Next Article in Journal
Droplet Digital PCR for BCR–ABL1 Monitoring in Diagnostic Routine: Ready to Start?
Next Article in Special Issue
New Scenarios in Pharmacological Treatments of Head and Neck Squamous Cell Carcinomas
Previous Article in Journal
Performance of Contrast-Enhanced Ultrasound in Thyroid Nodules: Review of Current State and Future Perspectives
Previous Article in Special Issue
Epithelial-to-Mesenchymal Transition-Derived Heterogeneity in Head and Neck Squamous Cell Carcinomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Emerging Molecular Therapies for Head and Neck Squamous Cell Carcinoma

by
Farzaneh Kordbacheh
1,2,3 and
Camile S. Farah
4,5,6,7,*
1
Broad Institute of MIT and Harvard, Boston, MA 02142, USA
2
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
3
ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT 0200, Australia
4
The Australian Centre for Oral Oncology Research & Education, Nedlands, WA 6009, Australia
5
Genomics for Life, Milton, QLD 4064, Australia
6
Anatomical Pathology, Australian Clinical Labs, Subiaco, WA 6009, Australia
7
Head and Neck Cancer Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(21), 5471; https://doi.org/10.3390/cancers13215471
Submission received: 2 July 2021 / Revised: 9 September 2021 / Accepted: 28 October 2021 / Published: 30 October 2021

Abstract

:

Simple Summary

Next-generation sequencing of head and neck squamous cell carcinoma has revealed multiple new gene mutations, while simultaneously confirming the role of others in head and neck cancer tumorigenesis. The ever-expanding number of actionable druggable targets has fuelled a plethora of clinical trials assessing various pharmacotherapeutics in the management of this group of cancers. This review paper summarizes the state of play of molecular therapies used in head and neck oncology with particular focus dedicated to current FDA-approved drugs and emerging therapies undergoing advanced clinical trials.

Abstract

Head and neck cancer affects nearly 750,000 patients, with more than 300,000 deaths annually. Advances in first line surgical treatment have improved survival rates marginally particularly in developed countries, however survival rates for aggressive locally advanced head and neck cancer are still poor. Recurrent and metastatic disease remains a significant problem for patients and the health system. As our knowledge of the genomic landscape of the head and neck cancers continues to expand, there are promising developments occurring in molecular therapies available for advanced or recalcitrant disease. The concept of precision medicine is underpinned by our ability to accurately sequence tumour samples to best understand individual patient genomic variations and to tailor targeted therapy for them based on such molecular profiling. Not only is their purported response to therapy a factor of their genomic variation, but so is their inclusion in biomarker-driven personalised medicine therapeutic trials. With the ever-expanding number of molecular druggable targets explored through advances in next generation sequencing, the number of clinical trials assessing these targets has significantly increased over recent years. Although some trials are focussed on first-line therapeutic approaches, a greater majority are focussed on locally advanced, recurrent or metastatic disease. Similarly, although single agent monotherapy has been found effective in some cases, it is the combination of drugs targeting different signalling pathways that seem to be more beneficial to patients. This paper outlines current and emerging molecular therapies for head and neck cancer, and updates readers on outcomes of the most pertinent clinical trials in this area while also summarising ongoing efforts to bring more molecular therapies into clinical practice.

1. Introduction

The druggable genome is defined as the altered genes or gene products that can interact with molecules exhibiting therapeutic properties [1,2]. Genomic alterations include gene deletions and amplifications that change gene abundance and their downstream products, alternative splicing or translocations that can create new proteins, and mutations such as single base changes that can modify protein activity [3,4]. Although genomic mutations are found in all cancers, they can be divided into two main groups. “Passenger” mutations, are the product of carcinogens and genomic instability that appear not to be involved in cancer progression, and “driver” mutations which are known to contribute to tumour development or progression [5,6,7]. The distinction between these mutations can change throughout the course of disease [8]. Driver mutations and their associated cellular pathways are known as “actionable” targets and may have significant diagnostic, prognostic, or therapeutic implications. A subset of these events may be druggable, making them valuable targets for therapeutic development and intervention [7,9].
Partial cancer genome datasets such as The Cancer Genome Atlas (TCGA, http://cancergenome.nih.gov) and the International Cancer Genome Consortium (ICGC, http://www.icgc.org) are available for many types of cancers [10,11]. TCGA data has more recently been used in large scale analysis of driver genes, incorporating over 9000 tumour exomes and resulting in identification of 299 unique cancer genes. This includes 59 novel driver genes and a number of known genes not previously associated with a specific tissue [12]. This comprehensive analysis across 33 cancer types highlighted the prevalence of clinically actionable cancer driver events in TCGA tumour samples [12]. It is estimated that nearly half the samples studied harboured a clinically relevant mutation, by predicting either sensitivity or resistance to certain treatments or clinical trial eligibility [12], with 57% of TCGA cases harbouring at least one potentially clinically actionable target.
Studies of whole-genome analysis of breast cancer patients has demonstrated new copy-number variations, descriptions of driver and other mutations, and elevated mutation rates in treatment-resistant tumours [13]. Banerji et al. has shown recurrent somatic mutations in PIK3CA, TP53, AKT1, GATA3 and MAP3K1 genes, and new recurrent mutations and deletions for CBFB and RUNX1 [14]. Furthermore, studies have also shown that only 36% of mutations are detected as transcribed [15], and that many mutations encode truncated proteins [16]. It is unlikely that these novel findings would have been discovered using conventional sequencing or genotyping approaches.
Some actionable mutations are commonly altered in several different cancer types, and it is reasonable to assume that a therapy effective for one may be applicable in another. BRAF V600E is a gene marker for vemurafenib therapy in melanoma [17], which can also be used in ovarian cancer [18]. On the other hand, a specific genetic abnormality may not confer the same sensitivity to an agent across all cancers. Trastuzumab (Herceptin—anti HER2 monoclonal antibody mAb) has been shown to benefit breast and gastric cancer patients with HER2-amplification, but not in patients with endometrial or ovarian cancer [19,20,21].
PanCancer analysis of driver genes in TCGA studies has grouped head and neck squamous cell carcinoma (HNSCC) with other cancers of squamous origin, where they displayed a higher proportion of mutations in immune and receptor tyrosine kinase signalling genes as well as genes involved in chromatin remodelling [12]. KIF1A, a kinesin, was identified as a novel HNSCC driver gene. Interestingly though, this analysis identified that 71% of the 502 HNSCC samples in the dataset had potentially actionable mutations (either SNPs/Indels, CNVs or all), but that only 16% had a druggable mutation at any stage of development from preclinical to U.S. Food and Drug Administration (FDA)-approved at time of publication. This is in contrast to cutaneous melanoma, for which 93% of mutations were potentially actionable, and 90% were druggable, including a high proportion already FDA-approved. The availability of these large cancer datasets has resulted in the validation of many new driver mutations, clinically useful biomarkers and pharmacotherapeutic developments. Those with specific application in the treatment of HNSCC are listed in Table 1.
This paper summarises current and emerging molecular therapies in head and neck oncology driven by our understanding of gene mutations and genomic oncological pathways relevant to head and neck cancers. FDA-approved therapies, in addition to those in Phase II and III clinical trials, are outlined. Some such as EGFR inhibitors are more common place and used widely, while others such as p53 stimulants utilising adenovirus vectors are more recent advancements in the field and have received limited exposure. The generally poor outcomes for head and neck cancers continue to drive innovation in targeted therapeutic approaches, repurposing of currently available pharmacotherapeutics, and clinical trials to test safety, efficacy and potential combination therapies. Understanding of these current and emerging therapies for HNSCC necessitates a sound understanding of the signaling pathways involved in these cancers. For this, readers are directed to earlier papers detailing the molecular landscape, oncological pathways and druggable targets for head and neck cancers [22,23]. This review is intended to act as a coherent comprehensive summary of current and emerging molecular therapies for head and neck cancers (HNC). It outlines clinical trials (with corresponding ClinicalTrials.gov Identifiers) for HNC when available, and for other cancers where relevant in order to highlight clinical utility for possible extrapolation of relevant findings such as progression free survival (PFS), overall survival (OS), toxicity and adverse effects. Trials with reported outcomes in the literature are addressed in more detail than those still ongoing or terminated.

2. Current and Emerging Molecular Therapies

2.1. EGFR Pathway

Mutation and overexpression of EGFR is associated with a number of tumours including breast, lung, colorectal, ovary and prostate [24,25,26]. The frequency of EGFR mutations is 4% in HNSCC. Notably they have been the most promising candidate for developing molecular therapies.
EGFR is a transmembrane receptor of the human epidermal receptor (HER) family of growth factor receptors. Formation of EGFR homodimers or heterodimers (i.e., with HER2) trigger intracellular pathways that lead to cancer cell proliferation, apoptotic arrest, activation of invasion and metastasis, and stimulation of tumour-induced neovascularisation [27,28].
Currently, two primary approaches have been taken to target EGFR with different mechanisms including inhibition of tyrosine kinase domain (intracellular) activity with small molecules, and inhibition of extracellular ligand binding using monoclonal antibodies (mAbs). Anti-EGFR monoclonal antibodies (cetuximab, panitumumab) recognise EGFR exclusively and bind to its extracellular domain, compete for receptor binding and block ligand-induced EGFR tyrosine kinase activation [29]. EGFR tyrosine kinase inhibitors (erlotinib, gefitinib) compete in a reversible fashion with ATP and bind to the intracellular domain of EGFR tyrosine kinase thus inhibiting EGFR autophosphorylation and downstream signalling. EGFR tyrosine kinase inhibitors can also block different growth factor receptor tyrosine kinases such as VEGFR. Cetuximab is FDA- and EMEA-approved, in combination with radiotherapy, to treat locally advanced, unresectable HNSCC. It is also approved as monotherapy for metastatic disease in patients who have failed to respond to chemotherapy.
An open-label phase II randomised trial applied cetuximab, 5-fluorouracil and cisplatin with or without docetaxel in patients with recurrent and/or metastatic HNSCC. The median follow-up was 2 years. The median overall survival for the group with docetaxel (DPFC) compared to that without (PFC), was 8.9 vs. 10.6 months and response rates were 38.2% vs. 31.9%, respectively [30].
Binding of EGFR to its ligands triggers two key signalling pathways, PI3K-AKT and MAPK/ERK. These signalling pathways can be initiated by mutations in intermediate molecules. Mellinghoff et al. showed that up to 20% of glioblastoma patients were responsive to EGFR kinase inhibition which was associated with coexpression of EGFR vIII and PTEN [25]. Additionally, activating mutations in KRAS results in EGFR-independent signal activation and has been noted in up to 30% and 45% of patients with small-cell lung cancer and colorectal cancer, respectively, with a history of smoking [31,32]. Such patients show resistance or limited efficacy to cetuximab and panitumumab therapy [33,34]. MET amplification also leads to EGFR-independent activation of the PI3K-AKT pathway through activation of HER3. Inhibition of MET signalling has been shown to restore the sensitivity of lung cancer cell lines to gefitinib [35].

2.1.1. Irreversible EGFR Inhibitors

In HNSCC, tyrosine kinase inhibitors (TKIs) are generally quinazoline-derived, low molecular weight synthetic molecules that competitively block the intracellular ATP binding domain of EGFR and consequently inhibit the activation and phosphorylation of EGFR and its downstream cellular intermediates [36,37]. These molecules can be administrated orally. Gefitinib and erlotinib are relatively specific for EGFR and are currently being used for lung cancer [37,38].
Gefitinib (Iressa, ZD1839, NSC 715055) is the first TKI to reach Phase III trials in HNSCC but according to several trial study failures it has been withdrawn in the United States. In a Phase III study (NCT00206219), 477 patients with recurrent or metastatic HNSCC were randomly assigned to compare survival rate after treating with gefitinib 250 mg/day (FDA-approved dose for advanced non-small-cell lung cancer (NSCLC)) or 500 mg/day (orally) or standard methotrexate (40 mg/m2 intravenously). The results showed that neither gefitinib 250 nor 500 mg/day improved overall survival compared with methotrexate [39]. Another Phase III trial conducted by the Eastern Cooperative Oncology Group (ECOG) with 270 patients was closed early at interim analysis due to the unlikelihood of meeting its endpoint goals. The results showed that adding gefitinib to docetaxel was well tolerated but did not improve outcomes in poor prognosis of patients with recurrent or metastatic HNSCC (NCT00088907) [40]. Therefore, clinical development of gefitinib for HNSCC is unlikely to proceed further.
Erlotinib (Tarceva, OSI-774) is an approved therapy in the U.S. for advanced pancreatic cancer [41] and NSCLC [42], and so far has demonstrated promising results in HNSCC clinical trials [43]. In a multicentre Phase II study, 115 patients were enrolled to determine the efficacy and safety profile of erlotinib as a single agent in advanced recurrent and/or metastatic HNSCC. Almost half of patients (46%) were treated with constant 150 mg/day and had a partial response rate of 4.3%. In addition, approximately one third of patients (38%) had disease stabilisation for a median duration of 16.1 weeks which was comparable with palliative chemotherapy [44]. In a Phase I/II study, the effect of erlotinib was assessed in combination with cisplatin to target patients with recurrent or metastatic HNSCC (NCT00030576). Fifty-one patients were treated with erlotinib 100 mg/day (orally) and cisplatin 75 mg/m2 (intravenously) every 21 days. The overall response rate was 21%, and disease stabilisation was observed for 49% of patients. Median PFS and median OS were 3.3 and 7.9 months, respectively. These results are comparable with a Phase III trial of cetuximab plus cisplatin [45,46].
In another Phase II trial, the effect of adding erlotinib to cisplatin and radiotherapy was investigated in 204 patients with locally advanced HNSCC. In this study, erlotinib (150 mg/day) did not elevate the toxicity of cisplatin and radiotherapy, however no significant increase was observed for complete response rate (CRR) or PFS [47]. A Phase II study of erlotinib as adjuvant treatment for locally advanced HNSCC (NCT01515137) significantly decreased proliferation in HNSCC, with additional effect from the nonsteroidal anti-inflammatory drug sulindac [48]. However, erlotinib therapy for patients with incurable cutaneous SCC in a single-arm Phase II clinical trial showed an overall response rate of 10% and a disease control rate (partial response + stable disease) of 72% [49]. In a recent study, HNSCC tumours collected from patients with MAPK1p.E322K mutation and treated with EGFR inhibitor, conferred heightened sensitivity to erlotinib in vivo and proceeding to patient recruitment in a clinical trial [43]. Its combination with other drugs such as docetaxel and cisplatin as an induction chemotherapy in loco-regional advanced HNC (Phase II, NCT00935961), or carboplatin and cetuximab in advanced HNC (Phase II, NCT01283334) [50] have been investigated.
In addition to gefitinib and erlotinib which are specific EGFR TKIs, lapatinib and vandetanib possess dual specificity for EGFR/HER2 and EGFR/VGFR2, respectively. Lapatinib (Tykerb) (GW572016) is FDA-approved for use in HER2-positive breast cancer in combination with capecitabine (Xeloda). In 2010, lapatinib received approval for the treatment with hormone receptor positive metastatic breast cancer in postmenopausal women with overexpression of HER2 receptor and for whom hormonal therapy was indicated (in combination with letrozole). In a multi-institutional Phase II study, 45 recurrent and/or metastatic HNSCC patients were recruited to determine efficacy and safety of lapatinib as a single agent (NCT00098631). Although lapatinib was well tolerated by HNSCC patients, it appeared to be inactive in EGFR-inhibitor naive or refractory subjects [51]. In a study of 44 patients with recurrent or metastatic HNSCC (NCT01044433), the combination of capecitabine and lapatinib as first-line therapy showed an overall response rate of 25% (90% CI, 15–38%) with a median OS of 10.7 months [52]. Addition of lapatinib to chemoradiotherapy and its use as long-term maintenance therapy (NCT00424255) in 688 patients (lapatinib, n = 346; placebo, n = 342) did not offer any efficacy benefits with a median follow-up time of 35.3 months, but had additional toxicity compared with placebo in patients with surgically treated high-risk HNSCC which resulted in the study being terminated early [53].
Vandetanib (ZD6474) is an orally bioavailable EGFR and VGFR-2 tyrosine kinase inhibitor. So far, it has been shown to effectively restore the sensitivity of HNSCC models to cisplatin and radiation in in vitro and in vivo studies [54]. It is important to note that trials for locally advanced HNSCC incorporating EGFR inhibition (cetuximab, erlotinib, panitumumab) with chemoradiation failed to show an advantage in PFS or OS over chemoradiation alone [55].

2.1.2. Anti-EGFR Monoclonal Antibodies

There are several anti-EGFR monoclonal antibodies developed for targeting human epithelial cancers of which at least five have been evaluated in clinical trials as potential EGFR-targeted therapy for HNSCC patients [36]. The initial concept for developing these therapies was that they would block receptor ligand interactions such as EGFR to EGF or TGF-α and consequently inhibit downstream signal transduction. However, the response to the antibody-EGFR interaction is complex in nature and is thought to act via different mechanisms. It has been shown that both monoclonal human immunoglobulin G1, IgG1 (cetuximab, matuzumab and nimatuzumab) and IgG2 antibodies (panitumumab) induce antibody-dependent cellular cytotoxicity (ADCC) [56,57]. Additionally, antibody structural differences and their EGFR epitope targets may affect the efficacy and toxicity of the drug [58].
Cetuximab is a chimeric human-murine IgG1 monoclonal antibody (mAb) (derivative of mAb225, C225) and a potent inhibitor of EGFR activation and blocks phosphorylation and activation of receptor-associated kinases and consequently inhibits cell cycle progression and proliferation, angiogenesis and vascular formation (decreased matrix metalloproteinase and vascular endothelial growth factor production) and metastasis [56,59]. EGFR is constitutively expressed in many normal epithelial tissues, including skin and hair follicles. Overexpression of EGFR is also detected in colon and rectum cancers.
Cetuximab (Erbitux®) was first approved by FDA in 2004 when it showed clinically significant activity when administrated alone or in combination with irinotecan (Camptosar) in patients with irinotecan-refractory colorectal cancer [60]. In 2006, it was FDA-approved for treatment of loco-regionally advanced HNSCC in combination with high dose radiotherapy (NCT00004227). The Phase III multicentre study was designed to evaluate the effect of radiotherapy with or without cetuximab (initial dose of 400 mg/m2 followed by 250 mg/m2 weekly) in treating 424 patients suffering from Stage III or IV oropharynx, hypopharynx, or larynx cancers [61]. Treatment with cetuximab plus radiation therapy (RT) (211 patients) increased the overall survival median duration to 49 months compared to 29.3 months for patients who were treated with radiotherapy alone (213 patients) [62]. Although this study showed a very positive outcome, treatment resistance and treatment failure must be viewed in the context of other mutations (such as p53) involved in HNSCC. One of the ways to overcome this issue might be adding agents to current known treatments. For example, cisplatin (a FDA-approved drug which induces apoptosis) can be employed in patients with mutated/damaged DNA [63].
In 2006, FDA also approved cetuximab for use in combination with platinum-based chemotherapy and 5-fluorouracil as first-line treatment of recurrent/metastatic conditions when it significantly increased the median overall survival from 7.4 months to 10.1 months, and as a single agent in recurrent/metastatic disease after failure of platinum-based chemotherapy when it increased the median progression-free survival time from 3.3 to 5.6 months as well as the response rate from 20% to 36% (p < 0.001) (EXTREME; NCT00122460) [64]. It was also approved by European Medicines Agency (EMA) for HNSCC and colorectal targeted therapy. Numerous Phase I, II and III studies are being performed on the effect of cetuximab as a single agent or with combination of radiotherapy and chemotherapy and other agents to treat HNSCC, but some side effects such as skin rash, drug resistance and infusional allergic reactions due to murine Ig component have been associated with this drug [46,62,65,66,67,68,69]. The effect of radiotherapy plus cetuximab or cisplatin was assessed in 849 HPV-positive oropharyngeal cancer patients in a Phase III randomised, multicentre, noninferiority trial (NCT01302834). 425 received radiotherapy plus cetuximab and 424 received radiotherapy plus cisplatin. Radiotherapy plus cetuximab showed inferior OS and PFS compared with radiotherapy plus cisplatin. Estimated 5-year overall survival was 77.9% in the cetuximab group versus 84.6% in the cisplatin group. Progression-free survival was significantly lower in the cetuximab group compared with the cisplatin group (5-year proportions 67.3% vs. 78.4%), and locoregional failure was significantly higher in the cetuximab group compared with the cisplatin group (5-year proportions 17.3% vs. 9.9%) [70]. Therefore, the use of cetuximab not only requires a comparison against other available conventional methods with or without alternative drugs, but also requires an understanding of each patient’s clinical status, history and risk profile [71].
Panitumumab (Vectibix®) is a fully humanised mAb that does not stimulate ADCC as strongly as cetuximab, and has been associated with minimal infusion-related reactions and may also reduce the production of neutralising antibodies [72]. Panitumumab was approved by FDA in 2006 for EGFR-expressing metastatic colorectal cancer (CRC) with disease progression on or following fluoropyrimidine-, oxaliplatin- and irinotecan containing chemotherapy regimens [73]. The approval was as a result of randomised Phase III multinational study with 463 patients suffering from metastatic colorectal cancer receiving either best supportive care (BSC) alone (chemotherapy regimens containing a fluoropyrimidine, irinotecan, and oxaliplatin) (232 patients) or BSC plus panitumumab, 6 mg/kg intravenously (231 patients). Median and mean PFS for patients receiving panitumumab and those receiving only BSC were 56 and 96.4 days, and 51 and 59.7 days, respectively [73].
Currently, panitumumab is in Phase III trials for HNSCC. A multinational Phase III study was performed to evaluate the efficacy and safety of panitumumab chemotherapy combined with cisplatin and 5-fluorouracil as first-line treatment for 658 patients with recurrent and/or metastatic HNSCC (NCT00460265) [74]. Both median OS (11.1 vs. 9 months) and median PFS (8.8 vs. 4.6 months) improved in those treated with additional panitumumab (327 patients) compared to the control group (a regimen of cisplatin and fluorouracil in 330 patients), but the improvements were not significant. In another Phase III trial evaluating the effect of combination of standard fractionation RT with cisplatin or panitumumab in 320 patients with locally advanced stage III and IV HNSCC (NCT00820248), panitumumab plus accelerated-fractionation RT was not superior to cisplatin plus standard-fractionation. The 2-year PFS was 73% in the cisplatin group and 76% in the panitumumab group, and 2-year overall survival was 85% and 88% in cisplatin and panitumumab groups, respectively [75].
Like cetuximab, many active Phase I and II trials are being performed to investigate the efficacy and safety of panitumumab as first or second line treatment for HNSCC despite development of some side effects including skin rashes which remains the main issue associated with EGFR-targeted antibodies and has even being considered as a marker for drug efficiency [29,76].
There are other promising mAbs being evaluated in Phase III trials for HNSCC including zalutumumab (fully humanised IgG1) and nimotuzumab (humanised murine IgG1) that have been tested as single agent or combinational therapy. Zalutumumab showed reasonable efficacy in HNSCC patients [77], extending progression-free survival in patients with recurrent HNSCC who had failed platinum-based chemotherapy (NCT00382031) but did not increase overall survival [78]. Nimotuzumab has shown less skin toxicity compared to cetuximab [79] and is currently in a Phase III trial recruiting patients aimed at improving loco-regional control rate and overall survival of locally advanced HNSCC compared to concurrent chemo-radiotherapy (NCT00957086).

