinations of PI3K and MET inhibitors in common scenarios of malignant neoplasms featuring aberrant MET expression and PIK3CA mutations. In conclusion, PI3K signaling pathway was a potential target for treatment optimization across all risk groups.

Activated PI3K/Akt causes resistance of EGFR inhibitors

Activated PI3K/Akt and RAS/MAPK/ERK pathways are associated with resistance to cetuximab-containing chemo-radiation (weekly cetuximab, paclitaxel, and carboplatin followed by chemoradiation with the same regimen) in operable stage III/IV HNSCC, implicated by Eastern Cooperative Oncology Group (ECOG) 2303 Phase II trial.Citation82 Inferior OS and/or PFS were observed in patients harboring activated PI3K/Akt and RAS/MAPK/ERK and maintained significance in multivariable analysis.Citation83 Preclinical studies suggest that PIK3CA and RAS mutations may predict intrinsic resistance to cetuximab, which may be prevented by combination of cetuximab and PI3K and/or mTOR inhibitors in HNSCC patients.Citation84

EGFR

EGFR plays an important role in the emergence and progression of epithelial malignancies and is the most frequently activated receptor tyrosine kinase in HNSCC.Citation85 The first molecular targeting approach to demonstrate a survival advantage for HNSCC patients has emerged in the context of EGFR biology. TCGA data have detected EGFR alteration in 14.34% cases including 65.0% with EGFR amplification. High-level EGFR amplifications in ~10% of HNSCC, and polysomy are present in even more.Citation13 Primary somatic mutations in EGFR are uncommon in HNSCC.Citation86 Most of the previous studies found that EGFR overexpression was associated with radioresistance, and worse locoregional and disease-free survival.Citation87Citation90 However, EGFR alteration also has no significant prognoses value in metastatic head and neck cancer analyzed in cBioPortal (). And because there has been no consistently identifiable alteration in the EGFR that correlates to sensitivity to EGFR inhibitors, EGFR analysis has not been incorporated into the pathologic evaluation of HNSCC.Citation85

EGFR amplification is highly associated with resistance to chemotherapy and radiation in HNSCC.Citation22,Citation91 Hypoxia induces radioresistance, directly as DNA damage is maximized in the presence of oxygen and indirectly by promoting genetic instabilityCitation92 (). Hypoxia induces the expression of EGFR and enhances the phosphorylation of EGFR thereby regulating intrinsic DNA-repair mechanisms, meanwhile EGFR can stabilize hypoxia-inducible factor 1a.Citation93,Citation94 These radioresistance mechanisms play a role in HNSCC because EGFR is overexpressed in most of these tumors. That is why EGFR inhibition has subsequently been explored as a potential therapeutic adjunct to radiotherapy in HNSCC.Citation95,Citation96

Targeting EGFR is lack of response evaluation

Anti-EGFR agents are largely divided into two classes of drugs: monoclonal antibodies and tyrosine kinase inhibitors (TKIs). EGFR inhibitors have been used to treat both lung and head and neck cancers with squamous cell histology.Citation97 These tumors often show high EGFR expression and/or increased gene copy number. The efficacy of cetuximab is demonstrated in the recurrent/metastatic (R/M) HNSCC settings of clinical trials: ECOG, Erbitux in First-line Treatment of Recurrent or Metastatic Head and Neck Cancer (EXTREME) trial. Recent developments included US Food and Drug Administration approval of pembrolizumab and cetuximab. Cetuximab has conferred a survival advantage when combined with platinum-based combination chemotherapy in the first-line R/M setting.Citation98 However, only a fraction of HNSCC are sensitive to the cetuximab, 5-year survival of HNSCC is not improved by the cetuximab, which is currently <40%.Citation82,Citation99

The use of EGFR as a biomarker for response to EGFR inhibitor is limited by the semiquantitative nature of and subjectivity of immunohistochemical scoring.Citation100Citation102 It might be due to inaccurate assays as well as the nature of increased gene copy number being a less predictive biomarker compared to activating mutations.Citation55 Several studies explored the biology, genomics, and patterns of response to EGFR inhibitors to inform identification of potential biomarkers that have shown promise in preclinical studies and clinical trials.Citation85,Citation97,Citation101,Citation103Citation105 But no biomarkers are known to predict response to the treatment.

