Highlights
-
•
Tobacco consumption and infection with high-risk human papillomavirus (HPV) are the major risk factors of the Head and neck squamous cell carcinoma (HNSCC).
-
•
In current clinical trials the three main therapeutic approaches being employed for the treatment of HNSCC include Chemotherapy, Molecular Targeted Therapy and Immunotherapy.
-
•
Immunotherapy has emerged as the most promising treatment of choice for the treatment of HNSCC. Currently, three monoclonal antibodies viz. cetuximab, pembrolizumab, and nivolumab have been approved by USFDA for the same.
Keywords: HNSCC, Immunotherapy, Clinical trial, PD-1, EGFR, Immune checkpoints
Abbreviations: HNSCC, Head and Neck Squamous Cell Carcinoma; HPV, Human Papillomaviruses; OS, Overall Survival; PFS, Progression-Free Survival; 5-FU, Fluorouracil; R/M, Recurrent/Metastatic; EGFR, Epidermal growth factor receptor; TKI, Tyrosine kinase inhibitor; CRR, Complete response rate; VEGF, Vascular endothelial growth factor; PDGFR, Platelet-derived growth factor receptor; CDK, Cyclin dependent kinase; ALK1, Activin receptor-like kinase-1; STAT3, Signal transducer and activator of transcription 3; IDO-1, Indoleamine 2, 3-dioxygenase 1; PD-1, Programmed cell death protein 1; PDL-1, Programmed death-ligand 1; CTLA4, Cytotoxic T lymphocyte antigen 4; LAG-3, Lymphocyte activated gene-3; NK, Natural killer; APCs, Antigen presenting cells; TIM-3, T cell immunoglobulin mucin-3; VISTA, V-domain immunoglobulin suppressor of T cell activation
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common non-skin cancer with a tobacco consumption and infection with high-risk human papillomavirus (HPV) being major risk factors. Despite advances in numerous therapy modalities, survival rates for HNSCC have not improved considerably; a vast number of clinical outcomes have demonstrated that a combination strategy (the most well-known docetaxel, cisplatin, and 5-fluorouracil) is the most effective treatment choice. Immunotherapy that targets immunological checkpoints is being tested in a number of clinical trials, either alone or in conjunction with chemotherapeutic or targeted therapeutic drugs. Various monoclonal antibodies, such as cetuximab and bevacizumab, which target the EGFR and VEGFR, respectively, as well as other signaling pathway inhibitors, such as temsirolimus and rapamycin, are also being studied for the treatment of HNSCC. We have reviewed the primary targets in active clinical studies in this study, with a particular focus on the medications and drug targets used.
Graphical abstract
Introduction
Squamous cell carcinoma of the head and neck (HNSCC) is one of the most common malignancies in the world, accounting for over 90% of head and neck tumors in Asia and Europe [85]. Tobacco usage, alcohol consumption, and infection with high-risk Human Papillomaviruses (HPV) are the main risk factors for HNSCC [86]. Furthermore, hypercalcemia and comorbidities are associated with poor clinical outcomes, increased relapse rates, and shorter survival times [87]. HNSCC is a kind of human cancer that is responsive to treatment. Despite the use of numerous treatment techniques in HNSCC patients, such as chemoradiotherapy, targeted therapy, and immunotherapy, overall clinical outcomes have not conclusively indicated a potential therapeutic benefit.
Furthermore, individuals with metastasis are usually incurable, and only a few therapy approaches have been shown to improve overall survival (OS) or progression-free survival (PFS) [88]. Several factors other than treatment appear to influence OS and time to progression, according to multivariable study [1]. Chemotherapy is primarily assessed using several prognostic markers to determine the clinical result; however, prior treatment (chemo/radiotherapy, surgery, or other) and stage of cancer are the most important factors influencing the response [87]. Despite these restrictions, a number of pharmacological compounds, particularly monoclonal antibodies, have showed great promise in the treatment of HNSCC, and many are currently undergoing clinical studies [89].
A large number of appropriately powered randomized and controlled clinical studies have established the therapeutic effects of numerous medications in various treatment cohorts such as chemotherapy, radiation, immunotherapy, and targeted therapy [[90], [91]]. So far, the most promising treatment is a combination of docetaxel, cisplatin, and 5-FU. However, new advancements in immunotherapy and targeted therapy suggest that monoclonal antibody-based medicines such as nivolumab, pembrolizumab, cetuximab, panitumumab, zalutumumab, and nimotuzumab could be used in the future [89]. We provide a synopsis of the primary therapeutic techniques used in clinical trials around the world, as well as their application in the treatment of HNSCC.
Recent clinical trials of HNSCC
A large number of clinical trials in patients with HNSCC have been undertaken around the world. In the clinicaltrials.gov database, 1266 clinical studies with the condition/disease 'HNSCC' and a'start date on or before 31 January 2022′ have been found. A total of 393 studies were completed out of 1266 trials registered, while 590 studies were still recruiting with the following recruitment statuses: recruiting (387), active–not recruiting (157), not yet recruiting (42), and enrolling by invitation (4). (Fig. 1). Notably, the majority of completed clinical trials have yet to be published. Docetaxel, cisplatin, and 5-fluorouracil (5-FU) have been identified as the most often used medications in clinical trials around the world [89]. Furthermore, the thorough examination of completed trials led to a greater understanding of the numerous therapeutic agents employed in chemotherapy, immunotherapy, and targeted therapy. Table 1 summarizes the results of selected clinical trials examining the efficacy of immunotherapy and molecular targeted treatments.
Table 1.
Target | Drug | Comparison treatment | Clinicaltrials.gov Identifier | Phase | Status | Treatment setting |
---|---|---|---|---|---|---|
PD-1 | Nivolumab | Therapy of investigator's choice (cetuximab, methotrexate, docetaxel) | NCT02105636 (CheckMate 141) | 3 | Active, not recruiting | As monotherapy in R/M platinum-refractory HNSCC |
Pembrolizumab | – | NCT01848834 (KEYNOTE-012) | 1 | Completed | Monotherapy for the treatment of advanced solid tumors | |
Sintilimab, nimotuzumab (anti-EGFR) combined with chemotherapy | – | NCT04882462 | 2 | Not yet recruiting | R/M HNSCC | |
CTLA-4 | Nivolumab, ipilimumab | Extreme study regimen (cetuximab +cisplatin/carboplatin + fluorouracil) | NCT02741570 (CheckMate 651) | 3 | Active, not recruiting | First line therapy of R/M HNSCC |
Nivolumab alone or in combination with ipilimumab | Surgical resection + adjuvant radio(-chemo) therapy | NCT03700905 (IMSTAR-HN) | 3 | Recruiting | In surgical resectable HNSCC after adjuvant therapy | |
LAG-3 | Relatlimab (BMS-986016) alone and in combination with anti-PD-1 monoclonal antibody nivolumab (BMS-936558) | – | NCT01968109 | 1/2a | Recruiting | Solid tumors that have spread and/or cannot be removed by surgery |
INCAGN02385 | – | NCT03538028 | 1 | Completed | Select advanced malignancies | |
XmAb®22841 monotherapy & in combination with pembrolizumab | – | NCT03849469 (DUET-4) | 1 | Recruiting | Subjects with selected advanced solid tumors | |
TIM-3 | TSR-022 | – | NCT02817633 (AMBER) | 1 | Recruiting | As a single agent and combination therapy in advanced solid tumors |
Siglec-15 | NC318 | – | NCT03665285 | 1/2 | Recruiting | Advanced or metastatic solid tumors |
VISTA | JNJ-61610588 | – | NCT02671955 | 1 | Terminated | Advanced cancer |
CI-8993 | – | NCT04475523 | 1 | Recruiting | Advanced solid tumor malignancies | |
STAT-3 | Durvalumab (MEDI4736, PD-L1 inhibitor) in combination with AZD9150 (STAT3 inhibitor) or AZD5069 (CXCR2 inhibitor) | – | NCT02499328 | 1b/2 | Active, not recruiting | In patients with advanced solid malignancies and subsequently comparing AZD9150 and AZD5069 both as monotherapy and in combination with MEDI4736 as second line treatment in patients with R/M HNSCC |
TTI-101 | – | NCT03195699 | 1 | Recruiting | Advanced cancers | |
IDO-1 | Pembrolizumab (MK-3475) in combination with INCB024360 (IDO-1 inhibitor) | – | NCT02178722 (ECHO-202/KEYNOTE-037) | 1/2 | Active, not recruiting | In participants with selected cancers |
Pembrolizumab plus epacadostat, pembrolizumab monotherapy | EXTREME Regimen | NCT03358472 (KEYNOTE-669/ECHO-304) | 3 | Active, not recruiting | As first line treatment for R/M HNSCC | |
combination of GDC-0919 and atezolizumab | – | NCT02471846 | 1 | Completed | In participants with locally advanced or metastatic solid tumors that has progressed after available standard therapy or for which standard therapy is ineffective, intolerable, or inappropriate | |