2.2. PI3K/AKT/mTOR Pathway

Alterations of PI3K pathway have been associated with many solid tumours, therefore it encompasses valuable therapeutic targets. So far, a number of therapeutic agents have been developed which target key molecules within the pathway including PI3K, AKT and mTOR. Moreover, there are also other targeted therapies targeting other intermediate proteins within the pathway. Some are approved for use while others are potential targeted therapeutics under investigation.

2.2.1. PI3K Inhibitors

PI3K inhibitors in HNSCC are currently directly or indirectly (solid tumours) in clinical trial investigations. They can be divided into three major classes:

PI3K/mTOR Inhibitors

GDC-0980 is being investigated in Phase I and II trials for conditions including renal cell carcinoma (RCC) (NCT01442090), solid tumours (NCT01301716 and NCT01332604) and currently recruiting patients for prostate cancer (NCT01485861).
NVP-BEZ235 could reach Phase II clinical trials, however has been delayed due to cases being terminated (transitional cell carcinoma, NCT01856101 and metastatic, castration-resistant prostate cancer, NCT01717898) or withdrawn (advanced endometrial carcinoma, NCT01290406 and malignant PEComa, NCT01690871) in order to reformulate the drug.
A Phase II comparative study of NVP-BEZ235 and everolimus in patients with advanced pancreatic neuroendocrine tumours did not demonstrate increased efficacy compared with everolimus which might be due to a poorer tolerability profile. The median PFS was 8.2 months with BEZ235 versus 10.8 months with everolimus [80]. BGT226 has completed a Phase I/II study sponsored by Novartis for advanced solid malignancies including advanced breast cancer (NCT00600275).

Pan-Class I PI3K Inhibitors

Pan PI3K inhibitors include PX-866, BKM120 and SAR245408 (XL147).
PX-866 showed modest activity as a single agent therapy in patients with recurrent or metastatic prostate cancer [81]. It is mostly in active clinical Phase II trials in combination with drugs such as vemurafenib for advanced melanoma (NCT01616199), cetuximab for incurable progressive, recurrent or metastatic HNSCC (NCT01252628) and docetaxel for HNSCC (NCT01204099).
BKM120 is in Phase II trials as a single or combination therapy for many cancers including head and neck cancers. BKM120 monotherapy is being evaluated for advanced HNSCC (NCT01527877) and metastatic and recurrent or progressive HNC (NCT01737450). Trials assessing BKM120 in combination with cetuximab (NCT01816984) and cisplatin (NCT02113878) for recurrent or metastatic HNC are also active.
SAR245408 (XL147) has not been tested in HNC directly but has successfully completed Phase I and/or II trials in other conditions including breast cancer in combination with letrozole (NCT01082068), locally advanced or metastatic solid tumours in combination with oral MSC1936369B (NCT01357330), and advanced or recurrent endometrial cancer as monotherapy (NCT01013324).

PI3Kα (p110α) Inhibitors

The p110α protein (protein phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform) is a subunit of the PI3K protein that is encoded by PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha). PIK3CA is one of the most commonly seen mutations in HNSCC (10–12%) alongside with BRCA1 (6%), and BRCA2 (7–9%) [82]. A comprehensive genomic study of HNSCC in 2015 showed 25% of mutated PIK3CA displayed concurrent amplification, with an additional 20% of tumours containing focal amplification without evidence of mutation. Seventy-three per cent of PIK3CA mutations localized to Glu542Lys, Glu545Lys and His1047Arg/Leu hotspots [83].
Alpelisib (BYL719) is an α-specific PI3K inhibitor, and was approved by the FDA in 2019 to treat certain types of breast cancer. Its combination with paclitaxel showed a challenging safety profile in patients with advanced solid tumours [84]. Currently, a Phase I trial of a combination of BYL719 with cetuximab and intensity-modulated radiation therapy is active for stage III/IVB HNSCC (NCT02282371).

2.2.2. AKT Inhibitors

Perifosine is a lipid-based AKT inhibitor which interacts with pleckstrin homology domain of AKT and inhibits its binding to PIP3 [85]. It has been successfully tested in Phase I and II trial studies including recurrent/progressive malignant gliomas (Phase II, NCT00590954), recurrent pediatric solid tumours (Phase I, NCT00776867) and non-small -cell lung cancer (Phase II, NCT00399789). In HNSCC, it has shown antiproliferative activity in cell lines by blocking cell cycle progression at G1-S and G2-M in preclinical studies [86]. However, a Phase II clinical study on recurrent or metastatic HNC (NCT00062387) was terminated due to lack of antitumor activity as a single agent [87] and currently has no clinical trial for HNC.
MK2206 is another AKT inhibitor that has reached Phase II for progressive, recurrent, or metastatic adenoid cystic carcinoma of the oral cavity (NCT01604772), recurrent or metastatic HNC (NCT01349933) and recurrent nasopharyngeal carcinoma (NCT01370070). Other examples of AKT inhibitors in Phase I or II clinical studies on a variety of cancers are AZD5363, triciribine, AR-67 and GSK690693.

2.2.3. mTOR Inhibitors

Rapamycin (Sirolimus, Rapamune®) received FDA approval in 1999 as an immuno-suppressant drug for the prophylaxis of organ rejection in patients receiving renal transplants. It binds to mTORC1 and consequently blocks the phosphorylation of its downstream substrates, S6K1 and eukaryotic initiation factor-4E (eIF-4E)-binding protein (4E-BP1) [88]. Due to its lipophilic structure [89] different formulations of rapamycin have been tested to improve its poor water solubility and bioavailability for clinical application. So far, three analogues of rapamycin have been developed; everolimus (RAD001, Afinitor®), temsirolimus (CCI-779, Torisel®) and deforolimus (ridaforolimus, AP23573). Although modified formulations of these drugs enhanced their bioavailability and half-life, as well as reducing their immunosuppressive effects compared to rapamycin, all target mTOR and result in arresting cell cycle at the G1 phase [90,91].
Everolimus is FDA-approved for five cancerous conditions including advanced RCC (2009), subependymal giant cell astrocytoma associated with tuberous sclerosis (2010), progressive neuroendocrine tumours of pancreatic origin (2011), advanced hormone receptor-positive HER2-negative breast cancer (2012), paediatric and adult patients with subependymal giant cell astrocytoma (2012), and neuroendocrine tumours of gastrointestinal or lung origin with unresectable, locally advanced or metastatic disease (2016). A Phase II trial (NCT00942734) assessing the combination of erlotinib and everolimus did not show significant benefit in unselected patients with platinum-resistant metastatic HNSCC despite an acceptable toxicity profile. Twelve-week PFS was 49%, median PFS 11.9 weeks, and median OS 10.25 months [92].
Temsirolimus was approved by the FDA in 2007 for treatment of advanced RCC [93]. Currently, in HNC patients it has reached Phase II clinical trials as monotherapy (NCT01172769) and combinational therapy with cetuximab (NCT01256385) and paclitaxel and carboplatin (NCT01016769) in recurrent and/or metastatic HNSCC. However, its Phase II trial study for patients with platinum-refractory/ineligible, advanced HNSCC in combination with erlotinib was terminated (NCT01009203) due to abrogation of the mTOR pathway [94].
It is noteworthy that one of the drawbacks of using mTOR inhibitors might be PI3K activation as a result of negative feedback response between phosphorylation of insulin receptor substrate protein 1 (IRS1) by S6K1 and blockage of insulin like growth factor 1 (IGF-1) signalling to PI3K [95]. Therefore, the search for agents with dual PI3K/mTOR inhibition activity might be a better approach for PI3K-pathway-targeted therapy.

2.3. RAS/RAF/MEK/MAPK Pathway

Mutations in RAS/RAF/MEK/ERK (MAPK) pathway are common in all cancer types including head and neck, breast and prostate to name a few. Targeted therapy has gained remarkable attention and many key protein inhibitors have been introduced including RAF and MEK1 and MEK2 inhibitors. Although RAS inhibitors may be of a great value in RAS pathway suppression, clinical trials have not achieved satisfactory results as the inhibitors could not appropriately affect protein targets. However, in 2013 data were presented suggesting that it may be feasible to target mutant RAS proteins directly or target other unique features of RAS-driven tumours. Here we give some examples of inhibition of two important downstream proteins of RAS pathway, including MEK and RAF.

2.3.1. RAS Inhibitors

RAS (KRAS, NRAS and HRAS) is the most frequently mutated gene family in cancers. Up until 10 years ago, RAS was termed ’undruggable’; however, the success of allele-specific covalent inhibitors against the most frequently mutated version of RAS provided the opportunity to evaluate the best therapeutic strategies to treat RAS-driven [96]. Mutations in HRAS occur in 4–8% of patients with recurrent and/or metastatic HNSCC [83,97].
Tipifarnib (Zarnestra) is a potent and highly selective inhibitor of farnesyltransferase (FTase). FTase facilitates the attachment of farnesyl groups to signalling proteins that are required for cell membrane localization. All RAS isoforms are FTase substrates; however, only HRAS is exclusively dependent upon farnesylation and therefore can be targeted via tipifarnib-mediated inhibition of FTase [98]. Tipifarnib demonstrated promising efficacy in a phase II clinical trial in patients with recurrent and/or metastatic HNSCC with HRAS mutations for whom limited therapeutic options exist (NCT02383927). Objective response rate for evaluable patients with high-VAF HNSCC was 55%. Median PFS on tipifarnib was 5.6 months versus 3.6 months on last prior therapy and median OS was 15.4 months. The most frequent adverse events among the 30 patients with HNSCC were anaemia (37%) and lymphopenia (13%) [97]. Currently a Phase II clinical trial is recruiting patients to assess the safety and efficacy of tipifarnib in HNSCC with HRAS mutations and impact of HRAS on response to therapy (AIM-HN/SEQ-HN) (NCT03719690).

2.3.2. RAF Inhibitors

As mutations in CRAF (RAF1) are extremely rare, whereas BRAF mutations are profoundly oncogenic, BRAF protein kinases have been targeted by a number of BRAF inhibitors including sorafenib, vemurafenib and dabrafenib.
Sorafenib (tosylate salt of BAY 43-9006, Nexavar®) is a small molecule which inhibits RAF kinase and VEGF receptor kinase. It has been used for Ras gene mutated tumours and tumours overexpressing growth factor receptors in the Ras/Raf/Mek pathway, and inhibiting tumour angiogenesis or neovascularization via inhibition of VEGFR-2, VEGFR-3, and/or PDGFR-β including advanced clear cell renal carcinoma and hepatocellular carcinoma as well as hepatocellular carcinoma [99]. Sorafenib was approved by FDA for the treatment of patients with advanced RCC in 2005, for the treatment of unresectable hepatocellular carcinoma in 2007, and for the treatment of locally recurrent or metastatic progressive differentiated thyroid carcinoma (DTC) in 2013 (http://www.cancer.gov/cancertopics/druginfo/fda-sorafenib-tosylate). It is currently in Phase II clinical studies for head and neck cancers. In 2007, the efficacy and safety of sorafenib was evaluated in 27 patients with recurrent and/or metastatic HNSCC and nasopharyngeal carcinoma (NPC) in a Phase II trial [100]. Five patients showed evidence of disruption of the EGFR-Ras-Raf-MEK-ERK signalling pathway, a proapoptotic effect, and less convincingly, an effect on angiogenic pathways [100]. Although, sorafenib was well tolerated by patients, it had a modest anticancer activity compared to other monotherapies for HNSCC in Phase II clinical trials [44,100,101,102,103]. Williamson et al. conducted a Phase II trial in 2010 to evaluate the efficacy and safety of sorafenib as a single agent (400 mg twice daily in 28-day cycles) in 41 patients with metastatic or recurrent HNSCC (NCT00096512) and achieved similar results to Elser et al. [99]. It is currently in active Phase II study in combination with carboplatin and paclitaxel to treat patients with HNSCC (NCT00494182), and has been assessed in a Phase II study in combination with cetuximab for recurrent or metastatic HNSCC and other types of oral and nasal cavity (NCT00939627).
Vemurafenib (Zelboraf®) was approved by FDA in 2011 for the treatment of unresectable or metastatic melanoma with the BRAF V600E mutation (http://www.cancer.gov/cancertopics/druginfo/fda-vemurafenib). It is currently being tested as monotherapy for locally advanced thyroid cancer (Phase II, NCT01709292), melanoma (Phase II, NCT01813214) and relapsed or refractory hairy cell leukaemia (Phase II, NCT01711632). It is also being evaluated in combination with other drugs such as everolimus or temsirolimus for advanced cancer and solid tumours (Phase I, NCT01596140), bevacizumab for stage IV BRAFV600 mutant metastatic melanoma (Phase II, NCT01495988) and cetuximab for advanced solid cancers (Phase I, NCT01787500). A Phase I study of vemurafenib plus HL-085 in solid tumours with BRAF V600 mutation is currently recruiting patients (NCT03781219).
In 2013, dabrafenib (Tafinlar®) was approved as a single agent for treatment of unresectable or metastatic melanoma with BRAF V600E mutation. In 2014 it was approved for use in combination with trametinib (MEK1/2 inhibitor) which was also FDA-approved in 2013 to treat patients with unresectable or metastatic melanoma with a BRAF V600E/K mutation. The combination of dabrafenib/trametinib received FDA approval in 2018 as an adjuvant treatment for BRAF V600E-mutated, stage III melanoma after surgical resection. This was based on the results of the COMBI-AD Phase III study making it the first oral chemotherapy regimen to prevent cancer relapse for node positive, BRAF-mutated melanoma. Like vemurafenib, it is currently recruiting patients for Phase II studies including use of dabrafenib with stereotactic radiosurgery in BRAF V600E melanoma brain metastases (NCT01721603), with or without trametinib for recurrent thyroid cancer (NCT01723202) and with trametinib and navitoclax for solid tumours which are metastatic or cannot be removed by surgery (NCT01989585).

2.3.3. MEK Inhibitors

In contrast to RAF and MEK1/2 which have narrow substrate specificities, activated ERK1/2 catalyse the phosphorylation of multiple cytoplasmic and nuclear substrates, and regulate diverse cellular responses such as mitosis, embryogenesis, cell differentiation, motility, metabolism, angiogenesis, and programmed cell death [104]. MEK1/2 represents a bottleneck in the activation of diverse cellular responses, many of which are of significantly important in tumorigenesis [105,106].
CI-1040 was the first MEK inhibitor to reach Phase I trials in 2000 [107], and since then many of its analogues have reached clinical trial studies. To date, trametinib is the only FDA-approved MEK inhibitor targeted therapy as the other agents either showed only limited efficiency as a single agent or failed to show satisfactory results in the examined tumour types [105]. Some examples of new and emerging MEK inhibitors are pimasertib, refametinib, selumetinib and MEK162.
Despite ongoing studies of MEK inhibitors, only a small group of cancers can be targeted by these molecules, including HNSCC. This can be improved by detection of biomarkers contributing to therapeutic responsiveness. So far, only NRAS and BRAF V600 mutations have shown a consistent correlation with MEK inhibitors [105].
Trametinib (Mekinist®, GSK1120212) is an orally bioavailable, allosteric and selective inhibitor of MEK1/2 enzymes. The FDA approved trametinib for the treatment of patients with unresectable or metastatic melanoma with BRAF V600E/K mutations as a single agent in 2013 and in combination with dabrafenib (Tafinlar®) in 2014. In 2018, the combination of dabrafenib/trametinib was approved by the FDA as adjuvant treatment for BRAF V600E-mutated, stage III melanoma after surgical resection [108].
Trametinib (Mekinist®) is indicated as monotherapy or combinational therapy for the treatment of patients with unresectable or metastatic melanoma with a BRAF V600 mutation.
Trametinib plus dabrafenib significantly prolonged PFS and OS, improved objective response rates and preserved health-related quality of life to a greater extent than dabrafenib (in the double-blind COMBI-d study) and vemurafenib (in the open-label COMBI-v study) in two large, randomized, Phase III studies in treatment-naïve patients with unresectable or metastatic melanoma with BRAF (V600E/K) mutation [109]. It is currently recruiting patients for Phase I, II and III trials as mono or combinational therapy.
Selumetinib (AZD6244, ARRY-142886) is a new potent MEK1/2 inhibitor and, like other MEK inhibitors, acts via the ATP noncompetitive mechanism and therefore has no significant inhibitory effect on other serine/threonine kinases [110]. In 2020, selumetinib received FDA approval for the treatment of children with neurofibromatosis type 1 (NF1) based on a clinical trial (NCT01362803). Selumetinib is now being tested for different cancer types including thyroid cancer [111] (NCT01843062), ovarian cancer, peritoneal cancer (NCT00551070), and advanced solid malignancies with cixutumumab combination (NCT01061749).
There are other MEK inhibitors which inhibit solid tumours such as cobimetinib in combination with pictilisib (PI3K inhibitor) in locally advanced or metastatic solid tumours (Phase I, NCT00996892), pimasertib and SAR245409 (PI3K/mTOR inhibitor) for locally advanced or metastatic solid tumours (Phase I, NCT01390818), and MEK162 and RAF265 (RAF inhibitor) in advanced solid tumours harbouring RAS or BRAF V600E mutations (Phase I, NCT01352273).