Vermorken et al demonstrated the superiority of incorporating cetuximab, in combination with cisplatin/carboplatin and fluorouracil in patients with recurrent/metastatic HNSCC.Citation97 Genomic studies suggest that EGFR alteration is more common in HPV-negative tumors compared to HPV-positive tumors.Citation74 Clinical data further contradict the HPV positivity as a biomarker for EGFR-inhibitor resistance.Citation97 Development of EGFR inhibitors could be more relevant in treating HPV-negative tumors.Citation81,Citation106,Citation107

MET

MET alterations as oncogenic driver of HNSCC

Hepatocyte growth factor (HGF) receptor (MET) is functionally novel in HNSCC with prominent overexpression, and increased copy number and mutations.Citation95,Citation108 It regulates cancer cell plasticity through reversible reprogramming of epithelial–mesenchymal transition (EMT).Citation109,Citation110 It is a process considered a critical step in the progression of HNSCC. Accumulating evidence suggests that EMT and its ligand HGF favor the distant dissemination of single carcinoma cells from the site of the primary tumor.Citation109,Citation111

MET/HGF pathway activation driven by MET overex-pression correlated with worse outcome in R/M HNSCC patients for progression-free survival and OS.Citation112 Although MET alterations are relatively low from querying 2,498 samples in 7 studies in TCGA head and neck carcinoma (1.3%) (), MET is proposed as an escape mechanism mediating drug resistance.Citation109,Citation110,Citation113 MET copy number gain or somatic mutations drive metastatic spread of HNSCCCitation108,Citation114 and are significantly associated with shorter disease-specific survival.Citation99 Therefore, MET may be a substantial ontogenetic driver of certain specific subtypes of HNSCC, which should be further explored.

Hence, multiple clinical scenarios targeting the MET in HNSCC have been undertaken because of significant preclinical work demonstrating a relationship between MET/HGF signaling and cancer cell survival (eg, tivantinib, cabozantinib, and crizotinib).Citation114 HGF/c-MET pathway mediates vascular endothelial growth factor receptor (VEGFR) inhibitor resistance and vascular remodeling in non-small-cell lung cancer (NSCLC). Forced HGF expression in NSCLC reduced tumor sensitivity to VEGFR TKIs and produced tumors with increased blood vessels. Dual VEGFR/c-MET signaling inhibition could prevent the vascular morphology alterations.Citation115 These preclinical results indicated that the level of MET expression is a determinant of outcome.Citation112

MET mutations associated with resistance to EGFR inhibitor

The cross-talk of MET and its ligand with EGFR, such as coactivation of alternative kinases, may contribute to EGFR inhibitor resistance.Citation82,Citation112 For example, MET mutations are present in up to 20% of patients with resistance to EGFR inhibitors, like cetuximab.Citation116 In head and neck cancer cell lines MET was shown to mediate resistance to dasatinib and erlotinib.Citation117,Citation118 HGF/MET pathway activation is associated with poorer outcome in R/M HNSCC patients. Therefore, patients treated with EGFR inhibitors have a worse prognosis.Citation119,Citation120 Experimental studies in vitro and in vivo of R/M HNSCC documented a synthetic lethal effect of dual blockade using c-MET (SU11274, PF2341066, and PF04217903) and EGFR TKIs (cetuximab, erlotinib, and gefitinib).Citation118,Citation121,Citation122

Conclusion

Precision medicine in oncology seeks to match each patient with the most effective and appropriate targeted therapies.Citation6 NGS data-guided therapy is promising, in revealing novel molecular alterations, which might be used as drug targets.Citation6

Since the last decade, concepts of cross-talk, co-occurrence, and interactions have been intensively introduced into biochemical research. Various etiologic factors are implicated in the genesis of distinct molecular subsets of HNSCC. Apparently, signaling pathways and transcription factors are not regulated by mutations of a specific gene. Multiple gene mutations, along with the corresponding protein dysfunction or molecular pathway dysregulations, are causative in the process of carcinogenesis or drug resistance.

HPV-positive and -negative HNSCCs are quite distinct clinically and biologically. However, they both can have deregulations within the PI3K pathway.Citation123,Citation124,Citation81 Our review paid less attention on HPV-positive HNSCC, because studies of the HPV-positive HNSCC are scarce. The TCGA cohort comprised 85% HPV-negative HNSCC.Citation3 HPV-negative HNSCC featured novel focal deletions in tumor suppressor genes (eg, NOTCH1 and CDKN2A).Citation13 Also, mutations of TP53, CDKN2A, PIK3CA, and NOTCH genes are enriched. Combination therapy is required. The major challenge is to identify and make good use of the novel genetic interactions, and develop more accurate model systems and innovative clinical trials.