BMS-986205 In combination with nivolumab | Nivolumab | NCT03854032 | 2 | Recruiting | In treating patients with stage II-IV squamous cell cancer of the head and neck | |
EGFR | Cetuximab | Radiotherapy | NCT00004227 | 3 | Terminated | In treating patients with stage III or stage IV cancer of the oropharynx, hypopharynx, or larynx |
Erlotinib in combination with chemoradiotherapy | Chemo-radiotherapy | NCT00410826 | 2 | Completed | In patients with stage III and IV squamous cell carcinoma of the head and neck | |
Afatinib | Placebo | NCT01345669 | 3 | Terminated | As adjuvant therapy after chemo-radiotherapy in primary unresected patients with stage III, IVa, or IVb loco-regionally advanced HNSCC | |
EGFR antisense DNA in combination with radiation and cetuximab | – | NCT01592721 | 1/2 | Active, not recruiting | In patients with locally advanced HNSCC | |
BB-401 | – | NCT03433027 | 2 | Completed | In patients with R/M HNSCC who have failed all available standard therapies | |
MRG003 | – | NCT04868162 | 2 | Recruiting | R/M HNSCC | |
Dacomitinib | – | NCT04946968 | 2 | Recruiting | Patients With EGFR-driven advanced solid tumours with low EGFR-AS1 IncRNA Expr or other novel emerging biomarkers | |
VEGF | Bevacizumab and erlotinib | – | NCT00055913 | 1/2 | Completed | R/M HNSCC |
Pemetrexed and bevacizumab | – | NCT00222729 | 2 | Completed | R/M HNSCC | |
Cetuximab and bevacizumab | – | NCT00409565 | 2 | Completed | R/M HNSCC | |
Cetuximab with or without sorafenib | – | NCT00939627 | 2 | Completed | R/M HNSCC | |
mTOR | Temsirolimus | – | NCT01172769 (TEMHEAD) | 2 | Completed | In Patients With relapsed/recurrent HNSCC |
Temsirolimus | – | NCT01016769 | 1/2 | Completed | In combination with weekly paclitaxel and carboplatin for R/M HNSCC | |
Everolimus (RAD001) | – | NCT01051791 | 2 | Terminated | Refractory, recurrent, locally advanced squamous cell carcinoma of the head and neck | |
CDK | Palbociclib (PD 0,332,991) | – | NCT02101034 | 1/2 | Active, not recruiting | With cetuximab in patients with incurable SCCHN |
Palbociclib | Cetuximab | NCT02499120 | 2 | Active, not recruiting | With cetuximab, in HPV negative, cetuximab naïve patients with R/M HNSCC after failure of one prior platinum containing chemotherapy regimen | |
P276–00 | – | NCT00899054 (SPARK) | 1/2 | Completed | In combination with radiation, in patients with recurrent and/or locally advanced HNSCC | |
P276–00 | – | NCT00824343 (MONARCH) | 2 | Completed | In Indian patients with recurrent, metastatic or unresectable locally advanced HNSCC | |
Flavopiridol | – | NCT00020189 | 2 | Completed | R/M HNSCC | |
ALK1 | Dalantercept | – | NCT01458392 | 2 | Completed | R/M HNSCC |
Chemotherapy
Localized HNSCC is usually treated with surgery and/or chemoradiotherapy in a multidisciplinary manner [92]. Concomitant chemotherapy consists of regular docetaxel, cisplatin, and 5-FU treatment followed by radiation, which has been shown to enhance clinical outcomes in post-operative or inoperable situations. Cisplatin-based treatment enhanced OS in patients with localized HNSCC, according to a meta-analysis of 50 trials [93]. Furthermore, in high-risk patients, chemoradiotherapy with cisplatin in the post-operative environment resulted in a considerable benefit. Five-year PFS increased from 36 to 47 percent in a phase III trial with 334 patients, while five-year OS climbed from 40 to 53 percent. However, cisplatin-treated patients had more adverse effects (hematologic toxicities, mucositis, and nausea/vomiting) than radiotherapy-only individuals [94].
Other platinum-based therapy schemes were developed due to toxicity concerns. Three cycles of carboplatin (70 mg/m2/day; 4d) and 5-FU (600 mg/m2/d; 4d) treatment in stage III/IV patients, for example, showed better 3-year PFS and OS. Mucositis of grade 3–4 looked to be a serious adverse occurrence once more [83]. Induction chemotherapy with cisplatin and 5-FU has been proven to be beneficial, as evidenced by a meta-analysis in the HNC cohort. In a phase III trial with 237 inoperable stage III/IV HNSCC patients, four rounds of cisplatin (100 mg/m2 q3w) and 5-FU (1000 mg/m2/days; 5days; q3w) followed by radiation increased 5-year OS by 13% as compared to radiotherapy alone [84].
Multi-combinatorial therapy have also been found to be extremely beneficial, as evidenced by the results of multiple clinical trials. In comparison to the cisplatin-5-FU combination, adding docetaxel to the above-mentioned cisplatin and 5-FU combination followed with chemoradiotherapy (weekly carboplatin) increased 3-year OS [95]. Only grade 3/4 neutropenia was frequently reported as an adverse occurrence [95]. Patients who received docetaxel, cisplatin, and 5-FU (DCF) followed by chemotherapy/radiotherapy or cetuximab/radiotherapy significantly improved their PFS or OS in previous trials [[96], [97]]. DCF toxicity, on the other hand, remained a worry, accounting for 25.8% of grade 3–4 neutropenia and, more crucially, 7% of treatment-related death [96]. Other similar trials, on the other hand, found no significant changes in PFS and OS [[98], [99]]. Furthermore, when compared to regular DCF, patients treated with modified DCF (leucovorin as an adjuvant) had a greater response and tolerability. Chemotherapy-induced toxicity appears to be a key issue in HNSCC treatment, highlighting the need for less toxic therapeutic options [100]. To date, no standard radiation regimen for post-induction chemotherapy has been created. To obtain a conclusion, experiments are being done employing cisplatin and/or cetuximab [[101], [102]].
Molecular targeted therapy
Despite advances in traditional therapies such as surgery, chemotherapy, and radiotherapy, HNSCC patients' overall survival has not improved considerably. Molecular targeted treatments have showed potential in HNSCC in the current scenario [89]. Several molecular targets have been implicated in the treatment of HNSCC, including epidermal growth factor receptor (EGFR), Vascular endothelial growth factor (VEGF) and phosphatidiylinositol 3-kinase (PI3K). Concurrent chemoradiotherapy has been linked to a slew of hazardous side effects in HNSCC patients, suggesting that exploring targeted therapies could be a good strategy in this cohort [89].
Epidermal growth factor receptor (EGFR)
EGFR is a well-known cancer drug target as it influences cell proliferation, apoptosis, angiogenesis, and metastasis of cancer cells. EGFR is a transmembrane protein, and after binding to the ligand (EGF and TGF-α), it forms a homo- or heterodimer with other Erb family proteins (ErbB2, ErbB3, ErbB4) and activates the downstream signaling through the mitogen-activated protein kinase (MAPK) cascades and PI3K/AKT/mTOR pathway. This signaling cascade leads to the activation of certain genes in the nucleus, which promotes tumorigenesis and metastases (Fig. 2) [1,2]. EGFR overexpression is a negative prognostic factor that can be seen in about 90% of the HNSCC cases [3]. Furthermore, its overexpression has been positively correlated with earlier relapse, reduced disease-free survival and OS. Considering the abovementioned facts, inhibition of EGFR was targeted in HNSCC therapy. For EGFR inhibition, drug molecules either bind to the extracellular domain of EGFR, disrupting the link between ligands, or the cytoplasmic region of EGFR and inhibit the EGFR autophosphorylation by competing with ATP and thus interfere with the downstream cell signaling cascade [4].
Monoclonal antibodies
Monoclonal antibodies form a major subset of the current EGFR targeted therapy regimen and have demonstrated significant clinical benefits. Cetuximab is a chimeric monoclonal antibody that binds to domain III of the extracellular region of EGFR, and its anti-cancer effects are based on apoptosis induction and inhibition of cancer proliferation and angiogenesis [5]. Cetuximab also inhibits the phosphorylation of EGFR and prevents signals from being transmitted to the cell [6]. Several clinical trials have demonstrated that cetuximab in combination with radiotherapy increased the median OS and PFS [7,8]. In 2006, after promising application of cetuximab in combination with radiotherapy, USFDA approved its use for HNSCC therapy (Table 2). Panitumumab [9], zalutumumab [10], and nimotuzumab [11] are other promising human EGFR monoclonal antibodies which have shown favorable but limited effects in HNSCC patients. Other investigational EGFR monoclonal antibodies are being evaluated as monotherapy as well as combination therapy in several phase 1/2 clinical trials (NCT02277197, NCT03491709, and NCT03744208) (Fig. 2).
Table 2.
