Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 18.
Published in final edited form as: Eur J Cancer. 2016 Oct 15;68:125–133. doi: 10.1016/j.ejca.2016.09.006

Treatment de-intensification strategies for head and neck cancer

Jacqueline R Kelly a, Zain A Husain a, Barbara Burtness b,*
PMCID: PMC5734050  NIHMSID: NIHMS925700  PMID: 27755996

Abstract

Increasingly, squamous cell carcinoma of the oropharynx (OPSCC) is attributable to transformation resulting from high-risk human papillomavirus (HPV) infection. Such cancers are significantly more responsive to treatment than traditional tobacco- and alcohol-associated squamous cell cancers of the head and neck. Conventional management with definitive chemoradiation, surgery and adjuvant radiation, or radiation given with altered fractionation schemes, while effective, incurs long-term morbidity that escalates with treatment intensity and significantly impairs quality of life. Recent trials have suggested that less intensive treatment regimens may achieve similar efficacy with decreased toxicity. In this article, we review the primary strategies used for de-escalation of treatment, which include the reduction of radiation dose, substitution and/or elimination of concurrent radiosensitising chemotherapy, and the use of minimally invasive surgery. We discuss the rationale behind these approaches and the preliminary data demonstrating the success of de-escalation, as well as potential considerations raised by treatment de-intensification in HPV-associated OPSCC.

Keywords: Head and neck, Oropharynx, Human papillomavirus, Oropharyngeal neoplasms, Clinical trial, Radiotherapy, Chemotherapy, Surgery, Carcinoma, Squamous cell

1. Introduction

Epidemiologic trends from the 1980s onward have demonstrated a decline in the incidence of squamous cell cancers in the larynx, hypopharynx, and oral cavity [1], related to the parallel decline in tobacco use. However, the incidence of oropharyngeal squamous cell carcinoma (OPSCC) over this same period has increased, attributable to the rise of high-risk human papilloma-virus (HPV) infection leading to HPV-associated OPSCC [2,3]. The incidence of HPV-associated OPSSC now accounts for over 70% of newly diagnosed OPSCC in the United States [3]. The virus is thought to be transmitted sexually, and a high lifetime number of sexual partners is associated with an increased risk of oropharyngeal cancer, regardless of the presence of established risk factors including tobacco and alcohol use [4,5]. Clinical and demographic factors differ between patients with HPV-positive and HPV-negative disease; patients with HPV-positive OPSCC are typically younger and more likely to have nodal involvement than their HPV-negative counterparts [6].

HPV is a DNA virus that is implicated in the carcinogenesis of neoplasms in the cervix, oropharynx, and anus [7,8]. HPV DNA integrates with the host DNA, allowing for the production of viral proteins E6 and E7. These proteins interfere with the activation of host tumour suppressor proteins p53 and Rb, respectively [9]. While HPV-negative tumours commonly harbour mutated p53, HPV-associated OPSCC is typically p53 wild type [10,11]. Inactivation of Rb by E7 results in the overexpression of p16, a marker commonly used to identify HPV-associated cases [12]. Data from The Cancer Genome Atlas observed complex mutational patterns including loss of TRAF3, activating mutations of PIK3CA, and amplification of E2F1 in HPV-associated oropharyngeal cancers, pointing to aberrant activation of NF-κB, other oncogenic pathways, and cell cycle dysregulation as critical in the pathogenesis of these tumours [11]. Non-HPV-associated cancers have also been shown to have higher expression of the epidermal growth factor receptor (EGFR) than their HPV-associated counterparts [13]. Additionally, HPV-associated tumours may be less hypoxic [14], another factor predicted to enhance response to radiotherapy.

As both EGFR expression and p53 status are correlated with treatment responsiveness and survival, these biologic differences would be expected to contribute to differential outcomes in OPSCC based on HPV status. In fact, in multiple prospective trials published over the past decade, HPV positivity has conferred improved prognosis for patients with OPSCC compared with patients with similar stage HPV-negative tumours [1423]. Eastern Cooperative Oncology Group (ECOG) 2399 was the first prospective study to demonstrate improved outcomes in patients with HPV-associated disease. In this trial, patients with HPV-positive tumours had a superior response to induction chemotherapy (82% versus 55%, p = 0.01), chemoradiotherapy (84% versus 57%, p = 0.007) and improved 2-year overall survival (OS) (95% versus 62%, p = 0.005) [16].

The effect of HPV status on clinical outcomes was validated by Ang et al. in their retrospective analysis of the Radiation Therapy Oncology Group (RTOG) 0129 study cohort, in which 63.8% of patients with stage III–IV OPSCC were found to have HPV-associated cancers [15]. OS at 3 years was 82.4% in patients with HPV-associated disease and 57.1% in patients with non-HPV-associated cancer (p < 0.001). The risk of death increased significantly with each additional pack-year of smoking. Patients were thus grouped by risk: those with HPV-associated disease and ≤ 10 pack-year smoking history as well as those with HPV-associated disease, >10 pack-year smoking history, and N0–N2a disease were deemed low-risk, with a 3-year OS of 93%; those with HPV-associated disease, >10 pack-year smoking history, and N2b-N3 disease were intermediate-risk, with a 3-year OS of 70.8%. Of note, no patients with HPV-associated disease were classified as high-risk, with a 3-year OS of 46.2%.

However, the current standard of care for OPSCC is derived from older trials of head and neck cancer patients with predominately HPV-negative disease, potentially representing overtreatment of favourable-risk, HPV-positive patients. Given the different aetiology, natural history [2426], biomolecular signature [11,27], and treatment responsiveness, it is now accepted that HPV-positive and HPV-negative OPSCCs are distinct diseases to be studied separately in trials. Consequently, a number of clinical trials are underway to investigate strategies for the de-intensification of treatment in patients with HPV-associated OPSCC in order to minimise morbidity while maintaining excellent outcomes.

2. Rationale for treatment de-intensification

In medically fit patients with locally advanced head and neck cancer, the non-surgical standard of care is radiotherapy to a dose of 70 Gy with concurrent cisplatin, determined by prospective randomised trials as well as a meta-analysis of over 17,000 patients [2830]. While these publications demonstrated improved OS in patients treated with concurrent chemoradiation (5-year OS 33.7% versus 27.2% without chemotherapy) [28], this regimen incurs notable acute and long-term morbidity. For the treatment-responsive HPV-associated cancers, current therapies may be more intensive than necessary to achieve cure. Increasing the intensity of therapy with some combination of surgery, chemotherapy and/or radiation therapy has been shown to increase treatment-induced toxicity [31] as well as cost [32]. Published rates of gastrostomy tube placement, for example, range from 5% [33] to as high as 85% [34], depending on the patient population and the therapeutic modalities used.