Acknowledgments

We sincerely thank Prof Dr Weidong Han for his suggestions on the theme and language modification.

This study is sponsored by grants from Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (grant no. 2019336033); Zhejiang Provincial Natural Science Foundation of China (grant no. LY15H160028); Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (grant no. 2014KYB129).

Disclosure

The authors report no conflicts of interests in this work.

References

  • BrauswetterDDánosKGurbiBCopy number gain of PIK3CA and Met is associated with poor prognosis in head and neck squamous cell carcinomaVirchows Arch2016468557958726832731
  • WalterVYinXWilkersonMDMolecular subtypes in head and neck cancer exhibit distinct patterns of chromosomal gain and loss of canonical cancer genesPLoS One201382e5682323451093
  • [No authors listed]TCGA releases head and neck cancer dataCancer Discov201554340341
  • LawrenceMSSougnezCLichtensteinLCancer Genome Atlas NetworkComprehensive genomic characterization of head and neck squamous cell carcinomasNature2015517753657658225631445
  • HymanDMSolitDBArcilaMEPrecision medicine at Memorial Sloan Kettering Cancer Center: clinical next-generation sequencing enabling next-generation targeted therapy trialsDrug Discov Today201520121422142826320725
  • MorrisLGChandramohanRWestLThe molecular landscape of recurrent and metastatic head and neck cancers: insights from a precision oncology sequencing platformJAMA Oncol201732244255
  • ZhouGLiuZMyersJNTP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment responseJ Cell Biochem2016117122682269227166782
  • WeissJHayesDNClassifying squamous cell carcinoma of the head and neck: prognosis, prediction and implications for therapyExpert Rev Anticancer Ther201414222923624313652
  • KonieczkowskiDJJohannessenCMGarrawayLAA convergence-based framework for cancer drug resistanceCancer Cell201833580181529763622
  • MuellnerMKUrasIZGappBVA chemical-genetic screen reveals a mechanism of resistance to PI3K inhibitors in cancerNat Chem Biol201171178779321946274
  • ZhouGLiuZMyersJNTP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment responseJ Cell Biochem2016117122682269227166782
  • PerriFIonnaFScarpatiGTranslational research: a future strategy for managing squamous cell carcinoma of the head and neck?Anticancer Agents Med Chem201818912201227
  • Cancer Genome Atlas NetworkComprehensive genomic characterization of head and neck squamous cell carcinomasNature2015517753657658225631445
  • StranskyNEgloffAMTwardADThe mutational landscape of head and neck squamous cell carcinomaScience201133360461157116021798893
  • AgrawalNFrederickMJPickeringCRExome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in Notch1Science201133360461154115721798897
  • OmuraGAndoMEbiharaYThe prognostic value of TP53 mutations in hypopharyngeal squamous cell carcinomaBMC Cancer201717189829282038
  • ShahnavazSARegeziJABradleyGDubéIDJordanRCP53 gene mutations in sequential oral epithelial dysplasias and squamous cell carcinomasJ Pathol2000190441742210699989
  • ManterolaLAguirrePLarreaEMutational profiling can identify laryngeal dysplasia at risk of progression to invasive carcinomaSci Rep201881661329700339
  • ZhaoDTahaneyWMMazumdarASavageMIBrownPHMolecularly targeted therapies for p53-mutant cancersCell Mol Life Sci201774224171418728643165
  • OckCYSonBKeamBIdentification of genomic mutations associated with clinical outcomes of induction chemotherapy in patients with head and neck squamous cell carcinomaJ Cancer Res Clin Oncol2016142487388326677030
  • NiehrFEderTPilzTMultilayered OMICs-based analysis of a head and neck cancer model of cisplatin resistance reveals intratumoral heterogeneity and treatment-induced clonal selectionClin Cancer Res201824115816829061642
  • GadhikarMASciutoMRAlvesMVChk1/2 inhibition overcomes the cisplatin resistance of head and neck cancer cells secondary to the loss of functional p53Mol Cancer Ther20131291860187323839309
  • StrojanPKarnerKSmidLConcomitant chemoradiotherapy with mitomycin C and cisplatin in advanced unresectable carcinoma of the head and neck: phase I-II clinical studyInt J Radiat Oncol Biol Phys200872236537218394816
  • van HoutenVMLeemansCRKummerJAMolecular diagnosis of surgical margins and local recurrence in head and neck cancer patients: a prospective studyClin Cancer Res200410113614362015173067
  • van HoutenVMLeemansCRKummerJAMolecular diagnosis of surgical margins and local recurrence in head and neck cancer patients: a prospective studyClin Cancer Res200410113614362015173067
  • LiZGonzalezCLWangBCdkn2a suppresses metastasis in squamous cell carcinomas induced by the gain-of-function mutant p53(R172H)J Pathol2016240222423427447534
  • ZhouGLiuZMyersJNTP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment responseJ Cell Biochem2016117122682269227166782
  • PerroneFBossiPCortelazziBTP53 mutations and pathologic complete response to neoadjuvant cisplatin and fluorouracil chemotherapy in resected oral cavity squamous cell carcinomaJ Clin Oncol201028576176620048189
  • HanelWMarchenkoNXuSYuSXWengWMollUTwo hot spot mutant p53 mouse models display differential gain of function in tumorigenesisCell Death Differ201320789890923538418
  • SongHHollsteinMXuYP53 gain-of-function cancer mutants induce genetic instability by inactivating ATMNat Cell Biol20079557358017417627
  • SkinnerHDSandulacheVCOwTJTP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescenceClin Cancer Res201218129030022090360
  • NeskeyDMOsmanAAOwTJEvolutionary action score of TP53 identifies high-risk mutations associated with decreased survival and increased distant metastases in head and neck cancerCancer Res20157571527153625634208
  • OsmanAAMonroeMMOrtega AlvesMVWee-1 kinase inhibition overcomes cisplatin resistance associated with high-risk TP53 mutations in head and neck cancer through mitotic arrest followed by senescenceMol Cancer Ther201514260861925504633
  • OsmanAANeskeyDMKatsonisPEvolutionary action score of TP53 coding variants is predictive of platinum response in head and neck cancer patientsCancer Res20157571205121525691460
  • LambertJMMoshfeghAHainautPWimanKGBykovVJMutant p53 reactivation by PRIMA-1MET induces multiple signaling pathways converging on apoptosisOncogene20102991329133819946333
  • RieberMStrasberg-RieberMHypoxia, Mn-SOD and H(2)O(2) regulate p53 reactivation and PRIMA-1 toxicity irrespective of p53 status in human breast cancer cellsBiochem Pharmacol201284121563157022982566
  • ZandiRSelivanovaGChristensenCLGerdsTAWillumsenBMPoulsenHSPRIMA-1Met/APR-246 induces apoptosis and tumor growth delay in small cell lung cancer expressing mutant p53Clin Cancer Res20111792830284121415220
  • HerzogABianYvander BroekRPI3K/mTOR inhibitor PF-04691502 antitumor activity is enhanced with induction of wild-type TP53 in human xenograft and murine knockout models of head and neck cancerClin Cancer Res201319143808381923640975
  • FriedmanJNottinghamLDuggalPDeficient TP53 expression, function, and cisplatin sensitivity are restored by quinacrine in head and neck cancerClin Cancer Res20071322 Pt 16568657818006756
  • SinghBReddyPGGoberdhanAP53 regulates cell survival by inhibiting PIK3CA in squamous cell carcinomasGenes Dev200216898499311959846
  • AndersonRTKeysarSBBowlesDWThe dual pathway inhibitor rigosertib is effective in direct patient tumor xenografts of head and neck squamous cell carcinomasMol Cancer Ther201312101994200523873848
  • TodorovaTAJordanovSHStanchevaGSMutational status of CDKN2A and TP53 genes in laryngeal squamous cell carcinomaPathol Oncol Res201521241342125149524
  • Ben-DayanMMOwTJBelbinTJNonpromoter methylation of the CDKN2A gene with active transcription is associated with improved locoregional control in laryngeal squamous cell carcinomaCancer Med20176239740728102032
  • LoyoMLiRJBettegowdaCLessons learned from next-generation sequencing in head and neck cancerHead Neck201335345446322907887