S. No. | Drug | Mechanism of action | Year | Disease conditions | Clinical trial | Ref. |
---|---|---|---|---|---|---|
1. | Cetuximab | EGFR inhibitor | 2006 | HNSCC after platinum-based therapy (in combination with radiotherapy) | NCT00004227 | [8] |
2009 | R/M HNSCC (in combination with platinum–fluorouracil chemotherapy) | NCT00122460 | [68] | |||
2. | Pembrolizumab | PD-1 inhibitor | 2016 | HNSCC after platinum-based chemotherapy | NCT01848834 | [66] |
2019 | Metastatic or unresectable recurrent HNSCC (in combination with platinum and fluorouracil (FU) for all patients and as a single agent for patients whose tumors express PD‑L1) | NCT02358031 | [71] | |||
3. | Nivolumab | PD-1 inhibitor | 2016 | R/M HNSCC with disease progression on or after a platinum-based therapy | NCT02105636 | [65] |
Tyrosine kinase inhibitors (TKIs)
Drugs of this class are under investigation in several early and late phase clinical trials. Gefitinib and erlotinib are the popular inhibitors which have shown potential for the treatment of HNSCC (Fig. 2) [12]. In a phase II study (NCT00410826), erlotinib in combination with cisplatin and radiotherapy was reported to improve complete response rate (CRR) as compared to the control group (only cisplatin and radiotherapy) but that was not significant [13]. Lapatinib is another TKI that inhibits EGFR (ErbB1) and HER2 (ErbB2) receptors by binding to the intracellular phosphorylation domain (ATP-binding pocket) preventing their self-phosphorylation and subsequent signaling activation [14]. Combinatorial therapy of lapatinib and capecitabine has been found effective and well-tolerated in HNSCC patients [15]. Afatinib, an irreversible ErbB family blocker, showed anti-proliferative and antitumor activity in in-vitro studies, which was further explored in clinical trials [16]. Efficacy of afatinib has been found comparable to cetuximab as evidenced by a phase II trial in recurrent HNSCC treated previously with platinum-based chemotherapy [17]. The utility of afatinib as an adjuvant has also been evaluated in a phase III trial (LUX-Head & Neck 2, NCT01345669) following chemoradiotherapy in the advanced stage of HNSCC [18]. Similarly, dacomitinib, an oral pan-EGFR inhibitor, has also shown clinical potential during the first line treatment of R/M HNSCC, warranting further studies as a part of combination therapy in advanced disease conditions (Fig. 2) [19].
EGFR anti-sense DNA
EGFR anti-sense DNA is a synthetic DNA sequence designed as an antisense to the DNA sequence of EGFR gene (Fig. 2). It inhibits the EGFR expression by tumor cells and thus inhibits tumor growth. Antisense therapy tends to be more effective due to the specificity and flexibility. Another advantage of this therapy is the ability to overcome drug resistance, since, the sequence of antisense DNA can be altered according to the genetic mutations of the targeted genes [20]. EGFR antisense DNA has been shown to reduce HNSCC proliferation and viability in preclinical studies [20,21]. Intratumoral EGFR antisense DNA has been evaluated in two phase 1/2 studies (NCT00903461 and NCT01592721), in combination with cetuximab and radiotherapy, in patients with locally advanced HNSCC and was well tolerated [22]. Intratumoral EGFR antisense DNA (BB-401) has completed a phase 2 trial (NCT03433027) evaluating the safety, tolerability, and efficacy in patients with R/M HNSCC who have failed all available standard therapies, and results have not been published yet.
Vascular endothelial growth factor (VEGF)
VEGF is a crucial signaling protein that stimulates angiogenesis and is responsible for the formation of new blood vessels [23]. Overexpression of VEGF has been positively correlated in majority of HNSCC cases and favors tumor growth, cell migration and metastases. Activated angiogenesis by tumor cells reduce the sensitivity to radiation in the patients undergoing radiotherapy treatment [24]. There are several clinical trials focused on VEGF, where targeted therapies have helped inhibiting the angiogenesis The most commonly used molecules in targeted therapies are bevacizumab, sunitinib, sorafenib and vandetanib (Fig. 2). [25]. Bevacizumab is USFDA approved monoclonal antibody and employed in the treatment of several cancer types including colon cancer, kidney cancer and cervical cancer. Although underlying mechanism remains unclear, pre-clinical studies have confirmed the ability of bevacizumab to improve the HNSCC sensitivity towards radiotherapy [26]. In a phase 1/2 trial (NCT00055913), bevacizumab in combination with erlotinib was well tolerated with four patients (out of 48) having a complete response. Median time of OS and PFS were 7.1 and 4.1 months [27]. Another phase 2 study (NCT00222729) investigating combinatorial therapy of bevacizumab and pemetrexed also has shown favorable outcomes [28]. Cetuximab (anti-EGFR) and bevacizumab (anti-VEGF) were evaluated in preclinical as well as phase 2 clinical study (NCT00409565). The combination enhanced growth inhibition both in-vitro and in-vivo, and also reduced tumor vascularization. Clinically, 16% of objective response rate (ORR) and 73% of disease control rate (DCR) were observed [29]. Furthermore, in a phase II trial, bevacizumab in combination with high doses of cisplatin and intensity-modulated radiation therapy (IMRT) delivered favorable clinical outcomes in advanced stage HNSCC [30]. It is worth adding that IMRT minimizes the radiation exposure to healthy area by a planned and focused photon irradiation to the tumor confined area [31].
VEGFR and platelet-derived growth factor receptor (PDGFR) inhibition
Several preclinical and clinical trials have indicated the importance of small molecules capable of modulating the signaling pathways acting through VEGFR and PDGFR [32]. Sorafenib is USFDA approved serine/threonine protein kinase inhibitor that targets VEGFR and inhibits tumor growth by inhibiting cell proliferation, cell migration, cell invasion (Fig. 2) [33] and inducing autophagy [34]. Furthermore, Sorafenib sensitizes the HNSCC to radiotherapy by its ability of double-stranded DNA break repair inhibition [35]. Sunitinib is another FDA approved oral kinase inhibitor that targets VEGFR, PDGFR and c-kit, however its activity is not explored much in HNSCC cases. During clinical trials, Sunitinib showed poor activity alone, but in combination with cetuximab, it showed significant synergic effects, and addition of radiotherapy completely abolished tumor growth [36]. Vandetanib is also a potential oral kinase inhibitor targeting EGFR, VEGFR, and RET-tyrosine kinase [37]. Pazipanib, nilotinib, axitinib and linifanib are other VEGF inhibitors which are under investigation in various clinical trials of HNSCC [32] (Fig. 2).
PI3K/AKT/mTOR pathway
The PI3K/AKT/mTOR pathway is an intracellular signaling pathway and serves a crucial role in the regulation of cell cycle [38]. Activation of this pathway leads to reduced apoptosis and increased cell proliferation in tumor. mTOR is a serine threonine kinase which has been found activated in HNSCC and thus a potential therapeutic target [39]. Primarily there are two types of mTOR inhibitors. First generation inhibitors are derived from rapamycin (antibiotic) while second generation is ATP competitive such as Torin1, PP242 and PP30. For mTOR inhibition in clinical trials, rapamycin analogues have been used including temsirolimus and everolimus (Fig. 2) [40]. Pre-clinical studies and patient derived xenograft models indicates that temsirolimus and everolimus is effective in inhibiting cell proliferation in HNC [41,42].
Several mTOR inhibitors are being evaluated alone or in combination therapies (with chemo/radiotherapy) in several phase1/2 clinical trials. Temsirolimus has been evaluated in a phase 2 study (NCT01172769) in platin- and cetuximab refractory R/M HNSCC patients and has shown tumor shrinking properties with clinical efficacy. However, the incidence of mutations (KRAS, BRAF, and PI3KCA) were lower than anticipated and showed no association with clinical outcome [43]. A combination therapy consisting of temsirolimus along with low-dose weekly carboplatin and paclitaxel was evaluated in a phase 2 study (NCT01016769) in R/M HNSCC patients and showed efficacy comparable to that of standard high-dose chemotherapy regimens and was well tolerated as well [44]. Everolimus failed to show efficacy as monotherapy in a phase 2 trial in previously treated R/M HNSCC patients (NCT01051791) [45]. Metformin, widely used drug for the treatment of type-2 diabetes worldwide, has shown to inhibit mTORC1 activity by both AMPK dependent and independent mechanisms and reduced the growth of HNSCC cells in in-vitro and in-vivo models [46,47]. Based on these studies, metformin is being evaluated in several phase 1/2 clinical trials alone and in combination with chemo/radiotherapy and immunotherapies [48].
Cyclin dependent kinases (CDK)
Cyclin dependent kinases play crucial roles in the cell-cycle regulation and are also involved in the pathogenesis of several diseases including cancer. USFDA approval of three CDK 4/6 inhibitors viz., palbociclib, ribociclib, and abemaciclib for the treatment of advanced and metastatic breast cancer has already proven the efficacy of this class of molecules as potential anticancer drugs (Fig. 3) [49]. Several CDK inhibitors have been evaluated in preclinical as well as clinical studies for the treatment of HNSCC. Ribociclib (LEE011), a CDK 4/6 inhibitor, has shown cytostatic effects in human papillomavirus (HPV) negative HNSCC models [50]. In another study, ribociclib was shown to induce cell-cycle arrest in HNSCC cell lines and also exhibited radiosensitization effects, suggesting that a combination of CDK 4/6 inhibitors with radiotherapy could be a promising option for the treatment of HNSCC [51].