Large portions of the pharyngeal axis and soft tissues in the neck receive high doses of radiation in patients undergoing definitive management for OPSCC; the toxic effects are augmented by radiosensitising chemotherapy. The rates of acute and late grade ≥ 3 toxicity are approximately 80% and 25%–60%, respectively [15,29,30]. Frequently occurring acute toxicities include dermatitis, mucositis, and dysphagia [23,24]. Long after the completion of definitive therapy, many patients endorse chronic dysphagia and xerostomia, which negatively impact their quality of life [35,36]. At 24 months after the receipt of radiotherapy (with or without chemotherapy) to the head and neck, 15% of patients were found to have grade ≥ 2 swallowing dysfunction, and 8% of patients had progressive dysphagia [37]. Patients with oropharyngeal primary cancers, who received chemoradiotherapy, or who underwent bilateral neck irradiation were more likely to have severe persistent dysphagia, underscoring the morbidity of present-day standard treatment for OPSCC patients.

Moreover, chronic effects such as dysphagia and speech-related toxicity have been shown to occur in a radiation dose-dependent fashion [3843]. Increasing dose to the supraglottic larynx is also associated with an increased likelihood of swallowing dysfunction [43]. Stricture and feeding tube dependence increase when the volume of pharyngeal constrictors receiving 70 Gy exceeds 50% and 30%, respectively, and aspiration increases when >50% of the pharyngeal constrictors receive 65 Gy [44,45]. Dysphagia increases with every 10 Gy above 55 Gy given to the superior and middle pharyngeal constrictors [46]. Therefore, a notable strategy for lessening treatment morbidity is to reduce the dose and volume of normal tissue irradiated. The adoption of intensity-modulated radiotherapy (IMRT) has already been shown to reduce the likelihood of late toxicities including xerostomia and dysphagia [43,47,48]. Nonetheless, further efforts should be made to reduce radiation-related morbidity, and radiation dose reduction may lead to improved functional outcomes, particularly if the dose is lowered below 55 Gy with regard to dysphagia.

The addition of chemotherapy to radiation is known to increase rates of acute and chronic toxicity. The Intergroup trial noted a 52% rate of grade ≥ 3 toxicity in the radiotherapy alone group, compared to a rate of 89.5% in the chemoradiotherapy cohort. Renal and hematologic toxicity, as well as nausea and vomiting, were significantly worse with the addition of cisplatin, and a trend for worsened mucositis was observed. Both arms including chemotherapy demonstrated a 10% increase in the placement of a gastrostomy tube when compared to radiotherapy alone (not significant) [30]. The Groupe Oncologie Radiotherapie Tete et Cou (GORTEC) 94-01 trial also demonstrated worse grade 3–4 toxicity (30%– 56%) with the addition of concurrent chemotherapy, though the number of patients in this subset was small, precluding detailed toxicity analysis [29]. Disturbingly, 10-year results of RTOG 91-11 demonstrated increased non-cancer mortality in the concomitant chemo-radiation arm (30.8%) when compared to the induction chemotherapy arm (20.8%) and the radiation-alone arm (16.9%), suggesting the current system of monitoring and grading late effects of treatment may be inadequate [49]. Worth noting, however, is that larynx cancer patients have higher baseline comorbidity and thus late cisplatin-related toxicity may not be mirrored in the more medically fit HPV-positive OPSCC patient population.

Concern for treatment-induced morbidity has prompted the initiation of multiple trials aiming to reduce treatment-related toxicity. Defining appropriate candidates for treatment de-intensification is critical in maintaining the excellent outcomes historically seen in patients with HPV-associated OPSCC. Several groups are currently examining how to best re-classify the American Joint Committee on Cancer (AJCC) staging groups for this patient population. The International Collaboration on Oropharyngeal cancer Network for Staging (ICON–S) group recently proposed a new staging system for HPV-associated OPSCC [50]. Examining the 5-year OS of a multi-institutional, international cohort of over 1900 patients with HPV-associated OPSCC, the authors used a recursive partitioning analysis to create the novel staging system. Using the AJCC 7th edition tumour node metastasis (TNM) classification, the proposed ICON–S stage groupings are stage I (T1–T2N0–N2b), stage II (T1–T2N2c or T3N0–N2c), and stage III (T4a–T4b or N3). The proportion of patients alive in the validation cohort at 5 years with ICON–S stage I, II, and III disease was 88%, 81%, and 65%, respectively. Metastatic disease (M1) is classified as ICON–S stage IV. It is worth noting that while smoking pack-years were shown to impact OS, the ICON–S staging system does not account for smoking history. An alternate grouping was proposed by Huang et al. using a recursive portioning analysis that included AJCC 7th edition TNM stage, age, and smoking pack-years to derive the following four valid prognostic groups for survival: group I (T1–3N0–N2c with ≤ 20 pack-years), group II (T1–3N0–N2c with >20 pack-years), group III (T4 or N3 and age ≤ 70 years), and group IVA (T4 or N3 and age > 70 years); with a 5-year median OS of 89%, 64%, 57%, and 40%, respectively [51]. Patients with lower T and N stage disease and minimal smoking history are thus the ideal candidates for possible de-intensification of therapy. Current trials explore de-intensification of treatment largely in this patient population (Table 1).

Table 1.

De-intensification trials in HPV-associated OPSCC.

