You are on page 1of 15

REVIEWS

The changing therapeutic landscape


of head and neck cancer
John D. Cramer1, Barbara Burtness2, Quynh Thu Le3 and Robert L. Ferris4,5,6*
Abstract | Head and neck cancers are a heterogeneous collection of malignancies of the upper
aerodigestive tract, salivary glands and thyroid. In this Review , we primarily focus on the changing
therapeutic landscape of head and neck squamous cell carcinomas (HNSCCs) that can arise in the
oral cavity , oropharynx, hypopharynx and larynx. We highlight developments in surgical and
non-​surgical therapies (mainly involving the combination of radiotherapy and chemotherapy),
outlining how these treatments are being used in the current era of widespread testing for the
presence of human papillomavirus infection in patients with HNSCC. Finally , we describe
the clinical trials that led to the approval of the first immunotherapeutic agents for HNSCC, and
discuss the development of strategies to decrease the toxicity of different treatment modalities.

Worldwide, >830,000 individuals are diagnosed with screening cannot be conducted by a corollary of the Pap
head and neck cancer and >430,000 patients die from smear test used for the early detection of uterine cer-
this disease each year1; the annual incidence in the USA vical cancer18. HPV vaccines are 90–100% effective at
is ~65,000 patients2. Head and neck squamous cell carci- preventing anogenital HPV infections and precancer-
nomas (HNSCCs) account for 90% of all head and neck ous lesions19. In 2018, the FDA extended the approved
cancers3 and can be detected at various anatomical sites age range of candidates for the vaccine against HPV,
(Fig. 1). Approximately 75% of HNSCCs are associated GARDASIL 9, to include men and women <45 years
with tobacco and alcohol use4,5; however, a minority of of age19,20. Epidemiological data suggest that prophy-
HNSCCs are caused by human papillomavirus (HPV) lactic HPV vaccination reduces the prevalence of oral
infection6,7. In the USA, the declining use of cigarettes HPV infection by 88–93%. Vaccination is expected to
has been associated with a decrease in HNSCC inci- reduce the incidence of oropharyngeal cancer by 2060,
dence, with the exception of HPV-​driven (HPV+) oro- on the basis of slow uptake and the latency period from
pharyngeal squamous cell carcinoma (OPSCC), which infection to clinical presentation with OPSCC12,21,22.
has a rapidly increasing incidence8. Patients with HPV+ OPSCC typically present with
1
Department of
Otolaryngology, Wayne State
HPV infection has been established to have a causal small primary tumours and cervical lymphadeno­
University School of Medicine, role in the development of HNSCC, primarily of the pathy23, and might initially be diagnosed with squamous
Detroit, MI, USA. oropharynx9. The epidemiology, pathophysiology and cell carcinoma of unknown primary (SCCUP) of the
2
Department of Medicine response to treatment of HPV+ OPSCC differ sharply head and neck. Despite the high frequency of spread
and Yale Cancer Center, from that of HPV− disease10,11. The demographic charac- to cervical lymph nodes, HPV+ OPSCC is associated
Yale School of Medicine,
teristics of patients with HPV+ OPSCC are also different, with a significantly more favourable prognosis than
New Haven, CT, USA.
including a higher incidence in men and a younger mean HPV− OPSCC (3-year overall survival (OS) 82.4%
3
Department of Radiation
Oncology, Stanford
age at diagnosis12. The prevalence of HPV+ OPSCC var- versus 57.1%; P < 0.001)10. In contrast to patients with
University, Palo Alto, CA, ies substantially by geographical location, with 60–70% HPV− HNSCC, those with HPV+ OPSCC presenting
USA. of OPSCCs in the USA13 and Western Europe14 but with cervical lymph node metastases have excellent
4
Department of <10% in low-​income and middle-​income countries15. survival outcomes (5-year OS of 80–87% for patients
Otolaryngology, University of HPV infection might have a role in a small proportion with N1–N2c HPV + OPSCC versus 37–58% for
Pittsburgh, Pittsburgh, PA, of HNSCCs located outside the oropharynx. Although those with N1–N2c HPV− OPSCC)24. The staging groups
USA.
the effect of HPV in cancers in these sites is less well defined by the American Joint Committee on Cancer
5
Department of Immunology,
understood, HPV positivity is also associated with more (AJCC) in 2010 (ref.25) failed to provide an accurate
University of Pittsburgh,
Pittsburgh, PA, USA. favourable outcomes16,17. prognosis of patients with HPV+ OPSCC23. As a result,
6
UPMC Hillman Cancer
The latency period between HPV infection and clin- the eighth edition of the AJCC staging manual uses a
Center, Pittsburgh, PA, USA. ical presentation with HPV+ OPSCC is estimated to be unique staging system for HPV+ OPSCC26. Application of
*e-​mail: ferrisrl@upmc.edu 10–30 years12. HPV+ OPSCC arises in deep crypts in the the new staging system resulted in the downstaging
https://doi.org/10.1038/ tonsil and the base of the tongue (BOT) and is not asso- of 92% of patients with HPV+ OPSCC, and the propor-
s41571-019-0227-z ciated with a pre-​malignant clinical lesion. Therefore, tion of patients with stage I disease increased from 3% to

Nature Reviews | Clinical Oncology


Reviews

that offer the possibility to limit the toxicity of treatment


Key points
and further improve outcomes of patients with HNSCCs
• Current treatments for human papillomavirus-​driven (HPV+) oropharyngeal squamous in one of the four major sites: the oral cavity, oropharynx,
cell carcinoma (OPSCC), primarily derived from those for the more aggressive HPV− hypopharynx or larynx. Noteworthy advances in head
head and neck squamous cell carcinoma (HSNCC), might be more intensive than and neck cancers of other histologies are also discussed.
necessary for patients with favourable risk features and an excellent prognosis.
• Multiple prospective studies have investigated various treatment de-​intensification Surgery
strategies with promising early results; however, pending definitive conclusions, HPV+
Surgery remains the preferred treatment for oral cavity
OPSCC should continue to be treated with established standard-​of-care therapies,
squamous cell carcinomas (OCSCCs)35 and advanced-​
known to yield very high survival.
stage (T4a) laryngeal or hypopharyngeal cancers36.
• Minimally invasive transoral surgical techniques provide an alternative treatment
Surgical treatment has improved dramatically over
option for OPSCC; retrospective evidence is promising, but results from prospective
randomized trials are required to fully integrate these approaches into the treatment the past 30 years. For advanced-​stage primary cancers,
armamentarium. microvascular reconstruction has become the standard-​
• More-​precisely targeted radiotherapy (with incorporation of intensity-​modulated of-care modality; this approach enables cancer resection
radiation therapy, molecular imaging-​guided therapy, adaptive therapy and proton with improved restoration of function37. For early-​stage
beam therapy) has the potential to decrease the long-​term toxicity of radiotherapy. cancers, minimally invasive surgical treatment options
• Anti-​EGFR therapy with cetuximab improves survival in the curative and recurrent have similarly improved and have now become a surgi-
and/or metastatic settings; however, no other molecularly targeted approach has cal option associated with reduced morbidity for many
prolonged survival in HNSCC. patients.
• Anti-​programmed cell death 1 (PD-1) therapies, currently approved for platinum-​
refractory recurrent and/or metastatic HNSCC, offer the prospect of long-​term Minimally invasive transoral surgery
remissions with fewer toxicities than traditional cytotoxic chemotherapy for a Definitive chemoradiotherapy was widely adopted
minority of patients, and are expected to advance to earlier lines of therapy. in the late 1990s in an effort to avoid the morbidities
associated with open surgery38,39. Transoral robotic sur-
gery (TORS) and transoral laser microsurgery (TLM)
64%27. Of note, 98% of patients from the cohort in which are surgical modalities that have emerged as promising
the new staging criteria were developed and validated alternatives with far fewer morbidities than open surgery
received non-​surgical therapy with definitive radio- (reduction in feeding tube support at 1 year from 31%
therapy or chemoradiotherapy24. Importantly, while to 3% and reduction in length of hospital stay from 8.0 to
the guidelines for HPV+ OPSCC in the eighth edition 3.8 days)40. These approaches enable improved visuali-
of the AJCC staging manual were designed to improve zation of deep regions of the upper airway while using
prognostication, they are based on multimodal therapy the mouth as a natural orifice41. Transoral surgery is
and do not provide sufficient data to support treatment primarily used to access primary OPSCCs of small-​to-­
de-​intensification on the basis of revised stage groups moderate size (T1–T2) and T1–T2 cancers of the larynx
alone. Treatment decisions should be driven by the clin- and hypopharynx42.
ical criteria used to select patients for inclusion in land- A retrospective analysis of records from several cen-
mark trials of definitive therapy, rather than by novel tres using TLM supports this modality as an effective
stage-​defined groups. Consequently, the identification and safe approach43. Several prospective trials of TLM
of clinical risk categories and/or biomarkers that can be from cooperative groups are ongoing (NCT01898494
used to modify the dose and duration of therapies in and NCT02215265), but no results have been published
patients with HPV+ OPSCC is a major interest of several yet. Since the FDA approval of the da Vinci surgical
research groups10,28,29. robotic system for transoral resection in the orophar-
In the setting of HPV− HNSCC, treatment with pri- ynx in 2009 (ref.44), TORS has been rapidly adopted by
mary surgery or radiotherapy improves the cure rate the surgical community for OPSCC45. A second robotic
of patients with early-​stage disease, with a 5-year OS of device by Medrobotics was approved in 2015 (ref.46).
70–90%30. Unfortunately, two-​thirds of patients present Compared with open approaches, transoral surgery
with advanced-​stage disease, with large primary disease improves the safety, efficacy and tolerability of surgery47.
(T3–T4) and/or cervical adenopathy31. The survival Most of the published evidence on transoral surgery
outcomes of patients with locoregionally advanced dis- comes from single-​institution series (with pooled results
ease are modest, ranging from a 5-year OS of 49% for published in ref.47). The indications for upfront surgi-
patients with laryngeal cancer to 25% for those with cal treatment of OPSCC are controversial. Proponents
hypopharyngeal cancer32. The standard-​of-care treat- of surgery point out that, when selected appropriately,
ment for locoregionally advanced disease is multimodal. 69–83% of patients who undergo surgery avoid the
Many studies have demonstrated that the addition of need for adjuvant chemotherapy and 11–47% also
concurrent chemotherapy to radiotherapy results in avoid the need for radiotherapy42,43,48,49. Retrospective
modest improvements in survival33. The awareness that studies have further suggested improved functional
more-​intense multimodal therapy regimens result in a results, including decreased dependency on a gastro­
higher incidence of severe acute and late toxicities34 has stomy tube for patients who undergo upfront transoral
generated renewed interest in therapeutic strategies that surgery compared with those who receive definitive
minimize toxicity while maintaining disease control. chemoradiotherapy 47,50,51. However, proponents of
In this Review, we focus on treatment advances from definitive chemoradiotherapy point out that 17–31%
clinical trials with results published in the past few years of patients who undergo surgery eventually need therapy

www.nature.com/nrclinonc
Reviews

Cancer subtype Annual incidence Annual mortality


Worldwide USA Worldwide USA

Oral cavity and lip 358,864 33,950 177,384 6,800


Nasopharynx
Laryngeal 177,422 13,150 94,771 3,710

Oral Nasopharyngeal 129,079 Rare 72,987 NA


cavity Oropharynx
Oropharyngeal 92,887 NA 51,005 NA

Hypopharyngeal 80,608 NA 34,984 NA


Hypopharynx
Larynx

Fig. 1 | incidence rates and mortality for patients with head and neck squamous carcinoma 1,2.  Results of worldwide
and US investigations of the epidemiology of head and neck squamous cell carcinoma are shown; national databases
frequently group different subtypes together (for example, oral cancers and pharyngeal cancers). For this reason, some
oropharyngeal cancers of the base of the tongue can be misclassified as cancers of the oral cavity. Furthermore, the
incidence of human papillomavirus-​associated oropharyngeal cancer is regionally specific: human papillomavirus
accounts for 70% of oropharyngeal cancers in the USA and for 10% in low-​income and middle-​income countries5,7,9.
The incidence of nasopharyngeal cancer is included as it significantly impacts the global burden of head and neck cancer,
particularly in Southeast Asia, although the incidence is low in the USA. NA , not applicable.

intensification to triple-​modality therapy (involving sur- reduces morbidity35. Historically, elective neck dissection
gery, chemotherapy and radiotherapy)52. The lack of data (END) has been advocated for patients with neck dis-
from prospective studies of transoral surgery led to the ease and no lymph node involvement (N0) and a >20%
development of several trials investigating TORS and risk of occult nodal disease62, although this approach is
TLM in patients with HPV+ OPSCC. not supported by data from randomized studies. D’Cruz
et al.63 performed a randomized trial of END versus
SCCUP of the head and neck observation in 500 patients with stage I/II OCSCC.
SCCUP accounts for 1–4% of all HNSCCs, and 69% Patients in the observation arm who developed recur-
of SCCUPs are attributable to HPV53,54. Historically, rent disease in the neck underwent salvage therapy with
if physical examination, fibre-​optic examination and therapeutic neck dissection63. At 3 years of follow-​up,
imaging were non-​revealing, patients underwent patients treated with END had a 12.5% improvement
examination under anaesthesia involving biopsy of the in OS compared with those in the observation group63.
nasopharynx, BOT, and pyriform sinus and palatine ton- Subgroup analysis showed that the improvement in
sillectomy55. This approach enabled the identification of OS was restricted to patients with a depth of tumour
a primary tumour in 24–39% of patients but many BOT invasion of >3 mm. Importantly, patients in the END
cancers were not detected54. Before examination under group were more likely to receive adjuvant radiotherapy
anaesthesia, tissue specimens should be tested for HPV than those in the observation group (48.6% versus 34%).
(either with p16 immunohistochemistry and/or PCR-​ The relative therapeutic contribution of END versus
based HPV testing) to provide an indication of the pri- adjuvant radiotherapy to survival was not explored in
mary site56. In single-​institution and multi-​institution this study. Nevertheless, a small prospective study of
retrospective studies, transoral surgery to excise lingual single-​modality elective neck treatment with radio-
tonsillar tissue in the BOT led to a twofold-​to-threefold therapy for patients with OCSCC demonstrated a high
improvement in the rates of detection of SCCUP to locoregional recurrence rate (23%), suggesting that adju-
72–89%57–61. Nevertheless, the addition of lingual ton- vant radiotherapy alone does not lead to a substantial
sillectomy increases the risk of acute morbidities and OS benefit64. Overall, these results provide important
can impair swallowing, and thus requires prospective level 1 evidence supporting END as the standard-​
evaluation. of-care approach in patients with node-​n egative
OCSCC with a depth of invasion of >3 mm.
Regional metastatic disease D’Cruz et al.63 did not evaluate alternative methods
Over the past 30 years, radical neck dissection has largely for the pathology-​based assessment of regional meta­
been supplanted by selective dissection as a therapeu- static disease, such as sentinel lymph node biopsy
tic procedure to eradicate lymph node metastases in (SLNB). While END is a safe operation, even the mor-
both the elective and the therapeutic settings; the latter bidities associated with selective neck dissections might
approach spares critical non-​lymphatic structures and be under-​appreciated65,66. In the upper aerodigestive

Nature Reviews | Clinical Oncology


Reviews

tract, SLNB has primarily been evaluated in patients schedules can enhance efficacy. Altered fractionation
with OCSCC who had primary tumours that were eas- includes hyperfractionation, in which the total radi-
ily accessible with radiotracer injections. Prospective, ation dose is increased by administering twice-​daily
single-​arm trials have found SLNB to be highly effec- fractions while keeping the overall treatment duration
tive for stage I/II OCSCC67–69, and a meta-​analysis the same at ~7 weeks, and accelerated fractionation, in
has shown that the sensitivity and negative predictive which the total dose is unchanged but delivered over a
value of this approach are 95% and 96%, respectively70. shorter duration (for example, 5–6 weeks). The updated
Unfortunately, no direct randomized comparison of MARCH meta-​analysis74 of data from 34 trials testing
END versus SLNB for OCSCC has fully examined the altered fractionation schedules in HNSCC showed an
risks and benefits of these approaches, although one improvement in 5-year OS favouring altered fraction-
such study is currently under development by the NRG ation over conventional fractionation without chemo-
Oncology group. A rando­mized controlled trial of neck therapy74. The OS benefit was restricted primarily to
dissections based on sentinel lymph node navigation hyperfractionation, with a 5-year absolute OS benefit
versus standard selective neck dissections in N0 oral of 8.1%, according to data from 8 trials involving 1,733
cancers conducted by a Japanese group has completed patients74. In a separate analysis of data from 5 trials
accrual, but the results are not yet available. including 986 patients comparing conventional radio-
therapy fractionation plus concomitant chemotherapy
Assessment after chemoradiotherapy versus altered fractionation alone, the latter was associ-
Evidence from retrospective studies suggests that ated with inferior outcomes74. Of note, these five trials
patients with a complete response to definitive chemo- primarily tested accelerated fractionation (shortening
radiotherapy detected on post-​t reatment PET–CT the radiation course by 1–2 weeks), which has not been
imaging might be able to avoid post-​treatment neck shown to increase OS compared with conventional frac-
dissection, but no prospective data are available. In a tionation74. Unfortunately, hyperfractionation has never
phase III non-​inferiority trial, 564 patients with HNSCC been tested in comparison with or as a component of
of an advanced nodal stage were randomly assigned to chemoradiotherapy and thus its efficacy and safety com-
receive definitive chemoradiotherapy followed by either pared with chemoradiotherapy are unclear. Moreover,
END within 4–8 weeks or PET–CT at 12 weeks71. In the hyperfractionation schedules using intensity-​modulated
PET–CT group, only patients with an incomplete or radiotherapy (IMRT) techniques are time-​consuming,
equivocal response on imaging underwent neck dissec- both in terms of institutional resources and for patients,
tion. Following definitive chemoradiotherapy, patients and have not been widely adopted.
in the PET–CT group had non-​inferior 2-year OS com-
pared with those in the END group (84.9% versus 81.5%; Intensity-​modulated radiotherapy
P = 0.004)71. In addition, patients in the PET–CT group Technological advances focusing on more precise delin-
underwent fewer surgical procedures (19.1% versus eation of targets and more accurate delivery of radio-
78.4%) and, correspondingly, had fewer surgical compli- therapy that enables sparing of non-​malignant tissues
cations (7.8% versus 29.4%), with a lower cost (savings of can reduce the morbidity of treatment. IMRT enables
US$2,190). Two important caveats merit emphasis. First, the delivery of high doses per fraction of radiation
the proportion of patients with N3 disease (>6 cm in any to the tumour while constraining the dose delivered to
dimension) included in this study was <4%; therefore, adjacent tissues. Several randomized trials, includ-
the results cannot be extrapolated to this disease setting. ing the PARSPORT trial75, have revealed a lower inci-
Second, 85% of patients enrolled had OPSCC, and the dence of xerostomia with IMRT than with conventional
majority of those patients had HPV+ disease; therefore, radiotherapy75–77. In addition, a large randomized trial
these results should be interpreted cautiously when conducted in China73 demonstrated an OS benefit
considering patients with HPV− disease. Nonetheless, and reduced toxicities with IMRT versus conven-
these results indicate that END after definitive chemo- tional radiotherapy78. Owing to these improvements
radiotherapy can be avoided in the majority of patients. and other quality of life (QOL) benefits, IMRT has
The American College of Radiology Imaging Network become a routine modality in the treatment of patients
(ACRIN) 6685 trial assessed the accuracy of PET–CT in with HNSCC79.
the preoperative management of N0 neck disease72. This
trial demonstrated a 92.2% negative predictive value of Adaptive radiotherapy
PET–CT for each side of the neck and also indicated a Radiotherapy treatment plans are traditionally designed
role of PET–CT in preoperative nodal staging, with an using a single pretreatment time point that does not take
improved negative predictive value of 94.2% demon- into account anatomical changes during the course of
strated by setting the threshold standardized uptake treatment. During treatment, tumour and nodal vol-
value intensity to 1.8 (ref.73). umes might contract by up to 3% per day80. Adaptive
radiotherapy is designed to alter treatment on the basis
Radiotherapy of such changes in tumour and non-​malignant tissues
Altered fractionation during the course of treatment. A prospective trial
Conventional fractionated radiotherapy for patients with involving 22 patients with OPSCC who received adap-
HNSCC involves the use of 2 Gy fractions delivered once tive radiotherapy showed improved dosimetry but no
daily to a total dose of 70 Gy. A major focus of investiga- differences in locoregional control (lack of disease recur-
tion has been to determine whether altered fractionation rence at the primary site or regional lymph nodes)81.

www.nature.com/nrclinonc
Reviews

While an appealing possibility for further investiga- High-​dose versus low-​dose cisplatin. Low-​dose weekly
tions, adaptive radiotherapy is not currently used on a cisplatin offers several advantages over high-​dose cis-
routine basis. platin, including ease of administration and reduced
toxicity90. Despite widespread use, this regimen has only
Proton beam therapy been evaluated previously in small-​cohort prospective
IMRT delivered using a photon beam can result in a trials88,91. Quon et al.92 investigated the safety and effi-
large volume of low-​dose radiation becoming deposi­ cacy of the addition of 20 mg/m2 weekly cisplatin to
ted in non-​malignant tissues located along the beam radiotherapy. This trial is important because it showed
path. Protons behave differently to photons, releasing that low-​dose radiosensitizers do not have a role in the
the greatest amount of energy at a defined depth, thus treatment of unresectable HNSCC. Between 1982 and
enabling reductions in the dose delivered to adjacent 1987, 308 eligible patients were accrued to this study,
non-​malignant tissues. Advances in the technology of which had a median follow-​up duration of 62 months.
proton beam delivery and planning during the past few Median failure-​f ree survival was 6.5 months and
years might enable further sparing of radiation to non-​ 7.2 months in the radiotherapy and radiotherapy plus
malignant tissue. Currently available proton beam treat- cisplatin groups, respectively (P = 0.30), and median
ments can use a combination of different beam angles OS was 13.3 months and 11.8 months, respectively
and/or intensity modulation similar to that used in pho- (P = 0.81). The incidence of expected acute toxicities
ton therapy, known as intensity-​modulated proton beam was significantly higher with the addition of cispla-
therapy (IMPT). Multiple comparative studies of treat- tin (for example, the incidences of grade 3–4 acute
ment planning approaches have shown that protons pro- haematological toxicities were 4% with radiotherapy
duce dose distributions that would deliver less radiation and 11% with radiotherapy plus cisplatin; P < 0.001).
to structures that are important for salivary function and Oesophageal and laryngeal late toxicities were more
swallowing (reviewed previously82). This observation has frequent in the radiotherapy plus cisplatin group (9%
resulted in widespread enthusiasm for proton therapy; versus 3%, P = 0.03; and 11% versus 4%, respectively,
however, evidence of the comparative effectiveness of P = 0.05). The results of this trial demonstrate that
IMPT versus standard IMRT is limited. With IMRT, the inadequate radiation sensitization with low-​dose plati-
incidence of chronic severe dysphagia in treated patients, nating agents can worsen toxicities without improving
including those with OPSCC, remains low (7–8% feed- survival; indeed, OS was shorter on a numerical basis
ing tube dependency 1 year after IMRT versus 2% with in those receiving low-​dose cisplatin, but this difference
IMPT)83,84. Some investigators have hypothesized that was not statistically significant.
this result is related to the dose delivered to the constric- Noronha et al.93, in a single-​centre randomized trial,
tor muscles and the oral cavity81,85,86. Therefore, a large compared low-​dose weekly cisplatin (30 mg/m2) and
multi-​institutional randomized phase II/III trial designed high-​dose cisplatin (100 mg/m2) every 3 weeks both
to determine whether IMPT is superior to IMRT concurrently with radiotherapy in 300 patients with
(NCT01893307) in decreasing late severe toxicities, and stage III/IV HNSCC. The primary end point of the
thus improving the QOL of survivors, is currently ongo- trial was non-​inferior locoregional control at 2 years
ing. These lines of investigation are important for the in a population of patients with OCSCC who predo­
integration of proton therapy into treatment algorithms. minantly received postoperative chemoradiotherapy
(93%). High-​dose cisplatin every 3 weeks resulted in a
Systemic therapy superior 2-year locoregional control rate (15% improve-
Cytotoxic chemotherapy ment; P = 0.014), although OS was not significantly
Over the past three decades, considerable effort has worse93. The doses used in the weekly cisplatin arm,
been devoted to understanding the benefit of adding however, might not have been adequate, as a retrospec-
systemic therapy to radiotherapy in the curative setting tive study88 comparing the efficacy of weekly 40 mg/m2
in patients with HNSCC. Cytotoxic chemotherapy can cisplatin and 100 mg/m2 cisplatin every 3 weeks demon-
be employed as induction, concurrently with definitive strated equivalent OS and locoregional control rates for
radiotherapy, or combined with adjuvant radiotherapy. the two regimens and fewer incidences of neutropenia,
A meta-​analysis of data from 93 randomized trials of nephrotoxicity and ototoxicity with low-​dose cisplatin
chemoradiotherapy81 identified a 6.5% absolute 5-year (unspecified grade of toxicities)94. Furthermore, the
OS benefit for concurrent chemoradiotherapy versus question as to whether the cumulative or peak dose
radiotherapy87. In this analysis, concurrent cisplatin is the more important variable remains to be defini-
monotherapy provided the same magnitude of benefit tively answered. In the RTOG 0129 trial, the efficacy
as combination chemoradiotherapy87,88. As a radiosen- of two doses of 100 mg/m2 cisplatin (cumulative dose of
sitizer, high-​dose cisplatin (100 mg/m2) administered 200 mg/m2) during accelerated radiotherapy was com-
every 3 weeks on days 1, 22 and 43 concurrently with pared with that of three doses of 100 mg/m2 cisplatin
conventionally fractionated radiotherapy over 7 weeks in (cumulative dose of 300 mg/m2) with conventional
the definitive setting, or on days 1 and 22 for accelerated fractionation. No significant difference was detected,
fractionation radiotherapy or as adjuvant radiotherapy albeit 30% of patients in the standard-​of-care arm did
over 6 weeks89, remains the gold standard. Nevertheless, not receive a third dose of cisplatin89. Higher dosing
the adverse effects of high-​dose cisplatin are substan- (40–50 mg/m2 per week) might improve the efficacy of
tial, thus fuelling the search for better-​tolerated dosing weekly cisplatin and is being investigated in an ongoing
schedules or agents. study90,95,96.

Nature Reviews | Clinical Oncology


Reviews

Induction chemotherapy. Induction chemotherapy between cetuximab and these other agents remains to
followed by radiotherapy alone has been extensively be determined110–112.
investigated in patients with locoregionally advanced In the curative setting, the addition of anti-​EGFR
HNSCC. Meta-​analyses of data from randomized agents to chemoradiotherapy has yielded conflict-
trials of intensive chemotherapy81,91 have identified ing results. The phase III RTOG 0522 trial106 failed to
a non-​s ignificant 2.2% OS benefit with induction demonstrate improved outcomes with the addition of
chemotherapy before radiotherapy compared with cetuximab to cisplatin and radiotherapy. Conversely,
radiotherapy alone87,97. Randomized trials have since an abstract presented in 2018 from a large phase III
established that the combination of docetaxel, cisplatin trial conducted in India involving 536 patients suggests
and 5-fluorouracil is a more active regimen than cispla- that nimotuzumab, which has lower EGFR-​binding
tin and 5-fluorouracil; however, no level 1 evidence is affinity and a more favourable toxicity profile than
available to support the routine use of either regimen as cetuximab, improved both PFS and disease-​free sur-
induction therapy98,99. Three phase III trials in patients vival (DFS) when added to radiotherapy plus weekly
with locoregionally advanced disease compared the cisplatin (30 mg/m2)113. The trial did not demonstrate
addition of taxane-​containing induction chemotherapy an improvement in OS and can be criticized for having
to cetuximab plus radiotherapy or to cisplatin plus radio­ a potentially inadequate control arm (weekly cisplatin
therapy on the effect of OS100–102. None of these stud- with radiotherapy)113. Other combination strategies
ies revealed a significant survival benefit of induction involving cetuximab have shown promising results; for
chemotherapy100–102. These studies were slightly under- example, the 2-year OS in a randomized phase II study
powered because baseline statistical assumptions had was higher for patients receiving postoperative chemo-
not accounted for the lower event rate associated with radiation who were treated with docetaxel plus cetuxi-
HPV+ disease, and two studies closed early; however, mab than for those treated with cisplatin plus cetuximab
the results consistently showed a lack of effect on OS, (79% versus 69%; P = 0.04)114.
with HRs of 0.91–1.12. Induction chemotherapy might
have lowered the rate of distant metastases102, although Other targeted agents. Chung et al.115, The Cancer
the morbidity rate increased (the incidence of serious Genome Atlas Network116 and others109,112,113 have delin-
adverse events increased from 28% to 47%)101. The eated the different genomic subtypes of HNSCC and
utility of induction chemotherapy for chemoselection provided guidance for the development of targeted
in patients with highly locally advanced disease, in which therapies for these cancers115–119. These data confirmed
patients with a >50% response to one cycle of induction the widespread prevalence of loss-​of-function muta-
chemotherapy are selected for organ preservation and tions in TP53 and CDKN2A in HPV− HNSCCs116 (Fig. 2).
patients with a <50% response are selected for surgery, In HPV+ HNSCCs, these groups identified deletions and
and its utility to tailor chemotherapy and radiotherapy point mutations in TRAF3, PIK3CA and E2F1 (ref.116).
regimens remains to be fully explored. Overall, the most promising actionable targets in
HNSCC include proteins involved in the PI3K pathway
Targeted therapies and cyclin-​dependent kinases 4/6 (refs120–122), although
Anti- ​ E GFR therapy. EGFR is overexpressed in up the most common genomic alterations affected tumour
to 90% of HNSCCs; high levels of this protein are suppressor genes, for which targeted therapies have had
correlated with decreased survival103,104. Cetuximab little success.
is a chimeric IgG1 monoclonal antibody that com-
petitively inhibits ligand binding to EGFR, fostering Therapy de-​intensification in HPV+ OPSCC
EGFR internalization and altering EGFR-​dependent Given the generally favourable prognosis of patients
signalling. Cetuximab also mediates antigen-​specific with HPV+ OPSCC and the considerable morbidities
immune responses, leading to cell death105. Cetuximab associated with current treatments, multiple studies
is approved by the FDA for the treatment of HNSCC have investigated whether therapy de-​intensification
in multiple settings. Treatment with cetuximab mono­ could provide similar levels of disease control to
therapy is associated with a 13% response rate in those achieved with conventional therapy, but with
patients with recurrent and/or metastatic HSNCC106. fewer acute and/or chronic toxicities (Table  1). De-​
The addition of cetuximab to first-​line chemother- intensification is probably most appropriate in patients
apy improves response rates, progression-​f ree sur- with HPV+ OPSCC with the lowest risk of disease
vival (PFS) and OS (median OS of 10.1 months versus recurrence, such as those with a smoking history of
7.4 months; P = 0.04) in patients with recurrent and/or <10 pack-​years and non-​T4 cancer10,123–126. Several strate-
metastatic HNSCC107,108. Finally, in patients with loco­ gies are currently used to reduce the toxicity of treatment.
regionally advanced disease, cetuximab plus radiother- The first possible strategy is reliance on resection alone
apy improves OS compared with radiotherapy alone in patients with HPV+ T1N0M0 OPSCC. The second
(49.0 months versus 29.3 months; P = 0.03)109. Impor­ approach involves the omission of cisplatin chemotherapy
tantly, numerous other anti-​EGFR agents (such as pani- from definitive radiotherapy regimens to reduce the
tumumab, zalutumumab, gefitinib, afatinib or lapatinib) risk of both acute and late effects. This exclusion can be
have failed to demonstrate notable clinical activity in achieved by omitting systemic therapy (NCT02254278;
patients with HNSCC. Whether differences in tumour ongoing) or by substituting cetuximab for cisplatin
penetration, immunological effects or toxicity pro- (NCT01302834; completed and published)127 or inhibi-
files contribute to the apparent differences in efficacy tors of programmed cell death 1 (PD-1) (NCT02764593;

www.nature.com/nrclinonc
Reviews

HPV– (n = 243) HPV+ (n = 36)


EGFR 15% v 6%
Receptor tyrosine kinases

FGFR1 10% 0%
ERBB2 5% 3%
IGF1R 4% 0%
EPHA2 4% 3%
DDR2 3% 6%
FGFR2 2% 0%
FGFR3 2% 11%
MET 2% 0%
Oncogenes

CCND1 31% 3%
MYC 14% 3%
HRAS 5% 0%

PIK3CA 34% 56%


PI3K

PTEN 12% 6%
PIK3R1 1% 3%

NF1 3% 0%
TSGs

TP53 84% 3%
CDKN2A 58% 0%

Homozygous Protein Protein EGFR vIII MET exon 14 FGFR3–TACC3


Amplification Mutation v
deletion upregulation downregulation deletion skipping fusion

Fig. 2 | Therapeutic targets and potential driver oncogenes in head and neck squamous carcinoma according to
The Cancer genome atlas network. The genes listed reflect somatic alterations and changes in protein expression
identified as reasonable therapeutic targets. The frequency of each genetic alteration is shown next to the gene name
on the far left for human papillomavirus negative (HPV−) and on the far right for HPV+. TSG, tumour supressor gene.
Reproduced from ref.116, Springer Nature Limited.

completed accrual). The third strategy is a mod- (Table 2). Substantial lifetime exposure to tobacco and
est reduction in the dose and/or field of radiation the presence of T4 cancer portended worse outcomes
(NCT02254278; ongoing), and the fourth is the admin- with this approach126. Induction chemoselection with
istration of systemic therapy followed by a more marked paclitaxel and carboplatin followed by paclitaxel plus
reduction in radiation dose delivered to tumours shown radiotherapy or radiotherapy alone have confirmed that
to be chemosensitive (NCT01706939; ongoing). Each induction chemotherapy followed by treatment with a
approach has merits and drawbacks, and none has been reduced radiation dose for patients with chemosensi-
compared with the others. Emerging biomolecular128 tive tumours maintains high cure rates, with 2-year PFS
and clinical risk28 categories and patient preferences will of 92% and 94.5%129,130, and favourable QOL outcomes
probably lead to tailored de-​intensification, according compared with historical controls131.
to the paradigm most suited to the individual’s risk of
locoregional recurrence, risk of distant metastases, and Concurrent chemoradiotherapy
predicted sensitivity to radiotherapy, chemotherapy An alternative attempt at de-​intensification involves
and/or immunotherapy. the substitution of concurrent cetuximab for cisplatin
in definitive chemoradiotherapy regimens. The results
Induction chemotherapy of the phase III RTOG 1016 trial127 comparing radio-
In the Eastern Cooperative Oncology Group (ECOG) therapy plus cetuximab with radiotherapy plus cispla-
1308 phase II trial, patients with HPV+ OPSCC received tin in 849 patients with HPV+ OPSCC were reported
induction chemotherapy with cisplatin, paclitaxel and in 2018. At 5 years, locoregional control, PFS and OS
cetuximab, followed by cetuximab plus radiotherapy were superior with cisplatin and no significant dif-
with reduced or full-​dose radiation to the primary ferences were reported in toxicity rates or swallowing
site on the basis of response: patients with a complete function (Table 2). However, for patients with preserved
response received a dose of 54 Gy, whereas those with- performance status, no significant differences in 5-year
out a complete response received a standard dose of OS were observed between the groups (84.6% versus
69.3 Gy126. As anticipated, 70% of patients achieved 84.0%)127. The results of the smaller De-​ESCALaTE
a clinical complete response at the primary site; these HPV trial132 were also reported in 2018, with similar dif-
patients had a longer PFS than those who did not have ferences in locoregional control observed with cisplatin
a complete response and required full-​dose radiation plus radiotherapy versus cetuximab plus radiotherapy

Nature Reviews | Clinical Oncology


Reviews

Table 1 | Summary of phase ii/iii trials of therapy de-​intensification in human papillomavirus-​driven oropharyngeal squamous cell carcinoma
Study Phase Patients Disease stage Treatment DFS, PFS and/or OS adverse
(n) locoregional events
control rate
Surgical approaches
Ma et al. IIb 80 <10 pack-​years, TORS + adjuvant 30 Gy radiotherapy 2-year DFS: 89% NR 0% grade 3–4 at
(2017)160a resected with (36 Gy for ENE) + docetaxel 2 years
negative margins
Siegel et al. IIb 20 III–IVac Induction chemotherapy (cisplatin, 21-month DFS: NR NR
(2018)161a docetaxel); if >80% response in 85%
primary site, then TORS + ND
Non-​surgical approaches
RTOG 1016 III 849 III–IVac Cisplatin (100 mg/m2 every PFS: 78.4% versus 5-year OS: Acute grade
(ref.127) 3 weeks) + 70 Gy radiotherapy versus 67.3% (P = 0.0002) 84.6% versus 3–4: 82% versus
cetuximab + 70 Gy radiotherapy 77.9% 77% (P = 0.16)
De-​ III 334 III–IVac Cisplatin (100 mg/m2 every 2-year recurrence 2-year No difference
ESCALaTE 3 weeks) + 70 Gy radiotherapy versus rate: 6% versus OS: 97.5% in acute or
HPV132 cetuximab + 70 Gy radiotherapy 16% (P = 0.0007) versus 89.4% late grade 3–5
(P = 0.001) toxicity
ECOG IIb 80 III–IVac Induction chemotherapy (cisplatin, 2-year PFS: 2-year OS: 1-year solid
1308 paclitaxel and cetuximab); if 96% versus 80% 96% versus food dysphagia:
(ref.126) CR in primary site, then 54 Gy (≤54 Gy versus 94% (≤54 Gy 40% versus
radiotherapy + cetuximab; if less >54 Gy) versus >54 Gy) 89%; P = 0.01
than CR in primary site, then 69.3 Gy (≤54 Gy versus
radiotherapy + cetuximab >54 Gy)
Chen et al. IIb 45 III–IVac Induction chemotherapy (2 cycles 2-year PFS: 92% NR Grade 3:
(2017)129 of carboplatin and paclitaxel); if PR 39% and 0%
or CR in primary site then 54 Gy dependency
radiotherapy + paclitaxel; if less on gastrostomy
than PR in primary site, then 60 Gy tube at
radiotherapy + paclitaxel 6 months
Chera et al. IIb 44 T0–3, N0–2c, M0 Cisplatin (30 mg/m2 weekly) + 60 Gy 3-year 95% at 3 years 0% dependency
(2018)133 and ≤10 pack-​years radiotherapy locoregional on gastrostomy
or >10 pack-​years control: 100% tube
and >5 years
smoke-​free
CR , complete response; DFS, disease-​free survival; ENE, clinical extranodal extension; ND, neck dissection; NR , not reported (can include immature data);
OS, overall survival; PFS, progression-​free survival; PR , partial response; TORS, transoral robotic surgery. aIncludes non-​peer-reviewed data from conference
presentations. bSingle-​arm trial. cRefers to NCCN guidelines, 7th edition25.

(Table 2). However, data were available for only 2 years (30 mg/m2), the 3-year OS was 95% with excellent QOL
of follow-​up monitoring and a substantial proportion of outcomes133.
the deaths were not caused by HNSCC, and thus the
efficacy end points were underpowered132. Of note, Transoral surgery
the De-​ESCALaTE HPV trial132 included a standard In response to the enthusiasm for transoral surgery
radiation scheme and three doses of high-​dose cispla- that has emerged, data from several ongoing prospec-
tin, while the RTOG 1016 trial127 involved the reduced tive trials investigating up-​front surgery followed by
cumulative cisplatin dose of 200 mg/m2, which had been de-​intensified adjuvant therapy should help to clarify
validated in the RTOG 0129 trial89 for use with acceler- the role of surgery. Early results from an innovative
ated radiation. These results confirm that cisplatin plus single-​institution phase II trial129 highlight the potential
radiotherapy remains the non-​surgical standard-​of-care of surgical de-​intensification: 80 patients underwent
treatment for unselected patients with HPV+ OPSCC. TORS and neck dissection followed by 30–36 Gy radio-
An alternative trial of de-​intensified standard chemo- therapy combined with docetaxel. Patients with clinical
radiotherapy, including definitive radiotherapy alone extra­nodal extension (ENE) received a boost to 36 Gy.
compared with chemoradiotherapy and reduction in At 2 years, DFS was 89% with no grade 3–4 toxicities. This
the dose of radiotherapy included in the chemoradio- small-​cohort trial highlights the potential to radically
therapy regimen (NCT02254278), is ongoing. Of note, reduce the toxicity of current treatments.
several single-​arm trials using reduced-​dose chemo- In the USA, the NCI sponsored the ECOG 3311 trial49,
radiotherapy have been reported with excellent out- a large-​cohort phase II trial of de-​intensified adjuvant
comes. For example, in a multicentre study involving therapy after transoral surgery in HPV+ OPSCC. In the
44 patients with T0–T3 N0–N2c (defined accord- UK, the Postoperative Adjuvant Treatment for HPV-​
ing to ref.25) OPSCC and a minimal smoking history positive Tumours (PATHOS) phase II/III trial134 used a
treated with 60 Gy radiotherapy and weekly cisplatin similar design. Both studies investigated the efficacy of

www.nature.com/nrclinonc
Reviews

Table 2 | Summary of data from clinical trials of anti-​PD-1 agents in head and neck squamous carcinoma
Study Phase Eligibility Patients Treatment arms OS ORR grade 3–4
(n) adverse events
First-​line therapy in refractory and/or metastatic disease
KEYNOTE-048 III – 882 Pembrolizumab (200 mg Pembrolizumab versus 17%, 36% 17%, 71%
(ref.150)a every 3 weeks) versus EXTREME: 14.9 months and 36% with and 69% with
pembrolizumab + versus 10.7 months pembrolizumab, pembrolizumab,
cisplatin or carboplatin + (P = 0.0007; patients pembrolizumab pembrolizumab
5-FU versus cetuximab + with CPS of ≥20) and + chemotherapy + chemotherapy
EXTREME 12.3 months versus and EXTREME, and EXTREME,
10.3 months (P = 0.0086; respectively respectively
patients with CPS of (overall
≥1); pembrolizumab + population)
chemotherapy versus
EXTREME: 13.0 months
versus 10.7 months
(P = 0.0034; overall
population)
Monotherapy in platinum-​refractory and/or metastatic disease
CheckMate 141 III – 361 Nivolumab versus 2-year OS: 17% versus 13% versus 6% 15% versus 37%
(refs160,162) investigators’ choice 6%
chemotherapy
KEYNOTE-012 Ibb PD-​L1 expression 60 Pembrolizumab (10 mg/kg Median OS: 8 months 18% 9%
(ref.138) >1% on tumour body weight every
and/or stromal 2 weeks)
cells by immuno-
histochemistry
KEYNOTE-012 Ibb – 132 Pembrolizumab (200 mg 6-month OS: 59% 18% 9%
(expansion every 3 weeks)
cohort)139
KEYNOTE-055 IIb Refractory to 171 Pembrolizumab (200 mg Median OS: 8 months 16% 15%
(ref.141) platinum and every 3 weeks)
cetuximab
KEYNOTE-040 III – 497 Pembrolizumab (200 mg Median OS: 8.4 months 15% versus 10% 13% versus 18%
(ref.142) every 3 weeks) versus versus 6.9 months
investigators’ choice (P = 0.02)
chemotherapy
Segal et al. I/IIb – 62 Durvalumab (10 mg/kg 12-month OS: 62% 11% 7%
(2015)143 body weight every
2 weeks)
HAWK163 IIb – 112 Durvalumab (10 mg/kg Immature data 14% 10%
body weight every
2 weeks)
Combination therapy in platinum-​refractory and/or metastatic disease
CONDOR145 II PD-​L1 expression 267 Durvalumab (20 mg/kg 7.6 months, 6.0 months 8%, 9% and 16%, 12%
<25% on tumour body weight every and 5.5 months 2% with and 17% with
cells by immuno- 4 weeks) + tremelimumab with combination, combination, combination,
histochemistry (1 mg/kg body weight durvalumab and durvalumab and durvalumab and
every 4 weeks) for tremelimumab, tremelimumab, tremelimumab,
four cycles followed respectively respectively respectively
by durvalumab
monotherapy
versus durvalumab
monotherapy versus
tremelimumab
monotherapy
5-FU, 5-fluorouracil; CPS, combined positive score; EXTREME, combination of cetuximab, 5-FU and cisplatin or carboplatin; ORR , overall response rate;
OS, overall survival; PD-1, programmed cell death 1; PD-​L1, programmed cell death 1 ligand 1. aIncludes non-​peer-reviewed data from conference presentation.
b
Single-​arm trial.

transoral surgical resection and neck dissection followed were randomly assigned to 50 Gy versus 60 Gy adjuvant
by pathological risk-​based adjuvant therapy in patients radiotherapy, and those with high-​risk disease (positive
with resectable HPV+ OPSCC with N0–N2b nodal surgical margins, five or more metastatic lymph nodes,
status. Patients with low-​risk disease (negative surgi- or >1 mm of ENE (in ECOG 3311) or any ENE (in
cal margins and no adverse pathology) did not receive PATHOS)) received adjuvant radiotherapy and cispla-
adjuvant therapy, those with intermediate-​risk disease tin (ECOG 3311) or were randomly assigned to adjuvant

Nature Reviews | Clinical Oncology


Reviews

chemoradiotherapy or radiotherapy (PATHOS). ECOG trial136 have been bolstered by evidence from an expan-
3311 has completed accrual of 519 patients and sion cohort, the phase II KEYNOTE-055 trial141, and
PATHOS has completed phase II accrual and is expected phase III data from the KEYNOTE-040 trial142. Of note,
to progress to phase III134. Furthermore, each trial used in the KEYNOTE-040 trial, 497 patients with platinum-​
a novel process to ensure consistent standards of sur- refractory and/or metastatic HNSCC were randomly
gery and patient safety49,134. The robust accrual outcomes assigned to receive pembrolizumab or standard-​of-care
and the existence of surgical quality assurance mecha- systemic therapy. Pembrolizumab resulted in improved
nisms provide proof of the concept that multicentre OS (8.4 months versus 6.9 months, HR 0.80; P = 0.016)
trials can be successfully completed with consistent and a significantly better toxicity profile142. Importantly,
surgical standards in the setting of head and neck surg­ the data for both nivolumab and pembrolizumab demon-
ical oncology. Additionally, in Ontario, Canada, the strate durable survival benefits in a substantial minority
Oropharynx: Radiotherapy Versus Trans-​Oral Robotic of patients137,138. Furthermore, patients receiving anti-​
Surgery (ORATOR) and the follow-​up ORATOR2 trials PD-1 agents reported fewer treatment-​related adverse
are single-​centre randomized trials directly comparing events142, and improved QOL outcomes in domains such
de-​escalated transoral surgery with de-​intensified adju- as swallowing, talking, eating and xero­stomia compared
vant therapy versus de-​intensified definitive chemo- with those receiving standard cytotoxic chemotherapy140.
radiotherapy (NCT01590355 and NCT03210103)135. A phase I/II trial138 showed an objective response rate of
These trials will provide a definitive comparison of 11% for durvalumab, an antibody against programmed
QOL outcomes of primary transoral surgery with those cell death 1 ligand 1 (PD-​L1), in the setting of advanced-​
of non-​surgical treatment. Unfortunately, these results stage disease, but this ICI has not yet been approved for
might not clearly favour any approach. While inter-​ use in patients with HNSCC143.
trial comparisons of standardized QOL measures can
be informative, no data from phase III studies directly Biomarkers and immunotherapy
comparing the major treatment paradigms are cur- Clinical response to immunomodulatory agents might
rently available. Owing to patient preferences, varia- be considerably improved with combination ther-
tions in the availability of expertise and other factors, apy in appropriately selected patients based on data
the field needs such comparisons to obtain level 1 evi- from other malignancies144, although the experience
dence for de-​intensification with different modalities, to with combinational therapy in HNSCC remains nas-
enable the better-​informed selection of surgical and/or cent145. Biomarkers might also enable the prediction
non-​surgical reduced-​dose therapies. of the principal immune phenotype (immune-​desert,
immune-​excluded or inflamed) for each patient146.
Immunotherapy The immune-​desert phenotype is characterized by a
The most remarkable therapeutic development in total absence of an immune response; in the immune-​
HNSCC over the past 10 years has been the advent of excluded phenotype, immune cells are unable to pene­-
immunotherapy. This therapeutic modality is based trate into the tumour microenvironment; and the
on the premise that alterations in immune surveil- inflamed phenotype is characterized by an active
lance, in cancer cell phenotype and/or in the tumour immune response within the tumour, but with the pres-
microenviron­ment are required for cancers to evade the ence of inhibitory factors that prevent a fully effective
immune system and manifest clinically. Indeed, antitumour immune response. High levels of PD-​L1
the immune system has been recognized to be key to the expression, typically only seen in the inflamed pheno­
development, establishment and spread of HNSCC136,137. type, are associated with an increased likelihood of
This biological rationale is supported by the results of response to anti-​PD-1 therapy. As noted, PD-​L1 is likely
large-​cohort clinical trials demonstrating consistent to be adopted as a predictive biomarker for the selection
efficacy of immune checkpoint inhibitors (ICIs). of patients with HNSCC for immunotherapy137.
PD-​L1 is expressed on 50–60% of HNSCCs147 and
Anti-​PD-1 is a key component of mechanisms of evasion of anti­
In 2016, the FDA approved the first anti-​PD-1 agents tumour immunity in HNSCC148. Data suggest that PD-1+
for the treatment of patients with HNSCC in the set- regulatory T cells infiltrate HPV+ HNSCCs more com-
ting of platinum-​refractory recurrent and/or metastatic monly than HPV− HNSCCs149. The PD-​L1 positivity of a
disease. The phase Ib trial KEYNOTE-012 (ref.138) and tumour can be measured with various scoring methods.
the expansion cohort of the same study139 showed dura- The tumour proportion score is a measure of the per-
ble responses to pembrolizumab (Table 2), leading to centage of PD-​L1-stained tumour cells142. The combined
its accelerated FDA approval. Soon after, nivolumab positive score (CPS) is a measure of the ratio of PD-​L1+
was approved in the same setting based on data from tumour and immune cells to the total number of tumour
the phase III CheckMate 141 trial140, which showed cells counted; the CPS has been shown to predict clinical
improved OS and QOL relative to the investigators’ outcomes150.
choice of standard-​of-care systemic therapy (Table 2). The importance of biomarker selection has been
In this randomized trial involving 361 patients, OS at shown in subgroup analyses from several trials. The
1 year was 36% in those treated with nivolumab com- survival benefit derived from anti-​PD-1 therapy is
pared with 16.6% in those treated with chemotherapy largely restricted to patients with ≥1% intratumoural
(HR 0.70, 95% CI 0.51–0.96)137. The phase Ib results PD-​L1 expression137,139: in the CheckMate 141 trial140, the
obtained with pembrolizumab in the KEYNOTE-012 median OS was 8.7 months with nivolumab treatment

www.nature.com/nrclinonc
Reviews

Paclitaxel inhibits
microtubule disaggregation
HNSCC
tumour cell
HNSCC tumour cell Pembrolizumab
overexpressing EGFR and nivolumab PD-1 MHC class I
block PD-1 and Antigen
Cetuximab PD-L1 interaction TCR
EGFR PD-L1 CTLA-4
Cisplatin inhibits B7
DNA replication and CD8+
mRNA transcription T cell

Ipilimumab DC
Cetuximab and EXTREME regimen
radiotherapy approved da Vinci robotic (cetuximab, 5-FU Pembrolizumab Data presented
in LRA HNSCC and as system approved for and cisplatin or and nivolumab showing superiority
monotherapy in transoral resection carboplatin) are approved in of pembrolizumab
platinum-refractory of oropharyngeal approved for first- platinum-refractory in first-line R/M
R/M HNSCC cancer line R/M HNSCC R/M HNSCC HNSCC

2006 2009 2011 2016 2018

Fig. 3 | Timeline showing the main therapeutic advances for patients with head and neck squamous carcinoma.
Cetuximab was the first major drug approved by the FDA for use in patients with head and neck squamous cell carcinoma
(HNSCC) since the approval of cisplatin in 1978. Cetuximab was approved in 2006 for use in patients with recurrent and/or
metastatic (R/M) HNSCC and as a radiosensitizer in patients with locally advanced HNSCC. In 2011, the EXTREME
regimen was also approved for use in patients with R/M HNSCC. The FDA had approved the da Vinci robotic system for
the resection of T1–T2 oropharyngeal cancer 2 years earlier. In 2016, both nivolumab and pembrolizumab were approved
by the FDA for use in patients with platinum-​refractory R/M HNSCC. In 2018, data were presented showing the superiority
of pembrolizumab over cisplatin as a first-​line treatment for R/M HNSCC; the FDA is currently reviewing these data. 5-FU,
5-fluorouracil; CTL A-4, cytotoxic T lymphocyte antigen 4; DC, dendritic cell; LRA , locoregionally advanced; PD-1,
programmed cell death 1; PD-​L1, programmed cell death 1 ligand 1; TCR , T cell receptor.

and 4.6 months with chemotherapy in the subgroup with respectively). New strategies to trigger an antitumour
≥1% intratumoural PD-​L1 expression but 5.7 months in immune response in patients with immune-​excluded
the subgroup with <1% intratumoural PD-​L1 expression, phenotypes are needed.
regardless of treatment.
Anti-​PD-1 therapy may soon be approved as a first-​ Combination with radiotherapy
line therapy in patients with recurrent and/or metastatic ICIs are rapidly being investigated in trials in the cura-
HNSCC. The KEYNOTE-048 trial (NCT02358031)150, a tive setting, and in various combinations with other
phase III randomized trial involving 825 patients, com- treatment modalities. Radiotherapy has complex effects
pared the efficacy of pembrolizumab monotherapy or of on malignant and non-​malignant tissues and might both
a novel combination of pembrolizumab, 5-fluorouracil enhance and suppress an antitumour immune response.
and cisplatin or carboplatin versus the EXTREME regi- Under certain conditions, radiotherapy might amplify
men (cetuximab, 5-fluorouracil and cisplatin or carbo­ the effect of immunotherapy by promoting the release
platin) in the first-​line setting for recurrent and/or of cytokines and tumour-​associated antigens. In the
metastatic HNSCC. Interim data presented in 2018 PACIFIC trial, durvalumab after definitive chemoradio-
(ref.146) indicate that pembrolizumab monotherapy signi­ therapy in patients with stage III non-​small-cell lung car-
ficantly improved OS compared with the EXTREME reg- cinoma improved 2-year OS compared with definitive
imen in patients with a PD-​L1 CPS of ≥20 or CPS of >1, chemoradiotherapy alone (66.3% versus 55.6%; HR 0.68,
and the addition of pembrolizumab to chemotherapy CI 0.47–0.99)153. Multiple phase I/II trials in the setting
significantly improved OS over the EXTREME regimen of locoregionally advanced HNSCC are investigating a
in unselected patients150 (Table 2). similar strategy, adding anti-​PD-1 antibodies to defini-
In other solid tumours, tumour mutational load tive chemoradiotherapy (NCT02252042, NCT02586207,
has been strongly associated with a response to ICIs151. NCT02819752, NCT02707588 and NCT02289209).
HNSCCs have a moderate-​to-high mutational load The published data currently available on the poten-
(the ninth-​highest average mutational load among 30 tial abscopal effect derived from the combination of
different tumour types studied)152 and harbour other immunotherapy and radiotherapy in HNSCC are lim-
genomic alterations (such as frameshifts)116 that might ited and emphasize the complex interactions between
also be associated with response to ICIs, although this radiotherapy and the immune system. In a randomized
hypothesis has not been directly tested. HPV status phase II study presented at the 2018 ASCO Annual
is another potential biomarker, with data from the Meeting154 in the setting of refractory and/or metastatic
CheckMate 141 trial133 indicating that HPV+ HNSCCs HNSCC, the addition of stereotactic body radiotherapy
are more likely to respond to anti-​P D-1 therapy to nivolumab failed to improve survival compared
than HPV− HNSCCs (HR for death of 0.56 and 0.73, with nivolumab alone.

Nature Reviews | Clinical Oncology


Reviews

Combination immunotherapy Other approaches, involving HPV peptide vaccines or


Unfortunately, the majority of patients with HNSCC co-​stimulatory agonists (such as Toll-​like receptor, OX40,
do not respond to single-​agent anti-​PD-1 therapy. In CD40L or CD137), are also being tested (NCT03906526,
an effort to improve response rates, ongoing trials are NCT02315066 and NCT03792724) 158 . In the
evaluating combinations involving ICIs, therapeutic Active8 study, 195 patients were randomly assigned (1:1)
vaccines, co-​stimulatory agonists and cytotoxic agents. to receive the EXTREME regimen with or without the
Combinations of anti-​cytotoxic T lymphocyte antigen 4 Toll-​like receptor 8 agonist motolimod159. No signi­ficant
(CTLA-4) and anti-​PD-1 agents have a synergistic benefit was derived from motolimod, but, in a prespeci­
effect in patients with melanoma144 and are being tested fied subgroup analysis, the addition of motolimod
in phase III studies involving patients with HNSCC improved PFS (7.8 months versus 5.9 months; P = 0.046)
(NCT02551159 and NCT027451570). In the phase II and OS (15.2 months versus 12.6 months; P = 0.03) in
CONDOR study, in which the anti-​CTLA-4 antibody patients with HPV+ HNSCC159.
tremelimumab plus the anti-​PD-L1 antibody dur-
valumab was investigated in patients with refractory Conclusions
and/or metastatic HNSCC and low or no (<25%) PD-​ The diversity of the therapeutic advances in HNSCC dis-
L1 tumour cell expression, overall response rates of 8% cussed in this Review is remarkable. These modalities
and 9% were observed with the combination and dur- illustrate important principles that have the potential to
valumab, respectively; a follow-​up phase III trial has dramatically alter treatment paradigms, improving both
completed accrual (NCT02369874)145. the oncological and functional outcomes of patients with
Multiple therapeutic vaccines are also under inves- HNSCC (Fig. 3). Technological advances in surgery and
tigation in phase I/II trials involving patients with radiotherapy have provided patients with multiple treat-
HNSCC. In a phase I study involving 16 patients, an ment options and have progressively decreased the risks
adjuvant peptide-​loaded dendritic cell-​based vaccine of morbidity that were previously associated with these
against p53 was well tolerated and the 2-year DFS was therapeutic modalities. De-​intensification of treatment
88%155. Another vaccine strategy involves talimogene for patients with a favourable prognosis might also have
laherparepvec, an oncolytic herpes simplex virus 1 a major effect on QOL after treatment. Furthermore,
vaccine that promotes the release of granulocyte– the advent of immunotherapy offers the potential for
macrophage colony-​stimulating factor that was FDA-​ long-​term remissions in the setting of refractory and/or
approved for use in patients with recurrent melanoma metastatic disease, for which a limited number of treat-
in 2015. In a phase I/II study in the setting of loco­ ment options was previously available. In the near future,
regionally advanced HNSCC, treatment with talimo- immunotherapy might be used to enhance the efficacy
gene laherparepvec in combination with cisplatin and of conventional treatments in the curative setting. To be
radio­therapy resulted in an OS of 70.5% at 29 months156. fully realized, these advances require continued, careful
A phase II study of a combination of nivolumab and evaluation. However, for the first time in decades, the
a synthetic long-​p eptide HPV-16 vaccine showed culmination of these technological advances offers
promising results with an overall response rate of the prospect of positively affecting the outcomes of
33% and a median OS of 17.5 months, both of which patients with HNSCC.
are higher than historically observed with anti-​PD-1
therapy alone157. Published online xx xx xxxx

1. Bray, F. et al. Global cancer statistics 2018: 10. Ang, K. K. et al. Human papillomavirus and survival of oropharyngeal “pap-​test equivalent” in high-​risk
GLOBOCAN estimates of incidence and mortality patients with oropharyngeal cancer. N. Engl. J. Med. populations. Cancer Prev. Res. (Phila) 4, 1378–1384
worldwide for 36 cancers in 185 countries. CA Cancer 363, 24–35 (2010). (2011).
J. Clin. 68, 394–424 (2018). 11. D’Souza, G. et al. Case-​control study of human 19. Harper, D. M. et al. Sustained efficacy up to 4.5 years
2. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, papillomavirus and oropharyngeal cancer. N. Engl. of a bivalent L1 virus-​like particle vaccine against
2017. CA Cancer J. Clin. 67, 7–30 (2017). J. Med. 356, 1944–1956 (2007). human papillomavirus types 16 and 18: follow-​up
3. Wyss, A. et al. Cigarette, cigar, and pipe smoking 12. Gillison, M. L. et al. Epidemiology of human from a randomised control trial. Lancet 367,
and the risk of head and neck cancers: pooled analysis papillomavirus-​positive head and neck squamous cell 1247–1255 (2006).
in the International Head and Neck Cancer carcinoma. J. Clin. Oncol. 33, 3235–3242 (2015). 20. Food and Drug Administration. FDA approves
Epidemiology Consortium. Am. J. Epidemiol. 178, 13. Mehanna, H. et al. Prevalence of human papillomavirus expanded use of Gardasil 9 to include individuals 27
679–690 (2013). in oropharyngeal and nonoropharyngeal head and through 45 years old. FDA https://www.fda.gov/news-​
4. Maier, H. et al. Tobacco and alcohol and the risk of neck cancer — systematic review and meta-​analysis of events/press-​announcements/fda-​approves-expanded-​
head and neck cancer. Clin. Investig. 70, 320–327 trends by time and region. Head Neck 35, 747–755 use-gardasil-9-include-​individuals-27-through-
(1992). (2013). 45-years-​old (2018).
5. Blot, W. J. et al. Smoking and drinking in relation to 14. Carlander, A. F. et al. Continuing rise in oropharyngeal 21. Chaturvedi, A. K. et al. Effect of prophylactic human
oral and pharyngeal cancer. Cancer Res. 48, cancer in a high HPV prevalence area: a Danish papillomavirus (HPV) vaccination on oral HPV
3282–3287 (1988). population-​based study from 2011 to 2014. infections among young adults in the United States.
6. Chaturvedi, A. K. et al. Human papillomavirus and Eur. J. Cancer 70, 75–82 (2017). J. Clin. Oncol. 36, 262–267 (2018).
rising oropharyngeal cancer incidence in the United 15. Ribeiro, K. B. et al. Low human papillomavirus 22. Herrero, R. et al. Reduced prevalence of oral human
States. J. Clin. Oncol. 29, 4294–4301 (2011). prevalence in head and neck cancer: results from two papillomavirus (HPV) 4 years after bivalent HPV
7. World Health Organization. IARC Monographs on the large case-​control studies in high-​incidence regions. vaccination in a randomized clinical trial in Costa Rica.
Evaluation of Carcinogenic Risks to Humans, in Int. J. Epidemiol. 40, 489–502 (2011). PLOS ONE 8, e68329 (2013).
Human Papillomaviruses (WHO, 2007). 16. Chung, C. H. et al. p16 protein expression and human 23. Huang, S. H. et al. Refining American Joint Committee
8. Sturgis, E. M. & Cinciripini, P. M. Trends in head and papillomavirus status as prognostic biomarkers of on Cancer/Union for International Cancer Control
neck cancer incidence in relation to smoking nonoropharyngeal head and neck squamous cell TNM stage and prognostic groups for human
prevalence: an emerging epidemic of human carcinoma. J. Clin. Oncol. 32, 3930–3938 (2014). papillomavirus-​related oropharyngeal carcinomas.
papillomavirus-​associated cancers? Cancer 110, 17. Li, H. et al. Association of human papillomavirus J. Clin. Oncol. 33, 836–845 (2015).
1429–1435 (2007). status at head and neck carcinoma subsites with 24. O’Sullivan, B. et al. Development and validation of a
9. Gillison, M. L. et al. Evidence for a causal association overall survival. JAMA Otolaryngol. Head Neck Surg. staging system for HPV-​related oropharyngeal cancer
between human papillomavirus and a subset of head 144, 519–525 (2018). by the International Collaboration on Oropharyngeal
and neck cancers. J. Natl Cancer Inst. 92, 709–720 18. Fakhry, C. et al. Associations between oral HPV16 cancer Network for Staging (ICON-​S): a multicentre
(2000). infection and cytopathology: evaluation of an cohort study. Lancet Oncol. 17, 440–451 (2016).

www.nature.com/nrclinonc
Reviews

25. American Joint Committee on Cancer. AJCC Cancer robotic surgery trial. Laryngoscope 120, 1749–1755 71. Mehanna, H. et al. PET-​CT surveillance versus neck
Staging Manual 7th edn (eds Edge, S. et al.) (Springer, (2010). dissection in advanced head and neck cancer. N. Engl.
2010). 49. Holsinger, F. C. & Ferris, R. L. Transoral endoscopic J. Med. 374, 1444–1454 (2016).
26. American Joint Committee on Cancer. AJCC Cancer head and neck surgery and its role within the 72. Stack, B. C. et al. The negative predictive value (NPV)
Staging Manual 8th edn (eds Amin, M. B. et al.) multidisciplinary treatment paradigm of oropharynx of FDG-​PET/CT in the head and neck squamous cell
(Springer, 2017). cancer: robotics, lasers, and clinical trials. J. Clin. carcinoma (HNSCC) N0 patient, the first report of the
27. Cramer, J. D. et al. Validation of the eighth edition Oncol. 33, 3285–3292 (2015). ACRIN 6685 trial. J. Clin. Oncol. 35, 6041–6041
American Joint Committee on Cancer staging 50. Albergotti, W. G. et al. A prospective evaluation of (2017).
system for human papillomavirus-​associated short-​term dysphagia after transoral robotic surgery 73. Lowe, V. J. et al. Multicenter trial of [18F]
oropharyngeal cancer. Head Neck 40, 457–466 for squamous cell carcinoma of the oropharynx. fluorodeoxyglucose positron emission tomography/
(2017). Cancer 123, 3132–3140 (2017). computed tomography staging of head and neck
28. Fakhry, C. et al. Development and validation of 51. Sharma, A. et al. Survival and gastrostomy prevalence cancer and negative predictive value and surgical
nomograms predictive of overall and progression-​free in patients with oropharyngeal cancer treated with impact in the N0 neck: results from ACRIN 6685.
survival in patients with oropharyngeal cancer. J. Clin. transoral robotic surgery versus chemoradiotherapy. J. Clin. Oncol. https://doi.org/10.1200/JCO.18.01182
Oncol. 35, 4057–4065 (2017). JAMA Otolaryngol. Head Neck Surg. 142, 691–697 (2019).
29. Gleber-​Netto, F. O. et al. Variations in HPV function (2016). 74. Lacas, B. et al. Role of radiotherapy fractionation in
are associated with survival in squamous cell 52. Kelly, J. R. et al. Upfront surgery versus definitive head and neck cancers (MARCH): an updated meta-​
carcinoma. JCI Insight 4, 124762 (2019). chemoradiotherapy in patients with human analysis. Lancet Oncol. 18, 1221–1237 (2017).
30. Tamura, Y. et al. Therapeutic outcomes of laryngeal papillomavirus-​associated oropharyngeal squamous 75. Nutting, C. M. et al. Parotid-​sparing intensity
cancer at Kyoto University Hospital for 10 years. Acta cell cancer. Oral Oncol. 79, 64–70 (2018). modulated versus conventional radiotherapy in head
Otolaryngol. Suppl. 127, 62–65 (2007). 53. Grau, C. et al. Cervical lymph node metastases from and neck cancer (PARSPORT): a phase 3 multicentre
31. Cognetti, D. M., Weber, R. S. & Lai, S. Y. Head and unknown primary tumours. Results from a national randomised controlled trial. Lancet Oncol. 12,
neck cancer: an evolving treatment paradigm. Cancer survey by the Danish Society for Head and Neck 127–136 (2011).
113, 1911–1932 (2008). Oncology. Radiother. Oncol. 55, 121–129 (2000). 76. Kam, M. K. et al. Prospective randomized study of
32. Gatta, G. et al. Prognoses and improvement for 54. Galloway, T. J. & Ridge, J. A. Management of intensity-​modulated radiotherapy on salivary gland
head and neck cancers diagnosed in Europe in early squamous cancer metastatic to cervical nodes with an function in early-​stage nasopharyngeal carcinoma
2000s: the EUROCARE-5 population-​based study. unknown primary site. J. Clin. Oncol. 33, 3328–3337 patients. J. Clin. Oncol. 25, 4873–4879 (2007).
Eur. J. Cancer 51, 2130–2143 (2015). (2015). 77. Pow, E. H. et al. Xerostomia and quality of life after
33. Blanchard, P. et al. Meta-​analysis of chemotherapy in 55. Kim, S. H. et al. HPV integration begins in the tonsillar intensity-​modulated radiotherapy versus conventional
head and neck cancer (MACH-​NC): a comprehensive crypt and leads to the alteration of p16, EGFR and radiotherapy for early-​stage nasopharyngeal
analysis by tumour site. Radiother. Oncol. 100, c-​myc during tumor formation. Int. J. Cancer 120, carcinoma: initial report on a randomized controlled
33–40 (2011). 1418–1425 (2007). clinical trial. Int. J. Radiat. Oncol. Biol. Phys. 66,
34. Machtay, M. et al. Factors associated with severe late 56. Fakhry, C. et al. Human papillomavirus testing in head 981–991 (2006).
toxicity after concurrent chemoradiation for locally and neck carcinomas: ASCO Clinical Practice Guideline 78. Peng, G. et al. A prospective, randomized study
advanced head and neck cancer: an RTOG analysis. Endorsement of the College of American Pathologists comparing outcomes and toxicities of intensity-​
J. Clin. Oncol. 26, 3582–3589 (2008). Guideline. J. Clin. Oncol. 36, 3152–3161 (2018). modulated radiotherapy versus conventional two-​
35. Chinn, S. B. & Myers, J. N. Oral cavity carcinoma: 57. Graboyes, E. M. et al. Management of human dimensional radiotherapy for the treatment of
current management, controversies, and future papillomavirus-​related unknown primaries of the head nasopharyngeal carcinoma. Radiother. Oncol. 104,
directions. J. Clin. Oncol. 33, 3269–3276 (2015). and neck with a transoral surgical approach. Head 286–293 (2012).
36. National Comprehensive Cancer Network. NCCN Neck 37, 1603–1611 (2015). 79. Gregoire, V., Langendijk, J. A. & Nuyts, S. Advances in
clinical practice guidelines in oncology: head and neck 58. Durmus, K. et al. Transoral robotic approach to radiotherapy for head and neck cancer. J. Clin. Oncol.
cancers. NCCN https://oncolife.com.ua/doc/nccn/Head_ carcinoma of unknown primary. Head Neck 36, 33, 3277–3284 (2015).
and_Neck_Cancers.pdf (2018). 848–852 (2014). 80. Castadot, P. et al. Assessment by a deformable
37. Hanasono, M. M. et al. Impact of reconstructive 59. Geltzeiler, M. et al. Transoral robotic surgery for registration method of the volumetric and positional
microsurgery in patients with advanced oral cavity management of cervical unknown primary squamous changes of target volumes and organs at risk in
cancers. Head Neck 31, 1289–1296 (2009). cell carcinoma: updates on efficacy, surgical technique pharyngo-​laryngeal tumors treated with concomitant
38. Calais, G. et al. Randomized trial of radiation therapy and margin status. Oral Oncol. 66, 9–13 (2017). chemo-​radiation. Radiother. Oncol. 95, 209–217
versus concomitant chemotherapy and radiation 60. Hatten, K. M. et al. Transoral robotic surgery-​assisted (2010).
therapy for advanced-​stage oropharynx carcinoma. endoscopy with primary site detection and treatment 81. Schwartz, D. L. et al. Adaptive radiotherapy for head-​
J. Natl Cancer Inst. 91, 2081–2086 (1999). in occult mucosal primaries. JAMA Otolaryngol. Head and-neck cancer: initial clinical outcomes from a
39. Haigentz, M. et al. Current trends in initial Neck Surg. 143, 267–273 (2017). prospective trial. Int. J. Radiat. Oncol. Biol. Phys. 83,
management of oropharyngeal cancer: the declining 61. Patel, S. A. et al. Robotic surgery for primary head 986–993 (2012).
use of open surgery. Eur. Arch. Otorhinolaryngol. 266, and neck squamous cell carcinoma of unknown site. 82. Kim, J. K. et al. Proton therapy for head and neck
1845–1855 (2009). JAMA Otolaryngol. Head Neck Surg. 139, cancer. Curr. Treat. Options Oncol. 19, 28 (2018).
40. White, H. et al. Salvage surgery for recurrent cancers 1203–1211 (2013). 83. Setton, J. et al. A multi-​institution pooled analysis of
of the oropharynx: comparing TORS with standard 62. Weiss, M. H., Harrison, L. B. & Isaacs, R. S. Use of gastrostomy tube dependence in patients with
open surgical approaches. JAMA Otolaryngol. Head decision analysis in planning a management strategy oropharyngeal cancer treated with definitive intensity-​
Neck Surg. 139, 773–778 (2013). for the stage N0 neck. Arch. Otolaryngol. Head Neck modulated radiotherapy. Cancer 121, 294–301
41. O’Malley, B. W. et al. Transoral robotic surgery (TORS) Surg. 120, 699–702 (1994). (2015).
for base of tongue neoplasms. Laryngoscope 116, 63. D’Cruz, A. K. et al. Elective versus therapeutic neck 84. Blanchard, P. et al. Intensity-​modulated proton
1465–1472 (2006). dissection in node-​negative oral cancer. N. Engl. beam therapy (IMPT) versus intensity-​modulated
42. de Almeida, J. R. et al. Oncologic outcomes after J. Med. 373, 521–529 (2015). photon therapy (IMRT) for patients with oropharynx
transoral robotic surgery: a multi-​institutional study. 64. Brennan, S. et al. Prospective trial to evaluate staged cancer — a case matched analysis. Radiother. Oncol.
JAMA Otolaryngol. Head Neck Surg. 141, neck dissection or elective neck radiotherapy in 120, 48–55 (2016).
1043–1051 (2015). patients with CT-​staged T1-2 N0 squamous cell 85. Vlacich, G. et al. Dose to the inferior pharyngeal
43. Haughey, B. H. et al. Transoral laser microsurgery as carcinoma of the oral tongue. Head Neck 32, constrictor predicts prolonged gastrostomy tube
primary treatment for advanced-​stage oropharyngeal 191–198 (2010). dependence with concurrent intensity-​modulated
cancer: a United States multicenter study. Head Neck 65. Schiefke, F. et al. Function, postoperative morbidity, radiation therapy and chemotherapy for locally-​
33, 1683–1694 (2011). and quality of life after cervical sentinel node advanced head and neck cancer. Radiother. Oncol.
44. US Food and Drug Administration. 501(k) summary, biopsy and after selective neck dissection. Head Neck 110, 435–440 (2014).
section III: indications for use for intuitive surgical 31, 503–512 (2009). 86. Feng, F. Y. et al. Intensity-​modulated
endoscopic instrument control system for transoral 66. Murer, K. et al. Comparison of morbidity between chemoradiotherapy aiming to reduce dysphagia in
otolaryngology procedures. FDA https://www. sentinel node biopsy and elective neck dissection for patients with oropharyngeal cancer: clinical and
accessdata.fda.gov/cdrh_docs/pdf9/K090993.pdf treatment of the N0 neck in patients with oral functional results. J. Clin. Oncol. 28, 2732–2738
(2009). squamous cell carcinoma. Head Neck 33, (2010).
45. Liederbach, E. et al. A contemporary analysis of 1260–1264 (2011). 87. Pignon, J. P. et al. Meta-​analysis of chemotherapy in
surgical trends in the treatment of squamous cell 67. Alkureishi, L. W. et al. Sentinel node biopsy in head head and neck cancer (MACH-​NC): an update on 93
carcinoma of the oropharynx from 1998 to 2012: and neck squamous cell cancer: 5-year follow-​up of a randomised trials and 17,346 patients. Radiother.
a report from the National Cancer Database. European multicenter trial. Ann. Surg. Oncol. 17, Oncol. 92, 4–14 (2009).
Ann. Surg. Oncol. 22, 4422–4431 (2015). 2459–2464 (2010). 88. Tsan, D. L. et al. The comparison between weekly and
46. US Food and Drug Administration. 510k summary 68. Civantos, F. J. et al. Sentinel lymph node biopsy three-​weekly cisplatin delivered concurrently with
medrobotics flex system (K150776). FDA https://www. accurately stages the regional lymph nodes for T1-T2 radiotherapy for patients with postoperative high-​risk
accessdata.fda.gov/cdrh_docs/pdf15/K150776.pdf oral squamous cell carcinomas: results of a squamous cell carcinoma of the oral cavity. Radiat.
(2015). prospective multi-​institutional trial. J. Clin. Oncol. 28, Oncol. 7, 215 (2012).
47. Weinstein, G. S. et al. Transoral robotic surgery: 1395–1400 (2010). 89. Nguyen-​Tan, P. F. et al. Randomized phase III trial to
a multicenter study to assess feasibility, safety, and 69. Schilling, C. et al. Sentinel European Node Trial test accelerated versus standard fractionation in
surgical margins. Laryngoscope 122, 1701–1707 (SENT): 3-year results of sentinel node biopsy in oral combination with concurrent cisplatin for head and
(2012). cancer. Eur. J. Cancer 51, 2777–2784 (2015). neck carcinomas in the Radiation Therapy Oncology
48. Weinstein, G. S. et al. Selective neck dissection and 70. Thompson, C. F. et al. Diagnostic value of sentinel Group 0129 trial: long-​term report of efficacy and
deintensified postoperative radiation and lymph node biopsy in head and neck cancer: a meta-​ toxicity. J. Clin. Oncol. 32, 3858–3866 (2014).
chemotherapy for oropharyngeal cancer: a subset analysis. Eur. Arch. Otorhinolaryngol. 270, 90. Szturz, P. et al. Weekly low-​dose versus three-​weekly
analysis of the University of Pennsylvania transoral 2115–2122 (2013). high-​dose cisplatin for concurrent chemoradiation in

Nature Reviews | Clinical Oncology


Reviews

locoregionally advanced non-​nasopharyngeal head 110. Vermorken, J. B. et al. Cisplatin and fluorouracil with 130. Seiwert, T. Y. et al. OPTIMA: a phase II dose and
and neck cancer: a systematic review and meta-​ or without panitumumab in patients with recurrent or volume de-​escalation trial for human papillomavirus-​
analysis of aggregate data. Oncologist 22, metastatic squamous-​cell carcinoma of the head and positive oropharyngeal cancer. Ann. Oncol. 30,
1056–1066 (2017). neck (SPECTRUM): an open-​label phase 3 randomised 297–302 (2019).
91. Uygun, K. et al. The comparison of weekly and three-​ trial. Lancet Oncol. 14, 697–710 (2013). 131. Hegde, J. V. et al. Patient-​reported quality-​of-life
weekly cisplatin chemotherapy concurrent with 111. Machiels, J. P. et al. Afatinib versus methotrexate as outcomes after de-​escalated chemoradiation for
radiotherapy in patients with previously untreated second-​line treatment in patients with recurrent or human papillomavirus-​positive oropharyngeal
inoperable non-​metastatic squamous cell carcinoma of metastatic squamous-​cell carcinoma of the head and carcinoma: findings from a phase 2 trial. Cancer 124,
the head and neck. Cancer Chemother. Pharmacol. neck progressing on or after platinum-​based therapy 521–529 (2018).
64, 601–605 (2009). (LUX-​Head & Neck 1): an open-​label, randomised 132. Mehanna, H. et al. Radiotherapy plus cisplatin or
92. Quon, H. et al. Phase III study of radiation therapy phase 3 trial. Lancet Oncol. 16, 583–594 (2015). cetuximab in low-​risk human papillomavirus-​positive
with or without cis-​platinum in patients with 112. Machiels, J. P. et al. Zalutumumab plus best oropharyngeal cancer (De-​ESCALaTE HPV): an open-​
unresectable squamous or undifferentiated carcinoma supportive care versus best supportive care alone in label randomised controlled phase 3 trial. Lancet
of the head and neck: an intergroup trial of the patients with recurrent or metastatic squamous-​cell 393, P51–P60 (2018).
Eastern Cooperative Oncology Group (E2382). carcinoma of the head and neck after failure of 133. Chera, B. S. et al. Mature results of a prospective
Int. J. Radiat. Oncol. Biol. Phys. 81, 719–725 (2011). platinum-​based chemotherapy: an open-​label, study of deintensified chemoradiotherapy for low-​risk
93. Noronha, V. et al. Once-​a-week versus once-​every-3- randomised phase 3 trial. Lancet Oncol. 12, human papillomavirus-​associated oropharyngeal
weeks cisplatin chemoradiation for locally advanced 333–343 (2011). squamous cell carcinoma. Cancer 124, 2347–2354
head and neck cancer: a phase III randomized 113. Patil, V. M. et al. Results of a randomized phase III (2018).
noninferiority trial. J. Clin. Oncol. 36, 1064–1072 study of nimotuzumab in combination with concurrent 134. Evans, M. et al. PATHOS: a phase II/III trial of risk-​
(2018). radiotherapy and cisplatin versus radiotherapy and stratified, reduced intensity adjuvant treatment in
94. Bauml, J. M. et al. Cisplatin every 3 weeks versus cisplatin alone, in locally advanced squamous cell patients undergoing transoral surgery for human
weekly with definitive concurrent radiotherapy for carcinoma of the head and neck [abstract]. J. Clin. papillomavirus (HPV)-positive oropharyngeal cancer
squamous cell carcinoma of the head and neck. J. Natl Oncol. 36 (Suppl. 15), 6000 (2018). [abstract]. J. Clin. Oncol. 36 (Suppl. 15), TPS6097
Cancer Inst. 111, 490–497 (2019). 114. Harari, P. M. et al. Postoperative chemoradiotherapy (2018).
95. Kunieda, F. et al. Randomized phase II/III trial of post-​ and cetuximab for high-​risk squamous cell carcinoma 135. Nichols, A. C. et al. Early-​stage squamous cell
operative chemoradiotherapy comparing 3-weekly of the head and neck: Radiation Therapy Oncology carcinoma of the oropharynx: radiotherapy versus
cisplatin with weekly cisplatin in high-​risk patients Group RTOG-0234. J. Clin. Oncol. 32, 2486–2495 trans-​oral robotic surgery (ORATOR) — study protocol
with squamous cell carcinoma of head and neck: (2014). for a randomized phase II trial. BMC Cancer 13, 133
Japan Clinical Oncology Group Study (JCOG1008). 115. Chung, C. H. et al. Molecular classification of head and (2013).
Jpn J. Clin. Oncol. 44, 770–774 (2014). neck squamous cell carcinomas using patterns of gene 136. Ferris, R. L. Immunology and immunotherapy of head
96. Quon, H. et al. Radiation therapy for oropharyngeal expression. Cancer Cell 5, 489–500 (2004). and neck cancer. J. Clin. Oncol. 33, 3293–3304
squamous cell carcinoma: American Society of Clinical 116. Cancer Genome Atlas Network. Comprehensive (2015).
Oncology Endorsement of the American Society for genomic characterization of head and neck squamous 137. Ferris, R. L. et al. Nivolumab for recurrent squamous-​
Radiation Oncology evidence-​based clinical practice cell carcinomas. Nature 517, 576–582 (2015). cell carcinoma of the head and neck. N. Engl. J. Med.
guideline. J. Clin. Oncol. 35, 4078–4090 (2017). 117. Agrawal, N. et al. Exome sequencing of head and neck 375, 1856–1867 (2016).
97. Blanchard, P. et al. Taxane-​cisplatin-fluorouracil as squamous cell carcinoma reveals inactivating 138. Seiwert, T. Y. et al. Safety and clinical activity of
induction chemotherapy in locally advanced head and mutations in NOTCH1. Science 333, 1154–1157 pembrolizumab for treatment of recurrent or
neck cancers: an individual patient data meta-​analysis (2011). metastatic squamous cell carcinoma of the head
of the Meta-​Analysis of Chemotherapy in Head and 118. Keck, M. K. et al. Integrative analysis of head and neck and neck (KEYNOTE-012): an open-​label,
Neck Cancer Group. J. Clin. Oncol. 31, 2854–2860 cancer identifies two biologically distinct HPV and multicentre, phase 1b trial. Lancet Oncol. 17,
(2013). three non-​HPV subtypes. Clin. Cancer Res. 21, 956–965 (2016).
98. Posner, M. R. et al. Cisplatin and fluorouracil alone 870–881 (2015). 139. Chow, L. Q. M. et al. Antitumor activity of
or with docetaxel in head and neck cancer. N. Engl. 119. Stransky, N. et al. The mutational landscape of head pembrolizumab in biomarker-​unselected patients with
J. Med. 357, 1705–1715 (2007). and neck squamous cell carcinoma. Science 333, recurrent and/or metastatic head and neck squamous
99. Vermorken, J. B. et al. Cisplatin, fluorouracil, and 1157–1160 (2011). cell carcinoma: results from the phase Ib
docetaxel in unresectable head and neck cancer. 120. Soulieres, D. et al. Buparlisib and paclitaxel in patients KEYNOTE-012 expansion cohort. J. Clin. Oncol. 34,
N. Engl. J. Med. 357, 1695–1704 (2007). with platinum-​pretreated recurrent or metastatic 3838–3845 (2016).
100. Haddad, R. et al. Induction chemotherapy followed squamous cell carcinoma of the head and neck (BERIL-1): 140. Harrington, K. J. et al. Nivolumab versus standard,
by concurrent chemoradiotherapy (sequential a randomised, double-​blind, placebo-​controlled phase single-​agent therapy of investigator’s choice in
chemoradiotherapy) versus concurrent 2 trial. Lancet Oncol. 18, 323–335 (2017). recurrent or metastatic squamous cell carcinoma
chemoradiotherapy alone in locally advanced head 121. Beck, T. N. et al. Phospho-​T356RB1 predicts survival of the head and neck (CheckMate 141): health-​
and neck cancer (PARADIGM): a randomised phase 3 in HPV-​negative squamous cell carcinoma of the head related quality-​of-life results from a randomised,
trial. Lancet Oncol. 14, 257–264 (2013). and neck. Oncotarget 6, 18863–18874 (2015). phase 3 trial. Lancet Oncol. 18, 1104–1115
101. Cohen, E. E. et al. Phase III randomized trial of 122. Michel, L. et al. Phase I trial of palbociclib, a selective (2017).
induction chemotherapy in patients with N2 or N3 cyclin dependent kinase 4/6 inhibitor, in combination 141. Bauml, J. et al. Pembrolizumab for platinum- and
locally advanced head and neck cancer. J. Clin. Oncol. with cetuximab in patients with recurrent/metastatic cetuximab-​refractory head and neck cancer: results
32, 2735–2743 (2014). head and neck squamous cell carcinoma. Oral Oncol. from a single-​arm, phase II study. J. Clin. Oncol. 35,
102. Geoffrois, L. et al. Induction chemotherapy followed by 58, 41–48 (2016). 1542–1549 (2017).
cetuximab radiotherapy is not superior to concurrent 123. Maxwell, J. H. et al. Tobacco use in human 142. Cohen, E. E. W. et al. Pembrolizumab versus
chemoradiotherapy for head and neck carcinomas: papillomavirus-​positive advanced oropharynx cancer methotrexate, docetaxel, or cetuximab for
results of the GORTEC 2007-02 phase III randomized patients related to increased risk of distant recurrent or metastatic head-​and-neck squamous
trial. J. Clin. Oncol. 36, 3077–3083 (2018). metastases and tumor recurrence. Clin. Cancer Res. cell carcinoma (KEYNOTE-040): a randomised, open-​
103. Kalyankrishna, S. & Grandis, J. R. Epidermal 16, 1226–1235 (2010). label, phase 3 study. Lancet 393, P156–P167
growth factor receptor biology in head and neck 124. Gillison, M. L. et al. Tobacco smoking and increased (2019).
cancer. J. Clin. Oncol. 24, 2666–2672 (2006). risk of death and progression for patients with p16- 143. Segal, N. S. et al. Safety and efficacy of MEDI4736,
104. Bossi, P. et al. Prognostic and predictive value of EGFR positive and p16-negative oropharyngeal cancer. an anti-​PD-L1 antibody, in patients from a squamous
in head and neck squamous cell carcinoma. J. Clin. Oncol. 30, 2102–2111 (2012). cell carcinoma of the head and neck (SCCHN)
Oncotarget 7, 74362–74379 (2016). 125. O’Sullivan, B. et al. Deintensification candidate expansion cohort [abstract]. J. Clin. Oncol. 33
105. Ferris, R. L., Jaffee, E. M. & Ferrone, S. Tumor antigen-​ subgroups in human papillomavirus-​related (Suppl. 15), 3011 (2015).
targeted, monoclonal antibody-​based immunotherapy: oropharyngeal cancer according to minimal risk of 144. Wolchok, J. D. et al. Nivolumab plus ipilimumab in
clinical response, cellular immunity, and distant metastasis. J. Clin. Oncol. 31, 543–550 advanced melanoma. N. Engl. J. Med. 369, 122–133
immunoescape. J. Clin. Oncol. 28, 4390–4399 (2013). (2013).
(2010). 126. Marur, S. et al. E1308: phase II trial of induction 145. Siu, L. L. et al. Safety and efficacy of durvalumab
106. Vermorken, J. B. et al. Open-​label, uncontrolled, chemotherapy followed by reduced-​dose radiation and with or without tremelimumab in patients with
multicenter phase II study to evaluate the efficacy and weekly cetuximab in patients with HPV-​associated PD-​L1-low/negative recurrent or metastatic HNSCC:
toxicity of cetuximab as a single agent in patients with resectable squamous cell carcinoma of the oropharynx the phase 2 CONDOR randomized clinical trial.
recurrent and/or metastatic squamous cell carcinoma — ECOG-​ACRIN Cancer Research Group. J. Clin. JAMA Oncol. 5, 195–203 (2018).
of the head and neck who failed to respond to Oncol. 35, 490–497 (2017). 146. Chen, D. S. & Mellman, I. Elements of cancer
platinum-​based therapy. J. Clin. Oncol. 25, 127. Gillison, M. L. et al. Radiotherapy plus cetuximab immunity and the cancer-​immune set point. Nature
2171–2177 (2007). Zor cisplatin in human papillomavirus-​positive 541, 321–330 (2017).
107. Burtness, B. et al. Phase III randomized trial of oropharyngeal cancer (NRG Oncology RTOG 1016): 147. Concha-​Benavente, F. et al. Identification of the
cisplatin plus placebo compared with cisplatin plus a randomised, multicentre, non-​inferiority trial. Lancet cell-​intrinsic and -extrinsic pathways downstream of
cetuximab in metastatic/recurrent head and neck 393, P40–P50 (2018). EGFR and IFNγ that induce PD-​L1 expression in head
cancer: an Eastern Cooperative Oncology Group study. 128. Hajek, M. et al. TRAF3/CYLD mutations identify a and neck cancer. Cancer Res. 76, 1031–1043 (2016).
J. Clin. Oncol. 23, 8646–8654 (2005). distinct subset of human papillomavirus-​associated 148. Dong, H. et al. Tumor-​associated B7-H1 promotes
108. Vermorken, J. B. et al. Platinum-​based chemotherapy head and neck squamous cell carcinoma. Cancer 123, T cell apoptosis: a potential mechanism of immune
plus cetuximab in head and neck cancer. N. Engl. 1778–1790 (2017). evasion. Nat. Med. 8, 793–800 (2002).
J. Med. 359, 1116–1127 (2008). 129. Chen, A. M. et al. Reduced-​dose radiotherapy for 149. Badoual, C. et al. PD-1-expressing tumor-​infiltrating
109. Bonner, J. A. et al. Radiotherapy plus cetuximab human papillomavirus-​associated squamous-​cell T cells are a favorable prognostic biomarker in HPV-​
for squamous-​cell carcinoma of the head and neck. carcinoma of the oropharynx: a single-​arm, phase 2 associated head and neck cancer. Cancer Res. 73,
N. Engl. J. Med. 354, 567–578 (2006). study. Lancet Oncol. 18, 803–811 (2017). 128–138 (2013).

www.nature.com/nrclinonc
Reviews

150. Burtness, B. et al. KEYNOTE-048: phase III study of 159. Ferris, R. L. et al. Effect of adding motolimod Competing interests
first-​line pembrolizumab (P) for recurrent/metastatic to standard combination chemotherapy and B.B. has received compensation for consulting from Aduro,
head and neck squamous cell carcinoma (R/M HNSCC) cetuximab treatment of patients with squamous cell Alligator Biosciences, AstraZeneca, Bayer, Boehringer
[abstract LBA8_PR]. Ann. Oncol. 29 (Suppl. 8), carcinoma of the head and neck: the Active8 Ingelheim, Bristol-​Myers Squibb, Celgene, DebioPharma,
mdy424.045 (2018). randomized clinical trial. JAMA Oncol. 4, 1583–1588 Genentech–Roche, Merck and TRM Oncology, has been a
151. Rizvi, N. A. et al. Mutational landscape determines (2018). non-​paid consultant of Adlai Norte and Celldex, has received
sensitivity to PD-1 blockade in non-​small cell lung 160. Ma, D. J. et al. Two-​year results for MC1273, a compensation for serving on data safety monitoring commit-
cancer. Science 348, 124–128 (2015). phase 2 evaluation of aggressive dose de-​escalation tees for IDDI (AstraZeneca–Medimmune) and VentiRx, and
152. Alexandrov, L. B. et al. Signatures of mutational for adjuvant chemoradiation in HPV+ oropharynx for medical education activities from CCO, Chemo Advisor,
processes in human cancer. Nature 500, 415–421 squamous cell carcinoma (OPSCC). Int. J. Radiat. PER, Peerview, Practice Update, Prime Oncology and
(2013). Oncol. Biol. Phys. 99, 1320 (2017). OncLive, and has received travel support from Boehringer
153. Antonia, S. J. et al. Overall survival with durvalumab 161. Siegel, R. et al. Phase II study: induction Ingelheim and Merck. Q.T.L. has received travel support for
after chemoradiotherapy in stage III NSCLC. N. Engl. chemotherapy and transoral surgery as definitive serving on the advisory board of Bristol-​Myers Squibb, serves
J. Med. 379, 2342–2350 (2018). treatment (Tx) for locally advanced oropharyngeal as a non-​paid consultant of Merck, and serves on the data
154. McBride, S. M. et al. A phase II randomized trial of squamous cell carcinoma (OPSCC): a novel approach safety monitoring committee for a clinical trial sponsored by
nivolumab with stereotactic body radiotherapy (SBRT) [abstract]. J. Clin. Oncol. 36 (Suppl. 15), 6004 Pfizer. R.L.F. has received compensation for consulting and/
versus nivolumab alone in metastatic (M1) head and (2018). or advisory board membership from AstraZeneca–
neck squamous cell carcinoma (HNSCC) [abstract]. 162. Ferris, R. L. et al. Nivolumab versus investigator’s Medimmune, Bristol-​M yers Squibb, Merck, PER, Pfizer,
J. Clin. Oncol. 36 (Suppl. 15), 6009 (2018). choice in recurrent or metastatic squamous cell Regeneron, Tesaro and TTMS Therapeutics. Through con-
155. Schuler, P. J. et al. Phase I dendritic cell p53 peptide carcinoma of the head and neck: 2-year long-​term tracts with the UPMC Hillman Cancer Center, his laboratory
vaccine for head and neck cancer. Clin. Cancer Res. survival update of CheckMate 141 with analyses by has also been funded to perform sponsored research by
20, 2433–2444 (2014). tumor PD-​L1 expression. Oral Oncol. 81, 45–51 AstraZeneca–Medimmune, Bristol-​Myers Squibb and Tesaro.
156. Harrington, K. J. et al. Phase I/II study of oncolytic (2018). J.D.C. declares no competing interests.
HSV GM-​CSF in combination with radiotherapy and 163. Zandberg, D. et al. Durvalumab for recurrent/
cisplatin in untreated stage III/IV squamous cell cancer metastatic (R/M) head and neck squamous cell Publisher’s note
of the head and neck. Clin. Cancer Res. 16, carcinoma (HNSCC): preliminary results from a single-​ Springer Nature remains neutral with regard to jurisdictional
4005–4015 (2010). arm, phase 2 study [abstract 10420]. Ann. Oncol. 28, claims in published maps and institutional affiliations.
157. Massarelli, E. et al. Combining immune checkpoint (Suppl. 5), v372–v394 (2017).
blockade and tumor-​specific vaccine for patients with Reviewer information
incurable human papillomavirus 16-related cancer: Acknowledgements Nature Reviews Clinical Oncology thanks A. Eisbruch and the
a phase 2 clinical trial. JAMA Oncol. 5, 67–73 The work of R.L.F. was supported by the US NIH grant P50 other anonymous reviewer(s), for their contribution to the
(2019). CA097190 and the University of Pittsburgh Cancer Center peer review of this work.
158. Davis, R. J., Ferris, R. L. & Schmitt, N. C. support grant P30CA047904.
Costimulatory and coinhibitory immune checkpoint
receptors in head and neck cancer: unleashing Author contributions Related links
immune responses through therapeutic combinations. All authors made a substantial contribution to all aspects of ClinicalTrials.gov: https://clinicaltrials.gov
Cancers Head Neck 1, 12 (2016). the preparation of the manuscript.

Nature Reviews | Clinical Oncology

You might also like