  • LarqueABCondeLHakimSP16(INK (4)a) overexpression is associated with CDKN2A mutation and worse prognosis in HPV-negative laryngeal squamous cell carcinomasVirchows Arch2015466437538225652585
  • PosnerMRLorchJHGoloubevaOSurvival and human papillomavirus in Oropharynx cancer in Tax 324: a subset analysis from an international phase III trialAnn Oncol20112251071107721317223
  • RischinDYoungRJFisherRPrognostic significance of p16INK4a and human papillomavirus in patients with oropharyngeal cancer treated on TROG 02.02 phase III trialJ Clin Oncol201028274142414820697079
  • ChungCHParkerJSKaracaGMolecular classification of head and neck squamous cell carcinomas using patterns of gene expressionCancer Cell20045548950015144956
  • JouAHessJEpidemiology and molecular biology of head and neck cancerOncol Res Treat201740632833228531899
  • HuangSHPerez-OrdonezBWeinrebINatural course of distant metastases following radiotherapy or chemoradiotherapy in HPV-related oropharyngeal cancerOral Oncol2013491798522917550
  • RiekeDTKeilholzUSystemic treatment in HPV-induced recurrent or metastatic HNSCCRecent Results Cancer Res201720614916027699536
  • LópezFSampedroTLlorenteJLHermsenMÁlvarez-MarcosCAlterations of p14 ARF, p15 INK4b, and p16 INK4a genes in primary laryngeal squamous cell carcinomaPathol Oncol Res2017231637127377733
  • SinhaPThorstadWTNussenbaumBDistant metastasis in p16-positive oropharyngeal squamous cell carcinoma: a critical analysis of patterns and outcomesOral Oncol2014501455124211084
  • TodorovaTAJordanovSHStanchevaGSMutational status of CDKN2A and TP53 genes in laryngeal squamous cell carcinomaPathol Oncol Res201521241342125149524
  • KangHKiessAChungCHEmerging biomarkers in head and neck cancer in the era of genomicsNat Rev Clin Oncol2015121112625403939
  • HuntJLBarnesLLewisJSMolecular diagnostic alterations in squamous cell carcinoma of the head and neck and potential diagnostic applicationsEur Arch Otorhinolaryngol2014271221122323467835
  • NamazieAAlaviSOlopadeOICyclin D1 amplification and p16(MTS1/CDK4I) deletion correlate with poor prognosis in head and neck tumorsLaryngoscope2002112347248112148857
  • NamazieAAlaviSOlopadeOICyclin D1 amplification and p16(MTS1/CDK4I) deletion correlate with poor prognosis in head and neck tumorsLaryngoscope2002112347248112148857
  • MaahsGSMachadoDCJeckel-NetoEAMichaelsesVSCyclin D1 expression and cervical metastases in squamous cell carcinoma of the mouthBraz J Otorhinolaryngol2007731879417505605
  • ZhangPZhangZZhouXQiuWChenFChenWIdentification of genes associated with cisplatin resistance in human oral squamous cell carcinoma cell lineBMC Cancer2006622416978399
  • NoelEEYeste-VelascoMMaoXThe association of CCND1 overexpression and cisplatin resistance in testicular germ cell tumors and other cancersAm J Pathol201017662607261520395447
  • Mielcarek-KuchtaDOlofssonJGolusinskiWP53, Ki67 and cyclin D1 as prognosticators of lymph node metastases in laryngeal carcinomaEur Arch Otorhinolaryngol20032601054955414551784
  • TiedemannREMaoXShiCXIdentification of kinetin riboside as a repressor of CCND1 and CCND2 with preclinical antimyeloma activityJ Clin Invest200811851750176418431519
  • KalishLHKwongRAColeIEGallagherRMSutherlandRLMusgroveEADeregulated cyclin D1 expression is associated with decreased efficacy of the selective epidermal growth factor receptor tyrosine kinase inhibitor gefitinib in head and neck squamous cell carcinoma cell linesClin Cancer Res200410227764777415570011
  • TimpsonPWilsonASLehrbachGMSutherlandRLMusgroveEADalyRJAberrant expression of cortactin in head and neck squamous cell carcinoma cells is associated with enhanced cell proliferation and resistance to the epidermal growth factor receptor inhibitor gefitinibCancer Res200767199304931417909038