Clinically, palbociclib, in combination with cetuximab, was evaluated in a phase 2 study (NCT02101034) in platinum- resistant and cetuximab-resistant HPV unrelated HNSCC patients. An objective response rate of 39% and 19% was observed in platinum-resistant and cetuximab-resistant groups, respectively, warranting further studies of CDK 4/6 inhibitors [52]. The same combination was investigated in another phase 2 study (PALATINUS, NCT02499120) in platinum-resistant, cetuximab-naive, HPV-unrelated HNSCC patients. Median OS was reported to be 9.7 months in palbociclib plus cetuximab group and 7.8 months in cetuximab group, showing a prolongation of median OS in combination group compared to cetuximab alone [53]. Riviciclib (P276–00), a novel inhibitor of CDK 1/4/9 [49], has been tested in two clinical studies (NCT00899054 and NCT00824343) in advanced HNSCC patients, and results are forthcoming. Flavopiridol, a CDK 4/9 inhibitor, has also completed a phase 2 trial (NCT00020189) focussed on the effectiveness in R/M HNSCC patients. Additional clinical trials investigating CDK inhibitors to treat HNSCC are ongoing (Fig. 3).
Activin receptor-like kinase-1 (ALK1)
The activin receptor-like kinase-1 (ALK1) belongs to TGF-β class and plays crucial role in modulating angiogenesis and vasculature development [54]. Dalantercept, an anti-angiogenic compound, has shown a significant potential as an ALK1 inhibitor in patients suffering from advanced solid tumors including HNSCC in a phase 2 clinical trial (NCT01458392) (Fig. 3) [55]. Apart from this several other line of targeted drugs such as proteasome inhibitors (e.g. bortezomib) and Notch inhibitors has also been explored [56], [57], [58], however an extensive clinical investigation is required in further studies.
Signal transducer and activator of transcription 3 (STAT3)
STAT3 is a transcriptional factor involved in various cellular processes including cell proliferation, survival, differentiation, and angiogenesis. In tumor cells, STAT3 becomes hyperactivated decreasing the expression of immunity factors like interferons, pro-inflammatory cytokines, and chemokines, while increasing the expression of growth factors [59]. STAT3 has also been found to be involved in the increased expression of various checkpoint molecules (PD-1, CTLA-4), the combination of STAT3 inhibitors with checkpoint inhibition therapy has shown encouraging results by increasing the therapeutic potential of checkpoint inhibitors and decreasing the resistance against them [59]. Various clinical studies are ongoing evaluating the potential of combination immunotherapy for the cancer treatment. A combination therapy of durvalumab (MEDI4736, PD-L1 inhibitor) and danvatirsen (AZD9150, STAT3 inhibitor) or AZD5069 (CXCR2 inhibitor) has been evaluated in a phase 1b/2 study (NCT02499328) in patients with advanced solid malignancies and HNSCC. This study showed improved anticancer activity as a result of combining PD-L1 inhibitor with STAT3 inhibitor as compared to PD-L1 monotherapy [60]. Further, enhanced activity was reported by the combination of PD-L1 inhibitor with STAT3 inhibitor compared to CXCR2 inhibitor plus PD-L1 inhibitor or PD-L1 monotherapy and warrants further investigation. Another STAT3 inhibitor, TTI-101, is undergoing a phase 1 evaluation in patients with advanced cancers including HNSCC (NCT03195699).
Indoleamine 2, 3-dioxygenase 1 (IDO-1)
IDO-1 is a tryptophan catabolizing enzyme that is expressed by the cancer cells and cancer-associated cells. It exerts immunosuppressive effects by suppressing activity of T-cells and NK cells, and activation of regulatory T-cells. Further, IDO-1 is involved in the development of resistance against checkpoint inhibitors. Combination of IDO-1 inhibitor with checkpoint inhibitors could be an alternative therapy for the cancer treatment. Several IDO-1 inhibitors are in currently clinical development.
Epacadostat is an investigational oral IDO-1 inhibitor; in a preliminary phase 1/2 study (ECHO-202/KEYNOTE-037, NCT02178722) Epacadostat in combination with pembrolizumab showed tolerable safety profile and encouraging antitumor activity in multiple advanced solid tumors [61]. A phase 3 clinical study (KEYNOTE-669/ECHO-304, NCT03358472) is evaluating the efficacy and safety of pembrolizumab and epacadostat combination therapy, pembrolizumab monotherapy, and the EXTREME regimen (cetuximab + cisplatin/carboplatin + 5-FU) as first-line treatment for R/M HNSCC. Another IDO-1 inhibitor, Navoximod (GDC-0919), has been studied in combination with a PD-L1 inhibitor atezolizumab in a phase 1 study (NCT02471846) in patients with advanced solid tumors, but failed to show additional benefit of combination [62]. BMS-986205 (linrodostat) is a selective and irreversible IDO-1 inhibitor that is being evaluated in a phase 2 clinical trial (NCT03854032) in combination with nivolumab in treating patients with stage II-IV HNSCC. Another phase 3 clinical study (NCT03386838) evaluating BMS-986205 plus nivolumab vs. standard of care EXTREME regimen in first line R/M HNSCC has been withdrawn.
Immunotherapy
Surgery and radiotherapy, with or without conventional chemotherapy, are the existing mainstays of advanced head and neck squamous cell carcinoma (HNSCC) treatment. Advanced human papillomavirus (HPV)-negative HNSCC has a dismal prognosis despite this multi-modality treatment. Treatment intensification with molecular targeted therapies is not enough to appreciably improve overall survival. Immunotherapy for head and neck cancer offers patients, particularly those with HPV-related malignancies, interesting new treatment options without the potentially fatal side effects of traditional treatments.
It is now widely accepted that a properly functioning immune system may effectively resist tumor cell effects. Blocking negative signaling pathways in effector cells (e.g., CTLA-4, PD-1/PD-L1) or inducing co-stimulatory signals could thus be used to boost host immunity. Agonistic monoclonal antibodies like MEDI0562 (against OX-40), urelumab and utomiliumab (against CD137), and motolimod (toll-like receptor-8 agonist) have been studied in multiple clinical studies in the co-stimulatory cohort [103]. It's worth noting that, in addition to cytotoxic effects, chemotherapy can also cause immunogenic changes that can stimulate the immune system. Cisplatin, for example, upregulates immunosuppressive signaling in the tumor microenvironment by activating the expression of major histocompatibility complex I and promoting the lytic activity of effector cells [104]. These findings suggest that immunotherapy could be combined with chemoradiotherapy to improve clinical outcomes in HNSCC. Immunotherapies are being tested on a variety of patient subgroups, including HPV-positive and HPV-negative subtypes. As indicated by multiple clinical trials [103], immunotherapy in HPV positive cases targets different non-host virus-specific tumor antigens E6 and E7. Recent clinical evidence from immunotherapy trials suggests that using the right checkpoint inhibitors can increase survival by many times.
Programmed cell death protein 1 (PD-1) and programmed death-ligand 1(PD-L1)
Programmed death-1 (PD-1) is an immunoreceptor which is expressed on T and B lymphocytes, as well as on monocytes and dendritic cells (Fig. 4) [63]. PD-1 is activated through binding of its ligands: PD-L1 and PD-L2, and as a result, suppress inflammatory response. Both PD-1 and PD-L1 have been found to be overexpressed in cancer cases, including head and neck, lung, colon, melanoma, breast, and kidney. In fact, overexpression of these receptors and ligands results in dysfunction of anti-cancer signaling, which allows tumor cells to escape from immune surveillance [64]. Thus PD-1 receptor and its ligands (PD-L1 and PD-L2) are attractive targets for various cancers, including HNSCC.
In a clinical trial (CheckMate 141, NCT02105636), an anti-PD-1 antibody nivolumab was evaluated in recurrent/metastatic (R/M) HNSCC patients at an intravenous dose of 3 mg/kg every 2 weeks and compared with weekly single agent intravenous chemotherapy using methotrexate (40–60 mg/m2), docetaxel (30–40 mg/m2) or cetuximab (first dose at 400 mg/m2 then 250 mg/m2). The group treated with nivolumab showed less adverse effects and increased OS as compared to other groups [65]. In another prospective trial (KEYNOTE-012, NCT01848834) with an anti-PD-1 antibody pembrolizumab (10 mg/kg every 2 weeks), clinical outcomes in terms of efficacy and toxicity were found similar to nivolumab [66]. Furthermore, an expanded study in similar cohort of R/M HNSCC patients at a fixed intravenous doses of 200 mg/kg every 3 weeks also showed promising clinical outcome in line with the nivolumab trial [67]. It is worth adding that several factors need to be determined before interpreting or comparing the results of different trials, as the inclusion criteria of trials may vary to a significant extent, which may affect the outcome across the trials. For example, pre-treatment status and age range of participants, randomized or non-randomized trials, drug doses, and treatment duration are major factors that directly impact final clinical outcomes. To date, cetuximab as an adjunct to platinum/5-FU, and, nivolumab monotherapy have responded to achieve significantly longer OS as compared to their respective control arms [68]. Immune checkpoint inhibitors usually elicit a delayed but a unique long-term off-treatment survival response than that of other targeted therapies (e.g., EGFR inhibitors) and conventional chemotherapies [69,70]. Both nivolumab and pembrolizumab are well tolerated and have gained FDA approval to be used as new care options for the treatment of HNSCC (Table 2) [32].
Cytotoxic T lymphocyte antigen 4 (CTLA4)
CTLA-4 is a receptor present on the surface of activated T-lymphocytes (Fig. 4), it competes with CD28 receptors to bind with B7 ligands present on the antigen presenting cells (APCs, tumor cells), inhibiting the activation signal required for T-cells. CTLA-4 has also been reported to inhibit IL-2 production, T-cell proliferation, and induce cell-cycle arrest [72]. Blocking CTLA-4 receptors can result in T-cell activation and immune response in the host cells. To date, only one CTLA-4 inhibitor, ipilimumab (Yervoy), has been approved for the treatment of unresectable or metastatic late-stage melanoma as well as adjuvant to surgery for high-risk stage III melanoma patients [73,74]. Recently, USFDA has approved the combination of ipilimumab and nivolumab as first-line treatment of metastatic non-small cell lung cancer (PD-L1 tumor expression ≥1%) after an investigation in a randomized, open-label, phase-3 clinical trial, CHECKMATE-227 (NCT02477826) [75].
CTLA-4 inhibitors are being evaluated in several clinical studies for the treatment of HNSCC. The combination of ipilimumab and nivolumab is undergoing a randomized, open-label phase 3 clinical trial as first-line treatment of R/M HNSCC compared to standard of care (CheckMate 651, NCT02741570). Another phase 3 clinical study, IMSTAR-HN (NCT03700905) is evaluating the potential of nivolumab, alone or in combination with ipilimumab, as immunotherapy vs standard follow up in surgical resectable HNSCC after adjuvant radio(-chemo)therapy [76].
Lymphocyte activated gene-3 (LAG-3)
LAG-3 is present on the surface of activated T-cells as well as natural killer (NK) cells, B cells, and dendritic cells (Fig. 4). It binds to MHC-II present on APCs inhibiting the interaction of T-cell receptors with MHC-II. Crosslinking of LAG-3 and T-cell receptors can impair cytokine production and T-cell proliferation [77]. Thus, LAG-3 interferes T-cell signaling in immune response. LAG-3 blockers can inhibit the binding of LAG-3 to MHC-II and provoke an immune response to the APCs. Anti-LAG-3 antibody relatlimab (BMS-986016) is being evaluated in a phase 1/2 study to assess the safety, tolerability, and effectiveness alone or in combination with anti-PD-1 nivolumab for the treatment of solid tumors (NCT01968109). INCAGN02385, an Fc-engineered IgG1κ antibody having high affinity to LAG-3, is being evaluated against advanced malignancies including HNSCC in a phase 1 study (NCT03538028). A bispecific antibody, XmAb®22841, targeting CTLA-4 and LAG-3 simultaneously has been developed and is being tested as monotherapy and in combination with pembrolizumab in patients with advanced solid tumors in an ongoing trial (DUET-4, NCT03849469). More clinical trials are undergoing to assess anti-LAG-3 antibodies alone or in combination with other drugs (nivolumab, relatlimab, ipilimumab; NCT04326257).
T cell immunoglobulin mucin-3 (TIM-3)
TIM-3 is an inhibitory protein expressed by CD4+ and CD8+ T-cells, and other immune cells including regulatory T-cells, mast cells, natural killer cells and dendritic cells (Fig. 4). Galectin-9 act as major ligand for TIM-3, and is produced by the APCs (tumor cells). Upon binding, galectin-9-TIM-3 complex suppresses immune responses by disrupting the formation of immune synapse, phosphatase recruitment, and ultimately, cell becomes anergic [78]. Blocking the interaction of galactin-9 and TIM-3 by anti-TIM-3 antibodies may facilitate the immune response against cancer, inhibiting tumor growth. In a preclinical study, PD-1 and TIM-3 have been reported to be co-expressed on T-cells of mice bearing solid tumors; and combined targeting both targets was highly effective in inhibiting tumor growth and restoring T-cell functions [79]. TIM-3 is also being targeted in multiple clinical trials for the treatment of various types of cancer. An anti-TIM-3 antibody TSR-022 is being evaluated in a phase 1 dose escalation and dose expansion in patients with advanced solid tumors (AMBER, NCT02817633). In another phase 1 clinical study, INCAGN02390 is undergoing safety, tolerability and preliminary effectiveness evaluation as a monotherapy in patients with advanced malignancies including HNSCC.
Siglec-15
Siglec-15 is an immunosuppressive protein expressed on tumor cells and/or tumor-associated macrophages (Fig. 4). It has been shown to suppress T-cell proliferation and activation in-vitro as well as in-vivo. On the other hand, siglec-15 deficiency has been shown to promote T-cell responses, and control tumor growth. Also, siglec-15 targeting with monoclonal antibody in murine model inhibited tumor growth by reversing T-cell suppression [80]. Thus, targeting siglec-15 by specific monoclonal antibodies can be a therapeutic option for cancer treatment, or it can provide additional support to the existing anti-cancer immunotherapy. Currently, anti-siglec-15 monoclonal antibody, NC318, is being evaluated in a phase 1/2 dose-escalation, safety and tolerability study, in patients with advanced or metastatic solid tumors including HNSCC cohort (NCT03665285).
V-domain immunoglobulin suppressor of T cell activation (VISTA)
VISTA is a negative checkpoint regulator, expressed on hematopoietic cells, mainly tumor infiltrating myeloid cells. It has been shown to inhibit T-cell proliferation and cytokine production in vitro. The same study highlighted that anti-VISTA mAb enhanced T-cell responses [81]. Another preclinical study demonstrated that VISTA mAb administration enhanced T-cell proliferation and function, as well as, suppressed tumor growth in transplantable and inducible tumor models [82]. Thus, VISTA could be an important target for the cancer therapeutics, either as a monotherapy or in combination with other therapies. To date, there has been two clinical studies utilizing anti-VISTA antibodies in subjects with advanced cancer, of which, a phase 1 study of monoclonal antibody JNJ-61610588 has been terminated (NCT02671955). Another phase 1 clinical trial evaluating the safety, tolerability, pharmacokinetics, pharmacodynamics, and anti-cancer activity of CI-8993, anti-VISTA antibody, in patients with advanced solid tumor malignancies is currently in the recruiting stage (NCT04475523).
Conclusion
Despite the fact that a variety of therapeutic techniques are being used to improve the prognosis of HNSCC patients, the treatment has yet to be revolutionized due to the disease's complexity and heterogeneity. Several medications have been studied in clinical trials around the world as monotherapies or in combination with other drugs. Chemotherapy, chemoradiotherapy, targeted therapy, and immunotherapy have all shown varying results depending on a variety of parameters such as the stage of HNSCC, comorbidities, age, and past treatment. Because of the critical involvement of the immune system in carcinogenesis, immunotherapy has emerged as a promising treatment option in recent years. The development of targeted therapeutics is based on a molecular understanding of the underlying biology of cancer progression, however the response is limited because to the complicated interplay of numerous cell-signaling cascades. To ensure their significance in diverse cohorts and stages of HNSCC, more clinical trials are needed. Docetaxel, cisplatin, and 5-FU were the most commonly used medications in the prospective and conventional treatment of HNSCC. Apart from these medications, the USFDA has only approved three others: cetuximab, pembrolizumab, and nivolumab. Furthermore, there is still room for advancement in immunotherapy and the addition of appropriate prognostic biomarkers for better therapeutic options in HNSCC patients. Biomarkers may play a crucial role in onco-immunotherapy, according to the suggestions of a research committee from the National Cancer Institute. For example, tumor-related biomarkers (e.g., interferon, PD-1/PD-L1, and CTLA-4 expression), peripheral blood mononuclear cell-related biomarkers (e.g., MDSCs and regulatory T lymphocytes, HPV virus peptides), serum biomarkers (e.g., cytokines, antibodies, and growth factors), bio-imaging (e.g., computed tomography and/or positron emission Although these biomarkers are not routinely used before and after diagnosis, they must be prospectively validated in well-designed controlled clinical trials. The direct link between exosomes, metabolomics, and the aggressiveness of HNSCC has become a hot area of research in recent years. Exosomes as HNSCC biomarkers, therapeutic targets, and drug carriers for HNSCC treatments require more research, and metabolomics must overcome a number of challenges before it can be extensively employed in clinical research and practice.
Funding
AK acknowledges the financial support from the Department of Biotechnology, Government of India, in the form of Ramalingaswami Fellowship. The funding agency played no role in the study design, analysis, and interpretation of results.
CRediT authorship contribution statement
Bharat Goel: Conceptualization, Methodology, Writing – original draft. Anoop Kumar Tiwari: Data curation, Writing – review & editing. Rajeev Kumar Pandey: Writing – review & editing. Akhand Pratap Singh: Conceptualization, Methodology, Writing – original draft. Sujeet Kumar: Writing – review & editing. Abhishek Sinha: Writing – review & editing. Shreyans K. Jain: Supervision, Writing – review & editing. Arun Khattri: Conceptualization, Methodology, Investigation, Supervision, Writing – review & editing, Project administration, Funding acquisition.
Declaration of Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
Figs. 2, 3 and 4 used elements from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (www.servier.fr/servier-medical-art).
