Αναζήτηση αυτού του ιστολογίου

Τετάρτη 30 Νοεμβρίου 2022

Nasal mucociliary clearance after extremely low frequency by scintigraphic and histopathologic evaluation

alexandrossfakianakis shared this article with you from Inoreader
Nasal mucociliary clearance after extremely low frequency by scintigraphic and histopathologic evaluation

To investigate effect of exposure to extremely low-frequency magnetic fields (ELF-MFs) on nasal mucociliary clearance in a rat model by rhinoscintigraphy and histopathology assessment. Our rat model has shown the nasal mucosa damage and decreased nasal mucociliary clearance rates and nasal mucociliary transport rate by rhinoscintigraphy as ELF-MFs intensity increases. It may be detrimental to mucociliary function depending on the ELF-MFs intensity.


Objective

To investigate the effect of exposure to extremely low-frequency magnetic fields (ELF-MFs) on nasal mucociliary clearance (MCC) by rhinosintigrapic and histopathological evaluation.

Materials and Methods

The rats were separated into three groups according to ELF-MFs intensity and control group. The exposure groups were standardized for the ELF-MFs of 1, 1.5, and 2 mT emitted by 3 Helmholtz coils for 4 h/day for 30 days. Rhinoscintigraphy was performed to measure nasal MCC. The nasal tissues were examined for edema, inflammation, hyperemia, necrosis, ciliary loss, goblet cell density, and fibroblast proliferation. The data were evaluated statistically (p < 0.05).

Results

Nasal mucociliary clearance rates (NMCR) were calculated as 33.13 ± 5.91% in control, 27.78 ± 4.7% in 1 mT, 22.67 ± 5.43% in 1.5 mT, and 18.11 ± 6.33% in 2 mT. NMCR were decreased with increasing ELF-MFs, in 1.5 and 2 mT groups (p < 0.05) compared to control. Nasal mucociliary transport rate (NMTR) values were found to be 2.17 ± 0.33 mm/min in control, 1.82 ± 0.32 mm/min in 1 mT, 1.46 ± 0.34 mm/min in 1.5 mT and 1.24 ± 0.29 mm/min in 2 mT. NMTR was decreased in the groups exposed to 1.5 and 2 mT (p < 0.05) compared to control. The edema, hyperemia, inflammation, ciliary loss, and goblet cell density were statistically significant differences between control and groups exposed to 1.5 and 2 mT (p < 0.05).

Conclusion

Our rat model has shown nasal mucosa damage and decreased NMCR and NMTR by rhinoscintigraphy as ELF-MFs intensity increases. It may be detrimental to nasal mucosa mucociliary function depending on the ELF-MFs intensity.

Level of Evidence

N/A Laryngoscope, 2022

View on Web

Use of Convolutional Neural Networks to Evaluate Auricular Reconstruction Outcomes for Microtia

alexandrossfakianakis shared this article with you from Inoreader
Use of Convolutional Neural Networks to Evaluate Auricular Reconstruction Outcomes for Microtia

We use convolutional neural networks to objectively assess aesthetic outcomes following ear reconstruction for microtia. This proof of concept paves the way for computer vision to be more broadly applied for us clinically and in outcomes research for reconstructive surgery.


Objectives

The objective of this study is to determine whether machine learning may be used for objective assessment of aesthetic outcomes of auricular reconstructive surgery.

Methods

Images of normal and reconstructed auricles were obtained from internet image search engines. Convolutional neural networks were constructed to identify auricles in 2D images in an auto-segmentation task and to evaluate whether an ear was normal versus reconstructed in a binary classification task. Images were then assigned a percent score for "normal" ear appearance based on confidence of the classification.

Results

Images of 1115 ears (600 normal and 515 reconstructed) were obtained. The auto-segmentation task identified auricles with 95.30% accuracy compared to manually segmented auricles. The binary classification task achieved 89.22% accuracy in identifying reconstructed ears. When the confidence of the classification was used to assign percent scores to "normal" appearance, the reconstructed ears were classified to a range of 2% (least like normal ears) to 98% (most like normal ears).

Conclusion

Image-based analysis using machine learning can offer objective assessment without the bias of the patient or the surgeon. This methodology could be adapted to be used by surgeons to assess quality of operative outcome in clinical and research settings.

Level of Evidence

4 Laryngoscope, 2022

View on Web

DURABILITY OF HUMORAL AND CELL‐MEDIATED IMMUNE RESPONSE AFTER SARS‐CoV‐2 mRNA VACCINE ADMINISTRATION

alexandrossfakianakis shared this article with you from Inoreader

ABSTRACT

Vaccination against the SARS-Cov-2 virus is an effective way to protect against the disease and the severe course of COVID-19. Forty-nine fully vaccinated with mRNA vaccines (BNT162b2 or mRNA-1273) SARS-CoV-2 infection-naïve volunteers aged 33–89 were enrolled in the study. Evaluation of the cellular and humoral immune response was performed within 1 to 3 months (T1) and 6-9 months (T2) after the second injection, and within 2-3 months (T3) after a booster dose. Additionally, a comparative analysis of the specific immune status was made between two age groups - below 60 (n=22) and over 60 (n=27) years. SARS-CoV-2-specific T-cell response was evaluated by IFN-γ-producing spot forming cells (SFCs) using a standardized ELISPOT assay. Virus neutralizing antibodies (VNA) against SARS-CoV-2 were measured by a blocking ELISA test and spike protein specific IgG (S-IgG) and IgA (S-IgA) antibodies - by semi-quantitative ELISA. IFN-γ-producing SFCs, S-IgG, S-IgA a nd VNA significantly decreased 6-9 months after the second dose. After the third injection S-IgG and S-IgA markedly increased compared to T2 and reached the levels at T1. Of note, the highest values of VNA were observed at T3. No differences in the tested immune parameters were found between the two age groups.

Data obtained showed that for a long period - 6-9 months after a full course of immunization with mRNA vaccine, immune reactivity is present, but both cellular and humoral immune responses gradually decrease. The administration of a third dose mainly restores the specific humoral immune response against the SARS-CoV-2 virus.

This article is protected by copyright. All rights reserved.

View on Web

Soft tissue features of peri‐implant diseases and related treatment

alexandrossfakianakis shared this article with you from Inoreader

Abstract

Background

The need for soft tissue grafting at implant sites for preventing and treating peri-implant diseases is a currently investigated and debated topic.

Purpose

The aim of this manuscript is to explore the inflammatory mechanisms at the peri-implant soft tissue compartment, to distinguish the structural components of the peri-implant soft tissue phenotype and their role on peri-implant health, and to appraise the clinical indications and expected outcomes of soft tissue augmentation procedures at peri-implant diseased sites.

Materials and Methods

This narrative review depicts the inflammatory biomarkers and mediators in the peri-implant crevicular fluid utilized to diagnose peri-implant disease and that have been shown to be associated with peri-implant soft tissue phenotype modification and disease resolution. The impact of the peri-implant soft tissue phenotype, involving keratinized mucosa (KM) width, attached mucosa (AM), mucosal thickness (MT), and supracrestal tissue height (STH), on peri-implant health, esthetic, patient's comfort and disease prevention are discussed. The manuscript also illustrates the use of ultrasonography for the detection of peri-implant health/disease and the evaluation of the treatment outcomes following surgical therapies.

Results

Current evidence indicates that soft tissue phenotype modification at implant sites with inadequate KM width, AM and MT can be beneficial for promoting peri-implant health and improving patient's comfort and hygiene procedures. Treatment approaches and outcomes from the available literature on soft tissue phenotype modification in combination with conventional techniques at sites with peri-implant mucositis or peri-implantitis are presented and discussed in detail.

Conclusions

Soft tissue grafting can be beneficial in preventing and treating peri-implant diseases. Clinical recommendations based on the disease, soft tissue phenotype characteristics and bone defect morphology are provided for a comprehensive hard- and soft-tissue-oriented treatment of peri-implant disease.

View on Web

Segmental mandibular advancement for moderate-to-severe obstructive sleep apnoea: a pilot study

alexandrossfakianakis shared this article with you from Inoreader
Segmental mandibular advancement (SMA) consists of a combination of bilateral sagittal split osteotomy, anterior subapical osteotomy with extraction of the first premolars, and genioplasty, to allow an extended advancement of the mandible for the improvement of tongue base obstruction in moderate-to-severe obstructive sleep apnoea (OSA) and to minimize any unfavourable aesthetic change due to the large jaw advancement. The aim of this pilot study was to evaluate the surgical outcomes and complications following SMA in OSA patients. (Source: International Journal of Oral and Maxillofacial Surgery)
View on Web

Paediatric differentiated thyroid carcinoma: a UK National Clinical Practice Consensus Guideline...in Endocrine-Related Cancer...Authors: Sasha R Howard , Sarah Freeston, Barney Harrison, Louise Izatt, Sonali Natu, Kate Newbold, Sabine Pomplun, Helen A Spoudeas, Sophie Wilne, Tom R Kurzawinski , and Mark N Gaze...Correspondence should be addressed to T R Kurzawinski: t.kurzawinski@ucl.ac.uk,DOI: https://doi.org/10.1530/ERC-22-0035,Volume/Issue: Volume 29: Issue 11,Online Publication Date: 07 Sep 2022

Abstract

This guideline is written as a reference document for clinicians presented with the challenge of managing paediatric patients with differentiated thyroid carcinoma up to the age of 19 years. Care of paediatric patients with differentiated thyroid carcinoma differs in key aspects from that of adults, and there have been several recent developments in the care pathways for this condition; this guideline has sought to identify and attend to these areas. It addresses the presentation, clinical assessment, diagnosis, management (both surgical and medical), genetic counselling, follow-up and prognosis of affected patients. The guideline development group formed of a multi-disciplinary panel of sub-speciality experts carried out a systematic primary literature review and Delphi Consensus exercise. The guideline was developed in accordance with The Appraisal of Guidelines Research and Evaluation Instrument II criteria, with input from stakeholders including charities and patient groups. Based on scientific evidence and expert opinion, 58 recommendations have been collected to produce a clear, pragmatic set of management guidelines. It is intended as an evidence base for future optimal management and to improve the quality of clinical care of paediatric patients with differentiated thyroid carcinoma.

Keywords: thyroid; papillary thyroid cancer; follicular thyroid cancer; paediatric cancer; differentiated thyroid cancer

Introduction

Differentiated thyroid cancer (DTC), the most common endocrine cancer in children, occurs in only a small number of patients in the United Kingdom. Approximately, 13 cases per year are seen in children aged less than 15 years and about 105 cases per year in those aged 15–24 years (children, teenagers and young adults UK cancer statistics report 2021: http://www.ncin.org.uk/cancer_type_and_topic_specific_work/cancer_type_specific_work/cancer_in_children_teenagers_and_young_adults/). It increases in frequency with age: of the total cases seen in those aged 0–24 years, 1% occur in those aged 0–4 years, 2% in those 5–9 years, 8% in those 10–14 years, 28% in teenagers 15–19 years and 61% in young adults 20–24 years. There is a female predominance which increases with age: 69% of cases aged 0–14 years and 80% of cases aged 15–24 years are female (children, teenagers and young adults UK cancer statistics report 2021: http://www.ncin.org.uk/cancer_type_and_topic_specific_work/cancer_type_specific_work/cancer_in_children_teenagers_and_young_adults/). The sex ratio is nearly equivalent in children under 10 years of age (Harach & Williams 1995). The overall incidence of thyroid cancer in the 0–24 age group is 1.2 per 100,000 in females and 0.5 per 100,000 in males (National Registry of Childhood Tumours 1991–2010). The incidence appears to be increasing (Vergamini et al. 2014, Golpanian et al. 2016) and this is largely thought to be due to better detection of asymptomatic disease through the increasing use of medical imaging. Less than 1% of thyroid cancers are linked to a risk factor such as previous thyroid irradiation (Lazar et al. 2009). Familial non-medullary thyroid cancer (NMTC) (is described in 3–9% of cases presenting at any age (Moses et al. 2011, Peiling Yang and Ngeow 2016). Genetic predisposition syndromes account for around 5% of NMTCs (Vriens et al. 2009, Richards 2010, Peiling Yang and Ngeow 2016); 95% is accounted for by non-syndromic forms (Peiling Yang & Ngeow 2016, Kamani et al. 2022); 80–90% of DTCs in children are papillary carcinoma and 5–20% follicular carcinoma (Hogan et al. 2009, Papendieck et al. 2011, Mussa et al. 2013, De Jong et al. 2021).

In all age groups in the United Kingdom, overall DTC mortality has decreased by 46% since the early 1970s (Lee et al. 2019). Although the risk of aggressive and recurrent disease in children and young people (CYP) with DTC is higher than in adults (Alessandri et al. 2000, Jarzab et al. 2000, Hay et al. 2010, Al-Qahtani et al. 2015, Lee et al. 2015), the 10-year cause-specific survival is better (Brink et al. 2000, Chow et al. 2004, Steliarova-Foucher et al. 2006, Huang et al. 2012, Shayota et al. 2013). No deaths were reported in children, teenagers and young adults with DTC from 2012 to 2014, and the age-standardised mortality rate for all thyroid cancer per 100,000 population is 0 for the 0–24 age group (data from the National Registry of Childhood Tumours). However, deaths from DTC in childhood may still occur after this age range.

In order to promote best practice standards for the diagnosis and management of thyroid cancers, the American Thyroid Association (ATA) (Haugen et al. 2016), the American Association of Clinical Endocrinologists (Gharib et al. 2016), the National Comprehensive Cancer Network (NCCN) (NCCN 2016) and the British Thyroid Association (BTA)/Royal College of Physicians (Perros et al. 2014) have published guidelines specifically addressing the evaluation, treatment and follow-up of thyroid nodules and DTC in adults.

In most cases, the evaluation, treatment and follow-up of children with thyroid cancer have followed adult guidelines. This approach results in excellent short- and intermediate-term outcomes but may have resulted in overtreatment for a disease with excellent prognosis in CYP, with significant late effects including second malignancy. More recently, specific paediatric guidelines have been provided by the ATA (Francis et al. 2015) and from the Netherlands (Lebbink et al. 2020), but many areas of the treatment of CYP with DTC remain controversial. With recent progress in the management of CYP with this condition, there is an ongoing need for up-to-date age-appropriate guidelines for the management of DTC in CYP that acknowledges the specific needs of this patient group.

Methods

A multidisciplinary Guideline Development Group (GDG) oversaw guideline development. The GDG members, as well as stakeholders and Delphi Consensus panel, are listed in Supplementary Appendix 1 (see section on supplementary materials given at the end of this article). This guideline was developed in accordance with The Appraisal of Guidelines Research and Evaluation Instrument II criteria, as specified in the Children's Cancer and Leukaemia (CCLG) guideline development standard operating procedure, version 6 (https://www.cclg.org.uk/write/MediaUploads/What%20we%20do%20section/CCLG_guideline_SOP_v_6.pdf). The methodology is summarised in Fig. 1. Different stages of the guideline development process were overseen and appraised by the Quality Improvement Committee of the Royal College of Paediatrics and Child Health (RCPCH).

View Full Size
Figure 1

Guideline development process. GDG, Guideline Development Group; GRADE, Grading of Recommendations, Assessment, Development and Evaluations (Guyatt et al. 2011).

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0035

Download Figure
Download figure as PowerPoint slide

Conflicts of interest

All GDG and Delphi Consensus group participants were asked to declare any conflicts of interest as per the National Institute of Health and Care Excellence (NICE 2018) conflicts of interest policy. Conflicts were reviewed and no relevant conflicts were identified. The CCLG provided administrative support throughout the guideline, and the RCPCH provided advice and appraised the guideline at different stages.

Developing the clinical questions

The GDG devised the scope and, subsequently, the Population, Intervention, Comparison, Outcome (PICO) questions (Akobeng 2005), which were sent out to stakeholders to ensure no relevant area had been omitted. Feedback from stakeholders was taken into account by the GDG in the finalisation of the PICO questions, which were used to direct a systematic literature search (Supplementary Appendix 2). Stakeholder involvement : Views from the target population (DTC patients, survivors and their families) were sought via the stakeholder consultation process through various patient support groups including the Royal College of Physicians Young Adult and Adolescent Steering Group. Stakeholders were given the opportunity to comment on the PICO questions being asked and on the final guideline recommendations made to facilitate brevity, clarity and fairness. Recommendation 22 and the section on health benefits (Recommendation 38) are specific to the needs of children and their parents.

Identifying the evidence

Literature searches were conducted as detailed in Supplementary Appendix 3. Of 2565 papers found using this search strategy, 238 papers met the inclusion criteria and were included in the guideline evidence base. The search strategy was limited by prior agreement of the overarching Project Board for all eight National Rare Paediatric Endocrine Tumour Guidelines to publications pertaining to CYP with DTC-related pathology before 19 years of age, including fully published case reports and case series. Full inclusion/exclusion criteria can be found in Supplementary Appendix 3. An additional 26 papers were included following the peer-review process prior to publication.

Reviewing and synthesising the evidence

The quality of evidence identified in the systematic search was appraised using the Grading of Recommendations, Assessment, Development and Evaluations criteria (Guyatt et al. 2011). Details of this process can be found in Supplementary Appendix 4.

Developing recommendations

Where the literature search identified evidence to answer the PICO questions, the GDG made a guideline recommendation. The strength of the recommendation was determined by the trade-off between the potential benefits and potential harms of the recommendation, taking into account the quality of the underpinning evidence. Where an evidence base to formulate recommendations was lacking (i.e., no evidence, contradictory evidence or very low-quality evidence), an expert consensus was necessary. These recommendations were evaluated using a formal Delphi Consensus process (Supplementary Appendix 5) (Okoli & Pawlowski 2004). A recommendation was deemed to have achieved consensus if 70% or more of the Delphi respondents (excluding those who indicated inadequate expertise in the posed question area to be able to comment) supported the recommendation as framed, or with minor modification.

The evidence supporting each recommendation is summarised following the recommendation. In situations where no or low-quality evidence was available, but a Delphi Consensus was not achieved and there was no possibility of near-future comparison trials, the recommendation was made by the GDG Consensus. If there was additionally a clear widespread clinical best practice, that was also used to support strengthening the recommendation. We followed a consistent NICE terminology, using the verbs 'offer' and 'consider' for strong and less strong interventions/actions, respectively and the verbs 'should' for strong and 'may' and 'consider' for moderate recommendations. All recommendations were reviewed by the Project Board and four selected peer experts, prior to guideline publication. Areas highlighted by the literature review and consensus process in which the GDG felt further research would be valuable are reported under the heading, Research Recommendations (Supplementary Appendix 6).

Recommendations

The GDG made 40 recommendations based on the identified evidence. Thirty further recommendations were made based on GDG expert opinion, and these were reviewed by two rounds of a Delphi Consensus process (Supplementary Appendix 5). Following this, 18 recommendations achieved consensus and were included in the guideline. Each recommendation is directly followed by a section discussing the related evidence and citations for this recommendation. One recommendation was made on the basis of GDG Consensus only. Areas highlighted by the literature review and consensus process where the GDG felt further research would be valuable have been proposed as research recommendations (Supplementary Appendix 6).

Health benefits, side effects and risks have been considered for all recommendations on the diagnosis, management and follow-up. The relevant factors are discussed for each recommendation in the general text. Examples include recommendations to assess vocal cord function pre-operatively; diagnosing and managing post-operative complications; safety precautions when using radioiodine, including fertility preservation; and how to follow-up these patients safely to balance over-investigation with timely diagnosis of recurrent disease. The guideline also highlights the need for surgery to take place at high-volume tertiary centres.

External review

The guideline was then externally peer-reviewed by four independent reviewers to improve the quality and applicability of the guideline (see Supplementary Appendix 1). The RCPCH, via the Quality Improvement Committee Clinical Leads for Evidence Medicine and Appraisals, provided advice on guideline development and appraised the draft for quality at different stages. Feedback from the endorsing body and experts was used to complete the final document.

Results

Differentiated thyroid cancer: presentation

Refer CYP with a diffusely or focally enlarged thyroid to an age-appropriate centre with expertise in the management of thyroid disease that can undertake all necessary investigations and treatment (Strong Recommendation, Delphi Consensus 93%)

Care for CYP with suspected or proven DTC in an age-appropriate tertiary centre linked to a paediatric or teenage and young adult oncology centre. A designated specialist clinician who has expertise in the investigation and treatment of patients with thyroid cancer should coordinate multidisciplinary care (endocrinology, surgery and oncology) (Strong Recommendation, Delphi Consensus 100%)

Recommendations 1 and 2 are based on national policy endorsed by NICE Improving Outcomes in Children and Young People with Cancer Guidelines, 2005 (NICE 2018).

Investigate CYP with the following presentations for DTC:

a solitary thyroid nodule (whether symptomatic or incidentally identified on imaging of the neck)

an enlarged nodular thyroid (Strong Recommendation, Very Low-Quality Evidence, Delphi Consensus 100%)

Less common presentations of thyroid cancer include cervical lymphadenopathy, dysphonia, dyspnoea and stridor, as reported in several retrospective cohort studies (Papendieck et al. 2011, Lazar et al. 2009). There is no reported association between nodule size and malignancy risk (Corrias & Mussa 2013, Chiu et al. 2012).

Clinicians should have a higher index of suspicion for DTC in CYP with a history of prior head and neck irradiation or family history of DTC (see Recommendation 10) (Strong Recommendation, Very Low-Quality Evidence, Delphi Consensus 100%)

Retrospective cohort studies of CYP previously treated for cancer with head and neck radiotherapy, especially those where the thyroid gland is included in the treatment field, have been shown to be at increased risk of developing DTC (Sigurdson et al. 2005, Brignardello et al. 2008, Lazar et al. 2009, Taylor et al. 2009, Bhatti et al. 2010, Papendieck et al. 2011, Veiga et al. 2012, Kiratli et al. 2013, Goldfarb & Freyer 2014, Klein Hesselink et al. 2016). Those at risk include patients treated for Hodgkin lymphoma, leukaemia, CNS tumours (Soberman et al. 1991, Solt et al. 2000, Sigurdson et al. 2005, Metzger et al. 2006, Taylor et al. 2009, Rose et al. 2012) and neuroblastoma patients treated with 131I-mIBG (Clement et al. 2015). Environmental radiation, such as occurred after the Chernobyl accident, significantly increases the risk of DTC in CYP (Nikiforov & Gnepp 1994). Endemic hypothyroidism secondary to iodine deficiency may also predispose to DTC (Santos et al. 2017).

Differentiated thyroid cancer: assessment of CYP with thyroid enlargement

5. Consider testing thyroid function in CYP presenting with a thyroid nodule or enlargement (Moderate Recommendation, Low-Quality Evidence, GDG Consensus)

Assessment of thyroid function is standard practice in CYP presenting with thyroid abnormalities such as a nodule or goitre, in order to diagnose hypo- or hyperthyroidism. However, most children with DTC are euthyroid at diagnosis (Papendieck et al. 2011, Suzuki et al. 2016). A correlation between higher TSH levels and risk of malignancy has been identified in one low and four very low-quality cohort studies of children with thyroid nodules (Chiu et al. 2012, Mussa et al. 2013, 2015, Papendieck et al. 2015, Ly et al. 2016) and between TSH levels and cervical lymph node metastases in a very low-quality cohort study of children diagnosed with differentiated thyroid carcinoma (Papendieck et al. 2011). In a CYP with a thyroid nodule found to have a low serum TSH, thyroid scintigraphy Iodine 123 or Technetium 99m pertechnetate can help to determine whether the nodule contains autonomously functioning tissue (Niedziela et al. 2002). Scintigraphy should be reserved only for children whose serum TSH is low. The majority of patients present with normal or high serum TSH thyroid, with ultrasound (US) being the optimal modality to confirm or refute the presence of thyroid nodule (see Recommendation 11).

6. Consider measurement of thyroid autoantibodies in CYP with thyroid enlargement, including those undergoing investigation for a thyroid malignancy (Moderate Recommendation, Delphi Consensus 73%)

Investigation of a CYP with thyroid enlargement will involve the measurement of thyroid autoantibodies to diagnose autoimmune disease. However, children with differentiated thyroid carcinoma may present with co-existing autoimmune thyroid disease (Lazar et al. 2009, Papendieck et al. 2011, Baş et al. 2012). Additionally, a high pre-operative thyroid autoantibody titre in an euthyroid patient indicates an increased risk of hypothyroidism post-surgery (Verloop et al. 2012).

7. Do not routinely measure calcitonin in CYP undergoing investigation of thyroid abnormalities (Strong Recommendation, Delphi Consensus 91%)

Calcitonin should be measured if a diagnosis of medullary thyroid cancer (MTC) is suspected or diagnosed, or if the patient has a genetic predisposition to MTC. Routine measurement of calcitonin in patients who present with a thyroid abnormality is not recommended by these Guidelines, in view of the very low likelihood of detecting MTC in the absence of other clinical indicators and risk of false-positive or indeterminate results, and the associated anxiety provoked by such results.

8. Discuss CYP diagnosed with DTC in both the adult thyroid and paediatric/Teenager and Young Adult (TYA) multidisciplinary team (MDT)s, or in a properly constituted all-age thyroid MDT (Strong Recommendation, Delphi Consensus 87%)

DTC is rare in children. The combination of specialist expertise in the diagnosis and treatment of thyroid cancer in the adult thyroid MDT, in addition to expertise in the care of CYP with malignancy in the paediatric/TYA MDT will provide the best care for young patients.

9. Take a three-generation family history for relevant conditions in CYP with DTC (Strong Recommendation, High-Quality Evidence)

10. Refer to a clinical geneticist for all CYP who have syndromic features (see Table 1), a family history of DTC or a family history of syndromic features associated with DTC (Strong Recommendation, High-Quality Evidence)

Genetic syndromes associated with differentiated thyroid cancer.

SyndromeGermline pathogenic variant and mode of inheritanceType of thyroid cancerSyndromic features noted on clinical examinationFurther clinical features
PTEN hamartoma tumour syndrome

(Includes Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome and PTEN-related Proteus syndrome. These overlapping phenotypes are all known to be due to pathogenic PTEN variants)PTEN

(autosomal dominant)Multinodular

goitre, adenomatous nodules and follicular adenomas

Papillary thyroid cancer (classical and follicular variant)

Follicular thyroid cancerMacrocephaly (OFC >97th centile) and dolichocephaly, learning difficulties, autism and developmental delay, lipomas, vascular features including haemangiomas and arteriovenous malformations, gingival hypertrophy, oral papillomas, facial papules, acral keratoses, palmoplantar keratosis, trichilemmomas, pigmented macules of the glans penis and overgrowth of tissues.Benign and malignant tumours of the breast, colon, endometrium and kidney, adult Lhermitte-Duclos disease due to cerebellar dysplastic gangliocytoma.
Familial adenomatous polyposis (FAP) (includes Gardner syndrome and Turcot syndrome. These overlapping phenotypes are all known to be due to pathogenic APC variants)APC

(autosomal dominant, with 20% cases arising de novo)Papillary thyroid cancer including cribiform pattern subtypeCongenital hypertrophy of the retinal pigment epithelium (CHRPE), congenital absence of teeth, delayed eruption of teeth, dentigerous cysts, supernumerary teeth, odontomas, epidermoid cysts, fibrous dysplasia of the skull, mandibular osteomas, fibromas, desmoid tumours and pilomatrixoma.Hepatoblastoma, medulloblastoma, multiple adenomatous polyps throughout the gastrointestinal tract, principally affecting the colon with high likelihood of malignant transformation, as well as upper GI tract adenomas and adrenal adenomas.
Carney complexPRKAR1A

(autosomal dominant, with 30% cases arising de novo)Papillary thyroid cancer, follicular thyroid cancer and follicular adenomaPale brown to black lentigines of skin, lips and oral mucosa, soft tissue myxomas, schwannomas and epithelioid-type blue nevi.Benign adrenal tumours (primary pigmented nodular adrenocortical disease), pituitary tumours (often somatotropinomas), large cell calcifying Sertoli cell tumours, breast ductal adenoma, osteochondromyxoma and Psammomatous melanotic schwannoma of the nerve sheath.
DICER1DICER1

(autosomal dominant)Multinodular goitre and papillary thyroid cancerNonePleuropulmonary blastoma, ovarian Sertoli-Leydig cell tumours, cystic nephroma, ciliary body medulloepithelioma, botryoid-type embryonal rhabdomyosarcoma, nasal chondromesenchymal hamartoma, pituitary blastoma, pineoblastoma, Wilms tumour and juvenile intestinal hamartomas.
WernerWRN

(autosomal recessive)Papillary thyroid cancer,

Follicular thyroid cancer and anaplastic thyroid cancerShort stature (lack of pubertal growth spurt), cataracts, premature aging, tight atrophic skin, ulceration, hyperkeratosis, pigmentary alterations, regional subcutaneous atrophy, and characteristic 'bird-like facies', hypogonadism, secondary sexual underdevelopment, premature greying and thinning of scalp hair, pes planus and abnormal voice.Malignant melanoma, meningioma, soft tissue sarcomas, leukaemia and pre-leukaemic conditions of the bone marrow, primary bone neoplasms, osteoporosis, soft tissue calcification, evidence of premature atherosclerosis and diabetes mellitus.

Approximately 5% of CYP with familial DTC will have an underlying syndromic genetic predisposition (Peiling Yang & Ngeow 2016). There is high-quality evidence for the association between certain syndromes and the development of DTC (Yamashita & Saenko 2007, Morrison & Atkinson 2009, Richards 2010, Lauper et al. 2013, Rutter et al. 2016) – Cowden syndrome (PTEN hamartoma syndrome) (Marsh et al. 1998, Ngeow et al. 2011, Smith et al. 2011), Familial Adenomatosis Polyposis (FAP) (Kennedy et al. 2014), Carney complex (Stratakis et al. 1997) and Multinodular goitre families (DICER1 pathogenic variants) (Rio Frio et al. 2011, De Kock et al. 2014, Stewart et al. 2019). Putative low-moderate penetrant NMTC susceptibility genes have recently been described in dominant papillary thyroid carcinoma (PTC) families (Fallah et al. 2013, Zivaljevic et al. 2013, Peiling Yang & Ngeow 2016). Werner syndrome, a rare autosomal recessive disease, is known to predispose to DTC (median age of onset 20 years) (Lauper et al. 2013). A CYP with DTC and syndromic features and no family history of the condition may represent a case of de novo presentation or somatic mosaicism. A de novo presentation of FAP is known to occur in 20–25% of cases (Aretz et al. 2004).

Clinical examination of a CYP with DTC should include measurement of their height, weight and head circumference (maximum occipito-frontal diameter), with the results plotted on an appropriate growth centile chart. The clinical features associated with rare syndromic forms of DTC predisposition are summarised in Table 1. Clinicians may be able to identify those patients with an underlying syndromic cause for their DTC by careful examination. However, as the penetrance of these features is variable and many clinical features will develop in older childhood or adulthood, the diagnosis may only become apparent if other family members are also examined. Family history should include details of previous genetic testing if cases of DTC or syndromic causes of DTC, multinodular goitre or thyroidectomy are identified in the patient's relatives. All possible efforts should be made to establish the precise thyroid tumour pathology and that of other relevant tumours previously diagnosed in the family.

The UK Genomic Medicine service strongly supports diagnostic testing in the clinical setting, that is, within the paediatric endocrine, surgical or oncology teams as well as within the genetics service. There are now several genetic panels that can be tested for in patients with paediatric DTC or multinodular goitre within the current test directory (https://www.england.nhs.uk/publication/national-genomic-test-directories/ (v3 April 2022)). Therefore, in the United Kingdom, referral to the clinical genetics service is not a requirement to carry out genetic testing on children with DTC or multinodular goitre, and the MDT can direct early genetic testing with additional referral to clinical genetics if there are variants of interest identified.

Differentiated thyroid cancer: imaging of CYP with thyroid enlargement

11. Undertake neck US in all CYP with thyroid enlargement with normal thyroid function if malignancy is suspected (Strong Recommendation, High-Quality Evidence)

US characteristics, as identified by an experienced head and neck or thyroid radiologist, are important in the differentiation of benign from malignant disease in CYP (Lyshchik et al. 2005, Corrias et al. 2010, Saavedra et al. 2011, Goldfarb et al. 2012, Gupta et al. 2013). US findings that predict an increased risk of thyroid cancer (Fig. 2) include solid nodules, nodules with internal calcification, the presence of enlarged or abnormal lymph node/s, irregular nodule margins, nodules that are taller than wide on transverse view (Al Nofal et al. 2016), hypoechoic nodules and increased intranodular blood flow on colour Doppler (Mussa et al. 2015, Koltin et al. 2016, Moudgil et al. 2016). US can assist to distinguish enlarged reactive lymph nodes, preventing their unnecessary biopsy, from DTC lymph node metastases, which have characteristic US appearances (Abbasian Ardakani et al. 2018).

View Full Size
Figure 2

British Thyroid Association 2014 classification ultrasound scoring of thyroid nodules. Reproduced, with permission, from Perros et al. (2014). Copyright 2014 John Wiley and Sons.

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0035

Download Figure
Download figure as PowerPoint slide

The differential diagnosis of multinodular goitre in CYP includes diffusely infiltrating papillary thyroid cancer that presents with enlargement of a lobe or the entire thyroid, very frequently associated with microcalcifications on neck US (Wang et al. 2022). Multinodular thyroid enlargement in CYP, especially if associated with palpable cervical lymph nodes, in a CYP with normal thyroid function therefore requires investigation by neck US.

The benefits of this recommendation are that the US provides excellent visualisation of structures in the neck without exposure to further ionising radiation and facilitates cytological examination of abnormal findings. There are no anticipated risks or side effects of following this recommendation.

12. Report neck US using the U1–U5 US reporting system as recommended by the British Thyroid Association (Strong Recommendation, Moderate Quality Evidence, GDG Consensus)

The use of the U1–U5 scoring/grading system is recommended for assessing the risk of malignancy in adults with thyroid enlargement (Fig. 2) (Perros et al. 2014). The GDG agreed it was appropriate to apply the same system to CYP undergoing investigation for DTC.

Differentiated thyroid cancer: pre-operative investigation of patients with known DTC

13. Use US-guidance in CYP undergoing fine-needle aspiration (FNA) for the investigation of DTC (Strong Recommendation, High-Quality Evidence)

The BTA and ATA paediatric guidelines both recommend US-guided fine-needle aspiration cytology (FNAC) to minimise rates of indeterminate samples and reduce rates of unsatisfactory samples (Izquierdo et al. 2009, Perros et al. 2014, Francis et al. 2015). As the use of US-guided FNAC is well established in the adult population to accurately identify targets for cytological assessment, it is logical to apply US-guided FNAC in CYP.

14. Offer sedation or general anaesthesia if FNA is required, depending on the needs of the individual child (Moderate Recommendation, Low-Quality Evidence, GDG Consensus)

There is no literature to clearly address the question of age cut-off for tolerating FNAC under local anaesthesia. In children under 10 years of age, and/ or in CYP who are unable to tolerate FNAC under local anaesthesia for other reasons, who require thyroid/neck FNA, sedation or general anaesthesia should be considered (Redlich et al. 2012). While general anaesthesia in a specialist centre carries a very low risk of harm, the associated risks and potential side effects should be discussed with patients and families as for any other procedure requiring general anaesthesia.

15. Undertake US-guided FNA on a thyroid nodule reported on US as U3- indeterminate, U4- suspicious or U5- malignant (Strong Recommendation, High-Quality Evidence)

The utility of pre-operative FNAC of thyroid nodules in CYP in differentiating benign and malignant disease has been evaluated via multiple studies including one meta-analysis (Raab et al. 1995, Khurana et al. 1999, Hosler et al. 2006, Stevens et al. 2009, Altincik et al. 2010, Bargren et al. 2010, Hoperia et al. 2010, Kapila et al. 2010, Gutnick et al. 2012, Monaco et al. 2012, Redlich et al. 2012, Cole & Wu 2014, Rossi et al. 2014, Buryk et al. 2015, Norlen et al. 2015, Partyka et al. 2016, Trahan et al. 2016). In CYP, pre-operative FNAC has high sensitivity and specificity, as well as high-positive predictive and negative predictive values for the diagnosis of malignancy (Stevens et al. 2009, Moudgil et al. 2016), although there is variability in the interpretation of cytopathology findings between institutions (Jia et al. 2021, Vuong et al. 2021, Cherella et al. 2022). In CYP, as in adults, the finding of follicular cytology on FNAC is not able to distinguish follicular adenoma from follicular carcinoma (Smith et al. 2013), and thus diagnostic hemithyroidectomy will be required. Observation with an interval US as an alternative to FNAC can be offered if a solitary sub-centimetre U3 indeterminate lesion is identified.

16. Report cytology samples using the Thy 1–5 grading system. Thyroid cytology must be reported by a cytopathologist with expertise in thyroid disease (Strong Recommendation, Delphi Consensus 93%)

The Royal College of Pathology document recommends thyroid cytology be reported in prose, together with an allocated Thy category (Thy 1−Thy 5) (https://www.rcpath.org/resourceLibrary/g089-guidancereportingthyroidcytology-jan16.htmln) (Table 2). If FNA sampling is inadequate (Thy1), in the absence of clinical or radiological features of concern, US-guided FNA may be repeated between 3 and 6 months (Perros et al. 2014, Francis et al. 2015).

Table 2

Royal College of Pathologists thyroid cytology reporting system.

Thyroid 1Non-diagnostic for cytological diagnosis
Thyroid 1cCystic lesion
Thyroid 2Non neoplastic
Thyroid 2cNon neoplastic, cystic lesion
Thyroid 3aNeoplasm possible-atypia/non-diagnostic
Thyroid 3fNeoplasm possible, suggesting follicular neoplasm
Thyroid 4Suspicious of malignancy
Thyroid 5Malignant

17. Consider diagnostic hemi-thyroidectomy in CYP if there is a discrepancy between clinical and/or radiological features and cytology (Moderate Recommendation, Delphi Consensus 71%)

Benign cytology (Thy2) that does not correlate with clinical/radiological findings of concern, or repeated findings of indeterminate cytology (Thy3f), indicates a need for diagnostic surgery (hemithyroidectomy) (Lale et al. 2015). If cytology demonstrates Thy3a, but clinical/radiological suspicion is assessed by the MDT to be low, US-guided FNA may be repeated between 3 and 6 months. This recommendation is supported by adult thyroid cancer guidelines (ATA & BTA) and ATA guidelines for children with thyroid nodules (Perros et al. 2014, Francis et al. 2015, Haugen et al. 2016). The MDT will need to review the management of patients with cytology that repeatedly demonstrates Thy3a on an individualised basis. There is growing evidence of the potential utility of somatic oncogene analysis to aid management decisions in FNA with indeterminate cytology (Mostoufi-Moab et al. 2018, Pekova et al. 2021, Mollen et al. 2022) (see Recommendation 34).

18. Consider the use of MRI neck with contrast prior to surgery in CYP with known DTC with evidence of local invasion or lymph node metastases (Moderate Recommendation, Low-Quality Evidence, GDG Consensus)

In CYP with DTC and features suggestive of extrathyroidal invasion, lymph node involvement is common (Jarzab et al. 2000, Koo et al. 2009, Hay et al. 2010). In the presence of clinically or radiologically detected nodal disease, for example, bilateral cervical lymphadenopathy, especially with the involvement of lower cervical and supraclavicular nodes, a large or fixed thyroid swelling, or symptoms of vocal cord paralysis (stridor, hoarse voice), further imaging with MRI contrast should be performed pre-operatively. This is particularly due to the importance of imaging the lateral neck and upper mediastinum, as knowledge of the extent of lymph node metastases is vital prior to surgery. Furthermore, local invasion of a tumour into local structures (trachea, oesophagus), while rare, is essential to guide the surgery (e.g., length of surgery, appropriate specialist backup).

Although the incidence of lung metastases in CYP presenting with DTC is much higher than in adults at approximately 12–23% (Vassilopouloii-Sellin et al. 1993, Bal et al. 2004, Leboulleux et al. 2005, Vali et al. 2015, De Jong et al. 2021, Nies et al. 2021), these will be accurately imaged at the staging radioiodine scan (Kim et al. 2011, Mostafa et al. 2016, Jiang et al. 2021), which now includes a SPECT/CT as well as planar scintigraphy as a matter of course. Rarely, large volume metastases visible on a plain chest radiograph may require cover with steroid therapy to avoid issues with flare/oedema with RAI (Fard-Esfahani et al. 2014).

19. Consider the use of pre-operative laryngoscopy to assess vocal cord function in CYP with DTC who have voice symptoms or extra-thyroidal disease or who have had previous neck surgery (Moderate Recommendation, Moderate Quality Evidence)

The Clinical Practice Guideline on improving voice outcomes after thyroid surgery (Chandrasekhar et al. 2013) recommends pre-operative examination of vocal fold mobility in patients with a normal voice when there is thyroid cancer with suspected extra-thyroidal extension or prior neck surgery. Children under the age of 18 years were specifically excluded from the target population of the guideline although the authors state that many of the findings might be applicable. The NCCN and the BTA have recommended pre-operative laryngeal examination for patients with proven or suspected thyroid malignancy.

Reasons given to support the use of pre-operative laryngoscopy are (i) to confirm in the event of a finding of post-operative vocal cord palsy (VCP) that it is indeed newly caused by the procedure and (ii) when a pre-operative VCP is identified, to raise awareness in the patient and surgeon of the risk of bilateral palsy if contralateral surgery is planned. Routine laryngoscopy prior to thyroid surgery performed on adult patients identified a 2.3–2.8% prevalence of pre-operative VCP (Lang et al. 2014, Heikkinen et al. 2019).

Studies that include routine pre-operative (and post-operative) laryngoscopy in paediatric patients prior to thyroidectomy are rare. Machens et al. do not report finding VCP on routine pre-operative laryngoscopy in 230 surgically treated CYP (Machens et al. 2016). A recent study of complication rates after 464 paediatric thyroidectomies reports pre-operative laryngoscopy was performed on patients due to undergoing completion surgery or on those with hoarseness or dysphagia pre-operatively (Baumgarten et al. 2019). The patient group included 178 operations for malignant disease. The paper does not include the number of pre-operative laryngoscopies performed or a finding of pre-operative VCP.

There is increasing evidence in adults that trans-laryngeal US is as effective as laryngoscopy in screening patients pre-operatively for VCP, especially if there is no clinical suspicion (Gambardella et al. 2020). It has very high sensitivity and specificity and is likely to be better tolerated than laryngoscopy, especially in younger children. Moreover, there is recent evidence in favour of the use of intra-operative neuromonitoring, particularly if continuous, to reduce the risk of VCP in CYP (Ritter et al. 2021, Schneider et al. 2021).

Differentiated thyroid cancer: surgical management

20. Surgery in CYP with DTC must be undertaken by a high-volume thyroid surgeon (>30 cervical endocrine procedures per year in adults and children) with collaborative care between adult and paediatric surgeons (Strong Recommendation, Moderate Quality Evidence, GDG Consensus)

Low-quality evidence from the United States indicates that high-volume surgeons (>30 cervical endocrine procedures per year in adults and children) have the best outcomes for CYP with DTC (Sosa et al. 2008, Burke et al. 2012, Breuer et al. 2013, Al-Qurayshi et al. 2016, Baumgarten et al. 2019, Maksimoski et al. 2022), especially in cases with active collaboration between endocrine and paediatric surgeons (Wood et al. 2011). In one US study, the case volume of the endocrine surgeons was an independent predictor of length of stay and costs, as well as reducing complications (Tuggle et al. 2008). Therefore, the risks of not following this recommendation are an increased incidence of short- and long-term complications of thyroid surgery (see Recommendation 22 for more details).

21. The surgical team must be led by a thyroid MDT nominated all-age thyroid surgeon (with paediatric, adolescent and adult experience) (Strong Recommendation, Delphi Consensus 88%)

The GDG agreed that a thyroid surgeon from the adult thyroid MDT nominated to operate on CYP with DTC should lead the designated surgical team. Due to the absence of UK evidence, this question was reviewed by a Delphi Consensus process.

22. Discuss with patients and their carers the risks of thyroid surgery. These include hypocalcaemia (transient or permanent hypoparathyroidism), recurrent laryngeal nerve injury (transient or permanent), post-operative bleed requiring emergency surgery, wound infection and the need for lifelong levothyroxine. If lateral neck lymph node dissection is planned, risks include injury to the spinal accessory/phrenic/sympathetic nerves and lymphatic leak(Strong Recommendation, Low-Quality Evidence, GDG Consensus)

The most commonly reported complications in CYP undergoing thyroid surgery are post-operative hypocalcaemia associated with transient or permanent hypoparathyroidism and injury to the recurrent laryngeal nerve/s (RNL) (transient or permanent hoarse voice, aspiration on swallowing and post-operative chest infection) (Newman et al. 1998, van Santen et al. 2004, Morris et al. 2012). Post-operative bleeding and wound infection are rare (1%) (Hanba et al. 2017, Schneider et al. 2018). Damage to the spinal accessory/phrenic or sympathetic nerves and lymphatic leak are specific complications of lateral neck dissections. Additionally, there is a risk of injury to the external branch of the superior laryngeal nerve which produces more subtle voice changes such as weakness of voice or inability to raise or project the voice.

CYP have higher endocrine-specific complication rates than adults after thyroidectomy (Sosa et al. 2008). Younger children (aged 0–6 years) have higher complication rates than those in mid-childhood (7–12 years) and older CYP (13–17 years). Analysis of nationwide outcomes (from the United States) after thyroidectomy in children <1 year old revealed infection rates of 29.9%, respiratory sequelae in 35.2% and a high incidence of VCP of 14.3% (Hanba et al. 2017).

The incidence of transient hypoparathyroidism ranges from 4.5 to 35%, permanent hypoparathyroidism from 0 to 32%, RLN injury from 0 to 25%, tracheostomy 8% and Horner's syndrome 2–8% (Newman et al. 1998, van Santen et al. 2004, Savio et al. 2005, Massimino et al. 2006, Burke et al. 2012, Morris et al. 2012).

Consent for surgery should be obtained as detailed in Section 3.5.1 – Establish and maintain partnerships with patients in 'Good Surgical Practice' (page 40–43, The Royal College of Surgeons of England 2014; https://www.rcseng.ac.uk/standards-and-research/gsp/).

23. Record in the surgical operation note whether or not the RLNs are dissected and preserved, and the number of parathyroid glands identified, preserved and autografted (Strong Recommendation, Delphi Consensus 100%)

Minimum standards for the content of operative notes are detailed in 'Good Surgical Practice' (page 21–22, The Royal College of Surgeons of England 2014; https://www.rcseng.ac.uk/standards-and-research/gsp/)

24. Offer total thyroidectomy for surgical management of CYP with cytologically proven DTC (Moderate Recommendation, Moderate Quality Evidence)

There are no randomised controlled trials to assess the optimal initial surgical management of DTC in CYP and there is no definitive evidence for the recommendation of more radical vs more conservative surgery. A systematic review (Jin et al. 2015) of seven retrospective case series (489 patients) found no evidence in CYP that overall survival is influenced by total thyroidectomy as compared to less extensive surgery such as hemithyroidectomy. Overall survival in 3861 cases from the National Cancer Database similarly failed to show an advantage from total thyroidectomy (Nice et al. 2015). However, selection bias may account for some of the lack of survival benefits from total thyroidectomy, and in some studies, follow-up may not have been sufficient to draw this conclusion (Nice et al. 2015). Nevertheless, there is some recent evidence to suggest there may be criteria by which to define very-low risk patients who may be candidates for lobectomy rather than total thyroidectomy (Kluijfhout et al. 2017).

PTC: In CYP, PTC is multifocal/bilateral in approximately 65 and 30% of patients, respectively. Lymph node metastasis at the time of diagnosis is evident in 40–90% and 20–30% present with distant metastasis (Dinauer et al. 2008, Rivkees et al. 2011). Total thyroidectomy performed by a high-volume surgeon is the optimal treatment for PTC in CYP (Welch Dinauer et al. 1999, Jarzab et al. 2000, Haveman et al. 2003, Kowalski et al. 2003, Hay et al. 2010, Astl et al. 2014). This surgical technique allows resection of the presenting macroscopic lesion in addition to the frequent microscopic foci of cancer, which can be found in the ipsilateral and contralateral lobes. Total or near-total thyroidectomy is advised to reduce the risk of local recurrence, to enable the use of radioiodine for ablation and therapy and allow subsequent monitoring of serum thyroglobulin (Bignol-Kologu et al. 2000, La Quaglia et al. 2000, Jarzab et al. 2005, Hogan et al. 2009, Mihailovic et al. 2014). As a significant number of CYP with PTC present with distant metastases, total or near-total thyroidectomy is advised in these patients as they will require radioiodine therapy. Management of patients who will require specific radiation protection (for example pregnant mothers) should be by an experienced paediatric molecular radiotherapy service.

Neck recurrence is more common in patients with lymph node involvement or multifocal disease at presentation, extrathyroidal invasion and distant metastases (Palmer et al. 2005, Demidchik et al. 2006, Spinelli et al. 2016). Low-quality studies have identified that lower recurrence rates and decreased need for reoperative surgery are associated with near/total thyroidectomy as compared to more conservative surgical approaches (Welch Dinauer et al. 1999, Jarzab et al. 2000, Haveman et al. 2003, Spinelli et al. 2004, Hay et al. 2010, Astl et al. 2014). Reoperative surgery is associated with higher rates of complications e.g. RLN injury and long-term hypoparathyroidism (Bignol-Kologu et al. 2000).

Follicular variant papillary thyroid carcinoma (FVPTC): FVPTC in CYP has a low risk for bilateral disease and metastasis (Lerner & Goldfarb 2015b , Samuels et al. 2018). The surgical treatment of these patients should be decided on a case-by-case basis.

Follicular thyroid cancer (FTC): In two retrospective series of CYP with FTC which included 50 patients (Enomoto et al. 2013, Spinelli et al. 2016) with a mean follow-up of 23.7 years and 6 years, respectively, cause-specific survival was 100% although the recurrent tumour was identified in three patients (two with distant metastases). Total thyroidectomy (single or two stages) was performed in 21 patients, external beam radiotherapy was used in eight cases and therapeutic RAI in eight cases. Only the patients in the more recent study received levothyroxine for TSH suppression. Total thyroidectomy was advised for patients with multifocal tumours, tumour diameter >4 cm, and for patients with >3 foci of vascular invasion, extrathyroidal tumour extension and distant metastases. Thyroid lobectomy and isthmectomy is appropriate treatment for patients with minimally invasive FTC and none of the above risk factors (Spinelli et al. 2019).

Data from observational studies suggest that the use of total thyroidectomy has increased from 50–60% to 85% in the last 20 years (Raval et al. 2010). Total thyroidectomy is more likely to be performed in high-volume centres (hospital factor) and if large tumours or nodal metastases are present at the time of resection (tumour factors) (Newman et al. 1998, Massimino et al. 2006, Raval et al. 2010, Burke et al. 2012).

Patients, parents and carers should be counselled that total thyroidectomy is associated with higher rates of permanent hypoparathyroidism and RLN injury (Newman et al. 1998, Welch Dinauer et al. 1999, Collini et al. 2006, Massimino et al. 2006, Canadian Pediatric Thyroid Nodule Study 2008, Enomoto et al. 2012) than lesser surgical procedures. This may be especially pertinent in young children (Scholz et al. 2011).

25. Undertake therapeutic central neck dissection in CYP with DTC and confirmed neck lymph node metastases (N1) (Strong Recommendation, Moderate Quality Evidence, GDG Consensus)

Increasing tumour size, extrathyroidal extension and multifocal disease are independent factors associated with nodal metastases in paediatric DTC. If these risk factors are present, children with DTC should undergo careful pre-operative evaluation for evidence of lateral cervical lymph node metastases, and the central compartment should be evaluated intraoperatively (Kim et al. 2017).

Cervical lymph node metastases (N1) are associated with reduced disease-free survival (Wada et al. 2009, Enomoto et al. 2012, Sugino et al. 2015, Spinelli et al. 2018). When therapeutic lymphadenectomy is performed, disease-free survival is equivalent to those patients without pre-operative evidence of lymph node disease (cN0) (Handkiewicz-Junak et al. 2007, Wada et al. 2009).

Central neck dissection (CND) has been demonstrated to reduce the rate of subsequent locoregional disease and increase the efficacy of radioiodine therapies in CYP with known lymph node metastases (Jarzab et al. 2000, Popovtzer et al. 2006, Handkiewicz-Junak et al. 2007). CND and the number of central nodes removed are significantly associated with permanent hypoparathyroidism in CYP with DTC (Ben Arush 2000, Machens et al. 2016) and it is important that the procedure is undertaken by an experienced surgeon.

Skip metastases (lateral neck node metastasis in the absence of central neck node involvement) in PTC are reported in adults in 7–20% (Chung et al. 2009, Park et al. 2012). On that basis, and that of low-quality evidence in CYP, CND should be considered when there is evidence of lateral neck node disease (Vassilopoulou-Sellin et al. 1998, Landau et al. 2000, Demidchik et al. 2006, Handkiewicz-Junak et al. 2007).

26. Consider selective lymphadenectomy of the lateral compartment in CYP with DTC with confirmed lateral neck node metastases (Moderate Recommendation, Moderate-Quality Evidence)

Moderate quality evidence suggests that lateral neck lymphadenectomy is associated with a significantly reduced rate of locoregional recurrence in cases of confirmed lymph node metastasis (Landau et al. 2000, Handkiewicz-Junak et al. 2007).

27. Consider prophylactic central neck node dissection in CYP with Papillary Thyroid Carcinoma (PTC), particularly those with multifocal disease (Moderate Recommendation, Moderate Quality Evidence)

28. Do not consider prophylactic lateral neck lymphadenectomy in CYP with DTC (Moderate Recommendation, Low-Moderate Quality Evidence)

CYP with DTC often have locoregional and distant spread at presentation, especially those with bilateral, multifocal or extrathyroidal disease (Ito et al. 2012, Jin et al. 2015, Balachandar et al. 2016, Jiang et al. 2016, Qu et al. 2016). However, it may be difficult to identify which cases are particularly at risk, in view of the observation that in CYP smaller tumour size may not correlate with reduced metastatic risk (Farahati et al. 1998, O'Gorman et al. 2010). Although the efficacy of radioiodine therapy can be increased after prophylactic node dissection, the MDT should consider carefully the morbidity of the procedure and potential benefits, particularly in younger children, with individualized decision-making (Machens et al. 2016). Recent evidence suggests there is no indication for prophylactic dissection in paediatric patients with FVPTC (Samuels et al. 2018).

29. Monitor serum calcium levels after total/completion thyroidectomy in the peri- and post-operative period until levels are stable and within the normal range (Strong Recommendation, Low-Quality Evidence, GDG Consensus)

Hypocalcaemia consequent to transient or permanent hypoparathyroidism may follow thyroid surgery in CYP; monitoring of serum calcium and intact parathyroid hormone levels is advised in the peri- and post- operative periods (Astl et al. 2014, Freire et al. 2014, Patel et al. 2018). 25-hydroxy vitamin D levels should be measured prior to surgery, and treatment or supplementation should be given as appropriate (Fig. 3).

View Full Size
Figure 3

Calcium monitoring flowchart for total/completion thyroidectomy. CCa, corrected serum calcium; PTH, parathyroid hormone.

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0035

Download Figure
Download figure as PowerPoint slide

30. Monitor calcium levels every 6–12 months in CYP with permanent hypoparathyroidism and stable calcium levels on treatment (Strong Recommendation, Delphi Consensus 92%)

31. Monitor calcium levels more frequently than every 6–12 months in younger patients and during puberty (Strong Recommendation, Delphi Consensus 92%)

The literature search did not identify any published evidence to support the surveillance frequency of CYP with permanent hypoparathyroidism, with biochemical plasma and urine calcium monitoring, or with renal US to monitor for nephrocalcinosis. This was therefore reviewed by the Delphi Consensus group who advised 6–12 monthly assessments. More frequent monitoring is advised in younger CYP and during puberty (Fig. 3).

There is one study with low-quality evidence that calcium and vitamin D supplementation improves bone–muscle proportionality in boys (but not in girls) with hypoparathyroidism following total thyroidectomy (Handkiewicz-Junak et al. 2007). There was no consensus on the utility or frequency of DEXA scanning in this group. If permanent hypoparathyroidism is confirmed CYP should be referred to and regularly reviewed by a paediatric endocrinologist (Schneider et al. 2004). This care should be transitioned to an adult endocrinologist when appropriate.

32. Undertake formal laryngeal examination in CYP who have a post-operative voice change following thyroidectomy and/or central neck dissection (Strong Recommendation, Low-Quality Evidence, Delphi Consensus 90%)

The incidence of post-operative VCP may be underestimated unless routine evaluation is performed. Institutions that practice routine post-operative laryngoscopy report almost two times higher rates of cord palsy than those that do not (Bergenfelz et al. 2008). Studies that report routine post-operative vocal fold examination post thyroidectomy identify transient and permanent VCP in 5.2–12.6% and 1.1–3.3% of patients, respectively (Lo et al. 2000, Joliat et al. 2017, Heikkinen et al. 2019). Routine post thyroidectomy laryngoscopy in CYP has not been reported.

Studies of post thyroidectomy outcomes in the paediatric setting that did not include a routine examination of the larynx include 186 paediatric thyroidectomies over a 20-year period, in which there were three cases (1.6%) of temporary RLN injury, but none were permanent (Chen et al. 2015).

Intraoperative and/or post-operative concern for nerve injury resulted in laryngoscopy in 16 of 464 patients (median age 15 years: range 2–24 years) post-thyroidectomy (Baumgarten et al. 2019). Ten patients were identified with unilateral vocal cord paresis and one patient with bilateral paresis. Two patients had persistent unilateral RLN deficit 6 months post-operatively (0.4%). The 'Kids Inpatient Database' study on thyroidectomy patients aged 1–20 years identified vocal cord paralysis in 1.7% of >2000 patients over a 2-year period. In patients <1 year of age, the incidence of VCP was 14.3% (Hanba et al. 2017).

Guideline recommendations for the adult population state that the surgeon should document whether there has been a change in voice between 2 weeks and 2 months following thyroid surgery (Chandrasekhar et al. 2013, Perros et al. 2014, Haugen et al. 2016, Sinclair et al. 2016).

Differentiated thyroid cancer: histology

33. Report DTC in CYP using The Royal College of Pathologists data set for thyroid cancerhistopathology reports (Strong Recommendation, High-Quality Evidence)

The Royal College of Pathologists' data set for thyroid cancer should be used to report thyroid pathology in CYP (https://www.rcpath.org/resourceLibrary/g089-guidancereportingthyroidcytology-jan16.html; https://www.rcpath.org/profession/guidelines/cancer-datasets-and-tissue-pathways.html).

34. Do not routinely use molecular genetic pathology testing (DNA/RNA tests looking for specific tumour mutations) in the assessment of histopathology samples for diagnostic purposes (Strong Recommendation, Delphi Consensus 93%)

Molecular testing as an ancillary investigation for the diagnosis of thyroid nodules in the adult population is currently under investigation. There is increasing evidence that molecular testing for somatic oncogenes in CYP with DTC may provide additional diagnostic information to aid management (Mostoufi-Moab et al. 2018, Pekova et al. 2021, Mollen et al. 2022), but this should be as advised by histopathology experts within the context of the MDT.

The evidence for mutations in the paediatric population includes studies of alterations of BRAF, RAS, RET/PTC, PAX8/PPAR, ALK, p53, CLIP2 and the sodium-iodide symporter (Suchy et al. 1998, Beimfohr et al. 1999, Fenton et al. 2000, Patel 2002, Hess et al. 2011, Vriens et al. 2011, Sassolas et al. 2012, Leeman-Neill et al. 2013, Henke et al. 2014, Cordioli et al. 2016, Patel et al. 2018, Nies et al. 2021, Stosic et al. 2021). In children, the BRAFV600E mutation has been demonstrated with a variable prevalence in DTC, and it has not been associated with more aggressive tumour behaviour (Penko et al. 2005, Rosenbaum et al. 2005, Vriens et al. 2011, Finkelstein et al. 2012, Henke et al. 2014, Ballester et al. 2016, Gertz et al. 2016, Mostoufi-Moab et al. 2018). RET fusion oncogenes have wide-ranging prevalence reported in CYP with DTC (Nikiforov et al. 1997, Fenton et al. 2000, Ricarte-Filho et al. 2013, Ballester et al. 2016, Gertz et al. 2016), with differences in the specific types of rearrangements linked to radiation-induced and sporadic tumours (Nikiforov et al. 1997). Neurotrophic tyrosine kinase receptor (NTRK) fusion oncogenes are more common in paediatric than adult DTC and were found in recent cohort studies to be associated with a 100% probability of malignancy, as well as more extensive disease and aggressive pathology in CYP with DTC (Prasad et al. 2016, Pekova et al. 2021).

Potential therapeutic options for management of DTC in CYP are continuing to evolve, and the availability of molecular genetic testing, both for inherited germline variants, via panel testing (https://panelapp.genomicsengland.co.uk/panels/171/) and somatic oncogene sequencing for small sequence and structural variants on pathology samples (https://www.england.nhs.uk/publication/national-genomic-test-directories/) is expanding in the UK and elsewhere. While it is rare in CYP with DTC to require non-standard therapy, molecular genetic testing is likely to be increasingly used to direct targeted palliative therapies (see Recommendation 57). Future trials can also consider the inclusion of molecular subtypes into risk stratification (Franco et al. 2022).

35. Report DTC in CYP using the TNM staging system (Strong Recommendation, Delphi Consensus 92%)

The Royal College of Pathologists recommends that TNM version 8 is used for the classification of all thyroid cancer after January 2018 (https://www.rcpath.org/profession/guidelines/cancer-datasets-and-tissue-pathways.html). The TNM system describes well the extent of disease and predicts mortality in adults, but there is less good correlation in CYP (Zimmerman et al. 1988, Dottorini et al. 1997, Chow et al. 2004, Vaisman et al. 2011). In the absence of a CYP-specific scoring system, the GDG recommends following Royal College of Pathologists guidelines. It is important to note that age is an important predictor of CYP in that most, albeit low quality, studies show that young children have a worse prognosis compared to young adult or adolescent patients (Welch-Dinauer et al. 1998, Bal et al. 2001, Borson-Chazot et al. 2004, Powers et al. 2004, Palmer et al. 2005, Jang et al. 2012, Silva-Vieira et al. 2015).

TNM particularly lacks the accuracy to determine disease-free survival in low-risk CYP, due to the high rate of lymph node involvement in this group, leading to the risk of under-diagnosis and under-treatment of this group (Oommen et al. 2008, Wada et al. 2009).

36. Consider recording a prognostic score (TNM) for CYP with DTC in the MDT (Moderate Recommendation, Delphi Consensus 73%)

There is no literature evidence to support this best practice recommendation and it was therefore reviewed by a Delphi Consensus process and 73% of respondents supported this recommendation.

Differentiated thyroid cancer: post-operative management

37. Assess the need for and nature of further treatment in the MDT (adult thyroid cancer and paediatric or TYA MDTs or properly constituted all-age thyroid MDT). This will be determined by the histopathology, TNM stage and risk stratification (Strong Recommendation, Moderate-Quality Evidence, GDG Consensus)

DTC in CYP is not a uniform entity, and the post-operative treatment course, assuming optimal surgery, needs to be individualised on a number of factors including age, stage and completeness of surgery. The patient's post-operative histology should be therefore discussed at an age-appropriate thyroid cancer MDT.

The BTA guidelines (Perros et al. 2014) recommend an adaptation of the ATA risk grouping, while the ATA paediatric guidelines recommend a specific paediatric risk grouping (Francis et al. 2015). There is no evidence that one system is superior to the other.

While both the BTA and ATA offer guidelines for risk stratification after surgery, these are not identical. There is therefore an option for individual clinicians or for MDTs to use one rather than the other, or to take both into account, and offer choices to patients' families. For example, a patient with pT1b pN1a M0 R0 disease may be classified as intermediate risk by the BTA system, and low risk by the ATA system. While radioactive iodine ablation has historically been recommended for the majority of DTC patients following surgery, there is recent evidence that some CYP with low-risk disease may not require it (Sohn et al. 2017, Sung et al. 2017). Further evidence is awaited from randomised trials. With this uncertainty, it would be reasonable to offer either radioactive iodine or close surveillance instead.

Health benefits: Children with DTC have specific paediatric needs and also specific disease-related needs. Expertise in both aspects is required. Their best care therefore requires expert input into both paediatric aspects of care and also disease-related aspects of care. A properly constituted all-age thyroid MDT will bring all the necessary expertise together in one setting, alternatively, this can be achieved by discussion in two different MDTs. There are no anticipated side effects or risks if this recommendation is followed. If children with DTC are considered only in an adult thyroid MDT, the focus may simply be on the disease, with neglect of paediatric aspects of care; if considered only in a paediatric or TYA MDT, then the disease-specific expertise may be lacking, so discussion in both is required to provide the best holistic care.

38. Undertake radioiodine remnant ablation (RAA) according to post-operative risk assessment (Strong Recommendation, Delphi Consensus 87%)

RRA is only applicable in those patients who have had either total thyroidectomy or lobectomy followed by completion thyroidectomy. Following total thyroidectomy, some radioiodine uptake is usually seen within the thyroid bed reflecting normal thyroid remnant tissue. Radioiodine-induced destruction of this remnant is known as 'radioiodine remnant ablation'. This term should not be used to describe the subsequent treatment, which is referred to as radioiodine 'therapy' where the intention is to treat residual, recurrent or metastatic disease. The principles and procedures are similar for the administration of RRA or therapy (Perros et al. 2014).

The advantages of RRA are defined by the BTA (Perros et al. 2014) as follows:

Eradication of all residual thyroid cells post-operatively with subsequent reduced risk of local and distant tumour recurrence

Possible prolonged survival

Reassurance to patients is provided by the knowledge that serum thyroglobulin is undetectable and neck US or diagnostic iodine scan imaging is negative, implying that all thyroid tissue has been destroyed

Increased sensitivity of thyroglobulin monitoring, facilitating early detection of recurrent or metastatic disease

Increased sensitivity of subsequent iodine scanning if required

Following definitive surgery, CYP with DTC will fall into one of the following groups:

No indication for RRA – observation only

Uncertain indication for RRA, management discussed with patient and family, with an individualized approach and consideration for trial entry.

Definite indication for RRA and then, follow-up and dynamic risk stratification.

Definite indication for RRA and subsequent radioactive iodine therapy administration

The indications for RRA are mainly TNM stage dependent with additional information from the pathological risk factors also contributing to the decision-making (see Table 3). The BTA guidelines define the indications for RRA in adults with DTC into three groups based on available evidence; 'no indication', 'definite indication' and 'uncertain indication' (Fig. 4) (Perros et al. 2014).

View Full Size
Figure 4

Decision-making flow chart for use of RRA, adapted from British Thyroid Association guidelines. RRA, radioiodine remnant ablation. Modified, with permission, from Perros et al. (2014). Copyright 2014 John Wiley and Sons.

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0035

Download Figure
Download figure as PowerPoint slide

Table 3

Comparison of risk stratification systems in British Thyroid Association (Perros et al. 2014) and American Thyroid Association Paediatric (Francis et al. 2015) guidelines for DTC.

Risk groupBTA-adapted ATA guidelinesATA Paediatric guidelines
Low riskNo local or distant metastases

All macroscopic tumour has been resected, that is, R0 or R1 resection (pathological definition)

No tumour invasion of loco-regional tissues or structures

The tumour does not have aggressive histology (tall cell, or columnar cell PTC, diffuse sclerosing PTC, poorly differentiated elements), or angioinvasionDisease grossly confined to the thyroid with N0/Nx disease or patients with incidental N1a disease (microscopic metastasis to a small number of central neck lymph nodes)
Intermediate riskMicroscopic invasion of tumour into the perithyroidal soft tissues (T3) at initial surgery

Cervical lymph node metastases (N1a or N1b)

Tumour with aggressive histology (tall cell, or columnar cell PTC, diffuse sclerosing PTC, poorly differentiated elements) or angioinvasionExtensive N1a or minimal N1b disease
High riskExtra-thyroidal invasion

Incomplete macroscopic tumour resection (R2)

Distant metastases (M1)Regionally extensive disease (extensive N1b) or locally invasive disease (T4 tumours), with or without distant metastasis

Most CYP with DTC present with locally advanced disease, with TNM tumour staging of T3 or T4, and/or N1 (Machens et al. 2010, Fridman et al. 2012, Markovina et al. 2014) and it is far less common to see microcarcinoma in the paediatric population (Lerner & Goldfarb 2015a ). It is likely that most CYP with DTC will fall into the 'definite' or 'uncertain' indication groups on the basis of the BTA classification. Those falling into the uncertain group are likely to have RRA recommended following case review due to greater rates of high-risk pathological features (as described in Table 3) seen in this population.

Following RRA, previously unknown disease status, for example unsuspected metastatic disease, may be detected on the post-ablation scan, or an incomplete response on dynamic risk assessment may indicate the need for subsequent radioactive iodine therapy.

However, evidence to support the use of RRA, particularly in early intra-thyroidal disease (cT1-T2 cN0 cM0), is limited (Newman et al. 1998, Welch Dinauer et al. 1999, Jarzab et al. 2000, Chow et al. 2004, Demidchik et al. 2006, Handkiewicz-Junak et al. 2007). In CYP, there is low quality and conflicting evidence on the impact of RRA on disease-free survival (Powers et al. 2003). Evidence on whether RRA is associated with increased risk of secondary malignancy is to date conflicting (Newman et al. 1998, Welch Dinauer et al. 1999, Jarzab et al. 2000). Data from the Iodine or Not study in which patients aged 16 or older with low-risk thyroid cancer were randomised to receive RRA or not, will provide further guidance as to which patients can safely avoid RRA (Mallick et al. 2012a ). Until this evidence is available, the standard UK approach in CYP is to treat any localised disease with definite or uncertain indications with RRA. However, there is room for individualisation of decision-making for the uncertain indications group (Perros et al. 2014) (see Fig. 4).

The ATA adult guidelines consider the use of post-operative thyroglobulin and US in relation to the need for RRA. This is on the basis that in adults, a post-treatment (surgery and RRA) stimulated thyroglobulin level <2 ng/mL and/or undetectable basal serum thyroglobulin measured with a highly sensitive assay are strong indicators of definitive cure and a very low risk of recurrent disease. A negative US scan adds to the sensitivity of this (Pacini et al. 2003, Smallridge et al. 2007, Spencer et al. 2010).

The optimal cut-off value or whether the stimulated or unstimulated thyroglobulin level should be used has not been defined, either in adults or children. It is critical to have a full discussion with the family to ensure all options and the risks and benefits of surveillance or treatment are conveyed.

The timing of RRA post-thyroidectomy is governed more by the availability of facilities and convenience for the patient than the biology of the tumour. There is no evidence that a delay of 1–2 months changes the outcome. Factors around schooling, childcare for siblings and preparing the child for a period of separation from family usually contribute to the selection of date for treatment. NHS cancer waiting targets stipulate treatment should be administered within 31 days of the decision to treat, but this is not based on the biology or outcomes of the disease.

While this recommendation is strongly made, it is important to acknowledge that decision-making is not always easy, especially when the indications for treatment are uncertain. Individual clinicians and families may have preconceived ideas, and families may have been influenced by what other clinicians have said, or by what they have read about the disease and its treatment. In these circumstances, when faced with the option of either radioactive iodine ablation or surveillance, it is the responsibility of the MDT to approach the family with options and to have a full and frank discussion of the uncertainties surrounding the evidence base. Some families will wish to be active participants in decision-making and will not wish to be told that they 'must' follow a certain course of action. Other families may feel unable to contribute to the decision-making, possibly for fear of making the wrong choice, and would like the clinician to make a definite decision. While families typically make the decisions on behalf of younger children, older teenagers who are Gillick competent, should be encouraged to express their opinions and contribute actively to decision-making where there are reasonable options.

39. Radioiodine must be prescribed and administered by professionals experienced in the dosing and administration of radioiodine in CYP (Strong Recommendation, High-Quality Evidence)

This recommendation ensures the least risk of the incorrect treatment being given. There is also a statutory responsibility in the prescription of radioactive iodine, as regulated by the Administration of Radioactive Substances Advisory Committee of the Department of Health and Social Care. There are no anticipated risks or side effects if this recommendation to limit prescribing and administration to experienced professionals is followed, but there is a significant risk of medication errors if this recommendation is not followed.

40. Administer radioiodine to children less than 16 years within a paediatric oncology centre with 24 h medical and nursing care (Strong Recommendation, High-Quality Evidence)

Children aged less than 16 years should receive radioiodine within a paediatric oncology centre with 24 h paediatric medical and nursing care. Older teenagers, from the age of 16 up to their 19th birthday, should receive care in a designated principal treatment centre for young people. From the age of 19–24 years (up to the 25th birthday), young adults should be offered radioiodine in a designated teenage and young adult cancer service but may elect for care in an adult environment that may be closer to home.

These recommendations are based on the Royal College of Radiologists Good Practice Guide for Paediatric Radiotherapy (https://www.rcr.ac.uk/system/files/publication/field_publication_files/bfco182_good_pract_paed_rt_second_ed.pdf), the NHS England: 2013/14 NHS Standard Contract for Cancer: Teenagers & Young Adults Section B Part 1 - service specifications (https://www.england.nhs.uk/wp-content/uploads/2013/09/b17.pdf), and also guidelines from the Intercollegiate Standing Committee on Nuclear Medicine (https://www.rcr.ac.uk/system/files/publication/field_publication_files/bfco199-icscnm-molecular-radiotherapy-guidance.pdf).

While there is a strong recommendation for younger children to be treated in a paediatric environment, older teenagers (over 18 years) should have the option to be treated in a specialist TYA centre with all the appropriate support for people in that age group or to be treated in an 'adult' centre which lacks TYA support.

41. Consider adjustment of the radioiodine activity for thyroid ablation and therapy based on the size of the CYP (Moderate recommendation, Low-Quality Evidence, Delphi Consensus 78%)

There are no standardised activities of RRA for children and there is no data to guide this. Following two randomised controlled trials in adult patients, HiLo (Mallick et al. 2012b ) and Estimabl 1 (Schlumberger et al. 2012), the standard activity for RRA in adults has reduced from 3.7 to 1.1 GBq for patients at low risk of recurrence of their thyroid cancer. In higher-risk diseases, 3.7 GBq remains the standard administered activity.

Historically the same activity has been considered for children and adults, but some adaptation according to body weight, body surface area and age has been applied by many specialists (Handkiewicz-Junak et al. 2007, Pawelczak et al. 2010), and Delphi Consensus and stakeholder review both gave further support for this practice.

For subsequent radioactive iodine for therapy in the setting of persistent or recurrent disease the recommended empirical administered activity is 5.5 GBq in adults (Perros et al. 2014). As with RRA, there is no good data for the adjustment of activity for children. Although there is interest in the dosimetric prescription of radioiodine, there is no evidence to date to show superiority over empiric activity prescription. Reports have suggested that treatment with at least 200 MBq/kg (5.4 mCi/kg) is possible without a risk of exceeding bone marrow tolerance limits (Verburg et al. 2011). Many patients will tolerate much higher activities. However, in children with extensive metastatic disease whole-body dosimetry may be employed to ensure total blood dose does not exceed 2 Gy and that the whole-body retention at 48 h does not exceed 4.44 or 2.86 Gy (in the case of no or miliary lung metastases respectively) (Verburg et al. 2011, Verburg et al. 2013). CYP with DTC, particularly those with pulmonary metastases and coexisting micronodular disease, often show excellent RAI uptake and thus may be more sensitive to 131I therapy than adults (Xu et al. 2016). In conclusion, there is no high-quality evidence to advise for or against the prescription of radioiodine based on empiric activity or informed by dosimetry. GDG Consensus opinion suggests that experts prescribe RRA as an empiric activity and reserve dosimetric methodology for patients with extensive disease and repeated therapies in centres with this expertise.

For the majority of patients, there are very few complications of radioiodine. The long-term toxicities are believed to be related to the absorbed radiation dose as a function of the administered activity and therefore the risk of toxicity will increase with cumulative radiation dose and with cumulative administered radioiodine activity. The risks of RRA are relatively low following a single administration. While the majority of DTC patients are teenagers for whom the adult activity is appropriate, very small children may get a good response from a size-adjusted dose and avoid the risks of radiation exposure beyond that which is necessary. There are no anticipated risks or side effects if this recommendation is followed.

42. Perform a blood or urine pregnancy test on all post-menarchal female patients prior to radioactive iodine administration (Strong Recommendation, Moderate-Quality Evidence, GDG Consensus)

The Royal College of Radiologists Good Practice Guide for Paediatric Radiotherapy (https://www.rcr.ac.uk/system/files/publication/field_publication_files/bfco182_good_pract_paed_rt_second_ed.pdf) states that radioactive iodine may not be administered to pregnant women. Post-pubertal girls should be advised to avoid pregnancy for 6 months after radioiodine (although it may be wise to advise to wait until post-risk assessment at 9–12 months as this will determine response to ablation and whether further treatment is required).

43. Consider fertility preservation with post-pubertal CYP if they are likely to receive more than two administrations of radioiodine (including the ablation administration) (Moderate Recommendation, Delphi Consensus 75%)

A single ablation dose of radioiodine should have no effect on male or female fertility (Landau et al. 2000). Sperm banking should be discussed with all post-pubertal males if they are likely to receive more than two administrations of radioiodine (including the ablation administration) (Wallace 2011). Referral to fertility units for expert advice may be warranted in young women with multiple treatments with radioiodine. Delphi Consensus supported this recommendation, although emphasised that the risks of infertility are low in those patients not requiring high cumulative activities of radioiodine.

While it is important to discuss fertility preservation options with post-pubertal patients and their families, the absolute risk of fertility impairment is low, and following discussion, patients and families should have the option of undergoing fertility-preserving features, or not.

44. Consider preparing CYP for radioactive iodine (RRA or therapy) with either thyroid hormone withdrawal or recombinant thyroid stimulating hormone (Moderate Recommendation, Moderate-Quality Evidence)

Historically, radioactive iodine was always administered after thyroid hormone withdrawal. Typically, levothyroxine is stopped 28 days prior to radioactive iodine administration, and/or liothyronine 10 days before. Most children tolerate thyroid hormone withdrawal well and reach the target TSH of ≥30 IU for radioiodine administration without problems (Kuijt & Huang 2005). More recently, recombinant TSH (Thyrogen) has become the standard of care in adult practice (Mallick et al. 2012b ). However, it is not licensed for use in children, and the data relating to its effectiveness have mostly related to the adult population. There is now increasing retrospective data suggesting the safety and efficacy of recombinant TSH in the paediatric population (Iorcansky et al. 2005, Luster et al. 2009, Rosario et al. 2012, Handkiewicz-Junak et al. 2015).

Uptake of radioactive iodine is poor without a raised TSH level. This recommendation ensures the maximum uptake of radioactive iodine and gives the best chance of successful treatment. While thyroid hormone withdrawal results in a feeling of tiredness and lethargy and may be associated with abnormal kidney function indicators on blood tests, thyrotropin alfa does not cause any significant side effects. However, two intramuscular injections are required.

45. Advise a low-iodine diet prior to radioiodine treatment in CYP with DTC (Strong Recommendation, High-Quality Evidence, Delphi Consensus 100%)

The promotion of low-iodine diets prior to radioiodine treatment in CYP with DTC is supported by adult (Perros et al. 2014) and paediatric guidelines (Pluijmen et al. 2003, Francis et al. 2015) and our Delphi Consensus. A high dietary iodine intake may reduce the therapeutic efficacy of radioactive iodine, and therefore a low iodine diet for 14 days before radioiodine treatment should maximise the chance of successful ablation or therapy (https://www.btf-thyroid.org/low-iodine-diet).

46. Perform a whole-body iodine 131 scan following RRA (Strong Recommendation, High-Quality Evidence)

Following RRA, a whole-body iodine 131 scan (preferably SPECT/CT with the CT component focusing on the neck and thorax, and any other body areas with possible abnormal uptake on planar scans) is indicated (Bal et al. 2004, Kim et al. 2011). This scan permits further staging of the disease, which may identify the presence of unsuspected distant metastases requiring subsequent therapy (Mostafa et al. 2016, Jiang et al. 2021). This will demonstrate localisation of uptake in the thyroid bed, thyroglossal tract remnants, cervical lymph nodes and pulmonary or other distant metastases. This is therefore an essential investigation for the complete staging of the disease and to predict prognosis. There are no side effects or risks anticipated if this recommendation is followed. There is a risk of missing metastatic disease if the recommendation is not followed.

47. Investigate residual disease on post-RRA 131 scan with additional imaging. Further management will be determined by imaging findings(Strong Recommendation, High-Quality Evidence)

Neck uptake within cervical lymph nodes requires anatomical imaging to localise uptake with SPECT CT/US/MRI as available to assess for macroscopic disease (Antonelli et al. 2003, Bal et al. 2004, Kim et al. 2011). If residual disease or distant metastases are identified, the CYP should be referred back to the MDT for consideration of further treatment, surgical or RAI. All other patients will then proceed to dynamic risk stratification (see the subsequent paragraphs).

Differentiated thyroid cancer: follow-up

48. Perform a dynamic risk assessment following RRA. Use this to guide further management and ongoing follow-up (Strong Recommendation, High-Quality Evidence)

At 9–12 months post-surgery and RRA, response to treatment should be assessed. If the suppressed thyroglobulin is undetectable, an US of the thyroid bed and bilateral neck together with the thyroglobulin following TSH stimulation should be performed. This is known as dynamic risk stratification, as recommended in the BTA and ATA Paediatrics guidelines (Perros et al. 2014, Francis et al. 2015). This assesses the response to treatment in low-risk diseases. There is very low-quality evidence that recombinant human TSH-stimulated thyroglobulin level is useful for disease surveillance in CYP (Hoe et al. 2006).

Patients are allocated to one of three groups following dynamic risk stratification: excellent response (no evidence of disease), indeterminate response (biochemical evidence of disease only) and incomplete response (imaging/structural evidence of disease with or without biochemical evidence) (see Table 4). These investigations allow an assessment of the completeness of ablation and facilitate allocation to a risk group for purposes of deciding the level of TSH suppression required and the frequency and intensity of follow-up going forward. There are no anticipated side effects or risks if this recommendation is followed. If the recommendation is not followed, patients may be under-treated, with the risk of disease progression or overtreated with the risk of treatment-related morbidity.

Table 4

Follow-up schedule for management of CYP with DTC.

Baseline risk groupDynamic risk assessment
Excellent responseIndeterminate responseIncomplete response
Low-risk(a) 6–12 monthly follow-up.

(b) At least 5 years.

(c) Clinic visit, TFTs and Tg. No routine imaging.

(d) TSH in normal range.(a) 6 monthly follow-up.

(b) At least 5 years – possibly longer depending on imaging and Tg trend. Consider repeating DRA at 5 years.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly if abnormal – increasing to annually if stable over time.

(d) TSH suppressed for at least 5 years.Consider further treatment in MDT depending on DRA findings. If active surveillance chosen over further treatment:

(a) 3–6 monthly follow-up.

(b) At least 5 years – more likely longer depending on imaging and Tg trend. Consider repeating DRA at 5 years.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly if abnormal – increasing to annually if stable over time.

(d) TSH suppressed indefinitely, unless repeat assessment shows improvement.
Intermediate risk(a) 6 monthly follow-up.

(b) at least 5 years.

(c) Clinic visit, TFTs and Tg. Annual US.

(d) TSH suppression may be slightly relaxed: target low-normal range.(a) 6 monthly follow-up.

(b) at least 5 years – possibly longer depending on imaging and Tg trend. Consider repeating DRA at 5 years.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly if abnormal – increasing to annually if stable over time.

(d) TSH suppressed for at least 5 years.Consider further treatment in MDT. depending on DRA findings. If active surveillance chosen over further treatment:

(a) 3–6 monthly follow-up.

(b) At least 5 years – more likely longer depending on imaging and Tg trend. Consider repeating DRA at 5 years.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly if abnormal – increasing to annually if stable over time.

(d) TSH suppressed indefinitely, unless repeat assessment shows improvement.
High-risk(a) 3–6 monthly follow-up.

(b) At least 10 years.

(c) Clinic visit, TFTs and Tg. US 6 monthly for 2 years, then annually to 5 years.

(d) TSH <0.1 to 10 years.(a) 3–6 monthly follow-up.

(b) At least 10 years – possibly longer depending on imaging and Tg trend. Consider repeating DRA at 5 years.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly – increasing to annually if stable over time.

(d) TSH <0.1 to 10 years.Consider further treatment in MDT depending on DRA findings. If active surveillance chosen over further treatment:

(a) 3–6 monthly follow-up.

(b) At least 10 years, probably lifelong.

(c) Clinic visit, TFTs and Tg. Repeat US initially 6 monthly if abnormal – increasing to annually if stable over time. Other imaging e.g. CT chest may need to be repeated periodically if clinically indicated.

(d) TSH <0.1 to at least 10 years, consider need for life-long suppression if there is still evidence of controlled disease.

DRA, dynamic risk assessment.

49. Use neck US as the first-line imaging modality for post-operative follow-up of CYP with DTC (Strong Recommendation, Moderate-Quality Evidence, GDG Consensus)

In CYP with DTC, US is sufficient for evaluation of loco-regional involvement in follow-up (Vali et al. 2015), with sensitivity of 85.7%, specificity of 89.4%, negative predictive value of 94.4% and positive predictive value of 75%. Neck US can be used to pinpoint the anatomic site of lymph node metastases (Antonelli et al. 2003). In adults, the combination of neck US and FNAC has been shown to detect cases of lymph node metastasis and local recurrence not found by whole-body scan or serum thyroglobulin determination (Durante et al. 2013). Neck US is also useful, particularly in young children, post-operatively to help differentiate pathological from reactive lymph nodes.

50. Measure thyroglobulin antibody in conjunction with serum thyroglobulin (Strong Recommendation, High-Quality Evidence)

Even very low thyroglobulin antibody concentrations can interfere with assay results and thyroglobulin antibodies are found in up to 25% of adult patients (Spencer et al. 1998, Vali et al. 2015, Haugen et al. 2016). Each specimen sent for thyroglobulin measurement requires concomitant thyroglobulin antibody testing because thyroglobulin antibody status can change over time. As variability exists between different thyroglobulin assays, there is a need to use the same assay for serial measurements. Thyroglobulin antibody titres can also correlate with disease burden.

51. Use the baseline risk grouping and subsequent dynamic risk stratification to determine the frequency and duration of follow-up (Strong Recommendation, Moderate-Quality Evidence, GDG Consensus)

There is a lack of high-level paediatric evidence on which to make clear-cut and highly detailed follow-up recommendations, but it is reasonable and pragmatic to base the long-term follow-up schedule of CYP evidence from adults with DTC. Prognostic stratification will be based on both the baseline risk group and also the results of dynamic risk assessment after treatment (see Table 4). CYP will be followed up until transition to adult services, and for low-risk adult patients, a decision can be taken about discharge to primary care. Life-long follow-up is advised in high-risk groups given that recurrence of DTC can occur 40 years after the initial disease (Landau et al. 2000, Hay et al. 2010).

The suggested follow-up schedule is given in Table 4, modified from BTA (Perros et al. 2014) and ATA Paediatrics (Francis et al. 2015) guidelines and recent review (Lee et al. 2019). Follow-up of patients will be in one of the three groups. These are, as above, no evidence of disease (excellent response), biochemical evidence of disease only (indeterminate response), and imaging/structural evidence of disease with or without biochemical evidence (incomplete response). The purpose of surveillance is to detect evidence of relapse/progression at a time point when further intervention may be of value and to ensure TSH levels are optimised to reduce the risk of recurrence.

If no structural disease is present and stimulated thyroglobulin is not detectable, this represents an excellent response to treatment, and follow-up intervals can be extended to 6 months during childhood (92% agreement on Delphi Consensus) and relaxation of TSH suppression may be considered (as per Table 4).

In patients with low-level TSH-stimulated thyroglobulin (<10 ng/mL), continued follow-up with serial TSH-suppressed thyroglobulin is indicated. The role of imaging with US in this situation is unclear, and this should be an individualised decision by the treating clinician.

If the unstimulated thyroglobulin is detectable, an US should be performed to localise persistent disease that may be surgically resectable (Vali et al. 2015). If no structural disease is present, a therapy dose of RAI may be considered (Francis et al. 2015).

Following this recommendation allows for personalisation of care, based on the extent of disease at the time of diagnosis and the response to treatment. This facilitates individualised follow-up schedules, which ensure those at the highest risk are followed more intensively to detect progression, while those at low risk are spared unnecessary hospital visits.

Differentiated thyroid cancer: metastatic, recurrent or persistent disease

52. Use serial thyroglobulin measurement with additional imaging if required to monitor CYP with DTC (Strong Recommendation, Moderate-Quality Evidence, GDG Consensus)

Serum thyroglobulin may continue to be detectable following RRA but may decline over time without additional therapy (Dottorini et al. 1997, Biko et al. 2011). A rise in thyroglobulin or thyroglobulin antibodies should trigger initial confirmation with repeat measurement within 2 months, followed by investigation with US of the thyroid bed and neck to exclude disease recurrence (Kirk et al. 1992, Xu et al. 2016). If neck US is normal, additional imaging such as CT or MRI can be considered to look for distant metastatic disease, especially lung metastases, if thyroglobulin levels suggest the disease may be present at a distant site. 123-I whole body scintigraphy and SPECT/CT may be added if the thyroglobulin measurement is considered to be unreliable because of a high antibody titre (Kim et al. 2011).

Sometimes the low administered activity of 123-I used for diagnostic imaging, and the resolution of the imaging techniques used, may result in small-volume disease being overlooked or mistaken for being iodine resistant. The use of 18F-FDG PET/CT may be a helpful additional investigation as it has high diagnostic accuracy for the detection of recurrent and/or metastatic diseases in DTC patients with thyroglobulin elevation and negative iodine scintigraphy (Larg et al. 2019, Qichang et al. 2019, Wang et al. 2021).

53. Consider further surgical resection for persistent local structural disease (Moderate Recommendation, GDG Consensus)

Structural disease refers to a definite abnormality on imaging, identifying that cancer has infiltrated anatomical structures such as jugular vein, trachea or oesophagus, or metastatic lymph nodes. If structural disease is detected on the neck US, MDT discussion about the role of further surgery is recommended.

54. Consider therapeutic radioiodine after further surgical resection (Moderate Recommendation, Moderate-Quality Evidence)

55. Administer radioiodine as first-line treatment for unresectable metastatic disease in CYP (Strong Recommendation, High-Quality Evidence)

Therapeutic choices after further surgical resection should be individualised, discussed with the MDT and agreed upon with the patient/family who may have strong views. The use of radioiodine should be considered and would normally be regarded as indicated if, following surgery, there is imaging or biochemical evidence of residual disease which was not felt to be amenable to surgery; whereas in the absence of such evidence, a policy of careful observation might be regarded as a safe and more conservative approach, although there is no evidence to show that the use of radioactive iodine is necessarily wrong. Most DTC in this age group responds well to radioactive iodine therapy and repeated therapy doses of radioactive iodine can be used after further surgical resection and to treat metastatic disease as long as a response is seen (Biko et al. 2011, Verburg et al. 2013). It is rare for DTC in CYP to become radioiodine refractory.

56. Consider chest imaging (chest X-ray or chest CT) in patients with high-risk disease or those with evidence of persistent or recurrent disease to diagnose and monitor metastatic lung disease (Moderate Recommendation, Moderate-Quality Evidence)

Chest X-ray is used to visualise macroscopic lung metastases. There remains controversy as to whether iodinated contrast should be used if a CT scan is undertaken due to concerns that this may lead to a delay in radioiodine therapy (Perros et al. 2014). If contrast enhancement will help define the extent of disease and help management decision-making, it should be used. Evidence is not clear as to the washout time required following intravenous contrast to prevent 'stunning' and reduced uptake of radioiodine therapy, but adult guidelines suggest waiting 8 weeks (Perros et al. 2014).

57. Consider the use of palliative targeted therapy in CYP with progressing radioiodine refractory DTC (Moderate Recommendation, Moderate-Quality Evidence)

Radioiodine refractory disease includes either the presence of at least one lesion that does not take up I-131 or clinical evidence that I-131 is no longer providing benefit. There is no evidence that traditional chemotherapeutic agents are an effective treatment of radioiodine refractory DTC in CYP. Targeted agents, sorafenib and lenvatinib, have been licensed more generally for the treatment of radioiodine refractory disease in adults but have not been proven in the paediatric population. In young adults over the age of 16 with progressing (i.e., with radiographic evidence of disease progression), radioiodine refractory DTC, sorafenib and lenvatinib can be considered as per marketing approval and based on phase III data from DECISION (Brose et al. 2014) and SELECT (Schlumberger et al. 2015) trials, respectively. These drugs should be administered under the supervision of clinicians with experience in managing these drugs and associated toxicities (Brose et al. 2012).

The use of next-generation sequencing to identify gene alterations, including BRAF mutations, RET, ALK and NTRK gene fusions, depends on the availability of such testing and NHS England is currently establishing a national test directory service over seven genomic hubs UK-wide to carry out cancer genomic testing by next-generation sequencing and interpret all results. Currently, the service offers testing in paediatric DTC via a multi-target next-generation sequencing panel for RET small and structural variants and NTRK1/2/3 structural variants (https://www.england.nhs.uk/publication/national-genomic-test-directories/). Future studies in CYP will likely help to direct targeted therapies for the treatment of individuals with particular somatic point mutations and fusion genes (Nies et al. 2021).

NICE has recommended the use of Larotrectinib (https://www.nice.org.uk/guidance/ta630) within the Cancer Drugs Fund as an option for treating NTRK fusion-positive solid tumours in adults and children if the disease is locally advanced or metastatic, or surgery could cause severe health problems and they have no satisfactory treatment options. Entrectinib has been recommended for use under similar circumstances in children over 12 years of age if they have not had treatment with an NTRK inhibitor previously (https://www.nice.org.uk/guidance/ta644). Clinical trials of RET inhibitors are ongoing.

In the rare situation where CYP are not cured of their DTC, palliative care teams should be involved in care at an early stage. Symptom control may include palliative radiotherapy, in a similar manner to as described above in Recommendation 55. Other locally ablative treatment modalities such as surgery, radiofrequency ablation and vertebroplasty can be considered to treat deposits of disease that are causing specific symptoms.

58. Consider the use of external beam radiotherapy for symptom control in the palliative setting (Moderate Recommendation, Low-Quality Evidence, Delphi Consensus 73%)

External beam radiotherapy is very rarely indicated in CYP with DTC in the primary or adjuvant setting because their disease is usually very iodine avid and sensitive so there is no benefit from the addition of external beam radiotherapy (Hay et al. 2010).

External beam radiotherapy to the neck can be of use in the palliative setting for symptom control, for example in cases of unresectable disease invading the larynx, trachea or oesophagus, where uncontrolled growth of the disease will cause life-threatening or distressing symptoms. There may also be a role in palliating the effects of more distant metastases for example painful bone metastases, bleeding or obstructing deposits of tumour or brain metastases. Any external beam radiotherapy administered should be delivered in a dedicated paediatric radiotherapy centre (Landau et al. 2000).

Discussion

The rarity of paediatric endocrine tumours like DTC makes their management challenging. During the process of guideline development, we have confirmed a general lack of high-quality evidence relating to this age group and identified, through the consensus surveys necessarily undertaken, a professional mandate for both national speciality advisory panels and most importantly, a national register and evaluation of key management outcomes in these rare, eminently curable young patients. If we are to enhance clinical trials and quality of evidence, improve the health-related quality of survival and improve access to, and equity of, expertise in care, such a national register and centralised, advisory panel needs to be expedited alongside the development of tertiary, dedicated and age-appropriate, endocrine oncology multidisciplinary teams and services.

The GDG believes that in the UK no child should be looked after without a full thyroid cancer MDT. If no CYP-specific MDT is available, then these patients should be discussed at an adult thyroid cancer MDT. Working through and in collaboration with existing MDTs for adult and paediatric thyroid cancer will ensure the best use of resources in the current financial climate. We also recommend treatment at a high-volume tertiary centre where technologies and expertise can be most easily accessed, and the process streamlined with regards to ease of decision making and patient flow. Some additional funding may be needed, for the attendance of additional personnel at the existing MDTs and an ongoing programme of evaluation and audit, but any cost implications for health care budgets must be considered alongside the likely benefits inherent in concentrating management of rare conditions in specific expert MDTs. These include improvements in clinical outcomes and reduced complication rates both of which also greatly improve the patient experience. Financial savings are also produced by more efficient referral pathways, reduction in unnecessary and inappropriate investigations and avoiding unnecessary surgical interventions. As the aim of these guidelines is to improve patient care and experience, long-term complications should be reduced with potentially associated improved costs.

Supplementary materials

This is linked to the online version of the paper at https://doi.org/10.1530/ERC-22-0035.

Declaration of interest

Dr Helen Spoudeas: founder of SUCCESS Charity – Life After Cure, www.successcharity.org advocating for the unmet health needs of patients surviving childhood brain tumours registered as LC in January 2019; founder of the national HPAT Virtual interest forum, piloting multidisciplinary virtual decision-making for children with complex hypothalamo-pituitary tumours. The other authors have no conflicts of interest to declare.

Funding

Dr M N Gaze is supported by the National Institute for Health Research, University College London Hospitals Biomedical Research Centre, London, UK and by the Radiation Research Unit at the Cancer Research UKK City of London Centre Award (C7893/A28990). The guideline development was sponsored by unrestricted grants from Sandoz Pharmaceuticals, the professional societies CCLG, BSPED and The Society of British Neurological Surgeons, and the patient support groups 'Association of Multiple Endocrine Neoplastic Disorders' (AMEND), 'SUCCESS Charity – Life After Cure' and 'The Pituitary Foundation'. Excepting as stakeholders, the sponsors had no role in development of guideline methodology or final guideline recommendations. The CCLG provided administrative support throughout the guideline and the RCPCH provided advice and appraised the guideline at different stages.

Endorsing organisations

RCPCH.

Author contribution statement

T R Kurzawinski and M N Gaze contributed equally to this work. HAS chaired the project board, obtained the funding and co-ordinated the GDG set up. SRH, TRK and MG led the GDG. SRH, TRK and MG were responsible for the study concept and design and drafted the manuscript. SRH and SF undertook the literature search. All authors took part in the grading process, interpreted the data, revised the manuscript critically for important intellectual content, approved the final version to be published, and agreed to be accountable for all aspects of the work.

Acknowledgements

The GDG would like to thank all stakeholders, the Project Board, Delphi panellists and our external peer reviewers for their input into this guideline. The authors also would like to thank the librarians of University College London and University Hospitals of Leicester NHS Trust and Dr Nirit Braha, Consultant Paediatrician, Royal Free Hospitals London NHS Foundation Trust for their help in the literature search process. The authors are particularly grateful to the Quality Improvement Committee Clinical Leads for Evidence Based Medicine and Appraisals at the RCPCH, for their advice and appraisal during the guideline development process, and for the final endorsement of the guideline.

References

Abbasian Ardakani A, Reiazi R & Mohammadi A 2018 A clinical decision support system using ultrasound textures and radiologic features to distinguish metastasis From tumor-free cervical lymph nodes in patients with papillary thyroid carcinoma. Journal of Ultrasound in Medicine 37 2527–2535. (https://doi.org/10.1002/jum.14610)

Search Google ScholarExport Citation

Akobeng AK 2005 Principles of evidence based medicine. Archives of Disease in Childhood 90 837–840. (https://doi.org/10.1136/adc.2005.071761)

Search Google ScholarExport Citation

Al Nofal A, Gionfriddo MR, Javed A, Haydour Q, Brito JP, Prokop LJ, Pittock ST & Murad MH 2016 Accuracy of thyroid nodule sonography for the detection of thyroid cancer in children: systematic review and meta-analysis. Clinical Endocrinology 84 423–430. (https://doi.org/10.1111/cen.12786)

Search Google ScholarExport Citation

Alessandri AJ, Goddard KJ, Blair GK, Fryer CJ & Schultz KR 2000 Age is the major determinant of recurrence in pediatric differentiated thyroid carcinoma. Medical and Pediatric Oncology 35 41–46. (https://doi.org/10.1002/1096-911x(200007)35:1<41::aid-mpo7>3.0.co;2-7)

Search Google ScholarExport Citation

Al-Qahtani KH, Tunio MA, Al Asiri M, Aljohani NJ, Bayoumi Y, Riaz K & Alshakweer W 2015 Clinicopathological features and treatment outcomes of differentiated thyroid cancer in Saudi children and adults. Journal of Otolaryngology – Head and Neck Surgery 44 48. (https://doi.org/10.1186/s40463-015-0102-6)

Search Google ScholarExport Citation

Al-Qurayshi Z, Hauch A, Srivastav S, Aslam R, Friedlander P & Kandil E 2016 A national perspective of the risk, presentation, and outcomes of pediatric thyroid cancer, presentation. JAMA Otolaryngology– Head and Neck Surgery 142 472–478. (https://doi.org/10.1001/jamaoto.2016.0104)

Search Google ScholarExport Citation

Altincik A, Demir K, Abaci A, Bober E & Buyukgebiz A 2010 Fine-needle aspiration biopsy in the diagnosis and follow-up of thyroid nodules in childhood. Journal of Clinical Research in Pediatric Endocrinology 2 78–80. (https://doi.org/10.4274/jcrpe.v2i2.78)

Search Google ScholarExport Citation

Antonelli A, Miccoli P, Fallahi P, Grosso M, Nesti C, Spinelli C & Ferrannini E 2003 Role of neck ultrasonography in the follow-up of children operated on for thyroid papillary cancer. Thyroid 13 479–484. (https://doi.org/10.1089/105072503322021142)

Search Google ScholarExport Citation

Aretz S, Uhlhaas S, Caspari R, Mangold E, Pagenstecher C, Propping P & Friedl W 2004 Frequency and parental origin of de novo APC mutations in familial adenomatous polyposis. European Journal of Human Genetics 12 52–58. (https://doi.org/10.1038/sj.ejhg.5201088)

Search Google ScholarExport Citation

Astl J, Chovanec M, Lukes P, Katra R, Dvorakova M, Vlcek P, Sykorova P & Betka J 2014 Thyroid carcinoma surgery in children and adolescents - 15 years experience surgery of pediatric thyroid carcinoma. International Journal of Pediatric Otorhinolaryngology 78 990–994. (https://doi.org/10.1016/j.ijporl.2014.03.005)

Search Google ScholarExport Citation

Bal CS, Padhy AK & Kumar A 2001 Clinical features of differentiated thyroid carcinoma in children and adolescents from a sub-Himalayan iodine-deficient endemic zone. Nuclear Medicine Communications 22 881–887. (https://doi.org/10.1097/00006231-200108000-00006)

Search Google ScholarExport Citation

Bal CS, Kumar A, Chandra P, Dwivedi SN & Mukhopadhyaya S 2004 Is chest x-ray or high-resolution computed tomography scan of the chest sufficient investigation to detect pulmonary metastasis in pediatric differentiated thyroid cancer? Thyroid 14 217–225. (https://doi.org/10.1089/105072504773297894)

Search Google ScholarExport Citation

Balachandar S, La Quaglia M, Tuttle RM, Heller G, Ghossein RA & Sklar CA 2016 Pediatric differentiated thyroid carcinoma of follicular cell origin: prognostic significance of histologic subtypes. Thyroid 26 219–226. (https://doi.org/10.1089/thy.2015.0287)

Search Google ScholarExport Citation

Ballester LY, Sarabia SF, Sayeed H, Patel N, Baalwa J, Athanassaki I, Hernandez JA, Fang E, Quintanilla NM & Roy A et al.2016 Integrating molecular testing in the diagnosis and management of children with thyroid lesions. Pediatric and Developmental Pathology 19 94–100. (https://doi.org/10.2350/15-05-1638-OA.1)

Search Google ScholarExport Citation

Bargren AE, Meyer-Rochow GY, Sywak MS, Delbridge LW, Chen H & Sidhu SB 2010 Diagnostic utility of fine-needle aspiration cytology in pediatric differentiated thyroid cancer. World Journal of Surgery 34 1254–1260. (https://doi.org/10.1007/s00268-010-0391-x)

Search Google ScholarExport Citation

Baş VN, Aycan Z, Cetinkaya S, Uner C, Cavuşoğlu YH & Arda N 2012. Thyroid nodules in children and adolescents a single institution's experience. Journal of Pediatric Endocrinology and Metabolism 25, 633. (https://doi.org/10.1515/jpem-2012-0132)

Search Google ScholarExport Citation

Baumgarten HD, Bauer AJ, Isaza A, Mostoufi-Moab S, Kazahaya K & Adzick NS 2019 Surgical management of pediatric thyroid disease: complication rates after thyroidectomy at the Children's Hospital of Philadelphia high-volume Pediatric Thyroid Center. Journal of Pediatric Surgery 54 1969–1975. (https://doi.org/10.1016/j.jpedsurg.2019.02.009)

Search Google ScholarExport Citation

Beimfohr C, Klugbauer S, Demidchik EP, Lengfelder E & Rabes HM 1999 NTRK1 re-arrangement in papillary thyroid carcinomas of children after the Chernobyl reactor accident. International Journal of Cancer 80 842–847. (https://doi.org/10.1002/(sici)1097-0215(19990315)80:6<842::aid-ijc7>3.0.co;2-z)

Search Google ScholarExport Citation

Ben Arush MW, Stein ME, Perez Nahum M, Zidan J & Kuten A 2000 Pediatric thyroid carcinoma 22 years of experience at the Northern Israel Oncology Center (1973–1995). Pediatric Hematology and Oncology 17 85–92. (https://doi.org/10.1080/088800100276695)

Search Google ScholarExport Citation

Bergenfelz A, Jansson S, Kristoffersson A, Martensson H, Reihner E, Wallin G & Lausen I 2008 Complications to thyroid surgery: results as reported in a database from a multicenter audit comprising 3,660 patients. Langenbeck's Archives of Surgery 393 667–673. (https://doi.org/10.1007/s00423-008-0366-7)

Search Google ScholarExport Citation

Bhatti P, Veiga LH, Ronckers CM, Sigurdson AJ, Stovall M, Smith SA, Weathers R, Leisenring W, Mertens AC, Hammond S, et al.2010 Risk of second primary thyroid cancer after radiotherapy for a childhood cancer in a large cohort study: an update from the childhood cancer survivor study. Radiation Research 174 741–752. (https://doi.org/10.1667/RR2240.1)

Search Google ScholarExport Citation

Bignol-Kologu M, Tanyel FC, Senocak ME, Büyükpamukc N & Hiçsönmez A 2000 Surgical treatment of differentiated thyroid carcinoma in children. European Journal of Pediatric Surgery 2000 347–452. (https://doi.org/10.1055/s-2000-12070)

Search Google ScholarExport Citation

Biko J, Reiners C, Kreissl MC, Verburg FA, Demidchik Y & Drozd V 2011 Favourable course of disease after incomplete remission on (131)I therapy in children with pulmonary metastases of papillary thyroid carcinoma: 10 years follow-up. European Journal of Nuclear Medicine and Molecular Imaging 38 651–655. (https://doi.org/10.1007/s00259-010-1669-9)

Search Google ScholarExport Citation

Borson-Chazot F, Causeret S, Lifante JC, Augros M, Berger N & Peix JL 2004 Predictive factors for recurrence from a series of 74 children and adolescents with differentiated thyroid cancer. World Journal of Surgery 28 1088–1092. (https://doi.org/10.1007/s00268-004-7630-y)

Search Google ScholarExport Citation

Breuer C, Tuggle C, Solomon D & Sosa JA 2013 Pediatric thyroid disease: when is surgery necessary, and who should be operating on our children? Journal of Clinical Research in Pediatric Endocrinology 5 (Supplement 1) 79–85. (https://doi.org/10.4274/jcrpe.817)

Search Google ScholarExport Citation

Brignardello E, Corrias A, Isolato G, Palestini N, Cordero Di Montezemolo L, Fagioli F & Boccuzzi G 2008 Ultrasound screening for thyroid carcinoma in childhood cancer survivors: a case series. Journal of Clinical Endocrinology and Metabolism 93 4840–4843. (https://doi.org/10.1210/jc.2008-1528)

Search Google ScholarExport Citation

Brink JS, Van Heerden JA, McIver B, Salomao DR, Farley DR, Grant CS, Thompson GB, Zimmerman D & Hay ID 2000 Papillary thyroid cancer with pulmonary metastases in children: long-term prognosis. Surgery 128 881–886; discussion 886–887. (https://doi.org/10.1067/msy.2000.109728)

Search Google ScholarExport Citation

Brose MS, Smit J, Capdevila J, Elisei R, Nutting C, Pitoia F, Robinson B, Schlumberger M, Shong YK & Takami H 2012 Regional approaches to the management of patients with advanced, radioactive iodine-refractory differentiated thyroid carcinoma. Expert Review of Anticancer Therapy 12 1137–1147. (https://doi.org/10.1586/era.12.96)

Search Google ScholarExport Citation

Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, De La Fouchardiere C, Pacini F, Paschke R, Shong YK, et al.2014 Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet 384 319–328. (https://doi.org/10.1016/S0140-6736(1460421-9)

Search Google ScholarExport Citation

Burke JF, Sippel RS & Chen H 2012 Evolution of pediatric thyroid surgery at a tertiary medical center. Journal of Surgical Research 177 268–274. (https://doi.org/10.1016/j.jss.2012.06.044)

Search Google ScholarExport Citation

Buryk MA, Simons JP, Picarsic J, Monaco SE, Ozolek JA, Joyce J, Gurtunca N, Nikiforov YE & Feldman Witchel S 2015 Can malignant thyroid nodules be distinguished from benign thyroid nodules in children and adolescents by clinical characteristics? A review of 89 pediatric patients with thyroid nodules. Thyroid 25 392–400. (https://doi.org/10.1089/thy.2014.0312)

Search Google ScholarExport Citation

Canadian Pediatric Thyroid Nodule ( CaPTN ) Study Group 2008 Canadian pediatric thyroid nodule study. Journal of Pediatric Surgery 43 826–830. (https://doi.org/10.1016/j.jpedsurg.2007.12.019)

Search Google ScholarExport Citation

Chandrasekhar SS, Randolph GW, Seidman MD, Rosenfeld RM, Angelos P, Barkmeier-Kraemer J, Benninger MS, Blumin JH, Dennis G, Hanks J, et al.2013 Clinical practice guideline: improving voice outcomes after thyroid surgery. Otolaryngology–Head and Neck Surgery 148 (Supplement) S1–S37. (https://doi.org/10.1177/0194599813487301)

Search Google ScholarExport Citation

Chen Y, Masiakos PT, Gaz RD, Hodin RA, Parangi S, Randolph GW, Sadow PM & Stephen AE 2015 Pediatric thyroidectomy in a high volume thyroid surgery center: risk factors for postoperative hypocalcemia. Journal of Pediatric Surgery 50 1316–1319. (https://doi.org/10.1016/j.jpedsurg.2014.10.056)

Search Google ScholarExport Citation

Cherella CE, Hollowell ML, Smith JR, Zendejas B, Modi BP, Cibas ES & Wassner AJ 2022 Subtype of atypia on cytology and risk of malignancy in pediatric thyroid nodules. Cancer Cytopathology 130 330–335. (https://doi.org/10.1002/cncy.22544)

Search Google ScholarExport Citation

Chiu HK, Sanda S, Fechner PY & Pihoker C 2012 Correlation of TSH with the risk of paediatric thyroid carcinoma. Clinical Endocrinology 77 316–322. (https://doi.org/10.1111/j.1365-2265.2012.04383.x)

Search Google ScholarExport Citation

Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Mang O & Lau WH 2004 Differentiated thyroid carcinoma in childhood and adolescence-clinical course and role of radioiodine. Pediatric Blood and Cancer 42 176–183. (https://doi.org/10.1002/pbc.10410)

Search Google ScholarExport Citation

Chung YS, Kim JY, Bae JS, Song BJ, Kim JS, Jeon HM, Jeong SS, Kim EK & Park WC 2009 Lateral lymph node metastasis in papillary thyroid carcinoma: results of therapeutic lymph node dissection. Thyroid 19 241–246. (https://doi.org/10.1089/thy.2008.0244)

Search Google ScholarExport Citation

Clement SC, Van Rijn RR, Van Eck-Smit BL, Van Trotsenburg AS, Caron HN, Tytgat GA & Van Santen HM 2015 Long-term efficacy of current thyroid prophylaxis and future perspectives on thyroid protection during 131I-metaiodobenzylguanidine treatment in children with neuroblastoma. European Journal of Nuclear Medicine and Molecular Imaging 42 706–715. (https://doi.org/10.1007/s00259-014-2967-4)

Search Google ScholarExport Citation

Cole CD & Wu HH 2014 Fine-needle aspiration in pediatric patients 12 years of age and younger: a 20-year retrospective study from a single tertiary medical center. Diagnostic Cytopathology 42 600–605. (https://doi.org/10.1002/dc.23085)

Search Google ScholarExport Citation

Collini P, Massimino M, Leite SF, Mattavelli F, Seregni E, Zucchini N, Spreafico F, Ferrari A, Castellani MR, Cantu G, et al.2006 Papillary thyroid carcinoma of childhood and adolescence: a 30-year experience at the Istituto Nazionale Tumori in Milan. Pediatric Blood and Cancer 46 300–306. (https://doi.org/10.1002/pbc.20474)

Search Google ScholarExport Citation

Cordioli MICV, Moraes L, Carvalheira G, Sisdelli L, Alves MTS, Delcelo R, Monte O, Longui CA, Cury AN & Cerutti JM 2016 AGK-BRAF gene fusion is a recurrent event in sporadic pediatric thyroid carcinoma. Cancer Medicine 5 1535–1541. (https://doi.org/10.1002/cam4.698)

Search Google ScholarExport Citation

Corrias A & Mussa A 2013 Thyroid nodules in pediatrics: which ones can be left alone, which ones must be investigated, when and how. Journal of Clinical Research in Pediatriatric Endocrinology 5 (Suppl 1) 57–69. (https://doi.org/10.4274/jcrpe.853)

Search Google ScholarExport Citation

Corrias A, Mussa A, Baronio F, Arrigo T, Salerno M, Segni M, Vigone MC, Gastaldi R, Zirilli G, Tuli G, et al.2010 Diagnostic features of thyroid nodules in pediatrics. Archives of Pediatrics and Adolescent Medicine 164 714–719. (https://doi.org/10.1001/archpediatrics.2010.114)

Search Google ScholarExport Citation

De Jong MC, Gaze MN, Szychot E, Rozalen Garcia V, Brain C, Dattani M, Spoudeas H, Hindmarsh P, Abdel-Aziz TE, Bomanji J, et al.2021 Treating papillary and follicular thyroid cancer in children and young people: single UK-center experience between 2003 and 2018. Journal of Pediatric Surgery 56 534–539. (https://doi.org/10.1016/j.jpedsurg.2020.07.034)

Search Google ScholarExport Citation

De Kock L, Sabbaghian N, Soglio DB, Guillerman RP, Park BK, Chami R, Deal CL, Priest JR & Foulkes WD 2014 Exploring the association Between DICER1 mutations and differentiated thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 99 E1072–E1077. (https://doi.org/10.1210/jc.2013-4206)

Search Google ScholarExport Citation

Demidchik YE, Demidchik EP, Reiners C, Biko J, Mine M, Saenko VA & Yamashita S 2006 Comprehensive clinical assessment of 740 cases of surgically treated thyroid cancer in children of Belarus. Annals of Surgery 243 525–532. (https://doi.org/10.1097/01.sla.0000205977.74806.0b)

Search Google ScholarExport Citation

Dinauer CA, Breuer C & Rivkees SA 2008 Differentiated thyroid cancer in children: diagnosis and management. Current Opinion in Oncology 20 59–65. (https://doi.org/10.1097/CCO.0b013e3282f30220)

Search Google ScholarExport Citation

Dottorini ME, Vignati A, Mazzucchelli L, Lomuscio G & Colombo L 1997 Differentiated thyroid carcinoma in children and adolescents a 37-year experience in 85 patients year experience in 85 patients. Journal of Nuclear Medicine 38 669–675.

Search Google ScholarExport Citation

Durante C, Costante G & Filetti S 2013 Differentiated thyroid carcinoma: defining new paradigms for postoperative management. Endocrine-Related Cancer 20 R141–R154. (https://doi.org/10.1530/ERC-13-0066)

Search Google ScholarExport Citation

Enomoto Y, Enomoto K, Uchino S, Shibuya H, Watanabe S & Noguchi S 2012 Clinical features, treatment, and long-term outcome of papillary thyroid cancer in children and adolescents without radiation exposure. World Journal of Surgery 36 1241–1246. (https://doi.org/10.1007/s00268-012-1558-4)

Search Google ScholarExport Citation

Enomoto K, Enomoto Y, Uchino S, Yamashita H & Noguchi S 2013 Follicular thyroid cancer in children and adolescents: clinicopathologic features, long-term survival, and risk factors for recurrence. Endocrine Journal 60 629–635. (https://doi.org/10.1507/endocrj.ej12-0372)

Search Google ScholarExport Citation

Fallah M, Pukkala E, Tryggvadottir L, Olsen JH, Tretli S, Sundquist K & Hemminki K 2013 Risk of thyroid cancer in first-degree relatives of patients with non-medullary thyroid cancer by histology type and age at diagnosis: a joint study from five Nordic countries. Journal of Medical Genetics 50 373–382. (https://doi.org/10.1136/jmedgenet-2012-101412)

Search Google ScholarExport Citation

Farahati J, Parlowsky T, Mäder U, Reiners C & Bucsky P 1998 Differentiated thyroid cancer in children and adolescents. Langenbeck's Archives of Surgery 383 235–239. (https://doi.org/10.1007/s004230050124)

Search Google ScholarExport Citation

Fard-Esfahani A, Emami-Ardekani A, Fallahi B, Fard-Esfahani P, Beiki D, Hassanzadeh-Rad A & Eftekhari M 2014 Adverse effects of radioactive iodine-131 treatment for differentiated thyroid carcinoma. Nuclear Medicine Communications 35 808–817. (https://doi.org/10.1097/MNM.0000000000000132)

Search Google ScholarExport Citation

Fenton CL, Lukes Y, Nicholson D, Dinauer CA, Francis GL & Tuttle RM 2000 The ret PTC mutations are common in sporadic papillary thyroid carcinoma of children and young adults. Journal of Clinical Endocrinology and Metabolism 85 1170–1175. (https://doi.org/10.1210/jcem.85.3.6472)

Search Google ScholarExport Citation

Finkelstein A, Levy GH, Hui P, Prasad A, Virk R, Chhieng DC, Carling T, Roman SA, Sosa JA, Udelsman R, et al.2012 Papillary thyroid carcinomas with and without BRAF V600E mutations are morphologically distinct. Histopathology 60 1052–1059. (https://doi.org/10.1111/j.1365-2559.2011.04149.x)

Search Google ScholarExport Citation

Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID, Luster M, et al.2015 Management guidelines for children with thyroid nodules and differentiated thyroid cancer. Thyroid 25 716–759. (https://doi.org/10.1089/thy.2014.0460)

Search Google ScholarExport Citation

Franco AT, Ricarte-Filho JC, Isaza A, Jones Z, Jain N, Mostoufi-Moab S, Surrey L, Laetsch TW, Li MM, Dehart JC, et al.2022 Fusion oncogenes are associated With increased metastatic capacity and persistent disease in pediatric thyroid cancers. Journal of Clinical Oncology 40 1081–1090. (https://doi.org/10.1200/JCO.21.01861)

Search Google ScholarExport Citation

Freire AV, Ropelato MG, Ballerini MG, Acha O, Bergada I, De Papendieck LG & Chiesa A 2014 Predicting hypocalcemia after thyroidectomy in children. Surgery 156 130–136. (https://doi.org/10.1016/j.surg.2014.02.016)

Search Google ScholarExport Citation

Fridman MV, Savva NN, Krasko OV, Zborovskaya AA, Mankovskaya SV, Schmid KW & Demidchik YE 2012 Clinical and pathologic features of "sporadic" papillary thyroid carcinoma registered in the years 2005 to 2008 in children and adolescents of Belarus. Thyroid 22 1016–1024. (https://doi.org/10.1089/thy.2011.0005)

Search Google ScholarExport Citation

Gambardella C, Offi C, Romano RM, De Palma M, Ruggiero R, Candela G, Puziello A, Docimo L, Grasso M & Docimo G 2020 Transcutaneous laryngeal ultrasonography: a reliable, non-invasive and inexpensive preoperative method in the evaluation of vocal cords motility-a prospective multicentric analysis on a large series and a literature review. Updates in Surgery 72 885–892. (https://doi.org/10.1007/s13304-020-00728-3)

Search Google ScholarExport Citation

Gertz RJ, Nikiforov Y, Rehrauer W, McDaniel L & Lloyd RV 2016 Mutation in BRAF and other members of the MAPK pathway in papillary thyroid carcinoma in the pediatric population. Archives of Pathology and Laboratory Medicine 140 134–139. (https://doi.org/10.5858/arpa.2014-0612-OA)

Search Google ScholarExport Citation

Gharib H, Papini E, Garber JR, Duick DS, Harrell RM, Hegedus L, Paschke R, Valcavi R, Vitti P & AACE/ACE/AME Task Force on Thyroid Nodules 2016 American Association of Clinical Endocrinologists, American college of endocrinology, and associazione medici endocrinologi medical guidelines for clinical practice for the diagnosis and management of thyroid nodules--2016 update. Endocrine Practice 22 622–639. (https://doi.org/10.4158/EP161208.GL)

Search Google ScholarExport Citation

Goldfarb M, Gondek SS, Sanchez Y & Lew JI 2012 Clinic-based ultrasound can predict malignancy in pediatric thyroid nodules. Thyroid 22 827–831. (https://doi.org/10.1089/thy.2011.0494)

Search Google ScholarExport Citation

Goldfarb M & Freyer DR 2014 Comparison of secondary and primary thyroid cancer in adolescents and young adults. Cancer 120 1155–1161. (https://doi.org/10.1002/cncr.28463)

Search Google ScholarExport Citation

Golpanian S, Perez EA, Tashiro J, Lew JI, Sola JE & Hogan AR 2016 Pediatric papillary thyroid carcinoma: outcomes and survival predictors in 2504 surgical patients. Pediatric Surgery International 32 201–208. (https://doi.org/10.1007/s00383-015-3855-0)

Search Google ScholarExport Citation

Gupta A, Ly S, Castroneves LA, Frates MC, Benson CB, Feldman HA, Wassner AJ, Smith JR, Marqusee E, Alexander EK, et al.2013 A standardized assessment of thyroid nodules in children confirms higher cancer prevalence than in adults. Journal of Clinical Endocrinology and Metabolism 98 3238–3245. (https://doi.org/10.1210/jc.2013-1796)

Search Google ScholarExport Citation

Gutnick J, Soldes O, Gupta M & Milas M 2012 Circulating thyrotropin receptor messenger RNA for evaluation of thyroid nodules and surveillance of thyroid cancer in children. Journal of Pediatric Surgery 47 171–176. (https://doi.org/10.1016/j.jpedsurg.2011.10.036)

Search Google ScholarExport Citation

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, Norris S, Falck-Ytter Y, Glasziou P, Debeer H, et al.2011. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 64 383–394. (https://doi.org/10.1016/j.jclinepi.2010.04.026)

Search Google ScholarExport Citation

Hanba C, Svider PF, Siegel B, Sheyn A, Shkoukani M, Lin HS & Raza SN 2017 Pediatric thyroidectomy. Otolaryngology–Head and Neck Surgery 156 360–367. (https://doi.org/10.1177/0194599816677527)

Search Google ScholarExport Citation

Handkiewicz-Junak D, Wloch J, Roskosz J, Krajewska J, Kropinska A, Pomorski L, Kukulska A, Prokurat A, Wygoda Z & Jarzab B 2007 Total thyroidectomy and adjuvant radioiodine treatment independently decrease locoregional recurrence risk in childhood and adolescent differentiated thyroid cancer. Journal of Nuclear Medicine 48 879–888. (https://doi.org/10.2967/jnumed.106.035535)

Search Google ScholarExport Citation

Handkiewicz-Junak D, Gawlik T, Rozkosz J, Puch Z, Michalik B, Gubala E, Krajewska J, Kluczewska A & Jarzab B 2015 Recombinant human thyrotropin preparation for adjuvant radioiodine treatment in children and adolescents with differentiated thyroid cancer. European Journal of Endocrinology 173 873–881. (https://doi.org/10.1530/EJE-15-0562)

Search Google ScholarExport Citation

Harach HR & Williams ED 1995 Childhood thyroid cancer in England and Wales. British Journal of Cancer 72 777–783. (https://doi.org/10.1038/bjc.1995.410)

Search Google ScholarExport Citation

Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, et al.2016 2015 American Thyroid Association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American Thyroid Association guidelines task force on thyroid nodules and differentiated thyroid cancer. Thyroid 26 1–133. (https://doi.org/10.1089/thy.2015.0020)

Search Google ScholarExport Citation

Haveman JW, Van Tol KM, Rouwé CW, Piers DA & Plukker JTM 2003 Surgical experience in children with differentiated thyroid carcinoma. Annals of Surgical Oncology 10 15–20. (https://doi.org/10.1245/aso.2003.03.002)

Search Google ScholarExport Citation

Hay ID, Gonzalez-Losada T, Reinalda MS, Honetschlager JA, Richards ML & Thompson GB 2010 Long-term outcome in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World Journal of Surgery 34 1192–1202. (https://doi.org/10.1007/s00268-009-0364-0)

Search Google ScholarExport Citation

Heikkinen M, Halttunen S, Terava M, Karkkainen JM, Lopponen H & Penttila E 2019 Vocal foldparesis as a surgical complication: our 10-year experience with 162 incidents. Clinical Otolaryngology 44 179–182. (https://doi.org/10.1111/coa.13251)

Search Google ScholarExport Citation

Henke LE, Perkins SM, Pfeifer JD, Ma C, Chen Y, DeWees T & Grigsby PW 2014 BRAF V600E mutational status in pediatric thyroid cancer. Pediatric Blood and Cancer 61 1168–1172. (https://doi.org/10.1002/pbc.24935)

Search Google ScholarExport Citation

Hess J, Thomas G, Braselmann H, Bauer V, Bogdanova T, Wienberg J, Zitzelsberger H & Unger K 2011 Gain of chromosome band 7q11 in papillary thyroid carcinomas of young patients is associated with exposure to low-dose irradiation. PNAS 108 9595–9600. (https://doi.org/10.1073/pnas.1017137108)

Search Google ScholarExport Citation

Hoe FM, Charron M & Moshang Jr T 2006 Use of the recombinant human TSH stimulated thyroglobulin level and diagnostic whole body scan in children with differentiated thyroid carcinoma. Journal of Pediatric Endocrinology and Metabolism 19 25–30. (https://doi.org/10.1515/jpem.2006.19.1.25)

Search Google ScholarExport Citation

Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI & Sola JE 2009 Pediatric thyroid carcinoma: incidence and outcomes in 1753 patients. Journal of Surgical Research 156 167–172. (https://doi.org/10.1016/j.jss.2009.03.098)

Search Google ScholarExport Citation

Hoperia V, Larin A, Jensen K, Bauer A & Vasko V 2010 Thyroid fine needle aspiration biopsies in children: study of cytological-histological correlation and immunostaining with thyroid peroxidase monoclonal antibodies. International Journal of Pediatric Endocrinology 2010 690108. (https://doi.org/10.1155/2010/690108)

Search Google ScholarExport Citation

Hosler GA, Clark I, Zakowski MF, Westra WH & Ali SZ 2006 Cytopathologic analysis of thyroid lesions in the pediatric population. Diagnostic Cytopathology 34 101–105. (https://doi.org/10.1002/dc.20388)

Search Google ScholarExport Citation

Huang CH, Chao TC, Hseuh C, Lin KJ, Ho TY, Lin SF & Lin JD 2012 Therapeutic outcome and prognosis in young patients with papillary and follicular thyroid cancer. Pediatric Surgery International 28 489–494. (https://doi.org/10.1007/s00383-012-3054-1)

Search Google ScholarExport Citation

Iorcansky S, Herzovich V, Qualey RR & Tuttle RM 2005 Serum thyrotropin (TSH) levels after recombinant human TSH injections in children and teenagers with papillary thyroid cancer. Journal of Clinical Endocrinology and Metabolism 90 6553–6555. (https://doi.org/10.1210/jc.2005-1550)

Search Google ScholarExport Citation

Ito Y, Kihara M, Takamura Y, Kobayashi K, Miya A, Hirokawa M & Miyauchi A 2012 Prognosis and prognostic factors of papillary thyroid carcinoma in patients under 20 years. Endocrine Journal 59 539–545. (https://doi.org/10.1507/endocrj.ej12-0086)

Search Google ScholarExport Citation

Izquierdo R, Shankar R, Kort K & Khurana K 2009 Ultrasound-guided fine-needle aspiration in the management of thyroid nodules in children and adolescents. Thyroid 19 703–705. (https://doi.org/10.1089/thy.2009.0058)

Search Google ScholarExport Citation

Jang HW, Lee JI, Kim HK, Oh YL, Choi YL, Jin DK, Kim JH, Chung JH & Kim SW 2012 Identification of a cut-off for the MACIS score to predict the prognosis of differentiated thyroid carcinoma in children and young adults. Head and Neck 34 696–701. (https://doi.org/10.1002/hed.21808)

Search Google ScholarExport Citation

Jarzab B, Handkiewicz Junak D, Włoch J, Kalemba B, Roskosz J, Kukulska A & Puch Z 2000 Multivariate analysis of prognostic factors for differentiated thyroid carcinoma in children. European Journal of Nuclear Medicine 27 833–841. (https://doi.org/10.1007/s002590000271)

Search Google ScholarExport Citation

Jarzab B, Handkiewicz-Junak D & Wloch J 2005 Juvenile differentiated thyroid carcinoma and the role of radioiodine in its treatment: a qualitative review. Endocrine-Related Cancer 12 773–803. (https://doi.org/10.1677/erc.1.00880)

Search Google ScholarExport Citation

Jia MR, Baran JA, Bauer AJ, Isaza A, Surrey LF, Bhatti T, McGrath C, Jalaly J, Mostoufi-Moab S, Adzick NS, et al.2021 Utility of fine-needle aspirations to diagnose pediatric thyroid nodules. Hormone Research in Paediatrics 94 263–274. (https://doi.org/10.1159/000519307)

Search Google ScholarExport Citation

Jiang W, Newbury RO & Newfield RS 2016 Pediatric thyroid surgery and management of thyroid nodules – an institutional experience over a 10-year period. International Journal of Pediatric Endocrinology 2016 1. (https://doi.org/10.1186/s13633-015-0019-x)

Search Google ScholarExport Citation

Jiang L, Xiang Y, Huang R, Tian R & Liu B 2021 Clinical applications of single-photon emission computed tomography/computed tomography in post-ablation (131)iodine scintigraphy in children and young adults with differentiated thyroid carcinoma. Pediatric Radiology 51 1724–1731. (https://doi.org/10.1007/s00247-021-05039-2)

Search Google ScholarExport Citation

Jin X, Masterson L, Patel A, Hook L, Nicholson J, Jefferies S, Gaze M, Nassif R, Eller R, Hulse T, et al.2015 Conservative or radical surgery for pediatric papillary thyroid carcinoma: a systematic review of the literature. International Journal of Pediatric Otorhinolaryngology 79 1620–1624. (https://doi.org/10.1016/j.ijporl.2015.08.004)

Search Google ScholarExport Citation

Joliat GR, Guarnero V, Demartines N, Schweizer V & Matter M 2017 Recurrent laryngeal nerve injury after thyroid and parathyroid surgery: incidence and postoperative evolution assessment. Medicine 96 e6674. (https://doi.org/10.1097/MD.0000000000006674)

Search Google ScholarExport Citation

Kamani T, Charkhchi P, Zahedi A & Akbari MR 2022 Genetic susceptibility to hereditary non-medullary thyroid cancer. Hereditary Cancer in Clinical Practice 20 9. (https://doi.org/10.1186/s13053-022-00215-3)

Search Google ScholarExport Citation

Kapila K, Pathan SK, George SS, Haji BE, Das DK & Qadan LR 2010 Fine needle aspiration cytology of the thyroid in children and adolescents experience with 792 aspirates. Acta Cytologica 54 569–574. (https://doi.org/10.1159/000325179)

Search Google ScholarExport Citation

Kennedy RD, Potter DD, Moir CR & El-Youssef M 2014 The natural history of familial adenomatous polyposis syndrome: a 24 year review of a single center experience in screening, diagnosis, and outcomes. Journal of Pediatric Surgery 49 82–86. (https://doi.org/10.1016/j.jpedsurg.2013.09.033)

Search Google ScholarExport Citation

Khurana KK, Labrador E, Izquierdo R, Mesonero CE & Pisharodi LR 1999 The role of fine-needle aspiration biopsy in the management of thyroid nodules in children, adolescents, and young adults: a multi-institutional study. Thyroid 9 383–386. (https://doi.org/10.1089/thy.1999.9.383)

Search Google ScholarExport Citation

Kim HY, Gelfand MJ & Sharp SE 2011 SPECT/CT imaging in children with papillary thyroid carcinoma. Pediatric Radiology 41 1008–1012. (https://doi.org/10.1007/s00247-011-2039-x)

Search Google ScholarExport Citation

Kim J, Sun Z, Adam MA, Adibe OO, Rice HE, Roman SA & Tracy ET 2017 Predictors of nodal metastasis in pediatric differentiated thyroid cancer. Journal of Pediatric Surgery 52 120–123. (https://doi.org/10.1016/j.jpedsurg.2016.10.033)

Search Google ScholarExport Citation

Kiratli PO, Volkan-Salanci B, Gunay EC, Varan A, Akyuz C & Buyukpamukcu M 2013 Thyroid cancer in pediatric age group an institutional experience and review of the literature. Journal of Pediatric Hematology/Oncology 35 93–97. (https://doi.org/10.1097/MPH.0b013e3182755d9e)

Search Google ScholarExport Citation

Kirk JM, Mort C, Grant DB, Touzel RJ & Plowman N 1992 The usefulness of serum thyroglobulin in the follow-up of differentiated thyroid carcinoma in children. Medical and Pediatric Oncology 20 201–208. (https://doi.org/10.1002/mpo.2950200304)

Search Google ScholarExport Citation

Klein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JGM, Van Dam EWCM, Havekes B, Van Den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, et al.2016 Pediatric differentiated thyroid carcinoma in the Netherlands: a nationwide follow-up study. Journal of Clinical Endocrinology and Metabolism 101 2031–2039. (https://doi.org/10.1210/jc.2015-3290)

Search Google ScholarExport Citation

Kluijfhout WP, Pasternak JD, Van Der Kaay D, Vriens MR, Propst EJ & Wasserman JD 2017 Is it time to reconsider lobectomy in low-risk paediatric thyroid cancer? Clinical Endocrinology 86 591–596. (https://doi.org/10.1111/cen.13287)

Search Google ScholarExport Citation

Koltin D, O'Gorman CS, Murphy A, Ngan B, Daneman A, Navarro OM, Garcia C, Atenafu EG, Wasserman JD, Hamilton J, et al.2016 Pediatric thyroid nodules: ultrasonographic characteristics and inter-observer variability in prediction of malignancy. Journal of Pediatric Endocrinology and Metabolism 29 789–794. (https://doi.org/10.1515/jpem-2015-0242)

Search Google ScholarExport Citation

Koo JS, Hong S & Park CS 2009 Diffuse sclerosing variant is a major subtype of papillary thyroid carcinoma in the young. Thyroid 19 1225–1231. (https://doi.org/10.1089/thy.2009.0073)

Search Google ScholarExport Citation

Kowalski LP, Goncalves Filho J, Pinto CA, Carvalho AL & De Camargo B 2003 Long-term survival rates in young patients with thyroid carcinoma. Archives of Otolaryngology–Head and Neck Surgery 129 746–749. (https://doi.org/10.1001/archotol.129.7.746)

Search Google ScholarExport Citation

Kuijt WJ & Huang SA 2005 Children with differentiated thyroid cancer achieve adequate hyperthyrotropinemia within 14 days of levothyroxine withdrawal. Journal of Clinical Endocrinology and Metabolism 90 6123–6125. (https://doi.org/10.1210/jc.2005-1085)

Search Google ScholarExport Citation

La Quaglia MP, Black T, Holcomb GWR, Sklar C, Azizkhan RG, Haase GM & Newman KD 2000 Differentiated thyroid cancer clinical characteristics, treatment, and outcome in patients under 21 years of age who present with distant metastases. A report from the surgical discipline committee of the children's cancer group. Journal of Pediatric Surgery, 35, 955–960. (https://doi.org/10.1053/jpsu.2000.6935)

Search Google ScholarExport Citation

Lale SA, Morgenstern NN, Chiara S & Wasserman P 2015 Fine needle aspiration of thyroid nodules in the pediatric population: a 12-year cyto-histological correlation experience at North Shore-Long Island Jewish Health System. Diagnostic Cytopathology 43 598–604. (https://doi.org/10.1002/dc.23265)

Search Google ScholarExport Citation

Landau D, Vini L, A'Hern R & Harmer C 2000 Thyroid cancer in children the Royal Marsden Hospital experience. European Journal of Cancer 36 214–220. (https://doi.org/10.1016/s0959-8049(9900281-6)

Search Google ScholarExport Citation

Lang BH, Chu KK, Tsang RK, Wong KP & Wong BY 2014 Evaluating the incidence, clinical significance and predictors for vocal cord palsy and incidental laryngopharyngeal conditions before elective thyroidectomy: is there a case for routine laryngoscopic examination? World Journal of Surgery 38 385–391. (https://doi.org/10.1007/s00268-013-2259-3)

Search Google ScholarExport Citation

Larg MI, Barbus E, Gabora K, Pestean C, Cheptea M & Piciu D 2019 18f-Fdg pet/Ct in differentiated thyroid carcinoma. Acta Endocrinologica 15 203–208. (https://doi.org/10.4183/aeb.2019.203)

Search Google ScholarExport Citation

Lauper JM, Krause A, Vaughan TL & Monnat Jr RJ 2013 Spectrum and risk of neoplasia in Werner syndrome: a systematic review. PLoS ONE 8 e59709. (https://doi.org/10.1371/journal.pone.0059709)

Search Google ScholarExport Citation

Lazar L, Lebenthal Y, Steinmetz A, Yackobovitch-Gavan M & Phillip M 2009 Differentiated thyroid carcinoma in pediatric patients: comparison of presentation and course between pre-pubertal children and adolescents. Journal of Pediatrics 154 708–714. (https://doi.org/10.1016/j.jpeds.2008.11.059)

Search Google ScholarExport Citation

Lebbink CA, Dekker BL, Bocca G, Braat AJAT, Derikx JPM, Dierselhuis MP, De Keizer B, Kruijff S, Kwast ABG, Van Nederveen FH, et al.2020 New national recommendations for the treatment of pediatric differentiated thyroid carcinoma in the Netherlands. European Journal of Endocrinology 183 P11–P18. (https://doi.org/10.1530/EJE-20-0191)

Search Google ScholarExport Citation

Leboulleux S, Baudin E, Hartl DW, Travagli JP & Schlumberger M 2005 Follicular cell-derived thyroid cancer in children. Hormone Research 63 145–151. (https://doi.org/10.1159/000084717)

Search Google ScholarExport Citation

Lee YA, Jung HW, Kim HY, Choi H, Kim HY, Hah JH, Park DJ, Chung JK, Yang SW, Shin CH, et al.2015 Pediatric patients with multifocal papillary thyroid cancer have higher recurrence rates than adult patients: a retrospective analysis of a large pediatric thyroid cancer cohort over 33 years. Journal of Clinical Endocrinology and Metabolism 100 1619–1629. (https://doi.org/10.1210/jc.2014-3647)

Search Google ScholarExport Citation

Lee KA, Sharabiani MTA, Tumino D, Wadsley J, Gill V, Gerrard G, Sindhu R, Gaze MN, Moss L & Newbold K 2019 Differentiated thyroid cancer in children: a UK multicentre review and review of the literature. Clinical Oncology 31 385–390. (https://doi.org/10.1016/j.clon.2019.02.005)

Search Google ScholarExport Citation

Leeman-Neill RJ, Brenner AV, Little MP, Bogdanova TI, Hatch M, Zurnadzy LY, Mabuchi K, Tronko MD & Nikiforov YE 2013 RET/PTC and PAX8/PPARgamma chromosomal rearrangements in post-Chernobyl thyroid cancer and their association with iodine-131 radiation dose and other characteristics. Cancer 119 1792–1799. (https://doi.org/10.1002/cncr.27893)

Search Google ScholarExport Citation

Lerner J & Goldfarb M 2015a Pediatric thyroid microcarcinoma. Annals of Surgical Oncology 22 4187–4192. (https://doi.org/10.1245/s10434-015-4546-8)

Search Google ScholarExport Citation

Lerner J & Goldfarb M 2015b Follicular variant papillary thyroid carcinoma in a pediatric population. Pediatric Blood and Cancer 62 1942–1946. (https://doi.org/10.1002/pbc.25623)

Search Google ScholarExport Citation

Lo CY, Kwok KF & Yuen PW 2000 A prospective evaluation of recurrent laryngeal nerve paralysis during thyroidectomy. Archives of Surgery 135 204–207. (https://doi.org/10.1001/archsurg.135.2.204)

Search Google ScholarExport Citation

Luster M, Handkiewicz-Junak D, Grossi A, Zacharin M, Taieb D, Cruz O, Hitzel A, Casas JA, Mader U, Dottorini ME, et al.2009 Recombinant thyrotropin use in children and adolescents with differentiated thyroid cancer: a multicenter retrospective study. Journal of Clinical Endocrinology and Metabolism 94 3948–3953. (https://doi.org/10.1210/jc.2009-0593)

Search Google ScholarExport Citation

Ly S, Frates MC, Benson CB, Peters HE, Grant FD, Drubach LA, Voss SD, Feldman HA, Smith JR, Barletta J, et al.2016 Features and outcome of autonomous thyroid nodules in children: 31 consecutive patients seen at a Single Center. Journal of Clinical Endocrinology and Metabolism 101 3856–3862. (https://doi.org/10.1210/jc.2016-1779)

Search Google ScholarExport Citation

Lyshchik A, Drozd V, Demidchik Y & Reiners C 2005 Diagnosis of thyroid cancer in children: value of gray-scale and power doppler US. Radiology 235 604–613. (https://doi.org/10.1148/radiol.2352031942)

Search Google ScholarExport Citation

Machens A, Lorenz K, Nguyen Thanh P, Brauckhoff M & Dralle H 2010 Papillary thyroid cancer in children and adolescents does not differ in growth pattern and metastatic behavior. Journal of Pediatrics 157 648–652. (https://doi.org/10.1016/j.jpeds.2010.04.026)

Search Google ScholarExport Citation

Machens A, Elwerr M, Thanh PN, Lorenz K, Schneider R & Dralle H 2016 Impact of central node dissection on postoperative morbidity in pediatric patients with suspected or proven thyroid cancer. Surgery 160 484–492. (https://doi.org/10.1016/j.surg.2016.03.007)

Search Google ScholarExport Citation

Maksimoski M, Bauer AJ, Kazahaya K, Manning SC, Parikh SR, Simons JP, D'Souza J, Maddalozzo J, Purkey MR, Rychlik K, et al.2022 Outcomes in pediatric thyroidectomy: results from a multinational, multi-institutional database. Otolaryngology–Head and Neck Surgery 1945998221076065. (https://doi.org/10.1177/01945998221076065)

Search Google ScholarExport Citation

Mallick U, Harmer C, Hackshaw A & & Moss L 2012a Iodine or Not (IoN) for low-risk differentiated thyroid cancer: the next UK National Cancer Research Network randomised trial following HiLo. Clinical Oncology 24 159–161. (https://doi.org/10.1016/j.clon.2012.01.001)

Search Google ScholarExport Citation

Mallick U, Harmer C, Yap B, Wadsley J, Clarke S, Moss L, Nicol A, Clark PM, Farnell K, McCready R, et al.2012b Ablation with low-dose radioiodine and thyrotropin alfa in thyroid cancer. New England Journal of Medicine 366 1674–1685. (https://doi.org/10.1056/NEJMoa1109589)

Search Google ScholarExport Citation

Markovina S, Grigsby PW, Schwarz JK, Dewees T, Moley JF, Siegel BA & Perkins SM 2014 Treatment approach, surveillance, and outcome of well-differentiated thyroid cancer in childhood and adolescence. Thyroid 24 1121–1126. (https://doi.org/10.1089/thy.2013.0297)

Search Google ScholarExport Citation

Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PL, Zheng Z, Liaw D, Caron S, Duboue B, Lin AY, et al.1998 Mutation spectrum and genotype-phenotype analyses in Cowden disease and Bannayan-Zonana syndrome, two hamartoma syndromes with germline PTEN mutation. Human Molecular Genetics 7 507–515. (https://doi.org/10.1093/hmg/7.3.507)

Search Google ScholarExport Citation

Massimino M, Collini P, Leite SF, Spreafico F, Zucchini N, Ferrari A, Mattavelli F, Seregni E, Castellani MR, Cantu G, et al.2006 Conservative surgical approach for thyroid and lymph-node involvement in papillary thyroid carcinoma of childhood and adolescence. Pediatric Blood and Cancer 46 307–313. (https://doi.org/10.1002/pbc.20438)

Search Google ScholarExport Citation

Metzger ML, Howard SC, Hudson MM, Gow KW, Li CS, Krasin MJ, Merchant T, Kun L, Shelso J, Pui CH, et al.2006 Natural history of thyroid nodules in survivors of pediatric Hodgkin lymphoma. Pediatric Blood and Cancer 46 314–319. (https://doi.org/10.1002/pbc.20541)

Search Google ScholarExport Citation

Mihailovic J, Nikoletic K & Srbovan D 2014 Recurrent disease in juvenile differentiated thyroid carcinoma: prognostic factors, treatments, and outcomes. Journal of Nuclear Medicine 55 710–717. (https://doi.org/10.2967/jnumed.113.130450)

Search Google ScholarExport Citation

Mollen KP, Shaffer AD, Yip L, Monaco SE, Huyett P, Viswanathan P, Witchel SF, Duvvuri U & Simons JP 2022 Unique molecular signatures are associated with aggressive histology in pediatric differentiated thyroid cancer. Thyroid 32 236–244. (https://doi.org/10.1089/thy.2021.0317)

Search Google ScholarExport Citation

Monaco SE, Pantanowitz L, Khalbuss WE, Benkovich VA, Ozolek J, Nikiforova MN, Simons JP & Nikiforov YE 2012 Cytomorphological and molecular genetic findings in pediatric thyroid fine-needle aspiration. Cancer Cytopathology 120 342–350. (https://doi.org/10.1002/cncy.21199)

Search Google ScholarExport Citation

Morris LF, Waguespack SG, Warneke CL, Ryu H, Ying AK, Anderson BJ, Sturgis EM, Clayman GL, Lee JE, Evans DB, et al.2012 Long-term follow-up data may help manage patient and parent expectations for pediatric patients undergoing thyroidectomy. Surgery 152 1165–1171. (https://doi.org/10.1016/j.surg.2012.08.056)

Search Google ScholarExport Citation

Morrison PJ & Atkinson AB 2009 Genetic aspects of familial thyroid cancer. Oncologist 14 571–577. (https://doi.org/10.1634/theoncologist.2009-0046)

Search Google ScholarExport Citation

Moses W, Weng J & Kebebew E 2011 Prevalence, clinicopathologic features, and somatic genetic mutation profile in familial versus sporadic nonmedullary thyroid cancer. Thyroid 21 367–371. (https://doi.org/10.1089/thy.2010.0256)

Search Google ScholarExport Citation

Mostafa M, Vali R, Chan J, Omarkhail Y & Shammas A 2016 Variants and pitfalls on radioiodine scans in pediatric patients with differentiated thyroid carcinoma. Pediatric Radiology 46 1579–1589. (https://doi.org/10.1007/s00247-016-3655-2)

Search Google ScholarExport Citation

Mostoufi-Moab S, Labourier E, Sullivan L, Livolsi V, Li Y, Xiao R, Beaudenon-Huibregtse S, Kazahaya K, Adzick NS, Baloch Z, et al.2018 Molecular testing for oncogenic gene alterations in pediatric thyroid lesions. Thyroid 28 60–67. (https://doi.org/10.1089/thy.2017.0059)

Search Google ScholarExport Citation

Moudgil P, Vellody R, Heider A, Smith EA, Grove JJ, Jarboe MD, Bruch SW & Dillman JR 2016 Ultrasound-guided fine-needle aspiration biopsy of pediatric thyroid nodules. Pediatric Radiology 46 365–371. (https://doi.org/10.1007/s00247-015-3478-6)

Search Google ScholarExport Citation

Mussa A, Salerno MC, Bona G, Wasniewska M, Segni M, Cassio A, Vigone MC, Gastaldi R, Iughetti L, Santanera A, et al.2013 Serum thyrotropin concentration in children with isolated thyroid nodules. Journal of Pediatrics 163 1465–1470. (https://doi.org/10.1016/j.jpeds.2013.07.003)

Search Google ScholarExport Citation

Mussa A, De Andrea M, Motta M, Mormile A, Palestini N & Corrias A 2015 Predictors of malignancy in children with thyroid nodules. Journal of Pediatrics 167 886.e1–892.e1. (https://doi.org/10.1016/j.jpeds.2015.06.026)

Search Google ScholarExport Citation

NCCN 2016 NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®): Thyroid Carcinoma, Version 1.2016. Plymouth Meeting, PA, USA: National Comprehensive Cancer Network. (avilable at: https://www.nccn.org/professionals/physician_gls/pdf/thyroid.pdf)

Search Google ScholarExport Citation

Newman KD, Black T, Heller G, Azizkhan RG, Holcomb GW, Sklar C, Vmamis V, Haase GM & Quaglia MPL 1998 Differentiated thyroid cancer determinants of disease progression in patients 21 years of age at diagnosis a report from the Surgical Discipline Committee of the Children ' s Cancer Group. Annals of Surgery 227 533–541. (https://doi.org/10.1097/00000658-199804000-00014)

Search Google ScholarExport Citation

Ngeow J, Mester J, Rybicki LA, Ni Y, Milas M & Eng C 2011 Incidence and clinical characteristics of thyroid cancer in prospective series of individuals with Cowden and Cowden-like syndrome characterized by germline PTEN, SDH, or KLLN alterations. Journal of Clinical Endocrinology and Metabolism 96 E2063–E2071. (https://doi.org/10.1210/jc.2011-1616)

Search Google ScholarExport Citation

Nice T, Pasara S, Goldfarb M, Doski J, Goldin A, Gow KW, Nuchtern JG, Vasudevan SA, Langer M & Beierle EA 2015 Pediatric papillary thyroid cancer >1 cm: is total thyroidectomy necessary? Journal of Pediatric Surgery 50 1009–1013. (https://doi.org/10.1016/j.jpedsurg.2015.03.031)

Search Google ScholarExport Citation

NICE 2018 Policies and procedures. London, UK: National Institute for Health and Care Excellence. (available at: https://www.nice.org.uk/about/who-we-are/policies-and-procedures)

Search Google ScholarExport Citation

Niedziela M, Breborowicz D, Trejster E & Korman E 2002 Hot nodules in children and adolescents in western Poland from 1996 to 2000: clinical analysis of 31 patients. Journal of Pediatric Endocrinology and Metabolism 15 823–830. (https://doi.org/10.1515/jpem.2002.15.6.823)

Search Google ScholarExport Citation

Nies M, Vassilopoulou-Sellin R, Bassett RL, Yedururi S, Zafereo ME, Cabanillas ME, Sherman SI, Links TP & Waguespack SG 2021 Distant metastases From childhood differentiated thyroid carcinoma: clinical course and mutational landscape. Journal of Clinical Endocrinology and Metabolism 106 e1683–e1697. (https://doi.org/10.1210/clinem/dgaa935)

Search Google ScholarExport Citation

Nikiforov YE & Gnepp DR 1994 Pediatric thyroid cancer after the Chernobyl disaster. Pathomorphologic study of 84 cases (1991–1992) from the Republic of Belarus. Cancer 74 748–766. (https://doi.org/10.1002/1097-0142(19940715)74:2<748::aid-cncr2820740231>3.0.co;2-h)

Search Google ScholarExport Citation

Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H & Fagin JA 1997 Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Research 57 1690–1694.

Search Google ScholarExport Citation

Norlen O, Charlton A, Sarkis LM, Henwood T, Shun A, Gill AJ & Delbridge L 2015 Risk of malignancy for each Bethesda class in pediatric thyroid nodules. Journal of Pediatric Surgery 50 1147–1149. (https://doi.org/10.1016/j.jpedsurg.2014.10.046)

Search Google ScholarExport Citation

O'Gorman CS, Hamilton J, Rachmiel M, Gupta A, Ngan BY & Daneman D 2010 Thyroid cancer in childhood: a retrospective review of childhood course. Thyroid 20 375–380. (https://doi.org/10.1089/thy.2009.0386)

Search Google ScholarExport Citation

Okoli CP & Pawlowski SD 2004 The Delphi method as a research tool: an example, design considerations and applications. Information and Management 42 15–29. (https://doi.org/10.1016/j.im.2003.11.002)

Search Google ScholarExport Citation

Oommen PT, Romahn A, Linden T, Fruhwald MC & Bucsky P 2008 UICC-2002 TNM classification is not suitable for differentiated thyroid cancer in children and adolescents. Pediatric Blood and Cancer 50 1159–1162. (https://doi.org/10.1002/pbc.21385)

Search Google ScholarExport Citation

Pacini F, Molinaro E, Castagna MG, Agate L, Elisei R, Ceccarelli C, Lippi F, Taddei D, Grasso L & Pinchera A 2003 Recombinant human thyrotropin-stimulated serum thyroglobulin combined with neck ultrasonography has the highest sensitivity in monitoring differentiated thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 88 3668–3673. (https://doi.org/10.1210/jc.2002-021925)

Search Google ScholarExport Citation

Palmer BA, Zarroug AE, Poley RN, Kollars JP & Moir CR 2005 Papillary thyroid carcinoma in children: risk factors and complications of disease recurrence. Journal of Pediatric Surgery 40 1284–1288. (https://doi.org/10.1016/j.jpedsurg.2005.05.012)

Search Google ScholarExport Citation

Papendieck P, Gruñeiro-Papendieck L, Venara M, Acha O, Maglio S, Bergadá I & Chiesa A 2011 Differentiated thyroid carcinoma: presentation and follow-up in children and adolescents. Journal of Pediatric Endocrinology and Metabolism 24 743–748. (https://doi.org/10.1515/jpem.2011.241)

Search Google ScholarExport Citation

Papendieck P, Gruñeiro-Papendieck L, Venara M, Acha O, Cozzani H, Mateos F, Maglio S, Calcagno ML, Bergada I & Chiesa A 2015 Differentiated thyroid cancer in children: prevalence and predictors in a large cohort with thyroid nodules followed prospectively. Journal of Pediatrics 167 199–201. (https://doi.org/10.1016/j.jpeds.2015.04.041)

Search Google ScholarExport Citation

Park JH, Lee YS, Kim BW, Chang HS & Park CS 2012 Skip lateral neck node metastases in papillary thyroid carcinoma. World Journal of Surgery 36 743–747. (https://doi.org/10.1007/s00268-012-1476-5)

Search Google ScholarExport Citation

Partyka KL, Huang EC, Cramer HM, Chen S & Wu HH 2016 Histologic and clinical follow-up of thyroid fine-needle aspirates in pediatric patients. Cancer Cytopathology 124 467–471. (https://doi.org/10.1002/cncy.21713)

Search Google ScholarExport Citation

Patel A, Jhiang S, Dogra S, Terrell R, Powers PA, Fenton C, Dinauer CA, Tuttle RM & Francis GL 2002 Differentiated thyroid carcinoma that express sodium-iodide symporter have a lower risk of recurrence for children and adolescents. Pediatric Research 52 737–744. (https://doi.org/10.1203/00006450-200211000-00021)

Search Google ScholarExport Citation

Patel NA, Bly RA, Adams S, Carlin K, Parikh SR, Dahl JP & Manning S 2018 A clinical pathway for the postoperative management of hypocalcemia after pediatric thyroidectomy reduces blood draws. International Journal of Pediatric Otorhinolaryngology 105 132–137. (https://doi.org/10.1016/j.ijporl.2017.12.011)

Search Google ScholarExport Citation

Pawelczak M, David R, Franklin B, Kessler M, Lam L & Shah B 2010 Outcomes of children and adolescents with well-differentiated thyroid carcinoma and pulmonary metastases following (1)(3). Thyroid 20 1095–1101. (https://doi.org/10.1089/thy.2009.0446)

Search Google ScholarExport Citation

Peiling Yang S & Ngeow J 2016 Familial non-medullary thyroid cancer: unraveling the genetic maze. Endocrine-Related Cancer 23 R577–R595. (https://doi.org/10.1530/ERC-16-0067)

Search Google ScholarExport Citation

Pekova B, Sykorova V, Mastnikova K, Vaclavikova E, Moravcova J, Vlcek P, Lastuvka P, Taudy M, Katra R, Bavor P, et al.2021 NTRK fusion genes in thyroid carcinomas: clinicopathological characteristics and their impacts on prognosis. Cancers 13 1932. (https://doi.org/10.3390/cancers13081932)

Search Google ScholarExport Citation

Penko K, Livezey J, Fenton C, Patel A, Nicholson D, Flora M, Oakley K, Tuttle RM & Francis G 2005 BRAF mutations are uncommon in papillary thyroid cancer of young patients. Thyroid 15 320–325. (https://doi.org/10.1089/thy.2005.15.320)

Search Google ScholarExport Citation

Perros P, Boelaert K, Colley S, Evans C, Evans RM, Gerrard Ba G, Gilbert J, Harrison B, Johnson SJ, Giles TE, et al.2014 Guidelines for the management of thyroid cancer. Clinical Endocrinology 81 (Supplement 1) 1–122. (https://doi.org/10.1111/cen.12515)

Search Google ScholarExport Citation

Pluijmen MJ, Eustatia-Rutten C, Goslings BM, Stokkel MP, Arias AM, Diamant M, Romijn JA & Smit JW 2003 Effects of low-iodide diet on postsurgical radioiodide ablation therapy in patients with differentiated thyroid carcinoma. Clinical Endocrinology 58 428–435. (https://doi.org/10.1046/j.1365-2265.2003.01735.x)

Search Google ScholarExport Citation

Popovtzer A, Shpitzer T, Bahar G, Feinmesser R & Segal K 2006 Thyroid cancer in children: management and outcome experience of a referral center. Otolaryngology–Head and Neck Surgery 135 581–584. (https://doi.org/10.1016/j.otohns.2006.04.004)

Search Google ScholarExport Citation

Powers PA, Dinauer CA, Tuttle RM, Robie DK, McClellan DR & Francis GL 2003 Tumor size and extent of disease at diagnosis predict the response to initial therapy for papillary thyroid carcinoma in children and adolescents. Journal of Pediatric Endocrinology and Metabolism 16 693–702. (https://doi.org/10.1515/jpem.2003.16.5.693)

Search Google ScholarExport Citation

Powers PA, Dinauer CA, Tuttle RM & Francis GL 2004 The MACIS score predicts the clinical course of papillary thyroid carcinoma in children and adolescents. Journal of Pediatric Endocrinology and Metabolism 17 339–343. (https://doi.org/10.1515/jpem.2004.17.3.339)

Search Google ScholarExport Citation

Prasad ML, Vyas M, Horne MJ, Virk RK, Morotti R, Liu Z, Tallini G, Nikiforova MN, Christison-Lagay ER, Udelsman R, et al.2016 NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 122 1097–1107. (https://doi.org/10.1002/cncr.29887)

Search Google ScholarExport Citation

Qichang W, Lin B, Gege Z, Youjia Z, Qingjie M, Renjie W & Bin J 2019 Diagnostic performance of 18F-FDG-PET/CT in DTC patients with thyroglobulin elevation and negative iodine scintigraphy: a meta-analysis. European Journal of Endocrinology 181 93–102. (https://doi.org/10.1530/EJE-19-0261)

Search Google ScholarExport Citation

Qu N, Zhang L, Lu ZW, Ji QH, Yang SW, Wei WJ & Zhang Y 2016 Predictive factors for recurrence of differentiated thyroid cancer in patients under 21 years of age and a meta-analysis of the current literature. Tumour Biology 37 7797–7808. (https://doi.org/10.1007/s13277-015-4532-6)

Search Google ScholarExport Citation

Raab SS, Silverman JF, Elsheikh TM, Thomas PA, Paul E, Raab S & Thomas A 1995 Pediatric thyroid nodules disease demographics and clinical management as determined by fine needle aspiration biopsy. Pediatrics 95 46–49. (https://doi.org/10.1542/peds.95.1.46)

Search Google ScholarExport Citation

Raval MV, Bentrem DJ, Stewart AK, Ko CY & Reynolds M 2010 Utilization of total thyroidectomy for differentiated thyroid cancer in children. Annals of Surgical Oncology 17 2545–2553. (https://doi.org/10.1245/s10434-010-1083-3)

Search Google ScholarExport Citation

Redlich A, Boxberger N, Schmid KW, Fruhwald M, Rohrer T & Vorwerk P 2012 Sensitivity of fine-needle biopsy in detecting pediatric differentiated thyroid carcinoma. Pediatric Blood and Cancer 59 233–237. (https://doi.org/10.1002/pbc.24051)

Search Google ScholarExport Citation

Ricarte-Filho JC, Li S, Garcia-Rendueles ME, Montero-Conde C, Voza F, Knauf JA, Heguy A, Viale A, Bogdanova T, Thomas GA, et al.2013 Identification of kinase fusion oncogenes in post-Chernobyl radiation-induced thyroid cancers. Journal of Clinical Investigation 123 4935–4944. (https://doi.org/10.1172/JCI69766)

Search Google ScholarExport Citation

Richards ML 2010 Familial syndromes associated with thyroid cancer in the era of personalized medicine. Thyroid 20 707–713. (https://doi.org/10.1089/thy.2010.1641)

Search Google ScholarExport Citation

Rio Frio T, Bahubeshi A, Kanellopoulou C, Hamel N, Niedziela M, Sabbaghian N, Pouchet C, Gilbert L, O'Brien PK, Serfas K, et al.2011 DICER1 mutations in familial multinodular goiter with and without ovarian Sertoli-Leydig cell tumors. JAMA 305 68–77. (https://doi.org/10.1001/jama.2010.1910)

Search Google ScholarExport Citation

Ritter A, Hod R, Reuven Y, Shpitzer T, Mizrachi A, Raveh E & Bachar G 2021 Role of intraoperative recurrent laryngeal nerve monitoring for pediatric thyroid surgery: comparative analysis. Head and Neck 43 849–857. (https://doi.org/10.1002/hed.26544)

Search Google ScholarExport Citation

Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA & Udelsman R 2011 The treatment of differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocrine Reviews 32 798–826. (https://doi.org/10.1210/er.2011-0011)

Search Google ScholarExport Citation

Rosario PW, Mineiro Filho AF, Lacerda RX & Calsolari MR 2012 Recombinant human TSH for thyroid remnant ablation with (131)I in children and adolescents with papillary carcinoma. Hormone Research in Paediatrics 77 59–62. (https://doi.org/10.1159/000335088)

Search Google ScholarExport Citation

Rose J, Wertheim BC & Guerrero MA 2012 Radiation treatment of patients with primary pediatric malignancies: risk of developing thyroid cancer as a secondary malignancy. American Journal of Surgery 204 881–886; discussion 886–887. (https://doi.org/10.1016/j.amjsurg.2012.07.030)

Search Google ScholarExport Citation

Rosenbaum E, Hosler G, Zahurak M, Cohen Y, Sidransky D & Westra WH 2005 Mutational activation of BRAF is not a major event in sporadic childhood papillary thyroid carcinoma. Modern Pathology 18 898–902. (https://doi.org/10.1038/modpathol.3800252)

Search Google ScholarExport Citation

Rossi ED, Straccia P, Martini M, Revelli L, Lombardi CP, Pontecorvi A & Fadda G 2014 The role of thyroid fine-needle aspiration cytology in the pediatric population: an institutional experience. Cancer Cytopathology 122 359–367. (https://doi.org/10.1002/cncy.21400)

Search Google ScholarExport Citation

Rutter MM, Jha P, Schultz KA, Sheil A, Harris AK, Bauer AJ, Field AL, Geller J & Hill DA 2016 DICER1 mutations and differentiated thyroid carcinoma: evidence of a direct association. Journal of Clinical Endocrinology and Metabolism 101 1–5. (https://doi.org/10.1210/jc.2015-2169)

Search Google ScholarExport Citation

Saavedra J, Deladoey J, Saint-Vil D, Boivin Y, Alos N, Deal C, Van Vliet G & Huot C 2011 Is ultrasonography useful in predicting thyroid cancer in children with thyroid nodules and apparently benign cytopathologic features? Hormone Research in Paediatrics 75 269–275. (https://doi.org/10.1159/000322877)

Search Google ScholarExport Citation

Samuels SL, Surrey LF, Hawkes CP, Amberge M, Mostoufi-Moab S, Langer JE, Adzick NS, Kazahaya K, Bhatti T, Baloch Z, et al.2018 Characteristics of follicular variant papillary thyroid carcinoma in a pediatric cohort. Journal of Clinical Endocrinology and Metabolism 103 1639–1648. (https://doi.org/10.1210/jc.2017-02454)

Search Google ScholarExport Citation

Santos JE, Freitas M, Fonseca CP, Castilho P, Carreira IM, Rombeau JL & Branco MC 2017 Iodine deficiency a persisting problem: assessment of iodine nutrition and evaluation of thyroid nodular pathology in Portugal. Journal of Endocrinological Investigation 40 185–191. (https://doi.org/10.1007/s40618-016-0545-2)

Search Google ScholarExport Citation

Sassolas G, Hafdi-Nejjari Z, Ferraro A, Decaussin-Petrucci M, Rousset B, Borson-Chazot F, Borbone E, Berger N & Fusco A 2012 Oncogenic alterations in papillary thyroid cancers of young patients. Thyroid 22 17–26. (https://doi.org/10.1089/thy.2011.0215)

Search Google ScholarExport Citation

Savio R, Gosnell J, Palazzo FF, Sywak M, Agarwal G, Cowell C, Shun A, Robinson B & Delbridge LW 2005 The role of a more extensive surgical approach in the initial multimodality management of papillary thyroid cancer in children. Journal of Pediatric Surgery 40 1696–1700. (https://doi.org/10.1016/j.jpedsurg.2005.07.029)

Search Google ScholarExport Citation

Schlumberger M, Catargi B, Borget I, Deandreis D, Zerdoud S, Bridji B, Bardet S, Leenhardt L, Bastie D, Schvartz C, et al.2012 Strategies of radioiodine ablation in patients with low-risk thyroid cancer. New England Journal of Medicine 366 1663–1673. (https://doi.org/10.1056/NEJMoa1108586)

Search Google ScholarExport Citation

Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, Habra MA, Newbold K, Shah MH, Hoff AO, et al.2015 Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. New England Journal of Medicine 372 621–630. (https://doi.org/10.1056/NEJMoa1406470)

Search Google ScholarExport Citation

Schneider P, Biko J, Reiners C, Demidchik YE, Drozd VM, Capozza RF, Cointry GR & Ferretti JL 2004 Impact of parathyroid status and Ca and vitamin-D supplementation on bone mass and muscle-bone relationships in 208 Belarussian children after thyroidectomy because of thyroid carcinoma. Experimental and Clinical Endocrinology and Diabetes 112 444–450. (https://doi.org/10.1055/s-2004-821204)

Search Google ScholarExport Citation

Schneider R, Machens A, Sekulla C, Lorenz K & Dralle H 2021 Recurrent laryngeal nerve Preservation Strategies in Pediatric thyroid Oncology: continuous vs. intermittent Nerve Monitoring. Cancers 13 4333. (https://doi.org/10.3390/cancers13174333)

Search Google ScholarExport Citation

Schneider R, Machens A, Sekulla C, Lorenz K, Weber F & Dralle H 2018 Twenty-year experience of paediatric thyroid surgery using intraoperative nerve monitoring. British Journal of Surgery 105 996–1005. (https://doi.org/10.1002/bjs.10792)

Search Google ScholarExport Citation

Scholz S, Smith JR, Chaignaud B, Shamberger RC & Huang SA 2011 Thyroid surgery at Children's Hospital Boston: a 35-year single-institution experience. Journal of Pediatric Surgery 46 437–442. (https://doi.org/10.1016/j.jpedsurg.2010.09.009)

Search Google ScholarExport Citation

Shayota BJ, Pawar SC & Chamberlain RS 2013 MeSS: a novel prognostic scale specific for pediatric well-differentiated thyroid cancer: a population-based, SEER outcomes study. Surgery 154 429–435. (https://doi.org/10.1016/j.surg.2013.04.047)

Search Google ScholarExport Citation

Sigurdson AJ, Ronckers CM, Mertens AC, Stovall M, Smith SA, Liu Y, Berkow RL, Hammond S, Neglia JP, Meadows AT, et al.2005 Primary thyroid cancer after a first tumour in childhood (the Childhood Cancer Survivor Study): a nested case-control study. Lancet 365 2014–2023. (https://doi.org/10.1016/S0140-6736(0566695-0)

Search Google ScholarExport Citation

Silva-Vieira M, Santos R, Leite V & Limbert E 2015 Review of clinical and pathological features of 93 cases of well-differentiated thyroid carcinoma in pediatric age at the Lisbon Centre of the Portuguese Institute of Oncology between 1964 and 2006. International Journal of Pediatric Otorhinolaryngology 79 1324–1329. (https://doi.org/10.1016/j.ijporl.2015.06.002)

Search Google ScholarExport Citation

Sinclair CF, Bumpous JM, Haugen BR, Chala A, Meltzer D, Miller BS, Tolley NS, Shin JJ, Woodson G & Randolph GW 2016 Laryngeal examination in thyroid and parathyroid surgery: an American Head and Neck Society consensus statement: AHNS Consensus Statement. Head and Neck 38 811–819. (https://doi.org/10.1002/hed.24409)

Search Google ScholarExport Citation

Smallridge RC, Meek SE, Morgan MA, Gates GS, Fox TP, Grebe S & Fatourechi V 2007 Monitoring thyroglobulin in a sensitive immunoassay has comparable sensitivity to recombinant human tsh-stimulated thyroglobulin in follow-up of thyroid cancer patients. Journal of Clinical Endocrinology and Metabolism 92 82–87. (https://doi.org/10.1210/jc.2006-0993)

Search Google ScholarExport Citation

Smith JR, Marqusee E, Webb S, Nose V, Fishman SJ, Shamberger RC, Frates MC & Huang SA 2011 Thyroid nodules and cancer in children with PTEN hamartoma tumor syndrome. Journal of Clinical Endocrinology and Metabolism 96 34–37. (https://doi.org/10.1210/jcem.96.3.zeg34a)

Search Google ScholarExport Citation

Smith M, Pantanowitz L, Khalbuss WE, Benkovich VA & Monaco SE 2013 Indeterminate pediatric thyroid fine needle aspirations: a study of 68 cases. Acta Cytologica 57 341–348. (https://doi.org/10.1159/000351029)

Search Google ScholarExport Citation

Soberman N, Leonidas JC, Cherrick I, Schiff R & Karayalcin G 1991 Sonographic abnormalities of the thyroid gland in longterm survivors of Hodgkin disease. Pediatric Radiology 21 250–253. (https://doi.org/10.1007/BF02018615)

Search Google ScholarExport Citation

Sohn SY, Kim YN, Kim HI, Kim TH, Kim SW & Chung JH 2017 Validation of dynamic risk stratification in pediatric differentiated thyroid cancer. Endocrine 58 167–175. (https://doi.org/10.1007/s12020-017-1381-7)

Search Google ScholarExport Citation

Solt I, Gaitini D, Pery M, Hochberg Z, Stein M & Arush MW 2000 Comparing thyroid ultrasonography to thyroid function in long-term survivors of childhood lymphoma. Medical and Pediatric Oncology 35 35–40. (https://doi.org/10.1002/1096-911x(200007)35:1<35::aid-mpo6>3.0.co;2-#)

Search Google ScholarExport Citation

Sosa JA, Tuggle CT, Wang TS, Thomas DC, Boudourakis L, Rivkees S & Roman SA 2008 Clinical and economic outcomes of thyroid and parathyroid surgery in children. Journal of Clinical Endocrinology and Metabolism 93 3058–3065. (https://doi.org/10.1210/jc.2008-0660)

Search Google ScholarExport Citation

Spencer CA, Takeuchi M, Kazarosyan M, Wang CC, Guttler RB, Singer PA, Fatemi S, Lopresti JS & Nicoloff JT 1998 Serum thyroglobulin autoantibodies: prevalence, influence on serum thyroglobulin measurement, and prognostic significance in patients with differentiated thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 83 1121–1127. (https://doi.org/10.1210/jcem.83.4.4683)

Search Google ScholarExport Citation

Spencer C, Fatemi S, Singer P, Nicoloff J & Lopresti J 2010 Serum Basal thyroglobulin measured by a second-generation assay correlates with the recombinant human thyrotropin-stimulated thyroglobulin response in patients treated for differentiated thyroid cancer. Thyroid 20 587–595. (https://doi.org/10.1089/thy.2009.0338)

Search Google ScholarExport Citation

Spinelli C, Bertocchini A, Antonelli A & Miccoli P 2004 Surgical therapy of the thyroid papillary carcinoma in children: experience with 56 patients ≤16 years old. Journal of Pediatric Surgery 39 1500–1505. (https://doi.org/10.1016/j.jpedsurg.2004.06.016)

Search Google ScholarExport Citation

Spinelli C, Rossi L, Piscioneri J, Strambi S, Antonelli A, Ferrari A, Massimino M & Miccoli P 2016 Pediatric differentiated thyroid cancer: when to perform conservative and radical surgery. Current Pediatric Reviews 12 247–252. (https://doi.org/10.2174/1573396312666161014092023)

Search Google ScholarExport Citation

Spinelli C, Tognetti F, Strambi S, Morganti R, Massimino M & Collini P 2018 Cervical lymph node metastases of papillary thyroid carcinoma, in the central and lateral compartments, in children and adolescents: predictive factors. World Journal of Surgery 42 2444–2453. (https://doi.org/10.1007/s00268-018-4487-z)

Search Google ScholarExport Citation

Spinelli C, Rallo L, Morganti R, Mazzotti V, Inserra A, Cecchetto G, Massimino M, Collini P & Strambi S 2019 Surgical management of follicular thyroid carcinoma in children and adolescents: a study of 30 cases. Journal of Pediatric Surgery 54 521–526. (https://doi.org/10.1016/j.jpedsurg.2018.05.017)

Search Google ScholarExport Citation

Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I & Parkin DM 2006 Thyroid cancer incidence and survival among European children and adolescents (1978–1997): report from the Automated Childhood Cancer Information System project. European Journal of Cancer 42 2150–2169. (https://doi.org/10.1016/j.ejca.2006.06.001)

Search Google ScholarExport Citation

Stevens C, Lee JK, Sadatsafavi M & Blair GK 2009 Pediatric thyroid fine-needle aspiration cytology: a meta-analysis. Journal of Pediatric Surgery 44 2184–2191. (https://doi.org/10.1016/j.jpedsurg.2009.07.022)

Search Google ScholarExport Citation

Stewart DR, Best AF, Williams GM, Harney LA, Carr AG, Harris AK, Kratz CP, Dehner LP, Messinger YH, Rosenberg PS, et al.2019 Neoplasm risk among individuals with a pathogenic germline variant in DICER1. Journal of Clinical Oncology 37 668–676. (https://doi.org/10.1200/JCO.2018.78.4678)

Search Google ScholarExport Citation

Stosic A, Fuligni F, Anderson ND, Davidson S, De Borja R, Acker M, Forte V, Campisi P, Propst EJ, Wolter NE, et al.2021 Diverse oncogenic fusions and distinct gene expression patterns define the genomic landscape of pediatric papillary thyroid carcinoma. Cancer Research 81 5625–5637. (https://doi.org/10.1158/0008-5472.CAN-21-0761)

Search Google ScholarExport Citation

Stratakis CA, Courcoutsakis NA, Abati A, Filie A, Doppman JL, Carney JA & Shawker T 1997 Thyroid gland abnormalities in patients with the syndrome of spotty skin pigmentation, myxomas, endocrine overactivity, and schwannomas (Carney complex). Journal of Clinical Endocrinology and Metabolism 82 2037–2043. (https://doi.org/10.1210/jcem.82.7.4079)

Search Google ScholarExport Citation

Suchy B, Waldmann V, Klugbauer S & Rabes HM 1998 Absence of RAS and p53 mutations in thyroid carcinomas of children after Chernobyl in contrast to adult thyroid tumours. British Journal of Cancer 77 952–955. (https://doi.org/10.1038/bjc.1998.157)

Search Google ScholarExport Citation

Sugino K, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, Uruno T, Suzuki A, Akaishi J, Masaki C, Matsuzu K-I, et al.2015 Papillary thyroid carcinoma in children and adolescents: long-term follow-up and clinical characteristics. World Journal of Surgery 39 2259–2265. (https://doi.org/10.1007/s00268-015-3042-4)

Search Google ScholarExport Citation

Sung TY, Jeon MJ, Lee YH, Lee YM, Kwon H, Yoon JH, Chung KW, Kim WG, Song DE & Hong SJ 2017 Initial and dynamic risk stratification of pediatric patients with differentiated thyroid cancer. Journal of Clinical Endocrinology and Metabolism 102 793–800. (https://doi.org/10.1210/jc.2016-2666)

Search Google ScholarExport Citation

Suzuki S, Nakamura I, Suzuki S, Ohkouchi C, Mizunuma H, Midorikawa S, Fukushima T, Ito Y, Shimura H, Ohira T, et al.2016 Inappropriate suppression of thyrotropin concentrations in young patients with thyroid nodules including thyroid cancer: the Fukushima health management survey. Thyroid 26 717–725. (https://doi.org/10.1089/thy.2015.0499)

Search Google ScholarExport Citation

Taylor AJ, Croft AP, Palace AM, Winter DL, Reulen RC, Stiller CA, Stevens MC & Hawkins MM 2009 Risk of thyroid cancer in survivors of childhood cancer: results from the British Childhood Cancer Survivor Study. International Journal of Cancer 125 2400–2405. (https://doi.org/10.1002/ijc.24581)

Search Google ScholarExport Citation

Trahan J, Reddy A, Chang E, Gomez R, Prasad P & Jeyakumar A 2016 Pediatric thyroid nodules: a single center experience. International Journal of Pediatric Otorhinolaryngology 87 94–97. (https://doi.org/10.1016/j.ijporl.2016.06.011)

Search Google ScholarExport Citation

Tuggle CT, Roman SA, Wang TS, Boudourakis L, Thomas DC, Udelsman R & Ann Sosa J 2008 Pediatric endocrine surgery: who is operating on our children? Surgery 144 869–877; discussion 877. (https://doi.org/10.1016/j.surg.2008.08.033)

Search Google ScholarExport Citation

Vaisman F, Bulzico DA, Pessoa CHCN, Bordallo MAN, Mendonça UBTD, Dias FL, Coeli CM, Corbo R & Vaisman M 2011 Prognostic factors of a good response to initial therapy in children and adolescents with differentiated thyroid cancer. Clinics 66 281–286. (https://doi.org/10.1590/s1807-59322011000200017)

Search Google ScholarExport Citation

Vali R, Rachmiel M, Hamilton J, El Zein M, Wasserman J, Costantini DL, Charron M & Daneman A 2015 The role of ultrasound in the follow-up of children with differentiated thyroid cancer. Pediatric Radiology 45 1039–1045. (https://doi.org/10.1007/s00247-014-3261-0)

Search Google ScholarExport Citation

Van Santen HM, Aronson DC, Vulsma T, Tummers RF, Geenen MM, De Vijlder JJ & Van Den Bos C 2004 Frequent adverse events after treatment for childhood-onset differentiated thyroid carcinoma: a single institute experience. European Journal of Cancer 40 1743–1751. (https://doi.org/10.1016/j.ejca.2004.03.006)

Search Google ScholarExport Citation

Vassilopoulou-Sellin R, Klein MJ, Smith TH, Samaan NA, Frankenthaler RA, Goepfert H, Cangir A & Haynie TP 1993 Pulmonary metastases in children and young adults with differentiated thyroid cancer. Cancer 71 1348–1352. (https://doi.org/10.1002/1097-0142(19930215)71:4<1348::aid-cncr2820710429>3.0.co;2-3)

Search Google ScholarExport Citation

Vassilopoulou-Sellin R, Goepfert H, Raney B & Schultz PN 1998 Differentiated thyroid cancer in children and adolescents clinical outcome and mortality after long-term follow-up. Head and Neck 20 549–555. (https://doi.org/10.1002/(sici)1097-0347(199809)20:6<549::aid-hed10>3.0.co;2-r)

Search Google ScholarExport Citation

Veiga LH, Lubin JH, Anderson H, De Vathaire F, Tucker M, Bhatti P, Schneider A, Johansson R, Inskip P, Kleinerman R, et al.2012 A pooled analysis of thyroid cancer incidence following radiotherapy for childhood cancer. Radiation Research 178 365–376. (https://doi.org/10.1667/rr2889.1)

Search Google ScholarExport Citation

Verburg FA, Biko J, Diessl S, Demidchik Y, Drozd V, Rivkees SA, Reiners C & Hanscheid H 2011 I-131 activities as high as safely administrable (AHASA) for the treatment of children and adolescents with advanced differentiated thyroid cancer. Journal of Clinical Endocrinology and Metabolism 96 E1268–E1271. (https://doi.org/10.1210/jc.2011-0520)

Search Google ScholarExport Citation

Verburg FA, Reiners C & Hanscheid H 2013 Approach to the patient: role of dosimetric RAI Rx in children with DTC. Journal of Clinical Endocrinology and Metabolism 98 3912–3919. (https://doi.org/10.1210/jc.2013-2259)

Search Google ScholarExport Citation

Vergamini LB, Frazier AL, Abrantes FL, Ribeiro KB & Rodriguez-Galindo C 2014 Increase in the incidence of differentiated thyroid carcinoma in children, adolescents, and young adults: a population-based study. Journal of Pediatrics 164 1481–1485. (https://doi.org/10.1016/j.jpeds.2014.01.059)

Search Google ScholarExport Citation

Verloop H, Louwerens M, Schoones JW, Kievit J, Smit JW & Dekkers OM 2012 Risk of hypothyroidism following hemithyroidectomy: systematic review and meta-analysis of prognostic studies. Journal of Clinical Endocrinology and Metabolism 97 2243–2255. (https://doi.org/10.1210/jc.2012-1063)

Search Google ScholarExport Citation

Vriens MR, Suh I, Moses W & Kebebew E 2009 Clinical features and genetic predisposition to hereditary nonmedullary thyroid cancer. Thyroid 19 1343–1349. (https://doi.org/10.1089/thy.2009.1607)

Search Google ScholarExport Citation

Vriens MR, Moses W, Weng J, Peng M, Griffin A, Bleyer A, Pollock BH, Indelicato DJ, Hwang J & Kebebew E 2011 Clinical and molecular features of papillary thyroid cancer in adolescents and young adults. Cancer 117 259–267. (https://doi.org/10.1002/cncr.25369)

Search Google ScholarExport Citation

Vuong HG, Chung DGB, Ngo LM, Bui TQ, Hassell L, Jung CK, Kakudo K & Bychkov A 2021 The use of the Bethesda system for reporting thyroid cytopathology in pediatric thyroid nodules: a meta-analysis. Thyroid 31 1203–1211. (https://doi.org/10.1089/thy.2020.0702)

Search Google ScholarExport Citation

Wada N, Sugino K, Mimura T, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, Nakayama H, Hirakawa S, Rino Y, et al.2009 Pediatric differentiated thyroid carcinoma in stage I: risk factor analysis for disease free survival. BMC Cancer 9 306. (https://doi.org/10.1186/1471-2407-9-306)

Search Google ScholarExport Citation

Wallace WH 2011 Oncofertility and preservation of reproductive capacity in children and young adults. Cancer 117 (Supplement) 2301–2310. (https://doi.org/10.1002/cncr.26045)

Search Google ScholarExport Citation

Wang H, Dai H, Li Q, Shen G, Shi L & Tian R 2021 Investigating (18)F-FDG PET/CT parameters as prognostic markers for differentiated thyroid cancer: a systematic review. Frontiers in Oncology 11 648658. (https://doi.org/10.3389/fonc.2021.648658)

Search Google ScholarExport Citation

Wang Q, Chang Q, Zhang R, Sun C, Li L, Wang S, Wang Q, Li Z & Niu L 2022 Diffuse sclerosing variant of papillary thyroid carcinoma: ultrasonographic and clinicopathological features in children/adolescents and adults. Clinical Radiology 77 e356–e362. (https://doi.org/10.1016/j.crad.2022.01.051)

Search Google ScholarExport Citation

Welch-Dinauer CA, Tuttle RM, Robie DK, McClellan DR, Svec RL, Adair C, Francis GL & He E 1998 Clinical features associated with metastasis and recurrence of differentiated thyroid cancer in children , adolescents and young adults. Clinical Endocrinology 49 619–628. (https://doi.org/10.1046/j.1365-2265.1998.00584.x)

Search Google ScholarExport Citation

Welch Dinauer CA, Tuttle RM, Robie DK, McClellan DR & Francis GL 1999 Extensive surgery improves recurrence-free survival for children and young patients with class I papillary thyroid carcinoma. Journal of Pediatric Surgery 34 1799–1804. (https://doi.org/10.1016/s0022-3468(9990316-0)

Search Google ScholarExport Citation

Wood JH, Partrick DA, Barham HP, Bensard DD, Travers SH, Bruny JL & McIntyre Jr RC 2011 Pediatric thyroidectomy: a collaborative surgical approach. Journal of Pediatric Surgery 46 823–828. (https://doi.org/10.1016/j.jpedsurg.2011.02.013)

Search Google ScholarExport Citation

Xu L, Liu Q, Liu Y & Pang H 2016 Parameters influencing curative effect of 131I therapy on pediatric differentiated thyroid carcinoma: a retrospective study. Medical Science Monitor 22 3079–3085. (https://doi.org/10.12659/msm.896876)

Search Google ScholarExport Citation

Yamashita S & Saenko V 2007 Mechanisms of Disease: molecular genetics of childhood thyroid cancers. Nature Clinical Practice: Endocrinology and Metabolism 3 422–429. (https://doi.org/10.1038/ncpendmet0499)

Search Google ScholarExport Citation

Zimmerman D, Hay ID, Gough IR, Goellner JR, Ryan JJ, Grant CS & McConahey WM 1988 Papillary thyroid carcinoma in children and adults: long-term follow-up of 1039 patients conservatively treated at one institution during three decades. Surgery 104 1157–1166.

Search Google ScholarExport Citation

Zivaljevic V, Tausanovic K, Sipetic S, Paunovic I, Diklic A, Kovacevic B, Stojanovic D, Zivic R, Stanojevic B & Kalezic N 2013 A case-control study of papillary thyroid cancer in children and adolescents. European Journal of Cancer Prevention 22 561–565. (https://doi.org/10.1097/CEJ.0b013e3283603494)

Search Google ScholarExport Citation


#
Medicine by Alexandros G. Sfakianakis,Anapafseos 5 Agios Nikolaos 72100 Crete Greece,00302841026182,00306932607174,alsfakia@gmail.com,