2.4. NOTCH Pathway

As the NOTCH pathway has dual oncogenic and tumour suppressor actions, therapeutically targeting this pathway is associated with some concern. To date, a variety of γ-secretase inhibitors (GSI) are being evaluated in preclinical and clinical trials to target NOTCH1 by inhibiting NICD cleavage and consequently its translocation into the nucleus [112,113]. Although there are no clinical trials currently being performed in HNSCC, there are studies targeting different tumour types including melanoma [114] and solid tumours [115]. Insights gleaned from these may inform therapeutic approaches in HNC, given the significance of the NOTCH pathway in HNSCC.

2.5. MET Pathway

MET regulation can be affected by overexpression, constitutive kinase activation, gene amplification, mutation or paracrine/autocrine activity via hepatocyte growth factor (HGF) [116,117]. The expression of MET can be targeted at RNA level by a novel approach of using small RNA molecules including small interfering RNA (siRNA) and micro RNA (miRNA) [118]. HGF competitors and antibodies are other therapeutic alternatives. NK4 is a member of N-terminal hairpin domain and Kringle domain (NK) inhibitor family which acts as a competitive antagonist for HGF. In vitro analysis has shown that NK4 inhibits angiogenesis induced by basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) as well as HGF [119]. AMG102 (rilotumumab) is a fully humanised monoclonal anti-HGF IgG which binds to β-chain of HGF and prevents its binding to MET. After positive results in preclinical and Phase I and II clinical studies on solid and lung tumours, it is currently recruiting patients for a Phase III trial for lung-MAP: S1400 biomarker-targeted second-line therapy in treating patients with recurrent stage IIIB-IV squamous cell lung cancer (NCT02154490).
Various MET inhibitors which act as kinase inhibitor at MET ATP binding sites, are currently under preclinical and clinical investigation. Tivantinib (ARQ197) is a MET kinase inhibitor which is being evaluated in different phases of clinical trials including investigating the effects of tivantinib plus erlotinib versus single agent chemotherapy in locally advanced or metastatic NSCLC (Phase II, NCT01395758), tivantinib in treating younger patients with relapsed or refractory solid tumours (Phase I, NCT01725191) and tivantinib in combination with chemotherapy in metastatic colorectal cancer (Phase I/II, NCT01075048). Patients are also being recruited for a Phase II randomized study to investigate the effects of cetuximab with or without tivantinib in HNC which is recurrent, metastatic, or cannot be removed by surgery (NCT01696955).
Ficlatuzumab is a humanised monoclonal antibody currently in a Phase II trial (NCT03422536), having been assessed in a Phase I trial assessing a combinational therapy with cetuximab in patients for recurrent/metastatic HNSCC which was completed in 2019 (NCT02277197). Results are not currently available for this study.
Foretinib is a c-Met receptor and vascular endothelial growth factor receptor 2 (VEGFR-2) inhibitor. A Phase II trial of single-agent foretinib (GSK1363089) in patients with recurrent or metastatic HNSCC (NCT00725764) was well tolerated and showed prolonged disease stabilisation [120].
Some other MET inhibitors that are currently in clinical investigation are crizotinib, exelixis (XL184) and MGCD265. The Phase I clinical trial of SGX523 to treat solid tumours (NCT00606879) however was terminated due to unexpected renal toxicity [121]. Moreover, there are other characterised small molecules including SU11274 [122] and PHA665752 [123] which so far have shown promising results in in vitro and in vivo studies.

2.6. JAK/STAT Pathway

The role of Janus kinase (JAK) and STAT pathway has been extensively studied in cancers including breast, lung, and head and neck (HNSCC). Activation of JAK leads to phosphorylation of STAT proteins, resulting in dimerization and translocation into the nucleus where they act as transcription factors with pleiotropic downstream effects. Activation of STAT3 and STAT5 results in elevated cell proliferation and survival, angiogenesis, and immune evasion [124].
Many molecules affecting STAT3, STAT5 and JAKs, particularly JAK2, are in preclinical and clinical studies. For example, it has been shown that combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition could reduce survival of HNC cell lines [125]. Therapies which inhibit STAT (mostly STAT3) are being researched in vitro and in vivo, however not many of these agents have reached clinical development. A novel STAT inhibitor, OPB-31121, strongly inhibited STAT3 and STAT5 phosphorylation and showed a significant antitumor effect on leukaemia with STAT-addictive oncokinases [126,127].
Oligonucleotide inhibitors of STAT3 such as siRNA and antisense RNA oligonucleotides have been investigated in order to inhibit transcriptional machinery of genes affected by JAK/STAT pathway. An example of this is use of a STAT3 decoy oligonucleotide with overall antitumor effects including inhibition of cyclin D1 and Bcl-xL transcription and angiogenesis in HNSCC both in in vitro and in vivo studies [128,129,130].
AZD9150, a synthetic antisense oligonucleotide molecule targeting STAT3 by inhibition of mRNA translation, has demonstrated antitumor activity in xenograft models. It is currently being studied in clinical trials of metastatic HNSCC as a monotherapy or combined with MED14736; an immunotherapy which blocks the interaction between PD-1 and PD-L1 (NCT02499328) [131].
Various in vitro and in vivo studies are being performed on JAK inhibitors (mostly JAK2) and many of these have reached preclinical and clinical studies. Currently, most in clinical development are orally available small molecule kinase inhibitors which act in an ATP-competitive manner [124,131,132]. Ruxolitinib (INCB018424) is an approved JAK (Jak2) inhibitor for myelofibrosis [133], with a study currently recruiting patients with HNSCC (NCT03153982). Fedratinib (SAR302503, TG101348) is a selective Jak2 inhibitor which has been studied in myelofibrosis and was approved by the FDA in 2019. In addition to myelofibrosis the effects of SAR302503 have also been investigated in solid tumours (Phase I, NCT01836705) and (Phase I, NCT01585623). Other JAK inhibitors in clinical studies are lestaurtinib (CEP-701) and XL019, while other agents are in early stages of clinical studies and development including CYT387, SB1518 and AZD1480 [124,134].

2.7. TP53/RB

Replacing a mutated TP53 gene with a wild-type gene in order to restore p53 activity has been studied for decades and now is a potential approach for HNC treatment. Adenovirus vectors expressing human full length wild-type p53 have been introduced, after initial efforts in NSCLC using a retroviral vector expressing human p53 under control of an actin promoter [135]. Unlike retroviruses that are potentially oncogenic, adenovirus does not integrate the vector DNA into host cell DNA. However, limited ability to infect all tumour cells as well as induction of the host immune system after injection and the need for repetition of the therapy are disadvantages associated with this approach [136]. Gendicine (recombinant human p53 adenovirus [Ad5RSV-p53]), was approved by the China Food and Drug Administration (CFDA) in 2003 for the treatment of HNSCC [137,138,139]. Wild-type p53 gene significantly enhanced radiotherapeutic effectiveness in patients with HNSCC [138]. Infusion of rAd-p53 and chemotherapy significantly increased survival rate of patients with stage III but not stage IV oral SCC, compared with intra-arterial chemotherapy alone (ChiCTR-TRC-09000392) [140].
A Phase III clinical trial for use of Advexin (adenovirus containing TP53) (INGN 201, Ad5CMV-p53) versus methotrexate in treating advanced recurrent HNSCC showed that patients with wild-type p53 had better response to Advexin, whereas patients carrying mutant p53 were more responsive to methotrexate chemotherapy which indicates the potential of p53 as a biomarker to design a targeted therapy [141]. This approach has subsequently been used in a Phase II trial (NCT03544723) investigating safety and efficacy of p53 gene therapy combined with immune checkpoint inhibitors in solid tumours including HNSCC [142]. Previous Ad-p53 clinical trials appear to have been negatively impacted by the inclusion of patients with unfavourable p53 biomarker profiles and by under dosing of Ad-p53 treatment [142]. This has important implications for Ad-p53 gene therapy and future p53-targeted therapy. Although Advexin has not been approved by the FDA [143], the use of adenovirus gene therapy for treatment of HNC was approved in 2003, and Gendicine has been commercially marketed since then and used as combinational therapy mostly with radiotherapy [136,139,144].
ONYX-015 is an adenovirus with deletion of its E1B region which inactivates p53 and therefore replicates in and lyses p53-deficient cancer cells [145]. A Phase II trial of a combination of intratumoural ONYX-015 injection with cisplatin and 5-fluorouracil in patients with recurrent HNSCC (NCT00006106) was withdrawn as it showed initial objective responses, however had no more progress after 6 months, whereas all noninjected tumours treated with chemotherapy alone had progressed [146]. Other approaches including p53-reactivating small molecules [147] targeting CDKN2A to reactivate p16/INK4A and CDK inhibitors are under investigation [38,148].
Head and neck cancers commonly display reduced MHC expression, hence MHC class I gene therapy may be a potential therapy to induce an antitumor response either by presenting tumour antigens or on its own as an antigen. Allovectin-7® is a gene therapy product that utilises a liposomal vector and encodes the class I MHC HLA-B7. A Phase II multi-institutional study of Allovectin-7 for HNC including SCC of the oral cavity or oropharynx showed that 10% of 60 HLA-B7-negative patients achieved partial response, and 23 patients had stable disease after one cycle of treatment. Responses persisted for 21 to 106 weeks [149].

2.8. HPV-Mediated Pathogenesis

Human papillomavirus (HPV) infection plays a critical role in a subset of HNSCC including the oropharynx (OPSCCs). HPV genome integration in the host genome not only results in amplification of oncogenes and disruption of tumour suppressor genes, but also derives inter- and intrachromosomal rearrangements [150]. A comprehensive study showed that HPV contributes to 22% of OPSCCs and 47% of tonsillar SCCs [151,152]. Therefore, understanding the prevention and treatment strategies that reflect the mechanism of HPV infection has become a matter of importance. Currently, two types of HPV vaccines are available including preventive vaccines which are based on HPV virus-like particles, and therapeutic vaccines which are being developed to eliminate established papillomavirus infection [153]. These vaccines were initially developed for anogenital malignancy, including cervical cancer and may be used for HPV associated head and neck SCC [154,155]. Prophylactic vaccines (preventive) were developed using HPV virus-like particles (VLP) to generate neutralizing antibodies against major capsid protein, L1 and L2. In 2006, the FDA approved the quadrivalent HPV vaccine Gardasil®/Silgard® (Merck) for the preventive control of HPV infections against four high risk HPV serotypes 6, 11, 16 and 18.
Cervarix® (GlaxoSmithKline) is a human papillomavirus bivalent vaccine which was approved by the FDA in 2009 and generates cellular immunity against HPV16/18 L1 protein [156]. It has also shown partial cross-protection against HPV types 31 and 45 as well [157,158]. Although, Gardasil and Cervarix have demonstrated excellent safety and efficacy profiles (up to 80% of all cervical cancers) [159,160], prophylactic HPV vaccines do not have therapeutic effects against existing HPV infections and HPV-associated lesions [160]. In addition to prevention, targeting HPV E6/E7 and induction of cell-mediated immunity response using live viral or bacterial peptide and nucleic acid vaccines may find a use in HPV associated HNSCC. Therapeutic vaccines targeting either HPV16 E6 or E7 have shown positive results in cervical cancer [161,162,163]. Simultaneous utilisation of HPV 16 genes, L1, E6, and E7 in combination therapy with archaeosomes can be an excellent strategy to stimulate immune responses against existing HPV16-associated malignancies, holding great promise for therapeutic vaccine development [164].
A Phase I clinical trial of E6 or E7 vaccine therapy in treating advanced or recurrent cancers including HNC has recently been completed (NCT00019110). A Phase I safety study of HPV E7 DNA vaccine to treat HNC patients is currently recruiting patients (NCT01493154). A Phase II trial of HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy for advanced or recurrent HPV16-induced gynaecological carcinoma, was well tolerated by patients and induced a broad IFN-γ-associated T-cell response. It was subsequently suggested to use this vaccine in combination with chemotherapy and immunomodulation [165]. Further studies for developing therapeutic vaccines to treat HPV16-induced malignancies are in progress [166,167].
ADX11-001 (Lovaxin-C) is a recombinant live-attenuated Listeria monocytogenes (Lm) that secretes the antigen HPV16 E7 fused to a nonhemolytic listeriolysin O protein. In a Phase I study, ADX11-001 was administered to 15 women with advanced cervical cancer and only flulike symptoms including fever and hypotension were observed [168]. A Phase II clinical trial of ADX11-001 for treatment of cervical cancer met the protocol specified benchmark for activity required and will be investigated further [169].

2.9. Cell Cycle Pathway

Targeting abrogated cell cycle control is a promising approach for cancer therapy particularly for HNSCC [170,171].
Carboplatin (Paraplatin) and cisplatin are the primary platinum agents used in the treatment of HNC. They interact with DNA nucleophilic groups such as GC-rich sites and form intra- and interstrand DNA and DNA-protein cross-links. These bifunctional covalent links interfere with DNA synthesis in the S phase of the cell cycle and confer tumouricidal effects [172]. Both cisplatin and carboplatin based regimens show 10% to 20% overall survival benefit about 3 years [173]. However, carboplatin shows more stability and less toxicity compared to cisplatin and consequently, carboplatin-based therapy is frequently administered to patients who are unable to tolerate cisplatin [173,174].
A phase III trial of 442 patients showed that combination of platinum-based chemotherapy (carboplatin/cisplatin) with cetuximab improved overall survival compared to fluorouracil combination in recurrent or metastatic HNSCC [64,175]. Additionally, in a multicentre, noninterventional trial, patients treated for recurrent or metastatic HNSCC either with platinum-based chemotherapy and cetuximab or radiotherapy and cetuximab showed lower tumour burden in responders [176]. Recently a Phase III trial to investigate the efficacy of adding carboplatin/5FU to Erbitux-radiotherapy in patients with locally advanced HNC was completed (NCT00609284), while more recently, a phase II trial evaluating the efficacy of carboplatin in combination with palbociclib for the treatment of unresectable recurrent or metastatic HNSCC was completed (NCT03194373). Currently, multiple trials are recruiting patients to perform comparative studies combining immunotherapy drugs (e.g., pembrolizumab), 5FU and radiation in HNSCC (NCT04428333, NCT03070366, NCT04671667).
Paclitaxel is one of a new class of agents known as taxanes, a natural product from the bark of the western yew tree. Paclitaxel stabilises microtubule polymerisation and arrests the cell cycle in mitosis. The prolonged activation of the mitotic checkpoint triggers apoptosis or reversion to the G0-phase of the cell cycle without cell division [177].
Phase II studies evaluating the efficacy and safety of weekly paclitaxel treatment in patients with recurrent or metastatic HNSCC showed promising activity with acceptable toxicity. The overall response rate was 29% according to RECIST (Response Evaluation Criteria in Solid Tumours). The median duration of response, median time to progression, and median survival time were 7.4, 3.4, and 14.3 months, respectively [178]. Currently multiple phase II-IIII trials are recruiting HNSCC patients for combination of paclitaxel with other drugs including camrelizumab (an anti-PD-1 immune checkpoint inhibitor) and cisplatin (Phase II, NCT04826679), pembrolizumab plus carboplatin (phase IIII, NCT04489888) and bleomycin and cisplatin or carboplatin (phase II, NCT03830385).

2.10. DNA Repair Pathway

DNA damage response pathways are activated following endogenous and exogenous damage of DNA [179]. Poly (ADP-ribose) polymerase 1 (PARP1) is a member of poly (ADP) ribose moiety enzyme superfamily which repairs single-strand and double-strand breaks [180]. Tumour cells use PARP to repair platinum-induced DNA damage and thus escape apoptosis. The activity of PARP inhibitors is based on the model of synthetic lethality, where an underlying homologous recombination repair deficiency (HRD) in tumour cells makes cells highly susceptible to PARP inhibition. The antitumor effects of PARP inhibitors are not dependent on a direct interaction with a mutated gene/protein, but instead on an underlying defect in the DNA damage repair mechanism of cancer cells themselves. Therefore, combination of platinum drugs and PARP inhibitors may block cancer cells from repairing damaged DNA ultimately leading to cellular apoptosis [181].
Olaparib (Lynparza) is a PARP inhibitor that acts against BRCA1 or BRCA2 mutations [182]. Olaparib received European Medicines Agency (EMA) and Food and Drug Administration (FDA) approval in 2014 for treatment of germline BRCA mutated (gBRCAm) advanced ovarian cancer with three or more prior lines of chemotherapy, gBRCAm metastatic breast cancer in 2018 and gBRCAm metastatic pancreatic adenocarcinoma in 2019. Olaparib in combination with temozolomide demonstrated substantial clinical activity in relapsed small-cell lung cancer [183].
Moreover, increasing evidence from trials in solid tumours suggests that DNA damage repair (DDR) alterations may predict response to immunotherapy [184]. Recently Psyrri et al., showed that changes in DDR signals are implicated in the response to HNSCC chemotherapy and can be exploited as novel therapeutic targets and sensitive/effective non-invasive biomarkers [179]. A Phase I trial of olaparib at 25 mg orally twice daily with concurrent cetuximab and radiation for heavy smoker patients with locally advanced HNC was well tolerated with reduced dermatitis within the radiation field. Two-year OS, PFS, local control, and distant control rates were 72%, 63%, 72%, and 79%, respectively [185]. Currently the combination of olaparib with cisplatin and durvalumab (Phase II, NCT02882308), or with pembrolizumab and carboplatin (Phase II, NCT04643379) are under investigation in patients with recurrent or metastatic HNSCC.
Other molecular targets including the Src family kinase (Dasatinib, SRC inhibitor, NCT00882583) and Cyclin Dependent Kinase complex (CDK) (pablociclib, selective CDK 4/6 Inhibitors, NCT03024489) are currently being investigated in combination with cetuximab and radiotherapy in locally advanced HNC patients.