  • KalishLHKwongRAColeIEGallagherRMSutherlandRLMusgroveEADeregulated cyclin D1 expression is associated with decreased efficacy of the selective epidermal growth factor receptor tyrosine kinase inhibitor gefitinib in head and neck squamous cell carcinoma cell linesClin Cancer Res200410227764777415570011
  • PickeringCRZhangJYooSYIntegrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic driversCancer Discov20133777078123619168
  • VermezovicJAdamowiczMSantarpiaLNotch is a direct negative regulator of the DNA-damage responseNat Struct Mol Biol201522541742425895060
  • TinhoferIBudachVSakiMTargeted next-generation sequencing of locally advanced squamous cell carcinomas of the head and neck reveals druggable targets for improving adjuvant chemoradiationEur J Cancer201657788626896955
  • TinhoferIStenzingerAEderTTargeted next-generation sequencing identifies molecular subgroups in squamous cell carcinoma of the head and neck with distinct outcome after concurrent chemoradiationAnn Oncol201627122262226827681865
  • VettoreALRamnarayananKPooreGMutational landscapes of tongue carcinoma reveal recurrent mutations in genes of therapeutic and prognostic relevanceGenome Med201579826395002
  • CortelazziBVerderioPCiniselliCMReceptor tyrosine kinase profiles and human papillomavirus status in oropharyngeal squamous cell carcinomaJ Oral Pathol Med201544973474525495427
  • SewellABrownBBiktasovaAReverse-phase protein array profiling of oropharyngeal cancer and significance of PIK3CA mutations in HPV-associated head and neck cancerClin Cancer Res20142092300231124599934
  • FeldmanRGatalicaZKnezeticJMolecular profiling of head and neck squamous cell carcinomaHead Neck201638Suppl 1E1625E163826614708
  • LuiVWHedbergMLLiHFrequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkersCancer Discov20133776176923619167
  • ChauNGLiYYJoVYIncorporation of next-generation sequencing into routine clinical care to direct treatment of head and neck squamous cell carcinomaClin Cancer Res201622122939294926763254
  • NisaLHäfligerPPoliakováMPIK3CA hotspot mutations differentially impact responses to Met targeting in MET-driven and non-driven preclinical cancer modelsMol Cancer20171619328532501
  • FokasEImJHHillSDual inhibition of the PI3K/mTOR pathway increases tumor radiosensitivity by normalizing tumor vasculatureCancer Res201272123924822108822
  • SoulièresDFaivreSMesíaRBuparlisib and paclitaxel in patients with platinum-pretreated recurrent or metastatic squamous cell carcinoma of the head and neck (BERIL-1): a randomised, double-blind, placebo-controlled phase 2 trialLancet Oncol201718332333528131786
  • SeiwertTYZuoZKeckMKIntegrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomasClin Cancer Res201521363264125056374
  • ArnoldLEndersJThomasSMActivated HGF-c-Met axis in head and neck cancerCancers2017912E169:16929231907
  • PsyrriALeeJWPectasidesEPrognostic biomarkers in phase II trial of cetuximab-containing induction and chemoradiation in resectable HNSCC: Eastern Cooperative Oncology Group E2303Clin Cancer Res201420113023303224700741
  • WangZMartinDMolinoloAAmTOR co-targeting in cetuximab resistance in head and neck cancers harboring PIK3CA and RAS mutations.J Natl Cancer Inst20141069
  • MisiukiewiczKDangRPParidesMEndothelial growth factor receptor inhibitors in recurrent metastatic cancer of the head and neckHead Neck201638Suppl 1E2221E222825900280
  • MartinDAbbaMCMolinoloAAThe head and neck cancer cell oncogenome: a platform for the development of precision molecular therapiesOncotarget20145198906892325275298
  • AhmadPSanaJSlavikMSlampaPSmilekPSlabyOMicroRNAs involvement in radioresistance of head and neck cancerDis Markers2017201718
  • FlezarMSKirbisISPopovićKSStrojanPRadiosensitivity of squamous cell carcinoma metastases to the neck assessed by immunocytochemical profiling of fine-needle aspiration biopsy cell specimens: a pilot studyRadiother Oncol200993357558019850367
  • MetheetrairutCSlackFJMicroRNAs in the ionizing radiation response and in radiotherapyCurr Opin Genet Dev2013231121923453900
  • SummererINiyaziMUngerKChanges in circulating microR-NAs after radiochemotherapy in head and neck cancer patientsRadiat Oncol2013829624373621
  • BrandTMIidaMLutharNStarrMMHuppertEJWheelerDLNuclear EGFR as a molecular target in cancerRadiother Oncol2013108337037723830194
  • ToulanyMKasten-PisulaUBrammerIBlockage of epidermal growth factor receptor-phosphatidylinositol 3-kinase-Akt signaling increases radiosensitivity of K-ras mutated human tumor cells in vitro by affecting DNA repairClin Cancer Res200612134119412616818713
  • JanssensGORademakersSETerhaardCHAccelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: results of a phase III randomized trialJ Clin Oncol201230151777178322508814
  • SwinsonDEO’ByrneKJInteractions between hypoxia and epidermal growth factor receptor in non-small-cell lung cancerClin Lung Cancer20067425025616512978
  • MoreiraJTobiasAO’BrienMPAgulnikMTargeted therapy in head and neck cancer: an update on current clinical developments in epidermal growth factor receptor-targeted therapy and immunothera-piesDrugs201777884385728382569
  • NijkampMMSpanPNTerhaardCHEpidermal growth factor receptor expression in laryngeal cancer predicts the effect of hypoxia modification as an additive to accelerated radiotherapy in a randomised controlled trialEur J Cancer201349153202320923867129
  • JuergensRABratmanSVTsaoMSBiology and patterns of response to EGFR-inhibition in squamous cell cancers of the lung and head & neckCancer Treat Rev201754435728192747
  • VermorkenJBMesiaRRiveraFPlatinum-based chemotherapy plus cetuximab in head and neck cancerN Engl J Med2008359111116112718784101
  • BrauswetterDDánosKGurbiBCopy number gain of PIK3CA and Met is associated with poor prognosis in head and neck squamous cell carcinomaVirchows Arch2016468557958726832731
  • MilikSNLasheenDSSeryaRATAbouzidKAMHow to train your inhibitor: design strategies to overcome resistance to epidermal growth factor receptor inhibitorsEur J Med Chem201714213115128754471
  • KontićMMilovanovićJČolovićZEpidermal growth factor receptor (EGFR) expression in patients with laryngeal squamous cell carcinomaEur Arch Otorhinolaryngol2015272240140525294054
  • MarimuthuAChavanSSatheGIdentification of head and neck squamous cell carcinoma biomarker candidates through proteomic analysis of cancer cell secretomeBiochim Biophys Acta20131834112308231623665456
  • AnsellAJedlinskiAJohanssonACRobergKEpidermal growth factor is a potential biomarker for poor cetuximab response in tongue cancer cellsJ Oral Pathol Med201645191625677871
  • ZanottiLPadernoAPiazzaCEpidermal growth factor receptor detection in serum and saliva as a diagnostic and prognostic tool in oral cancerLaryngoscope201712711E408E41428782189
  • van DijkLKBoermanOCKaandersJHBussinkJEpidermal growth factor receptor imaging in human head and neck cancer xenograftsActa Oncol20155491263126726248024
  • BeckTNGeorgopoulosRShagisultanovaEIEGFR and Rb1 as dual biomarkers in HPV-negative head and neck cancerMol Cancer Ther201615102486249727507850
  • RomanitanMNäsmanAMunck-WiklandEDalianisTRamqvistTEGFR and phosphorylated EGFR in relation to HPV and clinical outcome in tonsillar cancerAnticancer Res20133341575158323564800
  • SeiwertTYJagadeeswaranRFaoroLThe Met receptor tyrosine kinase is a potential novel therapeutic target for head and neck squamous cell carcinomaCancer Res20096973021303119318576
  • NietoMAHuangRYJacksonRAThieryJPEMT: 2016Cell20161661214527368099
  • TsaiJHYangJEpithelial-mesenchymal plasticity in carcinoma metastasisGenes Dev201327202192220624142872
  • TamWLWeinbergRAThe epigenetics of epithelial-mesenchymal plasticity in cancerNat Med201319111438144924202396
  • SzturzPRaymondEFaivreSc-MET-mediated resistance to EGFR inhibitors in head and neck cancer: how to move from bench to bedsideOral Oncol201659e12e1427302885
  • CitronFArmeniaJFranchinGAn integrated approach identifies mediators of local recurrence in head and neck squamous carcinomaClin Cancer Res201723143769378028174235
  • SzturzPRaymondEAbitbolCAlbertSde GramontAFaivreSUnderstanding c-MET signalling in squamous cell carcinoma of the head & neckCrit Rev Oncol Hematol2017111395128259294
  • GarufiATrisciuoglioDPorruMA fluorescent curcumin-based Zn(II)-complex reactivates mutant (R175H and R273H) p53 in cancer cellsJ Exp Clin Cancer Res2013327224220325
  • CañadasIRojoFArumí-UríaMRoviraAAlbanellJArriolaEC-Met as a new therapeutic target for the development of novel anticancer drugsClin Transl Oncol201012425326020462834
  • Madoz-GúrpideJZazoSChamizoCActivation of Met pathway predicts poor outcome to cetuximab in patients with recurrent or metastatic head and neck cancerJ Transl Med20151328226319934
  • XuHStabileLPGubishCTGoodingWEGrandisJRSiegfriedJMDual blockade of EGFR and c-Met abrogates redundant signaling and proliferation in head and neck carcinoma cellsClin Cancer Res201117134425443821622718
  • SeiwertTYJagadeeswaranRFaoroLThe Met receptor tyrosine kinase is a potential novel therapeutic target for head and neck squamous cell carcinomaCancer Res20096973021303119318576
  • StabileLPHeGLuiVWc-Src activation mediates erlotinib resistance in head and neck cancer by stimulating c-MetClin Cancer Res201319238039223213056
  • MoorenJJGültekinSEStraetmansJMP16(INK4A) immunostaining is a strong indicator for high-risk-HPV-associated oropharyngeal carcinomas and dysplasias, but is unreliable to predict low-risk-HPV-infection in head and neck papillomas and laryngeal dysplasiasInt J Cancer201413492108211724127203
  • WallineHMCareyTEGoudsmitCMHigh-risk HPV, biomarkers, and outcome in matched cohorts of head and neck cancer patients positive and negative for HIVMol Cancer Res201715217918827899422
  • OhnamiSOhshimaKNagashimaTComprehensive characterization of genes associated with the TP53 signal transduction pathway in various tumorsMol Cell Biochem20174311–2758528258440
  • VainshteinJMSpectorMEMcHughJBRefining risk stratification for locoregional failure after chemoradiotherapy in human papillomavirus-associated oropharyngeal cancerOral Oncol201450551351924565983
  • KanyilmazGEkinciOMugeACelikSOzturkFHPV-Associated p16INK4A Expression and Response to Therapy and Survival in Selected Head and Neck CancersAsian Pacific Journal of Cancer Prevention201516125325825640361
  • SenBPengSSaigalBWilliamsMDJohnsonFMDistinct Interactions Between c-Src and c-Met in Mediating Resistance to c-Src Inhibition in Head and Neck CancerClin Cancer Res201117351452421106725
  • CasconeTXuLLinHYThe HGF/c-MET Pathway Is a Driver and Biomarker of VEGFR-inhibitor Resistance and Vascular Remodeling in Non-Small Cell Lung CancerClin Cancer Res20172318489550127435400
  • HoadleyKAYauCHinoueTCell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancerCell201845173229130429625048
  • EllrottKBaileyMHSaksenaGScalable open science approach for mutation calling of tumor exomes using multiple genomic pipelinesCell systems20183286327128129596782
  • TaylorAMShihJHaGGenomic and functional approaches to understanding cancer aneuploidyCancer Cell20184933467668929622463
  • GaoQLiangWWFoltzSMDriver fusions and their implications in the development and treatment of human cancersCell reports20184323122733829617662
  • LiuJLichtenbergTHoadleyKAAn integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analyticsCell201845173240041629625055
  • Sanchez-VegaFMinaMArmeniaJOncogenic Signaling Pathways in The Cancer Genome AtlasCell201845173232133729625050
  • LiTWenHBraytonCEpidermal growth factor receptor and notch pathways participate in the tumor suppressor function of gamma-secretaseJ Biol Chem200728244322643227317827153
  • GaoJAksoyBADogrusozUIntegrative analysis of complex cancer genomics and clinical profiles using the cBioPortalSci Signal20136269l1
  • CeramiEGaoJDogrusozUThe cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics dataCancer Discov20122540140422588877