Trial Phase N Inclusion criteria Treatment
Chemotherapy de-intensification trials
RTOG 1016 (NCT01302834) III 706 T1–2, N2a–3, or T3–4, any N, HPV-positive OPSCC Cetuximab versus high-dose cisplatin concurrent with accelerated IMRT (70 Gy in 6 weeks)
De-ESCALaTE HPV (NCT01874171) III 304 Stage III–IVA HPV-positive OPSCC (T3N0–T4N0, T1N1 –T4N3). Excludes > N2b, >10 PY Cetuximab versus high-dose cisplatin concurrent with RT (70 Gy)
TROG 12.01 (NCT01855451) III 200 Stage III (excluding T1–2, N1) or IV (excluding T4, N3, or M1) HPV-positive OPSCC if ≤10 PY. If >10 PY, only N0 –2a Cetuximab versus weekly cisplatin concurrent with RT (70 Gy) once per week
Radiotherapy de-intensification trials
NRG HN-002 (NCT02254278) II 296 T1–2, N1–2b, or T3, N0–2b disease and <10 PY HPV-positive OPC Reduced-dose IMRT (60 Gy) with/without weekly cisplatin
NCT01530997 II 40 T1–3, N0–2c HPV-positive OPSCC if <10 PY or >5 years of abstinence IMRT (54–60 Gy) with weekly cisplatin (30 mg/m2)
ECOG 1308 (NCT01084083) II 80 Resectable stages IIIA/IIIB and IVA/IVB
HPV-positive OPSCC (p16-high or HPV-16 ISH positive)
IC, then response-adapted RT (54 or 66–70 Gy) with cetuximab
The Quarterback Trial (NCT01706939) III 365 Stage III/IV (M0) HPV-associated OPSCC/unknown primary/nasopharynx. Excludes active smokers/>20 PY IC with TPF: patients with CR/PR randomly assigned 2:1 to carboplatin with RT (56 versus 70 Gy) per week. Non-responders receive standard RT.
De-intensification of surgery/adjuvant therapy
ECOG 3311 (NCT01898494) II 377 Resectable stage III–IVB p16-positive OPSCC TORS then risk-adapted post-operative treatment (observation/50 versus 60/66 Gy with weekly platinum)
PATHOS trial (NCT02215265) II/III 242 Resectable T1–T3, N0–2b HPV-positive OPSCC.
Excludes active smokers with N2b disease
TORS then re-adapted post-operative treatment (observation/50 versus 60Gy/60 Gy with or without weekly cisplatin)
ADEPT (NCT01687413) III 500 Transoral resected p16-positive OPSCC (R0 margin), T1–4a, pN positive with ECE Post-operative adjuvant 60-Gy RT with or without weekly cisplatin
NCT01932697 II 40 P16-positive OPSCC (R0 margin), stage I–IVB. Excludes ≥ 10 PY or smoking within 5 years Surgery followed by hyperfractionated IMRT (36 Gy/20 fractions BID) + weekly docetaxel

Abbreviations: ADEPT, Post Operative Adjuvant Therapy De-intensification Trial for Human Papillomavirus-related P16 + Oropharynx Cancer; CR, complete response; De-ESCALATE, Determination of Cetuximab Versus Cisplatin Early and Late Toxicity Events in Human Papillomavirus + Oropharyngeal Squamous Cell Carcinoma; ECE, extracapsular extension; ECOG, Eastern Cooperative Oncology Group; Gy, Gray; HPV, human papillomavirus; IC, induction chemotherapy; IMRT, intensity-modulated radiotherapy; ISH, in situ hybridization; N, number of patients; NRG-HN002, HN002 study by NRG (group comprising National Surgical Adjuvant Breast and Bowel Project, Radiation Therapy Oncology Group, and Gynecologic Oncology Group); OPSSC, oropharyngeal squamous cell cancer; PATHOS, Post-operative Adjuvant Treatment for HPV-Positive Tumours; PR, partial response; PY, pack-years; RT, radiotherapy; RTOG, Radiation Therapy Oncology Group; TORS, transoral robotic surgery; TPF, docetaxel, cisplatin, fluorouracil; TROG, Trans Tasman Radiation Oncology Group.

Adapted from the review by Bhatia and Burtness [69].

3. De-intensification trials for HPV-associated OPSCC

3.1. Reducing chemotherapy-related toxicity

One strategy for reducing chemotherapy-related toxicity is the replacement of cisplatin with cetuximab, a monoclonal antibody targeting the EGFR extracellular ligand binding domain. This anti-EGFR therapy was approved by the regulatory bodies in both the United States and Europe for the treatment of patients with locally advanced head and neck squamous cell carcinoma on the basis of the Bonner trial, which demonstrated improved 3-year OS in those treated with combined cetuximab and radiotherapy when compared to radiotherapy alone, 55% versus 45%, respectively (p = 0.05) [52]. On subgroup analysis, the survival was greatest among patients with oropharyngeal primary cancers, younger age, high nodal stage, and low tumour stage—factors typical of HPV-associated cases. Furthermore, grade ≥ 3 toxicity was similar between the two groups, with the exception of higher rates of acneiform rash (17% versus 1%, p < 0.001) and infusion reaction (3% versus 0%, p =0.01) in the cetuximab arm. That cetuximab did not appear to increase the severity of typical radiation-induced toxicity, such as mucositis and dysphagia, rendered this therapy an enticing candidate for treatment de-escalation.

A recently published secondary analysis based on p16 status confirmed the association between p16 status and improved outcome; moreover, the addition of cetuximab to radiotherapy had its greatest benefit in HPV-associated OPSCC (locoregional control and OS hazard ratio 0.31 and 0.38, respectively) [53].

RTOG 1016 (NCT01302834) explored the replacement of cisplatin with cetuximab. This phase III non-inferiority trial stratified patient by T stage, N stage, Zubrod performance status, and smoking history and compared cetuximab concurrent with accelerated radiotherapy (70 Gy given six fractions per week for 6 weeks) to cisplatin days 1 and 22 with the same accelerated radiotherapy course for 987 patients with p16 positive stage III/IV OPSCC. Accrual for this trial has been completed, and the estimated primary completion date is in 2020. The Determination of Cetuximab Versus Cisplatin Early and Late Toxicity Events in Human Papillomavirus + Oropharyngeal Squamous Cell Carcinoma trial (NCT01874171) is a randomised phase III trial currently accruing patients with stage III–IVa HPV-associated OPSCC with ≤ 10 pack-year smoking history that used a similar study schema (cisplatin-based chemoradiotherapy to 70 Gy versus cetuximab concurrent with the same radiation) to compare toxicity outcomes between the two cohorts. The Trans-Tasman Radiation Oncology Group is also enrolling on a similar phase III trial (NCT01855451), but with weekly cisplatin, in order to examine symptom severity. Ultimately these studies will confirm or refute the idea that cetuximab plus 70 Gy achieves similar rates of cure as cisplatin plus radiation, and will also better delineate the toxicities associated with either approach, which may differ based on smoking history and T stage.

An alternative de-intensification approach is the omission of chemotherapy. The ongoing HN-002 trial (NCT02254278) is a large randomised phase II study of nearly 300 patients assessing two different de-escalation strategies. Eligible patients are those with HPV-associated T1–T3 tumours with N0–N2b nodal status and ≤ 10 pack-years smoking history, falling under the low-risk classification put forth by Ang et al. [15]. Patients are randomised to receive accelerated radiotherapy alone to 60 Gy given six fractions a week over 5 weeks versus 60 Gy using standard fractionation over 6 weeks with dose-reduced weekly cisplatin (40 mg/m2 weekly, total 240 mg/m [2]). Retrospective data suggest that the weekly chemotherapy schedule results in significantly reduced severity of mucositis compared to the traditional schedule of delivery every 3 weeks [54]. Notably, both arms represent a 10 Gy reduction in total radiotherapy dose. The primary end-point of the study is 2-year progression-free survival of at least 85% as well acceptable swallowing function based on the MD Anderson Dysphagia Inventory. This combination efficacy and toxicity end-point is novel, and demonstrates the emphasis of selecting therapies in which patients are likely to be cured, but also have minimal long-term morbidity.

3.2. Reduction of radiotherapy dose

Because radiotherapy-related toxicity is dose-dependent, Chera et al. conducted a phase II study examining the dose reduction of radiotherapy in low-risk patients with minimal smoking history who underwent chemo-radiation for T0–3 N0–2c M0 HPV-associated OPSCC (NCT01530997). Treatment was limited to 60 Gy IMRT delivered over 6 weeks with concurrent weekly cisplatin (30 mg/m2). All patients received post-treatment biopsy of the primary site and resection of involved nodes at 4–14 weeks following completion of therapy, possibly reflecting the investigators’ uncertainty about whether 60 Gy would suffice to eradicate gross disease. The pathologic complete response rate was 86%. The rates of grade ≥3 toxicities compare favourably to historical controls [55], with a gastrostomy tube rate of 39% and a 0% rate of long-term feeding tube dependence on this study. These results are particularly impressive given data demonstrating that undergoing post-treatment neck dissection after radiotherapy or chemo-radiotherapy significantly increases the likelihood of feeding tube dependence 24 months after completing therapy (relative risk [RR] 7.66, 95% confidence interval [CI] 2.07–10.89) [56]. The group is conducting larger follow-up studies without the need for biopsies and resection of neck nodes, however these early data suggest that de-escalation 60 Gy plus weekly cisplatin may suffice in appropriately selected patients.

The NRG HN-002 (HN002 study by NRG, group comprising National Surgical Adjuvant Breast and Bowel Project, Radiation Therapy Oncology Group, and Gynecologic Oncology Group) trial, previously mentioned for its omission of cisplatin, also explores radiotherapy dose reduction in both arms. In the low-risk arm, radiation-alone is given to a dose of only 60 Gy in standard 2 Gy fractions, albeit in an accelerated fashion with the delivery of six fractions per week rather than five. The standard five fractions per week schedule is used in the arm with concurrent dose-reduced cisplatin.

Chemoselection, the use of induction chemotherapy to identify patients with disease sensitive to therapy, has been incorporated in a novel paradigm administering lower doses of radiation in chemotherapy responsive HPV-associated cancer. ECOG 1308 (NCT01084083), a phase II trial, examined a de-intensification strategy of radiation dose reduction based on response to induction chemotherapy in patients with resectable stage III-IV T3–4b N0–3 HPV-associated OPSCC. In this study, patients received 3 cycles of induction chemotherapy with cisplatin, paclitaxel, and cetuximab. Patients with a clinical complete response at the primary site went on to receive radiotherapy to 54 Gy using standard 2 Gy fractions over 51/2 weeks with concurrent cetuximab. Patients with a clinical partial response or stable disease at the primary site received a more aggressive regimen, with radiotherapy to 69.3 Gy in 2.1 Gy fractions over 61/2 weeks with concurrent cetuximab.

Early results of this trial are excellent. Seventy-one percent of patients demonstrated a clinical complete response at the primary site. One-year progression-free survival rates ranged from 87% in patients without a complete response who received 69.3 Gy plus cetuximab up to 97% in the most favourable subset of patients. Toxicity results were also promising, with only 1% of patients exhibiting grade 4 toxicity (lymphopenia) and low rates of grade 3 toxicity [57]. The lower IMRT dose used in the favourable arm of this study has also been shown to ameliorate late toxicities, reaching a statistically significant improvement in patient-reported difficulty swallowing solids (100% versus 35%) [58]. This finding is critical, as it supports the hypothesis that doses <55 Gy are less likely to impair swallowing function [46], suggesting that de-escalation of radiotherapy dose to 60 Gy may not be sufficient to significantly improve the incidence of dysphagia.

The Quarterback Trial (NCT01706939), a phase III non-inferiority trial, is currently recruiting participants to further explore chemoselection using induction chemotherapy in patients with stage III–IV non-metastatic HPV-associated OPSCC, nasopharynx cancer, or head and neck cancer of an unknown primary with ≤20 pack-year smoking history. Patients will receive three cycles of docetaxel, cisplatin, and 5-FU induction therapy. Those with a clinical complete response will undergo a 2:1 randomization to reduced- (56 Gy) or standard- (70 Gy) dose radiotherapy with weekly carboplatin and cetuximab or carboplatin only.

3.3. Reducing surgical morbidity

Surgical resection is a modality often applied in the management of OPSCC. Until recently, a mandibulotomy was required to gain sufficient access to the oropharynx for resection, resulting in severe functional and cosmetic detriments [59]. Nearly 25% of these patients were noted to have severe complications, while operative mortality was around 3% [60]. Recent technological advancements led to the development of transoral approaches including transoral robotic surgery (TORS), which utilises miniaturised instruments to perform resection of select pharyngeal tumours through the open mouth. When coupled with magnification, it allows for accurate dissection without the morbidity and functional deficits of open surgery [59,61]. Some studies suggest that TORS may improve rates of margin-negative mucosal resections, potentially reducing the need for adjuvant chemotherapy in addition to radiotherapy [62].

While minimally invasive surgery reduces morbidity, the treatment-related toxicity is still considerable, particularly in patients who receive adjuvant treatment with radiotherapy or chemoradiation. For instance, one analysis showed a 0% rate of gastrostomy tube use in T1/T2 OPSCC patients treated with TORS alone versus a 44.4% rate of gastrostomy tube use and a 22.2% rate of gastrostomy tube dependence 1 year after TORS in patients who required adjuvant therapy [63]. To further reduce morbidity after surgery, ongoing trials explore reducing the dose of adjuvant radiotherapy or eliminating adjuvant chemotherapy in patients with pathologic evidence of extracapsular extension (ECE)—a departure from the standard of care adjuvant chemoradiation established for these patients based on the combined analysis of EORTC 22931 and RTOG 9501 [64].

ECOG 3311 (NCT01898494) was therefore designed to explore the combination of TORS and risk-based, de-intensified adjuvant radiotherapy or chemoradiotherapy in patients with clinical T1–2 N0–1 HPV-associated OPSCC. Patients deemed low-risk (margin-negative resection, N0 or N1 disease, no ECE) after resection are observed, intermediate-risk patients (negative but ≤ 1 mm margin, +ECE, N2a, or 2–4 involved lymph nodes) receive adjuvant radiotherapy alone with 50 versus 60 Gy using standard daily fractionation, and high-risk patients (>1 mm ECE, positive margins, or ≥ 5 lymph nodes positive) are treated with 66 Gy and weekly cisplatin. Results of this trial will determine if the combination of minimally invasive surgical techniques with reduced intensity adjuvant therapy proves to be less toxic than current standard of care management, and may justify a follow up phase III trial comparing TORS and reduced dose post-operative radiation therapy with standard chemoradiation.