2.11. Hypoxia and Angiogenesis

Cells under hypoxic conditions are known to be resistant to most anticancer drugs for several reasons [186] including their distant location from blood vessels which also result in decreased cell proliferation [187,188], increased survival of cells with loss of p53-mediated apoptosis sensitivity [189], and upregulation of genes involved in drug resistance such as genes encoding P-glycoprotein (a cell membrane protein that actively pumps many drugs out of the cell) [190,191,192]. Additionally, oxygen is required for effective action of some anticancer agents such as bleomycin [193], as well as tumour responsiveness to radiotherapy by stabilising free radicals produced by ionising radiation that causes DNA damage and cell death [194].
Radiosensitisation using hypoxic modifiers such as nitroimidazoles (5-nitroimidazole) have reached Phase I and II clinical evaluations. Among more than 7000 patients who have been studied in 50 randomized trials, treatment mostly benefitted head and neck lesions and to a lesser extent bladder tumours [195]. Radiopharmaceuticals including [(18)F]EF3 which is a labelled 2-nitroimidazole hypoxia marker [196], fluorodeoxyglucose ([18F]FDG, 18F-FDG or FDG) [197], fluoromisonidazole (F-FMISO) and F-fluoroazomycin-arabinoside (F-FAZA) [198] have been used for visualising hypoxia using positron emission tomography (PET) in head and neck tumours. The effect of other agents to improve HNC oxygenation have been studied in clinical trials including hyperbaric oxygen (HBO) (100% oxygen) [199,200,201,202,203,204], carbogen (95% oxygen plus 5% carbon dioxide) and nicotinamide [205,206], and hypoxic cytotoxin tirapazamine [207,208]. In addition to many studies that have been conducted to identify hypoxia gene expression signatures in HNC, reviewed by Toustrup et al. [209], they developed a 15-gene hypoxia gene expression classifier with prognostic and predictive impact for hypoxia-modifying therapy with nimorazole in conjunction with radiotherapy [210]. Furthermore, a 26-gene hypoxia signature showed benefit from hypoxia-modifying treatment including carbogen and nicotinamide in combination with accelerated radiotherapy in laryngeal cancer and is to be evaluated in a clinical trial [211].
To date, there are two main approaches available for targeting angiogenesis including targeting VEGF ligand and inhibition of VEGF receptor tyrosine kinase [212]. Bevacizumab (Avastin®) is a fully humanised IgG1 mAb that binds to all five forms of VEGF and prevents the interaction of VEGF to its receptors on endothelial cells surface. It has been FDA-approved for a variety of cancers including metastatic colorectal cancer (first line therapy, 2004/second line therapy, 2006), advanced nonsquamous non-small-cell lung cancer in combination with carboplatin/paclitaxel chemotherapy (first line therapy, 2006), glioblastoma (second line therapy, 2009), metastatic RCC (2009), metastatic HER2-negative breast cancer (2011), and metastatic colorectal cancer in combination with fluoropyrimidine-based chemotherapy (2013). A Phase III clinical trial for adding bevacizumab to chemotherapy for treatment of patients with recurrent or metastatic HNC (NCT00588770) did not improve OS but improved the response rate and PFS with increased toxicities. Median OS was 12.6 months with bevacizumab plus chemotherapy (BC) and 11.0 months with chemotherapy alone. At 2, 3, and 4 years, the OS rates were 25.2% v 18.1%, 16.4% v 10.0%, and 11.8% v 6.4% for BC versus chemotherapy, respectively [213].
Bevacizumab has reached Phase II trials in combination with other therapies including erlotinib [214], pemetrexed [215], cetuximab [216] and cisplatin and intensity-modulated radiation therapy for Stage III or Stage IV HNC (NCT00423930) [217]. Although, it has been proven to be effective, it has been associated with serious bleeding as a side effect [218].
Small molecules that inhibit VGFR are mostly multikinase inhibitors, tyrosine kinase inhibitors (TKI), including sorafenib, vatalanib, and sunitinib. Sorafenib is a Raf inhibitor which also inhibits VEGFR2, VGFR3 and PDGFR [219]. It has been used as a single agent [99] and is in active clinical trials in combination with other therapies such as carboplatin and paclitaxel (Phase II, NCT00494182) [220], cetuximab (Phase II, NCT00939627) [221] and cisplatin and docetaxel (Phase I/II, NCT02035527) [222] in HNSCC patients.
Sunitinib malate (Sutent®) is also a multikinase inhibitor which inhibits VGFR1, VGFR2, VGFR3, PDGFR, RET and c-KIT receptors [223]. It is an approved FDA agent for conditions including gastrointestinal stromal tumour (2006), advanced (metastatic) RCC (2006) and pancreatic neuroendocrine tumours (2011). Although, it has been well tolerated as a single agent therapy in HNSCC patients [224], it has been associated with significant bleeding side effect [225]. Currently, a Phase IB trial of sunitinib in combination with radiation therapy for HNC was recently completed (NCT00437372).
Vandetanib is a dual tyrosine kinase inhibitor that inhibits both EGFR and VEGFR simultaneously [226]. It was approved by FDA for the treatment of symptomatic or progressive medullary thyroid cancer in patients with unresectable, locally advanced, or metastatic disease in 2011. Sano et al. showed that adding vandetanib to cisplatin, and radiation could overcome cisplatin and radioresistance in in vitro and in vivo HNSCC models [54]. Vandetanib in combination with cisplatin and radiation (Phase II, NCT00720083) was withdrawn, while a Phase II clinical trial on preventing cancer in patients with precancerous head and neck lesions was recently completed (NCT01414426).
Vatalanib (PTK787/ZK-222584) is a small molecule that inhibits VGFR, EGFR, PDGFR, RET and c-KIT receptors and is under investigation for the treatment of solid tumours [227]. Vatalanib has successfully passed Phase II clinical trial as an oral angiogenesis inhibitor in patients with stage IIIB/IV NSCLC [228]. In vitro studies using HNC tumour cell lines showed that vatalanib could block VEGF downstream targets including Bcl-2 and CXCL8 expression in endothelial cells [229]. The Phase I study of vatalanib in combination with everolimus for advanced solid tumours (NCT00655655) was recently completed.
A Phase II trial of axitinib demonstrated a low objective response rate but favourable disease control rate of 77% and median OS of 10.9 months with an acceptable toxicity profile [230].
Other VEGFR inhibitors in clinical trials in HNC include cediranib (AZD-2171, recentin) (Phase II, NCT00458978) and pazopanib as monotherapy (Phase II, NCT01377298) and in combination with cetuximab (Phase I, NCT01716416), and nilotinib (Phase I, NCT01871311).

2.12. Host Immunity

Cancer immunotherapy utilizes the immune system to attack tumours. Immune checkpoint inhibitors provide a natural brake on the immune system so that immune cells (T cells) can recognize and target cancer cells. Head and neck cancers are immunosuppressive and therefore likely to respond well to immunotherapy [231]. The programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) and CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) are critical targets for immune checkpoint therapies.
Antibodies targeting the checkpoint molecules including PD-1, PD-L1 and CTLA-4 have resulted in durable responses and improved survival in patients with advanced cancer. Ipilimumab (Yervoy), an anti-CTLA-4 monoclonal antibody (human IgG1 k), targets CD28-CD80/CD86 co-stimulatory pathways and is the only CTLA-4 inhibitor to receive FDA approval in 2011 [232,233]. Tremelimumab, a human IgG2 anti-CTLA-4 antibody, has undergone various clinical trials for HNSCC (NCT02369874, NCT02551159, NCT02319044) [234,235,236].
In addition to ipilimumab, there are six approved monoclonal antibodies targeting PD-1 and PD-L1, namely, nivolumab (Opdivo), pembrolizumab (Keytruda), avelumab (Bavencio), atezolizumab (Tecentriq) durvalumab (Imfinzi) and cemiplimab (Libtayo). The first two are anti-PD-1 antibodies, and the latter four are anti-PD-L1 antibodies (Table 2). Nivolumab and pembrolizumab have been approved by the FDA for treatment of patients with HNSCC with relapse or metastasis who demonstrate cisplatin resistance [237].
Nivolumab, an anti-PD-1 monoclonal antibody, was the first immunotherapy approved by the FDA for the treatment of HNSCC following a Phase III clinical trial (Checkmate 141, NCT02105636) in 2016 [238]. Nivolumab treatment of recurrent and metastatic HNSCC has increased the overall survival time and disease-free survival regardless of the expression levels of PD-L1 or the p16 status of the cancer. Median OS was 7.5 months in the nivolumab group versus 5.1 months in the group that received standard therapy. OS was significantly longer with nivolumab than with standard therapy and the estimates of the 1-year survival rate were approximately 19 percentage points higher with nivolumab than with standard therapy (36.0% vs. 16.6%). Median PFS was 2.0 months with nivolumab versus 2.3 months with standard therapy. The rate of PFS at 6 months was 19.7% with nivolumab versus 9.9% with standard therapy. The response rate was 13.3% in the nivolumab group versus 5.8% in the standard-therapy group [238,239]. The combination of nivolumab and ipilimumab has shown effective results in melanoma [240,241] and RCC [242]. The combination is under investigation in other solid tumours including HNSCC.
Pembrolizumab, a humanised IgG4-κ monoclonal antibody targeting PD-1, was first approved by the FDA in 2017 for any unresectable or metastatic solid tumour with certain genetic anomalies such as mismatch repair deficiency or microsatellite instability [243]. A Phase III study of pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic HNSCC showed that pembrolizumab plus platinum and 5-fluorouracil is an appropriate first-line treatment for recurrent or metastatic HNSCC, and pembrolizumab monotherapy is an appropriate first-line treatment for PD-L1-positive recurrent or metastatic HNSCC [55]. Pembrolizumab showed significant treatment results in the treatment of relapsed or metastatic HNSCC in the Phase III clinical study (KEYNOTE 048; NCT02358031) in 2019. The combination of pembrolizumab and chemotherapy improved the OS to 13.0 months from 10.7 months for the combination of cetuximab and chemotherapy. Based on the observed efficacy and safety results, pembrolizumab and chemotherapy are now first-line treatment for patients with recurrent or metastatic HNSCC, whereas pembrolizumab monotherapy is the first-line treatment for patients with relapsed or metastatic PD-L1-positive HNSCC [55]. Other anti-PD-1 Phase II/III trials currently underway including NCT03813836 (pembrolizumab in recurrent/metastatic HNSCC with WHO performance status 2), NCT02521870 (pembrolizumab plus intratumoral SD-101 in anti-PD-1/PD-L1 treatment-naïve recurrent/metastatic HNSCC), NCT02741570 (nivolumab plus ipilimumab vs. the EXTREME regimen as first-line treatment in recurrent/metastatic HNSCC), NCT03040999 (pembrolizumab plus cisplatin plus RT vs. cisplatin plus RT alone in locally advanced HNC), and NCT02707588 (pembrolizumab plus RT vs. cetuximab plus RT in locally advanced HNC).
A Phase I immuno-radiotherapy with cetuximab and avelumab (Bavencio), a PD-L1 inhibitor, showed that cetuximab-RT plus avelumab is feasible in patients with advanced-stage HNSCC who are not good candidates for cisplatin treatment. Tumour recurrence was 50% after a median of 12 (8–26) months follow-up [244]. Patients are currently being recruited in a Phase I clinical trial to assess the feasibility of intratumoural administration of ipilimumab monotherapy prior to surgical resection, and to assess the immune system response to treatment (NCT02812524). Currently there are multiple Phase I/II clinical trials recruiting patients for combinational therapies, including Phase I/II trial of durvalumab, tremelimumab and radiation therapy in recurrent and metastatic HNSCC (NCT03522584), Phase II of nivolumab and ipilimumab for recurrent and metastatic HNSCC (NCT03620123), and Phase I of Interleukin-15 Superagonist (N-803) and ipilimumab in patients with advanced head and neck cancer (NCT04290546). This is in addition to a Phase III trial assessing atezolizumab after definitive local therapy in high-risk locally advanced HNC (NCT03452137).

3. Combination Therapies

It is clear from this paper and from many others detailing currently useful and agreed protocols for managing HNSCC, that a combination of treatment modalities and a combination of pharmacotherapeutics are required to manage and treat HNSCC effectively. Assessment of clinical trials registered with ClinicalTrials.gov shows that about 30% are assessing chemotherapeutic approaches, 15% immunotherapies, 15% targeted therapies, 10% radiotherapies, and 5% surgical approaches, with about another 15% assessing combination therapies. These combination therapies cross the traditional radiation oncology/medical oncology/surgery divides, and are aimed at maximizing the benefits of one treatment modality and limiting the adverse effects of another. These apply equally to radiation-based combinations, neoadjuvant chemotherapy, and adjuvant therapy, with a significant current focus on recurrent and metastatic disease. This is seen in routine oncological practice worldwide. Moving forward however, combination molecular therapies will become more common, as our understanding of the molecular and signalling pathways of HNSCC as a heterogeneous group of diseases increases. These can be applied to the same clinical settings and scenarios, but potentially as first line therapies.
Current treatment approaches are still very toxic, highlighting the need for more targeted approaches, stratification of tumour types and substratification of applicable and druggable biomarkers to reduce toxicity and improve outcomes. Further understanding of the tumour microenvironment and tumour immunology would allow better use of immunotherapies tailored appropriately to the patient and their tumour. Further exploration of the role of HPV and the oral microbiome in initiation of HNSCC is equally important as that may dictate combinations of molecular therapies with greater utility, depending on etiology, keratinocyte differentiation, pathogenesis and potential response to therapy. This may underpin the rationale for combination therapies where the input of the immune system and the influence of the microbiome are taken into account with the choice of PD-1/PD-L1 immunotherapies coupled with PI3K/AKT/mTOR inhibitors. An equally attractive approach is based on the activity of PARP inhibitors and radiation therapy, or PARP inhibitors and immunotherapies. Equally, combination approaches in HPV-negative tumours could make simultaneous use of EGFR inhibitors (Cetuximab), MET inhibitors (Ficlatuzumab) and p53-targetted therapies be it adenoviral p53 gene therapy or use of small molecules to restore TP53 function or disrupt inactivation of wild-type p53. Regardless of the combinations to be tested or trialled, the rationale for their combined use will depend on the overall molecular profile of the patient’s tumour gleaned through a precision medicine approach.

4. Conclusions and Future Directions

Currently, only cetuximab, pembrolizumab and nivolumab are approved by the FDA as molecular therapies for HNSCC. All three drugs are only indicated for locally advanced, recurrent or metastatic disease, and in combination with radiotherapy or other chemotherapeutic agents such as cisplatin or 5-fluorouracil. Surgery continues to be first line therapy for oral cavity SCC, with chemoradiotherapy indicated for oropharyngeal SCC. Although significant advances have been made in our understanding of the genes and associated signalling pathways involved in HNC, successful molecular therapies have not remarkably changed clinical practice as first line therapy. Molecular approaches however continue to serve patients with recalcitrant disease, or with particular mutational or immune profiles such as that seen in application of immune checkpoint inhibitors. More novel approaches with agents taking advantage of synthetic lethality or those exploiting p53 mutations utilising gene therapy hold hope for translation into clinical practice. While vaccination against oncogenic subtypes of HPV hold promise for a reduction in oropharyngeal and other HPV-induced HNCs, there remains a significant burden of disease that is nonresponsive to currently accepted standard therapies, and for which molecular therapies may still hold significant advantage. Taking full advantage of precision medicine in this regard is underpinned by our ability to accurately identify individual patient genomic variations and to tailor targeted therapy based on this. Incorporating screening for actionable genomic targets and treating with a matched targeted therapy in future clinical trials is but one approach to consider if there are to be significant benefits for patients moving forward. Design of new biomarker-driven clinical trials for HNSCC is urgently required if we are to successfully incorporate molecular therapies as standard treatment for head and neck cancers.

Author Contributions

Conceptualization, C.S.F.; methodology, C.S.F. and F.K.; investigation, C.S.F. and F.K., resources, C.S.F.; data curation, C.S.F. and F.K.; writing—original draft preparation, F.K. and C.S.F.; writing—review and editing, C.S.F. and F.K.; project administration, C.S.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors wish to thank Simon Fox for critical appraisal of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest in relation to the work presented in this paper. The authors have undertaken next generation sequencing of head and neck cancer and precancerous lesions utilising SOLiD™ and Ion™ technologies, funded by grants held by C.S.F awarded by the Queensland Government Smart Futures Co-Investment Fund and Cancer Australia, in collaboration with Life Technologies/Thermo Fisher Scientific and Agilent Technologies. No funding was received for this review.