References
- 1.Song J., Chen C., Raben D. Emerging role of EGFR-targeted therapies and radiation in head and neck cancer. Oncology. 2004;18:1757–1767. [PubMed] [Google Scholar]
- 2.Goerner M., Seiwert T.Y., Sudhoff H. Molecular targeted therapies in head and neck cancer-An update of recent developements. Head Neck Oncol. 2010;2:8. doi: 10.1186/1758-3284-2-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Argiris A., Karamouzis M.V., Raben D., Ferris R.L. Head and neck cancer. Lancet. 2008;371:1695–1709. doi: 10.1016/S0140-6736(08)60728-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lurje G., Lenz H.-.J. EGFR signaling and drug discovery. Oncology. 2009;77:400–410. doi: 10.1159/000279388. [DOI] [PubMed] [Google Scholar]
- 5.Matta A., Ralhan R. Overview of current and future biologically based targeted therapies in head and neck squamous cell carcinoma. Head Neck Oncol. 2009;1:6. doi: 10.1186/1758-3284-1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Huang S.-.M., Harari P.M. Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin. Cancer Res. 2000;6:2166–2174. [PubMed] [Google Scholar]
- 7.Cunningham D., Humblet Y., Siena S., et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N. Engl. J. Med. 2004;351:337–345. doi: 10.1056/NEJMoa033025. [DOI] [PubMed] [Google Scholar]
- 8.Bonner J.A., Harari P.M., Giralt J., et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2006;354:567–578. doi: 10.1056/NEJMoa053422. [DOI] [PubMed] [Google Scholar]
- 9.Vermorken J.B., Stöhlmacher-Williams J., Davidenko I., 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. doi: 10.1016/S1470-2045(13)70181-5. [DOI] [PubMed] [Google Scholar]
- 10.Eriksen J.G., Maare C., Johansen J., et al. Evaluation of the EGFR-Inhibitor Zalutumumab Given With Primary Curative (Chemo)radiation Therapy to Patients With Squamous Cell Carcinoma of the Head and Neck: results of the DAHANCA 19 Randomized Phase 3 Trial: definitive Management of Head-and-Neck Squamous Cell Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2014;88:465. [Google Scholar]
- 11.Subramanium S., Balasundaram V., Nithya S., Kiran P. Nimotuzumab with induction chemotherapy and chemo-radiation in patients with advanced head and neck cancer. J. Cancer Ther. 2015;6:146. [Google Scholar]
- 12.Mirghani H., Amen F., Blanchard P., et al. Treatment de-escalation in HPV-positive oropharyngeal carcinoma: ongoing trials, critical issues and perspectives. Int. J. Cancer. 2015;136:1494–1503. doi: 10.1002/ijc.28847. [DOI] [PubMed] [Google Scholar]
- 13.Martins R.G., Parvathaneni U., Bauman J.E., 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. doi: 10.1200/JCO.2012.46.3299. [DOI] [PubMed] [Google Scholar]
- 14.Fumagalli I., Dugue D., Bibault J.E., et al. Cytotoxic effect of lapatinib is restricted to human papillomavirus-positive head and neck squamous cell carcinoma cell lines. Onco. Targets Ther. 2015;8:335. doi: 10.2147/OTT.S68235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Weiss J.M., Bagley S., Hwang W.T., et al. Capecitabine and lapatinib for the first-line treatment of metastatic/recurrent head and neck squamous cell carcinoma. Cancer. 2016;122:2350–2355. doi: 10.1002/cncr.30067. [DOI] [PubMed] [Google Scholar]
- 16.Modjtahedi H., Cho B.C., Michel M.C., Solca F. A comprehensive review of the preclinical efficacy profile of the ErbB family blocker afatinib in cancer. Naunyn Schmiedebergs Arch. Pharmacol. 2014;387:505–521. doi: 10.1007/s00210-014-0967-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Seiwert T., Fayette J., Cupissol D., et al. A randomized, phase II study of afatinib versus cetuximab in metastatic or recurrent squamous cell carcinoma of the head and neck. Ann. Oncol. 2014;25:1813–1820. doi: 10.1093/annonc/mdu216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Burtness B., Bourhis J.P., Vermorken J.B., Harrington K.J., Cohen E.E. Afatinib versus placebo as adjuvant therapy after chemoradiation in a double-blind, phase III study (LUX-Head & Neck 2) in patients with primary unresected, clinically intermediate-to-high-risk head and neck cancer: study protocol for a randomized controlled trial. Trials. 2014;15:469. doi: 10.1186/1745-6215-15-469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Abdul Razak A., Soulieres D., Laurie S., et al. A phase II trial of dacomitinib, an oral pan-human EGF receptor (HER) inhibitor, as first-line treatment in recurrent and/or metastatic squamous-cell carcinoma of the head and neck. Ann. Oncol. 2013;24:761–769. doi: 10.1093/annonc/mds503. [DOI] [PubMed] [Google Scholar]
- 20.Thomas S.M., Sahu B., Rapireddy S., et al. Antitumor Effects of EGFR antisense guanidine-based peptide nucleic acids in cancer models. ACS Chem. Biol. 2013;8:345–352. doi: 10.1021/cb3003946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Thomas S.M., Grandis J.R. Pharmacokinetic and pharmacodynamic properties of EGFR inhibitors under clinical investigation. Cancer Treat. Rev. 2004;30:255–268. doi: 10.1016/j.ctrv.2003.10.003. [DOI] [PubMed] [Google Scholar]
- 22.Bauman J.E., Duvvuri U., Thomas S., et al. Phase 1 study of EGFR-antisense DNA, cetuximab, and radiotherapy in head and neck cancer with preclinical correlatives. Cancer. 2018;124:3881–3889. doi: 10.1002/cncr.31651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hinerman R.W., Mendenhall W.M., Morris C.G., Amdur R.J., Werning J.W., Villaret D.B. Postoperative irradiation for squamous cell carcinoma of the oral cavity: 35-year experience. Head Neck. 2004;26:984–994. doi: 10.1002/hed.20091. [DOI] [PubMed] [Google Scholar]
- 24.Minchenko A., Bauer T., Salceda S., Caro J. Hypoxic stimulation of vascular endothelial growth factor expression in vitro and in vivo. Lab. Invest. 1994;71:374–379. [PubMed] [Google Scholar]
- 25.Kyzas P.A., Cunha I.W., Ioannidis J.P. Prognostic significance of vascular endothelial growth factor immunohistochemical expression in head and neck squamous cell carcinoma: a meta-analysis. Clin. Cancer Res. 2005;11:1434–1440. doi: 10.1158/1078-0432.CCR-04-1870. [DOI] [PubMed] [Google Scholar]
- 26.Chen Z., Xu X.-H. Combining antiangiogenic therapy and radiation in nasopharyngeal carcinoma. Saudi Med. J. 2015;36:659. doi: 10.15537/smj.2015.6.11460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cohen E.E., Davis D.W., Karrison T.G., 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. doi: 10.1016/S1470-2045(09)70002-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Feinstein T., Raez L., Rajasenan K., et al. Pemetrexed (P) and bevacizumab (B) in patients (pts) with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC): updated results of a phase II trial. J. Clin. Oncol. 2008;26:6069. [Google Scholar]
- 29.Argiris A., Kotsakis A., Hoang T., 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. 2013;24:220–225. doi: 10.1093/annonc/mds245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fury M.G., Lee N.Y., Sherman E., 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. doi: 10.1002/cncr.27498. [DOI] [PubMed] [Google Scholar]
- 31.Cheung K. Intensity modulated radiotherapy: advantages, limitations and future developments. Biomed. Imaging Interv. J. 2006;2:e19. doi: 10.2349/biij.2.1.e19. -e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Szturz P., Vermorken J.B. Immunotherapy in head and neck cancer: aiming at EXTREME precision. BMC Med. 2017;15:110. doi: 10.1186/s12916-017-0879-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Möckelmann N., Rieckmann T., Busch C.-.J., et al. Effect of sorafenib on cisplatin-based chemoradiation in head and neck cancer cells. Oncotarget. 2016;7:23542. doi: 10.18632/oncotarget.8275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhang Y., Xu D., Wang X., Lu M., Gao B., Qiao X. Screening of kinase inhibitors targeting BRAF for regulating autophagy based on kinase pathways. Mol. Med. Rep. 2014;9:83–90. doi: 10.3892/mmr.2013.1781. [DOI] [PubMed] [Google Scholar]
- 35.Laban S., Steinmeister L., Gleißner L., et al. Sorafenib sensitizes head and neck squamous cell carcinoma cells to ionizing radiation. Radiother. Oncol. 2013;109:286–292. doi: 10.1016/j.radonc.2013.07.003. [DOI] [PubMed] [Google Scholar]
- 36.Bozec A., Sudaka A., Toussan N., Fischel J.-L., Etienne-Grimaldi M.-C., Milano G. Combination of sunitinib, cetuximab and irradiation in an orthotopic head and neck cancer model. Ann. Oncol. 2009;20:1703–1707. doi: 10.1093/annonc/mdp070. [DOI] [PubMed] [Google Scholar]
- 37.Heymach J.V., Johnson B.E., Prager D., et al. Randomized, placebo-controlled phase II study of vandetanib plus docetaxel in previously treated non–small-cell lung cancer. J. Clin. Oncol. 2007;25:4270–4277. doi: 10.1200/JCO.2006.10.5122. [DOI] [PubMed] [Google Scholar]
- 38.LoPiccolo J., Blumenthal G.M., Bernstein W.B., Dennis P.A. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist. Updat. 2008;11:32–50. doi: 10.1016/j.drup.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.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. doi: 10.1038/nrd2926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zaytseva Y.Y., Valentino J.D., Gulhati P., Evers B.M. mTOR inhibitors in cancer therapy. Cancer Lett. 2012;319:1–7. doi: 10.1016/j.canlet.2012.01.005. [DOI] [PubMed] [Google Scholar]
- 41.Niehr F., Weichert W., Stenzinger A., Budach V., Tinhofer I. CCI-779 (Temsirolimus) exhibits increased anti-tumor activity in low EGFR expressing HNSCC cell lines and is effective in cells with acquired resistance to cisplatin or cetuximab. J. Transl. Med. 2015;13:106. doi: 10.1186/s12967-015-0456-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Klinghammer K., Raguse J.D., Plath T., et al. A comprehensively characterized large panel of head and neck cancer patient-derived xenografts identifies the m TOR inhibitor everolimus as potential new treatment option. Int. J. Cancer. 2015;136:2940–2948. doi: 10.1002/ijc.29344. [DOI] [PubMed] [Google Scholar]
- 43.Grünwald V., Keilholz U., Boehm A., et al. TEMHEAD: a single-arm multicentre phase II study of temsirolimus in platin- and cetuximab refractory recurrent and/or metastatic squamous cell carcinoma of the head and neck (SCCHN) of the German SCCHN Group (AIO) Ann. Oncol. 2015;26:561–567. doi: 10.1093/annonc/mdu571. [DOI] [PubMed] [Google Scholar]
- 44.Fury M.G., Xiao H., Baxi S.S., et al. A phase II trial of temsirolimus plus low-dose weekly carboplatin and paclitaxel for recurrent/metastatic HNSCC. J. Clin. Oncol. 2014;32:6019. [Google Scholar]
- 45.Geiger J.L., Bauman J.E., Gibson M.K., et al. Phase II trial of everolimus in patients with previously treated recurrent or metastatic head and neck squamous cell carcinoma. Head Neck. 2016;38:1759–1764. doi: 10.1002/hed.24501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Vitale-Cross L., Molinolo A.A., Martin D., et al. Metformin prevents the development of oral squamous cell carcinomas from carcinogen-induced premalignant lesions. Cancer Prev. Res. 2012;5:562. doi: 10.1158/1940-6207.CAPR-11-0502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wu X., Yeerna H., Goto Y., et al. Metformin inhibits progression of head and neck squamous cell carcinoma by acting directly on carcinoma-initiating cells. Cancer Res. 2019;79:4360. doi: 10.1158/0008-5472.CAN-18-3525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wang Z., Valera J.C., Zhao X., Chen Q., Silvio Gutkind J. mTOR co-targeting strategies for head and neck cancer therapy. Cancer Metastasis Rev. 2017;36:491–502. doi: 10.1007/s10555-017-9688-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Goel B., Tripathi N., Bhardwaj N., Jain S.K. Small Molecule CDK Inhibitors for the Therapeutic Management of Cancer. Curr. Top. Med. Chem. 2020;20:1535–1563. doi: 10.2174/1568026620666200516152756. [DOI] [PubMed] [Google Scholar]
- 50.van Caloen G., Schmitz S., El Baroudi M., et al. Preclinical activity of ribociclib in squamous cell carcinoma of the head and neck. Mol. Cancer Ther. 2020;19:777. doi: 10.1158/1535-7163.MCT-19-0695. [DOI] [PubMed] [Google Scholar]
- 51.Tai T.S., Lin P.M., Wu C.F., et al. CDK4/6 Inhibitor LEE011 Is a potential radiation-sensitizer in head and neck squamous cell carcinoma: an In Vitro study. Anticancer Res. 2019;39:713–720. doi: 10.21873/anticanres.13167. [DOI] [PubMed] [Google Scholar]
- 52.Adkins D., Ley J., Neupane P., et al. Palbociclib and cetuximab in platinum-resistant and in cetuximab-resistant human papillomavirus-unrelated head and neck cancer: a multicentre, multigroup, phase 2 trial. Lancet Oncol. 2019;20:1295–1305. doi: 10.1016/S1470-2045(19)30405-X. [DOI] [PubMed] [Google Scholar]
- 53.Adkins D., Lin J.-.C., Sacco A.G., et al. Palbociclib plus cetuximab versus placebo plus cetuximab in platinum-resistant, cetuximab-naive, HPV-unrelated head and neck cancer: a double-blind randomized phase II trial (PALATINUS) J. Clin. Oncol. 2019;37:6013. [Google Scholar]
- 54.Cunha S.I., Pietras K. ALK1 as an emerging target for antiangiogenic therapy of cancer. Blood. 2011;117:6999–7006. doi: 10.1182/blood-2011-01-330142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jimeno A., Posner M.R., Weiss J., et al. Phase 2 study of dalantercept in recurrent or metastatic squamous cell carcinoma of the head and neck. J. Clin. Oncol. 2014;32:6045. [Google Scholar]
- 56.Benvenuto M., Ciuffa S., Focaccetti C., et al. Proteasome inhibition by bortezomib parallels a reduction in head and neck cancer cells growth, and an increase in tumor-infiltrating immune cells. Sci. Rep. 2021;11:19051. doi: 10.1038/s41598-021-98450-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhao Z.-.L., Zhang L., Huang C.-.F., et al. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci. Rep. 2016;6:24704. doi: 10.1038/srep24704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhao Y.Y., Yu G.T., Xiao T., Hu J. The Notch signaling pathway in head and neck squamous cell carcinoma: a meta-analysis. Adv. Clin. Exp. Med. 2017;26:881–887. doi: 10.17219/acem/64000. [DOI] [PubMed] [Google Scholar]
- 59.Zou S., Tong Q., Liu B., Huang W., Tian Y., Fu X. Targeting STAT3 in cancer immunotherapy. Mol. Cancer. 2020;19:145. doi: 10.1186/s12943-020-01258-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cohen E.E., Hong D.S., Wise Draper T., et al. Phase 1b/2 Study (SCORES) assessing safety, tolerability, and preliminary anti-tumor activity of durvalumab plus AZD9150 or AZD5069 in patients with advanced solid malignancies and squamous cell carcinoma of the head and neck (SCCHN) Ann. Oncol. 2017;28:V403. [Google Scholar]
- 61.Mitchell T.C., Hamid O., Smith D.C., et al. Epacadostat Plus Pembrolizumab in Patients With Advanced Solid Tumors: phase I Results From a Multicenter, Open-Label Phase I/II Trial (ECHO-202/KEYNOTE-037) J. Clin. Oncol. 2018;36:3223–3230. doi: 10.1200/JCO.2018.78.9602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jung K.H., LoRusso P., Burris H., et al. Phase I Study of the Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitor Navoximod (GDC-0919) Administered with PD-L1 Inhibitor (Atezolizumab) in Advanced Solid Tumors. Clin. Cancer Res. 2019;25:3220. doi: 10.1158/1078-0432.CCR-18-2740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Grzywnowicz M., Giannopoulos K. The role of receptor programmed death-1 and its ligands in immune system and tumors. Acta Haematol. Pol. 2012;43:132–145. [Google Scholar]
- 64.Dong H., Zhu G., Tamada K., Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999;5:1365–1369. doi: 10.1038/70932. [DOI] [PubMed] [Google Scholar]
- 65.Ferris R.L., Blumenschein G., Jr, Fayette J., et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2016;375:1856–1867. doi: 10.1056/NEJMoa1602252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Seiwert T.Y., Burtness B., Mehra R., et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17:956–965. doi: 10.1016/S1470-2045(16)30066-3. [DOI] [PubMed] [Google Scholar]
- 67.Chow L.Q., Haddad R., Gupta S., et al. Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J. Clin. Oncol. 2016;34:3838–3845. doi: 10.1200/JCO.2016.68.1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Vermorken J.B., Mesia R., Rivera F., et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N. Engl. J. Med. 2008;359:1116–1127. doi: 10.1056/NEJMoa0802656. [DOI] [PubMed] [Google Scholar]
- 69.McDermott D., Lebbé C., Hodi F.S., et al. Durable benefit and the potential for long-term survival with immunotherapy in advanced melanoma. Cancer Treat. Rev. 2014;40:1056–1064. doi: 10.1016/j.ctrv.2014.06.012. [DOI] [PubMed] [Google Scholar]
- 70.McKay R.R., Martini D., Moreira R.B., et al. Outcomes of PD-1/PD-L1 responders who discontinue therapy for immune-related adverse events (irAEs): results of a cohort of patients (pts) with metastatic renal cell carcinoma (mRCC) J. Clin. Oncol. 2017;35:467. [Google Scholar]
- 71.Burtness B., Harrington K.J., Greil R., et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet. 2019;394:1915–1928. doi: 10.1016/S0140-6736(19)32591-7. [DOI] [PubMed] [Google Scholar]
- 72.Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J. Exp. Clin. Cancer Res. 2019;38:255. doi: 10.1186/s13046-019-1259-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ascierto P.A., Del Vecchio M., Robert C., et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017;18:611–622. doi: 10.1016/S1470-2045(17)30231-0. [DOI] [PubMed] [Google Scholar]
- 74.Eggermont A.M.M., Chiarion-Sileni V., Grob J.-J., et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N. Engl. J. Med. 2016;375:1845–1855. doi: 10.1056/NEJMoa1611299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hellmann M.D., Paz-Ares L., Bernabe Caro R., et al. Nivolumab plus Ipilimumab in advanced non–small-cell lung cancer. N. Engl. J. Med. 2019;381:2020–2031. doi: 10.1056/NEJMoa1910231. [DOI] [PubMed] [Google Scholar]
- 76.Zech H.B., Moeckelmann N., Boettcher A., et al. Phase III study of nivolumab alone or combined with ipilimumab as immunotherapy versus standard of care in resectable head and neck squamous cell carcinoma. Future Oncol. 2020;16:3035–3043. doi: 10.2217/fon-2020-0595. [DOI] [PubMed] [Google Scholar]
- 77.Long L., Zhang X., Chen F., et al. The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer. 2018;9:176–189. doi: 10.18632/genesandcancer.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wolf Y., Anderson A.C., Kuchroo V.K. TIM3 comes of age as an inhibitory receptor. Nat. Rev. Immunol. 2020;20:173–185. doi: 10.1038/s41577-019-0224-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sakuishi K., Apetoh L., Sullivan J.M., Blazar B.R., Kuchroo V.K., Anderson A.C. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exper. Med. 2010;207:2187–2194. doi: 10.1084/jem.20100643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang J., Sun J., Liu L.N., et al. Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy. Nat. Med. 2019;25:656–666. doi: 10.1038/s41591-019-0374-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wang L., Rubinstein R., Lines J.L., et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011;208:577–592. doi: 10.1084/jem.20100619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Le Mercier I., Chen W., Lines J.L., et al. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014;74:1933. doi: 10.1158/0008-5472.CAN-13-1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Calais G., et al. Randomized trial of radiation therapy versus concomitant chemotherapy and radiation therapy for advanced-stage oropharynx carcinoma. J. Natl. Cancer Inst. 1999;91(24):2081–2086. doi: 10.1093/jnci/91.24.2081. [DOI] [PubMed] [Google Scholar]
- 84.Zorat P., et al. Randomized phase III trial of neoadjuvant chemotherapy in head and neck cancer: 10-year follow-up. J. Natl. Cancer Inst. 2004;96:1714–1717. doi: 10.1093/jnci/djh306. [DOI] [PubMed] [Google Scholar]
- 85.Patterson R., et al. Global burden of head and neck cancer: economic consequences, health, and the role of surgery. Otolaryngol Head Neck Surg. 2020;162(3):296–303. doi: 10.1177/0194599819897265. [DOI] [PubMed] [Google Scholar]
- 86.Du E., et al. Long‐term survival in head and neck cancer: impact of site, stage, smoking, and human papillomavirus status. Laryngoscope. 2019;129(11):2506–2513. doi: 10.1002/lary.27807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Colevas A.D. Chemotherapy options for patients with metastatic or recurrent squamous cell carcinoma of the head and neck. J. Clin. Oncol. 2006;24(17):2644–2652. doi: 10.1200/JCO.2005.05.3348. [DOI] [PubMed] [Google Scholar]
- 88.Marur S., Forastiere A.A. Head and neck squamous cell carcinoma: update on epidemiology, diagnosis, and treatment. Mayo Clin. Proc. 2016;91(3):386–396. doi: 10.1016/j.mayocp.2015.12.017. [DOI] [PubMed] [Google Scholar]
- 89.Kozakiewicz P., Grzybowska‑Szatkowska L. Application of molecular targeted therapies in the treatment of head and neck squamous cell carcinoma. Oncology letters. 2018;15(5):7497–7505. doi: 10.3892/ol.2018.8300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Moreira J., Tobias A., O’Brien M.P., Agulnik M. Targeted therapy in head and neck cancer: an update on current clinical developments in epidermal growth factor receptor-targeted therapy and immunotherapies. Drugs. 2017;77(8):843–857. doi: 10.1007/s40265-017-0734-0. [DOI] [PubMed] [Google Scholar]
- 91.Samra B., Tam E., Baseri B., Shapira I. Checkpoint inhibitors in head and neck cancer: current knowledge and perspectives. J. Invest. Med. 2018;66(7):1023–1030. doi: 10.1136/jim-2018-000743. [DOI] [PubMed] [Google Scholar]
- 92.Karabajakian A., et al. Chemotherapy for localized head and neck squamous cell cancers. Anticancer Drugs. 2017;28(4):362–368. doi: 10.1097/CAD.0000000000000480. [DOI] [PubMed] [Google Scholar]
- 93.Pignon J.-P., Aurelie L.M., Emilie M., et al. Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346 patients. Radiother oncol. 2009;92(1):4–14. doi: 10.1016/j.radonc.2009.04.014. [DOI] [PubMed] [Google Scholar]
- 94.Bernier J., Domenge C., Ozsahin M., et al. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N. Engl. J. Med. 2004;350(19):1945–1952. doi: 10.1056/NEJMoa032641. [DOI] [PubMed] [Google Scholar]
- 95.Posner M.R., Hershock D.M., Blajman C.R., et al. Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N. Engl. J. Med. 2007;357(17):1705–1715. doi: 10.1056/NEJMoa070956. [DOI] [PubMed] [Google Scholar]
- 96.Geoffrois L., Martin L., Garaud P., et al. Induction docetaxel platinum 5-FU (TPF) followed by cetuximab-radiotherapy (cetux-RT) versus concurrent chemo-radiotherapy (CT/RT) in patients with N2b/c-N3 non operated stage III-IV squamous cell cancer of the head and neck (SCCHN): Results of the GORTEC 2007-02 phase III randomized trial. J. Clin. Oncol. 2016;34(15_suppl):6000. doi: 10.1200/JCO.2016.34.15_suppl.6000. [DOI] [Google Scholar]
- 97.Ghi M.G., Paccagnella A., Ferrari D., et al. Concomitant chemoradiation (CRT) or cetuximab/RT (CET/RT) versus induction Docetaxel/ Cisplatin/5-Fluorouracil (TPF) followed by CRT or CET/RT in patients with Locally Advanced Squamous Cell Carcinoma of Head and Neck (LASCCHN). A randomized phase III factorial study ( NCT01086826) J. Clin. Oncol. 2014;32(15_suppl):6004. doi: 10.1200/jco.2014.32.15_suppl.6004. [DOI] [Google Scholar]
- 98.Cohen E.E.W., Karrison T.G., Kocherginsky M., et al. Phase III randomized trial of induction chemotherapy in patients with N2 or N3 locally advanced head and neck cancer. J. Clin. Oncol. 2014;32(25):2735. doi: 10.1200/JCO.2013.54.6309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Haddad R., O’Neil A., Rabinowits G., et al. Induction chemotherapy followed by concurrent chemoradiotherapy (sequential chemoradiotherapy) versus concurrent chemoradiotherapy alone in locally advanced head and neck cancer (PARADIGM): a randomised phase 3 trial. Lancet Oncol. 2013;14(3):257–264. doi: 10.1016/S1470-2045(13)70011-1. [DOI] [PubMed] [Google Scholar]
- 100.Fayette J., Fontaine-Delaruelle C., Ambrun A., et al. Neoadjuvant modified TPF (docetaxel, cisplatin, fluorouracil) for patients unfit to standard TPF in locally advanced head and neck squamous cell carcinoma: a study of 48 patients. Oncotarget. 2016;7(24):37297–37304. doi: 10.18632/oncotarget.8934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lefebvre J.L., Pointreau Y., Rolland F., et al. Induction chemotherapy followed by either chemoradiotherapy or bioradiotherapy for larynx preservation: the TREMPLIN randomized phase II study. J. Clin. Oncol. 2013;31(7):853–859. doi: 10.1200/JCO.2012.42.3988. [DOI] [PubMed] [Google Scholar]
- 102.Hitt R., Ricard M., Juan J.G., et al. Randomized phase III trial of induction chemotherapy (ICT) with docetaxel-cisplatin-5fluorouracil (DCF) followed by cisplatin-radiotherapy (CRT) or cetuximab-radiotherapy (CetRT) in patients (pts) with locally advanced unresectable head and neck cancer (LAUHNC) J. Clin. Oncol. 2016;34(15_suppl):6001. doi: 10.1200/JCO.2016.34.15_suppl.6001. [DOI] [Google Scholar]
- 103.Bauman J.E., Cohen E., Ferris R.L., et al. Immunotherapy of head and neck cancer: emerging clinical trials from a national cancer institute head and neck cancer steering committee planning meeting. Cancer. 2017;123(7):1259–1271. doi: 10.1002/cncr.30449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.de Biasi A.R., Villena-Vargas J., Adusumilli P.S. Cisplatin-induced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin Cancer Res. 2014;20(21):5384–5391. doi: 10.1158/1078-0432.CCR-14-1298. [DOI] [PMC free article] [PubMed] [Google Scholar]