Similar to ECOG 3311, the Post-operative Adjuvant Treatment for HPV-Positive Tumours trial (NCT02215265) also calls for risk-based stratification after TORS. While the low-risk classification is the same as ECOG 3311, the other risk group criteria vary. The intermediate-risk criteria are negative margin, ≥ pT3 disease, or pT1-2 disease with pN2a/b, perineural invasion, lymphovascular invasion, or negative but close margin (<5 mm). These patients receive either 60 Gy in 2 Gy fractions over 6 weeks or 50 Gy in 2 Gy fractions over 5 weeks. The high-risk criteria include positive margin or ECE, and these patients are assigned to receive 60 Gy with or without concurrent cisplatin.

Eliminating adjuvant chemotherapy in postoperative patients with ECE is yet another strategy for minimizing treatment-associated morbidity. The Washington University group published provocative data suggesting that the presence of ECE may not affect disease-free survival (DFS) in surgically managed patients. In their retrospectively assembled experience of 151 p16 + OPSCC patients after TORS, the 3-year DFS of patients with and without ECE were 89% (95% CI 84%–95%) and 94% (95% CI 83%–100%), respectively [65]. A study out of the University of Pittsburgh yielded similar findings [66], with a 5-year update showing a disease specific survival of 89.3% (95% CI 73.9%–98.1%) in patients with ECE and 84.8% (95% CI 64.4%–94.1%) in patients without ECE [67]. The Washington University group has also published a retrospective comparison of patients with ECE that received either adjuvant radiotherapy alone or chemoradiation. In the analysis, 48 patients received radiation alone while 65 were treated with chemoradiation. The 3-year DFS rates were similar at 94.5% in the radiation alone cohort and 91.8% in the chemoradiation group (p = 0.74) [65].

Bearing in mind these data from small, single institution retrospective series, the Post Operative Adjuvant Therapy De-intensification Trial for Human Papillomavirus-related P16 + Oropharynx Cancer trial (NCT01687413) was designed to investigate the role of adjuvant chemotherapy in patients with ECE. In this study, T1–4a node-positive patients with HPV-associated OPSCC with ECE but negative margins receive either standard 60 Gy with concurrent weekly cisplatin or 60 Gy without chemotherapy.

Finally, a Mayo Clinic phase II trial (NCT01932697) is exploring the use of an accelerated hyperfractionation with twice daily radiation in post-operative cases. The total radiation dose of 36 Gy is delivered in 20 fractions in 1.8 Gy twice daily dosing 5 d/week with concurrent weekly docetaxel. This reduced radiation dosing is inspired by studies showing lower doses are able to control disease in anal cancer, another HPV-associated squamous cell cancer [68]. Eligible patients must have at least one of the following risk factors: lymph node >3 cm, two or more metastatic lymph nodes, perineural invasion, lymphovascular invasion, ECE, pT3, or microscopic pT4a tumour stage. The combined results of these surgical trials should yield more information on proper risk stratification and adjuvant treatment selection in the coming years.

4. Conclusions

The aforementioned ongoing trials will elucidate many of the current questions regarding the standard of care management of HPV-associated OPSCC. In the future, these patients will likely be managed with de-intensified therapies, whether it is through de-intensification of surgery and adjuvant treatment, omission, substitution or reduction of the dose of chemotherapy, and/or the reduction radiotherapy dose. Appropriate selection of patients for treatment de-intensification is critical in order to avoid jeopardizing the excellent outcomes that are achieved with the current standard of care therapy in this patient population; therefore de-intensification is presently not recommended outside of a clinical trial. A better understanding of the molecular alterations underlying HPV-associated disease should be elucidated to provide further opportunities for targeted therapies with less toxicity. If these approaches prove adequate, the quality of life of patients should be significantly improved.

Acknowledgments

No funding sources have been utilised in the production of this manuscript.

Footnotes

Conflict of interest statement

None declared.

References

  • 1.Sturgis EM, Ang KK. The epidemic of HPV-associated oropharyngeal cancer is here: is it time to change our treatment paradigms? J Natl Compr Canc Netw. 2011;9(6):665–73. doi: 10.6004/jnccn.2011.0055. [DOI] [PubMed] [Google Scholar]
  • 2.Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29(32):4294–301. doi: 10.1200/JCO.2011.36.4596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mehanna H, Beech T, Nicholson T, El-Hariry I, McConkey C, Paleri V, et al. Prevalence of human papillomavirus in oropharyngeal and nonoropharyngeal head and neck cancer–systematic review and meta-analysis of trends by time and region. Head Neck. 2013;35(5):747–55. doi: 10.1002/hed.22015. [DOI] [PubMed] [Google Scholar]
  • 4.D’Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, et al. Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med. 2007;356(19):1944–56. doi: 10.1056/NEJMoa065497. [DOI] [PubMed] [Google Scholar]
  • 5.Gillison ML, D’Souza G, Westra W, Sugar E, Xiao W, Begum S, et al. Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers. J Natl Cancer Inst. 2008;100(6):407–20. doi: 10.1093/jnci/djn025. [DOI] [PubMed] [Google Scholar]
  • 6.Khode SR, Dwivedi RC, Rhys-Evans P, Kazi R. Exploring the link between human papilloma virus and oral and oropharyngeal cancers. J Cancer Res Ther. 2014;10(3):492–8. doi: 10.4103/0973-1482.138213. [DOI] [PubMed] [Google Scholar]
  • 7.Koerber SA, Schoneweg C, Slynko A, Krug D, Haefner MF, Herfarth K, et al. Influence of human papillomavirus and p16(INK4a) on treatment outcome of patients with anal cancer. Radiother Oncol. 2014;113(3):331–6. doi: 10.1016/j.radonc.2014.11.013. [DOI] [PubMed] [Google Scholar]
  • 8.Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, et al. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol. 1999;189(1):12–9. doi: 10.1002/(SICI)1096-9896(199909)189:1<12::AID-PATH431>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 9.Smeets SJ, van der Plas M, Schaaij-Visser TB, van Veen EA, van Meerloo J, Braakhuis BJ, et al. Immortalization of oral keratinocytes by functional inactivation of the p53 and pRb pathways. Int J Cancer. 2011;128(7):1596–605. doi: 10.1002/ijc.25474. [DOI] [PubMed] [Google Scholar]
  • 10.Licitra L, Perrone F, Bossi P, Suardi S, Mariani L, Artusi R, et al. High-risk human papillomavirus affects prognosis in patients with surgically treated oropharyngeal squamous cell carcinoma. J Clin Oncol. 2006;24(36):5630–6. doi: 10.1200/JCO.2005.04.6136. [DOI] [PubMed] [Google Scholar]
  • 11.Cancer Genome Atlas N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517(7536):576–82. doi: 10.1038/nature14129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Khleif SN, DeGregori J, Yee CL, Otterson GA, Kaye FJ, Nevins JR, et al. Inhibition of cyclin D-CDK4/CDK6 activity is associated with an E2F-mediated induction of cyclin kinase inhibitor activity. Proc Natl Acad Sci U S A. 1996;93(9):4350–4. doi: 10.1073/pnas.93.9.4350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kim SH, Koo BS, Kang S, Park K, Kim H, Lee KR, et al. HPV integration begins in the tonsillar crypt and leads to the alteration of p16, EGFR and c-myc during tumor formation. Int J Cancer. 2007;120(7):1418–25. doi: 10.1002/ijc.22464. [DOI] [PubMed] [Google Scholar]
  • 14.Rischin D, Young RJ, Fisher R, Fox SB, Le QT, Peters LJ, et al. Prognostic significance of p16INK4A and human papillomavirus in patients with oropharyngeal cancer treated on TROG 02. 02 phase III trial. J Clin Oncol. 2010;28(27):4142–8. doi: 10.1200/JCO.2010.29.2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tân PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35. doi: 10.1056/NEJMoa0912217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fakhry C, Westra WH, Li S, Cmelak A, Ridge JA, Pinto H, et al. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. 2008;100(4):261–9. doi: 10.1093/jnci/djn011. [DOI] [PubMed] [Google Scholar]
  • 17.Huang SH, O’Sullivan B, Xu W, Zhao H, Chen DD, Ringash J, et al. Temporal nodal regression and regional control after primary radiation therapy for N2-N3 head-and-neck cancer stratified by HPV status. Int J Radiat Oncol Biol Phys. 2013;87(5):1078–85. doi: 10.1016/j.ijrobp.2013.08.049. [DOI] [PubMed] [Google Scholar]
  • 18.Lassen P, Eriksen JG, Krogdahl A, Therkildsen MH, Ulhøi BP, Overgaard M, et al. The influence of HPV-associated p16-expression on accelerated fractionated radiotherapy in head and neck cancer: evaluation of the randomised DAHANCA 6&7 trial. Radiother Oncol. 2011;100(1):49–55. doi: 10.1016/j.radonc.2011.02.010. [DOI] [PubMed] [Google Scholar]
  • 19.Lorch JH, Goloubeva O, Haddad RI, Cullen K, Sarlis N, Tishler R, et al. Induction chemotherapy with cisplatin and fluorouracil alone or in combination with docetaxel in locally advanced squamous-cell cancer of the head and neck: long-term results of the TAX 324 randomised phase 3 trial. Lancet Oncol. 2011;12(2):153–9. doi: 10.1016/S1470-2045(10)70279-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nguyen-Tan PF, Zhang Q, Ang KK, Weber RS, Rosenthal DI, Soulieres D, et al. Randomized phase III trial to test accelerated versus standard fractionation in combination with concurrent cisplatin for head and neck carcinomas in the Radiation Therapy Oncology Group 0129 trial: long-term report of efficacy and toxicity. J Clin Oncol. 2014 doi: 10.1200/JCO.2014.55.3925. JCO. 2014.55. 3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Posner MR, Lorch JH, Goloubeva O, Tan M, Schumaker LM, Sarlis NJ, et al. Survival and human papillomavirus in oropharynx cancer in TAX 324: a subset analysis from an international phase III trial. Ann Oncol. 2011;22(5):1071–7. doi: 10.1093/annonc/mdr006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Vermorken JB, Psyrri A, Mesia R, Peyrade F, Beier F, de Blas B, et al. Impact of tumor HPV status on outcome in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck receiving chemotherapy with or without cetuximab: retrospective analysis of the phase III EXTREME trial. Ann Oncol. 2014;25(4):801–7. doi: 10.1093/annonc/mdt574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Egloff AM, Lee JW, Langer CJ, Quon H, Vaezi A, Grandis JR, et al. Phase II study of cetuximab in combination with cisplatin and radiation in unresectable, locally advanced head and neck squamous cell carcinoma: Eastern cooperative oncology group trial E3303. Clin Cancer Res. 2014;20(19):5041–51. doi: 10.1158/1078-0432.CCR-14-0051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fakhry C, Zhang Q, Nguyen-Tan PF, Rosenthal D, El-Naggar A, Garden AS, et al. Human papillomavirus and overall survival after progression of oropharyngeal squamous cell carcinoma. J Clin Oncol. 2014;32(30):3365–73. doi: 10.1200/JCO.2014.55.1937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.O’Sullivan B, Huang SH, Siu LL, Waldron J, Zhao H, Perez-Ordonez B, et al. Deintensification candidate subgroups in human papillomavirus-related oropharyngeal cancer according to minimal risk of distant metastasis. J Clin Oncol. 2013;31(5):543–50. doi: 10.1200/JCO.2012.44.0164. [DOI] [PubMed] [Google Scholar]
  • 26.Argiris A, Li S, Ghebremichael M, Egloff AM, Wang L, Forastiere AA, et al. Prognostic significance of human papillomavirus in recurrent or metastatic head and neck cancer: an analysis of Eastern Cooperative Oncology Group trials. Ann Oncol. 2014;25(7):1410–6. doi: 10.1093/annonc/mdu167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Slebos RJ, Yi Y, Ely K, Carter J, Evjen A, Zhang X, et al. Gene expression differences associated with human papillomavirus status in head and neck squamous cell carcinoma. Clin Cancer Res. 2006;12(3 Pt 1):701–9. doi: 10.1158/1078-0432.CCR-05-2017. [DOI] [PubMed] [Google Scholar]
  • 28.Pignon JP, le Maitre A, Maillard E, Bourhis J, Group M-NC. 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]
  • 29.Denis F, Garaud P, Bardet E, Alfonsi M, Sire C, Germain T, et al. Final results of the 94-01 French Head and Neck Oncology and Radiotherapy Group randomized trial comparing radiotherapy alone with concomitant radiochemotherapy in advanced-stage oropharynx carcinoma. J Clin Oncol. 2004;22(1):69–76. doi: 10.1200/JCO.2004.08.021. [DOI] [PubMed] [Google Scholar]
  • 30.Adelstein DJ, Li Y, Adams GL, Wagner H, Jr, Kish JA, Ensley JF, et al. An intergroup phase III comparison of standard radiation therapy and two schedules of concurrent chemoradiotherapy in patients with unresectable squamous cell head and neck cancer. J Clin Oncol. 2003;21(1):92–8. doi: 10.1200/JCO.2003.01.008. [DOI] [PubMed] [Google Scholar]
  • 31.Trotti A, Pajak TF, Gwede CK, Paulus R, Cooper J, Forastiere A, et al. TAME: development of a new method for summarising adverse events of cancer treatment by the Radiation Therapy Oncology Group. Lancet Oncol. 2007;8(7):613–24. doi: 10.1016/S1470-2045(07)70144-4. [DOI] [PubMed] [Google Scholar]
  • 32.Sher DJ, Fidler MJ, Tishler RB, Stenson K, al-Khudari S. Cost-effectiveness analysis of chemoradiation therapy versus transoral robotic surgery for human papillomavirus–associated, clinical N2 oropharyngeal cancer. Int J Radiat Oncol Biol Phys. 2016;94(3):512–22. doi: 10.1016/j.ijrobp.2015.11.006. [DOI] [PubMed] [Google Scholar]
  • 33.Kelly K, Johnson-Obaseki S, Lumingu J, Corsten M. Oncologic, functional and surgical outcomes of primary Transoral Robotic Surgery for early squamous cell cancer of the oropharynx: a systematic review. Oral Oncol. 2014;50(8):696–703. doi: 10.1016/j.oraloncology.2014.04.005. [DOI] [PubMed] [Google Scholar]
  • 34.Haddad R, O’Neill A, Rabinowits G, Tishler R, Khuri F, Adkins D, 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):257–64. doi: 10.1016/S1470-2045(13)70011-1. [DOI] [PubMed] [Google Scholar]
  • 35.Boscolo-Rizzo P, Stellin M, Fuson R, Marchiori C, Gava A, Da Mosto MC. Long-term quality of life after treatment for locally advanced oropharyngeal carcinoma: surgery and postoperative radiotherapy versus concurrent chemoradiation. Oral Oncol. 2009;45(11):953–7. doi: 10.1016/j.oraloncology.2009.06.005. [DOI] [PubMed] [Google Scholar]
  • 36.Verdonck-de Leeuw IM, Buffart LM, Heymans MW, Rietveld DH, Doornaert P, de Bree R, et al. The course of health-related quality of life in head and neck cancer patients treated with chemoradiation: a prospective cohort study. Radiother Oncol. 2014;110(3):422–8. doi: 10.1016/j.radonc.2014.01.002. [DOI] [PubMed] [Google Scholar]
  • 37.Christianen ME, Verdonck-de Leeuw IM, Doornaert P, Chouvalova O, Steenbakkers RJ, Koken PW, et al. Patterns of long-term swallowing dysfunction after definitive radiotherapy or chemoradiation. Radiother Oncol. 2015;117(1):139–44. doi: 10.1016/j.radonc.2015.07.042. [DOI] [PubMed] [Google Scholar]
  • 38.Mortensen HR, Jensen K, Aksglaede K, Behrens M, Grau C. Late dysphagia after IMRT for head and neck cancer and correlation with dose-volume parameters. Radiother Oncol. 2013;107(3):288–94. doi: 10.1016/j.radonc.2013.06.001. [DOI] [PubMed] [Google Scholar]
  • 39.Frowen J, Hornby C, Collins M, Senthi S, Cassumbhoy R, Corry J. Reducing posttreatment dysphagia: Support for the relationship between radiation dose to the pharyngeal constrictors and swallowing outcomes. Pract Radiat Oncol. 2013;3(4):e187–94. doi: 10.1016/j.prro.2012.11.009. [DOI] [PubMed] [Google Scholar]
  • 40.Vlacich G, Spratt DE, Diaz R, Phillips JG, Crass J, Li CI, et al. Dose to the inferior pharyngeal constrictor predicts prolonged gastrostomy tube dependence with concurrent intensity-modulated radiation therapy and chemotherapy for locally-advanced head and neck cancer. Radiother Oncol. 2014;110(3):435–40. doi: 10.1016/j.radonc.2013.12.007. [DOI] [PubMed] [Google Scholar]
  • 41.Mazzola R, Ricchetti F, Fiorentino A, Fersino S, Giaj Levra N, Naccarato S, et al. Dose-volume-related dysphagia after constrictor muscles definition in head and neck cancer intensity-modulated radiation treatment. Br J Radiol. 2014;87(1044):20140543. doi: 10.1259/bjr.20140543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Teguh DN, Levendag PC, Ghidey W, van Montfort K, Kwa SL. Risk model and nomogram for dysphagia and xerostomia prediction in head and neck cancer patients treated by radiotherapy and/or chemotherapy. Dysphagia. 2013;28(3):388–94. doi: 10.1007/s00455-012-9445-6. [DOI] [PubMed] [Google Scholar]
  • 43.Christianen ME, Schilstra C, Beetz I, Muijs CT, Chouvalova O, Burlage FR, et al. Predictive modelling for swallowing dysfunction after primary (chemo)radiation: results of a prospective observational study. Radiother Oncol. 2012;105(1):107–14. doi: 10.1016/j.radonc.2011.08.009. [DOI] [PubMed] [Google Scholar]
  • 44.Feng FY, Kim HM, Lyden TH, Haxer MJ, Worden FP, Feng M, et al. Intensity-modulated chemoradiotherapy aiming to reduce dysphagia in patients with oropharyngeal cancer: clinical and functional results. J Clin Oncol. 2010;28(16):2732–8. doi: 10.1200/JCO.2009.24.6199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Levendag PC, Teguh DN, Voet P, van der Est H, Noever I, de Kruijf WJ, et al. Dysphagia disorders in patients with cancer of the oropharynx are significantly affected by the radiation therapy dose to the superior and middle constrictor muscle: a dose-effect relationship. Radiother Oncol. 2007;85(1):64–73. doi: 10.1016/j.radonc.2007.07.009. [DOI] [PubMed] [Google Scholar]
  • 46.Eisbruch A, Schwartz M, Rasch C, Vineberg K, Damen E, Van As CJ, et al. Dysphagia and aspiration after chemoradiotherapy for head-and-neck cancer: which anatomic structures are affected and can they be spared by IMRT? Int J Radiat Oncol Biol Phys. 2004;60(5):1425–39. doi: 10.1016/j.ijrobp.2004.05.050. [DOI] [PubMed] [Google Scholar]
  • 47.McBride SM, Parambi RJ, Jang JW, Goldsmith T, Busse PM, Chan AW. Intensity-modulated versus conventional radiation therapy for oropharyngeal carcinoma: long-term dysphagia and tumor control outcomes. Head Neck. 2014;36(4):492–8. doi: 10.1002/hed.23319. [DOI] [PubMed] [Google Scholar]
  • 48.van Rij CM, Oughlane-Heemsbergen WD, Ackerstaff AH, Lamers EA, Balm AJ, Rasch CR. Parotid gland sparing IMRT for head and neck cancer improves xerostomia related quality of life. Radiat Oncol. 2008;3:41. doi: 10.1186/1748-717X-3-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Forastiere AA, Zhang Q, Weber RS, Maor MH, Goepfert H, Pajak TF, et al. Long-term results of RTOG 91-11: a comparison of three nonsurgical treatment strategies to preserve the larynx in patients with locally advanced larynx cancer. J Clin Oncol. 2013;31(7):845–52. doi: 10.1200/JCO.2012.43.6097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.O’Sullivan B, Huang SH, Su J, Garden AS, Sturgis EM, Dahlstrom K, et al. Development and validation of a staging system for HPV-related oropharyngeal cancer by the International Collaboration on Oropharyngeal cancer Network for Staging (ICON-S): a multicentre cohort study. Lancet Oncol. 2016;17(4):440–51. doi: 10.1016/S1470-2045(15)00560-4. [DOI] [PubMed] [Google Scholar]
  • 51.Huang SH, Xu W, Waldron J, Siu L, Shen X, Tong L, et al. Refining American Joint Committee on Cancer/Union for International Cancer Control TNM stage and prognostic groups for human papillomavirus-related oropharyngeal carcinomas. J Clin Oncol. 2015;33(8):836–45. doi: 10.1200/JCO.2014.58.6412. [DOI] [PubMed] [Google Scholar]
  • 52.Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med. 2006;354(6):567–78. doi: 10.1056/NEJMoa053422. [DOI] [PubMed] [Google Scholar]
  • 53.Rosenthal DI, Harari PM, Giralt J, Bell D, Raben D, Liu J, et al. Association of human papillomavirus and p16 status with outcomes in the IMCL-9815 phase III registration trial for patients with locoregionally advanced oropharyngeal squamous cell carcinoma of the head and neck treated with radiotherapy with or without cetuximab. J Clin Oncol. 2016;34(12):1300–8. doi: 10.1200/JCO.2015.62.5970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Geeta SN, Padmanabhan TK, Samuel J, Pavithran K, Iyer S, Kuriakose MA. Comparison of acute toxicities of two chemotherapy schedules for head and neck cancers. J Cancer Res Ther. 2006;2(3):100–4. doi: 10.4103/0973-1482.27584. [DOI] [PubMed] [Google Scholar]
  • 55.Machtay M, Moughan J, Trotti A, Garden AS, Weber RS, Cooper JS, et al. Factors associated with severe late toxicity after concurrent chemoradiation for locally advanced head and neck cancer: an RTOG analysis. J Clin Oncol. 2008;26(21):3582–9. doi: 10.1200/JCO.2007.14.8841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lango MN, Egleston B, Ende K, Feigenberg S, D’Ambrosio DJ, Cohen RB, et al. Impact of neck dissection on long-term feeding tube dependence in patients with head and neck cancer treated with primary radiation or chemoradiation. Head Neck. 2010;32(3):341–7. doi: 10.1002/hed.21188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Marur S, Lee J-W, Cmelak A, Zhao W, Westra WH, Chung CH, et al. ECOG 1308: a phase II trial of induction chemotherapy followed by cetuximab with low dose versus standard dose IMRT in patients with HPV-associated resectable squamous cell carcinoma of the oropharynx (OP). ASCO Annual Meeting Proceedings; 2012; 2012. p. 5566. [Google Scholar]
  • 58.Cmelak A, Li S, Marur S, Zhao W, Westra WH, Chung CH, et al. Symptom reduction from IMRT dose deintensification: results from ECOG 1308 using the Vanderbilt Head and Neck Symptom Survey version 2 (VHNSS V2). ASCO Annual Meeting Proceedings; 2015. p. 6021. [Google Scholar]
  • 59.Duek I, Billan S, Amit M, Gil Z. Transoral Robotic Surgery in the HPV Era. Rambam Maimonides Med J. 2014;5(2):e0010. doi: 10.5041/RMMJ.10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Parsons JT, Mendenhall WM, Stringer SP, Amdur RJ, Hinerman RW, Villaret DB, et al. Squamous cell carcinoma of the oropharynx. Cancer. 2002;94(11):2967–80. doi: 10.1002/cncr.10567. [DOI] [PubMed] [Google Scholar]
  • 61.Haigentz M, Jr, Silver CE, Corry J, Genden EM, Takes RP, Rinaldo A, et al. Current trends in initial management of oropharyngeal cancer: the declining use of open surgery. Eur Arch Otorhinolaryngol. 2009;266(12):1845–55. doi: 10.1007/s00405-009-1109-2. [DOI] [PubMed] [Google Scholar]
  • 62.Chen MM, Roman SA, Kraus DH, Sosa JA, Judson BL. Transoral robotic surgery: a population-level analysis. Otolaryngol Head Neck Surg. 2014;150(6):968–75. doi: 10.1177/0194599814525747. [DOI] [PubMed] [Google Scholar]
  • 63.Al-khudari S, Bendix S, Lindholm J, Simmerman E, Hall F, Ghanem T. Gastrostomy tube use after transoral robotic surgery for oropharyngeal cancer. ISRN Otolaryngology. 2013;2013:5. doi: 10.1155/2013/190364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bernier J, Cooper JS, Pajak TF, van Glabbeke M, Bourhis J, Forastiere A, et al. Defining risk levels in locally advanced head and neck cancers: a comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (# 9501) Head Neck. 2005;27(10):843–50. doi: 10.1002/hed.20279. [DOI] [PubMed] [Google Scholar]
  • 65.Sinha P, Lewis JS, Jr, Piccirillo JF, Kallogjeri D, Haughey BH. Extracapsular spread and adjuvant therapy in human papillomavirus-related, p16-positive oropharyngeal carcinoma. Cancer. 2012;118(14):3519–30. doi: 10.1002/cncr.26671. [DOI] [PubMed] [Google Scholar]
  • 66.Maxwell JH, Ferris RL, Gooding W, Cunningham D, Mehta V, Kim S, et al. Extracapsular spread in head and neck carcinoma: impact of site and human papillomavirus status. Cancer. 2013;119(18):3302–8. doi: 10.1002/cncr.28169. [DOI] [PubMed] [Google Scholar]
  • 67.ECOG3311 PIRToTSRfbL-doS-dIiRpLAOCTPA, editor. Addendum. 5 2015. Disease-specific survival estimates by ECS in HPV-positive and HPV-negative patients. [Google Scholar]
  • 68.Charnley N, Choudhury A, Chesser P, Cooper RA, Sebag-Montefiore D. Effective treatment of anal cancer in the elderly with low-dose chemoradiotherapy. Br J Cancer. 2005;92(7):1221–5. doi: 10.1038/sj.bjc.6602486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bhatia A, Burtness B. Human papillomavirus-associated oropharyngeal cancer: defining risk groups and clinical trials. J Clin Oncol. 2015;33(29):3243–50. doi: 10.1200/JCO.2015.61.2358. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES