Next Article in Journal
RNA Structures and Their Role in Selective Genome Packaging
Next Article in Special Issue
The Not-So-Good, the Bad and the Ugly: HPV E5, E6 and E7 Oncoproteins in the Orchestration of Carcinogenesis
Previous Article in Journal
Quantifying T Cell Cross-Reactivity: Influenza and Coronaviruses
Previous Article in Special Issue
Anti-HPV16 Antibody Titers Prior to an Incident Cervical HPV16/31 Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

De-Escalating Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma

by
Panagiota Economopoulou
,
Ioannis Kotsantis
and
Amanda Psyrri
*
Section of Medical Oncology, Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 1st Rimini St, Haidari, 12462 Athens, Greece
*
Author to whom correspondence should be addressed.
Viruses 2021, 13(9), 1787; https://doi.org/10.3390/v13091787
Submission received: 19 July 2021 / Revised: 2 September 2021 / Accepted: 6 September 2021 / Published: 8 September 2021
(This article belongs to the Special Issue HPV and HPV Vaccine)

Abstract

:
HPV-related head and neck squamous cell carcinoma (HNSCC) has emerged as a diverse clinical and biological disease entity, mainly in young patients with oropharyngeal tumors who are nonsmokers and nondrinkers. Indeed, during the past few years, the pendulum has shifted towards a new epidemiological reality, the “HPV pandemic”, where the majority of oropharyngeal squamous cell carcinomas (OPSCCs) are attributed to HPV. The oncogenic potential of the virus is associated to its capacity of integrating oncogenes E6 and E7 into the host cell, leading to the inactivation of several tumor suppressor genes, such as Rb. HPV status can affect prognosis in OPSCC, but its role as a predictive biomarker remains to be elucidated. Given the favorable prognosis associated with HPV-positive disease, the concept of de-escalation treatment strategies has been developed with the primary intent being the reduction of treatment-related long-term toxicities. In this review, we aim to depict current data regarding treatment de-escalation in HPV-associated OPSCC and discuss ongoing clinical trials.

1. Introduction

Recent epidemiological evidence suggests a substantial rise of Human Papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma (OPSCC), which accounts for more than 60% of OPSCC cases in the contemporary area [1,2,3]. Compared to tobacco-related disease, HPV-positive (HPV+) OPSCC is characterized by a distinct molecular landscape, including lower frequency of mutations in the TP53, EGFR and CDNK1A genes [4] and different demographic features, such as nonsignificant tobacco and alcohol consumption in a subset of cases and younger age at disease presentation, although an increased prevalence in older patients has been recently noted [5,6]. The combination of an early-stage primary tumor and bulky regional lymph nodes is a typical clinical presentation [7].
Most importantly, HPV+ OPSCC has been traditionally associated with significantly better outcomes [8,9,10,11]. In a landmark retrospective analysis of the Radiation Therapy Oncology Group (RTOG) 0129 trial in which patients with locally advanced head and neck squamous cell carcinoma (HNSCC) were randomized to radiotherapy (RT) with either accelerated fractionation with concomitant boost or standard fractionation with concurrent cisplatin, Ang et al. correlated HPV status with clinical outcome based on assessment of HPV DNA using in situ hybridization (ISH) [12]. It was postulated that patients with HPV+ OPSCC (63.8% of OPSCC cases who participated in the study) had a 58% decrease in the risk of death after adjusting for clinical and demographic features. Furthermore, Ang et al. suggested a patient risk stratification based on smoking status and TNM stage that defined patients with low- and intermediate-risk HPV+ OPSCC. Based on that classification, patients with intermediate-risk HPV+ disease are defined by a more than 10 pack/year smoking history and N2b/N3 or T4 disease by 7th edition staging; patients with HPV-OPSCC who are nonsmokers or have T1–T3 N0–N2 disease by TNM 7th edition belong to the low-risk group [12].
Given the remarkably better survival rates observed in HPV+ OPSCC, juxtaposed with the substantial morbidity caused by the standard of care treatment of cisplatin and RT combination in locally advanced disease, the concept of treatment de-escalation has been proposed as a new treatment paradigm. In this review, we seek to summarize current data on deintensification approaches in HPV+ OPSCC and discuss the goals of future de-escalation studies in this regard.

2. History and Rationale for Treatment De-Escalation-Selection of Patients

The retrospective analysis of the RTOG 0129 trial published by Ang et al. identified a low-risk group of HPV+ OPSCC patients who exhibit high survival rates (90–95% at 3 years), implying a high chance of cure [12]. These results denote that a steadily increasing group of patients will survive several decades, albeit with significant treatment sequelae and functional abnormalities related to cisplatin-based chemoradiotherapy (CRT). Indeed, a pooled analysis of RTOG trials that used conventional RT techniques showed that 43% of treated patients experienced a severe late toxicity [13]. Although the incorporation of intensity modulated radiation therapy (IMRT) in the treatment algorithm of HNSCC has reduced the burden of treatment-related xerostomia and weight loss [14], late toxicities of CRT including deficits in speech, shallowing and mobility, poor oral health, trismus, and dysphagia greatly influence functional capacity of the head and neck area and severely diminish quality of life in long-term survivors [8,15]. In this context and given the young age and nonsignificant comorbidity of patients with HPV+ OPSCC, a framework of deintensification strategies has been proposed and assessed in randomized clinical trials with the goal of decreasing treatment-associated sequalae, without compromising tumor control.
The choice of suitable candidates for treatment de-escalation trials has become a matter of paramount importance. Two groups of patients should probably be excluded from participation. The first group (a) consists of patients with a smoking history (more than 10 pack years) due to a substantially worse prognosis, with a 3-year OS of approximately 70% [16]. Of note, compelling evidence from RTOG 9003 and RTOG 0129 studies have demonstrated that each year of smoking increased risk of death by 2% between p16-positive patients [17]. The second group (b) consists of patients who display adverse pathological features following surgery, such as extracapsular extension and positive margins. Several trials have used risk classification by Ang as inclusion criteria [12]. Despite being prospectively validated by two other cohorts [18,19], this classification system fails to incorporate relevant prognostic factors, such as biological features, HPV type, age, comorbidities, and response to treatment.
Importantly, the recognition that the American Joint Committee on Cancer (AJCC) 7th edition staging system does not adequately reflect the prognosis of HPV+ OPSCC, given the paradoxically favorable survival associated with the advanced stage, led to the development of the modified 8th edition clinical stage paradigm for HPV+ disease that has been more recently adapted [20]. However, despite improved prognostication conferred by the 8th edition AJCC staging [21], it cannot be utilized for guidance regarding patient management until clinical trials have validated alternative treatments based on 8th edition staging. Thus, since treatment decisions are still based on 7th edition staging, the new staging system mainly provides information for patient prognosis.
Alternative methods for risk stratification that are currently under investigation include the incorporation of genomic signatures [22], expression of molecular markers [23], and radiologic biomarkers for evaluation of response to treatment [24]. In addition, better selection of patients included in clinical trials, with a more stringent definition of an HPV positive status, including the analysis of presence of HPV DNA/or HPV RNA together with p16+ status and limiting studies to patients with OPSCC only in the tonsillar or base of tongue sites, may result in more accurate information on treatment de-escalation [25,26].

3. De-Escalation Strategies

Although there are many ongoing de-escalation strategies, the most commonly assessed in clinical trials are reduction of RT dose, field, or schedule when given either as adjuvant or definitive therapy and omission, dose reduction, or replacement of concurrent chemotherapy. Some of these clinical trials are presented below and in Table 1.

3.1. Reducing Radiotherapy Dose/Schedule

The cumulative dose of delivered RT has been substantially correlated with long-term functional deficits, which are lessened with doses below 60 Gy [27]. The rationale for reducing the dose of RT in HPV+ tumors is based on accumulated preclinical data [28] and reported outcomes from clinical trials in HPV+ patients (Lassen, JCO 2009) that indicate a higher radiosensitivity of HPV+ compared to HPV- disease. Thus, RT at a reduced dose might save patients with HPV+ OPSCC from long-term sequelae of RT without compromising efficacy.
Definitive chemoradiotherapy. In the setting of definitive chemoradiation, the phase II NRG-HN002 (NCT02254278) trial evaluated the efficacy of a reduced-dose RT (60 Gy) in patients with low-risk disease based on stratification by Ang’s criteria, namely patients with 7th edition stage T1-2N1-N2bM0 or T3N0-N2bM0 OPSCC with a ≤10 pack-year history of tobacco consumption [29]. Patients were randomized to either IMRT over 6 weeks concurrently with weekly cisplatin 40 mg/m2 or to IMRT alone over 5 weeks. This study showed that despite the satisfactory efficacy of concurrent CRT arm (2-year PFS of 90.5%), the RT alone arm did not meet acceptability criteria (2-year PFS 87.6%). Of note, 2-year OS was similar between the two groups.
In the same context, Chera et al. reported the results of a phase II trial that evaluated the combination of reduced dose IMRT (60 Gy) with cisplatin at a lower dose (30 mg/m2 weekly) [30]. The primary study endpoint was the pathologic complete response rate based on biopsy of the primary site and dissection of pretreatment positive lymph node regions. Eligibility criteria included patients with stage T0-3N0-N2c (7th edition AJCC) HPV+ OSCC with ≤10 pack-years smoking history. Past smokers were also permitted if they had ceased more than 5 years ago. The study met its primary endpoint of complete pathological response rate (86%, similar to standard CRT), which was estimated based on biopsy at primary site and dissection of positive lymph nodes before the initiation of treatment. In addition, de-escalated CRT resulted in a 3-year tumor control and survival of 100% while providing an improved quality of life, inter alia a significant reduction in feeding tube placement. In an almost identical single arm study conducted by the same group of authors, where a PET-CT scan was used to evaluate the need for LN dissection, 2-year PFS and OS were 86% and 95%, respectively [31].
In a similar context, EVADER is a single arm phase II study that seeks to evaluate the effectiveness of reduced RT volume to selected lymph node regions in patients with early stage (T1-3 N0-1M0 AJCC 8th edition) HPV+ OPSCC [32]. On the other hand, LCCC 1612 is an ongoing clinical trial that is using smoking status and molecular-based stratification as selection tools for treatment de-escalation. More specifically, the efficacy of reduced dose CRT (60 Gy) is being evaluated in patients with HPV+ OPSCC and (a) a history of light smoking (<10 pack years) and (b) with a history of >10 pack years smoking and no p53 mutations. On the contrary, patients with a history of > 10 pack years and p53 mutations will receive a standard dose of 70 Gy cisplatin-based CRT (NCT03077243).
Deintensification of definitive RT after induction chemotherapy. Response to induction chemotherapy (IC) has been used as a biomarker for selection of appropriate candidates for deintensified RT in phase II trials. All trials demonstrate improved outcomes compared to standard controls, warranting the design of phase 3 trials.
The Quarterback trial was a phase II study that enrolled patients with HPV+ disease and a <20 pack year smoking history who received induction TPF. Following IC, clinical responders (20 out of 23 patients) were randomized to receive a standard dose (70 Gy) or reduced dose (54 Gy) RT with weekly carboplatin [33]. In a median follow-up of 56 months, PFS and OS were similar between the two groups. In the ongoing Quarterback 2b trial, a reduced dose RT schedule (56 Gy) is being evaluated after response to IC (NCT02945631).
E1308 was a larger phase II trial with 90 participants, all stage III-IV (AJCC 7th edition) HPV+ OPSCC patients who were assessed for response at primary site and lymph nodes after IC with cisplatin, paclitaxel, and cetuximab. Patients with complete response (CR) were treated with RT at a reduced dose of 54 Gy, whereas patients with less than CR received standard dose RT of 69.3 Gy; both RT schedules were given in combination with weekly cetuximab [34]. Importantly, this study showed promising results, with a 2-year 80% PFS and 94% OS in IC responders treated with reduced dose RT. Importantly, functional outcomes, such as nutrition and shallowing capacity, were substantially improved at patients receiving low dose RT.
On the other hand, the OPTIMA phase II trial included patients with low risk (defined as ≤T3, ≤N2b, ≤10 pack-year smoking history) and high risk (defined as T4 or ≥N2c or ≥10 pack-year smoking history) HPV+ OPSCC, who, depending on their response at IC with a carboplatin/nab-paclitaxel combination regimen, were treated with reduced dose RT (low-risk with good response) or CRT (low-risk with medium response or high-risk with good response) or standard dose CRT (patients with any other response) [35]. Two-year PFS and OS were favorable for both low (95% and 100% respectively) and high-risk patients (94% and 97% respectively) with decreased rate of mucositis and PEG-tube use. Interestingly, OPTIMA II (NCT03107182) was a phase II trial which evaluated the same IC in combination with nivolumab; subsequently, patients were randomized to three treatment arms based on risk stratification and response to IC [36]. Patients were included in the high risk group if they had any of the following: T4, N2c-N3 (AJCC 7th edition), >20 pack-year smoking history, and nonHPV16 subtype; the remaining were stratified as low risk. Low risk patients with ≥ 50% reduction in tumor size were treated with transoral robotic surgery (TORS) or reduced dose RT (50 Gy) (Arm A). High risk patients with ≥50% reduction in tumor size and low risk patients with <50% reduction in tumor size were treated with CRT 45–50 Gy (Arm B), and the remaining patients were treated with standard CRT 70–75 Gy (Arm C). Adjuvant nivolumab was also administered for 6 months. Preliminary results were presented at American Society for Clinical Oncology (ASCO) 2021. The primary endpoint of deep response rate (DDR) after nivolumab-IC combination regimen was 70.8%, and 2-year PFS and OS were excellent (PFS: 96.3%, 85.8%, and 100% for arms A, B, and C, respectively, and OS: 96%, 91.9%, and 100% for arms A, B, and C, respectively). In addition, induction immunochemotherapy yielded a high pCR rate for patients who underwent TORS (66.7%) [36].
Finally, the RAVD trial similarly sought to assess the efficacy of a de-escalated approach that incorporated a reduced RT volume areas based on response to IC in patients with locally advanced HNSCC [37]. Consequently, patients with ≥50 % CR based on RECIST criteria received combined chemotherapy with reduced RT planning target volume (targeting gross disease), whereas patients with lesser response were treated with combined CRT at a planning target volume including elective nodal basins. For patients with HPV+ OPSCC included in the analysis, 2-year PFS and OS were 93.1%/92.1%, respectively, for good responders and 74.0%/95.2%, respectively, for nonresponders; PFS did not differ significantly between the two groups.
Adjuvant therapy. Adjuvant CRT is currently indicated in patients whose tumor pathology includes positive surgical margins or extracapsular lymph node extension. On the other hand, adjuvant RT is considered in tumors with positive lymph nodes and deep invasion (for primary site of the oral cavity).
The ECOG-ACRIN 3311 trial included patients with resectable p16+ OPSCC and cT1-T2 stage III/IV 7th edition AJCC without matted nodes. After transoral resection (TORS), patients were stratified depending on their pathological features to low risk (clear margins, 0–1 positive lymph nodes (LNs), no extracapsular extension (ENE)) who had no additional treatment (Arm A), intermediate risk (clear/close margins, 2–4 positive LNs, ENE ≤ 1 mm) who were randomly assigned to either postoperative reduced dose RT 50 Gy (Arm B) or RT 60 Gy (Arm C), and high risk (positive margins, >5 positive LNs, ENE >1 mm) who were managed with adjuvant cisplatin-based CRT (Arm D) [38]. For intermediate-risk patients, reduced dose RT demonstrated promising results, with a 3-year PFS of 94.9%, which met the primary endpoint of the study. For low-risk patients, 3-year PFS was 96.9% without additional RT. Importantly, 56% of patients in arms B and C vs. 36% in arm D reported stable or improved functional outcomes (p = 0.011). Based on this study, TORS plus adjuvant reduced dose RT should be evaluated in a phase III trial in comparison to standard radical CRT in patients with HPV+ OPSCC.
MC1273 was a phase II trial that sought to assess the efficacy of a remarkably reduced RT dose administered as adjuvant treatment following surgical management of stage III-IV (AJCC 7th edition) HPV+ OPSCC [39]. Eligible patients were non/light smokers (≤10 pack years) and were grouped as intermediate or high risk based on pathology (intermediate-risk: ≥T3, ≥2 positive LNs, lymphovascular/perineural invasion, LN > 3 cm; high risk: ENE, all patients had negative surgical margins). Patients in the intermediate-risk group were treated with 30 Gy RT delivered twice a day for 2 weeks in combination with weekly docetaxel, whereas patients in the high-risk group received an additional boost to LNs with ENE. The two-year locoregional control rate was 96.2% and the 2-year PFS was 91.1% in the total population; these outcomes were comparable to historical controls using standard treatment.
On the other hand, the AVOID trial, a phase II trial with distinct design, focused on patients with LN involvement after surgical treatment with TORS [40]. Sixty patients with stage pT1-T2 N1-N3 HPV+ OPSCC without other pathological risk factors (lymphovascular/perineural invasion, involved margins) were included. Adjuvant treatment following TORS consisted of deintensified RT to areas at risk in the involved neck (60–64 Gy) and uninvolved neck (54 Gy), sparing the resected primary site. Results were promising, with a two-year local recurrence-free survival of 97.9% and OS of 100%. Toxicity was favorable, with no patient requiring a feeding tube during RT; however, two patients required a temporal feeding tube during follow up.
PATHOS is an ongoing trial which transitioned into phase III after successful completion of the phase II cohort. Patients with stage T1–T3 N0–N2b HPV+ OPSCC are stratified to pathological subgroups following TORS and LN dissection. Patients in the low-risk group receive no further treatment, patients in the intermediate-risk group are randomized to standard or reduced dose RT, and patients in the high-risk group are randomized to CRT or RT [41,42]. The primary endpoint is noninferiority for OS with coprimary endpoint shallowing function at 12 months.

3.2. Chemotherapy Replacing

Given the significant toxicity correlated with cisplatin-based CRT and the promising efficacy of cetuximab-based bioradiotherapy in patients with OPSCC demonstrated in the IMCL 9815 trial [43], the idea of substituting cisplatin with cetuximab in patients with HPV+ disease has emerged as a tempting approach. Two randomized phase III trials, which constitute the only completed phase III trials in HPV+ OPSCC, have failed to corroborate noninferiority of cetuximab as compared to cisplatin in combination with RT. In the De-ESCALaTE HPV trial, 334 patients with stage T3N0–T4N0, T1N1–T4N3 (AJCC 7th edition) HPV+ OPSCC and limited smoking history (low risk by Ang stratification) [12] were randomly allocated to either standard high dose cisplatin-based CRT or RT in combination with weekly cetuximab [44]. The primary endpoint was rate of severe adverse events at 24 months of treatment completion. Interestingly, not only was there not any significant difference in severe toxicity between treatment arms, but the experimental arm was also associated with less favorable OS and (97.5% vs. 89.4%, p = 0.001) and 2-year relapse rate (6% vs. 16.1%, p = 0.0007).
RTOG 1016 was a noninferiority phase III trial published in same year as De-ESCALAaTE-HPV, which randomized patients with HPV+ OPSCC to either two cycles of cisplatin in combination with RT or cetuximab plus RT [45]. It was a larger trial that included 849 patients as compared to 334 in De-ESCALaTE-HPV and was not focused on low-risk patients. Indeed, 29% of patients were intermediate-risk based on stratification by Ang [12], and 16% had advanced T or N stage. Interestingly, the cetuximab/RT arm did not meet noninferiority criteria for OS, which was the primary endpoint of the study, and the cisplatin/RT arm yielded superior OS (5-year OS 84.6% vs. 77.9% in the cetuximab arm), with no differences in toxicity. Follow-up was 5 years compared to 26 months in De-ESCALaTE-HPV.
In addition to these two trials, TROG 12.0, a phase III trial conducted in Australia was recently reported at ASCO 2021 [46]. In this trial, 189 patients with AJCC 7th edition stage III (excluding T3N1) and stage IV (except from distant metastases and T4 +/− N3 +/− N2b-c if >10 pack years smoking history) disease were randomized to either cetuximab/RT combination or weekly cisplatin 40 mg/m2 and RT; primary endpoint was symptom severity [46]. Similar to the aforementioned trials, symptom severity did not differ between the two treatment groups, and 3-year failure free survival was worse with cetuximab (80% in the cetuximab-arm vs. 93% in the cisplatin arm, p = 0.015).

4. Conclusions and Future Directions

As we discuss in detail a variety of heterogeneous deintensification trials in HPV-related OPSCC, we understand that it is currently not the proper time for a broad application of de-escalation therapy in HPV+ patients outside of clinical trials. However, any patient with low-risk disease and no/limited history of smoking should be considered a candidate for participation in ongoing trials. A very tempting approach is the incorporation of immunotherapy in de-escalation strategies, since HPV+ OPSCC has an immunogenic profile and has been found to produce high response rates with immunotherapy [47]. Two trials investigate the substitution of cisplatin with antiPDL1 antibody durvalumab combined with RT in locally advanced disease (NCT03410615, NCT03623646), whereas a phase II study is assessing the safety and efficacy of neoadjuvant ipilimumab followed by nivolumab and reduced dose RT (NCT03799445).
Nevertheless, in addition to including a more stringent definition of HPV positive status and accurate information of the OSCC subsite, the identification and evaluation of novel biomarkers is a most important step in de-escalation trials and patient selection. The diagnosis of an HPV+ tumor can be improved in future clinical trials by adding the analysis of HPV DNA or HPV RNA to p16 immunohistochemistry, since around 10–15% of all p16+ oropharyngeal cancer is not HPV DNA+ [25]. Clinical biomarkers, such as disease stage and smoking history, and dynamic biomarkers such as response to induction chemotherapy, have been already widely used in reported trials. Molecular markers could potentially replace clinical biomarkers such as tobacco consumption, which is not easily quantifiable, and quantification is subjected to personal bias. Additionally, it is likely that poor prognosis associated with smoking history is due to genetic changes caused by tobacco and not tobacco itself. Genomic biomarkers that have been shown to correlate with dismal prognosis, such as PI3KCA mutation, could be used to exclude patients from those trials [48]. Imaging biomarkers such as MRI and 18F-FMISO PET are currently under investigation in ongoing trials (NCT03323463, NCT03224000). Most importantly, dynamic molecular biomarkers such as plasma or saliva ctDNA that have been shown to predict tumor recurrence in prospective studies [49,50] must be investigated in clinical trial protocols. Collecting serum and tissue samples to identify the characteristics of responders and favorable prognosis patients is fundamental in the design of de-escalation trials in HPV+ OPSCC and may produce robust algorithms to improve survival and guide patient management. In this regard, the REACT study is using circulating tumor DNA as a biomarker for treatment deintensification in low-risk HPV+ OPSCC; patients with high ctDNA levels at week 4 of treatment will be treated with standard dose RT or CRT, whereas patients with low ctDNA levels will be treated with reduced dose and schedule RT or CRT; results of this trial are eagerly awaited.
Ongoing de-escalation clinical trials in HPV+ OPSCC are shown in Table 1.

Author Contributions

Conceptualization: P.E. and A.P.; Writing, original draft preparation: P.E.; Data collection: I.K.; Writing, review and editing: all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Hellenic Society of Medical Oncology.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gillison, M.L.; Chaturvedi, A.K.; Anderson, W.F.; Fakhry, C. Epidemiology of Human Papillomavirus-Positive Head and Neck Squamous Cell Carcinoma. J. Clin. Oncol. 2015, 33, 3235–3242. [Google Scholar] [CrossRef] [Green Version]
  2. Economopoulou, P.; Agelaki, S.; Perisanidis, C.; Giotakis, E.I.; Psyrri, A. The promise of immunotherapy in head and neck squamous cell carcinoma. Ann. Oncol. 2016, 27, 1675–1685. [Google Scholar] [CrossRef]
  3. Economopoulou, P.; Kotsantis, I.; Psyrri, A. Special Issue about Head and Neck Cancers: HPV Positive Cancers. Int. J. Mol. Sci. 2020, 21, 3388. [Google Scholar] [CrossRef]
  4. Stransky, N.; Egloff, A.M.; Tward, A.D.; Kostic, A.D.; Cibulskis, K.; Sivachenko, A.; Kryukov, G.V.; Lawrence, M.S.; Sougnez, C.; McKenna, A.; et al. The mutational landscape of head and neck squamous cell carcinoma. Science 2011, 333, 1157–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Strohl, M.P.; Wai, K.C.; Ha, P.K. De-intensification strategies in HPV-related oropharyngeal squamous cell carcinoma-a narrative review. Ann. Transl. Med. 2020, 8, 1601. [Google Scholar] [CrossRef] [PubMed]
  6. Windon, M.J.; D’Souza, G.; Rettig, E.M.; Westra, W.H.; van Zante, A.; Wang, S.J.; Ryan, W.R.; Mydlarz, W.K.; Ha, P.K.; Miles, B.A.; et al. Increasing prevalence of human papillomavirus-positive oropharyngeal cancers among older adults. Cancer 2018, 124, 2993–2999. [Google Scholar] [CrossRef] [Green Version]
  7. Gillison, M.L.; D’Souza, G.; Westra, W.; Sugar, E.; Xiao, W.; Begum, S.; Viscidi, R. 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, 407–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Rosenberg, A.J.; Vokes, E.E. Optimizing Treatment De-Escalation in Head and Neck Cancer: Current and Future Perspectives. Oncologist 2021, 26, 40–48. [Google Scholar] [CrossRef]
  9. Rischin, D. Oropharyngeal cancer, human papilloma virus, and clinical trials. J. Clin. Oncol. 2010, 28, 1–3. [Google Scholar] [CrossRef]
  10. Posner, M.R.; Lorch, J.H.; Goloubeva, O.; Tan, M.; Schumaker, L.M.; Sarlis, N.J.; Haddad, R.I.; Cullen, K.J. Survival and human papillomavirus in oropharynx cancer in TAX 324: A subset analysis from an international phase III trial. Ann. Oncol. 2011, 22, 1071–1077. [Google Scholar] [CrossRef]
  11. Nguyen-Tan, P.F.; Zhang, Q.; Ang, K.K.; Weber, R.S.; Rosenthal, D.I.; Soulieres, D.; Kim, H.; Silverman, C.; Raben, A.; Galloway, T.J.; 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, 32, 3858–3866. [Google Scholar] [CrossRef]
  12. Ang, K.K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tan, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef] [Green Version]
  13. Machtay, M.; Moughan, J.; Trotti, A.; Garden, A.S.; Weber, R.S.; Cooper, J.S.; Forastiere, A.; Ang, K.K. Factors associated with severe late toxicity after concurrent chemoradiation for locally advanced head and neck cancer: An RTOG analysis. J. Clin. Oncol. 2008, 26, 3582–3589. [Google Scholar] [CrossRef]
  14. van der Veen, J.; Nuyts, S. Can Intensity-Modulated-Radiotherapy Reduce Toxicity in Head and Neck Squamous Cell Carcinoma? Cancers 2017, 9, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chen, A.M.; Daly, M.E.; Farwell, D.G.; Vazquez, E.; Courquin, J.; Lau, D.H.; Purdy, J.A. Quality of life among long-term survivors of head and neck cancer treated by intensity-modulated radiotherapy. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 129–133. [Google Scholar] [CrossRef] [Green Version]
  16. O’Sullivan, B.; Huang, S.H.; Siu, L.L.; Waldron, J.; Zhao, H.; Perez-Ordonez, B.; Weinreb, I.; Kim, J.; Ringash, J.; Bayley, A.; et al. Deintensification candidate subgroups in human papillomavirus-related oropharyngeal cancer according to minimal risk of distant metastasis. J. Clin. Oncol. 2013, 31, 543–550. [Google Scholar] [CrossRef] [PubMed]
  17. Gillison, M.L.; Zhang, Q.; Jordan, R.; Xiao, W.; Westra, W.H.; Trotti, A.; Spencer, S.; Harris, J.; Chung, C.H.; Ang, K.K. Tobacco smoking and increased risk of death and progression for patients with p16-positive and p16-negative oropharyngeal cancer. J. Clin. Oncol. 2012, 30, 2102–2111. [Google Scholar] [CrossRef]
  18. Granata, R.; Miceli, R.; Orlandi, E.; Perrone, F.; Cortelazzi, B.; Franceschini, M.; Locati, L.D.; Bossi, P.; Bergamini, C.; Mirabile, A.; et al. Tumor stage, human papillomavirus and smoking status affect the survival of patients with oropharyngeal cancer: An Italian validation study. Ann. Oncol. 2012, 23, 1832–1837. [Google Scholar] [CrossRef]
  19. Fakhry, C.; Zhang, Q.; Gillison, M.L.; Nguyen-Tan, P.F.; Rosenthal, D.I.; Weber, R.S.; Lambert, L.; Trotti, A.M., 3rd; Barrett, W.L.; Thorstad, W.L.; et al. Validation of NRG oncology/RTOG-0129 risk groups for HPV-positive and HPV-negative oropharyngeal squamous cell cancer: Implications for risk-based therapeutic intensity trials. Cancer 2019, 125, 2027–2038. [Google Scholar] [CrossRef] [PubMed]
  20. Huang, S.H.; Xu, W.; Waldron, J.; Siu, L.; Shen, X.; Tong, L.; Ringash, J.; Bayley, A.; Kim, J.; Hope, A.; 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, 836–845. [Google Scholar] [CrossRef] [PubMed]
  21. Bigelow, E.O.; Seiwert, T.Y.; Fakhry, C. Deintensification of treatment for human papillomavirus-related oropharyngeal cancer: Current state and future directions. Oral Oncol. 2020, 105, 104652. [Google Scholar] [CrossRef]
  22. Gleber-Netto, F.O.; Rao, X.; Guo, T.; Xi, Y.; Gao, M.; Shen, L.; Erikson, K.; Kalu, N.N.; Ren, S.; Xu, G.; et al. Variations in HPV function are associated with survival in squamous cell carcinoma. JCI Insight 2019, 4, e124762. [Google Scholar] [CrossRef] [Green Version]
  23. Ramesh, P.S.; Devegowda, D.; Singh, A.; Thimmulappa, R.K. NRF2, p53, and p16: Predictive biomarkers to stratify human papillomavirus associated head and neck cancer patients for de-escalation of cancer therapy. Crit. Rev. Oncol. Hematol. 2020, 148, 102885. [Google Scholar] [CrossRef]
  24. Kwan, J.Y.Y.; Su, J.; Huang, S.H.; Ghoraie, L.S.; Xu, W.; Chan, B.; Yip, K.W.; Giuliani, M.; Bayley, A.; Kim, J.; et al. Radiomic Biomarkers to Refine Risk Models for Distant Metastasis in HPV-related Oropharyngeal Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1107–1116. [Google Scholar] [CrossRef]
  25. Smeets, S.J.; Hesselink, A.T.; Speel, E.J.; Haesevoets, A.; Snijders, P.J.; Pawlita, M.; Meijer, C.J.; Braakhuis, B.J.; Leemans, C.R.; Brakenhoff, R.H. A novel algorithm for reliable detection of human papillomavirus in paraffin embedded head and neck cancer specimen. Int. J. Cancer 2007, 121, 2465–2472. [Google Scholar] [CrossRef] [PubMed]
  26. Marklund, L.; Holzhauser, S.; de Flon, C.; Zupancic, M.; Landin, D.; Kolev, A.; Haeggblom, L.; Munck-Wikland, E.; Hammarstedt-Nordenvall, L.; Dalianis, T.; et al. Survival of patients with oropharyngeal squamous cell carcinomas (OPSCC) in relation to TNM 8-Risk of incorrect downstaging of HPV-mediated non-tonsillar, non-base of tongue carcinomas. Eur. J. Cancer 2020, 139, 192–200. [Google Scholar] [CrossRef] [PubMed]
  27. Mortensen, H.R.; 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, 288–294. [Google Scholar] [CrossRef] [PubMed]
  28. Kimple, R.J.; Smith, M.A.; Blitzer, G.C.; Torres, A.D.; Martin, J.A.; Yang, R.Z.; Peet, C.R.; Lorenz, L.D.; Nickel, K.P.; Klingelhutz, A.J.; et al. Enhanced radiation sensitivity in HPV-positive head and neck cancer. Cancer Res. 2013, 73, 4791–4800. [Google Scholar] [CrossRef] [Green Version]
  29. Yom, S.S.; Torres-Saavedra, P.; Caudell, J.J.; Waldron, J.N.; Gillison, M.L.; Xia, P.; Truong, M.T.; Kong, C.; Jordan, R.; Subramaniam, R.M.; et al. Reduced-Dose Radiation Therapy for HPV-Associated Oropharyngeal Carcinoma (NRG Oncology HN002). J. Clin. Oncol. 2021, 39, 956–965. [Google Scholar] [CrossRef]
  30. Chera, B.S.; Amdur, R.J.; Tepper, J.E.; Tan, X.; Weiss, J.; Grilley-Olson, J.E.; Hayes, D.N.; Zanation, A.; Hackman, T.G.; Patel, S.; et al. Mature results of a prospective study of deintensified chemoradiotherapy for low-risk human papillomavirus-associated oropharyngeal squamous cell carcinoma. Cancer 2018, 124, 2347–2354. [Google Scholar] [CrossRef] [Green Version]
  31. Chera, B.S.; Amdur, R.J.; Green, R.; Shen, C.; Gupta, G.; Tan, X.; Knowles, M.; Fried, D.; Hayes, N.; Weiss, J.; et al. Phase II Trial of De-Intensified Chemoradiotherapy for Human Papillomavirus-Associated Oropharyngeal Squamous Cell Carcinoma. J. Clin. Oncol. 2019, 37, 2661–2669. [Google Scholar] [CrossRef] [PubMed]
  32. Bratman, S.V.; Berthelet, E.; Butler, J.B.; de Almeida, J.R.; Karam, I.; Metser, U.; Olson, R.A.; Pochini, C.; Waldron, J.; Yu, E.; et al. CCTG HN.10: A phase II single-arm trial of elective volume adjusted de-escalation radiotherapy (EVADER) in patients with low-risk HPV-related oropharyngeal squamous cell carcinoma (NCT03822897). J. Clin. Oncol. 2020, 38 (Suppl. 15), TPS6592. [Google Scholar] [CrossRef]
  33. Misiukiewicz, K.; Gupta, V.; Miles, B.A.; Bakst, R.; Genden, E.; Selkridge, I.; Surgeon, J.T.; Rainey, H.; Camille, N.; Roy, E.; et al. Standard of care vs reduced-dose chemoradiation after induction chemotherapy in HPV+ oropharyngeal carcinoma patients: The Quarterback trial. Oral Oncol. 2019, 95, 170–177. [Google Scholar] [CrossRef]
  34. Marur, S.; Li, S.; Cmelak, A.J.; Gillison, M.L.; Zhao, W.J.; Ferris, R.L.; Westra, W.H.; Gilbert, J.; Bauman, J.E.; Wagner, L.I.; et al. E1308: Phase II Trial of Induction Chemotherapy Followed by Reduced-Dose Radiation and Weekly Cetuximab in Patients With HPV-Associated Resectable Squamous Cell Carcinoma of the Oropharynx- ECOG-ACRIN Cancer Research Group. J. Clin. Oncol. 2017, 35, 490–497. [Google Scholar] [CrossRef] [PubMed]
  35. Seiwert, T.Y.; Foster, C.C.; Blair, E.A.; Karrison, T.G.; Agrawal, N.; Melotek, J.M.; Portugal, L.; Brisson, R.J.; Dekker, A.; Kochanny, S.; et al. OPTIMA: A phase II dose and volume de-escalation trial for human papillomavirus-positive oropharyngeal cancer. Ann. Oncol. 2019, 30, 297–302. [Google Scholar] [CrossRef] [Green Version]
  36. Rosenberg, A.; Agrawal, N.; Pearson, A.T.; Seiwert, T.Y.; Gooi, Z.; Blair, E.A.; Ginat, D.; Howard, A.; Chin, J.; Kochanny, S.; et al. Nivolumab, nabpaclitaxel, and carboplatin followed by risk/response adaptive de-escalated locoregional therapy for HPV-associated oropharyngeal cancer: OPTIMA II trial. J. Clin. Oncol. 2021, 39 (Suppl. 15), 6011. [Google Scholar] [CrossRef]
  37. Villaflor, V.M.; Melotek, J.M.; Karrison, T.G.; Brisson, R.J.; Blair, E.A.; Portugal, L.; De Souza, J.A.; Ginat, D.T.; Stenson, K.M.; Langerman, A.; et al. Response-adapted volume de-escalation (RAVD) in locally advanced head and neck cancer. Ann. Oncol. 2016, 27, 908–913. [Google Scholar] [CrossRef]
  38. Ferris, R.L.; Flamand, Y.; Weinstein, G.S.; Li, S.; Quon, H.; Mehra, R.; Garcia, J.J.; Ringash, J.; Lewin, J.S.; Duvvuri, U.; et al. Updated report of a phase II randomized trial of transoral surgical resection followed by low-dose or standard postoperative therapy in resectable p16+ locally advanced oropharynx cancer: A trial of the ECOG-ACRIN cancer research group (E3311). J. Clin. Oncol. 2021, 39 (Suppl. 15), 6010. [Google Scholar] [CrossRef]
  39. Ma, D.J.; Price, K.A.; Moore, E.J.; Patel, S.H.; Hinni, M.L.; Garcia, J.J.; Graner, D.E.; Foster, N.R.; Ginos, B.; Neben-Wittich, M.; et al. Phase II Evaluation of Aggressive Dose De-Escalation for Adjuvant Chemoradiotherapy in Human Papillomavirus–Associated Oropharynx Squamous Cell Carcinoma. J. Clin. Oncol. 2019, 37, 1909–1918. [Google Scholar] [CrossRef]
  40. Swisher-McClure, S.; Lukens, J.N.; Aggarwal, C.; Ahn, P.; Basu, D.; Bauml, J.M.; Brody, R.; Chalian, A.; Cohen, R.B.; Fotouhi-Ghiam, A.; et al. A Phase 2 Trial of Alternative Volumes of Oropharyngeal Irradiation for De-intensification (AVOID): Omission of the Resected Primary Tumor Bed After Transoral Robotic Surgery for Human Papilloma Virus-Related Squamous Cell Carcinoma of the Oropharynx. Int. J. Radiat. Oncol. Biol. Phys. 2020, 106, 725–732. [Google Scholar] [CrossRef]
  41. Owadally, W.; Hurt, C.; Timmins, H.; Parsons, E.; Townsend, S.; Patterson, J.; Hutcheson, K.; Powell, N.; Beasley, M.; Palaniappan, N.; et al. PATHOS: A phase II/III trial of risk-stratified, reduced intensity adjuvant treatment in patients undergoing transoral surgery for Human papillomavirus (HPV) positive oropharyngeal cancer. BMC Cancer 2015, 15, 602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hargreaves, S.; Beasley, M.; Hurt, C.; Jones, T.M.; Evans, M. Deintensification of Adjuvant Treatment After Transoral Surgery in Patients With Human Papillomavirus-Positive Oropharyngeal Cancer: The Conception of the PATHOS Study and Its Development. Front. Oncol. 2019, 9, 936. [Google Scholar] [CrossRef] [Green Version]
  43. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mehanna, H.; Robinson, M.; Hartley, A.; Kong, A.; Foran, B.; Fulton-Lieuw, T.; Dalby, M.; Mistry, P.; Sen, M.; O’Toole, L.; et al. Radiotherapy plus cisplatin or cetuximab in low-risk human papillomavirus-positive oropharyngeal cancer (De-ESCALaTE HPV): An open-label randomised controlled phase 3 trial. Lancet 2019, 393, 51–60. [Google Scholar] [CrossRef] [Green Version]
  45. Gillison, M.L.; Trotti, A.M.; Harris, J.; Eisbruch, A.; Harari, P.M.; Adelstein, D.J.; Jordan, R.C.K.; Zhao, W.; Sturgis, E.M.; Burtness, B.; et al. Radiotherapy plus cetuximab or cisplatin in human papillomavirus-positive oropharyngeal cancer (NRG Oncology RTOG 1016): A randomised, multicentre, non-inferiority trial. Lancet 2019, 393, 40–50. [Google Scholar] [CrossRef]
  46. Rischin, D.; King, M.T.; Kenny, L.M.; Porceddu, S.; Wratten, C.; Macann, A.; Jackson, J.E.; Bressel, M.; Herschtal, A.; Richard, F.; et al. Randomized trial of radiotherapy with weekly cisplatin or cetuximab in low risk HPV associated oropharyngeal cancer (TROG 12.01): A Trans-Tasman Radiation Oncology Group study. J. Clin. Oncol. 2021, 39 (Suppl. 15), 6012. [Google Scholar] [CrossRef]
  47. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; 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. [Google Scholar] [CrossRef]
  48. Beaty, B.T.; Moon, D.H.; Shen, C.J.; Amdur, R.J.; Weiss, J.; Grilley-Olson, J.; Patel, S.; Zanation, A.; Hackman, T.G.; Thorp, B.; et al. PIK3CA Mutation in HPV-Associated OPSCC Patients Receiving Deintensified Chemoradiation. J. Natl. Cancer Inst. 2020, 112, 855–858. [Google Scholar] [CrossRef]
  49. Chera, B.S.; Kumar, S.; Shen, C.; Amdur, R.; Dagan, R.; Green, R.; Goldman, E.; Weiss, J.; Grilley-Olson, J.; Patel, S.; et al. Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV-Associated Oropharyngeal Cancer. J. Clin. Oncol. 2020, 38, 1050–1058. [Google Scholar] [CrossRef]
  50. Gerndt, S.P.; Ramirez, R.J.; Wahle, B.M.; Kuperwasser, C.; Gunning, A.; Chaudhuri, A.A.; Zevallos, J.P. Evaluating a clinically validated circulating tumor HPV DNA assay in saliva as a proximal biomarker in HPV+ oropharyngeal squamous cell carcinoma. J. Clin. Oncol. 2021, 39 (Suppl. 15), 6063. [Google Scholar] [CrossRef]
Table 1. Ongoing clinical trials of treatment deintensification in HPV+ OPSCC.
Table 1. Ongoing clinical trials of treatment deintensification in HPV+ OPSCC.
TableN (pts)PhaseStage/EligibilityTreatmentPrimary Endpoint
NCT04502407/
IIT2019-20-Zumsteg-HPVOPC
36IIT0-3N0-2 p16 + OPSCC or cancer of unknown primary (AJCC 8th edition) TORS→
-
High risk pts (positive margins, ECS, ≥5 LNs): RT 50 Gy in 25 fractions, cisplatin 40 mg/m2 d1, 8, 15, 22 and 29
-
All other pts: RT 30 Gy in 15 fractions, cisplatin 40 mg/m2 d1, 8, 15
2-year PFS
NCT02072148/
SIRS
200IIStage I, II, III or early and intermediate stage IVa (T1N0-2B, T2N0-2B) p16 + or HPV+ OPSCCTORS→
Based on pathological features:
Low risk: observation
Intermediate risk: RT 50 Gy
High risk: CRT 50 or 56 Gy
DFS and locoregional control at 3 years (high risk)
DFS and locoregional control at 5 years (low/intermediate risk)
NCT03210103/
ORATOR2
61IIStage c T1-T2, N0-2 (AJCC, 8th ed.) p16 + or HPV+ OPSCCRadiation +/− chemotherapy vs. transoral surgery+ LN dissection +/− RTOS at 2 years
NCT0321571954IIT1-T2, N1-N2b or T3, N1-N2b (AJCC 7th Edition) p16 + OPSCCInterval scan at 4 weeks post CRT:
≤40% nodal shrinkage: standard dose CRT
>40 % nodal shrinkage: reduced dose CRT
PFS at 2 years
NCT04444869/
ENID
28IIT1-T3, N0-N2c (AJCC, 7th ed.) p16 + OPSCCCisplatin-based CRT and RT dose de-escalation to clinically and radiologically uninvolved LNsRate of PEG tube placement
NCT03323463300IIT1-2, N1-2c HPV+ OPSCCRT 30 Gy in 3 weeks + chemotherapy (cisplatin or carbo/5FU)Effectiveness
NCT04900623/ReACT145IIStage I, II, III (AJCC, 8th ed.) p16+ HPV+ OPSCCBased on ctDNA levels
High risk: standard dose RT/CRT in 7–8 weeks
Low risk: low dose RT/CRT in 5–6 weeks
PFS at 2 years
NCT03875716/
ADAPT
111IIStage cT1-T2 cN0-N1 (AJCC, 8th ed.) p16 + or HPV ISH/PCR + cancer of tonsil/base of tongueBased on pathology following curative-intent surgery with anticipated negative margins
Low risk: observation
Intermediate risk: RT 46 Gy
High risk: RT 60 Gy without chemo
DFS at 2 years
NCT03410615180IIT1-2 N1 (smoking ≥ 10 pack years), T3 N0-N1 (smoking ≥ 10 pack years), T1-3 N2 (any smoking hx) (AJCC 8th edition) p16 + OPSCCStandard cisplatin-based CRT vs. Durvalumab+ RT and adjuvant durvalumab vs.NCT03410615
NCT0361813482Ib/IIStage cT0-3 cN0-2b p16 + OPSCCSBRT+ durvalumab → TORS + LN dissection (Cohort 1)
SBRT+ durvalumab/tremelimumab → TORS+ LN dissection (Cohort 2)
Incidence of AEs, PFS at 2 years
NCT03799445180IIT1N2a-N2CM0, T2N1-N2CM0, T3N0-N2CM0 (AJCC 7th Edition) p16 + HPV+ OPSCCNivolumab + Ipilimumab + Reduced dose RT 50–66 GyDLT (safety lead in phase), CR, PFS
NCT03623646/
CITHARE
11IINewly diagnosed T1 N1-N2 or T2-T3 N0 to N2 (AJCC 8th edition) p16 + OPSCCRadiotherapy + Cisplatin vs. Radiotherapy + DurvalumabPFS at 12 months
NCT046384651000observationalHPV+ OPSCC or unknown primaryBased on clinical stage:
A (cT1-3 N0-1 tonsillar, c T1-2, N0-1 non tonsillar, cT0N1 unknown primary): TORS + LN dissection
B (cT1-T3 N1 tonsillar, cT-T2 N1-N2):
TORS +6 cycles of Cisplatin 40 mg/m2
C (cT1-T2 N2 tonsillar, cT0 N2 unknown primary): 6 Cycles of Cisplatin 40 mg/m2 + 60 Gy RT
D (cT1-3 N3 cT4, any N tonsillar, cT3-4, any N, cAny T N3 non tonsillar, cT0N3 unknown primary): 7 Cycles of Cisplatin 40 mg/m2 + 70 Gy RT
OS and DFS at 10 years
NCT0360150714Ph II windowClinical Stage I-IVA p16 + OPSCCNeoadjuvant alpelisib → surgery Quantitative change in the sum of RECIST measurable lesions,
Change in tumor size in patients with genomic PIK3CA pathway alteration
NCT0334237824ObservationalStage III-IVB (AJCC 8th edition) intermediate or low risk HPV+ OPSCCCRT (70 Gy with cisplatin 40 mg/m2) →
PET/MRI prior the initiation of CRT, after 2 weeks of CRT and 3 months following the completion of CRT
Radiographic change in primary tumor and largest LN
NCT03952585711II/IIIClinical stage T1-2 N1M0 or T3 N0-1 M0 (AJCC 8th edition) p16 + OPSCCIMRT+ cisplatin vs. reduced dose IMRT+ cisplatin vs. reduced dose IMRT + nivolumab PFS, QOL
NCT0322400075IIClinical stage T1-2 N0-2b M0 (AJCC 7th edition) p16 + or HPV DNA ISH OPSCCMRI guided IMRT vs. standard IMRTLocoregional control, Composite dysphagia outcome
NCT03077243/
LCCC 1612
215IIT0-3 N0-2c HPV+ or p16 + OPSCCBased on smoking history/p53 status:
-
≤10 pack years → Reduced dose RT 60 Gy + cisplatin 30–40 mg/m2 weekly or cetuximab or Carbo/Paclitaxel or Carbo
-
10 pack years, no p53 mutation → Reduced dose RT 60 Gy + cisplatin 30–40 mg/m2 weekly or cetuximab or Carbo/Paclitaxel or Carbo
-
>10 pack years, p53 mutation → Standard dose RT 70 Gy + cisplatin 30–40 mg/m2 weekly or cetuximab or Carbo/Paclitaxel or Carbo
2 year PFS
NCT02945631/
Quarterback 2b
65IIStage III-IV p16 + and HPV+ OPSCC Reduced dose RT 56 Gy in 2 Gy fractions or 50.4 Gy in 1.8 fractionsPFS at 3 years
Abbreviations: AE = Adverse Event, AJCC = American Joint Committee of Cancer, CRT = ChemoRadiation, CR = Complete Response, DFS = Disease Free Survival, DLT = Dose Limiting Toxicities, HPV = Human Papilloma Virus, IMRT = Intensity Modulated Radiation Therapy, ISH = In Situ Hybridization, LN = Lymph Dissection, OPSCC = Oropharyngeal Squamous Cell Carcinoma, PEG = Percutaneous Endoscopic Gastrostomy, PFS = Progression Free Survival, TORS = TransoOral Surgery, QOL = Quality of life.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Economopoulou, P.; Kotsantis, I.; Psyrri, A. De-Escalating Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma. Viruses 2021, 13, 1787. https://doi.org/10.3390/v13091787

AMA Style

Economopoulou P, Kotsantis I, Psyrri A. De-Escalating Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma. Viruses. 2021; 13(9):1787. https://doi.org/10.3390/v13091787

Chicago/Turabian Style

Economopoulou, Panagiota, Ioannis Kotsantis, and Amanda Psyrri. 2021. "De-Escalating Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma" Viruses 13, no. 9: 1787. https://doi.org/10.3390/v13091787

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

Article Metrics

Back to TopTop