Post-therapeutic surveillance strategies in head and neck squamous cell carcinoma

14
1 23 European Archives of Oto-Rhino- Laryngology and Head & Neck ISSN 0937-4477 Eur Arch Otorhinolaryngol DOI 10.1007/s00405-012-2172-7 Post-therapeutic surveillance strategies in head and neck squamous cell carcinoma Antoine Digonnet, Marc Hamoir, Guy Andry, Missak Haigentz, Robert P. Takes, Carl E. Silver, Dana M. Hartl, Primož Strojan, et al.

Transcript of Post-therapeutic surveillance strategies in head and neck squamous cell carcinoma

1 23

European Archives of Oto-Rhino-Laryngologyand Head & Neck ISSN 0937-4477 Eur Arch OtorhinolaryngolDOI 10.1007/s00405-012-2172-7

Post-therapeutic surveillance strategies inhead and neck squamous cell carcinoma

Antoine Digonnet, Marc Hamoir,Guy Andry, Missak Haigentz, RobertP. Takes, Carl E. Silver, Dana M. Hartl,Primož Strojan, et al.

1 23

Your article is protected by copyright and

all rights are held exclusively by Springer-

Verlag. This e-offprint is for personal use only

and shall not be self-archived in electronic

repositories. If you wish to self-archive your

work, please use the accepted author’s

version for posting to your own website or

your institution’s repository. You may further

deposit the accepted author’s version on a

funder’s repository at a funder’s request,

provided it is not made publicly available until

12 months after publication.

REVIEW ARTICLE

Post-therapeutic surveillance strategies in head and necksquamous cell carcinoma

Antoine Digonnet • Marc Hamoir • Guy Andry • Missak Haigentz Jr. • Robert P. Takes • Carl E. Silver •

Dana M. Hartl • Primoz Strojan • Alessandra Rinaldo • Remco de Bree • Andreas Dietz • Vincent Gregoire •

Vinidh Paleri • Johannes A. Langendijk • Vincent Vander Poorten • Michael L. Hinni • Juan P. Rodrigo •

Carlos Suarez • William M. Mendenhall • Jochen A. Werner • Eric M. Genden • Alfio Ferlito

Received: 15 July 2012 / Accepted: 15 August 2012

� Springer-Verlag 2012

Abstract The management of head and neck squamous

cell carcinomas does not end with the completion of

ablative therapy. The oncologic objectives of post-treat-

ment follow-up are to detect recurrences and second pri-

mary tumors; beyond that, follow-up should evaluate acute

and chronic treatment-related side effects, guide the reha-

bilitation process, alleviate functional loss, manage pain,

restore nutritional status and assess psychosocial factors. In

this structured review, we address the questions of timing

and the tools required to achieve a complete and coherent

routine surveillance. Several guidelines and consensus

statements recommend clinical examination as the cor-

nerstone of follow-up which should be performed for at

least 5 years, although there are no data in favor of any one

particular follow-up program, and only low-level evidence

suggests an improvement in oncologic outcomes by close

follow-up. Baseline imaging (computed tomography andThis paper was written by members and invitees of the International

Head and Neck Scientific Group (http://www.IHNSG.com).

A. Digonnet � G. Andry

Department of Head and Neck and Thoracic Surgery,

Institut Jules Bordet, Brussels, Belgium

M. Hamoir

Department of Head and Neck Surgery,

Cancer Center, St. Luc University Hospital, Brussels, Belgium

M. Haigentz Jr.

Division of Oncology, Department of Medicine,

Montefiore Medical Center, Albert Einstein College

of Medicine, Bronx, NY, USA

R. P. Takes

Department of Otolaryngology-Head and Neck Surgery,

Nijmegen Medical Center, Radboud University,

Nijmegen, The Netherlands

C. E. Silver

Departments of Surgery and Otolaryngology-Head

and Neck Surgery, Montefiore Medical Center,

Albert Einstein College of Medicine, Bronx, NY, USA

D. M. Hartl

Department of Otolaryngology-Head and Neck Surgery,

Institut Gustave Roussy, Villejuif Cedex, France

D. M. Hartl

Laboratoire de Phonetique et de Phonologie,

Sorbonne Nouvelle, Paris, France

P. Strojan

Department of Radiation Oncology,

Institute of Oncology, Ljubljana, Slovenia

A. Rinaldo � A. Ferlito (&)

ENT Clinic, University of Udine,

Piazzale S. Maria della Misericordia, 33100 Udine, Italy

e-mail: [email protected]

R. de Bree

Department of Otolaryngology-Head and Neck Surgery,

VU University Medical Center, Amsterdam, The Netherlands

A. Dietz

Department of Otorhinolaryngology,

University of Leipzig, Leipzig, Germany

V. Gregoire

Radiation Oncology Department and Center for Molecular

Imaging and Experimental Radiotherapy, Universite Catholique

de Louvain, St-Luc University Hospital, Brussels, Belgium

V. Paleri

Department of Otolaryngology-Head and Neck Surgery,

Newcastle upon Tyne Foundation Hospitals NHS Trust,

Newcastle upon Tyne, UK

123

Eur Arch Otorhinolaryngol

DOI 10.1007/s00405-012-2172-7

Author's personal copy

magnetic resonance imaging) should be obtained within

2–6 months after definitive therapy if used for treatment

response evaluation. Metabolic response, if indicated,

should be assessed preferably after 3 months in patients

who undergo curative-intent therapy with (chemo)-radio-

therapy. Chest computed tomography is more sensitive

than plain radiography, if used in follow-up, but the benefit

and cost-effectiveness of routine chest computed tomog-

raphy has not been demonstrated. There are no current data

supporting modifications specific to the surveillance plan

of patients with human papillomavirus-associated disease.

Keywords Head and neck cancer � Surveillance strategy �Recurrence � HPV � Distant metastasis

Introduction

The care of patients with head and neck squamous cell

carcinoma (HNSCC) does not end with the completion of

definitive treatment and requires a period of post-thera-

peutic follow-up with three main objectives.

The first objective is to detect as soon as possible

recurrences, whether local, regional, and/or at distant sites.

It is widely accepted that patients who have received

potentially curative treatment for HNSCC are at risk for

loco-regional recurrences and second primary tumors.

Recurrence rates vary from \10 to 48 %, depending on

initial stage and primary tumor site [1, 2]. A prospective

study by Boysen et al. [3] published in 1992 found that

76 % of recurrences (all head and neck sites combined)

occurred in the first 2 years following treatment, with

another 11 % detected during the third year. de Visscher

and Manni [4] found that 76 % of recurrences, second

primary tumors or metastases (cancer-related ‘‘events’’)

occurred within 3 years of initial treatment. Similarly,

prospectively collected data analyzed by Lester and Wight

[5] found that 95 % of recurrences or second primaries

occurred within 2.7 years for oropharyngeal primaries,

2.3 years for hypopharyngeal primaries and 4.7 years for

laryngeal primaries. Wensing et al. [6] reported that 83 %

of recurrent disease in a series of 197 oral cavity squamous

cell cancer occurred within 2 years. Although early

detection of local and/or regional recurrence can offer the

possibility of successful salvage treatment with curative

intent [7], the early detection of distant metastasis is of

uncertain benefit to patients, as it implies incurable disease,

and in most cases treatment will be guided by symptoms in

a palliative setting [8]. Therapeutic options for locore-

gional recurrence continue to evolve. Recent demonstration

of promising new options such as re-irradiation [9, 10] with

or without targeted therapy and photodynamic therapy [11]

decrease the number of patients ‘‘without therapeutic

options.’’

The second objective of post-treatment surveillance lies

in the detection of second primary malignancies outside of

the head and neck region. In patients with primary tumors of

the upper aerodigestive tract (UADT), the reported rates of

second primary tumors in the esophagus or lung vary from 10

to 20 % [12, 13], depending on duration of follow-up.

The other objectives of surveillance are to evaluate

acute and chronic treatment-related side effects, guide the

rehabilitation process, alleviate functional loss, manage

pain, restore nutritional status, and assess the psychosocial

consequences of all these factors. The optimal surveillance

strategy for HNSCC patients who have undergone poten-

tially curative treatment is not clearly defined and varies

widely among practitioners [14–16].

J. A. Langendijk

Department of Radiation Oncology,

University Medical Center Groningen,

University of Groningen, Groningen,

The Netherlands

V. Vander Poorten

Department of Otorhinolaryngology-Head

and Neck Surgery and Leuven Cancer Institute,

University Hospitals Leuven,

Leuven, Belgium

M. L. Hinni

Department of Otolaryngology-Head and Neck Surgery,

Mayo Clinic, Phoenix, AZ, USA

J. P. Rodrigo � C. Suarez

Department of Otolaryngology,

Hospital Universitario Central de Asturias,

Oviedo, Spain

J. P. Rodrigo � C. Suarez

Instituto Universitario de Oncologıa del

Principado de Asturias, Oviedo, Spain

W. M. Mendenhall

Department of Radiation Oncology, University of Florida,

Gainesville, FL, USA

J. A. Werner

Department of Otolaryngology-Head and Neck Surgery,

Philipp University, Marburg, Germany

E. M. Genden

Department of Otolaryngology-Head and Neck Surgery,

The Mount Sinai Medical Center, New York, NY, USA

Eur Arch Otorhinolaryngol

123

Author's personal copy

Recently, the link between HNSCC, especially oropha-

ryngeal cancer (OPC), and human papillomavirus (HPV)

has been established [17]. HPV 16 is the most common

oncogenic genotype in these tumors. Patients with HPV-

associated HNSCC who are non-smokers seem to have a

better prognosis after completion of treatment [18]. Con-

versely, patients who have HPV-negative disease or those

who continue to smoke and drink after completion of

treatment are at high risk for recurrences. Guidelines for

surveillance stratified for HPV status are not well estab-

lished since epidemiological data in comparison with dif-

ferent continents (USA vs. Europe) and the individual mix

of risk factors are still confusing due to high heterogeneity.

In this structured review, we discuss the optimal timing for

visits and the tools required for surveillance.

Methods

A comprehensive literature search on PubMed/MEDLINE,

EMBASE and Cumulative index to Nursing and Allied

Health Science Literature (CINAHL) was performed dur-

ing the period of 1980–2011 for follow-up strategies in

patients with HNSCC. Keywords included head and neck

cancer, surveillance strategy, recurrence, and HPV. Selec-

ted articles fulfilled criteria of post-therapeutic surveillance

in HNSCC and use of any modality for surveillance of

HNSCC. Articles published in a language other than

English were not included. The reference list of the rele-

vant articles was analyzed and any other pertinent articles

were added to the review.

Results

Surveillance plan

There are several recommendations in the literature

regarding the post-treatment follow-up of HNSCC patients.

In their recent review, Manikantan et al. [19] concluded

that no one specific surveillance program is more efficient

in detecting recurrences or improving quality of life. Fur-

thermore, there is conflicting evidence as to whether

recurrences detected by the patient or those by routine

surveillance confer a survival advantage [2, 4]. Some

authors suggest that good patient education is important

and encourage patients to self-present at the first sign of

suspicious symptoms. In 2010, Flynn et al. [20] conducted

a study of 223 patients with locoregionally advanced (stage

III–IV) HNSCC to evaluate the utility of the follow-up

regimen used in their center. The recurrences were cate-

gorized into two groups: patient-detected cases were

defined as those for which the patients were symptomatic at

the time of the visit or were self-referred for pain or wor-

risome symptoms, and physician-detected cases were

defined as those for which patients had an asymptomatic

recurrence identified during follow-up consultation. The

authors found no improvement in disease-free or overall

survival in the physician-detected recurrence group when

compared with the patient-detected recurrence group. In

another study reported by Boysen et al. [3] which included

661 HNSCC patients considered free of disease at 6 weeks

after completion of therapy, the patients were seen every

2–3 months for the first 2 years and every 3–4 months for

the following 3 years. The authors found that the survival

rate was better in patients where recurrence was detected

through symptoms when compared with survival in

asymptomatic patients where recurrence was detected

through physical examination (PE). The majority of

recurrences (61 %) were diagnosed after self-referral for

specific symptoms. In this series, the authors did not

observe any recurrence after 3 years.

In a study on early stage (I and II) floor of mouth and

tongue cancer the effectiveness of a 10-year routine follow-

up was found to be limited, suggesting that visits on routine

basis can be stopped after 5 years [21]. In a study on lar-

yngeal carcinoma, a routine follow-up program did not lead

to survival benefit for asymptomatic patients with tumor

recurrence [22]. It was suggested that after the third year of

follow-up screening for recurrent tumor can be stopped [23]

and that an intensive and long follow-up schedule had lim-

ited influence on life expectancy in elderly people. The lack

of benefit of post-treatment oncologic surveillance programs

as compared with nationwide primary screening programs

may be explained by the fact that patients who have already

received oncologic treatment but develop a recurrence have a

worse prognosis compared with patients who have initial

malignancies. Other factors may be the limited therapeutic

options in the event of cancer recurrence, the detection of

slow-growing tumors (length–time bias) and the magnitude

and adjustment of lead time [23]. Also, for second primary

tumors, treatment options may be limited due to previous

treatments in the same anatomical site.

In contrast, Kissun et al. [24] retrospectively studied 278

patients treated for oral and OPC and reported that 54

patients (19.5 %) developed a recurrence within a median

of 8 months. Among them, only 20 patients were aware of

recurrent disease. Patients were considered aware of their

recurrence if they were experiencing new symptoms

defined by pain, lump, ulcer or swallowing difficulties. The

authors concluded that close clinical follow-up is needed

after treatment.

A study by de Visscher and Manni [4] of 428 patients

assessed whether curative treatment was possible in

patients in whom early recurrence, metastasis or second

primary tumors was detected by routine follow-up. The

Eur Arch Otorhinolaryngol

123

Author's personal copy

authors found that the mean survival of patients with

recurrences detected on routine follow-up was 58 months

while it was only 32 months in patients with recurrence

detected after self-referral.

These observed differences in the results of follow-up

strategies may reflect [25] that geographical and socio-

economic factors account for some of the variation in the

use of surveillance method. Lester and Wight [5] added

that routine follow-up must be planned with reference to

the local population.

Other authors propose a strict follow-up regimen during

the first 3 years only in patients where a salvage treatment

option could exist [26]. For patients without any potential

therapeutic option, follow-up should be focused more on

providing care and support than on detecting recurrence.

For other tumors resectability can only be determined after

restaging of the recurrent tumor.

In 2001, the British Association of Head and Neck On-

cologists (BAHNO) [27] advised a 4- to 6-week follow-up

schedule for the first 2 years, 3-month follow-up visits during

the third year, 6-month follow-up for the fourth and the fifth

years and annual visits thereafter. More recently, the National

Comprehensive Cancer Network (NCCN) [28] recom-

mended 1- to 3-month follow-up for the first year, 2- to

4-month follow-up for the second year, 4- to 6-month follow-

up for years 3 to 5 and every 6–12 months thereafter. The

most relevant schedules of follow-up visits reported in the

literature are summarized in Table 1 [5, 14, 16, 27, 28].

Some authors have proposed tailoring the frequency of

visits according to factors such as the primary tumor site and

stage of the disease [4, 29]. Although it would be reasonable

to tailor follow-up depending on risk-stratification for loco-

regional recurrences in particular, this could be difficult to

implement in clinical practice. Similarly, it may be argued

that the design of screening programs for distant metastasis

should be tailored to the risk of recurrence by identifying

those patients at highest risk for distant metastasis [30].

However, assessment of risk of recurrence would be more

relevant concerning locoregional recurrences, as patients will

only benefit if there are potentially curative treatment options

left for them. The presence of distant metastasis usually

implies incurable disease with the rare exception of oligo-

metastatic disease managed with locoregional therapy, and

management is often guided towards symptom control [8,

31]. The early detection of distant metastasis will not always

serve sensible purposes.

In our opinion, scheduled visits are still the best way to

offer an adequate follow-up to patients treated for HNSCC

as they address several purposes which are mentioned

above and not just for early detection of recurrence or

second primaries. Self-presentation at the first sign of

suspicious symptoms or other problems related to onco-

logical treatment must be used in combination with plan-

ned surveillance but should not replace it.

Surveillance tools

Physical examination

PE remains the cornerstone for detection of potential

recurrence. It is the most easily available and reliable

method for assessing superficial mucosal tumors and/or the

extent of mucosal involvement of other tumors [32]. Cur-

rent studies justify the need for PE in the routine follow-up,

but never provide exact details [15, 16, 19, 33]. As PE

should be easily standardized and reproducible, we propose

to present it as a checklist (Table 2) derived from the lit-

erature [27, 28]. In this proposal, PE should include cranial

nerve examination, an assessment of vocal, breathing and

swallowing functions, and a systematic pain evaluation

using a visual analogic scale (VAS). A complaint of new or

worsening pain should alert the physician to potential

disease recurrence. Flexible fiberoptic endoscopy is para-

mount for a comprehensive examination of the UADT [27]

as well as direct laryngoscopy when indicated.

Improvements in endoscopic evaluation for detection of

early recurrences or second primary tumors include tech-

niques like high-definition digital video endoscopy with

Table 1 Scheduled visits in the literature

Frequency of the visits

BAHNO [27] NCCN [28] ASHNS [16] SHNS [16] Lester and Wight [5] DHNS [14]

Year 1 4–6 weeks 1–3 months 1–3 months 1–3 months 1 month 2 months

Year 2 4–6 weeks 2–4 months 2–4 months 2–4 months 2 months 3 months

Year 3 3 months 4–6 months 3–6 months 3–6 months 3 months 4 months

Year 4 6 months 4–6 months 4–6 months 4–6 months 4 months 6 months

Year 5 6 months 4–6 months 4–6 months 4–6 months 6 months 6 months

[Year 6 1 year 6–12 months 1 year 1 year 1 year None

Eur Arch Otorhinolaryngol

123

Author's personal copy

narrow band imaging [34, 35] or laser-induced fluores-

cence endoscopy [36].

The weight of the patient should be noted at each visit, and

any unexplained weight loss should alert the physician.

Detection of functional outcomes remains a critical issue; in

2001, the World Health Organization (WHO) adopted the

International Classification of Functioning, Disability and

Health (ICF). The ICF has been tailored specifically for head

and neck cancer (ICF-HNC). The ICF core sets for head and

neck cancer are divided into four items: bodily functions, body

structures, participation in activities and environmental factors.

The ICF-HNC can be used routinely to provide a clear estimate

of early and late functional outcome measures [37, 38].

Dental status is an important and often overlooked aspect

of the clinical follow-up. Adequate evaluation and pre-

ventive periodontal care prior to treatment should be coupled

with a rigorous continued assessment of oral health during

follow-up visits, given the importance of oral health to self-

rated quality of life and to the potential complications arising

from failure to follow-up this aspect [39, 40].

Radiation-induced toxicity should also be objectively

monitored during follow-up evaluations. Toxicity reporting

systems such as the Common Terminology Criteria for

Adverse Events (CTCAE) scales, Late Effects of Normal

Tissues (LENT) and Subjective, Objective, Management

and Analytic (SOMA) scales have been shown to be a valid

measure of head and neck radiotherapy toxicity [41].

Bronchoscopy, esophagoscopy and panendoscopy

Historically, bronchoscopy and esophagoscopy have been

performed during initial evaluations of patients with

Table 2 Checklist for head and neck examination

Nodes Mass Ulceration

External fields

Nodes Level I-VIParotid gland

Examination-palpation

ThyroidOccipital fieldScalpFacial pedicles

Upper aerodigestive tract (UADT)

Oral cavity-oropharynxBispatulate examinationBimanual palpation

Nasal cavity

Flexible fiberoptic endoscopy

NasopharynxOropharynx

Hypopharynx, base of the tongue Rigid endoscopyLarynx

Functional outcomes

Weight (kg)Pain (VAS)Cranial nerve dysfunctionSwallowing disorder on indicationVoice assessment on indicationBreathing assessment on indication

Eur Arch Otorhinolaryngol

123

Author's personal copy

HNSCC to rule out the presence of synchronous second

primary tumors [42]. With regard to follow-up of patients

after definitive therapy, some authors have recommended

routine panendoscopy within 2 years of completion of

treatment [43], while others, despite high accuracy of the

procedure, do not support it, arguing that it is an aggressive

and unpleasant procedure [44]. Moreover, the yield is too

low to justify these invasive procedures. The use of routine

screening during follow-up for distant metastases and/or

second primary tumors in the esophagus and lower airway

in particular is questionable and, if indicated, is currently

often achieved with positron emission computed tomog-

raphy (PET-CT) (see below).

Another recent technique for close surveillance is the

use of autofluorescence bronchoscopy and esophagoscopy

that may detect bronchogenic and esophageal carcinoma at

an early stage in high-risk patients [45].

Thyroid function testing

Even the most modern techniques for delivery of curative

head and neck radiation therapy (RT) will inevitably

cause incidental exposure of non-target tissues and organs.

When conventional, non-intensity modulated radiation

therapy (IMRT) techniques were used, the prevalence of

radiation-induced hypothyroidism (RIHT) was reported to

range from 10 to 45 % among patients receiving RT to

the neck and its dose-dependent fields. The mean radia-

tion dose at which 50 % of patients will experience

hypothyroidism was estimated to be approximately 44 Gy

[46]. The risk is significantly higher in patients with lar-

yngeal cancer, especially those undergoing laryngectomy,

and does not seem to be associated with chemotherapy or

age [47].

Half of these events will appear within 5 years, with a

peak incidence after 2–3 years [48]. While surgery

including partial thyroidectomy is associated with an

increased risk of hypothyroidism in patients treated with

RT, even surgery sparing the thyroid gland is associated

with an increased risk of hypothyroidism when combined

with RT, possibly due to an increased risk of vascular

damage [49]. In a study dealing with 504 patients [50] in

which neck RT was part of the treatment for HNSCC, the

absence of hypothyroidism was 78 % at 5 years and 51 %

at 10 years. The authors concluded that serum thyroid

stimulating hormone (TSH) should be checked at 6-month

intervals for the first 5 years and yearly thereafter. The

NCCN [28] proposed TSH evaluations every 6–12 months

for the first 5 years. However, these recommendations are

just ‘‘guidelines’’ and are not applicable in patients that do

not have surgery in the thyroid bed or if no radiation has

been given. The point of initiating hormone replacement

therapy was proposed at a TSH value reaching 4.5 mIU/L.

Imaging studies

Computed tomography (CT) and magnetic resonance

imaging (MRI) A baseline imaging study is often recom-

mended after completion of therapy, allowing a compari-

son with subsequent images for earlier detection of lesions

[7]. These baseline studies could be particularly useful for

patients treated by (chemo)radiotherapy and for patients

treated with primary surgery for tumors in anatomical

regions difficult to evaluate on PE, such as the paranasal

sinuses and skull base. Accurate interpretation of imaging

is crucial to differentiate between post-treatment changes

and residual disease or recurrence. It is generally recom-

mended that the post-treatment ‘‘baseline’’ CT/MRI should

be performed 3-6 months after treatment [28, 32], and

subsequent imaging should be with the same modality.

Hermans et al. [51] reviewed the clinical records of 66

patients who underwent radiotherapy for laryngeal and

hypopharyngeal cancers. Twenty-nine patients experienced

local failure; in 12 patients suspicious CT findings were

apparent prior to physical findings and were confirmed by

biopsy. The authors concluded that imaging was necessary

for routine follow-up and recommended an interval of

3–4 months between studies for a duration of 2 years. Use

of imaging-based information could lead to earlier diag-

nosis of recurrence, allowing more efficient salvage sur-

gery and could improve the survival rate of these patients,

but this remains to be demonstrated with higher level

evidence (prospective randomized studies) and whether

this also applies to only surgically treated patients.

The aforementioned studies addressed either all sites

and stages of HNSCC or were limited to laryngeal and

hypopharyngeal cancers. Our review found no study

reporting the utility of imaging follow-up for early stage

oral carcinoma, the most common site of HNSCC. In the

particular subgroup of patients with carcinoma of the oral

cavity as well of the larynx and other sites easily accessible

for clinical examination, it may be appropriate to monitor

by PE alone. In 2011, the NCCN recommendations pro-

posed further reimaging based only on clinical signs and

symptoms. Routine imaging was not recommended for

asymptomatic patients [28].

There are no data suggesting the superiority of con-

ventional MRI over CT scan for follow-up assessment. It

appears logical, at least after radiotherapy or chemoradio-

therapy, to use the same imaging modality as the pre-

treatment study. Typically, MRI is recommended for

patients with sinonasal, skull base and nasopharyngeal

tumors and when there is suspicion of perineural or intra-

cranial spread [19]. Diffusion-weighted MRI (DW-MRI)

measures the diffusivity of water molecules in biologic

tissues and has been demonstrated to be effective in

detecting recurrences after completion of RT [52]. In this

Eur Arch Otorhinolaryngol

123

Author's personal copy

setting it has been shown to be superior to anatomical

imaging [53]. DW-MRI also shows promise for assessing

treatment response immediately after completion of che-

moradiotherapy at 3 weeks [54], whereas the window of

opportunity for PET-CT is only at 3 months after com-

pletion (see below).

Chest CT/chest X-ray The lungs are the most common

site of distant metastasis [55], and primary lung cancer

accounts for 23 % of metachronous second primaries in

HNSCC patients [55, 56]. Traditionally, conventional chest

radiography has been part of the follow-up. However, chest

X-ray has been reported as inadequate for the early

detection of synchronous and metachronous neoplasms at a

curative stage [57, 58].

In a recent study, only 33 % of intrathoracic lesions

picked up by CT were also detected by chest X-ray alone

[51]. In a retrospective study [59] of 26 patients undergoing

treatment for HNSCC, it was observed that 4 patients with

a normal chest X-ray had an abnormal CT. Similarly, in a

series of 102 with newly diagnosed mucosal head and neck

cancer patients, 11 were proven to have pulmonary

metastasis or primary lung tumor, and 7 had normal chest

X-rays. The authors concluded that chest CT should be

used rather than chest X-ray for the follow-up of HNSCC

patients. Notably, radiation doses delivered by CT have

decreased with the latest generation of CT scans [60].

However, in developing countries the availability of CT for

routine screening in asymptomatic patients remains a

limiting factor.

Aside from the question of which technique is best, the

question remains as to how effective is routine screening

for lung metastases or second primary lung cancer.

According to Hsu et al. [61] chest CT should be mandatory

in follow-up and is recommended every 6 months during

the first 2 years in high-risk (stage III–IV) patients. On the

other hand, screening by chest X-ray to detect lung cancer

in an asymptomatic stage after curative treatment for

squamous cell laryngeal cancer was reported to not

improve survival for patients who develop lung cancer

[62]. As only a limited number of patients with second

primary lung cancer were detected by annual radiography,

of which only a small percentage was eligible for curative

surgical treatment, screening by annual chest X-ray has

been reported to be of little benefit [63].

In summary, there is currently no high-level evidence to

support routine plain chest radiography, either for

improved oncologic outcomes or from a cost-benefit

standpoint.

Neck ultrasonography The accuracy of ultrasound (US)

and CT for detection of neck lymph node metastasis is

comparable [64–66]. Table 3 [64–66] reports the sensitiv-

ity and specificity of neck US and CT reported in the lit-

erature. With the use of color duplex ultrasonography,

Leuwer et al. [67] achieved a sensitivity of 90 % in

detecting neck metastasis. Neck US is frequently included

in the routine surveillance of patients treated for HNSCC,

avoiding radiation exposure and allowing fine needle

aspiration of suspicious lesions under US guidance (US-

gFNA). US-guided cytology of metastatic lymph nodes

that have been irradiated is less reliable than when applied

in the pre-treatment setting [68]. Park et al. [69] retro-

spectively reviewed the medical records of 140 HNSCC

patients followed at least for 5 years after completion of

definitive therapy. Patients were seen every month during

the first year, every 2 months during the second year, every

3 months during the third year, every 4 months during the

fourth year and every 6 months thereafter. Each visit

included medical history, PE, indirect or direct endoscopy

and neck US. A chest X-ray was taken every 6–12 months.

The authors reported that regional recurrences were diag-

nosed earlier than local failure (9 vs. 19 months after

completion of therapy), suggesting earlier detection of

small-sized nodal metastasis by neck ultrasonography. The

same authors concluded that neck ultrasonography has no

impact on the detection rate of nodal metastases but allows

earlier detection resulting in more rapid treatment. This

would allow, when suitable, less morbid neck dissection

with more favorable oncologic outcome as compared with

delayed diagnosis and more extended regional recurrence.

Nieuwenhuis et al. [70] reported a high salvage rate (79 %)

if delayed metastases are detected earlier by routine sur-

veillance USgFNA (each 3 months) in the previously

untreated neck. There is currently no high-level evidence,

however, regarding improvement in survival or cost-

effectiveness of routine follow-up neck screening using US

or neck CT to detect regional recurrences.

Finally, concerning the monitoring of treatment-related

morbidities, screening for extra-cranial carotid artery dis-

ease by specific US examination might be considered, as

patients with HNSCC treated with radiotherapy are at an

increased risk for this condition [71, 72]. Furthermore, the

physician should be aware that in this setting, the risk of

internal carotid stenosis increases following treatment.18Fluorodeoxyglucose (FDG)-PET-CT FDG-PET-CT

has been widely employed for the staging of HNSCC due

to the avidity of FDG for these tumors [73]. According to

Schoder et al. [74], one of the main applications of PET-CT

is the detection of residual disease in lymph nodes after

(chemo)radiotherapy. They found a high negative predic-

tive value (NPV [95 %) suggesting that planned neck

dissection could be avoided in many cases. Several post-

treatment studies [75–78] have reported that FDG-PET-CT

is very accurate for detection of locoregional and distant

recurrence of HNSCC. Gupta et al. [79] performed a sys-

tematic review and meta-analysis on the diagnostic per-

formance of post-treatment FDG-PET or FDG-PET-CT

Eur Arch Otorhinolaryngol

123

Author's personal copy

imaging in head and neck cancer. A total of 51 studies

involving 2,335 patients were included. The weighted

mean pooled sensitivity, specificity, positive predictive

value (PPV) and NPV for the primary site were 79.9, 87.5,

58.6 and 95.1 % and for the neck 72.2, 87.6, 52.1 and

94.5 %, respectively. These latter figures are also depen-

dent on the incidence of the disease, which may differ in

the different study populations. Meta-regression analysis

showed no significant difference between stand-alone PET

and integrated PET/CT, suggesting lack of impact of

technological and methodological advancements in PET

imaging on diagnostic performance in the post-treatment

setting [79].

The optimal timing for response assessment reported in

the literature ranges from 8 to 12 weeks [80], with most

authors agreeing that waiting at least 12 weeks allows one

to be sure that a negative result is truly negative [81–83].

Gupta et al. [79] found in a meta-analysis that scans done at

12 weeks or more after completion of definitive therapy

had a moderately higher diagnostic accuracy on meta-

regression analysis using time as covariate. The optimal

timing of post-treatment FDG-PET is still a subject of

debate [84].

Recently, Haerle et al. [83] studied the accuracy of

PET-CT in detecting distant metastasis in HNSCC

patients. This study retrospectively reviewed 299 patients

who underwent PET-CT imaging 3 months after com-

pletion of therapy and then every 6 months for 2 years.

The authors reported an NPV of 99.6 % and a PPV of

67.4 %. Although this study showed a high accuracy in

detecting distant metastasis, the drawback of using PET-

CT routinely in follow-up is the high number of false

positives resulting in increased emotional burden and

costs associated with follow-up and verification of all

suspicious lesions. Nevertheless, the authors recom-

mended screening for distant metastases with PET-CT at

least annually during the first 2 years after treatment [83].

However, it must be noted that in contrast to screening for

locoregional recurrences, screening for distant metastases

during follow-up remains debatable if no beneficial

treatment options for (asymptomatic) distant metastases

are available.

Lee et al. [85] tried to reach a consensus regarding the

interval and frequency of PET-CT for surveillance of

HNSCC patients. A review of 189 patients after completion

of treatment to detect residual cancer or recurrence was

performed. Twelve patients with inadequate clinical follow-

up or metastatic work up as well as 18 patients who under-

went PET-CT within 2 months to determine metabolic

response were excluded. A total of 159 charts of HNSCC

patients with a median follow-up of 24 months (3–65

months) were reviewed. One hundred twelve patients had

one PET-CT, 33 had 2 studies, 12 had 3 studies and 2 had 4

studies. The PET-CTs were divided into four groups

according to time after completion of treatment: 2–6 months

(group 1), 6–12 months (group 2), 12–24 months (group 3)

and [24 months (group 4). Each group was further subdi-

vided by indication: suspicion of recurrence versus ‘‘rou-

tine’’ PET-CT. The NPV for locoregional recurrence was 97

and 100 % for distant metastasis or second primary cancer.

The PPV for locoregional recurrence was 72 and 66 % for

distant metastasis and second primary cancer. Based on their

data, the authors concluded that routine PET-CT performed

within 2–6 months was highly accurate for detecting early

recurrences. However, most agree that it should not be

employed during the first 12 weeks after completion of

therapy [86]. In addition, of 106 patients who had a true

negative PET-CT, 37 (35 %) patients were in group 1, 27

(25 %) in group 2, 29 (27 %) in group 3 and 13 (12 %) in

group 4. Long-term follow-up of these patients suggested

that locoregional recurrence was unlikely for at least 1 year

after initial negative PET scans, suggesting that for patients

with negative PET-CT studies, conventional evaluation may

be sufficient for post-treatment surveillance during the next

year, unless obvious suspicious signs appear. The authors

concluded that initial PET-CT should be performed within

6 months after treatment and that the next routine PET-CT

should be performed 1 year later.

In summary, FDG PET-CT is highly effective for

assessing recurrence in patients treated for HNSCC.

However, cost effectiveness, impact on management and

survival impact remain to be evaluated more thoroughly.

No prospective trial comparing PET-CT to other imaging

modalities for follow-up has been conducted.

Patients with HPV-associated HNSCC

It is now widely acknowledged that a growing proportion

of HNSCC, particularly OPC, results from an infection by

HPV [6]. HPV 16 is the most common genotype of all HPV

DNA-positive cancers, accounting for 87 % of HPV-rela-

ted OPCs. HPV 18 is the second common genotype of

HPV-related cancer, occurring in 1 % of OPCs. HPV DNA

infection increases the relative risk of HPV-associated

HNSCC by 15- to 200-fold compared with the risk without

Table 3 Studies comparing accuracy of CT and US

Timing of

imaging

CT US

Di Martino

et al. [64]

Pre- and post-

treatment

Sens: 84 %

Spe: 91 %

Sens: 84 %

Spe: 87 %

Yoon et al.

[65]

Post-treatment Sens: 77 %

Spe: 99.4 %

Sens: 78.5 %

Spe: 98.5 %

Adams et al.

[66]

Post-treatment Sens: 82 % Sens: 72 %

Eur Arch Otorhinolaryngol

123

Author's personal copy

HPV infection [87]. Three types of tumors have been

defined: HPV-negative/p16-negative, HPV-negative/p16-

positive and HPV-positive/p16-positive. In a study dealing

with 323 patients, the authors reported the effect of tumor

HPV status on survival among patients with HNSCC [18].

The authors found 3-year overall survival of 82.4 % in the

HPV-positive group and 57.1 % in the HPV-negative

group. The 3-year progression-free survival was 73.7 % in

the HPV-positive group and 43.4 % in the HPV-negative

group. The authors also found that patients with HPV-

positive tumors had a 58 % reduction in the risk of death as

compared with patients with HPV-negative tumors.

Outcomes were also evaluated according to the

expression of p16 protein, associated with oncogenically

active HPV infection. P16 expression was associated with a

3-year overall survival of 83.6 % while the absence of p16

protein was associated with a 3-year overall survival of

51.3 %. This study provided strong evidence that tumor

HPV status was an independent prognostic factor for

overall and progression-free survival among patients with

HNSCC. Patients with HPV-positive tumors also have a

lower risk of developing metachronous second primaries

[88, 89]. According to our review, there are no data indi-

cating that the surveillance plan of patients with HPV-

positive disease should different than the follow-up of

patients with HPV-negative disease. However, HPV status,

in non-smokers particularly, is important information that

could be used by the clinicians to provide reassurance to

patients regarding prognosis. Notably, p16 status is an

independent predictor of outcome regardless of HPV status

and is generally much cheaper and easier to perform [90].

Conclusions

According to this literature review and from our own

experience, it is recommended that patients be followed in

the clinic every 2–3 months for the first 2 years, every

3–6 months for years three and four, every 6 months dur-

ing the fifth year and annually thereafter. However, other

frequencies and durations of follow-up may be followed

according to the literature [5, 16, 27–29]. An important

issue is which items are important to evaluate during these

visits. In our opinion, each visit should comprise a thor-

ough clinical history of symptoms and PE (Table 2).

Toxicity evaluation, rehabilitation of functional loss, pain

management, nutritional support and psychological support

including tobacco cessation should be addressed during

each of the visits. Weight should be assessed at each visit.

Systematic evaluation of radiation-induced late toxicity

should also be performed using published criteria.

Patients should be educated about relevant symptoms

and the need for additional visits if new symptoms appear.

In addition to palpation of the neck and direct visual

examination of the oral cavity, oropharynx and dental

status, flexible fiberoptic endoscopy of the UADT, should

be performed at each PE. A baseline post-treatment

imaging study (CT or MRI) should be performed within

3–6 months after completion of primary therapy in patients

treated for locoregionally advanced stage HNSCC in sites

not easily accessible to physical or fiberoptic examination

or when evaluating treatment response. Additional imaging

modalities should be performed based on clinical signs

and symptoms. In case of suspicious lymph nodes found

at clinical examination, ultrasonography can be performed

with fine needle aspiration biopsy. PET-CT provides high

accuracy for detecting residual disease and should be

performed at 12 weeks after treatment of patients who

have undergone definitive chemoradiotherapy. The value

of additional PET-CT evaluations for detection of loco-

regional and distant recurrences is debatable as its impact

on survival remains to be evaluated. Screening of distant

lung lesions may be performed by CT scan annually for

the first 2 years in high-risk patients. Serum TSH levels

should be checked at 6 monthly intervals for the first

5 years and yearly thereafter in patients treated with neck

radiotherapy.

Although non-smoking patients with HPV-positive

tumors have a better prognosis, they should have the same

routine follow-up plans as patients with HPV-negative

tumors, as there is no evidence to support other follow-up

regimens.

In the future, new modalities including biomarkers to

identify patients with low risk of developing distant

metastasis, molecular techniques for early detection of

recurrence, patient-based saliva proteomics and artificial

‘‘noses’’ (nanoparticle sensors for testing breath samples)

could lead to the development of cost-effective, fast, and

reliable methods for early detection of primary head and

neck cancer, recurrences or second primary tumors.

References

1. Cooney TR, Poulsen MG (1999) Is routine follow-up useful after

combined-modality therapy for advanced head and neck cancer?

Arch Otolaryngol Head Neck Surg 125:379–382

2. Virgo KS, Paniello RC, Johnson FE (1998) Costs of posttreat-

ment surveillance for patients with upper aerodigestive tract

cancer. Arch Otolaryngol Head Neck Surg 124:564–572

3. Boysen M, Lovdal O, Tausjo J, Winther F (1992) The value of

follow-up in patients treated for squamous cell carcinoma of the

head and neck. Eur J Cancer 28:426–430

4. de Visscher AV, Manni JJ (1994) Routine long-term follow-up in

patients treated with curative intent for squamous cell carcinoma

of the larynx, pharynx, and oral cavity. Does it make sense? Arch

Otolaryngol Head Neck Surg 120:934–939

5. Lester SE, Wight RG (2009) ‘When will I see you again?’ Using

local recurrence data to develop a regimen for routine

Eur Arch Otorhinolaryngol

123

Author's personal copy

surveillance in post-treatment head and neck cancer patients. Clin

Otolaryngol 34:546–551

6. Wensing BM, Merkx MA, Krabbe PF, Marres HA, Van den

Hoogen FJ (2011) Oral squamous cell carcinoma and a clinically

negative neck: the value of follow-up. Head Neck 33:1400–1405

7. Ridge JA (1993) Squamous cancer of the head and neck: surgical

treatment of local and regional recurrence. Semin Oncol 20:419–429

8. Haigentz M Jr, Hartl DM, Silver CE et al (2012) Distant

metastases from head and neck squamous cell carcinoma. Part III.

Treatment. Oral Oncol 48:787–793

9. Kurzweg T, Mockelmann N, Laban S, Knecht R (2012) Current

treatment options for recurrent/metastatic head and neck cancer: a

post-ASCO 2011 update and review of last year’s literature. Eur

Arch Otorhinolaryngol [Epub ahead of print]

10. Balermpas P, Keller C, Hambek M et al (2012) Reirradiation with

cetuximab in locoregional recurrent and inoperable squamous

cell carcinoma of the head and neck: feasibility and first efficacy

results. Int J Radiat Oncol Biol Phys 83:e377–e383

11. Tan IB, Dolivet G, Ceruse P, Vander Poorten V, Roest G,

Rauschning W (2010) Temoporfin-mediated photodynamic

therapy in patients with advanced, incurable head and neck

cancer: a multicenter study. Head Neck 2:1597–1604

12. Schwartz LH, Ozsahin M, Zhang GN et al (1994) Synchronous and

metachronous head and neck carcinomas. Cancer 74:1933–1938

13. Sturgis EM, Miller RH (1995) Second primary malignancies in

the head and neck cancer patient. Ann Otol Rhinol Laryngol

104:946–954

14. Kaanders JH, Dutch Cooperative Head and Neck Oncology

Group (2002) Carcinoma of the larynx: the Dutch national

guideline for diagnostics, treatment, supportive care and reha-

bilitation. Radiother Oncol 63:299–307

15. Johnson F, Johnson M, Virgo K (1997) Current follow-up strat-

egies after potentially curative resection of upper aerodigestive

tract epidermoid carcinoma. Int J Oncol 10:927–931

16. Paniello RC, Virgo KS, Johnson MH, Clemente MF, Johnson

FE (1999) Practice patterns and clinical guidelines for post-

treatment follow-up of head and neck cancers: a comparison of

2 professional societies. Arch Otolaryngol Head Neck Surg

125:309–313

17. Andl T, Kahn T, Pfuhl A et al (1998) Etiological involvement of

oncogenic human papillomavirus in tonsillar squamous cell car-

cinomas lacking retinoblastoma cell cycle control. Cancer Res

58:5–13

18. Ang KK, Harris J, Wheeler R et al (2010) Human papillomavirus

and survival of patients with oropharyngeal cancer. N Engl J Med

363:24–35

19. Manikantan K, Khode S, Dwivedi RC et al (2009) Making sense

of post-treatment surveillance in head and neck cancer: when and

what of follow-up. Cancer Treat Rev 35:744–753

20. Flynn CJ, Khaouam N, Gardner S et al (2010) The value of

periodic follow-up in the detection of recurrences after radical

treatment in locally advanced head and neck cancer. Clin Oncol

(R Coll Radiol) 22:868–873

21. Merkx MA, van Gulick JJ, Marres HA et al (2006) Effectiveness

of routine follow-up of patients treated for T1-2N0 oral squamous

cell carcinomas of the floor of mouth and tongue. Head Neck

28:1–7

22. Ritoe SC, Krabbe PF, Kaanders JH, van den Hoogen FJ, Verbeek

AL, Marres HA (2004) Value of routine follow-up for patients

cured of laryngeal carcinoma. Cancer 101:1382–1389

23. Ritoe SC, Verbeek AL, Krabbe PF, Kaanders JH, van den Hoo-

gen FJ, Marres HA (2007) Screening for local and regional

cancer recurrence in patients curatively treated for laryngeal

cancer: definition of a high-risk group and estimation of the lead

time. Head Neck 29:431–438

24. Kissun D, Magennis P, Lowe D, Brown JS, Vaughan ED, Rogers

SN (2006) Timing and presentation of recurrent oral and oro-

pharyngeal squamous cell carcinoma and awareness in the out-

patient clinic. Br J Oral Maxillofac Surg 44:371–376

25. Johnson FE, Johnson MH, Clemente MF, Paniello RC, Virgo KS

(2006) Geographical variation in surveillance strategies after

curative-intent surgery for upper aerodigestive tract cancer. Ann

Surg Oncol 13:1063–1071

26. Kazi R, Manikanthan K, Pathak KA, Dwivedi RC (2010) Head and

neck squamous cell cancers: need for an organised time-bound

surveillance plan. Eur Arch Otorhinolaryngol 267:1969–1971

27. British Association of Head and Neck Oncologists (2001) Prac-

tice care guidance for clinicians participating in the management

of head and neck cancer patients in the UK. Drawn up by a

consensus group of practising clinicians. Practice care guidance

for clinicians participating in the management of head and neck

cancer patients in the UK. EJSO 27(Suppl A):S1–S17

28. Pfister DG, Ang KK, Brizel DM et al (2011) National Compre-

hensive Cancer Network clinical practice guidelines in oncology.

Head and neck cancers. J Natl Compr Canc Netw 9:596–650

29. Johnson FE, Virgo KS, Clemente MF, Johnson MH, Paniello RC

(1998) How tumor stage affects surgeons’ surveillance strategies

after surgery for carcinoma of the upper aerodigestive tract.

Cancer 82:1932–1937

30. Coca-Pelaz A, Rodrigo JP, Suarez C (2012) Clinicopathologic

analysis and predictive factors for distant metastases in patients

with head and neck squamous cell carcinomas. Head Neck

34:771–775

31. de Bree R, Haigentz M Jr, Silver CE et al (2012) Distant

metastases from head and neck squamous cell carcinoma. Part II.

Diagnosis. Oral Oncol 48:780–786

32. Daisne JF, Duprez T, Weynand B et al (2004) Tumor volume in

pharyngolaryngeal squamous cell carcinoma: comparison at CT,

MR imaging, and FDG PET and validation with surgical speci-

men. Radiology 233:93–100

33. Manikantan K, Dwivedi RC, Sayed SI, Pathak KA, Kazi R (2011)

Current concepts of surveillance and its significance in head and

neck cancer. Ann R Coll Surg Engl 93:576–582

34. Chu PY, Tsai TL, Tai SK, Chang SY (2012) Effectiveness of

narrow band imaging in patients with oral squamous cell carci-

noma after treatment. Head Neck 34:155–161

35. Piazza C, Cocco D, De Benedetto L, Bon FD, Nicolai P, Peretti G

(2010) Role of narrow-band imaging and high-definition televi-

sion in the surveillance of head and neck squamous cell cancer

after chemo- and/or radiotherapy. Eur Arch Otorhinolaryngol

267:1423–1428

36. Rydell R, Eker C, Andersson-Engels S, Krogdahl A, Wahlberg P,

Svanberg K (2008) Fluorescence investigations to classify

malignant laryngeal lesions in vivo. Head Neck 30:419–426

37. Tschiesner U, Rogers S, Dietz A, Yueh B, Cieza A (2010)

Development of ICF core sets for head and neck cancer. Head

Neck 32:210–220

38. Leib A, Cieza A, Tschiesner U (2012) Perspective of physicians

within a multidisciplinary team: content validation of the com-

prehensive ICF core set for head and neck cancer. Head Neck

34:956–966

39. Schuurhuis JM, Stokman MA, Roodenburg JL et al (2011) Effi-

cacy of routine pre-radiation dental screening and dental follow-

up in head and neck oncology patients on intermediate and late

radiation effects. A retrospective evaluation. Radiother Oncol

101:403–409

40. Tolentino Ede S, Centurion BS, Ferreira LH, Souza AP, Damante

JH, Rubira-Bullen IR (2011) Oral adverse effects of head and neck

radiotherapy: literature review and suggestion of a clinical oral care

guideline for irradiated patients. J Appl Oral Sci 19:448–454

Eur Arch Otorhinolaryngol

123

Author's personal copy

41. Ho KF, Farnell DJ, Routledge JA et al (2009) Developing a

CTCAEs patient questionnaire for late toxicity after head and

neck radiotherapy. Eur J Cancer 45:1992–1998

42. Stoeckli SJ, Zimmermann R, Schmid S (2001) Role of routine

panendoscopy in cancer of the upper aerodigestive tract. Oto-

laryngol Head Neck Surg 124:208–212

43. Rachmat L, Vreeburg GC, de Vries N et al (1993) The value of

twice yearly bronchoscopy in the work-up and follow-up of

patients with laryngeal cancer. Eur J Cancer 29A:1096–1099

44. Haughey BH, Gates GA, Arfken CL, Harvey J (1992) Meta-

analysis of second malignant tumors in head and neck cancer: the

case for an endoscopic screening protocol. Ann Otol Rhinol

Laryngol 101:105–112

45. Lee P, de Bree R, Brokx HA, Leemans CR, Postmus PE, Sutedja

TG (2008) Primary lung cancer after treatment of head and neck

cancer without lymph node metastasis: is there a role for auto-

fluorescence bronchoscopy? Lung Cancer 62:309–315

46. Bakhshandeh M, Hashemi B, Mahdavi SR, Nikoofar A, Vashe-

ghani M, Kazemnejad A (2012) Normal tissue complication

probability modeling of radiation-induced hypothyroidism after

head-and-neck radiation therapy. Int J Radiat Oncol Biol Phys

[Epub ahead of print]

47. Vogelius IR, Bentzen SM, Maraldo MV, Petersen PM, Specht L

(2011) Risk factors for radiation-induced hypothyroidism. Cancer

117:5250–5260

48. Sinard RJ, Tobin EJ, Mazzaferri EL et al (2000) Hypothyroidism

after treatment for nonthyroid head and neck cancer. Arch Oto-

laryngol Head Neck Surg 126:652–657

49. Jereczek-Fossa BA, Alterio D, Jassem J, Gibelli B, Tradati N,

Orecchia R (2004) Radiotherapy-induced thyroid disorders.

Cancer Treat Rev 30:369–384

50. Garcia-Serra A, Amdur RJ, Morris CG, Mazzaferri E, Menden-

hall WM (2005) Thyroid function should be monitored following

radiotherapy to the low neck. Am J Clin Oncol 28:255–258

51. Hermans R, Pameijer FA, Mancuso AA, Parsons JT, Mendenhall

WM (2000) Laryngeal or hypopharyngeal squamous cell carci-

noma: can follow-up CT after definitive radiation therapy be used

to detect local failure earlier than clinical examination alone?

Radiology 214:683–687

52. Wang J, Takashima S, Takayama F et al (2001) Head and neck

lesions: characterization with diffusion-weighted echo-planar MR

imaging. Radiology 220:621–630

53. Vandecaveye V, De Keyzer F, Nuyts S et al (2007) Detection of

head and neck squamous cell carcinoma with diffusion weighted

MRI after (chemo)radiotherapy: correlation between radiologic

and histopathologic findings. Int J Radiat Oncol Biol Phys

67:960–971

54. Vandecaveye V, Dirix P, De Keyzer F et al (2012) Diffusion-

weighted magnetic resonance imaging early after chemoradio-

therapy to monitor treatment response in head-and-neck squa-

mous cell carcinoma. Int J Radiat Oncol Biol Phys 82:1098–1107

55. Leon X, Quer M, Orus C, del Prado Venegas M, Lopez M (2000)

Distant metastases in head and neck cancer patients who achieved

loco-regional control. Head Neck 22:680–686

56. Ferlito A, Shaha AR, Rinaldo A, Pellitteri PK, Mondin V, Byers

RM (2002) ‘‘Skip metastases’’ from head and neck cancers. Acta

Otolaryngol 122:788–791

57. Engelen AM, Stalpers LJ, Manni JJ, Ruijs JH, van Daal WA

(1992) Yearly chest radiography in the early detection of lung

cancer following laryngeal cancer. Eur Arch Otorhinolaryngol

249:364–369

58. Warner GC, Cox GJ (2003) Evaluation of chest radiography

versus chest computed tomography in screening for pulmonary

malignancy in advanced head and neck cancer. J Otolaryngol

32:107–109

59. Glynn F, Brennan S, O’Leary G (2006) CT staging and surveil-

lance of the thorax in patients with newly diagnosed and recurrent

squamous cell carcinoma of the head and neck: is it necessary?

Eur Arch Otorhinolaryngol 263:943–945

60. Loh KS, Brown DH, Baker JT, Gilbert RW, Gullane PJ, Irish JC

(2005) A rational approach to pulmonary screening in newly

diagnosed head and neck cancer. Head Neck 27:990–994

61. Hsu YB, Chu PY, Liu JC et al (2008) Role of chest computed

tomography in head and neck cancer. Arch Otolaryngol Head

Neck Surg 134:1050–1054

62. Ritoe SC, Krabbe PF, Jansen MM et al (2002) Screening for

second primary lung cancer after treatment of laryngeal cancer.

Laryngoscope 112:2002–2008

63. Buwalda J, Zuur CL, Lubsen H, Tijssen JG, Koole R, Hordijk GJ

(1999) Annual chest X-ray in patients after treatment for lar-

yngeal or oral cancer: only a limited number of second primary

lung cancers detected. Ned Tijdschr Geneeskd 143:1517–1522

(Article in Dutch)

64. Di Martino E, Nowak B, Hassan HA et al (2000) Diagnosis and

staging of head and neck cancer: a comparison of modern

imaging modalities (positron emission tomography, computed

tomography, color-coded duplex sonography) with panendo-

scopic and histopathologic findings. Arch Otolaryngol Head Neck

Surg 126:1457–1461

65. Yoon DY, Hwang HS, Chang SK et al (2009) CT, MR, US, 18F-

FDG PET/CT, and their combined use for the assessment of

cervical lymph node metastases in squamous cell carcinoma of

the head and neck. Eur Radiol 19:634–642

66. Adams S, Baum RP, Stuckensen T, Bitter K, Hor G (1998)

Prospective comparison of 18F-FDG PET with conventional

imaging modalities (CT, MRI, US) in lymph node staging of head

and neck cancer. Eur J Nucl Med 25:1255–1260

67. Leuwer RM, Westhofen M, Schade G (1997) Color duplex echog-

raphy in head and neck cancer. Am J Otolaryngol 18:254–257

68. van der Putten L, van den Broek GB, de Bree R et al (2009)

Effectiveness of salvage selective and modified radical neck

dissection for regional pathologic lymphadenopathy after che-

moradiation. Head Neck 31:593–603

69. Park JJ, Emmerling O, Westhofen M (2012) Role of neck ultra-

sound during follow-up care of head and neck squamous cell

carcinomas. Acta Otolaryngol 132:218–224

70. Nieuwenhuis EJ, Castelijns JA, Pijpers R et al (2002) Wait-and-

see policy for the N0 neck in early-stage oral and oropharyngeal

squamous cell carcinoma using ultrasonography-guided cytology:

is there a role for identification of the sentinel node? Head Neck

24:282–289

71. Smith GL, Smith BD, Buchholz TA et al (2008) Cerebrovascular

disease risk in older head and neck cancer patients after radio-

therapy. J Clin Oncol 26:5119–5125

72. Qureshi AI, Alexandrov AV, Tegeler CH et al (2007) Guidelines

for screening of extracranial carotid artery disease: a statement

for healthcare professionals from the multidisciplinary practice

guidelines committee of the American Society of Neuroimaging;

cosponsored by the Society of Vascular and Interventional Neu-

rology. J Neuroimaging 17:19–47

73. Zimmer LA, Branstetter BF, Nayak JV, Johnson JT (2005)

Current use of 18F-fluorodeoxyglucose positron emission

tomography and combined positron emission tomography and

computed tomography in squamous cell carcinoma of the head

and neck. Laryngoscope 115:2029–2034

74. Schoder H, Fury M, Lee N, Kraus D (2009) PET monitoring of

therapy response in head and neck squamous cell carcinoma.

J Nucl Med 50(Suppl 1):74S–88S

75. Connell CA, Corry J, Milner AD et al (2007) Clinical impact

of, and prognostic stratification by, F-18 FDG PET/CT in head

Eur Arch Otorhinolaryngol

123

Author's personal copy

and neck mucosal squamous cell carcinoma. Head Neck

29:986–995

76. Abgral R, Querellou S, Potard G et al (2009) Does 18F-FDG PET/

CT improve the detection of posttreatment recurrence of head and

neck squamous cell carcinoma in patients negative for disease on

clinical follow-up? J Nucl Med 50:24–29

77. Stokkel MP, Terhaard CH, Hordijk GJ, van Rijk PP (1999) The

detection of local recurrent head and neck cancer with fluorine-18

fluorodeoxyglucose dual-head positron emission tomography.

Eur J Nucl Med 26:767–773

78. Terhaard CH, Bongers V, van Rijk PP, Hordijk GJ (2001) F-18-

fluoro-deoxy-glucose positron-emission tomography scanning in

detection of local recurrence after radiotherapy for laryngeal/

pharyngeal cancer. Head Neck 23:933–941

79. Gupta T, Master Z, Kannan S et al (2011) Diagnostic perfor-

mance of post-treatment FDG PET or FDG PET/CT imaging in

head and neck cancer: a systematic review and meta-analysis. Eur

J Nucl Med Mol Imaging 38:2083–2095

80. Fukui MB, Blodgett TM, Meltzer CC (2003) PET/CT imaging in

recurrent head and neck cancer. Semin Ultrasound CT MR

24:157–163

81. Porceddu SV, Jarmolowski E, Hicks RJ et al (2005) Utility of

positron emission tomography for the detection of disease in

residual neck nodes after (chemo)radiotherapy in head and neck

cancer. Head Neck 27:175–181

82. Yao M, Smith RB, Graham MM et al (2005) The role of FDG

PET in management of neck metastasis from head-and-neck

cancer after definitive radiation treatment. Int J Radiat Oncol Biol

Phys 63:991–999

83. Haerle SK, Schmid DT, Ahmad N, Hany TF, Stoeckli SJ (2011)

The value of 18F-FDG PET-CT for the detection of distant

metastases in high-risk patients with head and neck squamous cell

carcinoma. Oral Oncol 47:653–659

84. de Bree R, van der Putten L, Brouwer J, Castelijns JA, Hoekstra

OS, Leemans CR (2009) Detection of locoregional recurrent head

and neck cancer after (chemo)radiotherapy using modern imag-

ing. Oral Oncol 45:386–393

85. Lee JC, Kim JS, Lee JH et al (2007) F-18 FDG-PET as a routine

surveillance tool for the detection of recurrent head and neck

squamous cell carcinoma. Oral Oncol 43:686–692

86. Wong RJ (2008) Current status of FDG-PET for head and neck

cancer. J Surg Oncol 97:649–652

87. D’Souza G, Kreimer AR, Viscidi R et al (2007) Case-control

study of human papillomavirus and oropharyngeal cancer. N Engl

J Med 356:1944–1956

88. Licitra L, Perrone F, Bossi P et al (2006) High-risk human pap-

illomavirus affects prognosis in patients with surgically treated

oropharyngeal squamous cell carcinoma. J Clin Oncol

24:5630–5636

89. Hafkamp HC, Manni JJ, Haesevoets A et al (2008) Marked differ-

ences in survival rate between smokers and nonsmokers with HPV

16-associated tonsillar carcinomas. Int J Cancer 122:2656–2664

90. Lewis JS Jr, Thorstad WL, Chernock RD et al (2010) p16 positive

oropharyngeal squamous cell carcinoma: an entity with a favor-

able prognosis regardless of tumor HPV status. Am J Surg Pathol

34:1088–1096

Eur Arch Otorhinolaryngol

123

Author's personal copy