References

  1. Hopkins, A.L.; Groom, C.R. The druggable genome. Nat. Rev. Drug Discov. 2002, 1, 727–730. [Google Scholar] [CrossRef] [PubMed]
  2. Russ, A.P.; Lampel, S. The druggable genome: An update. Drug Discov. Today 2005, 10, 1607–1610. [Google Scholar] [CrossRef]
  3. Stratton, M. Evolution of the Cancer Genome. J. Med. Genet. 2011, 48, S43. [Google Scholar] [CrossRef]
  4. Wong, K.M.; Hudson, T.J.; McPherson, J.D. Unraveling the Genetics of Cancer: Genome Sequencing and Beyond. Annu. Rev. Genom. Hum. Genet. 2011, 12, 407–430. [Google Scholar] [CrossRef]
  5. Pleasance, E.D.; Cheetham, R.K.; Stephens, P.J.; McBride, D.J.; Humphray, S.J.; Greenman, C.D.; Varela, I.; Lin, M.-L.; Ordóñez, G.R.; Bignell, G.R.; et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 2009, 463, 191–196. [Google Scholar] [CrossRef] [PubMed]
  6. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Dancey, J.E.; Bedard, P.L.; Onetto, N.; Hudson, T.J. The Genetic Basis for Cancer Treatment Decisions. Cell 2012, 148, 409–420. [Google Scholar] [CrossRef] [Green Version]
  8. Desmedt, C.; Voet, T.; Sotiriou, C.; Campbell, P.J. Next-generation sequencing in breast cancer: First take home messages. Curr. Opin. Oncol. 2012, 24, 597–604. [Google Scholar] [CrossRef] [PubMed]
  9. Mwenifumbo, J.C.; Marra, M.A. Cancer genome-sequencing study design. Nat. Rev. Genet. 2013, 14, 321–332. [Google Scholar] [CrossRef]
  10. Hudson, T.J.; Anderson, W.; Aretz, A.; Barker, A.D.; Bell, C.; Bernabe, R.R.; Bhan, M.K.; Calvo, F.; Eerola, I.; Gerhard, D.S.; et al. International network of cancer genome projects. Nature 2010, 464, 993–998. [Google Scholar]
  11. Mills, G.B. An emerging toolkit for targeted cancer therapies. Genome Res. 2012, 22, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bailey, M.H.; Tokheim, C.; Porta-Pardo, E.; Sengupta, S.; Bertrand, D.; Weerasinghe, A.; Colaprico, A.; Wendl, M.C.; Kim, J.; Reardon, B.; et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018, 174, 1034–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kaur, H.; Mao, S.; Shah, S.; Gorski, D.H.; Krawetz, S.A.; Sloane, B.F.; Mattingly, R.R. Next-generation sequencing: A powerful tool for the discovery of molecular markers in breast ductal carcinoma in situ. Expert Rev. Mol. Diagn. 2013, 13, 151–165. [Google Scholar] [CrossRef] [Green Version]
  14. Banerji, S.; Cibulskis, K.; Rangel-Escareno, C.; Brown, K.; Carter, S.L.; Frederick, A.M.; Lawrence, M.S.; Sivachenko, A.Y.; Sougnez, C.; Zou, L.; et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 2012, 486, 405–409. [Google Scholar] [CrossRef] [PubMed]
  15. Shah, S.P.; Roth, A.; Goya, R.; Oloumi, A.; Ha, G.; Zhao, Y.; Turashvili, G.; Ding, J.; Tse, K.; Haffari, G.; et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 2012, 486, 395–399. [Google Scholar] [CrossRef]
  16. Stephens, P.J.; The Oslo Breast Cancer Consortium (OSBREAC); Tarpey, P.S.; Davies, H.; Van Loo, P.; Greenman, C.; Wedge, D.C.; Nik-Zainal, S.; Martin, S.; Varela, I.; et al. The landscape of cancer genes and mutational processes in breast cancer. Nature 2012, 486, 400–404. [Google Scholar] [CrossRef] [PubMed]
  17. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [Green Version]
  18. Sieben, N.L.G.; Macropoulos, P.; Roemen, G.M.J.M.; Kolkman-Uljee, S.M.; Fleuren, G.J.; Houmadi, R.; Diss, T.; Warren, B.; Al Adnani, M.; De Goeij, A.P.M.; et al. In ovarian neoplasms, BRAF, but notKRAS, mutations are restricted to low-grade serous tumours. J. Pathol. 2004, 202, 336–340. [Google Scholar] [CrossRef]
  19. Bang, Y.-J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697. [Google Scholar] [CrossRef]
  20. Bookman, M.A.; Darcy, K.M.; Clarke-Pearson, D.; Boothby, R.A.; Horowitz, I.R. Evaluation of Monoclonal Humanized Anti-HER2 Antibody, Trastuzumab, in Patients with Recurrent or Refractory Ovarian or Primary Peritoneal Carcinoma with Overexpression of HER2: A Phase II Trial of the Gynecologic Oncology Group. J. Clin. Oncol. 2003, 21, 283–290. [Google Scholar] [CrossRef]
  21. Fleming, G.F.; Sill, M.W.; Darcy, K.M.; McMeekin, D.S.; Thigpen, J.T.; Adler, L.M.; Berek, J.S.; Chapman, J.A.; DiSilvestro, P.A.; Horowitz, I.R.; et al. Phase II trial of trastuzumab in women with advanced or recurrent, HER2-positive endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol. Oncol. 2010, 116, 15–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Farah, C.S. Molecular landscape of head and neck cancer and implications for therapy. Ann. Transl. Med. 2021, 9, 915. [Google Scholar] [CrossRef]
  23. Kordbacheh, F.; Farah, C. Molecular Pathways and Druggable Targets in Head and Neck Squamous Cell Carcinoma. Cancers 2021, 13, 3453. [Google Scholar] [CrossRef] [PubMed]
  24. Mendelsohn, J.; Baselga, J. The EGF receptor family as targets for cancer therapy. Oncogene 2000, 19, 6550–6565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mellinghoff, I.K.; Wang, M.Y.; Vivanco, I.; Haas-Kogan, D.A.; Zhu, S.; Dia, E.Q.; Lu, K.V.; Yoshimoto, K.; Huang, J.H.Y.; Chute, D.J.; et al. Molecular Determinants of the Response of Glioblastomas to EGFR Kinase Inhibitors. N. Engl. J. Med. 2005, 353, 2012–2024. [Google Scholar] [CrossRef] [Green Version]
  26. Yarden, Y. The EGFR family and its ligands in human cancer: Signalling mechanisms and therapeutic opportunities. Eur. J. Cancer 2001, 37, 3–8. [Google Scholar] [CrossRef]
  27. Citri, A.; Yarden, Y. EGF–ERBB signalling: Towards the systems level. Nat. Rev. Mol. Cell Biol. 2006, 7, 505–516. [Google Scholar] [CrossRef]
  28. Hynes, N.E.; Lane, H.A. ERBB receptors and cancer: The complexity of targeted inhibitors. Nat. Rev. Cancer 2005, 5, 341. [Google Scholar] [CrossRef]
  29. Garcia-Foncillas, J.; Sunakawa, Y.; Aderka, D.; Wainberg, Z.; Ronga, P.; Witzler, P.; Stintzing, S. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tu-mors. Front. Oncol. 2019, 9, 849. [Google Scholar] [CrossRef]
  30. Klinghammer, K.; Gauler, T.; Dietz, A.; Grünwald, V.; Stöhlmacher, J.; Knipping, S.; Schroeder, M.; Guntinas-Lichius, O.; Frickhofen, N.; Lindeman, H.-W.; et al. Cetuximab, fluorouracil and cisplatin with or without docetaxel for patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (CeFCiD): An open-label phase II randomised trial (AIO/IAG-KHT trial 1108). Eur. J. Cancer 2019, 122, 53–60. [Google Scholar] [CrossRef]
  31. Rodenhuis, S.; Slebos, R.J.; Boot, A.J.; Evers, S.G.; Mooi, W.J.; Wagenaar, S.S.; Van Bodegom, P.C.; Bos, J.L. Incidence and possible clinical significance of K-ras oncogene activation in adenocarcinoma of the human lung. Cancer Res. 1988, 48, 5738–5741. [Google Scholar]
  32. Ahrendt, S.A.; Decker, P.A.; Alawi, E.A.; Zhu, Y.R.; Sanchez-Cespedes, M.; Yang, S.C.; Haasler, G.B.; Kajdacsy-Balla, A.; Demeure, M.J.; Sidransky, D. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary ade-nocarcinoma of the lung. Cancer 2001, 92, 1525–1530. [Google Scholar] [CrossRef]
  33. Pao, W.; Wang, T.Y.; Riely, G.J.; Miller, V.A.; Pan, Q.; Ladanyi, M.; Zakowski, M.F.; Heelan, R.T.; Kris, M.; Varmus, H.E. KRAS Mutations and Primary Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib. PLoS Med. 2005, 2, e17. [Google Scholar] [CrossRef] [Green Version]
  34. De Roock, W.; Piessevaux, H.; De Schutter, J.; Janssens, M.; De Hertogh, G.; Personeni, N.; Biesmans, B.; Van Laethem, J.L.; Peeters, M.; Humblet, Y.; et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic col-orectal cancer treated with cetuximab. Ann. Oncol. 2008, 19, 508–515. [Google Scholar] [CrossRef]
  35. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.-M.; Zhao, X.; Christensen, J.; et al. MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  36. Fung, C.; Grandis, J.R. Emerging drugs to treat squamous cell carcinomas of the head and neck. Expert Opin. Emerg. Drugs 2010, 15, 355–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Byeon, H.K.; Ku, M.; Yang, J. Beyond EGFR inhibition: Multilateral combat strategies to stop the progression of head and neck cancer. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef] [Green Version]
  38. Suh, Y.; Amelio, I.; Urbano, T.G.; Tavassoli, M. Clinical update on cancer: Molecular oncology of head and neck cancer. Cell Death Dis. 2014, 5, e1018. [Google Scholar] [CrossRef] [Green Version]
  39. Stewart, J.S.W.; Cohen, E.E.; Licitra, L.; Van Herpen, C.M.; Khorprasert, C.; Soulieres, D.; Vodvarka, P.; Rischin, D.; Garin, A.M.; Hirsch, F.R.; et al. Phase III Study of Gefitinib Compared with Intravenous Methotrexate for Recurrent Squamous Cell Carcinoma of the Head and Neck. J. Clin. Oncol. 2009, 27, 1864–1871. [Google Scholar] [CrossRef]
  40. Argiris, A.; Ghebremichael, M.; Gilbert, J.; Lee, J.W.; Sachidanandam, K.; Kolesar, J.M.; Burtness, B.; Forastiere, A.A. Phase III Randomized, Placebo-Controlled Trial of Docetaxel with or without Gefitinib in Recurrent or Met-astatic Head and Neck Cancer: An Eastern Cooperative Oncology Group Trial. J. Clin. Oncol. 2013, 31, 1405–1414. [Google Scholar] [CrossRef] [PubMed]
  41. Moore, M.J.; Goldstein, D.; Hamm, J.; Figer, A.; Hecht, J.R.; Gallinger, S.; Au, H.J.; Murawa, P.; Walde, D.; Wolff, R.A.; et al. Erlotinib Plus Gemcitabine Compared with Gemcitabine Alone in Patients with Advanced Pancreatic Cancer: A Phase III Trial of the National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 2007, 25, 1960–1966. [Google Scholar] [CrossRef]
  42. Shepherd, F.A.; Pereira, J.R.; Ciuleanu, T.E.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in Previously Treated Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2005, 353, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ngan, H.-L.; Poon, P.H.Y.; Su, Y.-X.; Chan, J.Y.K.; Lo, K.-W.; Yeung, C.K.; Liu, Y.; Wong, E.; Li, H.; Lau, C.W.; et al. Erlotinib sensitivity of MAPK1p.D321N mutation in head and neck squamous cell carcinoma. NPJ Genom. Med. 2020, 5, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Soulieres, D.; Senzer, N.N.; Vokes, E.E.; Hidalgo, M.; Agarwala, S.S.; Siu, L.L. Multicenter Phase II Study of Erlotinib, an Oral Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor, in Patients with Recurrent or Metastatic Squamous Cell Cancer of the Head and Neck. J. Clin. Oncol. 2004, 22, 77–85. [Google Scholar] [CrossRef]
  45. Siu, L.L.; Soulieres, D.; Chen, E.X.; Pond, G.R.; Chin, S.F.; Francis, P.; Harvey, L.; Klein, M.; Zhang, W.; Dancey, J.; et al. Phase I/II Trial of Erlotinib and Cisplatin in Patients with Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: A Princess Margaret Hospital Phase II Consortium and National Cancer Institute of Canada Clinical Trials Group Study. J. Clin. Oncol. 2007, 25, 2178–2183. [Google Scholar] [CrossRef] [Green Version]
  46. Burtness, B.; Goldwasser, M.A.; Flood, W.; Mattar, B.; Forastiere, A.A. Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastat-ic/recurrent head and neck cancer: An eastern cooperative oncology group study. J. Clin. Oncol. 2005, 23, 8646–8654. [Google Scholar] [CrossRef]
  47. Martins, R.G.; Parvathaneni, U.; Bauman, J.E.; Sharma, A.K.; Raez, L.E.; Papagikos, M.A.; Yunus, F.; Kurland, B.F.; Eaton, K.D.; Liao, J.J.; et al. Cisplatin and Radiotherapy with or without Erlotinib in Locally Advanced Squamous Cell Carcinoma of the Head and Neck: A Randomized Phase II Trial. J. Clin. Oncol. 2013, 31, 1415–1421. [Google Scholar] [CrossRef]
  48. Gross, N.D.; Bauman, J.E.; Gooding, W.E.; Denq, W.; Thomas, S.; Wang, L.; Chiosea, S.; Hood, B.L.; Flint, M.; Sun, M.; et al. Erlotinib, Erlotinib–Sulindac versus Placebo: A Randomized, Double-Blind, Placebo-Controlled Window Trial in Operable Head and Neck Cancer. Clin. Cancer Res. 2014, 20, 3289–3298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gold, K.A.; Kies, M.S.; William, W.N.; Johnson, F.M.; Lee, J.J.; Glisson, B.S. Erlotinib in the treatment of recurrent or metastatic cutaneous squamous cell carcinoma: A single-arm phase 2 clinical trial. Cancer 2018, 124, 2169–2173. [Google Scholar] [CrossRef]
  50. Saba, N.F.; Hurwitz, S.J.; Magliocca, K.; Kim, S.; Owonikoko, T.K.; Harvey, D.; Ramalingam, S.S.; Chen, Z.; Rogerio, J.; Mendel, J.; et al. Phase 1 and pharmacokinetic study of everolimus in combination with cetuximab and carboplatin for re-current/metastatic squamous cell carcinoma of the head and neck. Cancer 2014, 120, 3940–3951. [Google Scholar] [CrossRef]
  51. De Souza, J.A.; Davis, D.W.; Zhang, Y.; Khattri, A.; Seiwert, T.Y.; Aktolga, S.; Wong, S.J.; Kozloff, M.F.; Nattam, S.; Lingen, M.W.; et al. A Phase II Study of Lapatinib in Recurrent/Metastatic Squamous Cell Carcinoma of the Head and Neck. Clin. Cancer Res. 2012, 18, 2336–2343. [Google Scholar] [CrossRef] [Green Version]
  52. Weiss, J.M.; Bagley, S.; Hwang, W.-T.; Bauml, J.; Olson, J.G.; Cohen, R.B.; Hayes, D.N.; Langer, C. Capecitabine and lapatinib for the first-line treatment of metastatic/recurrent head and neck squamous cell carcinoma. Cancer 2016, 122, 2350–2355. [Google Scholar] [CrossRef] [PubMed]
  53. Harrington, K.; Temam, S.; Mehanna, H.; D’Cruz, A.; Jain, M.; D’Onofrio, I.; Manikhas, G.; Horvath, Z.; Sun, Y.; Dietzsch, S.; et al. Postoperative Adjuvant Lapatinib and Concurrent Chemoradiotherapy Followed by Maintenance Lapatinib Monotherapy in High-Risk Patients with Resected Squamous Cell Carcinoma of the Head and Neck: A Phase III, Randomized, Double-Blind, Placebo-Controlled Study. J. Clin. Oncol. 2015, 33, 4202–4209. [Google Scholar] [CrossRef]
  54. Sano, D.; Matsumoto, F.; Valdecanas, D.R.; Zhao, M.; Molkentine, D.P.; Takahashi, Y.; Hanna, E.; Papadimitrakopoulou, V.; Heymach, J.; Milas, L.; et al. Vandetanib Restores Head and Neck Squamous Cell Carcinoma Cells’ Sensitivity to Cisplatin and Radiation In Vivo and In Vitro. Clin. Cancer Res. 2011, 17, 1815–1827. [Google Scholar] [CrossRef] [Green Version]
  55. Burtness, B.; Harrington, K.J.; Greil, R.; Soulieres, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Baste, N.; Neupane, P.; Bratland, A.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or meta-static squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  56. Vincenzi, B.; Zoccoli, A.; Pantano, F.; Venditti, O.; Galluzzo, S. CETUXIMAB: From Bench to Bedside. Curr. Cancer Drug Targets 2010, 10, 80–95. [Google Scholar] [CrossRef]
  57. Taylor, R.J.; Chan, S.L.; Wood, A.; Voskens, C.J.; Wolf, J.S.; Lin, W.; Chapoval, A.; Schulze, D.H.; Tian, G.L.; Strome, S.E. Fc gamma RIIIa polymorphisms and cetuximab induced cytotoxicity in squamous cell carcinoma of the head and neck. Cancer Immunol. Immunother. 2009, 58, 997–1006. [Google Scholar] [CrossRef]
  58. Cai, W.-Q.; Zeng, L.-S.; Wang, L.-F.; Wang, Y.-Y.; Cheng, J.-T.; Zhang, Y.; Han, Z.-W.; Zhou, Y.; Huang, S.-L.; Wang, X.-W.; et al. The Latest Battles Between EGFR Monoclonal Antibodies and Resistant Tumor Cells. Front. Oncol. 2020, 10, 1249. [Google Scholar] [CrossRef] [PubMed]
  59. Goldstein, N.; Prewett, M.; Zuklys, K.; Rockwell, P.; Mendelsohn, J. Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model. Clin. Cancer Res. 1995, 1, 1311–1318. [Google Scholar]
  60. Cunningham, D.; Humblet, Y.; Siena, S.; Khayat, D.; Bleiberg, H.; Santoro, A.; Bets, D.; Mueser, M.; Harstrick, A.; Verslype, C.; et al. Cetuximab Monotherapy and Cetuximab plus Irinotecan in Irinotecan-Refractory Metastatic Colorectal Cancer. N. Engl. J. Med. 2004, 351, 337–345. [Google Scholar] [CrossRef] [Green Version]
  61. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bonner, J.A.; Spencer, S.A.; Rowinsky, E.K. Rowinsky, Cetuximab plus radiotherapy for head and neck cancer—Reply. N. Engl. J. Med. 2006, 354, 2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Siddik, Z.H. Cisplatin: Mode of cytotoxic action and molecular basis of resistance. Oncogene 2003, 22, 7265–7279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Vermorken, J.B.; Mesia, R.; Rivera, F.; Remenar, E.; Kawecki, A.; Rottey, S.; Erfan, J.; Zabolotnyy, D.; Kienzer, H.-R.; Cupissol, D.; et al. Platinum-Based Chemotherapy plus Cetuximab in Head and Neck Cancer. N. Engl. J. Med. 2008, 359, 1116–1127. [Google Scholar] [CrossRef] [Green Version]
  65. Rampias, T.; Giagini, A.; Siolos, S.; Matsuzaki, H.; Sasaki, C.; Scorilas, A.; Psyrri, A. RAS/PI3K Crosstalk and Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2014, 20, 2933–2946. [Google Scholar] [CrossRef] [Green Version]
  66. Melosky, B.; Burkes, R.; Rayson, D.; Alcindor, T.; Shear, N.; Lacouture, M. Management of skin rash during EGFR-targeted monoclonal antibody treatment for gastrointestinal malig-nancies: Canadian recommendations. Curr. Oncol. 2009, 16, 16–26. [Google Scholar] [CrossRef] [Green Version]
  67. Pinto, C.; Barone, C.A.; Girolomoni, G.; Russi, E.G.; Merlano, M.C.; Ferrari, D.; Maiello, E. Management of Skin Toxicity Associated with Cetuximab Treatment in Combination with Chemotherapy or Radiotherapy. Oncologist 2011, 16, 228–238. [Google Scholar] [CrossRef] [Green Version]
  68. Segaert, S.; Van Cutsem, E. Clinical signs, pathophysiology and management of skin toxicity during therapy with epidermal growth factor receptor inhibitors. Ann. Oncol. 2005, 16, 1425–1433. [Google Scholar] [CrossRef]
  69. Jerhammar, F.; Johansson, A.-C.; Ceder, R.; Welander, J.; Jansson, A.; Grafström, R.C.; Söderkvist, P.; Roberg, K. YAP1 is a potential biomarker for cetuximab resistance in head and neck cancer. Oral Oncol. 2014, 50, 832–839. [Google Scholar] [CrossRef]
  70. Gillison, M.L.; Trotti, A.M.; Harris, J.; Eisbruch, A.; Harari, P.M.; Adelstein, D.J.; Jordan, R.C.K.; Zhao, W.; Sturgis, E.M.; Burtness, B.; et al. Radiotherapy plus cetuximab or cisplatin in human papillomavirus-positive oropharyngeal cancer (NRG Oncology RTOG 1016): A randomised, multicentre, non-inferiority trial. Lancet 2018, 393, 40–50. [Google Scholar] [CrossRef]
  71. Moon, C.; Chae, Y.K.; Lee, J. Targeting epidermal growth factor receptor in head and neck cancer: Lessons learned from ce-tuximab. Exp. Biol. Med. 2010, 235, 907–920. [Google Scholar] [CrossRef]
  72. Saltz, L.; Easley, C.; Kirkpatrick, P. Panitumumab. Nat. Rev. Drug Discov. 2006, 5, 987–988. [Google Scholar] [CrossRef] [PubMed]
  73. Giusti, R.M.; Shastri, K.A.; Cohen, M.H.; Keegan, P.; Pazdur, R. FDA drug approval summary: Panitumumab (Vectibix (TM)). Oncologist 2007, 12, 577–583. [Google Scholar] [CrossRef]
  74. Vermorken, J.B.; Stöhlmacher-Williams, J.; Davidenko, I.; Licitra, L.; Winquist, E.; Villanueva, C.; Foa, P.; Rottey, S.; Skladowski, K.; Tahara, M.; et al. Cisplatin and fluorouracil with or without panitumumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SPECTRUM): An open-label phase 3 randomised trial. Lancet Oncol. 2013, 14, 697–710. [Google Scholar] [CrossRef] [Green Version]
  75. Siu, L.L.; Waldron, J.N.; Chen, B.E.; Winquist, E.; Wright, J.R.; Nabid, A.; Hay, J.H.; Ringash, J.; Liu, G.; Johnson, A.; et al. Effect of Standard Radiotherapy with Cisplatin vs Accelerated Radiotherapy with Panitumumab in Locore-gionally Advanced Squamous Cell Head and Neck Carcinoma: A Randomized Clinical Trial. JAMA Oncol. 2017, 3, 220–226. [Google Scholar] [CrossRef] [PubMed]
  76. Siano, M.; Molinari, F.; Martin, V.; Mach, N.; Früh, M.; Freguia, S.; Corradino, I.; Ghielmini, M.; Frattini, M.; Espeli, V. Multicenter Phase II Study of Panitumumab in Platinum Pretreated, Advanced Head and Neck Squamous Cell Cancer. Oncologist 2017, 22, 782–e70. [Google Scholar] [CrossRef] [Green Version]
  77. Saloura, V.; Cohen, E.E.; Licitra, L.; Billan, S.; Dinis, J.; Lisby, S.; Gauler, T.C. An open-label single-arm, phase II trial of zalutumumab, a human monoclonal anti-EGFR antibody, in pa-tients with platinum-refractory squamous cell carcinoma of the head and neck. Cancer Chemother. Pharm. 2014, 73, 1227–1239. [Google Scholar] [CrossRef]
  78. Machiels, J.-P.; Subramanian, S.; Ruzsa, A.; Repassy, G.; Lifirenko, I.; Flygare, A.; Sørensen, P.; Nielsen, T.; Lisby, S.; Clement, P.M. Zalutumumab plus best supportive care versus best supportive care alone in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck after failure of platinum-based chemotherapy: An open-label, randomised phase 3 trial. Lancet Oncol. 2011, 12, 333–343. [Google Scholar] [CrossRef]
  79. Rivera, F.; Vega-Villegas, M.E.; López-Brea, M.F.; Márquez, R. Current situation of Panitumumab, Matuzumab, Nimotuzumab and Zalutumumab. Acta Oncol. 2008, 47, 9–19. [Google Scholar] [CrossRef] [PubMed]
  80. Salazar, R.; Garcia-Carbonero, R.; Libutti, S.K.; Hendifar, A.E.; Custodio, A.; Guimbaud, R.; Lombard-Bohas, C.; Ricci, S.; Klumpen, H.J.; Capdevila, J.; et al. Phase II Study of BEZ235 versus Everolimus in Patients with Mammalian Target of Rapamycin Inhibi-tor-Naive Advanced Pancreatic Neuroendocrine Tumors. Oncologist 2018, 23, 766.e90. [Google Scholar] [CrossRef] [Green Version]
  81. Hotte, S.J.; Chi, K.N.; Joshua, A.; Tu, D.; Macfarlane, R.J.; Gregg, R.W.; Ruether, J.D.; Basappa, N.S.; Finch, D.; Salim, M.; et al. A Phase II Study of PX-866 in Patients with Recurrent or Metastatic Castration-resistant Prostate Cancer: Canadian Cancer Trials Group Study IND205. Clin. Genitourin. Cancer 2019, 17, 201–208.e1. [Google Scholar] [CrossRef]
  82. Borkowska, E.M.; Barańska, M.; Kowalczyk, K.; Pietruszewska, W. Detection of PIK3CA Gene Mutation in Head and Neck Squamous Cell Carcinoma Using Droplet Digital PCR and RT-qPCR. Biomolecules 2021, 11, 818. [Google Scholar] [CrossRef]
  83. Network, T.C.G.A. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [Green Version]
  84. Rodon, J.; Curigliano, G.; Delord, J.-P.; Harb, W.; Azaro, A.; Han, Y.; Wilke, C.; Donnet, V.; Sellami, D.; Beck, T. A Phase Ib, open-label, dose-finding study of alpelisib in combination with paclitaxel in patients with advanced solid tumors. Oncotarget 2018, 9, 31709–31718. [Google Scholar] [CrossRef] [Green Version]
  85. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [Green Version]
  86. Patel, V.; Lahusen, T.; Sy, T.; Sausville, E.; Gutkind, J.S.; Senderowicz, A.M. Perifosine, a novel alkylphospholipid, induces p21(WAF1) expression in squamous carcinoma cells through a p53-independent pathway, leading to loss in cyclin-dependent kinase activity and cell cycle arrest. Cancer Res. 2002, 62, 1401–1409. [Google Scholar]
  87. Argiris, A.; Cohen, E.; Karrison, T.; Esparaz, B.; Mauer, A.; Ansari, R.; Wong, S.; Lu, Y.; Pins, M.; Dancey, J.; et al. A phase II trial of perifosine, an oral alkylphospholipid, in recurrent or metastatic head and neck cancer. Cancer Biol. Ther. 2006, 5, 766–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Faivre, S.; Kroemer, G.; Raymond, E. Current development of mTOR inhibitors as anticancer agents. Nat. Rev. Drug Discov. 2006, 5, 671–688. [Google Scholar] [CrossRef] [PubMed]
  89. Aoki, M.; Blazek, E.; Vogt, P.K. A role of the kinase mTOR in cellular transformation induced by the oncoproteins P3k and Akt. Proc. Natl. Acad. Sci. USA 2001, 98, 136–141. [Google Scholar] [CrossRef]
  90. Freudlsperger, C.; Burnett, J.R.; Friedman, J.A.; Kannabiran, V.R.; Chen, Z.; Van Waes, C. EGFR-PI3K-AKT-mTOR signaling in head and neck squamous cell carcinomas: Attractive targets for molecular-oriented therapy. Expert Opin. Ther. Targets 2011, 15, 63–74. [Google Scholar] [CrossRef] [Green Version]
  91. Seto, B. Rapamycin and mTOR: A serendipitous discovery and implications for breast cancer. Clin. Transl. Med. 2012, 1, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Massarelli, E.; Lin, H.; Ginsberg, L.E.; Tran, H.T.; Lee, J.J.; Canales, J.R.; Williams, M.D.; Blumenschein, G.R.; Lu, C.; Heymach, J.V.; et al. Phase II trial of everolimus and erlotinib in patients with platinum-resistant recurrent and/or metastatic head and neck squamous cell carcinoma. Ann. Oncol. 2015, 26, 1476–1480. [Google Scholar] [CrossRef] [PubMed]
  93. Kwitkowski, V.E.; Prowell, T.M.; Ibrahim, A.; Farrell, A.T.; Justice, R.; Mitchell, S.S.; Sridhara, R.; Pazdur, R. FDA Approval Summary: Temsirolimus as Treatment for Advanced Renal Cell Carcinoma. Oncol. 2010, 15, 428–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bauman, J.E.; Arias-Pulido, H.; Lee, S.-J.; Fekrazad, M.H.; Ozawa, H.; Fertig, E.; Howard, J.; Bishop, J.; Wang, H.; Olson, G.T.; et al. A phase II study of temsirolimus and erlotinib in patients with recurrent and/or metastatic, platinum-refractory head and neck squamous cell carcinoma. Oral Oncol. 2013, 49, 461–467. [Google Scholar] [CrossRef] [Green Version]
  95. O’Reilly, K.E.; Rojo, F.; She, Q.-B.; Solit, D.; Mills, G.B.; Smith, D.; Lane, H.; Hofmann, F.; Hicklin, D.J.; Ludwig, D.L.; et al. mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates Akt. Cancer Res. 2006, 66, 1500–1508. [Google Scholar] [CrossRef] [Green Version]
  96. Moore, A.R.; Rosenberg, S.C.; McCormick, F.; Malek, S. RAS-targeted therapies: Is the undruggable drugged? Nat. Rev. Drug Discov. 2020, 19, 533–552. [Google Scholar] [CrossRef] [PubMed]
  97. Ho, A.L.; Brana, I.; Haddad, R.; Bauman, J.; Bible, K.; Oosting, S.; Wong, D.J.; Ahn, M.-J.; Boni, V.; Even, C.; et al. Tipifarnib in Head and Neck Squamous Cell Carcinoma with HRAS Mutations. J. Clin. Oncol. 2021, 39, 1856–1864. [Google Scholar] [CrossRef]
  98. van Harten, A.M.; Brakenhoff, R.H. Tipifarnib as a Precision Therapy for HRAS-Mutant Head and Neck Squamous Cell Carcinomas. Mol. Cancer Ther. 2020, 19, 1784–1796. [Google Scholar] [CrossRef]
  99. Williamson, S.K.; Moon, J.; Huang, C.H.; Guaglianone, P.P.; Leblanc, M.; Wolf, G.T.; Urba, S.G. Phase II Evaluation of Sorafenib in Advanced and Metastatic Squamous Cell Carcinoma of the Head and Neck: Southwest Oncology Group Study S0420. J. Clin. Oncol. 2010, 28, 3330–3335. [Google Scholar] [CrossRef] [Green Version]
  100. Elser, C.; Siu, L.L.; Winquist, E.; Agulnik, M.; Pond, G.R.; Chin, S.F.; Francis, P.; Cheiken, R.; Elting, J.; McNabola, A.; et al. Phase II Trial of Sorafenib in Patients with Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck or Nasopharyngeal Carcinoma. J. Clin. Oncol. 2007, 25, 3766–3773. [Google Scholar] [CrossRef]
  101. Cohen, E.E.; Kane, M.A.; List, M.A.; Brockstein, B.E.; Mehrotra, B.; Huo, D.; Mauer, A.M.; Pierce, C.; Dekker, A.; Vokes, E.E. Phase II Trial of Gefitinib 250 mg Daily in Patients with Recurrent and/or Metastatic Squamous Cell Carcinoma of the Head and Neck. Clin. Cancer Res. 2005, 11, 8418–8424. [Google Scholar] [CrossRef] [Green Version]
  102. Abidoye, O.O.; Cohen, E.E.; Wong, S.J.; Kozloff, M.F.; Nattam, S.R.; Stenson, K.M.; Blair, E.A.; Day, S.; Dancey, J.E.; Vokes, E.E. A phase II study of lapatinib (GW572016) in recurrent/metastatic (RIM) squamous cell carcinoma of the head and neck (SCCHN). J. Clin. Oncol. 2006, 24, 297s. [Google Scholar] [CrossRef]
  103. Wheeler, R.H.; Jones, D.; Sharma, P.; Davis, R.K.; Spilker, H.; Boucher, K.; Leachman, S.; Grossman, D.; Salzman, K.; Akerley, W. Clinical and molecular phase II study of gefitinib in patients (pts) with recurrent squamous cell cancer of the head and neck (H&N Ca). J. Clin. Oncol. 2005, 23, 5531. [Google Scholar] [CrossRef]
  104. Rong, C.; Muller, M.F.; Xiang, F.; Jensen, A.; Weichert, W.; Major, G.; Plinkert, P.K.; Hess, J.; Affolter, A. Adaptive ERK signalling activation in response to therapy and in silico prognostic evaluation of EGFR-MAPK in HNSCC. Br. J. Cancer 2020, 123, 288–297. [Google Scholar] [CrossRef]
  105. Zhao, Y.; Adjei, A.A. The clinical development of MEK inhibitors. Nat. Rev. Clin. Oncol. 2014, 11, 385–400. [Google Scholar] [CrossRef] [PubMed]
  106. Guo, Y.; Pan, W.; Liu, S.; Shen, Z.; Xu, Y.; Hu, L. ERK/MAPK signalling pathway and tumorigenesis (Review). Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. LoRusso, P.M.; Adjei, A.A.; Varterasian, M.; Gadgeel, S.; Reid, J.; Mitchell, D.Y.; Hanson, L.; DeLuca, P.; Bruzek, L.; Piens, J.; et al. Phase I and pharmacodynamic study of the oral MEK inhibitor CI-1040 in patients with advanced ma-lignancies. J. Clin. Oncol. 2005, 23, 5281–5293. [Google Scholar] [CrossRef]
  108. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandala, M.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef] [Green Version]
  109. Dhillon, S. Dabrafenib plus Trametinib: A Review in Advanced Melanoma with a BRAF V600 Mutation. Target. Oncol. 2016, 11, 417–428. [Google Scholar] [CrossRef] [PubMed]
  110. Santarpia, L.; Lippman, S.M.; El-Naggar, A.K. El-Naggar, Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin. Ther. Targets 2012, 16, 103–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Ho, A.L.; Grewal, R.K.; Leboeuf, R.; Sherman, E.J.; Pfister, D.G.; Deandreis, D.; Pentlow, K.S.; Zanzonico, P.B.; Haque, S.; Gavane, S.; et al. Selumetinib-Enhanced Radioiodine Uptake in Advanced Thyroid Cancer. N. Engl. J. Med. 2013, 368, 623–632. [Google Scholar] [CrossRef] [Green Version]
  112. Fortini, M.E. gamma-secretase-mediated proteolysis in cell-surface-receptor signalling. Nat. Rev. Mol. Cell Biol. 2002, 3, 673–684. [Google Scholar] [CrossRef]
  113. Ran, Y.; Hossain, F.; Pannuti, A.; Lessard, C.B.; Ladd, G.Z.; Jung, J.I.; Minter, L.M.; Osborne, B.A.; Miele, L.; Golde, T.E. gamma-Secretase inhibitors in cancer clinical trials are pharmacologically and functionally distinct. EMBO Mol. Med. 2017, 9, 950–966. [Google Scholar] [CrossRef]
  114. Lee, S.M.; Moon, J.; Do, B.G.R.; Chidiac, T.; Flaherty, L.E.; Zha, Y.; Othus, M.; Ribas, A.; Sondak, V.K.; Gajewski, T.F.; et al. Phase 2 study of RO4929097, a gamma-secretase inhibitor, in metastatic melanoma: SWOG 0933. Cancer 2014, 121, 432–440. [Google Scholar] [CrossRef] [Green Version]
  115. Sahebjam, S.; Bedard, P.L.; Castonguay, V.; Chen, Z.; Reedijk, M.; Liu, G.; Cohen, B.; Zhang, W.-J.; Clarke, B.; Zhang, T.; et al. A phase I study of the combination of ro4929097 and cediranib in patients with advanced solid tumours (PJC-004/NCI 8503). Br. J. Cancer 2013, 109, 943–949. [Google Scholar] [CrossRef]
  116. Furge, K.A.; Kiewlich, D.; Le, P.; Vo, M.N.; Faure, M.; Howlett, A.R.; Lipson, K.; Woude, G.F.V.; Webb, C.P. Suppression of Ras-mediated tumorigenicity and metastasis through inhibition of the Met receptor tyrosine kinase. Proc. Natl. Acad. Sci. USA 2001, 98, 10722–10727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Moghul, A.; Lin, L.; Beedle, A.; Kanbourshakir, A.; Defrances, M.C.; Liu, Y.H.; Zarnegar, R. Modulation of C-Met Protooncogene (Hgf Receptor) Messenger-Rna Abundance by Cytokines and Hor-mones—Evidence for Rapid Decay of the 8 Kb C-Met Transcript. Oncogene 1994, 9, 2045–2052. [Google Scholar] [PubMed]
  118. Kim, E.S.; Salgia, R. MET Pathway as a Therapeutic Target. J. Thorac. Oncol. 2009, 4, 444–447. [Google Scholar] [CrossRef] [Green Version]
  119. Kuba, K.; Matsumoto, K.; Date, K.; Shimura, H.; Tanaka, M.; Nakamura, T. HGF/NK4, a four-kringle antagonist of hepatocyte growth factor, is an angiogenesis inhibitor that suppresses tumor growth and metastasis in mice. Cancer Res. 2000, 60, 6737–6743. [Google Scholar] [PubMed]
  120. Seiwert, T.; Sarantopoulos, J.; Kallender, H.; McCallum, S.; Keer, H.N.; Blumenschein, G. Phase II trial of single-agent foretinib (GSK1363089) in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. Investig. New Drugs 2012, 31, 417–424. [Google Scholar] [CrossRef] [Green Version]
  121. Infante, J.R.; Rugg, T.; Gordon, M.; Rooney, I.; Rosen, L.; Zeh, K.; Liu, R.; Burris, H.A.; Ramanathan, R.K. Unexpected renal toxicity associated with SGX523, a small molecule inhibitor of MET. Investig. New Drugs 2012, 31, 363–369. [Google Scholar] [CrossRef] [PubMed]
  122. Berthou, S.; Aebersold, D.M.; Schmidt, L.S.; Stroka, D.; Heigl, C.; Streit, B.; Stalder, D.; Gruber, G.; Liang, C.; Howlett, A.R.; et al. The Met kinase inhibitor SU11274 exhibits a selective inhibition pattern toward different receptor mutated variants. Oncogene 2004, 23, 5387–5393. [Google Scholar] [CrossRef] [Green Version]
  123. Puri, N.; Khramtsov, A.; Ahmed, S.; Nallasura, V.; Hetzel, J.T.; Jagadeeswaran, R.; Karczmar, G.; Salgia, R. A Selective Small Molecule Inhibitor of c-Met, PHA665752, Inhibits Tumorigenicity and Angiogenesis in Mouse Lung Cancer Xenografts. Cancer Res. 2007, 67, 3529–3534. [Google Scholar] [CrossRef] [Green Version]
  124. Lai, S.Y.; Johnson, F.M. Defining the role of the JAK-STAT pathway in head and neck and thoracic malignancies: Implications for future therapeutic approaches. Drug Resist. Updat. 2010, 13, 67–78. [Google Scholar] [CrossRef]
  125. Stegeman, H.; Kaanders, J.H.; Verheijen, M.M.; Peeters, W.J.; Wheeler, D.L.; Iida, M.; Grénman, R.; van der Kogel, A.J.; Span, P.N.; Bussink, J. Combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition reduces survival of head and neck cancer lines. Mol. Cancer 2013, 12, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hayakawa, F.; Sugimoto, K.; Harada, Y.; Hashimoto, N.; Ohi, N.; Kurahashi, S.; Naoe, T. A novel STAT inhibitor, OPB-31121, has a significant antitumor effect on leukemia with STAT-addictive oncokinases. Blood Cancer J. 2013, 3, e166. [Google Scholar] [CrossRef]
  127. Brambilla, L.; Lahiri, T.; Cammer, M.; Levy, D.E. STAT3 Inhibitor OPB-51602 Is Cytotoxic to Tumor Cells Through Inhibition of Complex I and ROS Induc-tion. iScience 2020, 23, 101822. [Google Scholar] [CrossRef] [PubMed]
  128. Leong, P.L.; Andrews, G.A.; Johnson, D.E.; Dyer, K.F.; Xi, S.; Mai, J.C.; Robbins, P.D.; Gadiparthi, S.; Burke, N.A.; Watkins, S.F.; et al. Targeted inhibition of Stat3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc. Natl. Acad. Sci. USA 2003, 100, 4138–4143. [Google Scholar] [CrossRef] [Green Version]
  129. Xi, S.; Gooding, W.; Grandis, J.R. In vivo antitumor efficacy of STAT3 blockade using a transcription factor decoy approach: Implications for cancer therapy. Oncogene 2004, 24, 970–979. [Google Scholar] [CrossRef] [Green Version]
  130. Klein, J.D.; Sano, D.; Sen, M.; Myers, J.N.; Grandis, J.R.; Kim, S. STAT3 Oligonucleotide Inhibits Tumor Angiogenesis in Preclinical Models of Squamous Cell Carcinoma. PLoS ONE 2014, 9, e81819. [Google Scholar] [CrossRef] [Green Version]
  131. Cristina, V.; Gómez, R.G.H.; Szturz, P.; Espeli, V.; Siano, M. Immunotherapies and Future Combination Strategies for Head and Neck Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 5399. [Google Scholar] [CrossRef] [Green Version]
  132. Solimani, F.; Meier, K.; Ghoreschi, K. Emerging Topical and Systemic JAK Inhibitors in Dermatology. Front. Immunol. 2019, 10, 2847. [Google Scholar] [CrossRef] [Green Version]
  133. Verstovsek, S.; Kantarjian, H.M.; Pardanani, A.; Thomas, D.A.; Cortes, J.E.; Mesa, R.; Hogan, W.; Redman, J.; Levy, R.; Vaddi, K.; et al. A phase I/II study of INCB018424, an oral, selective JAK inhibitor, in patients with primary myelofibrosis (PMF) and post polycythemia vera/essential thrombocythemia myelofibrosis (Post-PV/ET MF). J. Clin. Oncol. 2008, 26, 7004. [Google Scholar] [CrossRef]
  134. Sonbol, M.; Firwana, B.; Zarzour, A.; Morad, M.; Rana, V.; Tiu, R.V. Comprehensive review of JAK inhibitors in myeloproliferative neoplasms. Ther. Adv. Hematol. 2012, 4, 15–35. [Google Scholar] [CrossRef] [Green Version]
  135. Roth, J.; Nguyen, D.; Lawrence, D.; Kemp, B.; Carrasco, C.; Ferson, D.; Hong, W.; Komaki, R.; Lee, J.; Nesbitt, J.; et al. Retrovirus–mediated wild–type P53 gene transfer to tumors of patients with lung cancer. Nat. Med. 1996, 2, 985–991. [Google Scholar] [CrossRef] [PubMed]
  136. Lane, D.P.; Cheok, C.F.; Lain, S. p53-based Cancer Therapy. Cold Spring Harb. Perspect. Biol. 2010, 2, a001222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Zhang, X.-Z.; Lin, H.; Yang, X.-Y.; Wang, C.-H.; Zhang, M.-M.; Chen, P.-L.; Peng, Z.-H. Quality control of clinical-grade recombinant adenovirus used in gene therapy. Zhonghua Yi Xue Za Zhi 2004, 84, 849–852. [Google Scholar]
  138. Zhang, S.-W.; Xiao, S.-W.; Liu, C.-Q.; Sun, Y.; Su, X.; Li, D.-M.; Xu, G.; Zhu, G.-Y.; Xu, B. Recombinant adenovirus-p53 gene therapy combined with radiotherapy for head and neck squamous-cell carcinoma. Chinese J. Oncol. 2005, 27, 426–428. [Google Scholar]
  139. Zhang, W.-W.; Li, L.; Li, D.; Liu, J.; Li, X.; Li, W.; Xu, X.; Zhang, M.; Chandler, L.A.; Lin, H.; et al. The First Approved Gene Therapy Product for Cancer Ad-p53(Gendicine): 12 Years in the Clinic. Hum. Gene Ther. 2018, 29, 160–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Li, Y.; Li, L.J.; Wang, L.J.; Zhang, Z.; Gao, N.; Liang, C.Y.; Huang, Y.D.; Han, B. Selective intra-arterial infusion of rAd-p53 with chemotherapy for advanced oral cancer: A randomized clinical trial. BMC Med. 2014, 12, 16. [Google Scholar] [CrossRef] [Green Version]
  141. Nemunaitis, J.; Clayman, G.; Agarwala, S.S.; Hrushesky, W.; Wells, J.R.; Moore, C.; Hamm, J.; Yoo, G.; Baselga, J.; Murphy, B.A.; et al. Biomarkers Predict p53 Gene Therapy Efficacy in Recurrent Squamous Cell Carcinoma of the Head and Neck. Clin. Cancer Res. 2009, 15, 7719–7725. [Google Scholar] [CrossRef] [Green Version]
  142. Sobol, R.E.; Menander, K.B.; Chada, S.; Wiederhold, D.; Sellman, B.; Talbott, M.; Nemunaitis, J.J. Analysis Analysis of Adenoviral p53 Gene Therapy Clinical Trials in Recurrent Head and Neck Squamous Cell Car-cinoma. Front. Oncol. 2021, 11, 645745. [Google Scholar] [CrossRef] [PubMed]
  143. Senzer, N.; Nemunaitis, J.; Nemunaitis, M.; Lamont, J.; Gore, M.; Gabra, H.; Eeles, R.; Sodha, N.; Lynch, F.J.; Zumstein, L.A.; et al. p53 therapy in a patient with Li-Fraumeni syndrome. Mol. Cancer Ther. 2007, 6, 1478–1482. [Google Scholar] [CrossRef] [Green Version]
  144. Biaoxue, R.; Hui, P.; Wenlong, G.; Shuanying, Y. Evaluation of efficacy and safety for recombinant human adenovirus-p53 in the control of the malignant pleural effusions via thoracic perfusion. Sci. Rep. 2016, 6, 39355. [Google Scholar] [CrossRef]
  145. Arora, S.; Aggarwal, P.; Pathak, A.; Bhandari, R.; Duffoo, F.; Gulati, S.C. Molecular genetics of head and neck cancer (Review). Mol. Med. Rep. 2012, 6, 19–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Khuri, F.R.; Nemunaitis, J.; Ganly, I.; Arseneau, J.; Tannock, I.F.; Romel, L.; Gore, M.; Ironside, J.; MacDougall, R.H.; Heise, C.; et al. A controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cis-platin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat. Med. 2000, 6, 879–885. [Google Scholar] [CrossRef]
  147. Roh, J.L.; Kim, E.H.; Park, H.B.; Park, J.Y. The Hsp90 inhibitor 17-(allylamino)-17-demethoxy geldanamycin increases cisplatin antitumor activity by inducing p53-mediated apoptosis in head and neck cancer. Cell Death Dis. 2013, 4, e956. [Google Scholar] [CrossRef] [Green Version]
  148. Li, P.; Zhang, X.; Gu, L.; Zhou, J.; Deng, D. P16 methylation increases the sensitivity of cancer cells to the CDK4/6 inhibitor palbociclib. PLoS ONE 2019, 14, e0223084. [Google Scholar] [CrossRef] [Green Version]
  149. Gleich, L.L.; Gluckman, J.L.; Nemunaitis, J.; Suen, J.Y.; Hanna, E.; Wolf, G.T.; Coltrera, M.D.; Villaret, D.B.; Wagman, L.; Castro, D.; et al. Clinical experience with HLA-B7 plasmid DNA/lipid complex in advanced squamous cell carcinoma of the head and neck. Arch. Otolaryngol.-Head Neck Surg. 2001, 127, 775–779. [Google Scholar] [PubMed]
  150. Parfenov, M.; Pedamallu, C.S.; Gehlenborg, N.; Freeman, S.; Danilova, L.; Bristow, C.A.; Lee, S.; Hadjipanayis, A.G.; Ivanova, E.V.; Wilkerson, M.D.; et al. Characterization of HPV and host genome interactions in primary head and neck cancers. Proc. Natl. Acad. Sci. USA 2014, 111, 15544–15549. [Google Scholar] [CrossRef] [Green Version]
  151. Castellsagué, X.; Alemany, L.; Quer, M.; Halec, G.; Quirós, B.; Tous, S.; Clavero, O.; Alòs, L.; Biegner, T.; Szafarowski, T.; et al. HPV Involvement in Head and Neck Cancers: Comprehensive Assessment of Biomarkers in 3680 Patients. J. Natl. Cancer Inst. 2016, 108, djv403. [Google Scholar] [CrossRef]
  152. Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The molecular landscape of head and neck cancer. Nat. Rev. Cancer 2018, 18, 269–282. [Google Scholar] [CrossRef]
  153. Frazer, I.H. Prevention of cervical cancer through papillomavirus vaccination. Nat. Rev. Immunol. 2004, 4, 46–55. [Google Scholar] [CrossRef]
  154. Syrjänen, S. The role of human papillomavirus infection in head and neck cancers. Ann. Oncol. 2010, 21, vii243–vii245. [Google Scholar] [CrossRef]
  155. Lajer, C.B.; von Buchwald, C. The role of human papillomavirus in head and neck cancer. APMIS 2010, 118, 510–519. [Google Scholar] [CrossRef]
  156. McKeage, K.; Romanowski, B. AS04-Adjuvanted Human Papillomavirus (HPV) Types 16 and 18 Vaccine (Cervarix (R)) A Review of its Use in the Prevention of Premalignant Cervical Lesions and Cervical Cancer Causally Related to Certain On-cogenic HPV Types. Drugs 2011, 71, 465–488. [Google Scholar] [PubMed]
  157. Harper, D.M.; Franco, E.; Wheeler, C.M.; Moscicki, A.-B.; Romanowski, B.; Roteli-Martins, C.M.; Jenkins, D.; Schuind, A.; Clemens, S.A.C.; Dubin, G. Sustained efficacy up to 4·5 years of a bivalent L1 virus-like particle vaccine against human papillomavirus types 16 and 18: Follow-up from a randomised control trial. Lancet 2006, 367, 1247–1255. [Google Scholar] [CrossRef] [Green Version]
  158. Paavonen, J.; Jenkins, D.; Bosch, F.X. Efficacy of a prophylactic adjuvanted bivalent L1 virus-like-particle vaccine against infection with human papillomovirus types 16 and 18 in young women: An interim analysis of a phase III double-blind, ran-domised controlled trial. Lancet 2007, 370, 1414. [Google Scholar]
  159. Muñoz, N.; Bosch, F.X.; José, F.X.B.; Herrero, R.; Castellsagué, X.; Shah, K.V.; Snijders, P.J.; Meijer, C.J. Epidemiologic Classification of Human Papillomavirus Types Associated with Cervical Cancer. N. Engl. J. Med. 2003, 348, 518–527. [Google Scholar] [CrossRef] [Green Version]
  160. Hung, C.-F.; Ma, B.; Monie, A.; Tsen, S.-W.; Wu, T.-C. Therapeutic human papillomavirus vaccines: Current clinical trials and future directions. Expert Opin. Biol. Ther. 2008, 8, 421–439. [Google Scholar] [CrossRef]
  161. Peng, S.; Trimble, C.; He, L.; Tsai, Y.-C.; Lin, C.-T.; Boyd, D.A.K.; Pardoll, D.; Hung, C.-F.; Wu, T.-C. Characterization of HLA-A2-restricted HPV-16 E7-specific CD8+ T-cell immune responses induced by DNA vaccines in HLA-A2 transgenic mice. Gene Ther. 2005, 13, 67–77. [Google Scholar] [CrossRef] [PubMed]
  162. Lin, C.-T.; Tsai, Y.-C.; He, L.; Calizo, R.; Chou, H.-H.; Chang, T.-C.; Soong, Y.-K.; Hung, C.-F.; Lai, C.-H. A DNA Vaccine Encoding a Codon-Optimized Human Papillomavirus Type 16 E6 Gene Enhances CTL Response and Anti-tumor Activity. J. Biomed. Sci. 2006, 13, 481–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wu, A.; Zeng, Q.; Kang, T.H.; Peng, S.; Roosinovich, E.; Pai, S.I.; Hung, C.-F. Innovative DNA vaccine for human papillomavirus (HPV)-associated head and neck cancer. Gene Ther. 2010, 18, 304–312. [Google Scholar] [CrossRef] [Green Version]
  164. Karimi, H.; Soleimanjahi, H.; Abdoli, A.; Banijamali, R.S. Combination therapy using human papillomavirus L1/E6/E7 genes and archaeosome: A nanovaccine confer immuneadjuvanting effects to fight cervical cancer. Sci. Rep. 2020, 10, 5787. [Google Scholar] [CrossRef] [Green Version]
  165. van Poelgeest, M.I.E.; Welters, M.J.P.; van Esch, E.M.G.; Stynenbosch, L.F.M.; Kerpershoek, G.; van Meerten, E.L.V.; van den Hende, M.; Lowik, M.J.G.; Berends-van der Meer, D.M.A.; Fathers, L.M.; et al. HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J. Transl. Med. 2013, 11, 88. [Google Scholar] [CrossRef] [Green Version]
  166. Bonsack, M.; Mohan, N.; Öhlenschläger, K.; Lotsch, C.; Heinze, J.M.; Schmitt, J.; Hoppe, S.; Förster, J.; Blatnik, R.; Riemer, A.B. Application of individual HLA-binding prediction thresholds increases the detection of target cell sur-face-presented HPV16 E6- and E7-derived epitopes as a basis for therapeutic HPV vaccine design. J. Immunol. 2020, 204 (Suppl. 1), 169.8. [Google Scholar]
  167. Aggarwal, C.; Cohen, R.B.; Morrow, M.P.; Kraynyak, K.A.; Sylvester, A.J.; Cheung, J.; Dickerson, K.; Schulten, V.; Knoblock, D.; Gillespie, E. Immune Therapy Targeting E6/E7 Oncogenes of Human Paillomavirus Type 6 (HPV-6) Reduces or Eliminates the Need for Surgical Intervention in the Treatment of HPV-6 Associated Recurrent Respiratory Papillomatosis. Vaccines 2020, 8, 56. [Google Scholar] [CrossRef] [Green Version]
  168. Radulovic, S.; Brankovic-Magic, M.; Malisic, E.; Jankovic, R.; Dobricic, J.; Plesinac-Karapandzic, V.; Maciag, P.C.; Rothman, J. Therapeutic cancer vaccines in cervical cancer: Phase I study of Lovaxin-C. J. BUON 2009, 14, S165–S168. [Google Scholar]
  169. Huh, W.K.; Brady, W.E.; Fracasso, P.M.; Dizon, D.S.; Powell, M.A.; Monk, B.J.; Leath, C.A.; Landrum, L.M.; Tanner, E.J.; Crane, E.K.; et al. Phase II study of axalimogene filolisbac (ADXS-HPV) for platinum-refractory cervical carcinoma: An NRG oncology/gynecologic oncology group study. Gynecol. Oncol. 2020, 158, 562–569. [Google Scholar] [CrossRef]
  170. Otto, T.; Sicinski, P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 2017, 17, 93–115. [Google Scholar] [CrossRef] [Green Version]
  171. van Harten, A.M.; Buijze, M.; van der Mast, R.; Rooimans, M.A.; Martens-de Kemp, S.R.; Bachas, C.; Brink, A.; Stigter-van Walsum, M.; Wolthuis, R.M.F.; Brakenhoff, R.H. Targeting the cell cycle in head and neck cancer by Chk1 inhibition: A novel concept of bimodal cell death. Oncogenesis 2019, 8, 38. [Google Scholar] [CrossRef]
  172. Gomes, M.; Salazar, N.P. Chemotherapy: Principles in practice—A case study of the Philippines. Soc. Sci. Med. 1990, 30, 789–796. [Google Scholar] [CrossRef]
  173. Xiang, M.; Colevas, A.D.; Holsinger, F.C.; Le, Q.-T.X.; Beadle, B.M. Survival After Definitive Chemoradiotherapy with Concurrent Cisplatin or Carboplatin for Head and Neck Cancer. J. Natl. Compr. Cancer Netw. 2019, 17, 1065–1073. [Google Scholar] [CrossRef]
  174. Go, R.S.; Adjei, A.A. Review of the Comparative Pharmacology and Clinical Activity of Cisplatin and Carboplatin. J. Clin. Oncol. 1999, 17, 409. [Google Scholar] [CrossRef]
  175. Mesía, R.; Rivera, F.; Kawecki, A.; Rottey, S.; Hitt, R.; Kienzer, H.; Cupissol, D.; De Raucourt, D.; Benasso, M.; Koralewski, P.; et al. Quality of life of patients receiving platinum-based chemotherapy plus cetuximab first line for recurrent and/or metastatic squamous cell carcinoma of the head and neck. Ann. Oncol. 2010, 21, 1967–1973. [Google Scholar] [CrossRef]
  176. Hecht, M.; Hahn, D.; Wolber, P.; Hautmann, M.G.; Reichert, D.; Weniger, S.; Belka, C.; Bergmann, T.; Göhler, T.; Welslau, M.; et al. Treatment response lowers tumor symptom burden in recurrent and/or metastatic head and neck cancer. BMC Cancer 2020, 20, 1–11. [Google Scholar] [CrossRef]
  177. Schiff, P.; Fant, J.; Horwitz, S.B. Promotion of microtubule assembly in vitro by taxol. Nature 1979, 277, 665–667. [Google Scholar] [CrossRef] [PubMed]
  178. Tahara, M.; Minami, H.; Hasegawa, Y.; Tomita, K.; Watanabe, A.; Nibu, K.; Fujii, M.; Onozawa, Y.; Kurono, Y.; Sagae, D.; et al. Weekly paclitaxel in patients with recurrent or metastatic head and neck cancer. Cancer Chemother. Pharmacol. 2010, 68, 769–776. [Google Scholar] [CrossRef] [PubMed]
  179. Psyrri, A.; Gkotzamanidou, M.; Papaxoinis, G.; Krikoni, L.; Economopoulou, P.; Kotsantis, I.; Anastasiou, M.; Souliotis, V. The DNA damage response network in the treatment of head and neck squamous cell carcinoma. ESMO Open 2021, 6, 100075. [Google Scholar] [CrossRef] [PubMed]
  180. Chaudhuri, A.R.; Nussenzweig, A.R.C.A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 610–621. [Google Scholar] [CrossRef]
  181. Ledermann, J.A.; Pujade-Lauraine, E. Olaparib as maintenance treatment for patients with platinum-sensitive relapsed ovarian cancer. Ther. Adv. Med. Oncol. 2019, 11. [Google Scholar] [CrossRef]
  182. Fong, P.C.; Boss, D.S.; Yap, T.A.; Tutt, A.; Wu, P.; Mergui-Roelvink, M.; Mortimer, P.; Swaisland, H.; Lau, A.; O’Connor, M.J.; et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 2009, 361, 123–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Farago, A.F.; Yeap, B.Y.; Stanzione, M.; Hung, Y.P.; Heist, R.S.; Marcoux, J.P.; Zhong, J.; Rangachari, D.; Barbie, D.A.; Phat, S.; et al. Combination Olaparib and Temozolomide in Relapsed Small-Cell Lung Cancer. Cancer Discov. 2019, 9, 1372–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Mouw, K.W.; Goldberg, M.S.; Konstantinopoulos, P.A.; D’Andrea, A.D. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discov. 2017, 7, 675–693. [Google Scholar] [CrossRef] [Green Version]
  185. Karam, S.D.; Reddy, K.; Blatchford, P.J.; Waxweiler, T.; DeLouize, A.M.; Oweida, A.; Somerset, H.; Marshall, C.; Young, C.; Davies, K.D.; et al. Final Report of a Phase I Trial of Olaparib with Cetuximab and Radiation for Heavy Smoker Patients with Locally Advanced Head and Neck Cancer. Clin. Cancer Res. 2018, 24, 4949–4959. [Google Scholar] [CrossRef] [Green Version]
  186. Brown, J.M.; William, W.R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 2004, 4, 437–447. [Google Scholar] [CrossRef] [PubMed]
  187. Santilli, G.; Lamorte, G.; Carlessi, L.; Ferrari, D.; Nodari, L.R.; Binda, E.; Delia, D.; Vescovi, A.L.; De Filippis, L. Mild Hypoxia Enhances Proliferation and Multipotency of Human Neural Stem Cells. PLoS ONE 2010, 5, e8575. [Google Scholar] [CrossRef]
  188. Carmeliet, P.; Dor, Y.; Herbert, J.M.; Fukumura, D.; Brusselmans, K.; Dewerchin, M.; Neeman, M.; Bono, F.; Abramovitch, R.; Maxwell, P.; et al. Role of HIF-1 alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998, 394, 485–490. [Google Scholar] [CrossRef] [Green Version]
  189. Koumenis, C.; Alarcon, R.; Hammond, E.; Sutphin, P.; Hoffman, W.; Murphy, M.; Derr, J.; Taya, Y.; Lowe, S.W.; Kastan, M.; et al. Regulation of p53 by Hypoxia: Dissociation of Transcriptional Repression and Apoptosis from p53-Dependent Transactivation. Mol. Cell. Biol. 2001, 21, 1297–1310. [Google Scholar] [CrossRef] [Green Version]
  190. Comerford, K.M.; Wallace, T.J.; Karhausen, J.; Louis, N.; Montalto, M.C.; Colgan, S.P. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res. 2002, 62, 3387–3394. [Google Scholar]
  191. Wartenberg, M.; Ling, F.C.; Müschen, M.; Klein, F.; Acker, H.; Gassmann, M.; Petrat, K.; Pütz, V.; Hescheler, J.; Sauer, H. Regulation of the multidrug resistance transporter P-glycoprotein in multicellular tumor spheroids by hypoxia-inducible factor-1 and reactive oxygen species. FASEB J. 2003, 17, 1–22. [Google Scholar] [CrossRef]
  192. Xie, J.; Li, D.-W.; Chen, X.-W.; Wang, F.; Dong, P. Expression and significance of hypoxia-inducible factor-1α and MDR1/P-glycoprotein in laryngeal carcinoma tissue and hypoxic Hep-2 cells. Oncol. Lett. 2013, 6, 232–238. [Google Scholar] [CrossRef]
  193. Bristow, R.G.; Hill, R.P. Hypoxia, DNA repair and genetic instability. Nat. Rev. Cancer 2008, 8, 180–192. [Google Scholar] [CrossRef] [PubMed]
  194. Overgaard, J. Hypoxic Radiosensitization: Adored and Ignored. J. Clin. Oncol. 2007, 25, 4066–4074. [Google Scholar] [CrossRef] [Green Version]
  195. Overgaard, J. Clinical evaluation of nitroimidazoles as modifiers of hypoxia in solid tumors. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 1994, 6, 509–518. [Google Scholar]
  196. Mahy, P.; Geets, X.; Lonneux, M.; Leveque, P.; Christian, N.; De Bast, M.; Gillart, J.; Labar, D.; Lee, J.; Gregoire, V. Determination of tumour hypoxia with [F-18]EF3 in patients with head and neck tumours: A phase I study to assess the tracer pharmacokinetics, biodistribution and metabolism. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1282–1289. [Google Scholar] [CrossRef]
  197. Troost, E.G.; Schinagl, D.A.; Bussink, J.; Boerman, O.C.; van der Kogel, A.J.; Oyen, W.J.; Kaanders, J.H. Innovations in Radiotherapy Planning of Head and Neck Cancers: Role of PET. J. Nucl. Med. 2009, 51, 66–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Halmos, G.B.; de Bruin, L.B.; Langendijk, J.A.; van der Laan, B.F.A.M.; Pruim, J.; Steenbakkers, R.J.H.M. Head and Neck Tumor Hypoxia Imaging by F-18-Fluoroazomycin-arabinoside (F-18-FAZA)-PET A Re-view. Clin. Nucl. Med. 2014, 39, 44–48. [Google Scholar] [CrossRef]
  199. Henk, J.M. Late Results of a Trial of Hyperbaric-Oxygen and Radiotherapy in Head and Neck-Cancer—A Rationale for Hy-poxic Cell Sensitizers. Int. J. Radiat. Oncol. Biol. Phys. 1986, 12, 1339–1341. [Google Scholar] [CrossRef]
  200. Abe, M.; Shioyama, Y.; Terashima, K.; Matsuo, M.; Hara, I.; Uehara, S. Successful hyperbaric oxygen therapy for laryngeal radionecrosis after chemoradiotherapy for mesopharyngeal cancer: Case report and literature review. Jpn. J. Radiol. 2012, 30, 340–344. [Google Scholar] [CrossRef]
  201. Ferguson, B.J.; Hudson, W.R.; Farmer, J.C. Hyperbaric Oxygen Therapy for Laryngeal Radionecrosis. Ann. Otol. Rhinol. Laryngol. 1987, 96, 1–6. [Google Scholar] [CrossRef] [PubMed]
  202. Haffty, B.G.; Hurley, R.; Peters, L.J. Radiation therapy with hyperbaric oxygen at 4 atmospheres pressure in the management of squamous cell carcinoma of the head and neck: Results of a randomized clinical trial. Cancer J. Sci. Am. 1999, 5, 341–347. [Google Scholar]
  203. Schoen, P.J.; Raghoebar, G.M.; Bouma, J.; Reintsema, H.; Vissink, A.; Sterk, W.; Roodenburg, J.L. Rehabilitation of oral function in head and neck cancer patients after radiotherapy with implant-retained dentures: Effects of hyperbaric oxygen therapy. Oral Oncol. 2007, 43, 379–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Barkhuysen, R.; Gerlach, N.; Sterk, W.; Merkx, T. O92 The effect of hyperbaric oxygen therapy on quality of life in head and neck cancer patients treated with radiotherapy: A pilot study. Oral Oncol. Suppl. 2007, 2, 86–87. [Google Scholar] [CrossRef]
  205. Bernier, J.; Denekamp, J.; Rojas, A.; Minatel, E.; Horiot, J.C.; Hamers, H.; Antognoni, P.; Dahl, O.; Richaud, P.; van Glabbeke, M.; et al. ARCON: Accelerated radiotherapy with carbogen and nicotinamide in head and neck squamous cell carci-nomas. The experience of the Cooperative Group of Radiotherapy of the European Organization for Research and Treatment of Cancer (EORTC). Radiother. Oncol. 2000, 55, 111–119. [Google Scholar] [CrossRef]
  206. Kaanders, J.; Pop, L.; Marres, H.; Van Der Maazen, R.; Van Der Kogel, A.; Van Daal, W. Radiotherapy with carbogen breathing and nicotinamide in head and neck cancer: Feasibility and toxicity. Radiother. Oncol. 1995, 37, 190–198. [Google Scholar] [CrossRef] [Green Version]
  207. Rischin, D.; Peters, L.J.; O’Sullivan, B.; Giralt, J.; Fisher, R.; Yuen, K.; Trotti, A.; Bernier, J.; Bourhis, J.; Ringash, J.; et al. Tirapazamine, Cisplatin, and Radiation Versus Cisplatin and Radiation for Advanced Squamous Cell Car-cinoma of the Head and Neck (TROG 02.02, HeadSTART): A Phase III Trial of the Trans-Tasman Radiation Oncology Group. J. Clin. Oncol. 2010, 28, 2989–2995. [Google Scholar] [CrossRef]
  208. Cohen, E.E.; Rosine, D.; Haraf, D.J.; Loh, E.; Shen, L.; Lusinchi, A.; Vokes, E.E.; Bourhis, J. Phase I trial of tirapazamine, cisplatin, and concurrent accelerated boost reirradiation in patients with recurrent head and neck cancer. Int. J. Radiat. Oncol. 2007, 67, 678–684. [Google Scholar] [CrossRef]
  209. Toustrup, K.; Sørensen, B.S.; Alsner, J.; Overgaard, J. Hypoxia Gene Expression Signatures as Prognostic and Predictive Markers in Head and Neck Radiotherapy. Semin. Radiat. Oncol. 2012, 22, 119–127. [Google Scholar] [CrossRef]
  210. Toustrup, K.; Sørensen, B.S.; Nordsmark, M.; Busk, M.; Wiuf, C.; Alsner, J.; Overgaard, J. Development of a Hypoxia Gene Expression Classifier with Predictive Impact for Hypoxic Modification of Radiotherapy in Head and Neck Cancer. Cancer Res. 2011, 71, 5923–5931. [Google Scholar] [CrossRef] [Green Version]
  211. Eustace, A.; Mani, N.; Span, P.; Irlam, J.J.; Taylor, J.; Betts, G.N.; Denley, H.; Miller, C.; Homer, J.J.; Rojas, A.M.; et al. A 26-Gene Hypoxia Signature Predicts Benefit from Hypoxia-Modifying Therapy in Laryngeal Cancer but Not Bladder Cancer. Clin. Cancer Res. 2013, 19, 4879–4888. [Google Scholar] [CrossRef] [Green Version]
  212. von der Grun, J.; Rodel, F.; Brandts, C.; Fokas, E.; Guckenberger, M.; Rodel, C.; Balermpas, P. Targeted Therapies and Immune-Checkpoint Inhibition in Head and Neck Squamous Cell Carcinoma: Where Do We Stand Today and Where to Go? Cancer 2019, 11, 472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Argiris, A.; Li, S.; Savvides, P.; Ohr, J.P.; Gilbert, J.; Levine, M.A.; Chakravarti, A.; Jr, M.H.; Saba, N.F.; Ikpeazu, C.V.; et al. Phase III Randomized Trial of Chemotherapy with or Without Bevacizumab in Patients with Recurrent or Metastatic Head and Neck Cancer. J. Clin. Oncol. 2019, 37, 3266–3274. [Google Scholar] [CrossRef] [PubMed]
  214. Cohen, E.E.; Davis, D.W.; Karrison, T.G.; Seiwert, T.Y.; Wong, S.J.; Nattam, S.; Kozloff, M.F.; Clark, J.I.; Yan, D.-H.; Liu, W.; et al. Erlotinib and bevacizumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck: A phase I/II study. Lancet Oncol. 2009, 10, 247–257. [Google Scholar] [CrossRef] [Green Version]
  215. Argiris, A.; Karamouzis, M.V.; Gooding, W.E.; Branstetter, B.F.; Zhong, S.; Raez, L.E.; Savvides, P.; Romkes, M. Phase II Trial of Pemetrexed and Bevacizumab in Patients with Recurrent or Metastatic Head and Neck Cancer. J. Clin. Oncol. 2011, 29, 1140–1145. [Google Scholar] [CrossRef] [Green Version]
  216. Argiris, A.; Kotsakis, A.P.; Hoang, T.; Worden, F.P.; Savvides, P.; Gibson, M.K.; Gyanchandani, R.; Blumenschein, G.R.; Chen, H.X.; Grandis, J.R.; et al. Cetuximab and bevacizumab: Preclinical data and phase II trial in recurrent or metastatic squamous cell carcinoma of the head and neck. Ann. Oncol. 2012, 24, 220–225. [Google Scholar] [CrossRef]
  217. Fury, M.G.; Lee, N.Y.; Sherman, E.; Lisa, D.; Kelly, K.; Lipson, B.; Carlson, D.; Stambuk, H.; Haque, S.; Shen, R.; et al. A phase 2 study of bevacizumab with cisplatin plus intensity-modulated radiation therapy for stage III/IVB head and neck squamous cell cancer. Cancer 2012, 118, 5008–5014. [Google Scholar] [CrossRef] [Green Version]
  218. Leighl, N.B.; Bennouna, J.; Yi, J.; Moore, N.; Hambleton, J.; Hurwitz, H. Bleeding events in bevacizumab-treated cancer patients who received full-dose anticoagulation and re-mained on study. Br. J. Cancer 2011, 104, 413–418. [Google Scholar] [CrossRef]
  219. Wilhelm, S.M.; Adnane, L.; Newell, P.; Villanueva, A.; Llovet, J.M.; Lynch, M. Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF re-ceptor tyrosine kinase signaling. Mol. Cancer Ther. 2008, 7, 3129–3140. [Google Scholar] [CrossRef] [Green Version]
  220. Tahara, M.; Kiyota, N.; Yokota, T.; Hasegawa, Y.; Muro, K.; Takahashi, S.; Onoe, T.; Homma, A.; Taguchi, J.; Suzuki, M.; et al. Phase II trial of combination treatment with paclitaxel, carboplatin and cetuximab (PCE) as first-line treatment in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (CSPOR-HN02). Ann. Oncol. 2018, 29, 1004–1009. [Google Scholar] [CrossRef]
  221. Gilbert, J.; Schell, M.J.; Zhao, X.; Murphy, B.; Tanvetyanon, T.; Leon, M.E.; Hayes, D.N.; Haigentz, M.; Saba, N.; Nieva, J.; et al. A randomized phase II efficacy and correlative studies of cetuximab with or without sorafenib in recurrent and/or metastatic head and neck squamous cell carcinoma. Oral Oncol. 2015, 51, 376–382. [Google Scholar] [CrossRef] [Green Version]
  222. Limaye, S.; Riley, S.; Zhao, S.; O’Neill, A.; Posner, M.; Adkins, D.; Jaffa, Z.; Clark, J.; Haddad, R. A randomized phase II study of docetaxel with or without vandetanib in recurrent or metastatic squamous cell carcinoma of head and neck (SCCHN). Oral Oncol. 2013, 49, 835–841. [Google Scholar] [CrossRef]
  223. Roskoski, R. Sunitinib: A VEGF and PDGF receptor protein kinase and angiogenesis inhibitor. Biochem. Biophys. Res. Commun. 2007, 356, 323–328. [Google Scholar] [CrossRef]
  224. Choong, N.W.; Cohen, E.E.; Kozloff, M.F.; Taber, D.; Iii, J.L.W.; Hu, S.; Ivy, S.P.; Nichols, K.; Dekker, A.; Vokes, E.E. Phase II trial of sunitinib in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (SCCHN). J. Clin. Oncol. 2008, 26, 6064. [Google Scholar] [CrossRef]
  225. Machiels, J.-P.H.; Henry, S.; Zanetta, S.; Kaminsky, M.-C.; Michoux, N.; Rommel, D.; Schmitz, S.; Bompas, E.; Dillies, A.-F.; Faivre, S.; et al. Phase II Study of Sunitinib in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: GORTEC 2006-01. J. Clin. Oncol. 2010, 28, 21–28. [Google Scholar] [CrossRef] [PubMed]
  226. Klein, J.D.; Christopoulos, A.; Ahn, S.M.; Gooding, W.E.; Grandis, J.R.; Kim, S. Antitumor effect of vandetanib through EGFR inhibition in head and neck squamous cell carcinoma. J. Sci. Spec. Head Neck 2012, 34, 1269–1276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Jost, L.M.; Gschwind, H.-P.; Jalava, T.; Wang, Y.; Guenther, C.; Souppart, C.; Rottmann, A.; Denner, K.; Waldmeier, F.; Gross, G.; et al. Metabolism and Disposition of Vatalanib (PTK787/ZK-222584) in Cancer Patients. Drug Metab. Dispos. 2006, 34, 1817–1828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Gauler, T.C.; Besse, B.; Mauguen, A.; Meric, J.B.; Gounant, V.; Fischer, B.; Overbeck, T.R.; Krissel, H.; Laurent, D.; Tiainen, M.; et al. Phase II trial of PTK787/ZK 222584 (vatalanib) administered orally once-daily or in two divided daily doses as second-line monotherapy in relapsed or progressing patients with stage IIIB/IV non-small-cell lung cancer (NSCLC). Ann. Oncol. 2012, 23, 678–687. [Google Scholar] [CrossRef] [PubMed]
  229. Miyazawa, M.; Dong, Z.; Zhang, Z.; Neiva, K.G.; Cordeiro, M.M.; Oliveira, D.T.; Nor, J.E. Effect of PTK/ZK on the Angiogenic Switch in Head and Neck Tumors. J. Dent. Res. 2008, 87, 1166–1171. [Google Scholar] [CrossRef] [Green Version]
  230. Swiecicki, P.L.; Zhao, L.; Belile, E.; Sacco, A.G.; Chepeha, D.; Dobrosotskaya, I.Y.; Spector, M.; Shuman, A.G.; Malloy, K.M.; Moyer, J.S.; et al. A phase II study evaluating axitinib in patients with unresectable, recurrent or metastatic head and neck cancer. Investig. New Drugs 2015, 33, 1248–1256. [Google Scholar] [CrossRef]
  231. Mito, I.; Takahashi, H.; Kawabata-Iwakawa, R.; Ida, S.; Tada, H.; Chikamatsu, K. Comprehensive analysis of immune cell enrichment in the tumor microenvironment of head and neck squamous cell carcinoma. Sci. Rep. 2021, 11, 16134. [Google Scholar] [CrossRef]
  232. Vaddepally, R.K.; Kharel, P.; Pandey, R.; Garje, R.; Chandra, A.B. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers 2020, 12, 738. [Google Scholar] [CrossRef] [Green Version]
  233. Ritprajak, P.; Azuma, M. Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol. 2015, 51, 221–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Ferris, R.; Haddad, R.; Even, C.; Tahara, M.; Dvorkin, M.; Ciuleanu, T.E.; Clement, P.; Mesia, R.; Kutukova, S.; Zholudeva, L.; et al. Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann. Oncol. 2020, 31, 942–950. [Google Scholar] [CrossRef]
  235. Hwang, M.; Seiwert, T.Y. Are taxanes the future for head and neck cancer? Pragmatism in the immunotherapy era. Lancet Oncol. 2021, 22, 413–415. [Google Scholar] [CrossRef]
  236. Siu, L.L.; Even, C.; Mesía, R.; Remenar, E.; Daste, A.; Delord, J.-P.; Krauss, J.; Saba, N.F.; Nabell, L.; Ready, N.E.; et al. Safety and Efficacy of Durvalumab with or Without Tremelimumab in Patients with PD-L1–Low/Negative Recurrent or Metastatic HNSCC. JAMA Oncol. 2019, 5, 195–203. [Google Scholar] [CrossRef]
  237. Polverini, P.; D’Silva, N.; Lei, Y. Precision Therapy of Head and Neck Squamous Cell Carcinoma. J. Dent. Res. 2018, 97, 614–621. [Google Scholar] [CrossRef]
  238. Ferris, R.L.; Blumenschein, G.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  239. Mei, Z.; Huang, J.; Qiao, B.; Lam, A.K.-Y. Immune checkpoint pathways in immunotherapy for head and neck squamous cell carcinoma. Int. J. Oral Sci. 2020, 12, 1–9. [Google Scholar] [CrossRef] [PubMed]
  240. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [Green Version]
  241. Hodi, F.S.; Sileni, V.C.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
  242. Sheng, I.Y.; Ornstein, M.C. Ipilimumab and Nivolumab as First-Line Treatment of Patients with Renal Cell Carcinoma: The Evidence to Date. Cancer Manag. Res. 2020, 12, 4871–4881. [Google Scholar] [CrossRef] [PubMed]
  243. Goldberg, K.B.; Blumenthal, G.M.; McKee, A.; Pazdur, R. The FDA Oncology Center of Excellence and precision medicine. Exp. Biol. Med. 2017, 243, 308–312. [Google Scholar] [CrossRef] [PubMed]
  244. Elbers, J.B.; Al-Mamgani, A.; Tesseslaar, M.E.; Brekel, M.W.V.D.; Lange, C.A.; van der Wal, J.E.; Verheij, M.; Zuur, C.L.; de Boer, J.P. Immuno-radiotherapy with cetuximab and avelumab for advanced stage head and neck squamous cell carcinoma: Results from a phase-I trial. Radiother. Oncol. 2019, 142, 79–84. [Google Scholar] [CrossRef]
Table 1. Selected drugs and compounds currently available for treatment of head and neck squamous cell carcinoma.
Table 1. Selected drugs and compounds currently available for treatment of head and neck squamous cell carcinoma.
Drug (Brand Name)Mechanism of ActionFDA Approved for Head and Neck CancerFDA Approval Year/Progression PhaseKey ClinicalTrials.gov Identifiers
Docetaxel (Taxotere)Microtubule stimulantYes2006NCT00003888
Cetuximab (Erbitux)EGFR mAbYes2006NCT00004227
Cisplatin in combination with 5- Fluorouracil and CetuximabDNA synthesis and mRNA transcription inhibitor with EGFR mAbYes2011NCT00122460
Pembrolizumab (Keytruda)PD-1 inhibitorYes2017NCT02358031
NCT02252042
Nivolumab (Opdivo)PD-1 inhibitorYes2016NCT02105636
Zalutumumab (HuMax-EGFR)EGFR mAb-Phase IIINCT00382031
Panitumumab (Vectibix)EGFR mAb-Phase IIINCT00460265
NCT00820248
NimotuzumabEGFR mAb-Phase IIINCT00957086
Gefitinib (Iressa)EGFR TKI-Phase IIINCT00088907
NCT00206219
Erlotinib (Tarceva)EGFR TKI-Phase IINCT01515137
Lapatinib (Tykerb/Tyverb)EGFR/HER2 TKI-Phase IINCT00098631
NCT01044433
NCT00424255
Foretinibc-MET and VEGFR-2 inhibitor-Phase IINCT00725764
Bevacizumab (Avastin)Anti-VEGF mAb-Phase IIINCT00588770
Sunitinib (Sutent)Multi-target TKI (PDGFR, VEGFR, c-KIT)-Phase INCT00437372
Sorafenib (Nexavar)Multi-target TKI (VEGFR, PDGFR, RAF)-Phase IINCT00096512
NCT00494182
NCT00939627
Buparlisib aka BKM120PI3K inhibitor-Phase IINCT01527877
NCT01737450
NCT01816984
NCT02113878
Alpelisib aka BYL719 PI3K inhibitor-Phase INCT02282371
Sonolisib aka PX-866PI3K inhibitor-Phase IINCT01252628
NCT01204099
Temsirolimus (Torisel)mTOR kinase inhibitor-Phase IINCT01172769
NCT01256385
NCT01016769
Everolimus (Afinitor)mTOR kinase inhibitor-Phase IINCT00942734
Tipifarnib
(Zarnestra)
RAS inhibitor-Phase IINCT02383927
NCT03719690
Ad5RSV-p53 (Gendicine)p53 stimulant-Phase II/III-
Ad5CMV-p53 (Advexin)p53 stimulant-Phase II/IIINCT00041613
NCT00041626 NCT03544723
Lontucirev aka ONYX-015p53 stimulant-Phase IINCT00006106
Olaparib (Lynparza)PARP inhibitor-Phase IINCT02882308
NCT04643379
mAB: monoclonal antibody. TKI: tyrosine kinase inhibitor. FDA: U.S Food and Drug Administration.
Table 2. Immune checkpoint inhibitors.
Table 2. Immune checkpoint inhibitors.
Drug
(Brand Name)
TargetClassFirst FDA Approval Cancer Type
Nivolumab
(Opdivo)
PD-1IgG42014melanoma, lung cancer, renal cell carcinoma (RCC), Hodgkin lymphoma, head and neck cancer (HNC), colon cancer, and liver cancer
Pembrolizumab (Keytruda)PD-1IgG4-κ2017melanoma, lung cancer, head and neck cancer (HNC), Hodgkin lymphoma, stomach cancer and colorectal cancer
Atezolizumab (Tecentriq)PD-L1IgG12016urothelial carcinoma, non-small-cell lung cancer (NSCLC), triple-negative breast cancer (TNBC), small cell lung cancer (SCLC), and hepatocellular carcinoma (HCC)
Avelumab (Bavencio)PD-L1IgG12017Merkel-cell carcinoma (MCC)
Durvalumab (Imfinzi)PD-L1IgG12017bladder and lung cancer
Cemiplimab (Libtayo)PD-L1IgG12018squamous cell skin cancer
Ipilimumab
(Yervoy)
CTLA-4IgG12011melanoma, RCC, dMMR metastatic colorectal cancer
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kordbacheh, F.; Farah, C.S. Current and Emerging Molecular Therapies for Head and Neck Squamous Cell Carcinoma. Cancers 2021, 13, 5471. https://doi.org/10.3390/cancers13215471

AMA Style

Kordbacheh F, Farah CS. Current and Emerging Molecular Therapies for Head and Neck Squamous Cell Carcinoma. Cancers. 2021; 13(21):5471. https://doi.org/10.3390/cancers13215471

Chicago/Turabian Style

Kordbacheh, Farzaneh, and Camile S. Farah. 2021. "Current and Emerging Molecular Therapies for Head and Neck Squamous Cell Carcinoma" Cancers 13, no. 21: 5471. https://doi.org/10.3390/cancers13215471

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop