Childhood differentiated thyroid carcinoma: clinical course

Preview:

Citation preview

University of Groningen

Childhood differentiated thyroid carcinoma: clinical course and late effects of treatmentNies, Marloes

DOI:10.33612/diss.145080681

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Nies, M. (2020). Childhood differentiated thyroid carcinoma: clinical course and late effects of treatment.University of Groningen. https://doi.org/10.33612/diss.145080681

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license.More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne-amendment.

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 31-07-2022

CHILDHOOD DIFFERENTIATED THYROID CARCINOMA: CLINICAL COURSE AND

LATE EFFECTS OF TREATMENT

Marloes Nies

Childhood differentiated thyroid carcinoma: clinical course and late

effects of treatment

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de rector magnificus prof. dr. C. Wijmenga

en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op

woensdag 2 december 2020 om 16.15 uur

door

Marloes Nies

geboren op 9 mei 1991 te Enschede

Childhood differentiated thyroid carcinoma: clinical course and late effects of treatment

© Marloes Nies, 2020

All rights reserved. No part of this thesis may be reproduced, stored in a retrieval system or transmitted

in any form or by any means, without prior written permission of the authors.

The research in this thesis was financially supported by the Junior Scientific Masterclass Groningen,

Children Cancer-free Foundation, UMCG Cancer Research Fund, van der Meer-Boerema Foundation,

Prince Bernhard Culture Fund and Academy Ter Meulen Grant of the Royal Netherlands Academy of Arts

and Sciences.

Financial support for printing of this thesis was kindly provided by the University of Groningen Medical

Center, the University of Groningen, the Graduate School of Medical Sciences of the University of

Groningen, and the Endocrinology Fund (as part of the Ubbo Emmius Fund).

Cover, layout and printing: Off Page, Amsterdam

Childhood differentiated thyroid carcinoma: clinical course and late

effects of treatment

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de rector magnificus prof. dr. C. Wijmenga

en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op

woensdag 2 december 2020 om 16.15 uur

door

Marloes Nies

geboren op 9 mei 1991 te Enschede

Promotores Prof. dr. T.P. Links Prof. dr. W.J.E. Tissing

Copromotor

Dr. G. Bocca

Beoordelingscommissie

Prof. dr. R. Sanderman

Prof. dr. A. Hoek

Prof. dr. C.M. Zwaan

Paranimfen Tim Raveling Annet Wijnen

Promotores Prof. dr. T.P. Links Prof. dr. W.J.E. Tissing

Copromotor

Dr. G. Bocca

Beoordelingscommissie

Prof. dr. R. Sanderman

Prof. dr. A. Hoek

Prof. dr. C.M. Zwaan

Paranimfen Tim Raveling Annet Wijnen

TABLE OF CONTENTS

Chapter 1 General introduction 9

Chapter 2 Pediatric differentiated thyroid carcinoma in the Netherlands: 21 a nationwide follow-up study

Chapter 3 Distant metastases from childhood differentiated thyroid 43 carcinoma: clinical course and mutational landscape

Chapter 4 Long-term effects of radioiodine treatment on female fertility 79 in survivors of childhood differentiated thyroid carcinoma

Chapter 5 Long-term evaluation of male fertility after treatment with 99 radioactive iodine for differentiated thyroid carcinoma

Chapter 6 Cardiac dysfunction in survivors of pediatric differentiated 125 thyroid carcinoma

Chapter 7 Long-term quality of life in adult survivors of pediatric 139 differentiated thyroid carcinoma

Chapter 8 Psychosocial development in survivors of childhood 173 differentiated thyroid carcinoma: a cross-sectional study

Chapter 9 Summary and general discussion 205

Chapter 10 Appendices 227Nederlandse samenvatting 229Nederlandse samenvatting voor leken 235Dankwoord 241About the author 244List of publications 245

Chapter 1

Chapter 1

CHAPTER 1

General introduction

GENERAL INTRODUCTION

11

1The thyroid glandThe thyroid is an endocrine gland, located ventrally from the trachea. The gland consists of two lobes that are connected through the isthmus. The thyroid is composed of follicles, follicular cells, and parafollicular cells (also called C cells) (1). The follicular cells produce thyroxin (T4, thyroid hormone) and triiodothyronine (T3), which are composed of iodine and thyroglobulin (Tg, a precursor protein from the thyroid). T4 is the sole product of the thyroid gland, whereas T3 is produced in the thyroid and peripheral organs by deiodination of T4. Thyroid hormones are involved in a wide range of mechanisms within the body, and affect basal metabolic activity, growth, and neural development (2). Calcitonin, a hormone produced by the C cells and this hormone decreases the blood calcium concentration (3). The hypothalamic-pituitary-thyroid axis regulates the synthesis of the thyroid hormones. The hypothalamus releases thyrotropin-releasing hormone (TRH), causing the anterior pituitary gland to secrete thyroid-stimulating hormone (TSH). This results in thyroid hormone synthesis and secretion. T3 and T4 subsequently provide negative feedback to the hypothalamus and the pituitary gland (2).

Thyroid cancer and differentiated thyroid cancerThyroid cancer ensues when cells of the thyroid gland reproduce uncontrollably and develop the potential to spread (metastasize). Histologically, the most common subtype of thyroid cancer is (well-)differentiated thyroid cancer (DTC, which includes papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC)). DTC accounts for 90% of all thyroid cancers (4, 5). Differentiated cancers derive from the follicular cells. DTC can occur at all ages, but its peak incidence is from the 3rd to 5th life decades (4). Poorly differentiated thyroid cancers, such as medullary thyroid cancer (MTC, arising from C cells) and anaplastic thyroid cancer (ATC, arising from the follicular epithelium) are less common (5, 6).

Differentiated thyroid cancer in childrenDTC in children (diagnosed before the age of 19 years) is rare, but incidence rates are increasing (7). Age-adjusted incidence rates of childhood DTC are 0.6 to 11.0 per 100,000, varying between age group and country of origin (7-9). Most children diagnosed with DTC are post-pubertal. Up to puberty, the incidence of DTC in boys and girls is similar, but from puberty onwards most patients are female (6), making female sex the most important risk factor for DTC. The explanation for this sex-dependent diagnosis probably lies within the proliferative effect of estrogen on thyroid cells, but the exact mechanism is not completely understood (10-12). The emergence of thyroid cancer cannot always be explained, but known risk factors for developing childhood thyroid cancer are exposure to radiation, iodine deficiency, a positive family history for DTC, gene rearrangements, or a thyroid cancer syndrome (13-15).

CHAPTER 1

12

1Symptoms of childhood differentiated thyroid cancerChildren most often present with an asymptomatic solitary thyroid nodule or neck mass. Compressive symptoms, such as hoarseness, dysphagia, dyspnea, or experiencing a choking sensation are less common (16-18).

Diagnosis and treatment of childhood differentiated thyroid cancerUp to now, three official guidelines for the management of DTC in children have been published (19-21). Clinical evaluation, ultrasonography (US), and fine needle aspiration (FNA) are used to determine the origin of the thyroid nodule or neck mass (19). FNA can be makes it possible to obtain cells of the thyroid nodule and/or suspicious lymph node. The FNA of the thyroid nodule can then be evaluated according to the pathologic Bethesda System for Reporting Thyroid Cytopathology (22). Six different Bethesda diagnostic categories determine the follow-up measures, which range from clinical follow-up to surgical intervention (19, 22). When FNA indicates a strong possibility of malignant cells, treatment in children generally consists of a total thyroidectomy. Depending on the presence and the site of metastases, a central or (bi)lateral lymph node dissection can be performed (19). Children are postoperatively staged by means of the tumor-node-metastasis (TNM) classification (23, 24) and corresponding risk level of the disease (19), which determine the consecutive (intensity of the) treatment. After surgery, radioactive iodine (131I) can be administered. When administered in a high dose, the beta radiation of this radioisotope of iodine destroys thyroid cells (25), and may decrease the risk of recurrence of the disease (17, 26, 27). Although the precise role of 131I during treatment of low risk DTC has not yet been defined, the additional value of its administration in children with advanced disease is more established (27, 28). Subsequently, thyroid hormone supplementation with levothyroxine compensates the lack of thyroid hormone resulting from the thyroidectomy, but is also used to induce a certain level of TSH suppression (TSH suppression therapy). The aim of TSH suppression therapy is to suppress the growth-promoting effect of TSH on the thyroid cells, thereby preventing the (re)growth of malignant cells (29, 30). In high risk patients, a more intensive TSH suppression is advised. Recommendations are based on findings in adults, since no studies have as yet focused solely on evaluating children (19). Follow-up of the disease consists of clinical evaluation and neck palpation, US, and measuring of Tg during the thyroid hormone suppletion. Tg serves as a marker for residual or recurrent disease (19).

Outcome after treatment of childhood differentiated thyroid cancerSubsequent to treatment, survival rates of childhood DTC are up to 99% after 30 years of follow-up (6, 31). Although survival in children is excellent, a relatively high percentage (10 to 30%) of the children develops recurrent disease, occurring even decades after diagnosis (30, 32-34). Moreover, after treatment some patients still have

GENERAL INTRODUCTION

13

1evidence of disease, which is called persistent disease. Recurrent or persistent disease occurs more frequently in patients with advanced disease upon diagnosis (35, 36).

Differentiated thyroid cancer in children and adultsIn the past, DTC was presumed to be similar in children and adults. However, more advanced knowledge indicates great differences between DTC in children and adults.

Upon diagnosis, children present with more advanced and aggressive disease than do adults. Paradoxically, children have better overall survival rates in children than adults, but also more frequent persistent disease and recurrences (16-18, 31, 32, 34, 37-43). In childhood, the mutational landscape of DTC differs from that of adults (44-54). Table 1 presents an overview an overview of differences between DTC diagnosed during childhood and adulthood.

To date, however, no clear explanation can account for these differences between adult and childhood DTC. Although genetic alterations may play a role, studies are not conclusive. A higher expression of the sodium iodine symporter (NIS, essential in the uptake of iodine) in children may also help to explain their better responsiveness to 131I administrations, but ultimately the origin of the difference between adult and childhood DTC is probably multifactorial.

Adverse effects after childhood cancer Unfortunately, survivors of (childhood) cancer experience unwanted effects of the (treatment of the) cancer. These side effects are being increasingly recognized, as recent decades have seen an increase in the overall survival rate of childhood cancer (60). Side effects can occur during treatment, but may sometimes become manifest only years later. These late effects can be physical, mental, and/or psychosocial, such as cognitive impairment, fertility problems, diagnosis of a secondary malignancy,

Table 1. Differences between DTC diagnosed during childhood and adulthood

Childhood DTC Adult DTC

Malignant origin of thyroid nodules (16, 18, 39, 40) 19 to 26% 12 to 14%

Incidence of lymph node metastases (17, 31, 34, 41-43, 55) 40 to 90% 15 to 50%

Incidence of distant metastases (17, 56, 57) 20 to 30% 2 to 20%

Most prevalent mutational alteration (44-54) RET fusion BRAF V600E mutation

Recurrence rate (32, 34, 58) Up to 32% 5%

Rate of persistent disease (32, 36, 58, 59) 5 to 33% 2 to 3%

10-year survival rate (17, 32, 37, 38) 95 to 100% 85 to 91%

Abbreviations: DTC, differentiated thyroid carcinoma; RET, rearranged during transfection; BRAF, v-raf murine sarcoma

viral oncogene homolog B.

CHAPTER 1

14

1and fatigue (61). Depending on the type of late effect, treatment or support can be offered, but not all effects can be prevented or resolved.

Adverse and late effects after childhood differentiated thyroid cancerBecause the majority of childhood DTC patients will survive their disease, it is important to evaluate late effects in these survivors. However, in contrast to the knowledge of late effects in many other childhood malignancies, little is known about possible adverse effects of childhood DTC.

During treatment of DTC, surgical complications like surgical site infection, parathyroid damage (causing hypocalcaemia) and recurrent laryngeal nerve injury (causing hoarseness or loss of voice) can occur (62, 63). In addition, short-term side effects of 131I administration are radiation thyroiditis, nausea, vomiting, sialadenitis, gastro-intestinal symptoms, and bone-marrow suppression. In the long-term, administration of 131I for adult DTC is associated with salivary dysfunction or sialadenitis, pulmonary fibrosis, secondary malignancies, and gonadal damage in both men and women (causing fertility problems) (32, 64-69). Other long-term effects possibly induced by TSH suppression therapy are cardiovascular deterioration and loss of bone mineral density (also influenced by hypoparathyroidism) (29, 70-74). Moreover, general well-being or quality of life (QoL) can be affected by the diagnosis and the treatment of DTC (75-78).

Some studies have been performed in survivors of childhood DTC, but current knowledge is based mainly on late effects of DTC on adults. Because of the differences between childhood and adult DTC, as shown above, late effects may also differ. However, specific knowledge of the late effects of treatment for DTC during childhood is limited because of the scarcity of studies, the small number of patients evaluated, the lack of clear study definitions, or the poor quality of study designs.

Aims and outline of this thesisThe aim of the current thesis is to evaluate the clinical course and late effects of childhood DTC. The results will ultimately benefit newly diagnosed patients, patients previously treated for DTC, caregivers, and treating physicians.

A multicenter, cross-sectional study was conducted in the Netherlands. Patients diagnosed with DTC before the age of 19 years between 1970 and 2013 were included. Chapter 2 consists of an overview of the disease, treatment, outcomes, and follow-up characteristics of these patients. A minority of patients had distant metastases (DM). Chapter 3 specifically evaluates the clinical course of DTC in a large cohort of childhood DTC patients diagnosed with DM. This study was performed at the University of Texas MD Anderson Cancer Center in the United States.

Long-term treatment effects of 131I after childhood DTC in the Netherlands are evaluated in the subsequent chapters. Female fertility after treatment is studied in

GENERAL INTRODUCTION

15

1Chapter 4, where we evaluate reproductive characteristics in female survivors of childhood DTC, combined with levels of Anti-Müllerian hormone (AMH, a marker of ovarian reserve). Because the minority of childhood DTC patients is male, to attain a substantial and representative group of survivors, Chapter 5 includes a study of male fertility after treatment in survivors of adult DTC. Male fertility was evaluated by performing semen analyses, and assessing reproductive hormones and reproductive characteristics. Adverse effects of long-term TSH suppression therapy are evaluated in Chapter 6, including effects on cardiac function in survivors of childhood DTC. The first evaluation of these patients, performed five years after their childhood DTC diagnosis, showed that 21% of the survivors had asymptomatic diastolic dysfunction (79). Chapter 6 includes a re-evaluated of patients after a total follow-up period of 10 years to assess the course of their cardiac function. In Chapter 7, long-term thyroid cancer-specific QoL, health-related QoL, fatigue, and anxiety and depression are evaluated in survivors who were at least 5 years in follow-up after diagnosis. Because childhood cancer has been known to disrupt the course of life, Chapter 8 evaluates psychosocial developmental milestones in childhood DTC survivors. Chapter 9 contains the summary and general discussion of this thesis, and suggests it implications.

CHAPTER 1

16

1 REFERENCES 1. Schünke M, Schulte E, Schumacher U. Prometheus anatomische atlas 2 - Hoofd, hals en neuroanatomie. 2 ed.

Houten: Bohn Stafleu van Loghum; 2010. 200-203 p.

2. Brent GA. Mechanisms of thyroid hormone action. J Clin Invest. 2012;122(9):3035-3043.

3. Morton DA, Foreman KB, Albertine KH. Viscera of the Neck. The Big Picture: Gross Anatomy, 2e. New York, NY:

McGraw-Hill Education; 2019.

4. NKR Cijfers [Internet]. Integraal Kankercentrum Nederland. 2020 [cited April 2020]. Available from: https://www.

iknl.nl/nkr-cijfers.

5. Davies L, Welch HG. Increasing incidence of thyroid cancer in the United States, 1973-2002. JAMA.

2006;295(18):2164-2167.

6. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167-172.

7. Schmidt Jensen J, Gronhoj C, Mirian C, Jensen DH, Friborg J, Hahn CH, Agander TK, Hjuler T. Incidence and

survival of thyroid cancer in children, adolescents, and young adults in Denmark: a nationwide study from 1980 to

2014. Thyroid. 2018;28(9):1128-1133.

8. Dermody S, Walls A, Harley EH, Jr. Pediatric thyroid cancer: an update from the SEER database 2007-2012. Int J

Pediatr Otorhinolaryngol. 2016;89:121-126.

9. Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I, Parkin DM. Thyroid cancer incidence and survival

among European children and adolescents (1978-1997): report from the Automated Childhood Cancer Information

System project. Eur J Cancer. 2006;42(13):2150-2169.

10. Manole D, Schildknecht B, Gosnell B, Adams E, Derwahl M. Estrogen promotes growth of human thyroid tumor

cells by different molecular mechanisms. J Clin Endocrinol Metab. 2001;86(3):1072-1077.

11. Juvenal G, Christophe D, Roger P, Pisarev M. Thyroid function and growth regulation under normal and abnormal

conditions. J Thyroid Res. 2011;2011:805036-805036.

12. Derwahl M, Nicula D. Estrogen and its role in thyroid cancer. Endocr Relat Cancer. 2014;21(5):T273-283.

13. Liu Y, Su L, Xiao H. Review of factors related to the thyroid cancer epidemic. Int J Endocrinol. 2017;2017:5308635-5308635.

14. Tucker MA, Jones PHM, Boice JD, Robison LL, Stone BJ, Stovall M, Jenkin RDT, Lubin JH, Baum ES, Siegel SE,

Meadows AT, Hoover RN, Fraumeni JF, the Late Effects Study G. Therapeutic radiation at a young age is linked to

secondary thyroid cancer. Cancer Res. 1991;51(11):2885.

15. Zimmermann MB, Galetti V. Iodine intake as a risk factor for thyroid cancer: a comprehensive review of animal and

human studies. Thyroid Res. 2015;8(1):8.

16. Niedziela M. Pathogenesis, diagnosis and management of thyroid nodules in children. Endocr Relat

Cancer. 2006;13(2):427-453.

17. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA, Udelsman R. The treatment of

differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocr

Rev. 2011;32(6):798-826.

18. Bessey LJ, Lai NBK, Coorough NE, Chen H, Sippel RS. The incidence of thyroid cancer by fine needle aspiration

varies by age and gender. J Surg Res. 2013;184(2):761-765.

19. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management guidelines for children with thyroid nodules and differentiated thyroid cancer.

Thyroid. 2015;25(7):716-759.

20. Jarzab B, Dedecjus M, Slowinska-Klencka D, Lewinski A, Adamczewski Z, Anielski R, Baglaj M, Baldys-Waligorska

A, Barczynski M, Bednarczuk T, Bossowski A, Buziak-Bereza M, Chmielik E, Cichocki A, Czarniecka A, Czepczynski

R, Dzieciol J, Gawlik T, Handkiewicz-Junak D, Hasse-Lazar K, Hubalewska-Dydejczyk A, Jazdzewski K, Jurecka-

Lubieniecka B, Kalemba M, Kaminski G, Karbownik-Lewinska M, Klencki M, Kos-Kudla B, Kotecka-Blicharz

A, Kowalska A, Krajewska J, Kropinska A, Kukulska A, Kulik E, Kulakowski A, Kuzdak K, Lange D, Ledwon A,

Lewandowska-Jablonska E, Lacka K, Michalik B, Nasierowska-Guttmejer A, Nauman J, Niedziela M, Malecka-

Tendera E, Oczko-Wojciechowska M, Olczyk T, Paliczka-Cieslik E, Pomorski L, Puch Z, Roskosz J, Ruchala M,

Rusinek D, Sporny S, Stanek-Widera A, Stojcev Z, Sygula A, Syrenicz A, Szpak-Ulczok S, Tomkalski T, Wygoda

GENERAL INTRODUCTION

17

1Z, Wloch J, Zembala-Nozynska E. Guidelines of Polish National Societies diagnostics and treatment of thyroid

carcinoma. 2018 Update. Endokrynol Pol. 2018;69(1):34-74.

21. Lebbink CA, Dekker BL, Bocca G, Braat AJ, Derikx JP, Dierselhuis MP, de Keizer B, Kruijff S, Kwast AB, van Nederveen

FH, Nieveen van Dijkum E, Nievelstein RA, Peeters RP, Terwisscha van Scheltinga CE, Tissing WJ, van der Tuin K, Vriens

MR, Zsiros J, van Trotsenburg AS, Links TP, van Santen HM. New national recommendations for the treatment of

pediatric differentiated thyroid carcinoma in the Netherlands. Eur J Endocrinol. 2020;183(4):P11-P18.

22. Cibas ES, Ali SZ. The 2017 Bethesda System for reporting thyroid cytopathology. Thyroid. 2017;27(11):1341-1346.

23. Sobin LH, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours: John Wiley & Sons; 2009.

24. Brierley JD, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours: Wiley; 2017.

25. Preedy VR, Burrow GN, Watson RR. Comprehensive handbook of iodine: nutritional, biochemical, pathological

and therapeutic aspects: Academic Press; 2009.

26. Handkiewicz-Junak D, Wloch J, Roskosz J, Krajewska J, Kropinska A, Pomorski L, Kukulska A, Prokurat A, Wygoda

Z, Jarzab B. Total thyroidectomy and adjuvant radioiodine treatment independently decrease locoregional

recurrence risk in childhood and adolescent differentiated thyroid cancer. J Nucl Med. 2007;48(6):879-888.

27. Jarzab B, Handkiewicz-Junak D, Wloch J. Juvenile differentiated thyroid carcinoma and the role of radioiodine in

its treatment: a qualitative review. Endocr Relat Cancer. 2005;12(4):773-803.

28. Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Mang O, Lau WH. Differentiated thyroid carcinoma in childhood

and adolescence-clinical course and role of radioiodine. Pediatr Blood Cancer. 2004;42(2):176-183.

29. Brabant G. Thyrotropin suppressive therapy in thyroid carcinoma: what are the targets? J Clin Endocrinol

Metab. 2008;93(4):1167-1169.

30. Landau D, Vini L, A’Hern R, Harmer C. Thyroid cancer in children: the Royal Marsden Hospital experience. Eur J

Cancer. 2000;36(2):214-220.

31. Hay ID, Johnson TR, Kaggal S, Reinalda MS, Iniguez-Ariza NM, Grant CS, Pittock ST, Thompson GB. Papillary

thyroid carcinoma (PTC) in children and adults: comparison of initial presentation and long-term postoperative

outcome in 4432 patients consecutively treated at the Mayo Clinic during eight decades (1936-2015). World J

Surg. 2018;42(2):329-342.

32. Hay ID, Gonzalez-Losada T, Reinalda MS, Honetschlager JA, Richards ML, Thompson GB. Long-term outcome

in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J

Surg. 2010;34(6):1192-1202.

33. Grigsby PW, Gal-or A, Michalski JM, Doherty GM. Childhood and adolescent thyroid carcinoma. Cancer. 2002;95(4):724-729.

34. Welch Dinauer CA, Tuttle RM, Robie DK, McClellan DR, Svec RL, Adair C, Francis GL. Clinical features associated

with metastasis and recurrence of differentiated thyroid cancer in children, adolescents and young adults. Clin

Endocrinol (Oxf). 1998;49(5):619-628.

35. Russo M, Malandrino P, Moleti M, Vermiglio F, D’Angelo A, La Rosa G, Sapuppo G, Calaciura F, Regalbuto C, Belfiore

A, Vigneri R, Pellegriti G. Differentiated thyroid cancer in children: heterogeneity of predictive risk factors. Pediatr

Blood Cancer. 2018;65(9):e27226.

36. Verburg FA, de Keizer B, Lam MGEH, de Klerk JMH, Lips CJM, Borel-Rinkes IHM, van Isselt JW. Persistent disease in

patients with papillary thyroid carcinoma and lymph node metastases after surgery and Iodine-131 ablation. World

J Surg. 2007;31(12):2309-2314.

37. Eustatia-Rutten CFA, Corssmit EPM, Biermasz NR, Pereira AM, Romijn JA, Smit JW. Survival and death causes in

differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2006;91(1):313-319.

38. Links TP, van Tol KM, Jager PL, Plukker JT, Piers DA, Boezen HM, Dullaart RP, de Vries EG, Sluiter WJ. Life expectancy

in differentiated thyroid cancer: a novel approach to survival analysis. Endocr Relat Cancer. 2005;12(2):273-280.

39. Cherella CE, Angell TE, Richman DM, Frates MC, Benson CB, Moore FD, Barletta JA, Hollowell M, Smith JR,

Alexander EK, Cibas ES, Wassner AJ. Differences in thyroid nodule cytology and malignancy risk between children

and adults. Thyroid. 2019;29(8):1097-1104.

40. Gupta A, Ly S, Castroneves LA, Frates MC, Benson CB, Feldman HA, Wassner AJ, Smith JR, Marqusee E, Alexander

EK, Barletta J, Doubilet PM, Peters HE, Webb S, Modi BP, Paltiel HJ, Kozakewich H, Cibas ES, Moore FD, Jr.,

Shamberger RC, Larsen PR, Huang SA. A standardized assessment of thyroid nodules in children confirms higher

cancer prevalence than in adults. J Clin Endocrinol Metab. 2013;98(8):3238-3245.

41. Jarzab B, Handkiewicz-Junak D. Differentiated thyroid cancer in children and adults: same or distinct disease?

Hormones (Athens, Greece). 2007;6(3):200-209.

CHAPTER 1

18

142. LaFranchi SH. Inaugural management guidelines for children with thyroid nodules and differentiated thyroid

cancer: children are not small adults. Thyroid. 2015;25(7):713-715.

43. Vaisman F, Corbo R, Vaisman M. Thyroid carcinoma in children and adolescents-systematic review of the literature.

J Thyroid Res. 2011;2011:845362.

44. Yamashita S, Saenko V. Mechanisms of disease: molecular genetics of childhood thyroid cancers. Nat Clin Pract

Endocrinol Metab. 2007;3(5):422-429.

45. Fenton C, Anderson J, Lukes Y, Dinauer CA, Tuttle RM, Francis GL. Ras mutations are uncommon in sporadic

thyroid cancer in children and young adults. J Endocrinol Invest. 1999;22(10):781-789.

46. Fenton CL, Lukes Y, Nicholson D, Dinauer CA, Francis GL, Tuttle RM. The ret/PTC mutations are common in

sporadic papillary thyroid carcinoma of children and young adults. J Clin Endocrinol Metab. 2000;85(3):1170-1175.

47. Cordioli MI, Moraes L, Bastos AU, Besson P, Alves MT, Delcelo R, Monte O, Longui C, Cury AN, Cerutti JM.

Fusion oncogenes are the main genetic events found in sporadic papillary thyroid carcinomas from children.

Thyroid. 2017;27(2):182-188.

48. Diesen DL, Skinner MA. Pediatric thyroid cancer. Semin Pediatr Surg. 2012;21(1):44-50.

49. Sisdelli L, Cordioli M, Vaisman F, Moraes L, Colozza-Gama GA, Alves PAG, Jr., Araujo ML, Jr., Alves MTS, Monte O,

Longui CA, Cury AN, Carvalheira G, Cerutti JM. AGK-BRAF is associated with distant metastasis and younger age in

pediatric papillary thyroid carcinoma. Pediatr Blood Cancer. 2019;66(7):e27707.

50. Galuppini F, Vianello F, Censi S, Barollo S, Bertazza L, Carducci S, Colato C, Manso J, Rugge M, Iacobone M,

Watutantrige Fernando S, Pennelli G, Mian C. Differentiated thyroid carcinoma in pediatric age: genetic and

clinical scenario. Front Endocrinol (Lausanne). 2019;10:552.

51. Faggiano A, Coulot J, Bellon N, Talbot M, Caillou B, Ricard M, Bidart JM, Schlumberger M. Age-dependent variation

of follicular size and expression of iodine transporters in human thyroid tissue. J Nucl Med. 2004;45(2):232-237.

52. Pekova B, Dvorakova S, Sykorova V, Vacinova G, Vaclavikova E, Moravcova J, Katra R, Vlcek P, Sykorova P, Kodetova D, Vcelak

J, Bendlova B. Somatic genetic alterations in a large cohort of pediatric thyroid nodules. Endocr Connect. 2019;8(6):796-805

53. Alzahrani AS, Murugan AK, Qasem E, Alswailem M, Al-Hindi H, Shi Y. Single point mutations in pediatric

differentiated thyroid cancer. Thyroid. 2017;27(2):189-196.

54. Mostoufi-Moab S, Labourier E, Sullivan L, LiVolsi V, Li Y, Xiao R, Beaudenon-Huibregtse S, Kazahaya K, Adzick NS, Baloch

Z, Bauer AJ. Molecular testing for oncogenic gene alterations in pediatric thyroid lesions. Thyroid. 2018;28(1):60-67.

55. Wang TS, Dubner S, Sznyter LA, Heller KS. Incidence of metastatic well-differentiated thyroid cancer in cervical

lymph nodes. Arch Otolaryngol Head Neck Surg. 2004;130(1):110-113.

56. Farahati J, Bucsky P, Parlowsky T, Mader U, Reiners C. Characteristics of differentiated thyroid carcinoma in

children and adolescents with respect to age, gender, and histology. Cancer. 1997;80(11):2156-2162.

57. Durante C, Haddy N, Baudin E, Leboulleux S, Hartl D, Travagli JP, Caillou B, Ricard M, Lumbroso JD, De Vathaire F,

Schlumberger M. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid

carcinoma: benefits and limits of radioiodine therapy. J Clin Endocrinol Metab. 2006;91(8):2892-2899.

58. Medas F, Canu GL, Boi F, Lai ML, Erdas E, Calò PG. Predictive factors of recurrence in patients with differentiated

thyroid carcinoma: a retrospective analysis on 579 patients. Cancers (Basel). 2019;11(9):1230.

59. Nascimento C, Borget I, Ghuzlan AA, Deandreis D, Chami L, Travagli JP, Hartl D, Lumbroso J, Chougnet C, Lacroix

L, Baudin E, Schlumberger M, Leboulleux S. Persistent disease and recurrence in differentiated thyroid cancer

patients with undetectable postoperative stimulated thyroglobulin level. Endocrin Relat Cancer. 2011;18(2):R29.

60. Gatta G, Capocaccia R, Stiller C, Kaatsch P, Berrino F, Terenziani M, EUROCARE Working Group. Childhood cancer

survival trends in Europe: a EUROCARE working group study. J Clin Oncol. 2005;23(16):3742-3751.

61. Institute of Medicine (US) and National Research Council (US) National Cancer Policy Board, Hewitt M WS, Simone

JV. Childhood Cancer Survivorship: Improving Care and Quality of Life. Washington (DC): National Academies

Press (US); 2003. p. 49-89.

62. van Santen HM, Aronson DC, Vulsma T, Tummers RF, Geenen MM, de Vijlder JJ, van den Bos C. Frequent adverse

events after treatment for childhood-onset differentiated thyroid carcinoma: a single institute experience. Eur J

Cancer. 2004;40(11):1743-1751.

63. Elfenbein DM, Schneider DF, Chen H, Sippel RS. Surgical site infection after thyroidectomy: a rare but significant

complication. J Surg Res. 2014;190(1):170-176.

64. Albano D, Bertagna F, Panarotto MB, Giubbini R. Early and late adverse effects of radioiodine for pediatric

differentiated thyroid cancer. Pediatr Blood Cancer. 2017;64(11):e26595.

GENERAL INTRODUCTION

19

165. Sarkar SD, Beierwaltes WH, Gill SP, Cowley BJ. Subsequent fertility and birth histories of children and adolescents

treated with 131I for thyroid cancer. J Nucl Med. 1976;17(6):460-464.

66. Marti JL, Jain KS, Morris LGT. Increased risk of second primary malignancy in pediatric and young adult patients

treated with radioactive iodine for differentiated thyroid cancer. Thyroid. 2015;25(6):681-687.

67. Chen L, Shen Y, Luo Q, Yu Y, Lu H, Zhu R. Pulmonary fibrosis following radioiodine therapy of pulmonary metastases

from differentiated thyroid carcinoma. Thyroid. 2010;20(3):337-340.

68. Esfahani AF, Eftekhari M, Zenooz N, Saghari M. Gonadal function in patients with differentiated thyroid cancer

treated with (131)I. Hell J Nucl Med. 2004;7(1):52-55.

69. Bourcigaux N, Rubino C, Berthaud I, Toubert ME, Donadille B, Leenhardt L, Petrot-Keller I, Brailly-Tabard S,

Fromigue J, de Vathaire F, Simon T, Siffroi JP, Schlumberger M, Bouchard P, Christin-Maitre S. Impact on testicular

function of a single ablative activity of 3.7 GBq radioactive iodine for differentiated thyroid carcinoma. Hum

Reprod. 2018;33(8):1408-1416.

70. Leonova TA, Drozd VM, Saenko VA, Mine M, Biko J, Rogounovitch TI, Takamura N, Reiners C, Yamashita S. Bone

mineral density in treated at a young age for differentiated thyroid cancer after Chernobyl female patients on

TSH-suppressive therapy receiving or not Calcium-D3 supplementation. Endocr J. 2015;62(2):173-182.

71. Mendonca Monteiro de Barros G, Madeira M, Vieira Neto L, de Paula Paranhos Neto F, Carvalho Mendonca

LM, Correa Barbosa Lima I, Corbo R, Fleiuss Farias ML. Bone mineral density and bone microarchitecture after

long-term suppressive levothyroxine treatment of differentiated thyroid carcinoma in young adult patients. J

Bone Miner Metab. 2016;34(4):417-421.

72. Klein Hesselink EN, Klein Hesselink MS, de Bock GH, Gansevoort RT, Bakker SJ, Vredeveld EJ, van der Horst-Schrivers

AN, van der Horst IC, Kamphuisen PW, Plukker JT, Links TP, Lefrandt JD. Long-term cardiovascular mortality in

patients with differentiated thyroid carcinoma: an observational study. J Clin Oncol. 2013;31(32):4046-4053.

73. Parker WA, Edafe O, Balasubramanian SP. Long-term treatment-related morbidity in differentiated thyroid cancer:

a systematic review of the literature. Pragmatic and observational research. 2017;8:57-67.

74. Cooper DS. TSH suppressive therapy: an overview of long-term clinical consequences. Hormones (Athens). 2010;9(1):57-59.

75. Clement SC, Peeters RP, Ronckers CM, Links TP, van den Heuvel-Eibrink MM, Nieveen van Dijkum EJ, van

Rijn RR, van der Pal HJ, Neggers SJ, Kremer LC, van Eck-Smit BL, van Santen HM. Intermediate and long-term

adverse effects of radioiodine therapy for differentiated thyroid carcinoma-a systematic review. Cancer Treat

Rev. 2015;41(10):925-934.

76. Oren A, Benoit MA, Murphy A, Schulte F, Hamilton J. Quality of life and anxiety in adolescents with differentiated

thyroid cancer. J Clin Endocrinol Metab. 2012;97(10):1933.

77. Metallo M, Groza L, Brunaud L, Klein M, Weryha G, Feigerlova E. Long-term quality of life and pregnancy outcomes

of differentiated thyroid cancer survivors treated by total thyroidectomy and I(131) during adolescence and young

adulthood. Int J Endocrinol. 2016;2016:7586482.

78. de Oliveira Chachamovitz DS, dos Santos Vigario P, Nogueira Cordeiro MF, de Castro CL, Vaisman M, dos Santos

Teixeira Pde F. Quality of life, muscle strength, and fatigue perception in patients on suppressive therapy with

levothyroxine for differentiated thyroid carcinoma. Am J Clin Oncol. 2013;36(4):354-361.

79. Klein Hesselink MS, Bocca G, Hummel YM, Brouwers AH, Burgerhof JGM, van Dam EWCM, Gietema JA, Havekes

B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker

JTM, Ronckers CM, van Santen HM, van der Meer P, Links TP, Tissing WJE. Diastolic dysfunction is common in

survivors of pediatric differentiated thyroid carcinoma. Thyroid. 2017;27(12):1481-1489.

Chapter 2

Chapter 2

CHAPTER 2

Pediatric differentiated thyroid carcinoma in

the Netherlands: a nationwide follow-up study

Mariëlle S. Klein Hesselink, Marloes Nies, Gianni Bocca, Adrienne H. Brouwers, Johannes G.M. Burgerhof, Eveline W.C.M. van Dam, Bas Havekes, Marry M. van den Heuvel-Eibrink, Eleonora P.M. Corssmit, Leontien C.M. Kremer, Romana T. Netea-Maier, Heleen J.H. van der Pal, Robin P. Peeters, Kurt W. Schmid, Johannes W.A. Smit, Graham R. Williams, John T.M. Plukker, Cécile M. Ronckers, Hanneke M. van Santen, Wim J.E. Tissing, and Thera P. Links

J Clin Endocrinol Metab. 2016;101(5):2031-2039.

CHAPTER 2

22

2

ABSTRACTIntroductionTreatment for differentiated thyroid carcinoma (DTC) in pediatric patients is based mainly on evidence from adult series due to lack of data from pediatric cohorts. Our objective was to evaluate presentation, treatment-related complications, and long-term outcome in patients with pediatric DTC in the Netherlands.

Patients and methodsIn this nationwide study, presentation, complications and outcome of patients with pediatric DTC (age at diagnosis ≤18 years) treated in the Netherlands between 1970 and 2013 were assessed using medical records.

ResultsWe identified 170 patients. Overall survival was 99.4% after median follow-up of 13.5 years (range 0.3-44.7 years). Extensive follow-up data were available for 105 patients (83.8% women), treated in 39 hospitals. Median age at diagnosis was 15.6 years (range 5.8-18.9 years). At initial diagnosis, 43.8% of the patients had cervical lymph node metastases; 13.3% had distant metastases. All patients underwent total thyroidectomy. Radioactive iodine was administered to 97.1%, with a median cumulative activity of 5.66 GBq (range 0.74-35.15 GBq). Lifelong postoperative complications (permanent hypoparathyroidism and/or recurrent laryngeal nerve injury) were present in 32.4% of the patients. At last known follow-up, 8.6% of the patients had persistent disease and 7.6% experienced a recurrence. TSH suppression was not associated with recurrences (odds ratio 2.00, 95% CI 0.78 to 5.17, P = 0.152).

ConclusionsSurvival of pediatric DTC is excellent. Therefore, minimizing treatment-related morbidity takes major priority. Our study shows a frequent occurrence of lifelong postoperative complications. Adverse effects may be reduced by centralization of care, which is crucial for children with DTC.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

23

2

INTRODUCTIONDifferentiated thyroid carcinoma (DTC), which comprises papillary (PTC) and follicular thyroid carcinoma (FTC), is a rare disease during childhood. Age-standardized incidence rates for children 0-4 years of age are 0.4 per 100.000, and up to 1.5 per 100.000 for adolescents aged 15-19 years (1). However, DTC is the most common pediatric endocrine malignancy and its incidence is increasing (1-3). The prognosis in children has been reported to be excellent with 15-year survival rates greater than 95% (3). Due to sparse pediatric data, however, thyroid cancer care for pediatric patients is based predominantly on evidence from adult series. This year, the American Thyroid Association (ATA) published their first guidelines for children with DTC, thereby providing a thorough overview of the available literature (4).

The initial treatment for children with DTC generally consists of a (near) total thyroidectomy with or without lymph node dissection, although for patients with minimally invasive FTC 4 cm or less and lacking other adverse risk factors, a less aggressive treatment has recently been recommended (4, 5). Complication rates of thyroid surgery in children have been reported to be higher than those in adults (6). In most cases, surgery is followed by ablation therapy with radioactive iodine (131I) to destroy residual tumor foci and to facilitate disease monitoring by follow-up scans and measurement of serum thyroglobulin (Tg). However, nowadays 131I administration often depends on risk stratification (4, 5). Pediatric patients with residual tumor and/or metastases are generally treated by cyclic 131I administrations, with the activity of 131I being a matter of discussion (7). To decrease the risk of recurrent disease, TSH suppressive therapy with thyroid hormone has for decades been considered necessary during follow-up, but its use is currently tempered in patients showing no evidence of disease (4, 8-10).

Awareness regarding treatment-related morbidity is growing. However, long-term follow-up data, especially long-term data on morbidity, of children not exposed to the post-Chernobyl fallout are limited. Past studies in children 18 years old or younger at diagnosis have frequently been hampered by short follow-up, small patient series, series including patients with benign conditions, or treatment regimens not representative of current practice (e.g. including external beam radiotherapy) (11-15). Therefore, detailed insight into relevant clinical parameters of DTC in children is necessary to improve evidence-based treatment and follow-up strategies. The objective of this nationwide study was to evaluate the presentation, complications, and long-term outcome in patients with pediatric DTC in the Netherlands.

CHAPTER 2

24

2

PATIENTS AND METHODS Study design and populationIn this nationwide retrospective cohort study, children 18 years old or younger diagnosed with PTC or FTC between January 1970 and December 2013 and treated in the Netherlands were eligible for inclusion. The Dutch population is considered to be iodine-sufficient with virtually no exposure to the post-Chernobyl radioiodine fallout (16). To create a national cohort with coverage as high as reasonably feasible, patients were traced using data from the Netherlands Cancer Registry (1989-2013), and from hospital registries of the University Medical Centers (UMCs), in which patients were generally registered from 1970 onwards. Mortality data were obtained from electronic hospital patient information systems. For patients lost to follow-up, linkage with the database of the Central Bureau for Genealogy in the Netherlands was performed to identify deceased subjects. The Institutional Review Board of the University Medical Center Groningen approved the study. Informed consent was given by the patients and/or by their parents, for minors.

Data collectionMedical history, diagnosis, and treatment details were obtained from patients’ medical records. Histopathological data were obtained from the original pathology reports. Because the tumor node metastasis (TNM) classification of malignant tumors was changed several times within the period covered by this study, tumor stage was (re)classified according to the seventh edition of the TNM classification to facilitate comparison of the tumors (17). Data regarding 131I administrations (number and activities of 131I, and results of scans, both therapeutic and imaging) were collected from reports of the Departments of Nuclear Medicine. To calculate the cumulative administered 131I activity, only ablative and therapeutic 131I administrations were taken into account. TSH values were collected from the laboratory reports. In case of missing data, medical correspondence and the general practitioner were consulted.

Study definitionsDate of diagnosis was defined as the date of histological confirmation of thyroid carcinoma. Follow-up time was calculated from the date of diagnosis until the date of the patient’s last known assessment or the date of the patient’s death. Age at diagnosis was classified into 3 groups: age 0-10, 11-14 and 15-18 years. Transient and permanent hypoparathyroidism were defined by the use of calcium or vitamin D supplements for less than 6 months, and more than 6 months after thyroidectomy, respectively, or if these conditions were reported as such in the medical records. Recurrent laryngeal nerve (RLN) injury was defined as injury mentioned in the ear nose and throat report or, if this report was not available, in other medical records. Injury due to encasement by tumor was also defined as RLN injury. Remission was defined as the absence of clinical,

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

25

2

scintigraphical, or radiological evidence of disease and undetectable serum Tg under TSH suppressive therapy for at least 1 year after the last 131I therapy. Persistent disease was defined as the absence of remission. Recurrent disease was defined as histological, cytological, radiological, or biochemical evidence of disease after remission. Patients were classified according to risk of recurrence: low (T1-T2, N0, M0), intermediate (any T3 or N1 tumor), or high (any T4 or M1 tumor).

Statistical analysisGroups were compared using χ2 or Fisher’s exact tests (if conditions for χ2 test were not met) in the case of categorical variables. Mann-Whitney U and Kruskal-Wallis tests were performed for non-normally distributed continuous variables. Missing or unknown values were excluded from statistical testing. The TSH level for each year of follow-up was expressed as the geometric mean of the observed TSH values during that year. TSH values that were obtained before thyroidectomy until 12 weeks postoperatively were excluded, as well as TSH values obtained 6 weeks before until 12 weeks after thyroid hormone withdrawal or use of recombinant human TSH. The TSH level during the entire follow-up was defined as the geometric mean of the calculated TSH levels per year (18). Logistic regression analyses were performed to explore the associations between TSH level and recurrent disease, and between the occurrence of surgical complications and hospital volume, time of surgery, and age group. Regarding the association between TSH and recurrent disease, TSH was entered continuously in the crude model, followed by adjustment for risk classification. The associations between surgical complications and hospital volume, time of surgery, and age group were explored in a crude model, followed by adjustment for T stage (T1-T2 versus T3-T4) and the performance of lymph node dissection. Patients surgically treated in in more than one hospital were excluded from the analysis, as it was retrospectively unclear in which hospital the complication occurred. All tests were two sided. A P value of <0.05 was considered significant. IBM SPSS Statistics version 22 was used for statistical analyses.

RESULTSAs shown in the study flowchart (Figure 1), 170 patients with pediatric DTC were identified. One patient with familial adenomatous polyposis died at the age of 20 years due to complications of a colon carcinoma. He had been diagnosed 5 years earlier with PTC with lung metastases. Overall survival was 99.4% after a median follow-up of 13.5 years (range 0.3-44.7 years). Of the 169 survivors, 105 (62.1%) gave informed consent and were included in this study. The patients from whom no informed consent was obtained were more often male (29.7% vs. 16.2%, P = 0.038), and more often had distant metastases (P = 0.031) and a longer follow-up time (median 18.1 years (range 0.3-40.4 years) vs. 11.7 years (range 1.1-44.7 years), P = 0.043, Supplemental Table 1).

CHAPTER 2

26

2

Baseline characteristics Baseline characteristics are provided in Table 1. The female: male ratio was 5.2:1. Median age at diagnosis was 15.6 years (range 5.8-18.9 years). PTC was diagnosed in 81.0% of the patients; the remaining 19.0% had FTC. At initial diagnosis, histologically confirmed cervical lymph node metastases were found in 46 (43.8%) patients and distant metastases in 14 (13.3%) patients. Of these, 11 patients had lung metastases, one patient with FTC had a metastasis in the seventh thoracic vertebra and two patients with PTC and FTC, respectively, had both lung and bone metastases. Pathological features and TNM stage did not differ between the three age groups.

Medical historyFour (3.8%) patients developed DTC as a second malignant neoplasm (SMN); two of whom had been treated with cranial radiotherapy (Supplemental Table 2). One patient with a neuroblastoma had been treated with 131I-metaiodobenzylguanidine (previously reported (19)). The fourth patient had been treated for Langerhans cell histiocytosis. She did not receive radiotherapy. Two (1.9%) patients developed PTC after radiotherapy directed to the neck for benign conditions.

Surgical treatmentTotal thyroidectomy was performed in all patients. In 65 (61.9%) patients the total thyroidectomy was performed as a single procedure. In the remaining 40 (38.1%) patients a diagnostic hemithyroidectomy was performed, followed by a completion

Figure 1. Study flow chart, showing final number of included patients and reasons for nonparticipation

of eligible patients.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

27

2

thyroidectomy. The mean time span between both procedures was 31.5 days (range 2-210 days).

Lymph node dissection was performed as part of initial therapy in 46 (43.8%) patients, of whom 40 (87.0%) had histologically proven lymph node metastases. In 10 (9.5%) patients the central compartment (level VI) was dissected; in 36 (34.4%) patients a lateral lymph node dissection was performed, including other levels (II-V) on one or both sides of the neck (Table 1). In six patients not initially treated with a lymph node dissection, positive lymph nodes were found during histopathological examination. Central compartment dissection alone was performed more frequently in children aged 0-10 years, while in older children lateral levels were more often included in the lymph node dissection (P = 0.045, Table 1).

Throughout the entire study period, patients were surgically treated in 39 hospitals, including nine UMCs and 30 general hospitals. During this period, in our cohort, the median number of surgical procedures (hemi- or total thyroidectomy, or lymph node dissection, when performed at different dates) per hospital was two (range 1-30). Over the past decade, 50 patients were treated in 16 different hospitals, including nine UMCs and seven general hospitals.

Table 1. Baseline characteristics of patients with pediatric differentiated thyroid carcinoma

Characteristic All patients

(n = 105)

0-10 years

(n = 10)

11-14 years

(n = 34)

15-18 years

(n = 61)

P Valuea

Sex, n (%) 0.006

Male 17 (16.2) 5 (50.0) 6 (17.6) 6 (9.8)

Female 88 (83.8) 5 (50.0) 28 (82.4) 55 (90.2)

Age at diagnosis, years 15.6 (5.8-18.9) 9.5 (5.8-10.8) 13.0 (11.1-14.8) 17.1 (15.3-18.9) n.a.

Histology, n (%) 0.363

Papillary 85 (81.0) 9 (90.0) 25 (73.5) 51 (83.6)

Follicular 20 (19.0) 1 (10.0) 9 (26.5) 10 (16.4)

Primary tumor size, cm 2.5 (0.3-9.0) 1.4 (0.8-5.0) 2.9 (1.0-5.5) 2.5 (0.3-9.0)

Localization, n (%) 0.193

Unilateral 67 (63.8) 5 (50.0) 23 (67.7) 39 (63.9)

Bilateral 28 (26.7) 3 (30.0) 9 (26.5) 16 (26.2)

Otherb 0.493

Isthmus 3 (2.9) 1 (10.0) 0 (0.0) 2 (3.3)

Thyroglossal duct 1 (1.0) 0 (0.0) 0 (0.0) 1(1.6)

Unknown 6 (5.7) 1 (10.0) 2 (5.9) 3 (4.9)

Multifocality, n (%) 0.632

No 50 (47.6) 4 (40.0) 18 (52.9) 28 (45.9)

Yes 29 (27.6) 3 (30.0) 7 (20.6) 19 (31.1)

Unknown 26 (24.8) 3 (30.0) 9 (26.5) 14 (23.0)

Continues on next page

CHAPTER 2

28

2

Surgical complications As shown in Table 2, post-operative transient and permanent hypoparathyroidism were observed in 16 (15.2%) and 25 (23.8%) patients, respectively. Both transient and permanent hypoparathyroidism occurred more often in patients who underwent a lymph node dissection. Unilateral RLN injury occurred in 12 (11.4%) patients. Bilateral RLN injury occurred only in a 15-year-old patient with extended disease who was treated with a total thyroidectomy, a central compartment dissection and a bilateral modified lymph node dissection. The right RLN was encased by the tumor and was removed as part of the surgical procedure. RLN injury occurred more often in patients with tumors staged T3-T4 compared to stage T1-T2 (P <0.001), and in patients with lymph node involvement (P <0.001). The frequency of surgical complications did not differ between initial surgery performed before or during the last decade, either

Table 1. (continued)

Characteristic All patients

(n = 105)

0-10 years

(n = 10)

11-14 years

(n = 34)

15-18 years

(n = 61)

P Valuea

TNM stage, n (%)T 0.960

T1-T2 65 (61.9) 6 (60.0) 21(61.8) 38 (62.3)

T3-T4 26 (24.8) 2 (20.0) 9 (26.5) 15 (24.6)

Txc 14 (13.3) 2 (20.0) 4 (11.8) 8 (13.1)

N 0.339

N0 53 (50.5) 4 (40.0) 15 (44.1) 34 (55.7)

N1a-N1b 46 (43.8) 6 (60.0) 17 (50.0) 23 (37.7)

Nxc 6 (5.7) 0 (0.0) 2 (5.9) 4 (6.6)

M 0.752

M0 82 (78.1) 8 (80.0) 25 (73.5) 49 (80.3)

M1b 14 (13.3) 1 (10.0) 6 (17.6) 7 (11.5)

Lung 11 1 5 5

Bone 1 0 0 1

Lung and bone 2 0 1 1

Mxc 9 (8.6) 1 (10.0) 3 (8.8) 5 (8.2)

Surgery, n (%)Total thyroidectomy 105 (100) 10 (100) 34 (100) 61 (100) n.a.

Lymph node dissection 0.045

None 49 (46.7) 4 (40.0) 15 (44.1) 30(49.2)

Central LND 10 (9.5) 4 (40.0) 1 (2.9) 5 (8.2)

LND incl. lateral levels 36 (34.3) 2 (20.0) 15 (44.1) 19 (31.1)

Unknown 10 (9.5) 0 (0.0) 3 (8.8) 7 (11.5)

Numbers are expressed as median (range). Abbreviations: LND, lymph node dissection; n.a., not applicable. a Differences tested between the three age groups. Missing or unknown values excluded from statistical testing. b

Summarized as 1 variable for statistical testing. c ‘x’ indicates that there has been no assessment of that tumor

characteristic, or information about that characteristic was not available.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

29

2

Tab

le 2

. Sur

gica

l co

mpl

icat

ions

in p

atie

nts

wit

h p

edia

tric

diff

eren

tiat

ed th

yro

id c

arci

nom

a

Gro

upH

ypo

par

ath

yro

idis

m, n

(%

)R

ecur

ren

t la

ryn

gea

l ner

ve in

jury

, n (

%)

No

ne

Tran

sien

tPe

rman

ent

P V

alue

Un

kno

wn

No

ne

Left

Rig

ht

Bila

tera

lP

Val

ueU

nkn

ow

n

All

pat

ien

ts (

n =

10

5)51

(48.

6)16

(15

.2)

25 (

23.8

)13

(12

.4)

69 (

65.7

)5

(4.8

)7

(6.7

)1 (

1.0

)23

(21

.9)

T1-

T2

(n =

65)

36 (

55.4

)8

(12.

3)14

(21

.5)

0.14

37

(10

.8)

52 (

80.0

)1 (

1.5)

0 (

0.0

)0

(0

.0)

<0.0

01

12 (

18.5

)

T3-

T4

(n

= 2

6)10

(38

.5)

7 (2

6.9)

8 (3

0.8

)1 (

3.8)

14 (

53.8

)2

(7.7

)4

(15.

4)1 (

3.8)

5 (1

9.2)

Tx (

n =

14)

5 (3

5.7)

1 (7.

1)3

(21.4

)5

(35.

7)3

(21.4

)2

(14.

3)3

(21.4

)0

(0

.0)

6 (4

2.9)

No

LN

D (

n =

49)

32 (

65.3

)6

(12.

2)5

(10

.2)

0.0

01

6 (1

2.2)

39 (

79.6

)0

(0

.0)

0 (

0.0

)0

(0

.0)

<0.0

01

10 (

20.4

)

LND

(n

= 4

6)15

(32

.6)

10 (

21.7

)16

(34

.8)

5 (1

0.9

)28

(60

.9)

4 (8

.7)

6 (1

3.0

)1 (

2.2)

7 (1

5.2)

LND

un

kno

wn

(n

= 1

0)4

(40

.0)

0 (

0.0

)4

(40

.0)

2 (2

0.0

)2

(20

.0)

1 (10

.0)

1 (10

.0)

0 (

0.0

)6

(60

.0)

Abb

revi

atio

n: L

ND

, lym

ph n

od

e d

isse

ctio

n. D

iffer

ence

s te

sted

bet

wee

n T1

-T2

and

T3-

T4 a

nd n

o L

ND

and

LN

D M

issi

ng o

r un

kno

wn

valu

es e

xclu

ded

fro

m s

tati

stic

al te

stin

g.

CHAPTER 2

30

2

in a crude model or after adjustment for T stage and performance of lymph node dissection (odds ratio (OR) 0.96, 95% confidential interval (CI) 0.42 to 2.17, P = 0.912 and OR 0.94, 95% CI 0.32 to 2.72, P = 0.904, respectively). Finally, age group was not related to the occurrence of surgical complications, either in a crude or adjusted model (P = 0.550 and P = 0.189, respectively).

131I administrations and TSH suppressive therapyData regarding 131I administrations are provided in Table 3. One hundred and two (97.1%) patients were treated with 131I, with a median cumulative activity administered during initial treatment and follow-up of 5.66 GBq (range 0.74-35.15 GBq). The median number of 131I administrations was 1 (range 1-6). Ninety-four (89.5%) patients underwent 131I ablation therapy within 6 months after initial surgical treatment. By doctor’s choice, three patients did not receive 131I ablation therapy. Higher tumor stage (T3-T4), lymph node involvement, and distant metastases were independently associated with a higher administered cumulative 131I activity (P <0.001) and with an increase in the number of 131I administrations (P <0.001). The cumulative 131I activity and the number of 131I administrations during initial treatment and follow-up did not differ between age groups at diagnosis (P = 0.227 and P = 0.225, respectively, data not shown). Pulmonary fibrosis was not encountered in the medical charts. No patients were treated with external beam radiotherapy, chemotherapy or tyrosine kinase inhibitors for DTC. TSH values of 104 (99.0%) patients were available for analysis. Median TSH level during follow-up was 0.17 mU/L (range 0.01-4.74 mU/L). TSH was suppressed below 0.10 mU/L during the entire follow-up in 36 (34.6%) patients. The TSH level was not associated

Table 3. 131I administrations in patients with pediatric differentiated thyroid carcinoma

Group Cumulative 131I activity

(GBq)P Valuea 131I administrations, n P Valuea

All patients (n = 100)b 5.66 (0.74-35.15) 1 (1-6)

T1-T2 (n = 62) 5.55 (0.74-31.49) <0.001 1 (1-6) <0.001

T3-T4 (n = 26) 12.36 (1.78-35.15) 3 (1-6)

Tx (n = 12) 4.63 (1.22-27.85) 1 (1-5)

N0 (n = 49) 3.70 (1.00-21.46) <0.001 1 (1-6) <0.001

N1a-N1b (n = 45) 11.10 (0.74-35.15) 2 (1-6)

Nx (n = 6) 6.75 (1.85-8.33) 1 (1-2)

M0 (n = 77) 5.55 (0.74-35.15) <0.001 1 (1-6) <0.001

M1 (n = 14) 14.43 (6.11-29.79) 3 (1-5)

Mx (n = 9) 5.55 (1.85-27.85) 1 (1-5)

All data expressed as median (range). Abbreviation: 131I, radioactive iodine. a Differences tested between T1-T2 and

T3-T4, N0 and N1a-N1b, M0 and M1. Tx, Nx, Mx excluded from statistical testing. b Administered 131I activity was

unknown in two patients; three patients did not receive 131I treatment. Therefore n = 100 instead of n = 105.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

31

2

with recurrent disease, either in a crude model (OR 1.98, 95% CI 0.80 to 4.86, P = 0.140), or after adjustment for risk classification (OR 2.00, 95% CI 0.78 to 5.17, P = 0.152).

OutcomeAt last known follow-up, nine (8.6%) patients had persistent disease, six of whom were classified as such based on a detectable Tg level. Eight (7.6%) patients experienced a recurrence (Table 4). Recurrence free survival (RFS) from initial treatment to first recurrence ranged from 3.9 to 22.7 years. Three of eight patients relapsed within 5 years after initial treatment. Of the 11 (10.5%) patients who had not been treated with 131I within 6 months after total thyroidectomy, two developed a recurrence (P = 0.197, data not shown). As shown in Table 5, T3-T4 stage, lymph node involvement, and distant metastases stage were associated with persistent disease (P = 0.040, P = 0.010, and P = 0.020, respectively). No associations were found between initial TNM stage and recurrences. The three patients who initially presented with bone metastases became free of disease after administration of 13.7, 11.8, and 14.8 GBq 131I, and remained in remission after a follow-up of 2.9, 5.1, and 13.2 years, respectively. Outcome did not differ between patients with PTC and FTC, or between the three age groups (P = 0.411 and P = 0.789, respectively, data not shown). Outcome could not be assessed for two patients because of follow-up less than 1 year. For another patient, information to evaluate outcome was not available.

Median follow-up in patients with recurrent disease was 24.7 years (range 10.7-41.5 years), significantly longer than in patients who remained in remission (11.4 years, range 1.3-44.7 years) and in patients with persistent disease (5.5 years, range 1.7-36.1 years) (P = 0.030, data not shown).

Data regarding SMNs after pediatric DTC are provided in Supplemental Table 3.

DISCUSSIONThis nationwide study of pediatric patients with well-differentiated thyroid cancer in the Netherlands confirms an excellent overall survival. All patients underwent total thyroidectomy with nodal dissection in 43.8%, followed in the majority of patients by high-dose 131I ablation therapy. Over a 43-year period, 105 patients were surgically treated in 39 hospitals. In 32.4% of them life-long postoperative complications (permanent hypoparathyroidism and/or RLN injury) were present. A significant number of patients had persistent disease or experienced a recurrence. Despite small patient numbers, our cohort is one of the largest to be described for this rare disease in children. It gives insight into relevant clinical parameters that can be used to improve evidence-based treatment and follow-up strategies.

In our opinion, the incidence of hypoparathyroidism, using strict definitions in our cohort, seems to be relatively high compared to comparable cohorts of pediatric patients treated for DTC (transient and permanent hypoparathyroidism in 15.2% and

CHAPTER 2

32

2

Tab

le 4

. Pat

ient

s w

ith

recu

rren

t and

per

sist

ent d

isea

se in

pat

ient

s w

ith

ped

iatr

ic d

iffer

enti

ated

thyr

oid

car

cino

ma

Sex

and

ag

e at

d

iag

no

sis,

yea

rsFo

llow

-up

, ye

ars

His

tolo

gy,

TN

Ma

Init

ial

trea

tmen

tb

Evid

ence

of

dis

ease

Loca

lizat

ion

of r

ecur

ren

ceR

FSc ,

year

sR

emis

sio

n af

ter

recu

rren

ce

2nd

re

curr

ence

Rem

issi

on

afte

r 2n

d

recu

rren

ce

Recu

rren

t di

seas

e (n

= 8

)

F,18

.933

.3PT

C,

T2N

0M

0T

T +

131 I

Scin

tigr

aphi

cal

Thyr

oid

bed

3.92

yes

non.

a.

F,18

.310

.7PT

C,

T1bN

0M

0T

T +

131 I

Scin

tigr

aphi

cal

Cer

vica

l LN

4.16

yes

non.

a.

F,17

.117

.5PT

C,

T3N

1bM

0T

T +

131 I

Cyt

olo

gica

lC

ervi

cal L

N4.

81ye

sye

sye

s

F,13

.341

.5FT

C,

T4aN

1bM

0T

T +

131 Id

Scin

tigr

aphi

cal

Cer

vica

l LN

6.15

yes

yes

no

F,16

.312

.0PT

C,

T2N

0M

0T

T +

131 I

Scin

tigr

aphi

cal

Cer

vica

l LN

8.96

yes

non.

a.

F,13

.132

.4PT

C,

T1bN

1bM

0T

T +

LND

+ 13

1 IBi

och

emic

ale

No

t sp

ecifi

ed11

.04

yes

non.

a.

F,13

.125

.2PT

C,

T1bN

1bM

0T

T +

LND

His

tolo

gica

lC

ervi

cal L

N16

.62

yes

non.

a.

F,18

.824

.3PT

C,

T1bN

0M

xT

T H

isto

logi

cal +

rad

iolo

gica

lf

Cer

vica

l LN

,

lung

22.6

5no

n.a.

n.a.

Pers

iste

nt d

isea

se (

n =

9)

F,12

.523

.3PT

C,

T2N

1M0

TT

+ LN

D +

131 I

Rad

iolo

gica

l Lu

ng

M,

10.5

8.4

PTC

,T4

aN1b

M1

TT

+ LN

D +

131 I

Rad

iolo

gica

lLu

ng

F,18

.51.

7PT

C,

T4bN

1bM

1T

T +

LND

+ 13

1 ISc

inti

grap

hica

l

+ ra

dio

logi

calg

Cer

vica

l LN

,

lung

F,12

.14.

9PT

C,

T2N

1bM

0T

T +

LND

+ 13

1 IBi

och

emic

al

No

t sp

ecifi

ed

F,16

.15.

5PT

C,

T1N

1bM

0T

T +

LND

+ 13

1 IBi

och

emic

al

No

t sp

ecifi

ed

M,

12.7

14.8

PTC

,T3

N1b

M0

TT

+ LN

D +

131 I

Bio

chem

ical

N

ot

spec

ified

M,

14.7

4.4

PTC

,T3

N0

M1

TT

+ 13

1 IBi

och

emic

al

No

t sp

ecifi

ed

F,15

.92.

0PT

C,

T4aN

1bM

1T

T +

LND

+ 13

1 IBi

och

emic

al

No

t sp

ecifi

ed

F,16

.736

.1PT

C,

TxN

1bM

xT

T +

LND

+ 13

1 IBi

och

emic

al

No

t sp

ecifi

ed

Abb

revi

atio

ns: F

, fem

ale;

M, m

ale;

PTC

, pap

illar

y th

yro

id c

arci

nom

a; F

TC, f

olli

cula

r th

yro

id c

arci

nom

a; T

T, t

ota

l thy

roid

ecto

my;

LN

D, l

ymph

no

de

dis

sect

ion;

131 I,

rad

ioac

tive

iod

ine

adm

inis

trat

ion;

LN

, lym

ph n

od

e; R

FS, r

ecur

renc

e-fr

ee s

urvi

val;

and

n.a

., no

t ap

plic

able

. a

Init

ial T

NM

cla

ssifi

cati

on,

b 13

1 I is

men

tio

ned

as

part

of

init

ial t

hera

py if

per

form

ed le

ss t

han

6 m

ont

hs a

fter

sur

gica

l tre

atm

ent,

c RF

S ca

lcul

ated

fro

m d

ate

of

dia

gno

sis

unti

l firs

t

recu

rren

ce, d

Unk

now

n if

LND

was

per

form

ed; e

Pati

ent t

reat

ed w

ith

131 I b

ecau

se o

f inc

reas

ing

thyr

ogl

obu

lin le

vel;

unkn

ow

n if

po

st th

erap

y sc

an w

as p

erfo

rmed

; f Cer

vica

l lym

ph n

od

es

pro

ved

his

tolo

gica

lly. L

ung

met

asta

ses

wer

e vi

sibl

e o

n co

mpu

ted

tom

ogr

aphy

(CT)

; g C

ervi

cal l

ymph

no

des

det

ecte

d s

cint

igra

phic

ally

. Lun

g m

etas

tase

s w

ere

visi

ble

on

CT.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

33

2

23.8%, respectively, previously described in 7.4-32.7% and 0-32%, respectively) (12,

20-23). In adults, these complications were less frequently encountered (6.2-14.2%

and 0-4.0%, respectively) (24, 25). The occurrence of RLN injury in our cohort (12.4%)

was comparable with other pediatric cohorts (0-40%), as well as with reported adult

data (0-38.4%) (14, 21, 26, 27). However, it should be considered that definitions of

surgical complications are heterogeneous. Complication rates therefore vary widely

in the literature. Total thyroidectomy, which is recommended for the vast majority of

pediatric patients with DTC, is associated with a higher complication risk than partial

thyroidectomy, particularly when combined with lymph node dissection (4-6, 13).

Another major factor that could have contributed to our complication rates might

be the high number of centers where surgery was performed on this low-volume

high-risk patient group. Due to the low numbers of surgical treatments per hospital

in our cohort we were not able to analyze differences in the occurrence of surgical

complications between high- and low-volume centers. However, it has been shown

that complications of thyroid surgery in pediatric patients are reduced when surgery is

performed by high-volume surgeons (6). High-volume surgery may also be associated

with fewer incomplete resections. Therefore the need for further centralization of care

for pediatric patients with DTC is essential, as has been recommended by the ATA (4).

The cumulative therapeutic 131I activity during initial treatment and follow-up in our

cohort was relatively high. Given the good survival rate, it can be questioned whether

children could just as well be treated with lower therapeutic activities, as suggested by

recent guidelines (4, 5). The dosage of 131I is of importance, given that pulmonary fibrosis

Table 5. Outcome of patients with pediatric differentiated thyroid carcinoma

Group Remission,

n (%)

Recurrence,

n (%)

P Valuea,b Persistent

disease, n (%)

P Valuea,c Unknown,

n (%)d

All patients (n = 105) 85 (81.0) 8 (7.6) 9 (8.6) 3 (2.9)

T1-T2 (n = 65) 54 (83.1) 6 (9.2) 1.000 3 (4.6) 0.040 2 (3.1)

T3-T4 (n = 26) 18 (69.2) 2 (7.7) 5 (19.2) 1 (3.8)

Tx (n = 14) 13 (92.9) 0 (0.0) 1 (7.1) 0 (0.0)

N0 (n = 53) 47 (88.7) 4 (7.5) 0.713 1 (1.9) 0.010 1 (1.9)

N1a-N1b (n = 46) 32 (69.6) 4 (8.7) 8 (17.4) 2 (4.3)

Nx (n = 6) 6 (100.0) 0 (0.0) 0 (0.0) 0 (0.0)

M0 (n = 82) 70 (85.4) 7 (8.5) 1.000 4 (4.9) 0.020 1 (1.2)

M1 (n = 14) 10 (71.4) 0 (0.0) 4 (28.6) 0 (0.0)

Mx (n = 9) 5 (55.6) 1 (11.1) 1 (11.1) 2 (22.2)

a Differences tested between T1-T2 and T3-T4, N0 and N1a-N1b, M0 and M1. Tx, Nx, Mx and unknown outcome

excluded from statistical testing. b Patients in remission and with recurrent disease compared. c Patients in remission

and with persistent disease compared. d Outcome unknown in one patient and could not be assessed in two patients

due to follow-up less than 1 year (all summarized as unknown outcome).

CHAPTER 2

34

2

was observed as a side effect in 7.2% of patients with lung metastases in a high-risk Chernobyl-related pediatric cohort (28). In our cohort we did not observe pulmonary fibrosis after a similar follow-up period. As the presence of pulmonary fibrosis was assessed from medical records in our study, we may have missed subclinical cases. Nevertheless, it is our opinion that the administration of 131I should be considered very carefully in pediatric patients to prevent possible early and late adverse effects (29, 30). This is especially the case in children with low-risk DTC as no benefit of 131I ablation therapy has been shown in adults with low-risk disease (31). High 131I activities should be reserved for children with metastatic disease, as advocated earlier by Verburg et al. (7).

In about one-third of the patients in our cohort TSH was suppressed during the entire follow-up. To the best of our knowledge, our study is the first to report that TSH level is not associated with recurrent disease in children with DTC. This finding upholds the first ATA guidelines for pediatric patients with DTC, which recommend tempering TSH suppression in children showing no evidence of disease (4). However, this should be interpreted carefully as our study probably has a lack of power.

The prevalence of cervical lymph node metastases at initial diagnosis in our cohort (43.8%) is in the lower range of prevalences as reported in other pediatric series (39-90%) (15, 21, 22, 32-34). The prevalence of distant metastases in our study is comparable to that from other pediatric cohorts (32, 33).

Persistent disease was more often found in patients with higher T stage, cervical lymph node involvement and distant metastases at diagnosis. One-third of the patients with persistent disease had lung metastases at last known follow-up. The other two-thirds were classified as having persistent disease based on a detectable Tg level. It must be considered that Tg levels are not always analyzed in the literature, and that patients with detectable Tg levels are not always classified as having persistent disease in other studies.

We report a recurrence rate of 7.6%, which is low compared with a study involving comparable treatment and follow-up (18.8%) (33). Our study definitions of recurrent disease may have contributed to the low recurrence rate, as we did not interpret disease activity within 1 year after initial treatment as recurrent disease. In contrast to some other studies, in our cohort recurrences were not associated with initial TNM stage and did not differ between age groups (14, 33, 35). RFS ranged from 3.9 to 22.7 years. This favors lifelong follow-up of children. Patients with recurrent disease were significantly longer in follow-up compared to patients in remission and with persistent disease. This prolonged follow-up might be explained by the wish of physicians to follow patients with recurrent disease for a longer time than patients who remain in remission.

Possible differences in presentation of sporadic DTC compared to radiation-induced DTC could not be detected, as the number of patients with SMN in our cohort was low and the median follow-up was relatively short to develop a SMN.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

35

2

Limitations of this study are related to its retrospective design. As patients were treated in many hospitals over more than 40 years, complete information from medical records could not always be retrieved. Furthermore, preoperative staging and patient management were not similar in all hospitals and have evolved over time. In addition, the association between lymph node dissection and occurrence of surgical complications should be interpreted carefully as the number of procedures per hospital was small and information on the selection criteria for the extent of surgery was often lacking. However, over the last decades the general consensus on treatment for pediatric DTC has in the Netherlands remained roughly unchanged. Finally, various TSH assays have been used. However, we expect limited influence of the assay differences on the results, as the lower limits of the reference ranges remained more or less stable.

In conclusion, the life expectancy for children with DTC is excellent. However, many patients experience adverse effects from thyroid surgery, resulting in lifelong complications in 32.4%. Centralization of care for pediatric patients with DTC is crucial to reduce treatment-related damage in this young patient group. In the near future, treatment for pediatric patients with DTC will be further centralized in one or two hospitals in the Netherlands. Furthermore, the administration of 131I should be weighed very carefully to prevent early and late adverse events. Further studies are needed to evaluate long-term sequels. International collaboration using homogeneous definitions can accelerate evaluation and improvement of treatment for children with DTC.

CHAPTER 2

36

2

ACKNOWLEDGEMENTS We are grateful to our colleagues in the Netherlands for referring patients to this study. The authors thank the registration teams of the Comprehensive Cancer Centers for the collection of data for the Netherlands Cancer Registry and the scientific staff of the Netherlands Cancer Registry. Assistance provided by the Central Bureau for Genealogy in the Netherlands in identifying deceased persons was greatly appreciated.

FUNDINGThis work was supported by Stichting Kinderen Kankervrij (The Netherlands, Foundation Children Cancer-Free, Project 81). C.M.R. is supported by the Dutch Cancer Society.

CLINICAL TRIAL REGISTRYThis study had the clinical trial registration number of the Netherlands Trial Registry 3448/NL3280.

DISCLOSURE SUMMARYThe authors have nothing to disclose.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

37

2

REFERENCES1. Vergamini LB, Frazier AL, Abrantes FL, Ribeiro KB, Rodriguez-Galindo C. Increase in the incidence of differentiated

thyroid carcinoma in children, adolescents, and young adults: a population-based study. J Pediatr. 2014;164(6):1481-1485.

2. Siegel DA, King J, Tai E, Buchanan N, Ajani UA, Li J. Cancer incidence rates and trends among children and

adolescents in the United States, 2001-2009. Pediatrics. 2014;134(4):e945-e955.

3. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167-172.

4. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID, Luster

M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S. Management guidelines for children with thyroid nodules

and differentiated thyroid cancer. Thyroid. 2015;25(7):716-759.

5. Perros P, Colley S, Boelaert K, Evans C, Evans RM, Gerrard GE, Gilbert JA, Harrison B, Johnson SJ, Giles TE, Moss

L, Lewington V, Newbold KL, Taylor J, Thakker RV, Watkinson J, Williams GR. Guidelines for the management of

thyroid cancer. Clin Endocrinol (Oxf). 2014;81:1-122.

6. Sosa JA, Tuggle CT, Wang TS, Thomas DC, Boudourakis L, Rivkees S, Roman SA. Clinical and economic outcomes

of thyroid and parathyroid surgery in children. J Clin Endocrinol Metab. 2008;93(8):3058-3065.

7. Verburg FA, Reiners C, Hänscheid H. Approach to the patient: role of dosimetric RAI rx in children with DTC. J Clin

Endocrinol Metab. 2013;98(10):3912-3919.

8. Pujol P, Daures JP, Nsakala N, Baldet L, Bringer J, Jaffiol C. Degree of thyrotropin suppression as a prognostic

determinant in differentiated thyroid cancer. J Clin Endocrinol Metab. 1996;81(12):4318-4323.

9. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM,

Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L. 2015 American Thyroid

Association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer.

Thyroid. 2016;26(1):1-133.

10. Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW, Wiersinga W, European Thyroid Cancer Taskforce. European

consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur

J Endocrinol. 2006;154(6):787-803.

11. Vassilopoulou-Sellin R, Goepfert H, Raney B, Schultz PN. Differentiated thyroid cancer in children and adolescents:

clinical outcome and mortality after long-term follow-up. Head Neck. 1998;20(6):549-555.

12. van Santen HM, Aronson DC, Vulsma T, Tummers RF, Geenen MM, de Vijlder JJ, van den Bos C. Frequent adverse

events after treatment for childhood-onset differentiated thyroid carcinoma: a single institute experience. Eur J

Cancer. 2004;40(11):1743-1751.

13. Scholz S, Smith JR, Chaignaud B, Shamberger RC, Huang SA. Thyroid surgery at Children’s Hospital Boston:

a 35-year single-institution experience. J Pediatr Surg. 2011;46(3):437-442.

14. Enomoto Y, Enomoto K, Uchino S, Shibuya H, Watanabe S, Noguchi S. Clinical features, treatment, and

long-term outcome of papillary thyroid cancer in children and adolescents without radiation exposure. World J

Surg. 2012;36(6):1241-1246.

15. Fridman MV, Savva NN, Krasko OV, Zborovskaya AA, Mankovskaya SV, Schmid KW, Demidchik YE. Clinical and

pathologic features of “sporadic” papillary thyroid carcinoma registered in the years 2005 to 2008 in children and

adolescents of Belarus. Thyroid. 2012;22(10):1016-1024.

16. United Nations Scientific Committee on the Effects of Atomic Radiation. Sources and effects of ionizing radiation.

UNSCEAR 2008 report to the general assembly with scientific annexes, Volume II: Annex D: Health effects due to

radiation from the Chernobyl accident. New York: United Nations; 2011.

17. Sobin LH, Gospodarowicz MK, Wittekind C, eds. TNM Classification of Malignant Tumours. 7th ed. Oxford, UK:

Wiley-Blackwell; 2009.

18. Klein Hesselink EN, Klein Hesselink MS, de Bock GH, Gansevoort RT, Bakker SJ, Vredeveld EJ, van der Horst-Schrivers

AN, van der Horst IC, Kamphuisen PW, Plukker JT, Links TP, Lefrandt JD. Long-term cardiovascular mortality in

patients with differentiated thyroid carcinoma: an observational study. J Clin Oncol. 2013;31(32):4046-4053.

19. van Santen HM, Tytgat GA, van de Wetering MD, van Eck-Smit BL, Hopman SM, van der Steeg AF, Nieveen van

Dijkum EJ, van Trotsenburg AS. Differentiated thyroid carcinoma after 131I-MIBG treatment for neuroblastoma

during childhood: description of the first two cases. Thyroid. 2012;22(6):643-646.

CHAPTER 2

38

2

20. Haveman JW, van Tol KM, Rouwé CW, Piers DA, Plukker JT. Surgical experience in children with differentiated

thyroid carcinoma. Ann Surg Oncol. 2003;10(1):15-20.

21. Thompson GB, Hay ID. Current strategies for surgical management and adjuvant treatment of childhood papillary

thyroid carcinoma. World J Surg. 2004;28(12):1187-1198.

22. O’Gorman CS, Hamilton J, Rachmiel M, Gupta A, Ngan BY, Daneman D. Thyroid cancer in childhood: a retrospective

review of childhood course. Thyroid. 2010;20(4):375-380.

23. Kundel A, Thompson GB, Richards ML, Qiu LX, Cai Y, Schwenk FW, Lteif AN, Pittock ST, Kumar S, Tebben PJ,

Hay ID, Grant CS. Pediatric endocrine surgery: a 20-year experience at the Mayo Clinic. J Clin Endocrinol

Metab. 2014;99(2):399-406.

24. Hundahl SA, Cady B, Cunningham MP, Mazzaferri E, McKee RF, Rosai J, Shah JP, Fremgen AM, Stewart AK, Hölzer

S. Initial results from a prospective cohort study of 5583 cases of thyroid carcinoma treated in the United States

during 1996. U.S. and German Thyroid Cancer Study Group. An American College of Surgeons Commission on

Cancer Patient Care Evaluation Study. Cancer. 2000;89(1):202-217.

25. Kim SM, Kim HK, Kim KJ, Chang HJ, Kim BW, Lee YS, Chang YS, Park CS. Recovery from permanent

hypoparathyroidism after total thyroidectomy. Thyroid. 2015;25(7):830-833.

26. Jeannon JP, Orabi AA, Bruch GA, Abdalsalam HA, Simo R. Diagnosis of recurrent laryngeal nerve palsy after

thyroidectomy: a systematic review. Int J Clin Pract. 2009;63(4):624-629.

27. Bergenfelz A, Jansson S, Kristoffersson A, Mårtensson H, Reihnér E, Wallin G, Lausen I. Complications to thyroid

surgery: results as reported in a database from a multicenter audit comprising 3,660 patients. Langenbecks Arch

Surg. 2008;393(5):667-673.

28. Reiners C, Biko J, Haenscheid H, Hebestreit H, Kirinjuk S, Baranowski O, Marlowe RJ, Demidchik E, Drozd V,

Demidchik Y. Twenty-five years after Chernobyl: outcome of radioiodine treatment in children and adolescents with

very high-risk radiation-induced differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2013;98(7):3039-3048.

29. Hay ID, Gonzalez-Losada T, Reinalda MS, Honetschlager JA, Richards ML, Thompson GB. Long-term outcome

in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J

Surg. 2010;34(6):1192-1202.

30. Marti JL, Jain KS, Morris LG. Increased risk of second primary malignancy in pediatric and young adult patients

treated with radioactive iodine for differentiated thyroid cancer. Thyroid. 2015;25(6):681-687.

31. Jonklaas J, Cooper DS, Ain KB, Bigos T, Brierley JD, Haugen BR, Ladenson PW, Magner J, Ross DS, Skarulis MC,

Steward DL, Maxon HR, Sherman SI. Radioiodine therapy in patients with stage I differentiated thyroid cancer.

Thyroid. 2010;20(12):1423-1424.

32. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA, Udelsman R. The treatment of

differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocr

Rev. 2011;32(6):798-826.

33. Welch Dinauer CA, Tuttle RM, Robie DK, McClellan DR, Svec RL, Adair C, Francis GL. Clinical features associated

with metastasis and recurrence of differentiated thyroid cancer in children, adolescents and young adults. Clin

Endocrinol (Oxf). 1998;49(5):619-628.

34. La Quaglia MP, Black T, Holcomb GW 3rd, Sklar C, Azizkhan RG, Haase GM, Newman KD. 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. J Pediatr

Surg. 2000;35(6):955-959.

35. Fridman M, Sawa N, Krasko O, Mankovskaya S, Branovan DI, Schmid KW, Demidchik Y. Initial presentation and late results

of treatment of post-Chernobyl papillary thyroid carcinoma in children and adolescents of Belarus. J Clin Endocrinol

Metab. 2014;99(8):2932-2941.

CLINICAL COURSE OF CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

39

2

SUPPLEMENTARY DATA

Supplemental Table 1. Participants versus non-participants

Characteristic Participants

(n = 105)

Non-participants

(n = 64)

P Valuea

Sex, n (%) 0.038

Male 17 (16.2) 19 (29.7)

Female 88 (83.8) 45 (70.3)

Age at diagnosis, years 15.6 (5.8-18.9) 15.5 (4.9-18.8) 0.498

Histology, n (%) 0.394

Papillary 85 (81.0) 50 (78.1)

Follicular 20 (19.0) 8 (12.5)

Unknown 0 (0.0) 6 (9.4)

TNM stage, n (%)T 0.960

T1-T2 65 (61.9) 21 (32.8)

T3-T4 26 (24.8) 14 (21.9)

Tx 14 (13.3) 29 (45.3)

N 0.339

N0 53 (50.5) 17 (26.6)

N1a-N1b 46 (43.8) 24 (37.5)

Nx 6 (5.7) 23 (35.9)

M 0.752

M0 82 (78.1) 27 (42.2)

M1b 14 (13.3) 12 (18.8)

Mx 9 (8.6) 25 (39.1)

Follow-up, years 11.7 (1.1-44.7) 18.1 (0.3-40.4)b 0.043

Numbers are expressed as median (range). a Missing or unknown values and Tx, Nx, Mx excluded from statistical

testing. b If date of diagnosis was available (n = 61). c ‘x’ indicates that there has been no assessment of that tumor

characteristic, or information about that characteristic was not available.

CHAPTER 2

40

2

Supplemental Table 2. History of radiotherapy and malignant neoplasms during childhood

Variable Patients

(n = 105)

Age at diagnosis

primary malignancy

other than DTC

Age at

diagnosis

DTC

Histology and

TNM DTC

DTC as primary malignancy, n (%)No history of radiation 99 (94.3) n.a. n.a. n.a.

History of cervical radiation for

benign conditionsa

2 (1.9) n.a. 17 y; 16 y PTC, TxN1bM0;

PTC, T1bN0M0

DTC as Second Malignant Neoplasm,

n (%)Langerhans cell histiocytosis Chemob 1 (0.95) 8 y 18 y FTC, T2N0M0

Acute Lymphoblastic Leukemia

Chemo + RTc

1 (0.95) 5 y 10 y PTC T1bN0M0

Medulloblastoma Chemo + RTd 1 (0.95) 9 y 18 y PTC, T3N1bM1

Neuroblastoma 131I-MIBG + chemoe 1 (0.95) 4 m 6 y PTC, T1N1M0

Abbreviations: DTC, differentiated thyroid carcinoma; Chemo, chemotherapy; RT, radiotherapy; 131I-MIBG, 131I-metaiodobenzylguanidine; n.a., not applicable; y, years; PTC, papillary thyroid carcinoma; FTC, follicular

thyroid carcinoma.a One patient received cervical RT for eczema (total dose 7.2 Gy) between ages 4-17 years. The second patient received

cervical RT (total dose breast and cervical region 32 Gy between ages 1-12 months for a nevus flammeus. Patients

received RT between 1958 and 1970. b Vinblastine, prednisolone, cyclophosphamide. c Rubidomycin, adriamycin,

cyclophosphamide + cranial RT dose 18 Gy. d Vincristine, cysplatinum + craniospinal RT dose 23.4 Gy. e 4x 131I-MIBG +

carboplatin, vincristine, etoposide, carboplatin, ifosfamide (VECI).

Supplemental Table 3. Second malignant neoplasms after pediatric differentiated thyroid carcinoma

Sex Histology

and TNM

Age at diagnosis

DTC, years

Cumulative 131I activity (GBq)

before SMN

SMN Age at

diagnosis

SMN, years

F PTC, TxNxMx 11 8.33 Breast cancer 39

F FTC, T4aN1bM0 13 35.15a Breast cancer 53

F PTC, T1bN1bM0 13 0 High-grade cervical

intraepithelial neoplasia

26

Abbreviations: DTC, differentiated thyroid carcinoma; SMN, second malignant neoplasm; F, female; PTC, papillary

thyroid carcinoma; FTC, follicular thyroid carcinoma. a During follow-up, this patient developed 2 recurrences, for

which she was treated with 131I.

Chapter 3

Chapter 3

CHAPTER 3

Distant metastases from childhood differentiated

thyroid carcinoma: clinical course and

mutational landscape

Marloes Nies, Rena Vassilopoulou-Sellin, Roland L. Bassett, Sireesha Yedururi, Mark E. Zafereo, Maria E. Cabanillas, Steven I. Sherman, Thera P. Links, and Steven G. Waguespack

Revised version submitted

CHAPTER 3

44

3

ABSTRACT ContextDistant metastases (DM) from childhood differentiated thyroid carcinoma (DTC) are uncommon and published studies are limited.

ObjectiveTo describe the long-term outcomes of patients with DM from childhood DTC and to evaluate the molecular landscape of these tumors.

Design, Setting, and PatientsRetrospective study at a tertiary cancer center including patients with pediatric DTC (diagnosed at age <19 years from 1946-2019) and DM.

ResultsWe identified 148 patients; 144 (97%) had papillary thyroid carcinoma (PTC) and 104 (70%) were female. Median age at DTC diagnosis was 13.4 (IQR 9.9-15.9) years. Evaluable subjects received a median of two (IQR 1-3) radioactive iodine (RAI) treatments at a median cumulative administered activity of 238.0 (IQR 147.5-351.0) mCi. The oncogenic driver was determined in 64/69 PTC samples: RET fusion (38/64; 59%), NTRK1/3 fusions (18/64; 28%), and the BRAF V600E mutation (8/64; 13%). At last evaluation, 93% had persistent disease. The median overall and disease-specific survival after DTC diagnosis were 50.7 and 52.8 years, respectively. Eight (5%) PTC patients died of disease after a median of 30.7 (IQR 20.6-37.6) years.

ConclusionsChildhood DTC with DM persists in most patients despite multiple courses of RAI, but disease-specific death is uncommon, typically occurring decades after diagnosis. Fusions are highly prevalent in PTC and all identified molecular alterations have appropriate targeted therapies. Future studies should focus on expanding genotype-phenotype correlations, determining how to integrate molecularly-targeted therapy into treatment paradigms, and relying less on repeated courses of RAI to achieve cure in patients with DM from childhood DTC.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

45

3

INTRODUCTION The most common endocrine malignancy, differentiated thyroid carcinoma (DTC) is rarely diagnosed during childhood and accounts for about 4% of all pediatric cancers (1). Approximately 90% of children with DTC have papillary thyroid carcinoma (PTC) whereas <10% are diagnosed with follicular thyroid carcinoma (FTC) (2,3). From 2012-2016, the age-adjusted incidence rates of thyroid cancer in the United States ranged from 1.6 cases/million/year in children ages 5-9 years to 34 cases/million/year in adolescents (ages 15-19), the most commonly affected pediatric age group (4). Rates of pediatric DTC have been increasing over the decades, a phenomenon that cannot solely be explained by an increased diagnosis of small or incidental tumors (1,2,4,5).

At diagnosis, children present with more advanced disease compared with adults. Children with PTC have larger primary tumor sizes with frequent extrathyroidal extension and are more often diagnosed with lymph node and distant metastases (DM) (6-17). The lungs are the primary site of DM, which is identified in up to ~25% of childhood DTC patients (7,13,15-34), depending on the series. The risk of DM correlates with the presence and extent of lymph node metastases (7,9,20,34). Despite more extensive disease at presentation, children have an extremely low disease-specific mortality regardless of the fact that most patients with DM from pediatric DTC do not reach remission (10,13,15,16,23,25,26,30,34-36).

The molecular pathogenesis of DTC, especially PTC, has become better elucidated over

the years. Point mutations and gene fusions that activate the mitogen-activated protein kinase

pathway play a major role in thyroid cancer development and propagation (37,38). In pediatric PTC, chromosomal rearrangements involving the REarranged during Transfection (RET) proto-oncogene and the neurotropic tyrosine receptor kinase (NTRK) genes (NTRK1 and NTRK3) are the most common oncogenic drivers (14,32,39-47). The BRAF V600E point mutation is also prevalent in childhood PTC (32,40-43,45-50), but at a much lower rate than adult PTC. These differences in molecular pathogenesis most likely explain the differences in clinical behavior observed between older and younger patients with PTC. Knowledge regarding tumor genotype may inform the expected clinical course, response to radioactive iodine (RAI), and potential for gene-targeted therapy for progressive DM. However, there has been very limited research in this area as relates to pediatric DTC.

The management of pediatric thyroid cancer with DM is challenging, given the excellent prognosis and protracted clinical course of childhood DTC, coupled with unique concerns regarding the potential long-term sequelae of overzealous treatment during childhood. To date, there have been few large-scale studies with comprehensive clinical data and extended follow-up periods in patients with DM from childhood DTC, and none of these incorporated testing for somatic molecular alterations. Therefore, data on clinical outcomes and tumor genotype are lacking in these patients. In the current study, we sought to describe the clinical characteristics

CHAPTER 3

46

3

and long-term outcomes of patients with DM and a diagnosis of pediatric DTC. The second aim was to evaluate the molecular genotype of pediatric PTC in a large subset of study subjects and to assess possible relationships between mutational status and clinical characteristics.

MATERIALS AND METHODSThis study was performed at The University of Texas MD Anderson Cancer Center, a tertiary referral center for thyroid cancer located in Houston, Texas, USA. Using institutional databases, we identified subjects diagnosed with pediatric DTC from 1946 to 2019 and retrospectively reviewed their medical records for eligibility. Patients were considered eligible for study inclusion if they had been diagnosed with DTC during childhood (defined as <19 years old) and were found to have DM at any time point of their follow-up, including adulthood. Subjects were excluded if they did not have at least one in-person clinic visit or if there was insufficient clinical information to determine DM status. Some subjects included in the current study have been reported in prior published studies and case reports (20,21,36,51,52). This study was approved by the MD Anderson Institutional Review Board. A waiver of informed consent was requested and granted by the IRB for the data collection and retrospective review.

Data retrievalData regarding diagnosis, pathology, molecular test results, surgical treatment, administration of RAI, additional therapies (surgery, systemic therapy, external beam radiation therapy or other interventions), and clinical follow-up (imaging; levels of thyroid-stimulating hormone (TSH), thyroglobulin (Tg), and thyroglobulin antibodies (TgAb)) were extracted from the electronic or paper medical records. If medical records were incomplete, subjects were contacted for written consent, and additional records were obtained from outside institutions to supplement the MD Anderson data.

Study definitionsDate of diagnosis was defined as the date of initial histologic confirmation of DTC resulting from primary thyroid surgery or core thyroid biopsy, excisional or core lymph node biopsy, or biopsy of DM. For staging, we used the 8th edition of the tumor node metastases classification of the American Joint Committee on Cancer (AJCC) (53). TNM stage was scored based on the maximal known disease extent during clinical follow-up. For example, if lateral neck lymph node or distant metastases were identified at any moment after initial therapy, the node stage would be scored as N1b or M1, respectively, with the assumption that this disease was present (but unrecognized) at the time of diagnosis. By definition, all patients included in this study were stage group II, even though DM may not have been identified within the first four months of diagnosis (as is used for AJCC staging). This was done because, due to the nature

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

47

3

of pediatric DTC and its indolent clinical course, exact staging at diagnosis was not available or inaccurate in some cases due to the delay in recognition of other sites of disease, especially decades ago when sensitive diagnostic imaging and tumor markers were unavailable. For TNM staging, if surgical and pathological data were incomplete or unavailable, or if the patient received systemic therapy prior to surgery, clinical data such as physical exam findings and imaging studies were used to complete staging. If the pathology report at MD Anderson was incongruent with the outside report, we used the MD Anderson interpretation for histopathologic characterization. Patients with PTC were categorized according to the American Thyroid Association (ATA) pediatric risk level classification (low-, intermediate-, or high-risk level) (11). ATA risk level was determined only by the initial clinical and histopathologic findings.

The date of DM diagnosis was the date of first confirmation of distant disease. If the date of the identification of the DM preceded the histological confirmation of DTC, the date of DTC diagnosis was considered to be the date of DM diagnosis. Distant metastases were defined as the presence of at least one of the following: i) RAI uptake consistent with iodine-avid thyroid cancer metastases on the diagnostic and/or therapeutic whole body scan outside of the thyroid bed or cervical/mediastinal lymph nodes; ii) pathologically-proven thyroid cancer tissue in the lung, bone, brain, or any other organ; iii) enlarging, discrete pulmonary nodules consistent with metastatic disease, coupled with a detectable Tg or persistently detectable or rising TgAb; iv) multiple (>10) non-calcified solid pulmonary nodules on imaging predominantly in the lower lung distribution and determined by the collaborating radiologist (SY) to be consistent with pulmonary metastases and associated with a detectable Tg or persistently detectable or rising TgAb. When pulmonary nodules were predominantly pleural based, they were not considered to be metastases. If imaging or RAI scans were ambiguous with regard to the diagnosis of DM, a radiologist (SY) reviewed the chest CT images to classify the patient as having DM or not.

Testing for the molecular oncogenic driver, when obtained, was done as part of routine patient care in Clinical Laboratory Improvement Amendments (CLIA)-certified laboratories. This analysis was done across a variety of testing platforms and included immunohistochemistry (IHC; primarily for the BRAF V600E mutation but in one case was also done to look for an NTRK fusion) as well as DNA and/or RNA sequencing. If a tumor tested positive for a known oncogenic mutation or fusion, this was considered to be a true positive result. A negative result was only considered as a true negative result when the tumor was “comprehensively” tested for all relevant oncogenic drivers, defined for this study as testing for BRAF and (N/K/H)RAS mutations and fusions involving the RET, NTRK1, NTRK3, and anaplastic lymphoma kinase (ALK) genes. When patients’ tumors tested negative and were not “comprehensively” tested, we considered them as not evaluated.

We scored disease status at last clinical evaluation as shown in Table 1. These categories were adapted from previously published dynamic staging definitions

CHAPTER 3

48

3

Table 1. Categories of disease status at last known clinical evaluation of patients with childhood DTC and

distant metastases

Category Imaging Thyroglobulin Thyroglobulin antibodies

No evidence of disease negative below LLNa,b below ULN

Persistent disease

Biochemical disease negative above LLNa,b any level

negative below LLN positive (stable or rising)

Structural disease positive above LLNa,b any level

positive any level positive (stable or rising)

Unable to determine known unknown unknown

unknown known known

Abbreviations: DTC, differentiated thyroid carcinoma; LLN, lower limit of normal; ULN, upper limit of normal. LLN and

ULN were determined by the laboratory-provided normal reference ranges for the particular assay used. a using the athyrotic range for athyrotic patients, if available. b includes both suppressed and stimulated thyroglobulin.

(54,55). Evaluable patients were considered to have both tumor markers and at least one type of imaging available for review; if data were missing, the disease status of these subjects was considered undeterminable. We did not incorporate an “indeterminate” category because we felt it highly unlikely that any durable detectable Tg or non-declining TgAb after long-term follow-up in a patient with known structural DM would represent anything other than persistent disease.

Deaths were classified as death from DTC, death from a cause other than DTC, or death from an unknown cause. Disease-specific survival and overall survival were evaluated, the interval of time defined as the date of diagnosis to the date of last contact date with the patient (last date of written or verbal contact with patient; contact with parent(s) in regard to the patient; or a completed clinic visit) or date of death. For disease-specific survival, patients with an unknown cause of death were excluded from the analysis. The length of follow-up for disease status was defined as the time between the date of diagnosis and the date of the last known clinical evaluation. Follow-up to last known disease status and follow-up to last known vital status could therefore differ.

Statistical analysesStudy data were collected and managed using REDCap electronic data capture tools (56,57) hosted at MD Anderson. Descriptive statistics were used to describe diagnostic, pathological, treatment, and outcome variables. Continuous variables are summarized with the median and interquartile range (IQR), unless otherwise specified. Mann–Whitney U tests were performed for continuous variables. We considered differences to be statistically significant at P < 0.05 (two-sided). R software (version

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

49

3

3.6.1) and Microsoft Office Professional Plus Excel 2016 (Microsoft Corporation) was used for statistical analyses.

RESULTSStudy SubjectsFrom 1946 to 2019, 602 patients with a new diagnosis or history of pediatric DTC (PTC=568; FTC=34) were registered and seen at least once at MD Anderson. Of these, 148 subjects (24.6%) had been diagnosed with DM at some point during their follow-up. Demographic and clinical characteristics are shown in Table 2. The majority of patients were female (n=104, 70.3%). Median age at DTC diagnosis was 13.4 years (IQR 9.9 to 15.9 years); the youngest subject was 2.8 years at diagnosis. Median age at DTC diagnosis did not significantly differ between females and males (13.6 years with IQR 10.2 to 16.4 years vs. 12.5 years with IQR 9.3 to 15.9 years, respectively, P = 0.197). Regarding age of diagnosis, 39 patients (26.4%) were diagnosed before the age of 10, 55 patients (37.2%) were diagnosed from 10 to 15 years, and 54 participants (36.5%) were diagnosed at

Table 2. Characteristics of patients with childhood DTC and distant metastases

Characteristic All patients

n = 148

PTC

n = 144

FTC

n = 4

Sex, n (%)Female 104 (70.3) 100 (69.4) 4 (100)

Male 44 (29.7) 44 (30.6) 0

Race/Ethnicity, n (%)White 89 (60.1) 85 (59.0) 4 (100)

Hispanic or Latino 41 (27.7) 41 (28.5) 0

Black 6 (4.1) 6 (4.2) 0

Asian 6 (4.1) 6 (4.2) 0

Half Black, half White 2 (1.4) 2 (1.4) 0

Other 4 (2.7)a 4 (2.8)a 0

Age at DTC diagnosis (years) 13.4 (9.9-15.9) 13.4 (9.8-15.9) 14.3 (12.2-16.8)

Range 2.8-18.9 2.8-18.9 12.2-18.5

Clinical presentation at diagnosis, n (%)Palpable thyroid nodule or neck mass 121 (87.1) 118 (87.4) 3 (75.0)

Incidental finding by imaging 6 (4.3) 6 (4.4) 0

Compressive symptoms 3 (2.2) 3 (2.2) 0

Nodule/neck mass with compressive symptoms 8 (5.8) 8 (5.9) 0

Other 1 (0.7) 0 1 (25.0)b

Unknown 9 9 0

Age at diagnosis is shown as median (interquartile range). Abbreviations: DTC, differentiated thyroid carcinoma;

PTC, papillary thyroid carcinoma; FTC, follicular thyroid carcinoma. a Pacific Islander (n=1), half Asian/half White (n=1),

half Hispanic/half Black (n=1), and half Hispanic/half White (n=1).b In 1 patient, the DTC diagnosis was made after

evaluation of overt hyperthyroidism and diagnosis of a functioning thyroid nodule.

CHAPTER 3

50

3

age 15 or older. White (60.1%), Hispanic/Latino (27.4%), Black (4.1%), and Asian (4.1%) were the most prevalent races/ethnicities. Information about clinical presentation was present for 139/148 patients. Most of these 139 patients (87.1%) presented with only a palpable nodule or neck mass that led to the diagnosis of DTC.

Only 13 of 142 (9.2%) patients with available information (all PTC) had a history of external beam radiation therapy (EBRT) with exposure to the neck prior to their cancer diagnosis (Supplemental Table 1 (58)). For seven cases, EBRT was part of their cancer therapy (acute myeloid leukemia (n=2), alveolar rhabdomyosarcoma, B-cell acute lymphocytic leukemia, Hodgkin lymphoma (n=2), neuroblastoma, and retinoblastoma). An eighth patient diagnosed with Hodgkin lymphoma before PTC diagnosis was not treated with EBRT. In six cases, patients received EBRT for benign conditions. A pre-existing thyroid diagnosis was reported in 16/138 (11.6%) evaluable patients (hypothyroidism/Hashimoto’s disease [n=8], hyperthyroidism/Graves’ disease [n=4], and goiter not otherwise specified [n=4]). A total of 11/133 (8.3%) evaluable patients reported to have family members with thyroid cancer; none of these reported DTC in a first-degree relative and seven patients had a second-degree family member with thyroid cancer. One patient was known to have familial Brugada syndrome at diagnosis but none of the other patients had a known familial syndrome at diagnosis.

Treatment (Table 3)Diagnostic surgeryDefinitive thyroid surgery was preceded by another diagnostic surgical procedure in 39 cases (26.4%, fine needle aspirations not included). For 32/39 patients, the first histologic confirmation of DTC was after a lymph node biopsy; 4/39 patients had a lung biopsy; 2/39 underwent a Sistrunk procedure with removal of an ectopic PTC; and one patient with widespread DM had the diagnosis confirmed after adrenalectomy. The median number of days between diagnostic surgery and initial thyroid surgery was 19 days (IQR 10 to 32 days; range 3 to 449 days).

Initial surgical treatmentThe majority of patients (n=114/148, 77.0%) had their initial thyroid surgery outside of MD Anderson, and 144/147 (98.0%) had a total thyroidectomy, including a completion thyroidectomy after lobectomy in 11 subjects. Details regarding the initial thyroid surgery were unknown in one case. Three patients (2.0%) with PTC did not have a total thyroidectomy after diagnosis. In one patient, only a neck dissection was performed since the intended total thyroidectomy could not be accomplished due to the invasiveness of the primary disease. In another patient, the intent of surgery was a total thyroidectomy, but the pathology report revealed no normal thyroid tissue and a residual thyroid lobe was appreciated during RAI therapy; it was assumed that

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

51

3

Table 3. Treatment of patients with childhood DTC and distant metastases

Treatment All patients

n = 148

PTC

n = 144

FTC

n = 4

Initial thyroid surgery, n (%)Total/subtotal thyroidectomy 133 (90.5) 131 (91.0) 2 (50.0)

Thyroid Lobectomy 11 (7.5) 9 (6.3) 2 (50.0)

Followed by completion

thyroidectomy

11 9 2

Other 3 (2.0)a 3 (2.1)a 0

Unknown 1 1 0

Initial lymph node resection, n (%)Yes 119 (86.9) 118 (88.1) 1 (33.3)

Only central compartment 13 13 -

Only lateral neck (uni/bilateral) 29 28 1

Both central and lateral neck 64 64 -

Unspecified lymph node resection 13b 13b -

No 18 (13.1) 16 (11.9) 2 (66.7)

Unknown 11 10 1

RAI, n (%)Yes 146 (98.6) 142 (98.6) 4 (100)

No 2 (1.4) 2 (1.4) 0

Time diagnosis DTC to first RAI (months) 2.7 (1.6-4.5)c 2.7 (1.6-4.4)d 3.2 (1.4-18.1)

Range 0.4-474.0 0.4-474.0 1.0-57.9

Number of RAI administrations/patient 2 (1-3)c 2 (1-3)d 1 (1-1.3)

Range 1-9 1-9 1-2

Activity of RAI per administration/

patient (mCi)

143.8 (97.4-158.0)e 143.6 (97.0-157.0)f 150.0 (114.8-175.9)

Range 26.4-532.0 26.4-532.0 104.5-190.0

Total cumulative activity (mCi)/patient 238.0 (147.5-351.0)g 241.4 (147.9-352.1)h 143.4 (119.9-206.3)

Range 29.1-1538.7 29.1-1538.7 104.5-340.0

Numbers are shown as median (interquartile range). Abbreviations: DTC, differentiated thyroid carcinoma; PTC,

papillary thyroid carcinoma; FTC, follicular thyroid carcinoma; RAI, radioactive iodine. a In 1 patient, the intended

total thyroidectomy could not be accomplished due to the invasiveness of the disease. In another patient, the intent

of the surgery was a total thyroidectomy, but the pathology report revealed no normal thyroid tissue. In a third

patient initially treated in 1946, only a nodulectomy was performed. b In 1 case, only a Delphian node was removed.

For the other 12 cases an unspecified selective lymph node excision was performed. c n=146 because RAI was not

administered in 2 cases. (See text.) d n=142, because RAI was not administered in 2 cases (See text). e 11/307 doses were

excluded from analysis due to missing data regarding the administered activity. 11/302 doses were excluded from

analysis due to missing data regarding the administered activity. g n=139, because 7 patients had at least one missing

administered activity of RAI and RAI was not administered in 2 cases. h n=135, because 7 patients had at least one

missing administered activity of RAI and RAI was not administered in 2 cases.

CHAPTER 3

52

3

tumor had completely replaced the bulk of the thyroid gland. In a third patient who was initially treated in 1946, only a nodulectomy was performed. Some extent of lymph node resection was done in 119/137 (86.9%) of the study subjects at the initial thyroid surgery (Table 3).

Radioactive iodine In the 146/148 (98.6%) patients treated with RAI, the first therapeutic RAI was administered after a median of 2.6 months from surgery (IQR 1.6 to 4.5 months). The median number of therapeutic RAI administrations was 2 (IQR 1 to 3; range 1 to 9). In evaluable subjects, the median administered activity per therapeutic session was 143.8 mCi (IQR 98.0 to 157.3 mCi, range 26.4 to 532.0 mCi). The median cumulative administered RAI activity of 139 patients with all dosing data known was 238.0 mCi (IQR 147.5 to 351.0 mCi, range 29.1 to 1538.7 mCi).

Two patients with PTC (1.4%) did not receive RAI. One recently-diagnosed patient, whose tumor harbored an NTRK1 fusion, had variable RAI uptake in the pulmonary metastases on a diagnostic scan; systemic therapy was started and RAI had not yet been administered. The second patient with a RET fusion positive tumor did not receive RAI because thyroidectomy could not be accomplished; her disease was treated systemically.

Additional treatmentAdditional surgeries besides the initial diagnostic and therapeutic surgeries took place in 84 (56.8%) patients. EBRT was given to 14 (9.5%) PTC patients (unknown in one patient). This approach was used as adjuvant therapy in five patients who were treated between 1946 and 1960. Nine PTC patients had palliative radiation therapy for metastatic disease between 1976 and 2013. Cytotoxic or targeted therapy was given to 23/143 (16.2%) PTC patients with DM (unknown in one PTC patient). None of the FTC patients were treated with systemic therapies (outside of RAI) or external beam therapy.

PathologyPathology characteristics are shown in Table 4 and Supplemental Table 2 (58). PTC was diagnosed in 144 (97.3%) patients and FTC in four subjects (2.7%). The most commons subtypes of PTC were the conventional (n=37, 25.7%) and follicular variants (n=27, 18.8%). All four patients with FTC (100%) had tumors that were encapsulated and angioinvasive, including one patient with an insular variant. The median tumor size for 115 evaluable PTC patients was 3.5 cm (IQR 2.3 to 5.5 cm) and 3.0 cm (IQR 2.5 to 3.5) for the four FTC patients. In terms of TNM staging, the majority of PTC tumors were staged as T3 (n=56, 38.9%) and 126 (87.5%) PTC patients had stage N1b disease. All four (100%) FTC tumors were staged as T2 and one (25.0%) had N1b disease. In three

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

53

3

Table 4. Pathology characteristics of patients with childhood DTC and distant metastases

Characteristic All patients

n = 148

PTC

n = 144

FTC

n = 4

Primary tumor size (cm) 3.5 (2.3-5.5)a 3.5 (2.3-5.5)b 3.0 (2.5-3.5)

Tumor stage, n (%)T1, T1a, or T1b 21 (14.2) 21 (14.6) 0

T2 40 (27.0) 36 (25.0) 4 (100)

T3, T3a, or T3b 56 (37.8) 56 (38.9) 0

T4a or T4b 16 (10.8) 16 (11.1) 0

Txc 15 (10.1) 15 (10.4) 0

Node stage, n (%)N0 6 (4.1) 3 (2.1) 3 (75.0)

N1 5 (3.4) 5 (3.5) 0

N1a 7 (4.7) 7 (4.9) 0

N1b 127 (85.8) 126 (87.5) 1 (25.0)

Nx 3 (2.0) 3 (2.1) 0

Focality, n (%)Unifocal 22 (23.7) 18 (20.2) 4 (100)

Multifocal, unilateral 17 (18.3) 17 (19.1) 0

Multifocal, bilateral 54 (58.1) 54 (60.7) 0

Unable to determine/unknown 55 55 0

Primary tumor size is show as median (interquartile range). Tumor and node stages represent maximal scores during

follow-up. Patients were scored according to the 8th edition of the American Joint Committee on Cancer staging

system. Pathological staging was leading. If surgical and pathological data were incomplete, clinical data were

used to complete staging. Diffusely infiltrating tumors were scored as multifocal and bilateral. Abbreviations: DTC,

differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma; FTC, follicular thyroid carcinoma. a 119 cases. b 115

cases. c Includes two patients with ectopic PTC.

(2.1%) PTC patients and 3 (75.0%) FTC patients, no metastases in lymph nodes were diagnosed. Most PTC patients (76.3%) had multifocal disease and all FTC cases were unifocal. ATA Pediatric Risk level was determinable in 126/144 (87.5%) PTC patients: 109/126 (86.5%) were high risk, 13/126 (10.3%) were intermediate risk, and 4/126 (3.2%) patients were low risk.

Distant Metastases The median time from initial DTC diagnosis to DM diagnosis was 2.6 months (IQR 0.6 to 18.5 months) for all patients; PTC patients had a median time to DM diagnosis of 2.6 months (IQR 0.5 to 15.6 months) whereas FTC patients were diagnosed with DM after a median time of 81.5 months (IQR 38.7 to 140.3 months). The median age at diagnosis of DM from DTC was 14.0 years (IQR 11.1 to 17.2 years, range 3.1 to 69.4 years); PTC patients had a median age at diagnosis of DM from DTC of 14.0 years (IQR 11.0 to 17.1 years, range 3.1 to 69.4 years) whereas FTC patients were diagnosed with DM at a median age

CHAPTER 3

54

3

of 22.1 years (IQR 15.4 to 29.6 years, range 12.6 to 34.8 years). The diagnosis of DM was made in 127/148 subjects (85.8%) before the age of 19 years. 26/148 patients (17.6%) were diagnosed with DM at age <10 years, 56 patients (37.8%) between 10 and 15 years, and 66 patients (44.6%) were diagnosed with DM at the age of 15 years or older. For 22 (15%) DTC patients, DM were identified before the histological confirmation of DTC. In these patients, DM were identified at a median of 0.4 months (IQR 0.2 to 1.2 months) before initial thyroid surgery.

All 144 PTC patients were diagnosed with DM to the lung, including 129/144 patients (89.6%) with lung metastases exclusively. Other sites of DM in PTC patients included bone (n=13; 9.0%), brain (n=8; 5.6%), liver (n=3; 2.1%), and adrenal and renal metastases in a single patient (0.7%) (Supplemental Table 3 (58)). Two out of the four FTC patients solely had lung metastases and the other two had bone metastases alone.

The majority (76.4%) of subjects were diagnosed with DM within two years after their cancer diagnosis. After four months, one, five, 10 and 20 years from DTC diagnosis, the proportion of patients diagnosed with DM was 60.1%, 68.2%, 90.5%, 95.3% and 96.6%, respectively. Details regarding the patients diagnosed with DM beyond 10 years after diagnosis (n=10, 6.8%) are shown in Supplemental Table 4 (58). Almost all patients with a delayed diagnosis of DM were diagnosed in an earlier era when less sensitive diagnostic testing was available, and the delay did not apparently lead to worse outcomes as five out of these seven patients were alive at last contact with their follow-up ranging from 19.2 to 66.1 years.

Mutational analysisThe tumors from most patients (95/148; 64.2%; PTC=94; FTC=1) underwent any testing to identify the oncogenic driver, and of these, 64 specimens (all PTC) were found to have a mutation or gene fusion (Table 5). Fusions were identified in 87.5% (56/64) whereas 8/64 (12.5%) tumors had the BRAF V600E mutation. Fusions involved RET in 38/64 (59.4%) cases (the most common partner being NCOA4) followed by NTRK1 (11/64; 17.2%) and NTRK3 (7/64; 10.9%). One tumor with a BRAF V600E mutation also had a second point mutation (p.E17K) in the v-akt murine thymoma viral oncogene homolog 1 (AKT1) gene. A RET fusion was identified in 3/13 tumors (not tested [n=8], not comprehensively tested [n=2]) from patients who had received EBRT before their DTC diagnosis. Of the 64 patients with a true positive result, the tissue tested included the primary tumor (n=17), lymph node metastasis (n=29), either primary tumor or lymph node (n=12; exact site unknown), or distant metastasis (n=6). The methodology that identified the oncogenic drivers was RNA sequencing in 46 cases, solely DNA sequencing in 16 cases, and BRAF V600E IHC in two cases.

Of the 31 tumors (PTC=30) that tested negatively for oncogenic drivers, only five PTC cases were comprehensively tested (i.e. tested for BRAF/RAS mutations and RET, NTRK1/3, and ALK fusions). All 31 tumors were tested for the BRAF V600E mutation and

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

55

3

Table 5. Somatic molecular analysis in patients with childhood papillary thyroid carcinoma and

distant metastases

Molecular analysis n = 144

Not tested 50 (34.7)

Tested with oncogenic driver identified 64 (45.8)

RET fusion 38 RET fusion 59%NCOA4/RET 21

CCDC6/RET 11

TRIM24/RET 2

ERC1/RET 2

PRKAR1A/RET 1

EML4/RET 1

NTRK1 fusion 11 NTRK1 fusion 17%TPR/NTRK1 4

TPM3/NTRK1 3

IRF2BP2/NTRK1 2

TFG/NTRK1 1

SQSTM1/NTRK1 1

NTRK3 fusion 7 NRTK3 fusion 11%ETV6/NTRK3 5

SQSTM1/NTRK3 2

BRAF V600E mutation 8a BRAF mutation 13%Tested without oncogenic driver identified 30 (20.8)

Not comprehensively testedb 25

Comprehensively testedc 5

Abbreviations: RET, rearranged during transfection; NTRK, neurotrophic tyrosine kinase receptor; BRAF, v-Raf murine

sarcoma viral oncogene homolog B. a In one patient, a BRAF V600E and a v-akt murine thymoma viral oncogene

homolog 1 (AKT1) mutation were found. b 20/25 patients were tested only for the BRAF V600E mutation and were

negative. c Tested for BRAF and RAS mutations and RET, NTRK1/3, and ALK fusions.

were found to be wild type. Of the 95 tested tumors, 62 samples (65.3%; all PTC) were tested for (N/K/H)RAS and all were found to be negative.

Supplemental Table 5 (58) shows the disease characteristics of the various oncogenic drivers. The youngest patient to have a tumor with an identified oncogenic driver (NTRK3 fusion) was diagnosed with PTC and widely metastatic DM at age four. The smallest median tumor size (2.9 cm) was found in patients with a BRAF mutation (RET fusion 4.1 cm; NTRK1 fusion 5.2 cm; and NTRK3 fusion 4.5 cm). The median time to diagnosis of DM was also longest in patients with a BRAF mutation (11.4 months vs. 3.1 months in RET fusions; 1.6 months in NTRK1 fusions; and 1.2 months in NTRK3 fusions). Given the small numbers of patients, we did not test whether these characteristics differed statistically.

CHAPTER 3

56

3

OutcomeFor all 148 patients, the median time from diagnosis to last known vital status was 11.8 years (IQR 7.2 to 18.3 years; range 0.8 to 66.6 years). Sixteen patients (10.8%) died during follow-up, including eight (5.4%) patients who died from PTC directly and two patients (1.4%) who died from complications of therapy: one at age 10 from respiratory failure caused by RAI-induced pulmonary fibrosis and the other at age 70 due to massive hemorrhage as a side effect from an anti-angiogenic tyrosine kinase inhibitor. Three deaths were not DTC related and the cause of death was unknown for three patients. None of the FTC patients died. The median overall survival after DTC diagnosis was 50.7 years, whereas the median disease-specific survival (excluding the three subjects in whom the cause of death was unknown) was 52.8 years (Figure 1). The 5-, 10-, 15-, 20-, 25-, and 30-year overall survival rates were 98.5%, 97.7%, 96.1%, 93.5%, 90.6%, and 86.8%, respectively. For disease specific deaths, these rates were 99.2%, 99.2%, 99.2%, 96.3%, 93.0%, and 93.0%, respectively. Overall and disease-specific survival did not differ for sex or age group (<10, 10-15, and ≥15 years) at DTC or DM diagnosis (Supplemental Figure 1 (58)). For the 132 survivors, the median follow-up to last known vital status was 11.1 years (IQR 7.0 to 16.1 years, range 0.8 to 65.3 years).

The patients who died from PTC (Supplemental Table 6 (58)) were diagnosed between 1956 and 2004. Median follow-up from diagnosis to death from PTC was 30.7 years (IQR 20.6 to 37.6 years, range 4.5 to 52.8 years) at a median age of 44.5 years (IQR 36.1 to 49.9 years, range 20.6 to 68.2 years). The youngest patient to die was 20.6 years of age and time from diagnosis to death was only 4.5 years; his was an extraordinary case of RAI-avid disease in a patient who had been heavily treated for stage IV neuroblastoma. All PTC patients who died from their disease had pulmonary and extrapulmonary DM at the time of death. Patients who died from PTC received a median cumulative administered RAI activity of 473.5 mCi (IQR 250.0 to 805.5 mCi) compared with a median of 226.7 mCi (141.8 to 331.0 mCi) in patients who were alive at last contact (reported on the seven and 125 patients with a known cumulative RAI activity, respectively (Supplemental Table 7 (58)). Molecular testing was done in tumors from only 2/8 patients who died from DTC, but testing was not comprehensive as defined by this study and no oncogenic driver was identified. Palliative EBRT was administered to 6/8 patients and 5/8 of these patients were treated with systemic therapy.

Median follow-up from diagnosis of DTC to last clinical evaluation was 10.5 years (IQR 6.3 to 16.2 years; range 0.8 to 66.1 years). We were unable to score disease status in 22/148 (14.9%) patients because insufficient data were available. At last clinical evaluation, 117/126 (92.9%) had persistent disease, of which 110 had structural evidence of disease and seven had biochemical evidence of disease. Only 9/126 evaluable subjects (7.1%) had no evidence of disease as strictly defined by the study (Supplemental Table 8 (58)). These nine patients were diagnosed from 1986 to 2014 at

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

57

3

Figure 1. Kaplan-Meier survival curves of patients diagnosed with distant metastases from childhood

differentiated thyroid carcinoma. (A) Overall survival (n=148). Sixteen patients died during follow-up.

The median overall survival was 50.7 years. (B) Disease-specific survival (n=145). Eight patients died

from PTC. No FTC-related deaths occurred. The median disease-specific survival was 52.8 years. The 5-,

10-, 15-, 20-, 25-, and 30-year overall survival rates were 98.5%, 97.7%, 96.1%, 93.5%, 90.6%, and 86.8%,

respectively. For disease specific deaths, these rates were 99.2%, 99.2%, 99.2%, 96.3%, 93.0%, and 93.0%,

respectively. For disease-specific survival, patients with an unknown cause of death (n=3) were excluded

from the analysis.

a median age of 15.5 years and, of the eight patients with complete records, the median cumulative administered activity of RAI was 125.2 mCi (66.5 to 175.6 mCi, Supplemental Table 7 (58)). Only spread of DTC to the lungs was identified in these patients. Median follow-up to last known vital status was 15.5 years for these nine patients having no evidence of disease.

DISCUSSIONChildhood DTC resulting in DM is rare, and previous studies on this topic have been limited by small numbers of subjects, inconsistent testing for molecular alterations, short follow-up periods in many pediatric series, and/or a lack of detailed clinical data

CHAPTER 3

58

3

(especially when derived from large cancer registries). To our knowledge, this single-center study, which focused on the clinical course and the mutational landscape of DTC in 148 patients diagnosed with DM from childhood DTC at any time point, is the largest of its kind with extended follow-up as long as six decades in some cases

The prevalence of DM from childhood DTC diagnosed at any time during follow-up has been reported to be as low as 5% (59) and as high as 42% (18), depending on the population studied and the treating center. In the current study, DM were identified in 24.6% of the subjects diagnosed with DTC of pediatric onset, and over two-thirds of these patients had their DM recognized within the first year after diagnosis. In some cases the DM will not be recognized until years into clinical follow-up, especially in patients diagnosed at a time when less sensitive diagnostic testing was available. We believe our study overestimates the true prevalence of DM in pediatric thyroid cancer patients, which is likely to be lower in the larger cohort of all children diagnosed with DTC. This overestimation is most certainly due to a referral bias, in addition to not knowing the exact denominator of all patients ever seen at our institution with a history of DTC diagnosis before age 19 years. Furthermore, we included patients with DM diagnosed during adulthood, which in turn will artificially elevate the proportion of patients who are likely to have more advanced disease (leading to their referral to a tertiary cancer center). Similar to other series, pulmonary metastases were nearly universal, especially in PTC where all patients in our study had lung metastases, and the identification of extra-pulmonary sites of disease (most commonly bone followed by brain; 11.5% in our series and 10.4% of PTC cases) was less common (6,17,23,24,28,31,59-62).

FTC represents a minority of DTC cases during childhood and has been reported even less frequently to be associated with DM, occurring in only 2/20 cases in a large Japanese series (63). In our study, only four FTC subjects (2.7 % of the entire DM group; 11.8% of all the pediatric FTC patients known in our center) were confirmed to have DM, and the sites of DM were equally split between the lungs and the skeleton. As would be anticipated, and similar to the study by Enomoto et al. (63) and others (6), our four cases of FTC were unifocal tumors with evidence for vascular invasion in all cases and a low prevalence of cervical lymph node disease. Although death due to pediatric FTC has been reported (23,29,59), we had no deaths from FTC in our series. Therefore, if deaths from FTC do occur, they appear to be extraordinarily rare. Given the small number of FTC cases, comparisons with PTC could not be made but it was noted that the latency period from FTC diagnosis to DM diagnosis was longer in the FTC patients, which underscores the importance of long-term follow-up of children with FTC, especially in the presence of angioinvasive tumors.

The current study was not designed to compare the clinical characteristics of patients with stage II DTC (DM+) with those of stage I (DM-) disease, but similar to prior studies (7,14,21,23,25,28,34,64), patients with DM had larger tumors and a very high rate of lymph node metastases to the lateral neck. The vast majority (86.5%) of

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

59

3

evaluable subjects were considered to be ATA high-risk at diagnosis. Patients with

DM treated at our center were more likely to be female, in keeping with the known

female predilection in DTC (4). Most of the distantly metastatic tumors comprised

conventional PTC followed by the follicular and diffuse sclerosing variants of PTC.

Outcomes from pediatric DTC are consistently reported to be excellent, even when

DM is present at diagnosis (3,10,13,30,34,35). However, most patients are not cured of

their disease and, similar to other recently published studies (14-16,23,25,26,34), we

identified a high prevalence of persistent disease (93%) in this population. In addition

to a referral bias, this high rate of persistent disease may reflect the strict definitions

used for the study and the better diagnostic studies available in the modern era of

DTC care such as more sensitive Tg assays and computed tomography. There is also

less reliance on diagnostic and post-treatment RAI scans for the identification of

pulmonary metastatic disease, recognizing that RAI non-avid disease does indeed

occur in children (18,19,34,52). The DTC was indolent in the vast majority of cases with

a notable long-term survival that exceeds any other distantly metastatic pediatric

solid tumor.

Death from pediatric DTC was uncommon; the median overall survival was 50.7

years and the median disease-specific survival was 52.8 years after diagnosis. Survival

did not differ based on the age at DTC or DM diagnosis and sex of the patient. When

death from disease occurred, it was attributed to PTC in all cases after a median of

three decades after initial diagnosis. In our series, 5.5% of evaluable subjects with

DM from pediatric-onset DTC died of disease. This is consistent with some studies

(6,15,25), higher than many others (14,17,19,24,26,28,29,61), and lower than some series

where death from childhood DTC occurred in 8-22% of cases with DM (10,13,18,27,32-

34). An interesting observation in our study was that those who died of their disease

all developed extrapulmonary metastatic disease. Most of the patients in the current

study who died of PTC were diagnosed in an earlier era when less sensitive clinical

tools were available for disease monitoring, when higher cumulative RAI activities

were given (thus theoretically increasing the risk of secondary tumor mutations and

more aggressive disease), and before there were advances in the understanding of

the molecular alterations that drive PTC and the availability of molecularly targeted

therapy. Moving forward, it is likely that the death rate will be lower as knowledge

regarding pediatric DTC expands and treatment approaches evolve. Notably, two

patients died of complications related to therapy, one from hemorrhage due to

a tyrosine kinase inhibitor prescribed for progressive disease and the other from

pulmonary fibrosis caused by overaggressive therapy with RAI. In the Chernobyl

pediatric cohort, pulmonary fibrosis was identified in 7.2% of 69 patients and resulted

in the death of one (1.5% of the 69 subjects) (65). Therefore, in children with diffuse

pulmonary disease that is RAI-avid, additional careful consideration must be given to

RAI dosing and frequency and monitoring for pulmonary complications of treatment.

CHAPTER 3

60

3

Our study is one of the first to report the underlying oncogenic driver in a large number of patients with DM from childhood DTC. Not unexpectedly, the most frequently found oncogenic driver was a fusion (RET fusions being predominant, representing 59% of PTC samples with a known driver). NTRK fusions were also common (28% of known drivers; NTRK1 > NTRK3) but the BRAF V600E mutation was surprisingly seen in ~13% of these tumors. As understood for PTC in general (37,45), oncogenic mutations are usually mutually exclusive. We found the same to hold true in this study since only one tumor in a patient with an aggressive presentation of metastatic PTC had two concurrent somatic mutations (BRAF V600E and AKT1). Of 69 PTC samples comprehensively tested in our study, the oncogenic driver was identified in 93% of cases. In another recent study with comprehensive molecular testing (32), all 10 patients with DM were found to have a molecular alteration, suggesting that the molecular driver can be identified in almost all cases of pediatric stage II PTC after comprehensive testing. Importantly, this has treatment implications since the most commonly identified fusions and point mutations all have commercially available, molecularly targeted therapies that can be used for systemic therapy if clinically indicated.

The medical literature is sparse as relates to the molecular profiles of distantly metastatic tumors and most prior studies have been limited by the lack of comprehensive molecular testing, especially for NTRK fusions. In line with previous studies (25,32), RET fusions were the most commonly found oncogenic drivers in patients with DM from childhood DTC. A RAS mutation was not found in any of our cases, and we are aware of only two reported cases of a RAS mutation (one NRAS and one HRAS) in young patients with DM (25,32). ALK fusions were also not identified in our patient population. Although ALK fusions in pediatric PTC have been described (45), unequivocal cases of ALK fusion positive distantly metastatic childhood PTC have not yet been published. Therefore, it can be concluded that point mutations in the RAS gene and ALK fusions are rare in this population. Furthermore, there have been cases of distantly metastatic tumors with MET (32) and AGK-BRAF fusions (50,66). In the five PTC patients comprehensively tested for somatic molecular alterations with negative results, we did not identify a BRAF (n=4) or MET fusion (n=3). In three of these cases, the tumor was tested in a laboratory (using DNA and RNA sequencing) where we have seen false negative results, in one the testing was primarily done via analysis of cell free DNA (liquid biopsy), and the fifth case had DNA-based testing alone. Therefore we cannot rule out a false negative result in these cases. In the setting of an uninformative test result when systemic therapy is warranted and knowledge of the oncogenic driver paramount, repeat testing in a different laboratory using RNA-based next generation sequencing should be considered, understanding that this approach provides optimal screening for fusions in genes such as NTRK1 and NTRK3 (67).

Our sample size was too small to statistically analyze genotype-phenotype correlations. However, it appears that patients with BRAF-mutated tumors had smaller

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

61

3

tumor sizes and were diagnosed with DM later than patients with other identified

oncogenic drivers. It is hypothesized that, due to the increased risk of RAI refractory

disease in BRAF-mutated PTC (68,69), it takes longer to recognize DM in these

cases due to the poor sensitivity of RAI scans. There were no obvious differences in

the clinical characteristics between RET and NTRK fusions.

This study was not designed to evaluate the prevalence of RAIR refractory disease

since the images of diagnostic and post-therapy RAI scans were not available in

all cases and for all episodes of therapy. Although the criteria for RAI-refractory

disease have been defined in adults (68), this has not been studied in children. Some

of the adult criteria, such as lack of RAI uptake in known structural disease, would

certainly be applicable to pediatric DTC but others such as the 600 mCi threshold is not

translatable to children, understanding that the cumulative mCi activity that defines

RAI refractory disease in an eight-year-old would be quite different from an 18-year-

old. Given the high rate of persistent disease in our study despite a median of two RAI

treatments and a median cumulative activity of 248.9 mCi, it would appear that RAI-

refractory disease is more common than previously recognized. However, unlike older

patients with DTC, RAI-refractory disease in children, even if fluorodeoxyglucose-avid,

can remain indolent for decades (70) and can be associated with declining Tg levels

despite no further RAI therapy (71). Understanding that the long-term prognosis in

patients with DM from DTC diagnosed during childhood is excellent and recognizing

the high rates of persistent disease despite aggressive RAI therapy, one could strongly

argue that repeated RAI therapy is unlikely to benefit these patients and may result

in more harm than good over the course of their lives. Ultimately, further studies of

the genotype-phenotype correlations in pediatric DTC and a better understanding of

what distinguishes those cured of their disease from those with persistent disease will

help to inform decisions regarding RAI during childhood.

Strengths and limitationsThe main strengths of the current study are the size of the cohort, the length of follow-

up, and the large group of patients whose tumors were tested molecularly. We were

able to describe the clinical outcomes of 148 patients with DM from childhood DTC

through decades of follow-up in many cases. We also identified the oncogenic drivers

in the largest number of childhood DTC patients with DM. Limitations of the study are

as would be expected for a retrospective study wherein data was not prospectively

collected or documented in a systematic fashion. We also had several patients in whom

we could not determine disease status due to incomplete staging and, in exceptional

cases, structural disease may have been overcalled (for example, an abnormal cervical

US without confirmatory biopsy; persistent but stable lung nodules that may or may

not be active sites of cancer); in these cases the Tg or TgAb was still abnormal and

therefore, at a minimum, would still be categorized as persistent disease. Furthermore,

CHAPTER 3

62

3

our patient population is likely to be sicker with more advanced disease compared with other academic centers due to our practice in a comprehensive cancer center with known expertise in thyroid cancer management. Thus our reported outcomes may be worse.

In conclusion, at a tertiary cancer center, DM occurs in up to 25% of pediatric DTC cases. These cases are more enriched for PTC compared with the expected distribution in DTC as a whole and the lungs are universally affected in PTC. Although most cases are diagnosed within a year after DTC diagnosis, the recognition of DM can be delayed by years, especially in FTC, which highlights the importance of long-term follow-up in children with DTC. Fortunately, despite a high prevalence of persistent biochemical and structural disease, the prognosis remains excellent with death from DTC occurring in few patients and typically decades after diagnosis. Given the excellent long-term survival outcomes and high proportion of persistent disease as seen in our study and others, it behooves us to reconsider the aggressiveness of treatment at a young age when the patient may be more at risk for the life-long sequelae of surgical therapy and RAI. Gene rearrangements (i.e. RET and NTRK1/3 fusions) occurred in 88% of PTC cases with a known oncogenic driver, and all identified alterations currently have targeted systemic therapies available. Future studies should focus on expanding the genotype-phenotype correlations in stage II pediatric DTC, determining the best way to integrate molecularly-targeted therapy into treatment paradigms, and relying less on repeated courses of RAI to achieve cure in patients with DM from childhood DTC.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

63

3

ACKNOWLEDGEMENTSWe are forever grateful to the patients and families who contributed to the current study; your courage in the face of adversity inspires us. This study could not have been completed without the help of Pamela Waddilove, Danielle Litofsky, Jennifer Ellefson, and Damaris Cruz-Goldberg. We also acknowledge our colleagues in Pathology and Endocrine Neoplasia who helped to answer questions and cared for patients on the study. We thank the Prince Bernhard Culture Fund and the Academy Ter Meulen Grant of the Royal Netherlands Academy of Arts and Sciences for their financial support. This work was also supported in part by the NIH/NCI Cancer Center Support Grant (award number P30CA016672) and used the MD Anderson Biostatistics Resource Group. We thank the Junior Scientific Masterclass Groningen for extending the MD/PhD-program to allow Dr. Nies to perform this research. Finally, we are very grateful to Ms. Sandra Montez and Dr. David Hong in Investigational Cancer Therapeutics for their assistance with molecular testing.

DATA AVAILABILITYRestrictions apply to the availability of data generated or analyzed during this study to preserve patient confidentiality or because they were used under license. The corresponding author will upon request detail the restrictions and any conditions under which access to some data may be provided.

DISCLOSURE SUMMARYThe authors have nothing to disclose.

CHAPTER 3

64

3

REFERENCES1. Siegel DA, King J, Tai E, Buchanan N, Ajani UA, Li J. Cancer incidence rates and trends among children and

adolescents in the United States, 2001-2009. Pediatrics. 2014;134(4):e945-955.

2. Bernier MO, Withrow DR, Berrington de Gonzalez A, Lam CJK, Linet MS, Kitahara CM, Shiels MS. Trends in pediatric

thyroid cancer incidence in the United States, 1998-2013. Cancer. 2019;125(14):2497-2505.

3. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167-172.

4. Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS,

Feuer EJ, Cronin KAe. SEER Cancer Statistics Review, 1975-2016, based on November 2018 SEER data submission,

posted to the SEER web site, April 2019. Vol 2020. Bethesda, MD: National Cancer Institute; 2019.

5. Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I, Parkin DM. Thyroid cancer incidence and survival

among European children and adolescents (1978-1997): report from the Automated Childhood Cancer Information

System project. Eur J Cancer. 2006;42(13):2150-2169.

6. Welch Dinauer CA, Tuttle RM, Robie DK, McClellan DR, Svec RL, Adair C, Francis GL. Clinical features associated

with metastasis and recurrence of differentiated thyroid cancer in children, adolescents and young adults. Clin

Endocrinol. 1998;49(5):619-628.

7. Wada N, Sugino K, Mimura T, Nagahama M, Kitagawa W, Shibuya H, Ohkuwa K, Nakayama H, Hirakawa S, Yukawa

N, Rino Y, Masuda M, Ito K. Treatment strategy of papillary thyroid carcinoma in children and adolescents: clinical

significance of the initial nodal manifestation. Ann Surg Onc. 2009;16(12):3442-3449.

8. Jarzab B, Handkiewicz-Junak D. Differentiated thyroid cancer in children and adults: same or distinct disease?

Hormones (Athens, Greece). 2007;6(3):200-209.

9. Machens A, Lorenz K, Nguyen Thanh P, Brauckhoff M, Dralle H. Papillary thyroid cancer in children and adolescents

does not differ in growth pattern and metastatic behavior. J Pediatr. 2010;157(4):648-652.

10. Hay ID, Gonzalez-Losada T, Reinalda MS, Honetschlager JA, Richards ML, Thompson GB. Long-term outcome

in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J

Surg. 2010;34(6):1192-1202.

11. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management Guidelines for Children with Thyroid Nodules and Differentiated Thyroid Cancer.

Thyroid. 2015;25(7):716-759.

12. Spinelli C, Strambi S, Rossi L, Bakkar S, Massimino M, Ferrari A, Collini P, Cecchetto G, Bisogno G, Inserra A,

Bianco F, Miccoli P. Surgical management of papillary thyroid carcinoma in childhood and adolescence: an Italian

multicenter study on 250 patients. J Endocrinol Invest. 2016;39(9):1055-1059.

13. Hay ID, Johnson TR, Kaggal S, Reinalda MS, Iniguez-Ariza NM, Grant CS, Pittock ST, Thompson GB. Papillary

thyroid carcinoma (PTC) in children and adults: comparison of initial presentation and long-term postoperative

outcome in 4432 patients consecutively treated at the Mayo Clinic during eight decades (1936-2015). World J

Surg. 2018;42(2):329-342.

14. Galuppini F, Vianello F, Censi S, Barollo S, Bertazza L, Carducci S, Colato C, Manso J, Rugge M, Iacobone M,

Watutantrige Fernando S, Pennelli G, Mian C. Differentiated thyroid carcinoma in pediatric age: genetic and

clinical scenario. Front Endocrinol (Lausanne). 2019;10:552.

15. Alzahrani AS, Alswailem M, Moria Y, Almutairi R, Alotaibi M, Murugan AK, Qasem E, Alghamdi B, Al-Hindi H.

Lung metastasis in pediatric thyroid cancer: radiological pattern, molecular genetics, response to therapy, and

outcome. J Clin Endocrinol Metab. 2019;104(1):103-110.

16. Zhang XY, Song HJ, Qiu ZL, Shen CT, Chen XY, Sun ZK, Wei WJ, Zhang GQ, Luo QY. Pulmonary metastases in

children and adolescents with papillary thyroid cancer in China: prognostic factors and outcomes from treatment

with (131)I. Endocrine. 2018;62(1):149-158.

17. Bal CS, Garg A, Chopra S, Ballal S, Soundararajan R. Prognostic factors in pediatric differentiated thyroid cancer

patients with pulmonary metastases. J Pediatr Endocrinol Metab. 2014.

18. Schlumberger M, De Vathaire F, Travagli JP, Vassal G, Lemerle J, Parmentier C, Tubiana M. Differentiated thyroid

carcinoma in childhood: long term follow-up of 72 patients. J Clin Endocrinol Metab. 1987;65(6):1088-1094.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

65

3

19. Dottorini ME, Vignati A, Mazzucchelli L, Lomuscio G, Colombo L. Differentiated thyroid carcinoma in children

and adolescents: a 37-year experience in 85 patients. J Nucl Med. 1997;38(5):669-675.

20. Vassilopoulou-Sellin R, Klein MJ, Smith TH, Samaan NA, Frankenthaler RA, Goepfert H, Cangir A, Haynie TP.

Pulmonary metastases in children and young adults with differentiated thyroid cancer. Cancer. 1993;71(4):1348-1352.

21. La Quaglia MP, Black T, Holcomb GW, 3rd, Sklar C, Azizkhan RG, Haase GM, Newman KD. 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. J Pediatr Surg.

2000;35(6):955-959; discussion 960.

22. Bal CS, Kumar A, Chandra P, Dwivedi SN, Mukhopadhyaya S. 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. 2004;14(3):217-225.

23. Demidchik YE, Demidchik EP, Reiners C, Biko J, Mine M, Saenko VA, Yamashita S. Comprehensive clinical

assessment of 740 cases of surgically treated thyroid cancer in children of Belarus. Ann Surg. 2006;243(4):525-532.

24. Klein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-

Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Schmid KW, Smit JW, Williams

GR, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP. Pediatric differentiated thyroid carcinoma in

the Netherlands: a nationwide follow-up study. J Clin Endocrinol Metab. 2016;101(5):2031-2039.

25. Chesover AD, Vali R, Hemmati SH, Wasserman JD. Lung metastasis in children with differentiated thyroid cancer: factors

associated with diagnosis and outcomes of therapy. Thyroid. 2020. Online ahead of print. doi: 10.1089/thy.2020.0002

26. Vaisman F, Bulzico DA, Pessoa CH, Bordallo MA, Mendonca UB, Dias FL, Coeli CM, Corbo R, Vaisman M. Prognostic

factors of a good response to initial therapy in children and adolescents with differentiated thyroid cancer. Clinics

(Sao Paulo). 2011;66(2):281-286.

27. Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Mang O, Lau WH. Differentiated thyroid carcinoma in childhood

and adolescence-clinical course and role of radioiodine. Pediatr Blood Cancer. 2004;42(2):176-183.

28. Prpic M, Franceschi M, Jukic T, Kust D, Dabelic N, Varjacic T, Lucijanic M, Bolanca A, Kusic Z. Differentiated Thyroid

Cancer in Pediatric Population (</=18 Years): Postoperative Treatment with Radioactive Iodine (I-131). Acta Clin

Croat. 2019;58(1):119-127.

29. Tamam M, Uyanik E, Edis N, Mulazimoglu M, Ozpacaci T. Differentiated thyroid carcinoma in children: Clinical

characteristics and long-term follow-up. World J Nucl Med. 2020;19(1):28-35.

30. Golpanian S, Perez EA, Tashiro J, Lew JI, Sola JE, Hogan AR. Pediatric papillary thyroid carcinoma: outcomes and

survival predictors in 2504 surgical patients. Pediatr Surg Int. 2016;32(3):201-208.

31. Handkiewicz-Junak D, Wloch J, Roskosz J, Krajewska J, Kropinska A, Pomorski L, Kukulska A, Prokurat A, Wygoda

Z, Jarzab B. Total thyroidectomy and adjuvant radioiodine treatment independently decrease locoregional

recurrence risk in childhood and adolescent differentiated thyroid cancer. J Nucl Med. 2007;48(6):879-888.

32. Pekova B, Sykorova V, Dvorakova S, Vaclavikova E, Moravcova J, Katra R, Astl J, Vlcek P, Kodetova D, Vcelak J,

Bendlova B. RET, NTRK, ALK, BRAF and MET fusions in a large cohort of pediatric papillary thyroid carcinomas.

Thyroid. 2020. Online ahead of print. doi: 10.1089/thy.2019.0802.

33. Pekova B, Dvorakova S, Sykorova V, Vacinova G, Vaclavikova E, Moravcova J, Katra R, Vlcek P, Sykorova P, Kodetova

D, Vcelak J, Bendlova B. Somatic genetic alterations in a large cohort of pediatric thyroid nodules. Endocr

Connect. 2019;8(6):796-805.

34. Sugino K, Nagahama M, Kitagawa W, Ohkuwa K, Uruno T, Matsuzu K, Suzuki A, Tomoda C, Hames KY, Akaishi J,

Masaki C, Ito K. Distant metastasis in pediatric and adolescent differentiated thyroid cancer: Clinical outcomes

and risk factor analyses. J Clin Endocrinol Metab. 2020; dgaa545.

35. Pawelczak M, David R, Franklin B, Kessler M, Lam L, Shah B. Outcomes of children and adolescents with well-

differentiated thyroid carcinoma and pulmonary metastases following (1)(3)(1)I treatment: a systematic review.

Thyroid. 2010;20(10):1095-1101.

36. Vassilopoulou-Sellin R, Goepfert H, Raney B, Schultz PN. Differentiated thyroid cancer in children and adolescents:

clinical outcome and mortality after long-term follow-up. Head Neck. 1998;20(6):549-555.

37. Cancer Genome Atlas Research N. Integrated genomic characterization of papillary thyroid carcinoma.

Cell. 2014;159(3):676-690.

CHAPTER 3

66

3

38. Yoo SK, Lee S, Kim SJ, Jee HG, Kim BA, Cho H, Song YS, Cho SW, Won JK, Shin JY, Park do J, Kim JI, Lee KE, Park YJ,

Seo JS. Comprehensive analysis of the transcriptional and mutational landscape of follicular and papillary thyroid

cancers. PLoS genetics. 2016;12(8):e1006239.

39. Hardee S, Prasad ML, Hui P, Dinauer CA, Morotti RA. Pathologic characteristics, natural history, and prognostic

implications of brafv600e mutation in pediatric papillary thyroid carcinoma. Pediatr Dev Pathol. 2017;20(3):206-212.

40. Paulson VA, Rudzinski ER, Hawkins DS. Thyroid cancer in the pediatric population. Genes (Basel). 2019;10(9).

41. Bauer AJ. Molecular genetics of thyroid cancer in children and adolescents. Endocrinol Metab Clin North

Am. 2017;46(2):389-403.

42. Picarsic JL, Buryk MA, Ozolek J, Ranganathan S, Monaco SE, Simons JP, Witchel SF, Gurtunca N, Joyce J, Zhong S,

Nikiforova MN, Nikiforov YE. Molecular Characterization of Sporadic Pediatric Thyroid Carcinoma with the DNA/

RNA ThyroSeq v2 Next-Generation Sequencing Assay. Pediatr Dev Pathol. 2016;19(2):115-122.

43. Nikita ME, Jiang W, Cheng SM, Hantash FM, McPhaul MJ, Newbury RO, Phillips SA, Reitz RE, Waldman FM, Newfield

RS. Mutational analysis in pediatric thyroid cancer and correlations with age, ethnicity, and clinical presentation.

Thyroid. 2016;26(2):227-234.

44. Cordioli MI, Moraes L, Bastos AU, Besson P, Alves MT, Delcelo R, Monte O, Longui C, Cury AN, Cerutti JM.

Fusion oncogenes are the main genetic events found in sporadic papillary thyroid carcinomas from children.

Thyroid. 2017;27(2):182-188.

45. Vanden Borre P, Schrock AB, Anderson PM, Morris JC, 3rd, Heilmann AM, Holmes O, Wang K, Johnson A,

Waguespack SG, Ou SI, Khan S, Fung KM, Stephens PJ, Erlich RL, Miller VA, Ross JS, Ali SM. Pediatric, adolescent,

and young adult thyroid carcinoma harbors frequent and diverse targetable genomic alterations, including kinase

fusions. Oncologist. 2017;22(3):255-263.

46. Mostoufi-Moab S, Labourier E, Sullivan L, LiVolsi V, Li Y, Xiao R, Beaudenon-Huibregtse S, Kazahaya K,

Adzick NS, Baloch Z, Bauer AJ. Molecular testing for oncogenic gene alterations in pediatric thyroid lesions.

Thyroid. 2018;28(1):60-67.

47. Alzahrani AS, Alswailem M, Alswailem AA, Al-Hindi H, Goljan E, Alsudairy N, Abouelhoda M. Genetic

alterations in pediatric thyroid cancer using a comprehensive childhood cancer gene panel. J Clin Endocrinol

Metab. 2020;105(10):dgaa389.

48. Cordioli MI, Moraes L, Cury AN, Cerutti JM. Are we really at the dawn of understanding sporadic pediatric thyroid

carcinoma? Endocr Relat Cancer. 2015;22(6):R311-324.

49. Alzahrani AS, Murugan AK, Qasem E, Alswailem M, Al-Hindi H, Shi Y. Single point mutations in pediatric

differentiated thyroid cancer. Thyroid. 2017;27(2):189-196.

50. Sisdelli L, Cordioli M, Vaisman F, Moraes L, Colozza-Gama GA, Alves PAG, Jr., Araujo ML, Jr., Alves MTS, Monte O,

Longui CA, Cury AN, Carvalheira G, Cerutti JM. AGK-BRAF is associated with distant metastasis and younger age in

pediatric papillary thyroid carcinoma. Pediatr Blood Cancer. 2019;66(7):e27707.

51. Vassilopoulou-Sellin R, Libshitz HI, Haynie TP. Papillary thyroid cancer with pulmonary metastases beginning in

childhood: clinical course over three decades. Med Pediat Oncol. 1995;24(2):119-122.

52. Waguespack SG, Sherman SI, Williams MD, Clayman GL, Herzog CE. The successful use of sorafenib to treat

pediatric papillary thyroid carcinoma. Thyroid. 2009;19(4):407-412.

53. AJCC Cancer Staging Manual. 8th ed: Springer International Publishing.

54. Tuttle RM, Leboeuf R. Follow up approaches in thyroid cancer: a risk adapted paradigm. Endocrinology and

metabolism clinics of North America. 2008;37(2):419-435, ix-x.

55. Sung TY, Jeon MJ, Lee YH, Lee YM, Kwon H, Yoon JH, Chung KW, Kim WG, Song DE, Hong SJ. Initial and

Dynamic Risk Stratification of Pediatric Patients With Differentiated Thyroid Cancer. J Clin Endocrinol

Metab. 2017;102(3):793-800.

56. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)--a

metadata-driven methodology and workflow process for providing translational research informatics support. J

Biomed Inform. 2009;42(2):377-381.

57. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O’Neal L, McLeod L, Delacqua G, Delacqua F, Kirby J, Duda SN. The REDCap

consortium: Building an international community of software platform partners. J Biomed Inform. 2019;95:103208.

58. Nies M, Vassilopoulou-Sellin R, Bassett RL, Yedururi S, Zafereo ME, Cabanillas ME, Sherman SI, Links TP,

Waguespack SG. Data from: Distant Metastases from Childhood DTC - Supplemental Material. 2020. Deposited

September 12, 2020. https://doi.org/10.6084/m9.figshare.12948386.v1.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

67

3

59. Popovtzer A, Shpitzer T, Bahar G, Feinmesser R, Segal K. Thyroid cancer in children: management and outcome

experience of a referral center. Otolaryngol Head Neck Surg. 2006;135(4):581-584.

60. Park S, Jeong JS, Ryu HR, Lee CR, Park JH, Kang SW, Jeong JJ, Nam KH, Chung WY, Park CS. Differentiated thyroid

carcinoma of children and adolescents: 27-year experience in the yonsei university health system. J Korean Med

Sci. 2013;28(5):693-699.

61. Reiners C, Biko J, Haenscheid H, Hebestreit H, Kirinjuk S, Baranowski O, Marlowe RJ, Demidchik E, Drozd V,

Demidchik Y. Twenty-five years after Chernobyl: outcome of radioiodine treatment in children and adolescents with

very high-risk radiation-induced differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2013;98(7):3039-3048.

62. Schlumberger M, Brose M, Elisei R, Leboulleux S, Luster M, Pitoia F, Pacini F. Definition and management of

radioactive iodine-refractory differentiated thyroid cancer. Lancet Diabetes Endocrinol. 2014;2(5):356-358.

63. Enomoto K, Enomoto Y, Uchino S, Yamashita H, Noguchi S. Follicular thyroid cancer in children and adolescents:

clinicopathologic features, long-term survival, and risk factors for recurrence. Endocr J. 2013;60(5):629-635.

64. Livhits MJ, Pasternak JD, Xiong M, Li N, Gosnell JE, Yeh MW, Chiu HK. Pre-ablation Thyroglobulin and Thyroglobulin

to thyroid-stimulating hormone ratio may be associated with pulmonary metastases in children with differentiated

thyroid cancer. Endocr Pract. 2016;22(11):1259-1266.

65. Hebestreit H, Biko J, Drozd V, Demidchik Y, Burkhardt A, Trusen A, Beer M, Reiners C. Pulmonary fibrosis in youth

treated with radioiodine for juvenile thyroid cancer and lung metastases after Chernobyl. Eur J Nucl Med Mol

Imaging. 2011;38(9):1683-1690.

66. Chu YH, Wirth LJ, Farahani AA, Nose V, Faquin WC, Dias-Santagata D, Sadow PM. Clinicopathologic features of

kinase fusion-related thyroid carcinomas: an integrative analysis with molecular characterization. Mod Pathol.

2020. Online ehead of print. doi: 10.1038/s41379-020-0638-5.

67. Penault-Llorca F, Rudzinski ER, Sepulveda AR. Testing algorithm for identification of patients with TRK fusion

cancer. J Clin Pathol. 2019;72(7):460-467.

68. Aashiq M, Silverman DA, Na’ara S, Takahashi H, Amit M. Radioiodine-Refractory Thyroid Cancer: Molecular Basis of

Redifferentiation Therapies, Management, and Novel Therapies. Cancers (Basel). 2019;11(9).

69. Sabra MM, Dominguez JM, Grewal RK, Larson SM, Ghossein RA, Tuttle RM, Fagin JA. Clinical outcomes and

molecular profile of differentiated thyroid cancers with radioiodine-avid distant metastases. J Clin Endocrinol

Metab. 2013;98(5):E829-836.

70. Kwong N, Marqusee E, Gordon MS, Larsen PR, Garber JR, Kim MI, Alexander EK. Long-term, treatment-free

survival in select patients with distant metastatic papillary thyroid cancer. Endocr Connect. 2014;3(4):207-214.

71. Biko J, Reiners C, Kreissl MC, Verburg FA, Demidchik Y, Drozd V. Favourable course of disease after incomplete

remission on (131)I therapy in children with pulmonary metastases of papillary thyroid carcinoma: 10 years follow-

up. Eur J Nucl Med Mol Imaging. 2011;38(4):651-655.

CHAPTER 3

68

3

SUPPLEMENTARY DATADOI: https://doi.org/10.6084/m9.figshare.12948386

Supplemental Table 1. Prior Malignancies and Risk Factors for Childhood DTC prior to Diagnosis

All patients

n = 148

PTC

n = 144

FTC

n = 4

History of EBRT to the neck, n (%)No 129 (90.8) 126 (90.6) 3 (100)

Yes 13 (9.2) 13 (9.4) 0

Malignant condition 7 7 -

Benign condition 6a 6a -

Unknown 6 5 1

History of previous cancer, n (%)No 137 (94.5) 133 (94.3) 4 (100)

Yes 8 (5.5) 8 (5.7) 0

Acute myeloid leukemia 2 2 -

Alveolar rhabdomyosarcoma 1 1 -

B-cell acute lymphocytic leukemia 1 1 -

Hodgkin lymphoma 2 2 -

Neuroblastoma 1 1 -

Retinoblastoma 1 1 -

Unknown 3 3 0

History of RAI treatment or exposure, n (%)No 140 (100) 137 (100) 3 (100)

Unknown 8 7 1

Previous thyroid disease, n (%)No 122 (88.4) 121 (89.0) 1 (50.0)

Yes 16 (11.6) 15 (11.0) 1 (50.0)

Hypothyroidism/Hashimoto’s Disease 8 8 0

Hyperthyroidism/Graves’ Disease 4 3 1b

Goiter not otherwise specified 4 4 0

Unknown 10 8 2

Abbreviations: DTC, differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma; FTC, follicular thyroid

carcinoma; EBRT, external beam radiation therapy, and RAI, radioactive iodine. a ‘Enlarged glands of the neck’,

enlarged thymus, hemangioma, tinea, seizures, and tonsillitis. b In one patient, hyperthyroidism was caused by

the functioning malignant thyroid nodule.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

69

3

Supplemental Table 2. Additional Pathological Characteristics of Patients with Childhood DTC and

Distant Metastases

Characteristic All patients

n = 148

PTC

n = 144

FTC

n = 4

Tumor stage, n (%)T1a 3 (2.0) 3 (2.1) 0

T1b 18 (12.2) 18 (12.5) 0

T2 40 (27.0) 36 (25.0) 4 (100)

T3 6 (4.1) 6 (4.2) 0

T3a 34 (23.0) 34 (23.6) 0

T3b 16 (10.8) 16 (11.1) 0

T4a 16 (10.8) 16 (11.1) 0

T4b 0 0 0

Txa 15 (10.1) 15 (10.4) 0

Subtypes PTCNot otherwise specified - 41 (28.5) -

Conventional variant - 37 (25.7) -

Follicular variant - 27 (18.8) -

Conventional and other subtypeb - 19 (13.2) -

Diffuse sclerosing variant - 16 (11.1) -

Tall cell variant - 1 (0.7) -

Solid and follicular variant - 1 (0.7) -

Subtypes FTCEncapsulated angioinvasive - - 4 (100)c

Abbreviations: DTC, differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma; FTC, follicular thyroid

carcinoma. Patients were scored according to the 8th edition of the American Joint Committee on Cancer staging

system. Pathological staging was leading. If surgical and pathological data were incomplete, clinical data were used

to complete tumor and node staging. a Two patients with ectopic PTC were considered Tx. Subtypes of FTC were

classified according to the latest classification of the World Health Organization (minimally invasive FTC; encapsulated

angioinvasive FTC; or widely invasive FTC). b Cases that included conventional PTC mixed with another subtype. c For

one patient, the FTC insular subtype was favored.

CHAPTER 3

70

3

Supplemental Table 3. Patterns of Distant Metastases in Patients with Childhood DTC

Site of Distant Metastases All patients

n = 148

PTC

n = 144

FTC

n = 4

Maximal spread of DM, n (%)Lung only 131 (88.5) 129 (89.6) 2 (50.0)

Bone only 2 (1.4) 0 2 (50.0)

Lung and bone 7 (4.7) 7 (4.9) 0

Lung and brain 2 (1.4) 2 (1.4) 0

Lung, bone, and brain 2 (1.4) 2 (1.4) 0

Lung, bone, brain, and liver 3 (2.0) 3 (2.1) 0

Lung, bone, brain, adrenal, and kidney 1 (0.7) 1 (0.7) 0

Abbreviations: DTC, differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma; FTC, follicular thyroid

carcinoma; DM, distant metastases.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

71

3

Sup

ple

men

tal T

able

4. P

atie

nts

wit

h C

hild

hoo

d D

TC w

ith

Dia

gno

sis

of D

M >

10 y

ears

aft

er in

itia

l dia

gno

sis

(n=7

)

IDA

ge

at D

TC

Dia

gn

osi

s

(yrs

.)

Year

of D

TC

Dia

gn

osi

s

Sex

Init

ial S

urg

ical

Trea

tmen

t

RA

I tre

atm

ent

T s

tag

eN

sta

ge

Tim

e to

dia

gn

osi

s D

M

(yrs

.)

DM

max

Follo

w-u

p

(yrs

.)

Die

d

fro

m D

TC

146

15.4

1962

FT

T3;

473

.5T1

bN

1b13

lung

, liv

er, b

one

, bra

in52

.7Y

130

116

.120

00

FH

T+H

T1;

125.

0T2

N0

19b

one

19.2

N

107

13.4

1978

FT

T+U

2; U

TxN

1b26

lung

41.4

N

150

17.2

1946

FT

T+U

2; 2

84.4

TxN

x38

lung

66.1

N

125

18.9

1956

MT

T+U

2; U

TxN

x39

lung

, bo

ne, b

rain

39.2

Y

4912

.519

68F

TT+

U2;

187.

0TX

N1a

44lu

ng51

.7N

4417

.119

64F

U2;

UT4

a1b

52lu

ng52

.8N

All

had

str

uctu

ral

dis

ease

at

last

fo

llow

-up.

Abb

revi

atio

ns:

DTC

, d

iffer

enti

ated

thy

roid

car

cino

ma;

RA

I, ra

dio

acti

ve i

od

ine;

DM

, d

ista

nt m

etas

tase

s; A

TA,

Am

eric

an T

hyro

id

Ass

oci

atio

n; M

, m

ale;

F,

fem

ale;

TT,

to

tal t

hyro

idec

tom

y; C

ND

, ce

ntra

l nec

k d

isse

ctio

n; B

LND

, bi

late

ral l

ater

al n

eck

dis

sect

ion;

T s

tage

, tu

mo

r st

age;

N s

tage

, no

de

stag

e; L

ND

,

late

ral n

eck

dis

sect

ion;

HT,

hem

ithy

roid

ecto

my;

and

U, u

nkno

wn

thyr

oid

sur

gery

and

nec

k d

isse

ctio

n (I

D #

44)

or

unkn

ow

n ne

ck d

isse

ctio

n (I

Ds

#10

7, 1

50, 1

25, a

nd 4

9).

1 Fo

llicu

lar

thyr

oid

car

cino

ma.

CHAPTER 3

72

3

Supplemental Table 5. Identified Oncogenic Driver and Disease Characteristics of Patients with Childhood

PTC and Distant Metastases

RET fusion

n = 38

NTRK1 fusion

n = 11

NTRK3

fusion

n = 7

BRAF V600E

mutation

n = 8a

Sex, n (%)Female 23 (60.5) 6 (54.4) 4 (57.1) 7 (87.5)

Male 15 (39.5) 5 (45.5) 3 (42.9) 1 (12.5)

Age at diagnosis (years) 12.6 (9.9-15.0) 11.9 (8.1-16.1) 12.5 (7.9-14.3) 12.5 (10.9-13.5)

Range 6.8-18.9 6.2-17.5 4.0-15.8 7.6-15.7

Primary tumor size (cm) 4.1 (2.5-7.0)b 5.2 (4.6-6.9)c 4.5 (3.3-5.0) 2.9 (2.3-3.2)d

Lymph node status, n (%)N0 0 0 0 0

N1a 0 0 0 1 (12.5)

N1b 38 (100) 11 (100) 7 (100) 7 (87.5)

Time to diagnosis DM (months) 3.1 (0.8-14.4) 1.6 (0.4-4.4) 1.2 (0.7-7.7) 11.4 (0.8-32.9)

Last disease status, n (%)Structural disease 36 (97.3) 11 (100) 7 (100) 7 (100)

Biochemical disease 1 (2.7) 0 0 0

No evidence of disease 0 0 0 0

Unable to determine 1 0 0 1

Died of DTC, n (%)Yes 0 0 0 0

No 38 (100) 11 (100) 7 (100) 8 (100)

Follow-up time (years)e 8.4 (5.8-14.0) 5.9 (3.8-8.9) 9.8 (5.8-10.7) 11.5 (5.8-19.5)

Numbers shown as median (interquartile range). Abbreviations: PTC, papillary thyroid carcinoma; RET, rearranged

during transfection; NTRK, neurotrophic tyrosine kinase receptor; BRAF, v-Raf murine sarcoma viral oncogene

homolog B; DM, distant metastases. a In one patient, both a BRAF V600E and a v-akt murine thymoma viral oncogene

homolog 1 (AKT1) mutation were found. b 34 cases. c 10 cases. d 6 cases.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

73

3

Supplemental Figure 1. Overall survival (n=148) and disease-specific survival (n=145) of patients

diagnosed with distant metastases from childhood differentiated thyroid carcinoma (DTC). (A-C) Overall

survival, specified by (A) sex, (B) age at DTC diagnosis, and (C) age at diagnosis of distant metastases.

(D-F) Disease-specific survival, specified by (D) sex, (E) age at DTC diagnosis, and (F) age at diagnosis of

distant metastases. Categories: Sex; females and males. Age at diagnosis; <10 years, 10-15 years, and ≥15

years. Age at diagnosis of distant metastases; <10 years, 10-15 years, and ≥15 years. Three patients were

excluded from disease-specific survival because they had an unknown cause of death.

CHAPTER 3

74

3

Supplemental Table 6. Patients with Distant Metastases from Childhood PTC who Died from Disease (n=8)

ID Sex Year of

diagnosis

Age at PTC

Diagnosis

(yrs.)

Previous

disease

before

PTC

Initial Treatment # of RAI

administrations,

cumulative activity

[mCi]

T stage N stage Time to

diagnosis

DM (mos.)

Maximal DM spread Systemic

Therapy

EBRT Follow-up

diagnosis to

Death (years)

Age at

Death

(years)

91 M 2004 16.0 Ya TT 2, 227 T2 N1b 36.2 lung, liver, bone and brain Y Palliative 4.5 20.6

110 M 1993 15.9 N TT + CND + BLND 4, 754 T4a N1b 31.0 lung, liver, bone and brain Y Palliative 18.5 34.4

125 M 1956 18.8 N TT + U 3, U Tx Nx 470.0 lung, bone, and brain Y Initial 44.5 63.3

128 F 1960 9.6 Yb TT + CND 3, 350c T3b N1 133.0 lung and bone N Initial 35.3 45.0

145 F 1959 15.3 N TT + CND 5, 806 T3 N1b 15.0 lung and bone N Palliative 21.3 36.6

146 F 1962 15.4 Yd TT 3, 474 T1b N1b 152.2 lung, liver, bone and brain Y Palliative 52.8 68.2

152 F 1970 13.4 N TT + CND + LND 2, 250 T1a N1b 2.4 lung, bone, brain Y Palliative 30.7 44.1

155 F 1972 14.9 N TT + BLND 4, 718 T3b N1b 0.9 lung and bone N Palliative 30.6 45.5

Median 15.4 33.6 30.7 44.6

Abbreviations; PTC, papillary thyroid carcinoma; U, Unknown; Y, Yes; N, No; TT, total thyroidectomy; CND, central neck lymph

node dissection; LND, lateral neck lymph node dissection; EBRT, external beam radiation therapy. a Stage IV Neuroblastoma

treated with chemotherapy and whole body radiation prior to autologous stem cell transplant. b Hemangioma treated with

neck radiation. c Patient received two RAI treatments with a cumulative administered activity of 350 mCi and one dose with an

unknown activity. d Seizures treated with radiation therapy at an unknown dose. None of the patients with follicular thyroid

carcinoma died from their disease. IDs 110 and 146 were evaluated for mutations or fusions, but had a negative result after not

being comprehensively tested for fusions. All other patients who died from DTC were not tested for molecular alterations.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

75

3

Supplemental Table 6. Patients with Distant Metastases from Childhood PTC who Died from Disease (n=8)

ID Sex Year of

diagnosis

Age at PTC

Diagnosis

(yrs.)

Previous

disease

before

PTC

Initial Treatment # of RAI

administrations,

cumulative activity

[mCi]

T stage N stage Time to

diagnosis

DM (mos.)

Maximal DM spread Systemic

Therapy

EBRT Follow-up

diagnosis to

Death (years)

Age at

Death

(years)

91 M 2004 16.0 Ya TT 2, 227 T2 N1b 36.2 lung, liver, bone and brain Y Palliative 4.5 20.6

110 M 1993 15.9 N TT + CND + BLND 4, 754 T4a N1b 31.0 lung, liver, bone and brain Y Palliative 18.5 34.4

125 M 1956 18.8 N TT + U 3, U Tx Nx 470.0 lung, bone, and brain Y Initial 44.5 63.3

128 F 1960 9.6 Yb TT + CND 3, 350c T3b N1 133.0 lung and bone N Initial 35.3 45.0

145 F 1959 15.3 N TT + CND 5, 806 T3 N1b 15.0 lung and bone N Palliative 21.3 36.6

146 F 1962 15.4 Yd TT 3, 474 T1b N1b 152.2 lung, liver, bone and brain Y Palliative 52.8 68.2

152 F 1970 13.4 N TT + CND + LND 2, 250 T1a N1b 2.4 lung, bone, brain Y Palliative 30.7 44.1

155 F 1972 14.9 N TT + BLND 4, 718 T3b N1b 0.9 lung and bone N Palliative 30.6 45.5

Median 15.4 33.6 30.7 44.6

Abbreviations; PTC, papillary thyroid carcinoma; U, Unknown; Y, Yes; N, No; TT, total thyroidectomy; CND, central neck lymph

node dissection; LND, lateral neck lymph node dissection; EBRT, external beam radiation therapy. a Stage IV Neuroblastoma

treated with chemotherapy and whole body radiation prior to autologous stem cell transplant. b Hemangioma treated with

neck radiation. c Patient received two RAI treatments with a cumulative administered activity of 350 mCi and one dose with an

unknown activity. d Seizures treated with radiation therapy at an unknown dose. None of the patients with follicular thyroid

carcinoma died from their disease. IDs 110 and 146 were evaluated for mutations or fusions, but had a negative result after not

being comprehensively tested for fusions. All other patients who died from DTC were not tested for molecular alterations.

Supplemental Table 7. Cumulative Activity and Number of Radioactive Iodine Administrations per Disease

and Vital Status

Cumulative RAI activity (mCi) No. of RAI doses, n

Disease status at last clinical evaluationNo evidence of disease (n=9) 150.0 (75.0-182.1)1 1 (1-2)

Persistent disease (n=118) 238.0 (139.7-352.0) 2 2 (1-3)

Indeterminate (n=21) 253.4 (150.6-340.0) 2 (1-3)

Vital status3

Died of DTC (n=8)4 462.8 (244.2-739.9)5 3 (2-4)

Alive at last contact (n=132) 211.3 (137.7-309.9)6 2 (1-3)

Numbers are shown as median (interquartile range). 1 One of the 9 patients who had no evidence of disease had an

unknown administered activity of RAI. 2 Of the 117 patients with persistent disease, 2 patients did not receive RAI and

6 had an unknown cumulative activity of RAI (n=109). 3 Patients who died of other causes or unknown causes were

excluded from this analysis (n=8). All patients had PTC. 5 One of the 8 patients who died of DTC had an unknown

cumulative administered RAI activity. 6 Five patients alive at last contact had an unknown cumulative administered

activity of RAI and 2 did not receive RAI (n=125).

CHAPTER 3

76

3

Supplemental Table 8. Patients with Childhood PTC with Distant Metastases with

No Evidence of Disease at Last Clinical Evaluation (n=9)

ID Sex Year

of DTC

diagnosis

Age at DTC

diagnosis

(yrs.)

Previous

disease

before

DTC

Initial treatment # of RAI

administration;

cumulative activity

(mCi)

T stage N stage Time diagnosis

to diagnosis DM

(mos.)

Maximal DM spread Follow-up to last clinical

evaluation (yrs.)

Follow-up last known

vital status (yrs.)

67 M 2010 13.5 N TT + CND + BLND 1; 75 T1b N1b 2.6 Lung 5.2 9.2

37 M 2014 10.9 N TT + CND + LND 1; 64 T2 N1b 6.7 Lung 5.56 5.6

90 F 2008 16 N TT + CND + LND 1; 100 T3a N1b 2.1 Lung 7.8 11.6

58 M 2012 16.4 Ya TT + CND + LND 1; 150 T3 N1b 1.3 Lung 7.9 7.9

97 F 2007 2.8 N HT + HT 1; 29 T2 N1a 3.3 Lung 8.3 12.9

129 M 1989 15.5 N TT + LND 1; 156 T1b N1b 0.9 Lung 11.9 30.9

51 M 2007 15.9 N HT + HT 2; 182 T3a N1a 7.8 Lung 12.4 12.4

140 F 1986 9.5 N TT + LND 2; 257 T3a N1b 1.9 Lung 26.0 31.0

103 M 1970 17.8 N HT + HT 2; U  Tx N1b 0.0 Lung 49.6 49.8

Median 15.5         2.13   8.26 15.5

Abbreviations: PTC, papillary thyroid carcinoma; DTC, differentiated thyroid carcinoma; RAI, radioactive iodine; DM, distant

metastases; ATA, American thyroid association; M, male; F, female; TT, total thyroidectomy; CND, central neck dissection; BLND,

bilateral neck dissection; T stage, tumor stage; N stage, node stage; LND, lateral neck dissection; HT, hemithyroidectomy; and

U, unknown. a Hodgkin lymphoma diagnosed at age of one year, treated with chemotherapy. None of the patients received

systemic therapy or external radiation therapy. All patients with follicular thyroid carcinoma had persistent disease at last

known clinical evaluation.

DISTANT METASTASES FROM CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

77

3

Supplemental Table 8. Patients with Childhood PTC with Distant Metastases with

No Evidence of Disease at Last Clinical Evaluation (n=9)

ID Sex Year

of DTC

diagnosis

Age at DTC

diagnosis

(yrs.)

Previous

disease

before

DTC

Initial treatment # of RAI

administration;

cumulative activity

(mCi)

T stage N stage Time diagnosis

to diagnosis DM

(mos.)

Maximal DM spread Follow-up to last clinical

evaluation (yrs.)

Follow-up last known

vital status (yrs.)

67 M 2010 13.5 N TT + CND + BLND 1; 75 T1b N1b 2.6 Lung 5.2 9.2

37 M 2014 10.9 N TT + CND + LND 1; 64 T2 N1b 6.7 Lung 5.56 5.6

90 F 2008 16 N TT + CND + LND 1; 100 T3a N1b 2.1 Lung 7.8 11.6

58 M 2012 16.4 Ya TT + CND + LND 1; 150 T3 N1b 1.3 Lung 7.9 7.9

97 F 2007 2.8 N HT + HT 1; 29 T2 N1a 3.3 Lung 8.3 12.9

129 M 1989 15.5 N TT + LND 1; 156 T1b N1b 0.9 Lung 11.9 30.9

51 M 2007 15.9 N HT + HT 2; 182 T3a N1a 7.8 Lung 12.4 12.4

140 F 1986 9.5 N TT + LND 2; 257 T3a N1b 1.9 Lung 26.0 31.0

103 M 1970 17.8 N HT + HT 2; U  Tx N1b 0.0 Lung 49.6 49.8

Median 15.5         2.13   8.26 15.5

Abbreviations: PTC, papillary thyroid carcinoma; DTC, differentiated thyroid carcinoma; RAI, radioactive iodine; DM, distant

metastases; ATA, American thyroid association; M, male; F, female; TT, total thyroidectomy; CND, central neck dissection; BLND,

bilateral neck dissection; T stage, tumor stage; N stage, node stage; LND, lateral neck dissection; HT, hemithyroidectomy; and

U, unknown. a Hodgkin lymphoma diagnosed at age of one year, treated with chemotherapy. None of the patients received

systemic therapy or external radiation therapy. All patients with follicular thyroid carcinoma had persistent disease at last

known clinical evaluation.

Chapter 4

Chapter 4

CHAPTER 4

Long-term effects of radioiodine treatment on

female fertility in survivors of childhood differentiated

thyroid carcinoma

Marloes Nies, Astrid E.P. Cantineau*, Eus G.J.M. Arts*, Marleen H. van den Berg, Flora E. van Leeuwen, Anneke C. Muller Kobold, Mariëlle S. Klein Hesselink, Johannes G.M. Burgerhof, Adrienne H. Brouwers, Eveline W.C.M. van Dam, Bas Havekes, Marry M. van den Heuvel-Eibrink, Eleonora P.M. Corssmit, Leontien C.M. Kremer, Romana T. Netea-Maier, Helena J.H. van der Pal, Robin P. Peeters, John T.M. Plukker, Cécile M. Ronckers, Hanneke M. van Santen, Anouk N.A. van der Horst-Schrivers, Wim J.E. Tissing, Gianni Bocca, Eline van Dulmen-den Broeder**, and Thera P. Links**

* These authors contributed equally to this study.** These authors contributed equally to this study.

Thyroid. 2020;30(8):1169-1176.

CHAPTER 4

80

4

ABSTRACTBackgroundDifferentiated thyroid carcinoma (DTC) during childhood is a rare disease. Its excellent survival rate requires a focus on possible long-term adverse effects. This study aimed to evaluate fertility in female survivors of childhood DTC by assessing various reproductive characteristics combined with anti-Müllerian hormone (AMH) levels (a marker of ovarian reserve).

MethodsFemale survivors of childhood DTC, diagnosed at ≤18 years of age between 1970 and 2013, were included. Survivors were excluded when follow-up time was less than five years or if they developed other malignancies before or after diagnosis of DTC. Survivors filled out a questionnaire regarding reproductive characteristics (e.g. age at menarche and menopause, pregnancies, pregnancy outcomes, need for assisted reproductive therapy). Survivors aged <18 years during evaluation received an altered questionnaire without questions regarding pregnancy and pregnancy outcomes. These data were combined with information from medical records. AMH levels were measured in serum samples and were compared with AMH levels from 420 women not treated for cancer.

ResultsFifty-six survivors with a median age of 31.0 (interquartile range, IQR, 25.1–39.6) years were evaluated after a median follow-up of 15.4 (IQR 8.3–24.7) years. The median cumulative dose of 131I administered was 7.4 (IQR 3.7–13.0) GBq/200.0 (IQR 100.0–350.0) mCi. Twenty-five of the 55 survivors aged 18 years or older during evaluation reported 64 pregnancies, 45 of which resulted in live birth. Of these 55, 10.9% visited a fertility clinic. None of the survivors reported premature menopause. Age at AMH evaluation did not differ between DTC survivors and the comparison group (P = 0.268). Median AMH levels did not differ between DTC survivors and the comparison group [2.0 (IQR 1.0–3.7) µg/L vs. 1.6 (IQR 0.6–3.1) µg/L, respectively, P = 0.244]. The cumulative dose of 131I was not associated with AMH levels in DTC survivors (r

s = 0.210, P = 0.130).

ConclusionsFemale survivors of DTC who received 131I treatment during childhood do not appear to have major abnormalities in reproductive characteristics nor in predictors of ovarian failure.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

81

4

INTRODUCTIONChildhood differentiated thyroid carcinoma (DTC) is rare, with age-adjusted incidences

reported between 0.6 and 1.2 per 100 000 per year (1,2). Up to puberty, the female to

male ratio is similar, but after puberty, mainly females are diagnosed with the disease

(3). In all age groups, a rise in incidence rates of thyroid cancer has been reported (4).

Treatment of pediatric DTC most commonly consists of total thyroidectomy with or

without central or lateral neck dissection (5). After surgery, 131I is often administered.

Depending on the risk classification of the patient, a certain intensity of thyrotropin

(TSH) suppression is pursued. Although this treatment results in excellent survival

rates, up to 99% after 30 years of follow-up (6), DTC treatment, especially therapy

with 131I, calls for examination of possible long-term adverse effects: reproductive

characteristics, secondary cancers, salivary dysfunction, bone marrow suppression,

and alterations in quality of life (7–12).

Female fertility after treatment with 131I has been evaluated in survivors of childhood

and adult DTC. Only two studies examined survivors of childhood DTC and were limited

by small numbers of patients and unclear or ill-defined endpoints (9,13). Studies in

survivors of adult DTC found conflicting results regarding the effect of 131I on female

fertility, although permanent impairment of fertility is not common (14–21).

Anti-Müllerian hormone (AMH) is released by the granulosa cells and is a reflection

of the number of antral follicles in the ovaries. Although there is no consensus on

the clinical value of AMH, it is a commonly used marker for ovarian reserve in cancer

survivors (22), partly because AMH is not influenced by menstrual cycle fluctuations

(23,24). AMH levels in adult DTC patients decrease after treatment with 131I (25–27),

although it is unclear whether this decrease is transient or permanent or even clinically

relevant (21,27). AMH levels show a greater decrease after 131I in women aged 35 years

or older during treatment (26).

There is a need for well-defined and systematically performed studies regarding

effects on long-term fertility in female survivors of childhood DTC. Therefore,

the primary aim of the current study was to assess the reproductive characteristics

(pregnancies, number of live births, pregnancy outcomes, and health of offspring) in

female survivors of childhood DTC treated with 131I. The secondary aim was to compare

AMH levels (as a measure of ovarian reserve) in female survivors of childhood DTC

with a group consisting of women who had not been treated for cancer.

MATERIALS AND METHODSThis research is part of a nationwide, long-term follow-up study on childhood DTC

in the Netherlands, previously described in detail (28). The institutional review

board of the University Medical Center Groningen approved the study on behalf of

all participating institutions (ABR NL40572.042.12, file number 2012/183). This study

CHAPTER 4

82

4

has been registered in the Netherlands Trial Registry (trial NL3280). Written informed consent was obtained from all subjects before participation in the study.

ParticipantsDTC survivorsIncluded were female patients diagnosed with DTC between 1970 and 2013 at age ≤18 years and treated in the Netherlands. Treatment most commonly consisted of total thyroidectomy, 131I, and TSH suppression therapy (28). Specific exclusion criteria in this study were as follows: less than five years since diagnosis, diagnosis of other malignancy before or after the DTC diagnosis, thyroid hormone withdrawal or recombinant human TSH administration within three months before evaluation, not being able to complete a Dutch questionnaire, and not being treated with 131I for DTC. Patients were evaluated from February 2013 until November 2014.

Fertility assessmentFertility was assessed by means of a self-administered questionnaire, information from medical records, and a hormonal evaluation.

QuestionnaireSurvivors were asked to complete a questionnaire regarding their use of current medication (thyroid hormone, contraceptives, or other medications), smoking, and reproductive characteristics: obstetric and gynecological medical history (menarche, menstrual cycles, age at first pregnancy, children conceived, birth defects and major health problems, and visiting a fertility clinic due to problems with conceiving). Survivors aged <18 years during evaluation received an altered questionnaire without questions regarding pregnancy and pregnancy outcomes.

Medical dataMedical records were accessed to obtain information regarding survivors’ characteristics: thyroid carcinoma histology, tumor node metastasis (TNM) classification (redefined to the seventh edition of the TNM since the seventh edition was current during initial evaluation), treatment modalities (type of surgery and details of 131I administrations), and survivors’ outcomes (remission, recurrence, or persistent disease, defined as previously described) (28). Comorbidities interacting with fertility (e.g. endometriosis or gynecological surgery) were also documented.

Clinical evaluationSurvivors were evaluated during a visit to an outpatient clinic, in the context of the study of long-term treatment effects. Height and weight were measured by one of the researchers (M.S.K.H.). Fasting blood samples were drawn by venipuncture. Blood

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

83

4

samples were subsequently stored in a −80°C environment until processed. Blood sampling was performed at a random time during the menstrual cycle for logistical reasons. Luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estradiol (E2) measurements were performed in survivors who did not use contraceptives containing hormones.

AMH, LH, FSH, and E2 analyses of DTC survivors were centrally performed in one run in the laboratory of the University Medical Center Groningen, the Netherlands, by electrochemiluminescence immunoassay on a Roche Cobas analysis platform. Limit of detection (LoD) and intra-assay variation of these assays were 0.010 µg/L and <1.3% for AMH, 0.3 IU/L and <1% for higher ranges and 2.2% for values below 1.0 IU/L for LH, 0.100 IU/L and <2.5% for FSH, and for E2 an LoD of 0.018 nmol/L and an intra-assay coefficient of variability (CV) of 1.1–1.6% over the measuring range, whereas values below 0.07 nmol/L had a CV of 2.4–6.7%. Reference norms per age group (in years) for AMH (in µg/L, 2.5th to 97.5th percentile) were: 15–18.9: 0.34–10.39; 20–24: 1.22–11.7; 25–29: 0.89–9.85; 30–34: 0.58–8.13; 35–39: 0.15–7.49; 40–44: 0.03–5.47; 45–50: 0.01–2.71 (29,30). Smoking and body mass index may influence AMH levels and were therefore also evaluated (31–34).

Comparison group for AMH levelsThe comparison group consisted of sisters of childhood cancer survivors (n = 196) and women from the general population (n = 224) who participated in a previous nationwide cohort study among Dutch female five-year survivors of childhood cancer aiming to evaluate the effects of childhood cancer treatment on fertility (the DCOG-LATER VEVO-study) (35,36). Participants of the comparison group were aged ≥18 years and had not been treated for cancer.

AMH analyses of the comparison group were performed in one run using an ultra-sensitive Elecsys AMH assay (Roche Diagnostics GmbH, Mannheim, Germany) in the laboratory of the VU Medical Center Amsterdam, the Netherlands. The LoD of this assay was 0.01 µg/L, the intra-assay CV of this assay was 0.5–1.8%, and the limit of quantitation of 0.03 µg/L. Reference norms per age group (in years) for AMH (in µg/L, 2.5th–97.5th percentile) were: 15–25: 0.26–11; 25–30: 0.49–14; 30–35: 0.14–13; 35–40: <11; 40–45: <6; 45 and older: <0.48. There was a good agreement between the two AMH assays. The Passing–Bablok regression intercept did not differ significantly from 0 (−0.003, [95% confidence interval −0.075 to 0.021]) and slope 1.092 [95% confidence interval 1.049 to 1.143].

Study definitionsEvaluation date was the date of blood sampling or, in case of lacking blood sample, the date of filling in the questionnaire. Follow-up time was defined as the period between the date of diagnosis and the date of evaluation. Dosages of 131I of 0.9 GBq

CHAPTER 4

84

4

(25 mCi) or higher were considered as therapeutic doses. Women were considered postmenopausal if they reported 12 months of amenorrhea without any other obvious pathological or physiological cause (22). Premature ovarian insufficiency was defined as start of menopause before the age of 40 years (22).

Statistical analysesDescriptive statistics regarding disease, treatment, reproductive characteristics, and AMH levels are presented as median (interquartile range [IQR]), unless otherwise specified. Cutoff scores for “low AMH levels” were calculated, based on the 10th (0.22 µg/L) and 25th percentile (0.64 µg/L) of AMH levels of the complete comparison group. Categorical variables were compared using chi-square test or Fisher’s exact test (if >20% of the cells had an expected count of <5). Mann–Whitney U test was performed for nonnormally distributed continuous or ordinal variables. When variables were normally distributed, an independent sample t-test was performed. To correlate two nonnormally distributed continuous and/or ordinal variables, Spearman’s rank correlation coefficient (r

s) was used. Simple linear regression

analysis was performed to evaluate associations between attained age (in years) and cumulative 131I dose (in GBq) as predictors and AMH as outcome measure. In the first multiple linear regression analysis, log-transformed AMH was predicted by attained age in years and group (coded as 0 = comparison group, 1 = DTC survivors). A second multiple linear regression analysis predicted log-transformed AMH by independent variables: attained age (years) and cumulative dose of 131I (in GBq). A P value of <0.05 was considered statistically significant. All tests were performed two-sided. IBM SPSS Statistics version 23.0.0.3 for Windows (IBM, Armonk, NY) was used for statistical analyses.

RESULTSParticipantsSixty-two of the 105 survivors of the nationwide follow-up study were eligible for this substudy. Four survivors declined participation, and two were late for inclusion. Thus, 56 of the 62 (90.3%) female survivors were included (Supplemental Figure 1). Two of the 56 subjects only completed the questionnaire, declining participation in the clinical evaluation. Table 1 shows clinical and treatment characteristics of the included survivors. The median age of survivors at evaluation was 31.0 (IQR 25.1–39.6) years after a median follow-up period of 15.4 (IQR 8.3–24.7) years. The median cumulative activity of 131I administered was 7.4 GBq/200.0 mCi (IQR 3.7–13.0 GBq/IQR 100.0–350.0 mCi, respectively). Half of the survivors received multiple administrations of 131I.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

85

4

Reproductive characteristicsFifty-six DTC survivors reported their reproductive characteristics in the administered questionnaire (Table 2). Four (7.1%) women reported being postmenopausal. Ages at menopause were 45, 51, and 52 years, with one age at menopause missing. Of the 55 survivors aged ≥18 years during evaluation, 25 (45.5%) reported one or more pregnancies. The median age at first pregnancy was 25.5 (IQR 22.5–30.0) years. Sixty-four pregnancies were reported (2.6 pregnancies per survivor who reported to ever having been pregnant), of which 1 was a twin pregnancy. Subsequently, 45 live births were reported. Other pregnancy outcomes were miscarriage (n = 13), induced abortion (n = 3), unknown outcome (n = 3), and pregnant at evaluation (n = 1). Six (10.9%) survivors had visited a fertility doctor or clinic because of problems with conceiving. Birth defects and major health problems of children reported by the survivors are shown in Supplementary Table 1.

Table 1. Characteristics of survivors of childhood differentiated thyroid carcinoma

Characteristic n = 56

Age at evaluation, years 31.0 (25.1-39.6)

Age at diagnosis, years 16.0 (13.7-17.5)

Follow-up duration, years 15.4 (8.3-24.7)

Histology, n (%)Papillary 47 (83.9)

Follicular 9 (16.1)

Tumor node metastasis stage, n (%)T

T1-T2 37 (66.1)

T3-T4 11 (19.6)

Tx 8 (14.3)

NN0 27 (48.2)

N1 25 (44.6)

Nx 4 (7.1)

MM0 45 (80.4)

M1 6 (10.7)

Mx 5 (8.9)

Cumulative 131I activity, GBqa 7.4 (3.7-13.0)

Cumulative 131I activity, mCia 200.0 (100.0-350.0)

Multiple 131I administrations, n (%) 28 (50.0)

Numbers shown as median (interquartile range). a the dose of administered was 131I unknown in one survivor, therefore

n=55. Abbreviation: 131I, radioactive iodine.

CHAPTER 4

86

4

Hormonal evaluationCharacteristics and AMH levels of the female survivors of childhood DTC and the comparison group are shown in Table 3. DTC survivors who provided blood samples had a median age of 29.4 years (n = 54, IQR 24.8–38.3 years) upon evaluation. The median age of the comparison group was 33.1 (IQR 26.8–39.3) years. There were no statistically significant differences between the two groups for nationality (predominantly Dutch, P = 1.000, data not shown), age upon evaluation, smoking, and body mass index. The median AMH levels did not differ between DTC survivors and the comparison group (2.0 µg/L vs. 1.6 µg/L, respectively, P = 0.244).

The cumulative dosage of 131I did not correlate with AMH levels (rs = 0.210, P = 0.130).

In the DTC group, age was negatively correlated with AMH levels (rs = −0.480, P< 0.001).

Eight (14.8%) and 10 (18.5%) of the DTC survivors had an AMH level below the cutoff value based on the 10th and 25th percentiles of the comparison group, respectively. The number of DTC survivors with “low AMH levels” did not significantly differ from those in the comparison group (P = 0.278 and P = 0.296, respectively) (Supplementary Table 2).

Because the data of AMH were positively skewed, the values were log transformed. Subsequently, all assumptions for linear regression analysis were met. Log-transformed AMH levels did not differ between DTC survivors and the comparison group (median ln(AMH) 0.7 vs. 0.5, respectively, P = 0.696; Table 3). Results of the simple and multiple

Table 2. Reproductive characteristics of survivors of childhood differentiated thyroid carcinoma

Characteristic n = 56

Age at menarche, yearsa 13.0 (12.0-13.0)

Postmenopausal, n (%) 4 (7.1)

Use of contraceptives, n (%)Hormonal contraceptives 24 (42.9)

Non-hormonal contraceptivesb 1 (1.8)

No contraceptives 31 (55.4)

Visited doctor for subfertility (yes), n (%)c 6 (10.9)

Ever been pregnant (yes), n (%)c 25 (45.5)

Age at first pregnancy, yearsc,d 25.5 (22.5-30.0)

Number of pregnancies, nc 64e

Live births, n 45

Women reporting miscarriage, n 8f

Induced abortion, n 3

Pregnant during evaluation, n 1

Unknown pregnancy outcome, n 3

Numbers shown as median (interquartile range). a n=55 because one missing value. b copper intrauterine device.c not applicable in one participant because age <18y during evaluation, n=55. d n=22 because age first pregnancy

missing for 3 participants. e 1 twin pregnancy. f 7 women reported 1 miscarriage, 1 woman reported 6 miscarriages.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

87

4Table 3. Anti-Müllerian hormone levels in survivors of childhood differentiated thyroid carcinoma compared

to the comparison group

Characteristic DTC survivors

n = 54a

Comparison group

n = 420

P Value

Age at evaluation, years 29.4 (24.8–38.3) 33.1 (26.8–39.3) 0.268b

Smoking, n (%) 0.392c

Current 7 (13.0) 78 (18.6)

Ever 13 (24.1) 123 (28.8)

Never 33 (61.1) 221 (52.6)

Missing 1 (1.9) 0 (0)

Body Mass Index, kg/m2 23.8 (21.2–26.8)d 23.0 (21.2–25.9) 0.428b

Type of control, n (%) n.a.

General population - 224 (53.3)

Sister - 196 (46.7)

AMH level, µg/L 2.0 (1.0–3.7) 1.6 (0.6–3.1) 0.244b

ln(AMH) 0.7 (0.0–1.3) 0.5 (-0.4–1.1) 0.696e

Numbers shown as median (interquartile range). a two participants participated only in questionnaire part of study. b Mann-Whitney U test; c Pearson Chi-Square Test (Asymptotic Significance). d Length and weight self-reported by 2

participants. e Independent samples t-test. Abbreviations: AMH, anti-Müllerian hormone; DTC, differentiated thyroid

carcinoma; and n.a., not applicable.

Table 4. Simple and multiple linear regression analyses for log transformed anti-Müllerian hormone in 54

survivors of childhood differentiated thyroid carcinoma

Variable Intercept βa 95% CI R2 P Value

Simple linear regressionAge at evaluation, years 4.12 -0.12 [-0.13; -0.11] 0.418 <0.001

Cumulative 131I activity, GBq -0.28 0.06 [-0.01; 0.12] 0.056 0.089

Multiple linear regression1. Age at evaluation, years

Groupb

4.13 -0.12

-0.06

[-0.13; -0.11]

[-0.40; 0.29]

0.418 <0.001

2. Age at evaluation, yearsc

Cumulative 131I activity, GBq

3.73 -0.12

0.02

[-0.16; -0.07]

[-0.04; 0.07]

0.414 <0.001

Abbreviations: CI, 95% confidence interval and 131I, radioactive iodine. a Unstandardized coefficients β. b comparison group = 0, DTC survivors = 1. c ‘Group’ removed from this analysis, because comparison group did not

receive 131I administrations.

linear regression analyses for log-transformed AMH are shown in Table 4. Simple linear regression showed that age was a significant predictor of log-transformed AMH, but cumulative dose of 131I was not. The first multiple linear regression analysis showed that age was a significant predictor of log-transformed AMH, but group (i.e. survivor vs.

CHAPTER 4

88

4

comparison group) was not. In model 2, when log-transformed AMH levels in only DTC survivors were predicted by age and cumulative dose of 131I, age remained a significant predictor of log-transformed AMH, but cumulative dose of 131I was not.

There was no difference in AMH levels between DTC survivors who did or did not use contraceptives containing hormones, or between survivors who had received single or multiple doses of 131I (data shown in Supplementary Table 3). LH, FSH, and E2 levels of DTC survivors who did not use contraceptives or used nonhormonal contraceptives, obtained at a random time during the menstrual cycle, were within the reference range (Supplementary Table 4).

DISCUSSIONThe current study, focusing on various aspects of female fertility after treatment with 131I for childhood DTC, shows no major abnormalities in reproductive characteristics and no difference in AMH levels between long-term DTC survivors and a comparison group after a median follow-up period of 15 years.

In this unique series of patients, the number of live births per pregnancy in the current study is comparable to those in the normal population: 70% of pregnancies in the DTC survivors resulted in a live birth, which corresponds with the 71% in an earlier prospective register-based study (37). The 10% of female DTC survivors who visited a fertility clinic or doctor because of problems with conception corresponds with that of other couples in the Netherlands who are trying to become pregnant, in whom this percentage is around 15% (38,39).

Comparing current results with findings of previous studies among survivors of childhood DTC is complicated by the fact that the earlier studies lack concrete definitions, report on only a small number of patients, or evaluated only a selection of reproductive characteristics (9,13). The cumulative dose of 131I administered to the current survivors is similar to the dose administered in the study of Sarkar et al. (13). Overall, no clear impairments of fertility have been observed in the current or previous studies in female survivors of childhood DTC (9,13).

It is unclear whether adverse effects of 131I have similar consequences in children and adults. Quantitatively, damage to the ovaries caused by 131I could be relatively less severe in younger women, since girls and adolescent women still have a greater number of primary oocytes/primordial follicles than adult women (40). As oocytes decrease in quality with increasing age (41), a higher quality of primary oocytes/primordial follicles in pre-adult women may also be beneficial. Studies in women aged >35 years who were treated with 131I for DTC observed a more pronounced negative effect on AMH levels (26) and birth rates (14). Negative effects on fertility in women treated with chemotherapy for other types of cancer also increase with age at treatment (42).

In this group of childhood DTC survivors, evaluation of AMH levels is a measure for ovarian reserve, and this hormone is not significantly affected by menstrual cycle

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

89

4

variations (23,24). However, as AMH levels are strongly affected by age, we adjusted our analyses accordingly.

The mere determination of AMH levels as outcome measure in the assessment of female fertility provides an incomplete representation. AMH levels have been shown to be decreased up to one year after treatment with 131I for DTC in adults (25,26). This reflects damage to the secondary and early antral follicles of the ovary since AMH expression is highest during these follicular stages (Supplementary Table 5) (43,44). Primordial follicles are probably less prone to the effects of 131I treatment, and these unharmed primordial follicles can subsequently develop into secondary and early antral follicles after therapy, resulting in normal AMH expression over the long term. A slight rise in AMH levels in survivors of adult DTC one year after 131I treatment was seen in only one study (25); this was not confirmed in another study (26). Seven years after treatment at adult ages, AMH levels did not differ between 131I-treated females and their controls (21). In the current study, 15 years after treatment during childhood, AMH levels were similar to those of the comparison group. Moreover, studies have also reported recovery of AMH levels after other anticancer treatments (45–47). Evaluation of AMH levels soon after treatment may indicate some form of ovarian damage, but long-term evaluation of AMH levels, combined with reproductive characteristics, is more appropriate in providing information on possible irrecoverable damage to the ovary and subsequent reproductive health.

Unfortunately, we did not evaluate the effects of TSH suppression therapy on fertility in the current survivors (28), although effects of subclinical hyperthyroidism on fertility have not been proven (48,49), other than the well-known effects of overt hyperthyroidism causing, for instance, menstrual disturbances or amenorrhea (50).

Strengths of the present study include the cohort size, given the rarity of DTC in childhood, and the availability of an appropriate comparison group for AMH levels. Minor limitations deserve consideration as well. The reported reproductive characteristics may be subject to change since many of the evaluated survivors in this study were of reproductive age, but may not have conceived yet owing to other factors. The chosen reproductive characteristics were well defined, based on current knowledge. Thereby, we evaluated a broad range of reproductive characteristics that determine fertility, including objective outcome measures, such as live births and hormonal evaluation.

Although no major impact on fertility after 131I treatment for childhood DTC was observed, this does not necessarily imply that 131I can be administered without restriction in young female patients. Sparse data show that 131I therapy seems to have no adverse effects on the risk of congenital abnormalities in offspring of DTC survivors (51). Other adverse effects of 131I (i.e., salivary gland dysfunction, bone marrow suppression) do increase with multiple or higher doses (11,12). This study, in accordance with previous studies, did not find a dose–response relationship between cumulative administrated

CHAPTER 4

90

4

131I and AMH levels (21,25,26). Follow-up beyond menopause of the survivors in this cross-sectional study will shed light on the full reproductive period. The current study can serve as a basis for this evaluation.

To conclude, the current study found no abnormalities in fertility in long-term female survivors of childhood DTC. Our conclusions are based on evaluation of a broad range of reproductive characteristics: fertility outcomes, parameters of reproductive health, indications of impaired fertility, and AMH as a marker of ovarian reserve. Altogether, these results regarding long-term reproductive outcomes seem to be reassuring for females receiving 131I for childhood DTC.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

91

4

ACKNOWLEDGMENTSThe authors are grateful to their colleagues in the Netherlands for referring patients for this study. We would like to thank Dr. Annemieke C. Heijboer for her laboratory support.

FUNDING This work was supported by the Stichting Kinderen Kankervrij (Foundation Children Cancer-free, The Netherlands, project no. 81). C.M.R. is supported by the Dutch Cancer Society.

AUTHOR DISCLOSURE STATEMENTNo competing financial interests exist.

CHAPTER 4

92

4

REFERENCES1. Dermody S, Walls A, Harley EH, Jr. Pediatric thyroid cancer: an update from the SEER database 2007-2012. Int J

Pediatr Otorhinolaryngol. 2016;89:121-126.

2. Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I, Parkin DM. Thyroid cancer incidence and survival

among European children and adolescents (1978-1997): report from the Automated Childhood Cancer Information

System project. Eur J Cancer. 2006;42(13):2150-2169.

3. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167-172.

4. Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM. Trends in Thyroid Cancer Incidence and Mortality in the United

States, 1974-2013. JAMA. 2017;317(13):1338-1348.

5. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management guidelines for children with thyroid nodules and differentiated thyroid cancer.

Thyroid. 2015;25(7):716-759.

6. Hay ID, Johnson TR, Kaggal S, Reinalda MS, Iniguez-Ariza NM, Grant CS, Pittock ST, Thompson GB. Papillary

thyroid carcinoma (PTC) in children and adults: comparison of initial presentation and long-term postoperative

outcome in 4432 patients consecutively treated at the Mayo Clinic during eight decades (1936-2015). World J

Surg. 2018;42(2):329-342.

7. Fard-Esfahani A, Emami-Ardekani A, Fallahi B, Fard-Esfahani P, Beiki D, Hassanzadeh-Rad A, Eftekhari

M. Adverse effects of radioactive iodine-131 treatment for differentiated thyroid carcinoma. Nucl Med

Commun. 2014;35(8):808-817.

8. Klein Hesselink EN, Links TP. Radioiodine treatment and thyroid hormone suppression therapy for differentiated

thyroid carcinoma: adverse effects support the trend toward less aggressive treatment for low-risk patients. Eur

Thyroid J. 2015;4(2):82-92.

9. Albano D, Bertagna F, Panarotto MB, Giubbini R. Early and late adverse effects of radioiodine for pediatric

differentiated thyroid cancer. Pediatr Blood Cancer. 2017;64(11):e26595.

10. Nies M, Klein Hesselink MS, Huizinga GA, Sulkers E, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den

Heuvel-Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Plukker JT, Ronckers CM,

van Santen HM, Tissing WJ, Links TP, Bocca G. Long-term quality of life in adult survivors of pediatric differentiated

thyroid carcinoma. J Clin Endocrinol Metab. 2016;102(4):1218–1226.

11. Prinsen HT, Klein Hesselink EN, Brouwers AH, Plukker JT, Sluiter WJ, van der Horst-Schrivers AN, van Imhoff

GW, Links TP. Bone marrow function after (131)I therapy in patients with differentiated thyroid carcinoma. J Clin

Endocrinol Metab. 2015;100(10):3911-3917.

12. Selvakumar T, Nies M, Klein Hesselink MS, Brouwers AH, van der Horst-Schrivers ANA, Klein Hesselink EN, Tissing

WJE, Vissink A, Links TP, Dutch Pediatric Thyroid Cancer study consortium. Long-term effects of radioiodine

treatment on salivary gland function in adult survivors of pediatric differentiated thyroid carcinoma. J Nucl

Med. 2018;60(2):172-177.

13. Sarkar SD, Beierwaltes WH, Gill SP, Cowley BJ. Subsequent fertility and birth histories of children and adolescents

treated with 131I for thyroid cancer. J Nucl Med. 1976;17(6):460-464.

14. Wu JX, Young S, Ro K, Li N, Leung AM, Chiu HK, Harari A, Yeh MW. Reproductive outcomes and nononcologic

complications after radioactive iodine ablation for well-differentiated thyroid cancer. Thyroid. 2015;25(1):133-138.

15. Anderson C, Engel SM, Weaver MA, Zevallos JP, Nichols HB. Birth rates after radioactive iodine treatment for

differentiated thyroid cancer. Int J Cancer. 2017;141(11):2291-2295.

16. Ko KY, Yen RF, Lin CL, Cheng MF, Huang WS, Kao CH. Pregnancy outcome after I-131 therapy for patients with

thyroid cancer: a nationwide population-based cohort study. Medicine. 2016;95(5):e2685.

17. Garsi JP, Schlumberger M, Rubino C, Ricard M, Labbe M, Ceccarelli C, Schvartz C, Henri-Amar M, Bardet S, de

Vathaire F. Therapeutic administration of 131I for differentiated thyroid cancer: radiation dose to ovaries and

outcome of pregnancies. J Nucl Med. 2008;49(5):845-852.

18. Schlumberger M, De Vathaire F, Ceccarelli C, Delisle MJ, Francese C, Couette JE, Pinchera A, Parmentier C.

Exposure to radioactive iodine-131 for scintigraphy or therapy does not preclude pregnancy in thyroid cancer

patients. J Nucl Med. 1996;37(4):606-612.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

93

4

19. Sioka C, Fotopoulos A. Effects of I-131 therapy on gonads and pregnancy outcome in patients with thyroid cancer.

Fertil Steril. 2011;95(5):1552-1559.

20. Sawka AM, Lakra DC, Lea J, Alshehri B, Tsang RW, Brierley JD, Straus S, Thabane L, Gafni A, Ezzat S, George SR,

Goldstein DP. A systematic review examining the effects of therapeutic radioactive iodine on ovarian function and

future pregnancy in female thyroid cancer survivors. Clin Endocrinol (Oxf). 2008;69(3):479-490.

21. Giusti M, Mittica M, Comite P, Campana C, Gay S, Mussap M. Anti-Müllerian hormone in pre-menopausal females

after ablative radioiodine treatment for differentiated thyroid cancer. Endocrine. 2018;60(3):516-523.

22. Research on the menopause in the 1990s. Report of a WHO Scientific Group. World Health Organ Tech Rep

Ser. 1996;866:1-107.

23. La Marca A, Stabile G, Artenisio AC, Volpe A. Serum anti-Mullerian hormone throughout the human menstrual

cycle. Hum Reprod. 2006;21(12):3103-3107.

24. Lambert-Messerlian G, Plante B, Eklund EE, Raker C, Moore RG. Levels of antimullerian hormone in serum during

the normal menstrual cycle. Fertil Steril. 2016;105(1):208-213.e201.

25. Evranos B, Faki S, Polat SB, Bestepe N, Ersoy R, Cakir B. Effects of radioactive iodine therapy on ovarian reserve:

a prospective pilot study. Thyroid. 2018;28(12):1702-1707.

26. Yaish I, Azem F, Gutfeld O, Silman Z, Serebro M, Sharon O, Shefer G, Limor R, Stern N, Tordjman KM. A single

radioactive iodine treatment has a deleterious effect on ovarian reserve in women with thyroid cancer: results of

a prospective pilot study. Thyroid. 2018;28(4):522-527.

27. Acibucu F, Acibucu DO, Akkar OB, Dokmetas HS. Evaluation of ovarian reserve with amh level in patients

with well-differentiated thyroid cancer receiving radioactive iodine ablation treatment. Exp Clin Endocrinol

Diabetes. 2016;124(10):593-596.

28. Klein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-

Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Schmid KW, Smit JW, Williams

GR, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP. Pediatric differentiated thyroid carcinoma in

the Netherlands: a nationwide follow-up study. J Clin Endocrinol Metab. 2016;101(5):2031-2039.

29. Anckaert E, Oktem M, Thies A, Cohen-Bacrie M, Daan NM, Schiettecatte J, Muller C, Topcu D, Groning A, Ternaux

F, Engel C, Engelmann S, Milczynski C. Multicenter analytical performance evaluation of a fully automated anti-

Mullerian hormone assay and reference interval determination. Clin Biochem. 2016;49(3):260-267.

30. Jopling H, Yates A, Burgoyne N, Hayden K, Chaloner C, Tetlow L. Paediatric Anti-Müllerian hormone measurement:

male and female reference intervals established using the automated Beckman Coulter Access AMH assay.

Endocrinol Diabetes Metab. 2018;1(4):e00021-e00021.

31. Pasternak MC, Christos P, Spandorfer SD. The relationship between body mass index and anti-mullerian hormone

levels in reproductive-age women; is there a negative correlation? Fertil Steril. 2018;109(3):e42-e43.

32. Barriere P, Freour T, Masson D, Mirallie S, Jean M. Normal anti-mullerian hormone (AMH) levels in young

smoking women undergoing IVF have no predictive value for ovarian response, inversely to non smokers. Fertil

Steril. 2007;88:S172.

33. White AJ, Sandler DP, D’Aloisio AA, Stanczyk F, Whitworth KW, Baird DD, Nichols HB. Antimüllerian hormone in

relation to tobacco and marijuana use and sources of indoor heating/cooking. Fertil Steril. 2016;106(3):723-730.

34. Simoes-Pereira J, Nunes J, Aguiar A, Sousa S, Rodrigues C, Sampaio Matias J, Calhaz-Jorge C. Influence of

body mass index in anti-Mullerian hormone levels in 951 non-polycystic ovarian syndrome women followed at

a reproductive medicine unit. Endocrine. 2018;61(1):144-148.

35. van den Berg MH, Overbeek A, Lambalk CB, Kaspers GJL, Bresters D, van den Heuvel-Eibrink MM, Kremer LC,

Loonen JJ, van der Pal HJ, Ronckers CM, Tissing WJE, Versluys AB, van der Heiden-van der Loo M, Heijboer AC,

Hauptmann M, Twisk JWR, Laven JSE, Beerendonk CCM, van Leeuwen FE, van Dulmen-den Broeder E, DCOG

LATER-VEVO study group. Long-term effects of childhood cancer treatment on hormonal and ultrasound markers

of ovarian reserve. Hum Rep 2018;33(8):1474-1488.

36. Overbeek A, van den Berg MH, Kremer LC, van den Heuvel-Eibrink MM, Tissing WJ, Loonen JJ, Versluys B, Bresters

D, Kaspers GJ, Lambalk CB, van Leeuwen FE, van Dulmen-den Broeder E. A nationwide study on reproductive

function, ovarian reserve, and risk of premature menopause in female survivors of childhood cancer: design and

methodological challenges. BMC Cancer. 2012;12:363.

37. Magnus MC, Wilcox AJ, Morken N-H, Weinberg CR, Håberg SE. Role of maternal age and pregnancy history in risk

of miscarriage: prospective register based study. BMJ. 2019;364:l869-l869.

CHAPTER 4

94

4

38. Gnoth C, Godehardt E, Frank-Herrmann P, Friol K, Tigges J, Freundl G. Definition and prevalence of subfertility and

infertility. Hum Reprod. 2005;20(5):1144-1147.

39. Beurskens MP, Maas JW, Evers JL. Subfertility in South Limburg: calculation of incidence and appeal for specialist

care. Ned Tijdschr Geneeskd. 1995;139(5):235-238.

40. Scheffer GJ, Broekmans FJ, Looman CW, Blankenstein M, Fauser BC, de Jong FH, te Velde ER. The number

of antral follicles in normal women with proven fertility is the best reflection of reproductive age. Hum

Reprod. 2003;18(4):700-706.

41. Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr

Rev. 2009;30(5):465-493.

42. Lee SJ, Schover LR, Partridge AH, Patrizio P, Wallace WH, Hagerty K, Beck LN, Brennan LV, Oktay K. American Society

of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol. 2006;24(18):2917-2931.

43. Weenen C, Laven JS, Von Bergh AR, Cranfield M, Groome NP, Visser JA, Kramer P, Fauser BC, Themmen AP. Anti-

Mullerian hormone expression pattern in the human ovary: potential implications for initial and cyclic follicle

recruitment. Mol Hum Reprod. 2004;10(2):77-83.

44. Andersen CY, Schmidt KT, Kristensen SG, Rosendahl M, Byskov AG, Ernst E. Concentrations of AMH and inhibin-B

in relation to follicular diameter in normal human small antral follicles. Hum Reprod. 2010;25(5):1282-1287.

45. Dillon KE, Sammel MD, Prewitt M, Ginsberg JP, Walker D, Mersereau JE, Gosiengfiao Y, Gracia CR. Pre-treatment

anti-Müllerian hormone levels determine rate of post-therapy ovarian reserve recovery: acute changes in ovarian

reserve during and after chemotherapy. Fertil Steril. 2013;99(2):477-483.

46. Bedoschi G, Navarro PA, Oktay K. Chemotherapy-induced damage to ovary: mechanisms and clinical impact.

Future Oncol. 2016;12(20):2333-2344.

47. Decanter C, Cloquet M, Dassonneville A, D’Orazio E, Mailliez A, Pigny P. Different patterns of ovarian recovery

after cancer treatment suggest various individual ovarian susceptibilities to chemotherapy. Reprod Biomed

Online. 2018;36(6):711-718.

48. Casey BM, Dashe JS, Wells CE, McIntire DD, Leveno KJ, Cunningham FG. Subclinical hyperthyroidism and

pregnancy outcomes. Obstet Gynecol. 2006;107(2 Pt 1):337-341.

49. Cho MK. Thyroid dysfunction and subfertility. Clinical and experimental reproductive medicine. 2015;42(4):131-135.

50. Krassas GE. Thyroid disease and female reproduction. Fertil Steril. 2000;74(6):1063-1070.

51. Clement SC, Peeters RP, Ronckers CM, Links TP, van den Heuvel-Eibrink MM, Nieveen van Dijkum EJ, van

Rijn RR, van der Pal HJ, Neggers SJ, Kremer LC, van Eck-Smit BL, van Santen HM. Intermediate and long-term

adverse effects of radioiodine therapy for differentiated thyroid carcinoma--a systematic review. Cancer Treat

Rev. 2015;41(10):925-934.

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

95

4

SUPPLEMENTARY DATA

Supplemental Figure 1. Flowchart of inclusion. Abbreviations: DTC, differentiated thyroid carcinoma and

SMN, secondary malignant neoplasm.

Supplemental Table 1. Parentally self-reported birth defects and major health problems in 45 children of

survivors of childhood differentiated thyroid carcinoma

Reported health problem n

Dilated ureter by birth 1

Blount disease 1

PDD-NOS 1

Lung tumor 1

Hypermobile Ehlers-Danlos syndrome 2a

Facial paralysis 1

Total 7

Abbreviation: PDD-NOS, pervasive developmental disorder-not otherwise specified. aChildren from the same mother.

CHAPTER 4

96

4

Supplemental Table 2. Percentages of differentiated thyroid carcinoma survivors with low AMH based on

10th percentile and 25th percentile of comparison group

DTC survivors

n = 54a

Comparison group

n = 420

P Value

10th percentile, n (%)b 0.278c

Low AMH 8 (14.8) 42 (10.0)

No low AMH 45 (85.2) 378 (90.0)

25th percentile, n (%)d 0.296c

Low AMH 10 (18.5) 105 (25.0)

No low AMH 44 (81.5) 315 (90.0)

Abbreviations; AMH, anti-Müllerian hormone and DTC, differentiated thyroid carcinoma. a two participants

participated only in questionnaire part of study. b threshold 10th percentile = 0.22 µg/L. c Pearson Chi-Square Test

(Asymptotic Significance). d threshold 25th percentile = 0.64 µg/L.

Supplemental Table 3. Anti-Müllerian hormone levels in subgroups of survivors of childhood differentiated

thyroid carcinoma

No hormonal contraceptive

n = 30

Hormonal contraceptive

n = 24

P Value

AMH, µg/L 2.3 (0.7-3.7) 1.9 (1.2-3.8) 0.676a

Single dose 131I

n = 28

Multiple doses 131I

n = 26

AMH, µg/L 1.5 (0.4-3.6) 2.3 (1.3-3.9) 0.319a

Abbreviations; AMH, Anti-Müllerian Hormone and 131I, radioactive iodine. Numbers shown as median (interquartile

range). a Mann-Whitney U test.

Supplemental Table 4. Hormonal evaluation in survivors of differentiated thyroid carcinoma during

childhood not using hormone contraceptives

Hormone n = 30

Follicle-stimulating hormone, IU/L 5.4 (3.2-13.2)

Luteinizing hormone, IU/L 8.6 (4.5-14.9)

Estradiol, nmol/L 0.4 (0.1-0.7)

Numbers are shown as median (interquartile range).

FEMALE FERTILITY AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

97

4

Supplemental Table 5. Folliculogenesis and anti-Müllerian hormone secretion

Follicular stage AMH secretion

Primordial follicle -

Primary follicle +

Secondary follicle ++

Early tertiary/ antral follicle +++

Late tertiary/pre-ovulatory follicle -

References: Andersen et al. Hum Reprod. 2010;25(5):1282-7.

Weenen et al. Mol Hum Reprod. 2004;10(2):77-83.

Chapter 5

Chapter 5

CHAPTER 5

Long-term evaluation of male fertility after

treatment with radioactive iodine for differentiated

thyroid carcinoma

Marloes Nies, Eus G.J.M. Arts, Evert F.S. van Velsen, Johannes G.M. Burgerhof, Anneke C. Muller Kobold, Eleonora P.M. Corssmit, Romana T. Netea-Maier, Robin P. Peeters, Anouk N.A. van der Horst-Schrivers, Astrid E.P. Cantineau*, and Thera P. Links*

*These authors contributed equally to this study.

Revised version submitted

CHAPTER 5

100

5

ABSTRACT BackgroundRadioactive iodine (131I) is often administered as part of the treatment for differentiated thyroid carcinoma (DTC). Short-term studies show a decrease in male fertility after administration of 131I, but long-term data are scarce. This study evaluated long-term male fertility after 131I for DTC, and compared semen quality before and after 131I administration.

MethodsThis multicenter study involving males diagnosed with DTC was conducted at least two years after their final 131I treatment with a total cumulative dose of 131I equal to or higher than 100 mCi/3.7 GBq. Evaluation of fertility consisted of semen analysis, hormonal evaluation, and a fertility-focused questionnaire. Cut-off scores for ‘low semen quality’ were based on reference values of the general population as defined by the World Health Organization.

ResultsThe 51 participants had a median age of 40.5 (interquartile range, IQR, 34.0-49.6) years upon evaluation, and a median follow-up of 5.8 (IQR 3.0-9.5) years after their last 131I administration. The median cumulative dose of 131I was 200.0 mCi/7.4 GBq. The proportion of males with a low semen volume, concentration, progressive motility, or total motile sperm count (TMSC) did not differ from the 10th percentile cut-off based on reference ranges of a general population (P=0.500, P=0.131, P=0.094, and P=0.500, respectively). However, a significantly and clinically relevant higher proportion of participants had semen concentration and motility below the 5th percentile (11.8% vs. 5%, P=0.029 and 15.0% vs. 5%, P=0.006). The live birth rate before and after 131I were 82.5% and 78.9%, respectively. Cryopreserved semen was used by one participant of the twenty who had preserved their semen. Median TMSC upon follow-up was significantly increased compared to TMSC upon semen preservation (121.2 vs. 66.9 million, respectively. P=0.036); 66.7% of these participants were hypothyroid at the moment of preservation.

ConclusionsAlthough a minority of participants treated with 131I for DTC experienced long-term poor semen quality, most participants had a normal long-term semen quality. Cryopreservation of semen of males with DTC is not crucial for conceiving a child after 131I administrations, but may be performed as a precaution.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

101

5

INTRODUCTIONAfter a total thyroidectomy, for the majority of patients treated for differentiated

thyroid carcinoma (DTC) further treatment consists of administration of radioactive

iodine (131I) (1). 131I destroys remaining thyroid (cancer) tissue, treats local or distant

metastases, and/or decreases the chance of recurrence of DTC (2-4). Although

increasing emphasis is being focused on the adverse effects on female fertility caused

by administration of 131I (5-7), little research has been performed regarding its effect

on fertility in males. 131I treatment can affect male fertility in various ways, mediated by absorption of β

and γ radiation (8, 9). 131I is excreted primarily through the urine (10), indirectly affecting

the testes. Therefore, adequate hydration, frequent micturition, and prevention of

constipation during 131I therapy are recommended to stimulate the excretion of 131I and

thus prevent its accumulation near the testes (1, 9, 11, 12). In addition, expression of

the sodium iodine symporter (NIS) has been found in germinal and Leydig cells, but

whether the NIS facilitates the uptake of 131I in the testes is unknown (13).

Damage to testicular cells induced by the administration of 131I probably causes

transient subfertility, but not permanent infertility (9, 11, 12, 14-16). Studies up to 18

months after 131I treatment show that administration of 131I causes a decreased semen

quality, elevated levels of luteinizing hormone (LH) and follicle-stimulating hormone

(FSH), and decreased levels of testosterone (T) (9, 11, 12, 14-18). However, previous

studies have also shown that males treated with 131I for DTC were later able to conceive

children (9, 19, 20). The short-term impairment of male fertility has been described

as dose-dependent (11, 12, 14, 16). Overall, studies on long-term male fertility after 131I

treatment are scarce.

These limited data seem to suggest that patients who receive multiple or high dose

treatment may be at risk of permanent impairment of fertility. Fertility preservation

has therefore been recommended for this group (1).

The primary aim of this study was to evaluate long-term male fertility after

administration of a high cumulative dose of 131I (equal to or higher than 100 mCi/3.7

GBq) for DTC by evaluating semen quality, serum endocrine markers (free T, LH and

FSH), and reproductive characteristics. The secondary aim was to compare semen

quality before and after treatment with 131I.

MATERIALS AND METHODSThis multicenter study was initiated by the University Medical Center Groningen

(UMCG) in the Netherlands. The institutional review board of the UMCG approved

the protocol on behalf of all participating centers (file number 2016/685), including

the Erasmus Medical Center, the Radboud University Medical Center, and

the Leiden University Medical Center. The study was registered in the Netherlands

CHAPTER 5

102

5

Trial Register, trial ID NL5966. All included patients signed written informed consent before participation.

ParticipantsTo select eligible participants, we consulted local databases. All identified male patients with an attained age of 18 years or older, diagnosed with DTC as of January 2000, who had been at least two years in follow-up since their final 131I treatment, and had had a total cumulative dose of 131I equal to or higher than 100 mCi/3.7 GBq, were eligible for inclusion. Participants were excluded if they used testosterone suppletion, chemotherapy, or anabolic steroids; if they had diseases or treatments potentially associated with impairment of semen quality (e.g. vasectomy or treatment with chemotherapy); if they were under treatment for active or widespread DTC; if they had received administrations of 131I only after use of recombinant human thyroid-stimulating hormone (rhTSH); or if they had noncompliance of thyroid hormone substitution, resulting in several measurements of TSH >10 mU/L in the year before participation. If a participant had had a fever (body temperature higher than 38.5°C) in the three months before the scheduled semen analysis, the semen analysis was postponed. Participants were included from May 2017 to March 2019.

Semen analysisParticipants were asked to abstain from ejaculation for a period of two to four days before participation. Semen samples were obtained through masturbation and were produced at the fertility departments of each participating center. Semen analysis took place within one hour after production. Semen analyses were performed according to the laboratory manual for the examination and processing of human semen of the World Health Organization, with additional guidelines provided by Björndahl et al. and Barrat et al. (21-23). We evaluated whether semen analyses were performed according to our protocol, and described protocol deviations. Semen samples were evaluated for pH, viscosity (scored as thin liquid, moderately viscous, or very viscous), volume (in mL), concentration (per mL), motility (percentage of rapid progressive forward, slow progressive forward, non-progressive, and immotile sperm cells), round cells (per mL), and morphology (percentage of sperm cells with normal morphology). Concentration and motility were evaluated in duplicate (200 sperm cells). Motility was assessed at a room temperature of 37°C, except for one center, where this was not possible for logistical reasons. Assessment of the percentage sperm of cells with a normal morphology was performed centrally in the UMCG according to Tygerberg criteria (24) and using Papanicolaou staining.

Blood sampling and hormonal measurementsBlood was drawn from participants, and samples were stored in a -80°C environment. We asked participants to provide the serum samples before noon. All blood samples

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

103

5

were then centrally measured in the UMCG. LH, FSH, total and free T, albumin, sex hormone-binding globulin (SHBG), TSH, and free thyroxine (FT4) were measured in the samples.

LH, FSH, TSH, and FT4 were analyzed by electrochemiluminescent immunoassay on the Cobas 6000 immunoanalyzer (Roche Diagnostics, Almere, the Netherlands). The interassay coefficients of variation (interCV, %) were 3.6 to 2.3% at levels ranging from 1.86 to 72.63 U/L, 2.8 to 2.7% at levels 2.15 to 41.74 U/L, 2.0 to 1.9% at levels 0.52 to 40.8 mU/L, and 4.2 to 4.5% at levels of 10.58 to 62.72 pmol/L, respectively. Plasma albumin was analyzed using the Bromocresol green method on the Cobas 6000 chemistry analyzer. The interCV ranged from 2.0 to 1.6% at levels ranging from 27.4 to 46.9 g/L. SHBG was analyzed by chemiluminescent microparticle immunoassay in serum on the Architect immunoanalyzer (Abbott Laboratories, Hoofddorp, the Netherlands). The interCV ranged from 5.91 to 2.29% at levels ranging from 5.46 to 117.8 nmol/L. Total T was analyzed by LC-MS/MS. The interCV were 2.4 to 5.6% at levels of 0.29 to 49.2 nmol/L. Free T was calculated using the formula of Vermeulen (25) using the total T value, SHBG and albumin. Reference ranges of the assays are reported in the relevant tables.

QuestionnaireEach participant was asked to complete a questionnaire regarding general health (medical history, use of drugs, intoxications, height, and weight), reproductive health (diseases, professional or leisure activities associated with impaired male fertility), and reproductive characteristics (conceived pregnancies and outcomes, fertility treatment, and fertility preservation).

Medical records and results of other semen analysesConsent to access medical records of the participants was obtained. Medical records were evaluated to obtain information about diagnostic, treatment, and follow-up characteristics. Additional consent was given to obtain information regarding semen analyses outside of the current study. If participants had preserved semen, information regarding the quality of the semen during preservation was collected.

Study definitionsDate of diagnosis was defined as the first histological confirmation of DTC. Age upon evaluation was defined as age at the moment of the semen analysis. Follow-up was defined as time from diagnosis or last 131I administration to semen analysis. The 8th edition of the American Joint Committee on Cancer (AJCC) tumor node metastasis scoring system was used (26). Total sperm count was defined as the product of the concentration and volume of the ejaculate. Total motile sperm count was defined as the product of the volume, concentration, and percentage of progressive spermatozoa of the ejaculate, divided by 100%.

CHAPTER 5

104

5

“Normal semen quality” has a wide range, and only having low semen quality will have clinical consequences. We determined cut-off values for having “low semen quality” based on the 10th percentile of the reference values of a general population of unscreened males for semen characteristics, provided by the World Health Organization (27). Based on this 10th percentile, a total motile sperm count below 10.6 million was considered as “low semen quality”. We considered a doubling of the proportion of males scoring below the 10.6 million cut-off value to be a clinically relevant finding (i.e. 20% of the participants). The cut-off values for volume, concentration, progressive motility, and total sperm count were also based on the 10th percentile.

Statistical analysisCharacteristics of participants, and treatment and outcome variables, were described using median and interquartile range (IQR), unless stated otherwise. In case of protocol violations these parameters were excluded from analysis. For our primary aim, we used a binomial test to evaluate whether the proportion of participants having a total motile sperm count below 10.6 million significantly differed from the proportion in the general population (10%) (27). The Clopper-Pearson binomial proportion 95% confidence interval (CI) was used to show estimated proportions. Thereby, we also evaluated whether the proportion of participants with a low volume, concentration, progressive motility, or total sperm count differed from the 10% in the general population. To evaluate whether results changed when we based the cut-off value on the 5th or 25th percentiles, we performed binomial tests with these cut-off values.

Subsequently, using Mann-Whitney U tests we evaluated whether median values of age upon evaluation, body mass index (BMI), follow-up period since the last 131I administration, or the cumulative dose of 131I differed between the groups that scored below or above the 10th percentile cut-off value of semen parameters (volume, concentration, motility, and total motile sperm count) or between the participants who did or did not conceive children. We evaluated whether the proportion of participants scoring below cut-off values differed depending on whether they had undergone single or multiple administrations of 131I; for this we used Chi-square tests or Fisher’s exact tests (if more than 20% of the cells had an expected count below 5). We described results of the questionnaire regarding reproductive characteristics.

We made paired comparisons between the median total motile sperm count upon semen cryopreservation and the median total motile sperm count during follow-up using a Wilcoxon signed-rank test. For this analysis, we evaluated only participants who cryopreserved semen before their first administration of 131I. If semen was preserved multiple times before 131I, we calculated the median total motile sperm count. A P value ≤0.05 was considered statistically significant. For statistical analyses, IBM SPSS Statistics version 23.0.0.3 for Windows (IBM, Armonk, NY, USA) and Microsoft Excel Professional Plus 2016 were used.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

105

5

RESULTSParticipantsWe identified 236 males who met the inclusion criteria. Seventy-one males met one or more exclusion criteria. Most excluded males had undergone a vasectomy (n=25) or had a comorbidity interfering with their ability to participate, or they had a disease or treatment that impaired semen quality (n=24). Twenty-two males were excluded for other reasons. Of the 165 eligible males, 51 (30.9%) gave informed consent and completed their participation, see Supplementary Figure 1.

All 51 participants had the Dutch nationality. The median age of participants upon evaluation was 40.5 years (IQR 34.0 to 49.6). Participants were diagnosed at a median age of 33.7 years (IQR 26.9 to 39.8 years). Participants had a median BMI of 26.8 kg/m2

(IQR 24.0 to 29.4 kg/m2, unknown in two participants). At the moment of evaluation one of the participants (2.0%) was an active smoker, 13 participants (25.5%) had previously smoked, and 32 participants (62.7%) had never smoked. The smoking status of five participants was unknown. Four participants reported having had an orchidopexy due to cryptorchidism. The orchidopexy was performed on both testes in three of these four participants. One participant reported having used anabolic steroids; his latest use had been three years before evaluation. One participant had been treated with testosterone during puberty to decrease his final height; he participated in the study at the age of 36.

Treatment and pathology characteristicsTreatment characteristics of the participants are shown in Table 1. All participants were treated with a total thyroidectomy, 32 of whom (62.7%) had a lymph node dissection. Most participants received one administration of 131I (n=26, 51.0%), and none of them had more than four 131I administrations. The median cumulative dose of 131I was 200.0 mCi/7.4 GBq (IQR 150.0 to 300.0 mCi/5.6 to 11.1 GBq). Median follow-up since the last treatment with 131I was 5.8 years (IQR 3.0 to 9.5 years).

Papillary thyroid carcinoma was the most common histology type, with 49 (96.1%) cases. One participant (2.0%) had follicular thyroid carcinoma and one (2.0%) was diagnosed with Hürthle cell carcinoma. T2 and T3a/T3b were the most common tumor stages (n=20, 39.2% and n=18, 35.3%, respectively). Thirty participants (58.8%) had metastases of DTC to the regional lymph nodes, and three (5.9%) had lung metastases (Supplementary Table 1).

Semen analysisNone of the subjects reported having had a fever within three months before evaluation. The median abstinence period was four days (IQR 3 to 6 days). All samples were obtained through masturbation. Semen analysis took place within one hour after ejaculation for 46 (90.2%) participants. Semen samples were analyzed between

CHAPTER 5

106

5

Table 1. Treatment Characteristics of Participants treated with Radioactive Iodine for Differentiated

Thyroid Carcinoma

Characteristic Participants

n=51

Total thyroidectomy1, n (%) 51 (100.0)

Lymph node dissection accompanying thyroid surgery, n (%)Only central 7 (13.7)

Only (bi)lateral 4 (7.8)

Central and (bi)lateral 21 (41.2)

None 19 (37.3)

Number of 131I administrations, n (%)One administration 26 (51.0)

Two administrations 17 (33.3)

Three administrations 4 (7.8)

Four administrations 4 (7.8)

Dosage per 131I administration, mCi 150.0 (143.0-150.0)

Cumulative 131I dose, mCi 200.0 (150.0-300.0)

Follow-up since last 131I therapy, years 5.8 (3.0-9.5)

Follow-up since diagnosis, years 6.5 (3.9-11.0)

Numbers shown as median (interquartile range). Abbreviations: 131I, radioactive iodine. 1 15 participants (29.4%) had

a total thyroidectomy in two tempi.

one and two hours in four participants. Time to analysis can affect the motility of spermatozoa, but the progressive motility percentages did not significantly differ between semen samples analyzed within one hour and samples analyzed between one and two hours (P=0.447). Time to analysis was unknown in one participant. Semen characteristics of the participants are shown in Supplementary Table 2.

The proportions of participants scoring below the 10th percentile cut-off point for volume, concentration, and progressive motile spermatozoa were 9.8%, 15.7% and 17.5%, respectively (see Table 2). The binomial test showed that these proportions did not differ significantly from the 10% in the general population (P=0.500, P=0.131, and P=0.094, respectively) (27). The proportion of participants scoring below the 10.6 million cut-off point for total motile sperm count was 10.0% (n=4). A binomial test showed that this proportion did not significantly differ from the 10% in the general population (P=0.500) (27). The proportion of participants with a total motile sperm count below 10.6 million was lower than the 20% that we defined as clinically relevant.

When cut-off points for volume, concentration, progressive motility, total sperm count, and total motile sperm count were based on the 5th or 25th percentile of the general population, the proportion of males with low concentration (11.8% and 37.3%, respectively) and low progressive motility (15.0% and 37.5% respectively) was statistically significantly higher than the 5% or 25% in the general population

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

107

5

(P=0.029, P=0.006, P=0.031, and P=0.050). However, only for the 5th percentile cut-off value, the proportions of participants with low concentration and progressive motility were higher than the proportion of 10% that we considered clinically relevant (Supplementary Table 3). For volume, total sperm count, and total motile sperm count, the proportion of participants did not show statistically or clinically significant differences.

Evaluation of serum hormone levelsHormonal evaluation was performed in 48 participants (Table 3). The median values of FSH, LH, total T, and free T were within reference values of the corresponding assays, except for the median value of total T for participants aged 50 years or older (9.7 nmol/L, reference range 10.90 to 30.79 nmol/L). The median FT4 of 23.3 pmol/L was outside the reference range of 11.0 to 19.5 pmol/L.

Table 2. Semen Characteristics of 51 Participants treated with Radioactive Iodine for Differentiated Thyroid

Carcinoma compared to the 10th Percentile of Reference Values based on a General Population1

10th Percentile cut-off points Participants

n (%)

P Value 95% CI of estimated proportion

Volume, mL 0.5002

≥ 1.6 mL 46 (90.2)

< 1.6 mL 5 (9.8) [3.3 – 21.4%]

Concentration, x106 0.1312

≥ 17 million 43 (84.3)

< 17 million 8 (15.7) [7.0 – 28.6%]

Progressive motility3, percentage 0.0942

≥ 39% 33 (82.5)

< 39% 7 (17.5) [7.3 – 32.8%]

Total sperm count, x106 0.4262

≥ 27.2 million 45 (88.2)

< 27.2 million 6 (11.8) [4.4 – 23.9%]

Total motile sperm count3,4, x106 0.5002

≥ 10.6 million 36 (90.0)

< 10.6 million 4 (10.0) [2.8 – 23.7%]

P values in bold indicate a statistically significant difference (P < 0.05). Abbreviations: CI, confidence interval. 1

Cut-off points were based on the 10th percentile of a general population, as described by Cooper TG et al., Human

Reproduction update, 2010. 16(3): p. 231-245. 2 Binomial tests with the hypothesized proportion of 0.1. Clopper-Pearson

binomial proportion confidence intervals are shown. 3 n=40; it was not possible to perform motility evaluations at

a temperature of 37°C in one center, we excluded these cases from analysis (n=11). 4 Total motile sperm count is

defined as the product of the cut-off values of the 10th percentile as defined by Cooper et al. for concentration, and

percentage of progressive spermatozoa of ejaculate, divided by 100%.

CHAPTER 5

108

5

Characteristics associated with male fertilityThe proportion of participants scoring above the 10th percentile cut-off value for volume had a higher median BMI than the proportion scoring higher than the cut-off value (27.1 vs. 22.2 kg/m2, P = 0.009). Participants who conceived children had a higher median age upon evaluation than participants who never conceived children (44.7 vs. 33.6 years, P = 0.005). Other associations between participant or treatment characteristics and semen characteristics were not statistically significant (see Supplementary Table 4).

Reproductive characteristicsOf 51 participants, 50 completed the questionnaire. Thirty of these 50 participants (60.0%) reported having conceived a pregnancy. Three of the 50 participants (6.0%) reported having an unfulfilled desire to have (more) children, two due to the lack of a partner and one due to a miscarriage. Nine of the 50 participants (18.0%) reported that their desire to have children had changed because of their diagnosis and treatment of DTC. Some reported having (temporarily or permanently) put off conceiving; others reported having expedited conceiving.

Six of the 50 participants (12.0%) had previously visited a fertility clinic or physician for not being able to conceive naturally; three of them underwent fertility treatment and three never had a fertility treatment. One participant and his partner underwent in vitro fertilization and intracytoplasmic sperm injection treatments in the years after 131I treatment. Another participant and his partner had intra uterine insemination (IUI) treatments. One participant did not specify the type of fertility treatment.

Table 3. Hormonal Evaluation of Participants treated with Radioactive Iodine for Differentiated

Thyroid Carcinoma

Hormone Participants

n=481

Reference

range

median (IQR) range

Follicle-stimulating hormone, U/L 3.7 (2.9-5.9) 1.5-23.5 1.5-12.4

Luteinizing hormone, IU/L 4.9 (4.0-6.8) 2.6-11.9 1.7-8.6

Total Testosterone, nmol/L 13.6 (9.8-16.7) 6.3-28.2 n.a.

<50 years old2 14.7 (10.6-17.1) 6.9-28.2 11.37-34.32

≥50 years old3 9.7 (9.3-15.0) 6.3-24.8 10.90-30.79

Free Testosterone, nmol/L 0.3 (0.2-0.4) 0.2-0.6 0.3-0.64

Thyroid-stimulating hormone, mU/L 0.5 (0.1-1.6) <0.1-7.7 0.5-4.0

Free thyroxine, pmol/L 23.3 (21.4-25.6) 18.3-32.4 11.0-19.5

Abbreviations: IQR, interquartile range and n.a., not applicable. 1 48 of 51 participants had a serum sample drawn. 2 n = 37. 3 n = 11.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

109

5

An overview of conceived pregnancies and pregnancy outcomes before and after 131I administrations is shown in Supplementary Figure 2. The live birth rate before 131I administration was 82.5%, and the live birth rate after 131I administration was 78.9%. None of the participants reported having fathered children with congenital malformations or major health problems.

Semen cryopreservation and longitudinal evaluation of semen qualitySemen cryopreservation was performed in 20/50 participants (40%). For one participant, it was unknown whether semen cryopreservation was performed. Participants had a median age of 32.4 years upon semen cryopreservation (IQR 23.6 to 33.8 years, range 16.6 to 50.1 years).

One participant (5.0%) reported having used his cryopreserved semen and having subsequent IUI treatments. Seventeen participants (85.0%) did not use their cryopreserved semen and two (10%) did not report their usage.

For the second aim of the study, we compared semen quality before and after 131I. We evaluated only the 15/20 participants in whom semen cryopreservation took place before their first administration of 131I. For these 15 participants, the median total motile sperm count significantly increased from 66.9 million at semen cryopreservation to 121.2 million (IQR 17.1 to 115.1 million and 48.4 to 204.6 million, P=0.036) upon follow-up after a median follow-up period of 4.1 years (IQR 3.1 to 6.4 years). Figure 1 shows the individual changes in semen quality over time from the moment of semen cryopreservation to follow-up. Based on available TSH values, 10/15 participants were hypothyroid at the moment of semen cryopreservation, 3/15 were euthyroid, and for the remaining 2/15 TSH levels were not available at the time of semen cryopreservation.

We asked participants whether they found it important to be offered the possibility of semen preservation. 58% reported finding this very important, 18% found it fairly important, 18% had a neutral opinion, and 4% found it not important at all. One participant did not answer this question.

DISCUSSIONThe current study assessed long-term male fertility after treatment with a high dose of 131I for DTC evaluating semen quality, hormone levels, and reproductive characteristics. A relatively small number of participants treated with 131I for DTC had long-term poor semen quality, depicted by the higher number of participants who had semen motility and concentration below the cut-off value based on the 5th percentile of a general population (27). No statistically or clinically relevant differences in proportions were seen between the participants and the general population, based on the 10th or 25th percentile for having low semen quality. Ultimately, the clinical consequences of this finding may be limited since we found no detrimental effects on reproductive

CHAPTER 5

110

5

Fig

ure

1. T

ota

l mo

tile

sp

erm

co

unt (

TMSC

, per

mill

ion)

of 1

5 pa

rtic

ipan

ts a

t the

mo

men

t of s

emen

pre

serv

atio

n (t

=0) a

nd a

t las

t kno

wn

sem

en a

naly

sis.

Gre

en li

nes

repr

esen

t pa

rtic

ipan

ts t

reat

ed w

ith

a cu

mul

ativ

e d

ose

of 1

50 m

Ci o

r le

ss, b

lue

lines

rep

rese

nt p

arti

cipa

nts

trea

ted

wit

h 15

0-2

99 m

Ci,

and

red

line

s re

pres

ent

do

ses

of 3

00

mC

i or

mo

re. T

he v

alue

of t

he 10

th p

erce

ntile

for

TMSC

of a

gen

eral

po

pula

tio

n (a

s d

efine

d b

y C

oo

per

TG

et a

l. H

uman

Rep

rod

ucti

on

upd

ate,

20

10.16

(3):

231-

245)

is 1

0.6

mill

ion,

dep

icte

d b

y th

e so

lid h

ori

zont

al li

ne. A

ll bu

t fiv

e pa

rtic

ipan

ts w

ere

hyp

oth

yro

id a

t m

om

ent

of s

emen

cry

opr

eser

vati

on.

So

lid li

nes

repr

esen

t

hyp

oth

yro

id p

arti

cipa

nts,

das

hed

line

s eu

thyr

oid

par

tici

pant

s, a

nd d

ott

ed li

nes

part

icip

ants

wit

h un

kno

wn

thyr

oid

al s

tate

.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

111

5

characteristics. The evaluation of LH, FSH, total T, and free T did not show clinically relevant abnormalities.

Evaluated after a median of five years after their final treatment with 131I, the normal male fertility in the majority of current survivors seems to confirm the prospect of recovery of parameters of male fertility over time, as also indicated by other studies. Previous studies evaluating semen quality or the functioning of the testes had a follow-up to a maximum of 18 months (11, 12, 14-18). All studies reported transient damage of male fertility. Some reported normalization of parameters over time; others found a persistent disturbance upon 12 or 18 months of follow-up, especially in patients treated with higher or multiple administrations of 131I (14, 16), suggesting a dose-dependent effect (11, 12, 14, 16). However, this effect was confirmed neither in the current study nor a previous study (18).

By defining various cut-off values for low semen quality, we were able to detect subtle disturbances in semen quality of males treated with DTC as we found that there were more participants with poor semen motility and concentration in the lower range. No predictors for poor semen quality were identified. No relation between BMI and semen volume has previously been described (28), our results might be explained by a multiple testing effect.

The fact that we did not find a dose-dependent relationship between 131I and impairment of male fertility may be explained by our long-term follow-up. The increase of semen quality after treatment is the result of spermatogenesis: self-renewal and differentiation of spermatogonial stem cells into new spermatozoa (29). The magnitude of damage caused to the testes by radiation depends on several factors, such as radiation dose, the number of doses, and the surface of the testes exposed to the radiation (30). The duration of recovery of male fertility increases when the damage caused by radiation is greater (16). In the current study, the long-term follow-up period apparently allowed for continued spermatogenesis in most participants with subsequent normal findings.

A low usage rate of the cryopreserved semen, as in our study, was previously described in a study evaluating the usage rate of cryopreserved semen in males treated for various cancer types (31). Possible explanations for this low usage rate may be that males do not use their preserved semen because they have conceived naturally, but it may also indicate a change in the wish to conceive children. Considering the age of the participants, future use of cryopreserved semen can, however, not be ruled out.

The reported reproductive characteristics did not show major abnormalities in the 12% of participants who sought medical assistance for not being able to conceive. This number is comparable to other couples In the Netherlands (32). Thereby, live birth rate before and after 131I administration did not differ.

The management guidelines of the American Thyroid Association state that semen preservation can be considered for men who receive cumulative 131I doses of more than

CHAPTER 5

112

5

400 mCi (1). Current Dutch guidelines recommend discussing semen cryopreservation with all males to be treated with 131I (33). Based on current and previous results, no firm evidence indicates that fertility preservation in males is necessary for conceiving a child after treatment with 131I for DTC. However, semen cryopreservation can be performed as a precaution, considering the higher occurrence of low semen quality after long-term follow-up in some males. Another reason to perform semen cryopreservation may be that the patient in question has a desire to have children sooner after 131I treatment. One additional argument to continue semen preservation in males treated with 131I may be the slight (but significant) elevation of numerical chromosomal aberrations in spermatozoa still observed one year after 131I administration, with its initial peak at three months after 131I administration (15). Theoretically, such chromosomal aberrations can affect fecundity and offspring, but to date there is no proof that that this clinically affects males treated with 131I for DTC, as congenital malformations or health defects were not associated with children fathered by patients treated with 131I for DTC in the current or previous studies (9, 20).

Our data indicate that the impairment in semen quality caused by hypothyroidism is more relevant than possible long-term impairment of semen quality caused by 131I. If semen is cryopreserved, a euthyroid state of the patient would provide semen of optimal quality (34). However, since cryopreserved semen is usually applied in an assisted reproduction procedure, preservation of semen with a suboptimal quality is also acceptable.

Strengths and limitationsThis is the first study to evaluate a broad range of aspects of male fertility after a long-term follow-up period subsequent to administration of 131I for DTC. We specifically focused on males treated with a relatively high cumulative dose of 131I. A limitation of this study was the lack of a control group. Although the reference ranges used to compare current results are also currently used in daily practice, it would be preferable to have a comparison or control group. By defining several cut-off values for our analysis, we avoided basing our results on an arbitrary cut-off point. The number of centers eligible to participate in the current study was restricted by the fact that not all laboratories were able to perform semen analyses according to our protocol. This subsequently limited the number of participants. To provide highly accurate data, we excluded results that were not acquired according to the protocol. This resulted in the inclusion of fewer participants for the analysis of some parameters

In conclusion, administration of 131I for DTC does not impair long-term male fertility in the majority of the patients. However, cryopreservation of semen before administration of 131I for DTC should be discussed with all male patients treated with a dose of 100 mCi or more, since we could not identify clear risk factors for impaired semen quality. Therefore selection of males for fertility preservation is not possible.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

113

5

ACKNOWLEDGEMENTSWe are grateful to the participants for selflessly committing to this study and sharing their information with us. This work could not have been completed without the help of Lammie Stoffer, Janny G. Wijchman, Gerlinde J. Knol, and Bettien M. van Hemel. We thank the embryologists and laboratory analysts from the other participating centers for their valuable contributions.

AUTHOR CONTRIBUTIONSConception or design of the work: MN, EGJMA, JGMB, ANAHS, AEPC, and TPL; data collection: MN, EGJMA, EFSV, ACMK, EPMC, RTNM, RPP, and TPL; data analysis and interpretation: MN, EGJMA, JGMB, ACMK, AEPC, and TPL; drafting the article: MN, EGJMA, AEPC, and TPL; critical revision of the article: EFSV, JGMB, ACMK, EPMC, RTNM, RPP, and ANAHS; Final approval of the version to be published: all authors.

AUTHOR DISCLOSURE STATEMENTNo competing financial interests exist for all authors.

FUNDINGThis work was supported by the University Medical Center Groningen Cancer Research Fund and the Junior Scientific Masterclass Groningen.

CHAPTER 5

114

5

REFERENCES 1. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM,

Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L. 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. 2016;26(1):1-133.

2. Orosco RK, Hussain T, Noel JE, Chang DC, Dosiou C, Mittra E, Divi V, Orloff LA. Radioactive iodine in differentiated

thyroid cancer: a national database perspective. Endocr Relat Cancer. 2019;26(10):795-802.

3. Van Nostrand D. The benefits and risks of I-131 therapy in patients with well-differentiated thyroid cancer.

Thyroid. 2009;19(12):1381-1391.

4. Banerjee M, Reyes-Gastelum D, Haymart MR. Treatment-free survival in patients with differentiated thyroid

cancer. J Clin Endocrinol Metab. 2018;103(7):2720-2727.

5. Evranos B, Faki S, Polat SB, Bestepe N, Ersoy R, Cakir B. Effects of radioactive iodine therapy on ovarian reserve:

a prospective pilot study. Thyroid. 2018;28(12):1702-1707.

6. Yaish I, Azem F, Gutfeld O, Silman Z, Serebro M, Sharon O, Shefer G, Limor R, Stern N, Tordjman KM. A single

radioactive iodine treatment has a deleterious effect on ovarian reserve in women with thyroid cancer: results of

a prospective pilot study. Thyroid. 2018;28(4):522-527.

7. van Velsen EFS, Visser WE, van den Berg SAA, Kam BLR, van Ginhoven TM, Massolt ET, Peeters RP. Longitudinal

analysis of the effect of radioiodine therapy on ovarian reserve in females with differentiated thyroid cancer.

Thyroid. 2020;30(4):580-587.

8. Ceccarelli C, Battisti P, Gasperi M, Fantuzzi E, Pacini F, Gualdrini G, Pierantoni MC, Luciani A, Djokich D, Pinchera

A. Radiation dose to the testes after 131I therapy for ablation of postsurgical thyroid remnants in patients with

differentiated thyroid cancer. J Nucl Med. 1999;40(10):1716-1721.

9. Hyer S, Vini L, O’Connell M, Pratt B, Harmer C. Testicular dose and fertility in men following I(131) therapy for

thyroid cancer. Clin Endocrinol (Oxf). 2002;56(6):755-758.

10. Riggs DS. Quantitative aspects of iodine metabolism in man. Pharmacol Rev. 1952;4(3):284-370.

11. Pacini F, Gasperi M, Fugazzola L, Ceccarelli C, Lippi F, Centoni R, Martino E, Pinchera A. Testicular function in

patients with differentiated thyroid carcinoma treated with radioiodine. J Nucl Med. 1994;35(9):1418-1422.

12. Rosario PW, Barroso AL, Rezende LL, Padrao EL, Borges MA, Guimaraes VC, Purisch S. Testicular function after

radioiodine therapy in patients with thyroid cancer. Thyroid. 2006;16(7):667-670.

13. Russo D, Scipioni A, Durante C, Ferretti E, Gandini L, Maggisano V, Paoli D, Verrienti A, Costante G, Lenzi A, Filetti S.

Expression and localization of the sodium/iodide symporter (NIS) in testicular cells. Endocrine. 2011;40(1):35-40.

14. Canale D, Ceccarelli C, Caglieresi C, Moscatelli A, Gavioli S, Santini P, Elisei R, Vitti P. Effects of radioiodine

treatment for differentiated thyroid cancer on testis function. Clin Endocrinol (Oxf). 2015;82(2):295-299.

15. Bourcigaux N, Rubino C, Berthaud I, Toubert ME, Donadille B, Leenhardt L, Petrot-Keller I, Barlly-Tabard S,

Fromigue J, de Vathaire F, Simon T, Siffroi JP, Schlumberger M, Bouchard P, Christin-Maitre S. Impact on testicular

function of a single ablative activity of 3.7 GBq radioactive iodine for differentiated thyroid carcinoma. Hum

Reprod. 2018;33(8):1408-1416.

16. Handelsman DJ, Turtle JR. Testicular damage after radioactive iodine (I-131) therapy for thyroid cancer. Clin

Endocrinol (Oxf). 1983;18(5):465-472.

17. Esfahani AF, Eftekhari M, Zenooz N, Saghari M. Gonadal function in patients with differentiated thyroid cancer

treated with (131)I. Hell J Nucl Med. 2004;7(1):52-55.

18. Wichers M, Benz E, Palmedo H, Biersack HJ, Grunwald F, Klingmuller D. Testicular function after radioiodine

therapy for thyroid carcinoma. Eur J Nucl Med. 2000;27(5):503-507.

19. Dottorini ME, Vignati A, Mazzucchelli L, Lomuscio G, Colombo L. Differentiated thyroid carcinoma in children and

adolescents: a 37-year experience in 85 patients. J Nucl Med. 1997;38(5):669-675.

20. Sarkar SD, Beierwaltes WH, Gill SP, Cowley BJ. Subsequent fertility and birth histories of children and adolescents

treated with 131I for thyroid cancer. J Nucl Med. 1976;17(6):460-464.

21. World Health O. WHO laboratory manual for the examination and processing of human semen. 2010 (5th ed.).

22. Bjorndahl L, Barratt CL, Mortimer D, Jouannet P. ‘How to count sperm properly’: checklist for acceptability of

studies based on human semen analysis. Hum Reprod. 2016;31(2):227-232.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

115

5

23. Barratt CLR, Björndahl L, Menkveld R, Mortimer D. ESHRE special interest group for andrology basic semen analysis

course: a continued focus on accuracy, quality, efficiency and clinical relevance†. Hum Reprod. 2011;26(12):3207-3212.

24. Menkveld R, Stander FS, Kotze TJ, Kruger TF, van Zyl JA. The evaluation of morphological characteristics of human

spermatozoa according to stricter criteria. Hum Reprod. 1990;5(5):586-592.

25. Vermeulen A, Verdonck L, Kaufman JM. A critical evaluation of simple methods for the estimation of free

testosterone in serum. J Clin Endocrinol Metab. 1999;84(10):3666-3672.

26. Amin Mahul B. AJCC Cancer Staging Manual: Springer; 2017.

27. Cooper TG, Noonan E, von Eckardstein S, Auger J, Baker HW, Behre HM, Haugen TB, Kruger T, Wang C, Mbizvo

MT, Vogelsong KM. World Health Organization reference values for human semen characteristics. Hum Reprod

Update. 2010;16(3):231-245.

28. Jensen TK, Andersson A-M, Jørgensen N, Andersen A-G, Carlsen E, Skakkebæk NE. Body mass index in relation to

semen quality and reproductive hormones among 1,558 Danish men. Fertil Steril. 2004;82(4):863-870.

29. De Felice F, Marchetti C, Marampon F, Cascialli G, Muzii L, Tombolini V. Radiation effects on male fertility.

Andrology. 2019;7(1):2-7.

30. Okada K, Fujisawa M. Recovery of spermatogenesis following cancer treatment with cytotoxic chemotherapy and

radiotherapy. World J Mens Health. 2019;37(2):166-174.

31. Muller I, Oude Ophuis RJ, Broekmans FJ, Lock TM. Semen cryopreservation and usage rate for assisted reproductive

technology in 898 men with cancer. Reprod Biomed Online. 2016;32(2):147-153.

32. Beurskens MP, Maas JW, Evers JL. Subfertility in South Limburg: calculation of incidence and appeal for specialist

care. Ned Tijdschr Geneeskd. 1995;139(5):235-238.

33. Oncoline, Cancer Clinical Practice Guidelines. Richtlijn Schildkliercarcinoom 2.0. 2015.

34. Krassas GE, Papadopoulou F, Tziomalos K, Zeginiadou T, Pontikides N. Hypothyroidism has an adverse effect on

human spermatogenesis: a prospective, controlled study. Thyroid. 2008;18(12):1255-1259.

CHAPTER 5

116

5

SUPPLEMENTARY DATA

Supplemental Figure 1. Flowchart of inclusion. Abbreviations: DTC, differentiated thyroid carcinoma;

rhTSH, recombinant human thyrotropin; RAI, radioactive iodine. 1 All identified males aged 18 to 60 years

old, diagnosed with differentiated thyroid carcinoma, treated with a cumulative dose of radioactive iodine

≥100 mCi/3.7 GBq and at least two years after their last treatment with radioactive iodine. 2 Comorbidities:

malignancy or current or previous treatment with drugs impairing fertility (n=16), erectile dysfunction

(n=3), psychiatric disorder interfering with ability to participate (n=2), spinal cord injury/paraplegia (n=1),

treatment with testosterone (n=1), mentally disabled (n=1). 3 Decision physician: vulnerable patient (n=3),

non-compliant to therapy (n=1).

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

117

5

Supplemental Figure 2. Overview of number of conceived pregnancies and pregnancy outcomes before

and after treatment with a cumulative dose of at least 100 mCi of radioactive iodine (RAI) in 50 males

diagnosed with differentiated thyroid carcinoma. For one participant who conceived two pregnancies,

it was unknown if the pregnancies were conceived before or after RAI treatments. No participants

reported having fathered children with congenital malformations or major health problems. One out of

51 participants did not report on his conceived pregnancies.

CHAPTER 5

118

5

Supplemental Table 1. Pathology Characteristics of Male Participants treated with Radioactive Iodine for

Differentiated Thyroid Carcinoma

Characteristic Participants

n=51

Histology, n (%)Papillary 49 (96.1)

Classical variant 31

Follicular variant 9

Tall cell variant 1

Diffuse sclerosing variant 1

Other1 2

Unknown 5

Follicular 1 (2.0)

Minimally invasive variant 1

Hürthle cell carcinoma 1 (2.0)

Primary tumor size2, mm 28.0 (19.3-49.5)

Multifocality, n (%)Yes 26 (51.0)

No 25 (49.0)

Extranodal growth of lymph nodes3

Yes 9 (17.6)

No 10 (19.6)

Unknown 11 (21.6)

Tumor node metastasis stage, n (%)T stage

T0 1 (2.0)

T1a 6 (11.8)

T1b 5 (9.8)

T2 20 (39.2)

T3a 16 (31.4)

T3b 2 (3.9)

Tx 1 (2.0)

N stage

N0 21 (41.2)

N1 1 (2.0)

N1a 3 (5.9)

N1b4 26 (51.0)

M stage

M0 48 (4.1)

M15 3 (5.9)

Primary tumor size shown as median (interquartile range).1 Mixed classical and follicular (n=1), Warthin-like subtype of papillary thyroid carcinoma (n=1). 2 n=48; tumor size was

unknown in three patients. 3 n=30; no malignant lymph nodes were present in 21 patients. 3 In one patient, a lymph

node metastasis was found upon reviewing pathology after a parathyroidectomy, therefore N1b. No lymph node

dissection was performed. 5 All participants with distant metastases had metastases to the lungs.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

119

5

Supplemental Table 2. Semen Characteristics of Participants treated with Radioactive Iodine for Differentiated

Thyroid Carcinoma

Semen Characteristic Participants

n=51

50th percentile of the General

Population1

pH of semen 7.7 (7.5-8.0) -

Viscosity, n (%) -

Thin liquid 36 (70.6)

Moderately viscous 12 (23.5)

Very viscous 2 (3.9)

Unknown 1 (2.0)

Volume, mL 3.6 (2.7-4.5) 3.2

Concentration, x106/mL 49.0 (26.6-90.0) 64

Motility2, percentageGrade A3 35.5 (12.3-53.8) 57 (A+B)

Grade B4 8.5 (2.3-28.8)

Grade C5 3.0 (1.0-5.0)

Grade D6 42.5 (32.8-52.5)

Total sperm count, x106 191.7 (71.6-357.0) 196

Total motile sperm count2,7, x106 116.1 (44.4-172.4) 116.7

Round cells8, x106/mL 0.5 (0.2-1.5) -

Morphology9, percentage normal 8.5 (5.8-11.0) -

Numbers are shown as median (interquartile range). Assessment of motility and total motile sperm count shown

for 40 participants; in one laboratory, evaluation of motility at 37°C was not possible. 1 As described by Cooper TG et

al., Human Reproduction update, 2010. 16(3): p. 231-245. 2 n=40; in one laboratory, evaluation of motility at 37°C was

not possible and these cases were excluded from this analysis (n=11). 3 Rapid progressive forward. 4 Slow progressive

forward. 5 Non-progressive. 6 Immotile. 7 Total motile sperm count is defined as the product of the cut-off values of

the 10th percentile as defined by Cooper et al. for concentration, and percentage of progressive spermatozoa of

the ejaculate, divided by 100%. 8 n=39; presence of round cells was unknown in 12 participants. 9 n=50; this parameter

could not be evaluated in one participant, since only a very low number of immotile spermatozoa were present in

the ejaculate.

CHAPTER 5

120

5

Supplemental Table 3. Semen Characteristics of 51 Male Participants treated with Radioactive Iodine for

Differentiated Thyroid Carcinoma, compared to the 5th and 25th Percentile of Reference Ranges based on

a General Population1

Cut-off points Participants

n (%)

P Value 95% CI of

estimated proportion

5TH PERCENTILE

Volume, mL 0.4872

≥ 1.2 mL 49 (96.1)

< 1.2 mL 2 (3.9) [0.5 – 13.5%]

Concentration, x 106 0.0292

≥ 9 million 45 (88.2)

< 9 million 6 (11.8) [4.4 – 23.9%]

Progressive motility3, percentage 0.0062

≥ 31% 34 (85.0)

< 31% 6 (15.0) [5.7 – 29.8%]

Total sperm count, x106 0.4872

≥ 10.8 million 49 (96.1)

< 10.8 million 2 (3.9) [0.5 – 13.5%]

Total motile sperm count3,4, x106 0.5002

≥ 3.3 million 38 (95.0)

< 3.3 million 2 (5.0) [0.6 – 16.9%]

25TH PERCENTILE

Volume, mL 0.3435

≥ 2.2 mL 40 (78.4)

< 2.2 mL 11 (21.6) [11.3– 35.3%]

Concentration, x 106 0.0315

≥ 36 million 32 (62.7)

< 36 million 19 (37.3) [24.1 – 51.9%]

Progressive motility3, percentage 0.0505

≥ 49% 25 (62.5)

< 49% 15 (37.5) [22.7 – 54.2%]

Total sperm count, x106 0.4045

≥ 79.2 million 37 (72.5)

< 79.2 million 14 (27.5) [15.9 – 41.7%]

Total motile sperm count3,4, x106 0.4285

≥ 38.8 million 31 (77.5)

< 38.8 million 9 (22.5) [10.8 – 38.5%]

P values in bold indicate a statistically significant difference (P < 0.05). Abbreviations: CI, confidence interval. 1 Cut-off

points were based on the 5th and 25th percentile of a general population, as described by Cooper TG et al., Human

Reproduction update, 2010. 16(3): p. 231-245. 2 Binomial test with the hypothesized proportion of 0.05. 3 n=40; in one

laboratory, evaluation of motility at 37°C was not possible and these cases were excluded from this analysis (n=11). 4Total motile sperm count is defined as the product of cut-off values of the 10th percentile as defined by Cooper et al.

for concentration, and percentage of progressive spermatozoa of the ejaculate, divided by 100%. 5 Binomial test with

the hypothesized proportion of 0.25. Clopper-Pearson binomial proportion confidence intervals are shown.

MALE FERTILITY AFTER ADULT DIFFERENTIATED THYROID CARCINOMA

121

5

Supplemental Table 4. Associations between Treatment and Semen Characteristics in Participants treated

with Radioactive Iodine for Differentiated Thyroid Carcinoma

Volume <10th

percentile

n = 5

Volume ≥10th

percentile

n = 46

P Value

Age upon Evaluation, years 49.6 (34.9-55.0) 40.4 (33.8-48.6) 0.2411

Body mass index2, kg/m2 22.2 (19.5-24.7) 27.1 (24.4-29.9) 0.0091

Follow-up since last RAI treatment, years 10.5 (4.3-16.0) 5.6 (3.0-8.5) 0.1281

Cumulative RAI dose, mCi 300.0 (175.0-411.2) 200.0 (149.7-300.0) 0.2381

Number of RAI administrations, n (%) 0.6683

Single administration 2 (40.0) 24 (52.2)

Multiple administrations 3 (60.0) 22 (47.8)

Concentration <10th

percentile

n = 8

Concentration ≥10th

percentile

n = 43

P Value

Age upon Evaluation, years 34.4 (28.9-42.2) 42.0 (36.1-49.6) 0.1141

Body mass index2, kg/m2 25.3 (23.4-27.8) 27.1 (24.2-30.2) 0.3581

Follow-up since last RAI treatment, years 6.1 (2.3-11.3) 5.8 (3.5-9.5) 0.9171

Cumulative RAI dose, mCi 299.0 (212.5-421.5) 200.0 (148.6-300.0) 0.1311

Number of RAI administrations, n (%) 0.1403

Single administration 2 (25.0) 24 (55.8)

Multiple administrations 6 (75.0) 19 (44.2)

A+B Motility <10th

percentile

n = 7

A+B Motility ≥10th

percentile

n = 33

P Value

Age upon Evaluation, years 43.0 (34.7-46.7) 37.2 (33.1-44.9) 0.5331

Body mass index2, kg/m2 27.2 (23.3-32.1) 25.9 (24.3-28.4) 0.9851

Follow-up since last RAI treatment, years 3.5 (2.1-4.6) 5.7 (3.0-7.2) 0.1051

Cumulative RAI dose, mCi 300.0 (200.0-300.0) 200.0 (149.0-300.0) 0.1901

Number of RAI administrations, n (%) 0.4073

Single administration 2 (28.6) 18 (54.5)

Multiple administrations 5 (71.4) 15 (45.5)

Total motile

sperm count

<10th percentile

n = 4

Total motile

sperm count

≥10th percentile

n = 36

P Value

Age upon Evaluation, years 38.1 (25.0-52.7) 38.1 (33.5-45.3) 0.8221

Body mass index2, kg/m2 24.3 (21.5-27.1) 27.0 (24.2-28.8) 0.1991

Follow-up since last RAI treatment, years 4.1 (2.2-6.2) 5.0 (2.9-7.3) 0.3921

Cumulative RAI dose, mCi 299.0 (224.5-300.0) 200.0 (150.0-300.0) 0.4391

CHAPTER 5

122

5

Supplemental Table 4. (continued)

Total motile

sperm count

<10th percentile

n = 4

Total motile

sperm count

≥10th percentile

n = 36

P Value

Number of RAI administrations, n (%) 0.6053

Single administration 1 (25.0) 19 (52.8)

Multiple administrations 3 (75.0) 17 (47.2)

Ever conceived

child(ren)

n = 30

Never conceived

child(ren)

n = 20

P Value

Age upon Evaluation, years 44.7 (37.1-49.8) 33.6 (26.0-45.5) 0.0051

Body mass index2, kg/m2 27.1 (23.9-29.8) 25.4 (24.1-29.4) 0.7761

Follow-up since last RAI treatment, years 6.6 (3.7-10.4) 4.6 (2.5-6.4) 0.1781

Cumulative RAI dose, mCi 199.9 (142.8-300.0) 293.0 (150.0-340.5) 0.1061

Number of RAI administrations, n (%) 0.2484

Single administration 17 (56.7) 8 (40.0)

Multiple administrations 13 (43.3) 12 (60.0)

Numbers are shown as median (interquartile range). For A+B motility (progressively motile spermatozoa) and total

motile sperm count n=40; in one laboratory, evaluation of motility at 37°C was not possible and these cases were

excluded from this analysis (n=11). P values in bold indicate a statistically significant difference (P < 0.05).

Abbreviations: RAI, radioactive iodine.1 Mann-Whitney U test. 2 n=49; one participant did not complete the questionnaire; one did not report his current

weight. 3 Fischer’s Exact test. 4 Pearson Chi-Square test (Asymptotic Significance).

Chapter 6

Chapter 6

CHAPTER 6

Cardiac dysfunction in survivors of

pediatric differentiated thyroid carcinoma

Antoinette D. Reichert*, Marloes Nies*, Wim J.E. Tissing, Anneke C. Muller Kobold, Mariëlle S. Klein Hesselink, Adrienne H. Brouwers, Bas Havekes, Marry M. van den Heuvel-Eibrink, Helena J.H. van der Pal, John T.M. Plukker, Hanneke M. van Santen, Johannes G.M. Burgerhof, Peter van der Meer, Eleonora P.M. Corssmit, Romana T. Netea-Maier, Robin P. Peeters, Eveline W.C.M. van Dam, Gianni Bocca, and Thera P. Links

*These authors contributed equally to this study.

Submitted

CHAPTER 6

126

6

ABSTRACTImportancePediatric differentiated thyroid cancer (DTC) has an excellent prognosis, requiring focus on late effects of treatment. In an initial cardiac evaluation one in five survivors were found to have diastolic dysfunction. Compared to age- and sex-matched controls, diastolic function was significantly worse in survivors of pediatric DTC.

ObjectiveTo determine the clinical course of the diastolic dysfunction in survivors of pediatric DTC.

DesignThis follow-up study conducted between 2018 and 2020, re-evaluated survivors initially studied between 2012 and 2014.

SettingNationwide multicenter study in the Netherlands.

ParticipantsThis evaluation was completed in 47 (71.2%) of 66 long-term survivors of pediatric DTC who had completed the first cardiac evaluation within the previous study.

Main outcome and measuresThe primary endpoint was diastolic cardiac function (depicted by the mean of the early diastolic septal and early diastolic lateral tissue velocity [e’ mean]) assessed using echocardiography. Secondary endpoints were other echocardiographical parameters, plasma biomarkers (e.g. N-terminal pro–B-type natriuretic peptide, NT-proBNP), and survivors’ cardiovascular risk factors.

ResultsOf the 47 survivors completing both cardiac evaluations, 41 were women (87.2%). The median age was 39.8 (range 18.8-60.3) years and the median follow-up after initial diagnosis was 23.4 (range 10.2-48.8) years. Between the first and second evaluation, the e’ mean significantly declined with 2.1 cm/s (SD±2.3 cm/s, P<0.001). The early diastolic septal tissue velocity (e’ septal) and the early diastolic lateral tissue velocity (e’ lateral) also significantly declined (-2.1±2.3 cm/s, P<0.001 and -1.7±2.9 cm/s, P<0.001, respectively). The median left ventricular ejection fraction (LVEF) did not significantly differ between the two evaluations (58.0% vs. 59.0%, P=0.084). The median NT-proBNP level significantly increased between the two evaluations (51.5 to 62.5 ng/L, P=0.023). In the best explanatory model of the multivariate linear regression analysis, body

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

127

6

mass index and age were significantly associated with e’ mean (β coefficient -0.169 [95% confidence interval, CI -0.292;-0.047], P=0.008 and β coefficient -0.177 [95% CI -0.240;-0.113], P<0.001, respectively).

Conclusions and relevanceDiastolic function continues to decline significantly in survivors of pediatric DTC. This finding requires further follow-up to assess its clinical consequences.

CHAPTER 6

128

6

INTRODUCTIONWorldwide, the incidence of pediatric differentiated thyroid carcinoma (DTC) shows a sustained increase (1). Fortunately, the prognosis is excellent with a 15-year overall survival rate exceeding 95% (2) .

In survivors of adult DTC, thyroid-stimulating hormone (TSH) suppression following initial therapy is associated with diastolic dysfunction and an increased risk of cardiovascular disease and cardiovascular mortality (3, 4). In a cross-sectional cardiac evaluation of long-term survivors of pediatric DTC, one out of five survivors had diastolic dysfunction without cardiac arrhythmias or overt systolic dysfunction (5). Compared to sex- and age-matched controls, diastolic function of survivors was significantly worse.

We aimed to assess the clinical course of this finding by evaluating the five-year follow-up cardiac (diastolic) function in these long-term survivors of pediatric DTC.

METHODSAll 66 survivors of pediatric DTC participating in the initial evaluation were invited to complete a second evaluation (5). This five-year follow-up evaluation consisted of a physical examination (blood pressure, waist and hip circumference, heart rate, length, and body weight), measurement of serum biomarkers (cholesterol, glucose, and N-terminal pro–B-type natriuretic peptide [NT-proBNP]) and echocardiography. Smoking status was obtained from a questionnaire completed by the survivor. Details regarding treatment and medical history were obtained from survivors’ medical records. The echocardiographic evaluation consisted of systolic and diastolic function, left ventricle ejection fraction (LVEF), dimensions, wall thickness, tissue Doppler I, and tissue velocity imaging. Diastolic function was marked by the diastolic septal (e’ septal) and lateral tissue velocity (e’ lateral), and the E/A ratio. Difference values between both evaluations are calculated as second minus first evaluation. Diastolic function was considered abnormal when values were less than 2 SD of the mean age-adjusted reference data (6). The mean TSH level represents the area under the curve (AUC) for survivors with at least one available TSH measurement per year from diagnosis to second evaluation. A univariate linear regression analysis was performed to evaluate whether attained age, sex, smoking, diastolic blood pressure, waist circumference, body mass index (BMI) or TSH were significant predictors of diastolic function (e’ mean).(7) Variables that were significantly associated (P<0.1) with diastolic function were included in a multivariate analysis. Based on reported literature, TSH and sex were added to the multivariate model.(7, 8) Missing or unknown cases were excluded from statistical testing. Two-sided P values <0.05 were considered statistically significant. Performed statistical tests are described in the table legends. IBM SPSS Statistics (version 23.0. Armonk, NY: IBM Corp) was used for statistical analysis. This study was

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

129

6

approved by the local institutional review board and registered in the Netherlands Trial Register (Trial NL3280). Written informed consent was obtained from all survivors.

RESULTS The second assessment was completed for 47/66 of the initially evaluated pediatric DTC survivors. Comparing clinical and treatment characteristics, and cardiovascular risk factors, only systolic blood pressure and e’ lateral significantly differed between the participating and the non-participating survivors (114.7 vs. 120.0 mmHg, respectively; P=0.049 and 17.0 vs. 15.9 cm/s, respectively; P=0.035).

Of the 47 participating survivors, 41 were women (87.2%). The median age of survivors upon second evaluation was 39.8 (range 18.8-60.3) years, and median follow-up was 23.4 (range 10.2-48.8) years after initial diagnosis (Table 1).

Table 1 shows the treatment characteristics, physical examination, serum biomarkers, and smoking status. In the five-year follow-up period, median NT-proBNP levels significantly increased from 51.5 to 62.5 ng/L (P=0.023).

Echocardiographic characteristics are shown in Table 2. For diastolic function, the mean e’ septal declined with 2.1 cm/s (SD±2.3 cm/s, P<0.001), the e’ lateral declined with 1.7 cm/s (SD±2.9 cm/s, P<0.001), and the E/A ratio declined with 0.2 (SD±0.4, P=0.002) between the first and the second evaluation. The LVEF did not significantly change between the first and second evaluations (58.0%, range 51-73% vs. 59.0%, range 51-70%, respectively; P=0.084). For e’ lateral as well as e’ septal, 20 survivors showed a decline of more than 2 cm/s, and three of the 20 had a decline greater than 6 cm/s (Figure 1).

Based on age-adjusted reference ranges (6), diastolic dysfunction in the second evaluation was present in 23.9% for the e’ septal, in 40.4% for the e’ lateral, and 12.8% for the E/A ratio, compared to 13.0%, 19.1%, and 14.9% upon first evaluation, respectively.

In the best explanatory multivariate model, body mass index and attained age were significantly associated with e’ mean (β coefficient -0.169 [95% confidence interval, CI -0.292;-0.047], P=0.008, and β coefficient -0.177 [95% CI -0.240;-0.113], P<0.001, respectively). The median AUC of the TSH values was not significantly associated with the e’ mean (Supplemental Table 1).

DISCUSSIONIn this longitudinal study of cardiac function in these relatively young survivors of childhood DTC, we found a deterioration of the already impaired diastolic function. None of the survivors currently had a known medical history of cardiovascular disease. To assess the clinical consequences, these findings require continued follow-up, since the presence of diastolic dysfunction is a known risk factor for heart failure and accelerated cardiac aging (7-10).

CHAPTER 6

130

6

Table 1. Clinical and treatment characteristics and cardiovascular risk factors in survivors of pediatric

differentiated thyroid carcinoma

Survivors

n=47

Characteristic Characteristic

Sex (female), n (%) 41 (87.2) TNM stage, n (%)Age upon diagnosis, y 16.1 (8.7–18.9) T T1-T2 27 (57.4)

Follow-up since diagnosis, y 23.4 (10.2–48.8) T3-T4 11 (23.4)

Histology, n (%) Tx 9 (19.1)

Papillary 40 (85.1) N N0 20 (42.6)

Follicular 7 (14.9) N1a-N1b 22 (46.8)

Total thyroidectomy, n (%) 47 (100.0) Nx 5 (10.6)

Radioiodine treatment, n (%) 45 (95.7) M M0 40 (85.1)

Cumulative activitya, GBq 5.8 (1.3–35.2) M1b 2 (4.3)

TSH levels, mU/L Mx 5 (10.6)

Total follow-up period, AUCc 0.5 (0.2–2.8) Disease remission, n (%) 47 (100.0)

Till first evaluation, AUCd 0.6 (0.1–3.9) Time between evaluations, y 5.5 (4.7–6.7)

Cardiovascular risk factor First evaluation

median (range)

Second evaluation

median (range)

Difference

median (IQR)

P Value

Age upon evaluation, y 34.3 (18.8–60.3) 39.8 (24.6–65.9) 5.5 (5.2–6.2) n.a.

Heart rate, bpm 68.2 (43.5–93.7) 66.0 (52.7–88.8) 2.1 (-7.8–7.1) 0.878e

Body mass index, kg/m2 24.0 (18.9–40.4) 25.6 (19.3–39.4) 1.5 (-0.1–2.4) <0.001e

Overweight (BMI≥25 kg/m2), n (%) 21 (44.7) 24 (51.1) 0.219f

Body surface area, m2 1.8 (1.6–2.4) 1.9 (1.6–2.4) 0.0 (-0.0–0.1)g <0.001e

Smokingh, n (%)No 32 (74.4) 31 (72.1) 0.317i

Current 1 (2.3) 1 (2.3)

Past 10 (23.3) 11 (25.6)

NT-proBNPj, ng/L 51.5 (7.0–225.0) 62.5 (5.0–653.0) 8.5 (-12.0–31.0) 0.023e

NT-proBNP ≥125 ng/L, n (%) 5 (10.9) 8 (17.4) 0.508f.

Fasting glucosek, mmol/L 4.9 (4.2–5.8) 5.3 (3.7–6.9) 0.4 (0.0–0.7) <0.001e

Cholesterol, mmol/L 4.6 (3.2–7.1) 5.1 (3.8–7.6) 0.3 (-0.1–0.8) 0.002e

Systolic blood pressurel, mmHg 120.0 (100.0–145.0) 121.0 (98.0–173.0) 0.1 (-5.4–10.9) 0.249e

Diastolic blood pressurel, mmHg 77.0 (53.0–100.0) 78.0 (57.0–105.0) 1.0 (-1.5–5.9) 0.056e

Waist circumferencem, cm 83.0 (67.0–123.0) 87.0 (67.0–123.0) 1.5 (-2.5–7.1) 0.044e

Hip circumferencen, cm 102.0 (77.0–134.0) 104.0 (79.0–132.0) 0.5 (-2.5–5.0) 0.404e

History of cardiovascular

disease, n (%)

0.0 (0.0) 0.0 (0.0) 0.0 (0.0) 1.000e

In these survivors of pediatric DTC we found no treatment-related risk factors to be associated with diastolic function. Moreover, we found no clinically relevant increase in cardiovascular risk factors between the two evaluations. However, in survivors of adult DTC, the prolonged subclinical hyperthyroidism induced by TSH

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

131

6

Table 1. (continued)

P Values in bold indicate a significant difference (<0.05). Variables are shown as median (range). Difference values

between the two evaluations are calculated as second evaluation minus first evaluation. Difference values are shown as

median (interquartile range). Abbreviations: IQR, interquartile range; TSH, thyroid stimulating hormone; AUC, mean

area under the curve; and TNM, tumor node metastasis; y, years; n.a., not applicable; bpm, beats per minute; and NT-

proBNP, N-terminal pro–B-type natriuretic peptide. a 2 survivors had no radioiodine treatment; n=45. b Both survivors

had lung metastases. c n=10, 37 survivors had incomplete TSH levels. d n=22 survivors, 25 survivors had incomplete TSH

levels. e Wilcoxon Signed-Rank test. f McNemar test. g Unrounded median difference: 0.04 (-0.01–0.08) m2. h n=43; 4

survivors excluded from analysis for inconsistent answers. i McNemar-Bowker test. j n=46; blood sample too small in

one participant. k n=45; blood sample too small in one participant and non-fasting glucose in one participant. l n=44;

three survivors had no complete physical examination. For two survivors, blood pressure at first or second evaluation

measured using right arm only. This measurement taken into analysis. m n=42; waist circumference was not measured

in four survivors and measured incorrectly in one survivor. n n=43; in four survivors hip circumference not measured.

Figure 1. Difference (delta) in diastolic function in 47 survivors of childhood differentiated thyroid

carcinoma over a median period of five years. Dashed vertical line displays difference of zero cm/s

between evaluations. Negative values indicate a decrease in diastolic function. Graph A. Difference in

early diastolic lateral (grey bars, n=47) and septal (black bars, n=46) tissue velocity in cm/s, plotted against

frequency. Graph B. Difference in mean of early diastolic lateral and septal tissue velocity in cm/s (black

bars, n=46), plotted against frequency. Abbreviations: e’ lateral, early diastolic lateral tissue velocity; and

e’ septal, early diastolic septal tissue velocity. For one survivor, data regarding the e’ septal was missing.

CHAPTER 6

132

6

suppression therapy is associated with disturbed myocardial relaxation, which can eventually result in diastolic dysfunction (3, 11), and an increased risk of cardiovascular disease and mortality (12-15).

LimitationsThe retrospective aspect of this study may be considered a limitation, creating a limited availability of TSH values and resulting in an incomplete representation of

Table 2. Echocardiographic measurements of survivors of pediatric differentiated thyroid carcinoma

Variable Survivors

n=47

Difference

mean ± SD

P Value

First

evaluation

median (range)

Second

evaluation

median (range)

Dimensions and volumes Left ventricular end diastolic

diameter, mm

47.0 (39.0–58.0) 47.0 (39.0–54.0) -1.4 ± 3.2 0.006

Left ventricular mass at end diastole, g 121.0 (71.0–226.0) 124.0 (78.0–208.0) 7.2 ± 24.7 0.052

Left ventricular mass at end diastole

index, g/m2

66.2 (41.0–106.0) 65.9 (44.9–114.4) 2.5 ± 13.2 0.196

Left ventricular end systolic

diameter, mm

31.0 (22.0–46.0) 30.0 (23.0–38.0) -1.5 ± 4.4 0.026

Left atrial volume indexa, mL/m2 28.1 (12.0–46.0) 28.5 (16.0–38.0) 0.5 ± 6.3 0.596

Systolic functionLeft ventricular ejection fraction, n (%) 58.0 (51.0–73.0) 59.0 (51.0-70.0) 1.5 ± 5.6 0.084

Diastolic functionEarly diastolic mitral valve inflow, m/s 0.8 (0.4–1.2) 0.8 (0.5–1.2) -0.1 ± 0.1b 0.009Late diastolic mitral valve inflow, m/s 0.5 (0.3–0.9) 0.6 (0.3–1.1) 0.0 ± 0.1c 0.124

Deceleration time of mitral valve

early inflow, m/s

195.4 (137.8–329.4) 184.9 (76.0–337.3) -11.4 ± 55.2 0.163

E/A ratio 1.6 (0.7–3.3) 1.4 (0.7–3.2) -0.2 ± 0.4 0.002E/e’ ratiod 6.1 (3.7–9.5) 5.9 (2.8–9.3) -0.3 ± 1.4 0.133

Early diastolic septal tissue

velocityd, cm/s

12.6 (5.6–19.9) 10.2 (5.1–15.7) -2.1 ± 2.3 <0.001

Early diastolic lateral tissue

velocity, cm/s

15.9 (6.1–24.6) 13.4 (5.8–23.3) -1.7 ± 2.9 <0.001

Mean of septal and lateral early

diastolic tissue velocityd, cm/s

14.5 (6.3–20.4) 11.7 (5.5–18.2) -1.9 ± 2.2 <0.001

P Values in bold indicate a significant difference (<0.05). Variables are shown as median (range). Difference values

between the two evaluations are calculated as second evaluation minus first evaluation. Difference values are shown

as mean ± standard deviation. Abbreviation: SD, standard deviation. a Left atrial volume index was available for 40 patients. b Unrounded mean difference: -0.06 m/s. c Unrounded mean

difference: 0.003 m/s. d n=46; for one survivor the required echocardiographic image was missing.

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

133

6

actual TSH levels and suppression. Another limitation includes the lack of age- and gender-specific reference values for the decline of diastolic function over time.

ConclusionThis study showed a significant decline in diastolic function in survivors of pediatric DTC after a median follow-up of 23 years. Although the pathophysiological mechanisms and clinical consequences have yet to be assessed, this outcome requires attention, since diastolic dysfunction is associated with accelerated cardiac aging and the survivors included in our study had a median age of only 40 years. Further research and follow-up is needed.

CHAPTER 6

134

6

ACKNOWLEDGEMENTSWe thank J. G. Jongsma, C. Hooghiemstra, and H. Renkema, Groningen Imaging Core Laboratory, for their help with the echocardiographic measurements. We thank the cardiology departments of the participating centers for their collaboration.

AUTHOR CONTRIBUTIONSReichert and Nies had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: Reichert, Nies, Tissing, Burgerhof, Bocca, Links. Acquisition, analysis, or interpretation of data: Reichert, Nies, Tissing, Muller Kobold, Klein Hesselink, Burgerhof, van der Meer, Corssmit, Netea-Maier, Peeters, van Dam, Bocca, Links. Drafting of the manuscript: Reichert, Nies, Tissing, Bocca, Links. Critical revision of the manuscript for important intellectual content: Muller Kobold, Klein Hesselink, Brouwers, Havekes, van den Heuvel-Eibrink, van der Pal, Plukker, Ronckers, van Santen, Corssmit, Netea-Maier, Peeters, van Dam. Statistical analysis: Reichert, Nies, Burgerhof. Obtained funding: Nies, Tissing, Klein Hesselink, Bocca, Links. Administrative, technical, or material support: Reichert, Nies, Tissing, Muller Kobold, Klein Hesselink, Burgerhof, van der Meer, Corssmit, Netea-Maier, Peeters, van Dam, Bocca, Links. Study supervision: Tissing, van der Meer, Bocca, Links. Final approval of the version to be published: all authors.

CONFLICTS OF INTEREST DISCLOSURESThe authors declare that there is no conflict of interest.

FUNDING/SUPPORTThis study was funded by Stichting Kinderen Kankervrij (Foundation Children Cancer-Free, the Netherlands, project no. 81), the Van der Meer-Boerema Foundation, and the Junior Scientific Masterclass Groningen.

TRIAL REGISTRATIONThis follow-up study was registered in the Netherlands Trial Register under number NL3280 (www.trialregister.nl/trial/3280).

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

135

6

REFERENCES1. Vergamini LB, Frazier AL, Abrantes FL, Ribeiro KB, Rodriguez-Galindo C. Increase in the incidence of differentiated

thyroid carcinoma in children, adolescents, and young adults: a population-based study. J Pediatr. 2014;164(6):1481-1485.

2. Dermody S, Walls A, Harley EH. Pediatric thyroid cancer: an update from the SEER database 2007–2012. Int J of

Pediatr Otorhinolaryngol. 2016;89:121-126.

3. Smit JWA, Eustatia-Rutten CFA, Corssmit EPM, Pereira AM, Frölich M, Bleeker GB, Holman ER, van der Wall EE,

Romijn JA, Bax JJ. Reversible diastolic dysfunction after long-term exogenous subclinical hyperthyroidism:

a randomized, placebo-controlled study. J Clin Endocrinol Metab. 2005;90(11):6041-6047.

4. Klein Hesselink EN, Klein Hesselink MS, de Bock GH, Gansevoort RT, Bakker SJL, Vredeveld EJ, van der Horst-Schrivers

ANA, van der Horst ICC, Kamphuisen PW, Plukker JTM, Links TP, Lefrandt JD. Long-term cardiovascular mortality in

patients with differentiated thyroid carcinoma: an observational study. J Clin Oncol. 2013;31(32):4046-4053.

5. Klein Hesselink MS, Bocca G, Hummel YM, Brouwers AH, Burgerhof JGM, van Dam EWCM, Gietema JA, Havekes

B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker

JTM, Ronckers CM, van Santen HM, van der Meer P, Links TP, Tissing WJE. Diastolic dysfunction is common in

survivors of pediatric differentiated thyroid carcinoma. Thyroid. 2017;27(12):1481-1489.

6. Nagueh SF, Appleton CP, Gillebert TC, Marino PN, Oh JK, Smiseth OA, Waggoner AD, Flachskampf FA, Pellikka PA,

Evangelista A. Recommendations for the evaluation of left ventricular diastolic function by echocardiography. J

Am Soc Echocardiogr. 2009;22(2):107-133.

7. Wan SH, Vogel MW, Chen HH. Pre-clinical diastolic dysfunction. J Am Coll Cardiol. 2014;63(5):407-416.

8. Malhotra Y, Kaushik RM, Kaushik R. Echocardiographic evaluation of left ventricular diastolic dysfunction in

subclinical hypothyroidism: A case-control study. Endocr Res. 2017;42(3):198-208.

9. Bella JN, Palmieri V, Roman MJ, Liu JE, Welty TK, Lee ET, Fabsitz RR, Howard BV, Devereux RB. Mitral ratio of peak

early to late diastolic filling velocity as a predictor of mortality in middle-aged and elderly adults: the strong heart

study. Circulation. 2002;105(16):1928-1933.

10. Halley CM, Houghtaling PL, Khalil MK, Thomas JD, Jaber WA. Mortality rate in patients with diastolic dysfunction

and normal systolic function. Arch Intern Med. 2011;171(12):1082-1087.

11. Fazio S, Biondi B, Carella C, Sabatini D, Cittadini A, Panza N, Lombardi G, Saccà L. Diastolic dysfunction in patients

on thyroid-stimulating hormone suppressive therapy with levothyroxine: Beneficial effect of beta-blockade. J Clin

Endocrinol Metab. 1995;80(7):2222-2226.

12. Collet T-H, Gussekloo J, Bauer DC, den Elzen WPJ, Cappola AR, Balmer P, Iervasi G, Åsvold BO, Sgarbi JA, Völzke

H, Gencer B, Maciel RMB, Molinaro S, Bremner A, Luben RN, Maisonneuve P, Cornuz J, Newman AB, Khaw K-T,

Westendorp RGJ, Franklyn JA, Vittinghoff E, Walsh JP, Rodondi N, Thyroid Studies C. Subclinical hyperthyroidism

and the risk of coronary heart disease and mortality. Arch Intern Med. 2012;172(10):799-809.

13. Klein Hesselink EN, van der Horst-Schrivers ANA, van der Horst ICC, Bakker SJL, Muller Kobold AC, Brouwers

AH, de Bock GH, Gietema JA, Dullaart RPF, Links TP, Lefrandt JD. NT-proBNP is increased in differentiated thyroid

carcinoma patients and may predict cardiovascular risk. Clin Biochem. 2017;50(12):696-702.

14. Pajamäki N, Metso S, Hakala T, Ebeling T, Huhtala H, Ryödi E, Sand J, Jukkola-Vuorinen A, Kellokumpu-Lehtinen

P-L, Jaatinen P. Long-term cardiovascular morbidity and mortality in patients treated for differentiated thyroid

cancer. Clin Endocrinol (Oxf). 2018;88(2):303-310.

15. Wang R, Yang L, Jin S, Han X, Liu B. Thyroid stimulating hormone suppression time on cardiac function of patients

with differentiated thyroid carcinoma. Cancer Cell Int. 2018;18:189-189.

CHAPTER 6

136

6

Supplemental Table 1. Univariate and multivariate linear regression models for diastolic function (e’ mean)

among 47 survivors of pediatric DTC

Variable Coefficient βa. 95% CI P-value R² Constant B

Univariate models    

Attained age, years -0.199 [-0.264 ; -0.133] <0.001 0.462 20.032

Sex -1.072 [-3.917 ; 1.773] 0.452 0.013 12.239

TSH- Till first evaluation, mU/L

(n=22)

-0.948 [-2.277 ; 0.381] 0.152 0.100 14.297

TSH- Between evaluations,

mU/L (n=19)

0.070 [-2.023 ; 2.163] 0.944 0.000 12.046

Diastolic blood pressure, mmHg

(n=44)

-0.169 [-0.267 ; -0.070] 0.001 0.225 25.240

Current smoking 5.134 [-1.358 ; 11.626] 0.118 0.056 12.016

BMI, kg/m2 -0.258 [-0.410 ; -0.106] 0.001 0.211 19.021

Waist circumference, mm (n=42) -0.109 [-0.172 ; -0.046] <0.001 0.237 21.784

Glucose, mmol/L (n=45) -0.031 [-1.902 ; 1.840] 0.973 0.000 12.306

Cholesterol, mmol/L (n=45) -1.588 [-2.681 ; -0.495] 0.005 0.166 20.248

Multivariate modelsModel 1 (n=18) 0.112 0.588 26.060

Attained age -0.168 [-0.315 ; -0.021] 0.028Diastolic blood pressure -0.028 [-0.240 ; 0.184] 0.774

BMI -0.134 [-0.700 ; 0.432] 0.613

Sex -1.651 [-6.056 ; 2.754] 0.427

TSH- Till first evaluation -0.102 [-1.714 ; 1.510] 0.891

Cholesterol -0.173 [-2.302 ; 1.955] 0.8861

Waist circumference -0.003 [-0.227 ; 0.221] 0.978

Model 2 (n=20) 0.030 0.588 26.027

Attained age -0.169 [-0.294 ; -0.043] 0.012Diastolic blood pressure -0.029 [-0.210 ; 0.152] 0.739

BMI -0.140 [-0.423 ; 0.143] 0.307

Sex -1.657 [-5.444 ; 2.131] 0.364

TSH- Till first evaluation -0.101 [-1.488 ; 1.287] 0.879

Cholesterol -0.177 [-1.997 ; 1.642] 0.838

Model 3 (n=42) <0.001 0.587 26.251

Attained age -0.171 [-0.241 ; -0.101] <0.001Diastolic blood pressure -0.029 [-0.134 ; 0.076] 0.580

BMI -0.139 [-0.303 ; 0.026] 0.096

Sex -1.596 [-3.745 ; 0.553] 0.141

Cholesterol -0.223 [-1.216 ; 0.771] 0.653

Model 4 (n=42) <0.001 0.585 25.739

Attained age -0.175 [-0.242 ; -0.107] <0.001Diastolic blood pressure -0.032 [-0.135 ; 0.071] 0.537

SUPPLEMENTARY DATA

CARDIAC FUNCTION AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

137

6

Supplemental Table 1. (continued)

Variable Coefficient βa. 95% CI P-value R² Constant B

BMI -0.148 [-0.305 ; 0.009] 0.064

Sex -1.732 [-3.770 ; 0.307] 0.094

Model 5 (n=45) <0.001 0.580 24.294

Attained age -0.180 [-0.242 ; -0.118] <0.001BMI -0.177 [-0.296 ; -0.058] 0.004Sex -1.805 [-3.735 ; 0.125] 0.066

Model 6 (n=45) <0.001 0.545 23.719

Attained age -0.177 [-0.240 ; -0.113] <0.001BMI -0.169 [-0.292 ; -0.047] 0.008

Abbreviations: DTC, Differentiated thyroid carcinoma; CI, confidence interval; TSH, Thyroid stimulating hormone;

BMI, body mass index. P Values in bold represent statistically significant values (<0.05). Variables in italic in

the multivariate models represent the variable excluded in the successive model. a. Unstandardized coefficient β.

Chapter 7

Chapter 7

CHAPTER 7

Long-term quality of life in adult survivors of pediatric

differentiated thyroid carcinoma

Marloes Nies*, Mariëlle S. Klein Hesselink*, Gea A. Huizinga, Esther Sulkers, Adrienne H. Brouwers, Johannes G.M. Burgerhof, Eveline W.C.M. van Dam, Bas Havekes, Marry M. van den Heuvel-Eibrink, Eleonora P.M. Corssmit, Leontien C.M. Kremer, Romana T. Netea-Maier, Heleen J.H. van der Pal, Robin P. Peeters, John T.M. Plukker, Cécile M. Ronckers, Hanneke M. van Santen, Wim J.E. Tissing, Thera P. Links, and Gianni Bocca

*These authors contributed equally to this study.

J Clin Endocrinol Metab. 2017, 102: 1218–1226.

CHAPTER 7

140

7

ABSTRACT ContextLittle is known about long-term quality of life (QoL) of survivors of pediatric differentiated thyroid carcinoma (DTC). Therefore, this study aimed to evaluate generic health-related QoL (HRQoL), fatigue, anxiety, and depression in these survivors compared with matched controls, and to evaluate thyroid cancer-specific HRQoL in survivors only.

DesignSurvivors diagnosed between 1970 and 2013 at age ≤18 years, were included. Exclusion criteria were a follow-up <5 years, attained age <18 years, or diagnosis of DTC as a second malignant neoplasm. Controls were matched by age, sex, and socioeconomic status. Survivors and controls were asked to complete 3 questionnaires [Short-Form 36 (HRQoL), Multidimensional Fatigue Inventory 20 (fatigue), and Hospital Anxiety and Depression scale (anxiety/depression)]. Survivors completed a thyroid cancer–specific HRQoL questionnaire.

ResultsSixty-seven survivors and 56 controls. Median age of survivors at evaluation was 34.2 years (range 18.8 to 61.7 years). Median follow-up was 17.8 years (range 5.0 to 44.7 years). On most QoL subscales, scores of survivors and controls did not differ significantly. However, survivors had more physical problems (P = 0.031), role limitations due to physical problems (P = 0.021), and mental fatigue (P = 0.016) than controls. Some thyroid cancer-specific complaints (e.g. sensory complaints and chilliness) were present in survivors. Unemployment and more extensive disease or treatment characteristics were most frequently associated with worse QoL.

ConclusionsOverall, long-term QoL in survivors of pediatric DTC was normal. Survivors experienced mild impairment of QoL in some domains (physical problems, mental fatigue, and various thyroid cancer-specific complaints). Factors possibly affecting QoL need further exploration.

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

141

7

INTRODUCTIONThe diagnosis and treatment of childhood cancer constitute a major life event that may influence psychosocial functioning and quality of life (QoL) long after initial treatment has been completed. Pediatric differentiated thyroid carcinoma (DTC) is a rare malignancy with an excellent survival rate (1, 2). The incidence of this disease is rising (3, 4). At present there is an increased focus on the long-term consequences of this disease and its treatment. After initial treatment, which most often consists of a total thyroidectomy followed by radioactive iodine (131I) administration, life-long follow-up is initiated (5). Moreover, about a third of the pediatric DTC survivors in The Netherlands have postoperative complications, including permanent hypoparathyroidism, which also require medical support (2). Both hypothyroidism and hypoparathyroidism have been described as negatively affecting the QoL of DTC survivors (6, 7).

The available data on QoL in survivors of pediatric DTC are limited. Most studies evaluate QoL in survivors of adolescent and adult DTC. QoL in survivors of adolescent DTC was found to be normal compared to control groups (8, 9). Survivors of adult DTC reported lower health-related QoL (HRQoL) and higher levels of fatigue, anxiety, and depression compared with healthy controls (5, 10-12). For DTC survivors, thyroid cancer-specific complaints may arise after treatment and during follow-up. Most long-term survivors of adult DTC with a median follow-up period of 9.6 years experienced disease specific symptoms (13). In survivors of pediatric DTC, thyroid cancer-specific complaints have not yet been evaluated.

QoL studies performed in survivors of other childhood cancers showed conflicting results. QoL varied from no differences to lower QoL in survivors, compared to a control or comparison group; this may be explained by the differences in specific clinical characteristics of the different types of cancers in which this has been studied (14-20).

Several specific survivor characteristics may influence QoL during follow-up. QoL may improve in survivors of adult DTC after a longer time following diagnosis, especially ≥5 years after diagnosis (10, 21). Employment and a higher educational level were found to be associated with better QoL in survivors of adult DTC (12, 21, 22). Young adult survivors reported other thyroid cancer-specific complaints than did older survivors. For example, young adult survivors reported more scar complaints, but less voice complaints (22). QoL in DTC survivors seems to be independent of thyrotropin (TSH) levels (8, 10).

So far, information is limited on long-term QoL in survivors of pediatric DTC. Therefore, the first aim of our study was to evaluate QoL by means of the self-reported levels of generic HRQoL, fatigue, anxiety, and depression in adult survivors of pediatric DTC compared with those of matched controls. The second aim was to evaluate thyroid cancer-specific HRQoL in adult survivors of pediatric DTC. Our third aim was to evaluate whether long-term generic HRQoL, fatigue, anxiety, depression,

CHAPTER 7

142

7

and thyroid cancer-specific HRQoL were associated with survivor, tumor, treatment, or follow-up characteristics.

MATERIALS AND METHODSSurvivorsAdult survivors of pediatric DTC who participated in a nationwide follow-up study were asked to participate in this substudy (2). Inclusion criteria for the nationwide study were: diagnosis of DTC between January 1970 and December 2013 at the age of ≤18 years, and treatment in The Netherlands [as described earlier (2)]. Exclusion criteria for this substudy were: follow-up <5 years after diagnosis, attained age <18 years, diagnosis of DTC as a second malignant neoplasm, lack of command of the Dutch language, and 131I administration within 3 months before evaluation.

ControlsTo include controls, survivors were asked to approach 1 or 2 persons of similar sex and age (± 5 years of the survivors’ age) at time of follow-up. The only exclusion criterion for controls was having a medical history of malignancy. Preferably, peers were included as controls, trying to match on socioeconomic status. If this was not possible, siblings were allowed to be matched controls.

ParticipantsAll survivors and controls gave informed consent before participating in the study. The Institutional Review Board of the University Medical Center Groningen approved the study on behalf of all participating institutions.

Medical dataData on survivor and tumor characteristics (tumor node metastasis classification, histology, and age at diagnosis), treatment (surgical complications, i.e. hypoparathyroidism and recurrent laryngeal nerve injury, and cumulative 131I dose), and follow-up (follow-up time; outcome: i.e. remission, recurrence or persistent disease; and level of TSH suppression during follow-up) were retrieved from medical records. Data on marital status, job status, and level of education were retrieved from a questionnaire.

QoL assessmentGeneric HRQoL, fatigue, anxiety, and depression questionnaires were assessed for survivors and controls. The thyroid cancer-specific HRQoL questionnaire was assessed for survivors only. The questionnaires delineated below were used to assess QoL.

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

143

7

Short Form 36 The Short Form 36 (SF-36) (23) is a 36-item health survey, validated for the Dutch population (24). This questionnaire measures self-reported HRQoL on 8 domains: physical functioning, role limitations due to physical problems, bodily pain, general health, vitality, social functioning, role limitations due to emotional problems and mental health. Scores of domains are presented on a scale from 0 to 100. A higher score indicates better HR-QoL. The scores of the 8 subscales were converted into the physical and mental component scales by means of the algorithm of Ware and Kosinski (25), based on Dutch norms (24). An example of a question in the SF-36 questionnaire is: “In general, would you say your health is…” Possible answers are “poor,” “fair,” “good,” “very good,” “excellent.”

Multidimensional Fatigue Inventory 20The Multidimensional Fatigue Inventory 20 (MFI-20) (26) self-report instrument was designed to measure fatigue. Fatigue is scored on 5 domains: general fatigue, physical fatigue, mental fatigue, reduced motivation and reduced activity. The MFI-20 was proven to be valid for the Dutch population (26). The total score, ranging from 20 to 100 (higher scores indicating more fatigue), is calculated as the sum of the 5 domains, and each domain (range 4 to 20) is the sum of 4 items. Each domain includes 4 items on a 1 to 5-point scale ranging from “no, that is not true” to “yes, that is true.” An example of an item is “thinking requires effort.”

Hospital Anxiety and Depression Scale The Hospital Anxiety and Depression Scale (HADS) (27) is a 14-item self-assessment scale measuring levels of depression and anxiety. This questionnaire has been validated for Dutch subjects (28). Scores on subscales range from 0 to 21. Scores for the total scale can range from 0 to 42. A higher score indicates a higher level of anxiety and/or depression. Answers can be given on a 4-point Likert scale. For example, for “I feel tense or ‘wound up’,” possible answers are “not at all,” “from time to time, occasionally,” “a lot of the time,” or “most of the time.”

Thyroid cancer-specific HRQoL The thyroid cancer-specific HRQoL questionnaire (THYCA-QoL) (29) was designed to assess treatment- and cancer-specific side effects of thyroid cancer. For a time frame dealing with the previous week (except in the case of the sexual interest item, which refers to the previous 4 weeks) items can be scored on a 4-point scale ranging from 1 (“not at all”) to 4 (“very much”). Items are summarized and translated into 7 scales (neuromuscular, voice, concentration, sympathetic, throat/mouth, psychological, and sensory problems) and 6 single items (problems with scar, felt chilly, tingling hands/feet, gained weight, headache, interest in sex). Scales range from 0 to 100.

CHAPTER 7

144

7

A higher score on this scale indicates more complaints. An example of a question in the THYCA-QoL questionnaire is “Have you had a dry mouth?”

Statistical analysisCharacteristics of survivors included in the QoL study were compared to characteristics of the excluded survivors from the nationwide study and to the eligible, but not included, survivors in the QoL study. Characteristics of survivors and controls were compared to evaluate whether controls were a good comparison group for the included survivors. Scores on HRQoL, fatigue, anxiety, and depression of survivors and matched controls were compared. Scores on thyroid cancer-specific HRQoL were analyzed using descriptive statistics. Associations between survivor, tumor, treatment, and follow-up characteristics and scores on QoL were assessed. Single items of the THYCA-QoL were dichotomized (not at all and a little = no; quite a bit and very much = yes) before testing these associations. Comparisons were made using χ2

tests or a Fisher’s exact test (if >20% of the cells had an expected count of <5) in case of categorical variables. Mann-Whitney U and Kruskal-Wallis tests were performed because normality and homogeneity assumptions of dependent variables had been violated. For analyses of possible correlations between 2 variables with ordinal scales, the Spearman rank correlation coefficient was used. The TSH level for each year of follow-up was expressed as the geometric mean of the observed TSH values during that year, as published previously (30).

Missing or unknown values were excluded (listwise deletion) from analyses or, in case of the 4 questionnaires, imputed using questionnaire-specific guidelines. For SF-36 and the THYCA-QoL, scores were imputed when ≤50% values of the scores for this subscale were missing. For MFI-20 and HADS, scores were imputed when ≥2 values of the subscale were not missing. When these criteria were not met, survivors or controls were excluded from analyses of the relevant subscale. IBM SPSS Statistics for Windows version 23 (IBM, Armonk, NY) was used for statistical analyses. Differences were considered statistically significant at P <0.05. To adjust for the multiple testing, a significance level of 1% (P <0.01) was used for the tested associations between survivor, tumor, treatment, and follow-up characteristics and QoL.

RESULTS Characteristics of survivors of pediatric DTC and matched controlsOut of 105 survivors participating in the nationwide study (2), 75 survivors were eligible for this long-term QoL substudy, 67 of whom (89.3%) agreed to participate. Eight survivors eligible for the QoL study were not included (Figure 1). As expected, survivors of the nationwide study, excluded for the QoL study, and participating survivors in the QoL study differed in age at evaluation (median 19.1 vs. 34.2 years, respectively; P <0.001) and follow-up duration (median 2.8 vs. 17.8 years, respectively.

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

145

7Figure 1. Flowchart of inclusion of adult survivors of pediatric differentiated thyroid carcinoma in

the quality of life study.

Table 1. Characteristics of adult survivors of pediatric differentiated thyroid carcinoma and

matched controls

Characteristic Survivors

n = 67

Controls

n = 56

P Value

Sex, n (%) 0.878a

Female 58 (86.6) 49 (87.5)

Male 9 (13.4) 7 (12.5)

Age at evaluation, years 34.2 (18.8-61.7) 34.0 (19.4-60.2) 0.431b

Age at diagnosis, years 15.8 (7.9-18.8) - n.a.

Follow-up duration, years 17.8 (5.0-44.7) - n.a.

Nationality, n (%) n.a.

Dutch 67 (100) 56 (100)

Marital status, n (%) 0.129a

Relationship 43 (64.2) 43 (76.8)

No relationship 24 (35.8) 13 (23.2)

Completed education, n (%) 0.128a

Low level 16 (23.9) 6 (10.7)

Medium level 25 (37.3) 21 (37.5)

High level 26 (38.8) 29 (51.8)

Employment, n (%) 0.743a

Employed or full-time student 61 (91.0) 50 (89.3)

Unemployed and no full-time student 6 (9.0) 6 (10.7)

Relationship with survivor, n (%) n.a.

Peer - 48 (85.7)

Sibling - 8 (14.3)

Numbers are shown as median (range). Abbreviation: n.a., not applicable. a Chi-square test. b Mann-Whitney U test.

CHAPTER 7

146

7

P <0.001). For all other survivor, tumor, treatment, or follow-up characteristics, survivors of the 3 groups did not differ (Supplemental Table 1). Table 1 shows the demographic characteristics of the survivors and their matched controls. Median age of survivors at evaluation was 34.2 years (range 18.8 to 61.7). Most survivors were female (86.6%), were married or in a relationship (64.2%), and had a paid job or were full-time students (91.0%). Tumor, treatment, and follow-up characteristics of survivors are shown in Supplemental Table 1. Survivors had a median follow-up time of 17.8 years (range 5.0 to 44.7) after diagnosis. All survivors underwent a total thyroidectomy and 131I was administered to 97.0%. Of the survivors, 82.1% remained in remission during follow-up. Ten percent had recurrence of the disease and 7% had persistent disease. The 67 survivors gathered 59 controls. Three controls were not included because matching criteria were not met. The survivor and control groups did not differ significantly with respect to sex, age, marital status, employment/education, nationality, or completed education (Table 1). Almost 14% of the controls were siblings of the survivors.

QoL in adult survivors of pediatric DTC (vs. matched controls)Table 2 summarizes self-reported QoL measurements in survivors and controls. For the subscales of the questionnaires, survivors had significantly lower scores on physical functioning and role limitations due to physical problems as compared with controls (P = 0.031 and P = 0.021, lower scores represent worse HRQoL) [Figure 2(A) and 2(B)]. Mental fatigue scores were significantly higher in survivors (P = 0.012, higher scores represent more fatigue) [Figure 2(C)]. Scores on the remaining SF-36 and MFI-20 subscales and all HADS subscales did not differ significantly between survivors and controls. In 12 out of 15 subscales on generic HRQoL, fatigue, anxiety, and depression, the interquartile range of scores was larger for survivors than for controls; for none

Table 2. Quality of life in adult survivors of pediatric differentiated thyroid carcinoma and matched

controls

 Questionnaire Survivors

n = 67

Controls

n = 56

P Value

Survivors vs. controlsSF-36

Physical functioning 95 (85-100)a 100 (95-100)b 0.031c

Role limitations due to physical problems 100 (75-100)a 100 (100-100)b 0.021c

Bodily pain 84 (72-100)a 100 (74-100)d 0.104

Social functioning 88 (63-100) 100 (81-100) 0.065

Mental health 84 (72-92)a 84 (76-92) 0.723

Role limitations due to emotional problems 100 (67-100)a 100 (67-100)b 0.711

General health perceptions 72 (57-87)a 77 (67-89) 0.231

Vitality 65 (50-78)a 70 (60-80) 0.194

Mental component summary scale 54 (41-56)e 53 (47-56)b 0.961

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

147

7

Table 2. (continued)

Survivors

n = 67

Controls

n = 56

P Value

Physical component summary scale 53 (47-57)e 57 (53-59)b 0.024c

MFI-20

General fatigue 10 (8-15) 9 (5-12) 0.075

Physical fatigue 8 (5-12) 6 (4-10) 0.083

Reduced activity 8 (5-11) 8 (5-11) 0.613

Reduced motivation 6 (4-9) 6 (4-9) 0.879

Mental fatigue 9 (5-15) 7 (4-10) 0.012c

Total 41 (31-57) 36 (27-54) 0.129

HADS

Anxiety 4 (2-9) 3 (2-6) 0.317

Depression 1 (0-4) 1 (0-3) 0.964

Total 6 (2-11) 4 (3-9) 0.392

Survivors only THYCA-QoL scales

Neuromuscular 11 (0-22)

Voice 0 (0-17)

Concentration 0 (0-17)

Sympathetic 0 (0-17)

Throat/mouth 11 (0-22)

Psychological 8 (0-17)

Sensory 17 (0-33)

THYCA-QoL single items, n (%) No A Little Quite a Bit Very Much

Problems with scara 56 (84) 8 (12) 2 (3) 0 (0)

Felt chilly 39 (58) 13 (19) 9 (13) 6 (9)

Tingling hands/feet 42 (63) 19 (28) 5 (8) 1 (2)

Gained weight 47 (70) 16 (24) 3 (5) 1 (2)

Headache 34 (51) 29 (43) 2 (3) 1 (2)

Interested in sex 8 (12) 37 (55) 21 (31) 1 (2)

Numbers are shown as median (interquartile range). Abbreviations: SF-36, short form 36; MFI-20, multidimensional

fatigue inventory 20; HADS, hospital anxiety and depression scale; and THYCA-QoL; thyroid cancer-specific

health-related quality of life. For SF-36, higher scores indicate a better HRQoL; subscales range from 0 to 100. For

MFI-20, higher scores indicate more fatigue; subscales range from 4 to 20; the total scale ranges from 20 to 100.

For HADS, higher scores indicate higher levels of anxiety and/or depression; subscales range from 0 to 21; the total

scale ranges from 0 to 42. For THYCA-QoL, a higher score indicates more symptoms; subscales range from 0 to

100. Scores on the SF-36, MFI-20, and HADS were compared using the Mann–Whitney U test. a n = 66 because 1

survivor filled in the questionnaire erroneously. b n = 54 because 2 controls filled in the questionnaire erroneously. c P < 0.05. d n = 53 because 3 controls filled in the questionnaire erroneously. e n = 65 because 2 survivors filled in

the questionnaire erroneously.

of the 15 subscales the interquartile range of scores for controls was larger than that for the survivors. For the component scales, scores on the physical component scale were significantly lower in survivors compared to controls (P = 0.024; lower scores

CHAPTER 7

148

7

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

149

7

represent worse HRQoL). Scores of other summary scales (mental component scale,

total MFI-20, and total HADS scores) did not differ significantly between survivors and

controls. Scores of survivors with a second malignant neoplasm (SMN) are reported in

Supplemental Table 2. Three survivors developed a SMN after DTC. Scores of QoL of

the 2 women diagnosed with breast cancer were similar compared with the survivors

that were not diagnosed with a SMN. For 1 woman diagnosed with a high-grade cervical

intraepithelial neoplasia, QoL scores were lower compared with the survivors that were

not diagnosed with a SMN after DTC. Siblings of survivors did not differ significantly

from nonsiblings in scores on questionnaires (data not shown). For thyroid cancer-

specific QoL, scores of survivors are shown in Table 2 and Figure 2(D). Most frequently

reported were neuromuscular, throat/mouth, psychological and sensory complaints,

feeling chilly, and headache.

Characteristics associated with QoL in adult survivors of pediatric DTC Characteristics that were possibly associated with QoL in adult survivors of pediatric

DTC are shown in Supplemental Table 3. Male survivors reported higher levels

of reduced motivation and more depression than did females (P = 0.007 and P =

0.009). Unemployment was associated with lower scores on physical functioning

and the physical component scale (P = 0.005 and P = 0.003) and higher levels of

general fatigue, physical fatigue and total fatigue (P = 0.009, P = 0.009, and, P = 0.004,

respectively). Treatment with a higher cumulative dose of 131I was associated with more

complaints of headache (P = 0.006). Survivors with recurrent or persistent disease

and survivors with a higher level of TSH suppression reported more complaints

about the scars in their necks (P <0.001 and P = 0.002). Marital status at follow-up,

educational level, age at follow-up, all tumor characteristics (i.e. age at diagnosis,

tumor node metastasis stage, and histology), surgical complications (permanent

hypoparathyroidism and recurrent laryngeal nerve injury), and follow-up duration

showed no significant associations with QoL (HRQoL, fatigue, anxiety, depression,

and/or thyroid cancer-specific HRQoL).

Figure 2. Quality of Life (QoL) scores in survivors of pediatric differentiated thyroid carcinoma. An

asterisk (*) indicates a statistically significant difference. (A-C) Scores of survivors vs. controls. (A) Physical

functioning (Short Form 36, health-related QoL) scores of survivors of vs. matched controls. Scale ranges

from 0 to 100. A higher score indicates a better QoL. (B) Limitations in role functioning due to physical

health (Short Form 36, health-related QoL) scores of survivors vs. matched controls. Scale ranges from

0 to 100. A higher score indicates a better QoL. (C) Mental fatigue (Multidimensional Fatigue Inventory,

fatigue) scores of survivors vs. matched controls. Scale ranges from 4 to 20. A higher score indicates

a worse QoL. (D) Scores of survivors only (THYCA-QoL, thyroid cancer-specific health-related QoL).

Scores of survivors on neuromuscular, throat/mouth, psychological, and sensory complaints are shown.

Scales range from 0 to 100. A higher score indicates a worse QoL.

CHAPTER 7

150

7

DISCUSSIONIn the present study, we focus on QoL in long-term adult survivors of pediatric DTC, and our results show overall normal HRQoL, fatigue, anxiety, and depression in these survivors compared to matched controls. This normal QoL is similar to that found in studies performed in survivors of adolescent DTC (8, 9). Although survivors reported more physical problems, role limitations due to physical problems, and mental fatigue, the overall scores of survivors on these domains were still within the normal range. Thyroid cancer-specific complaints were present in some survivors, but most survivors reported relatively few or no complaints [Table 2 and Figure 2(D)]. Another reflection of the normal QoL of the survivors is their full participation in society, exemplified in most of these survivors by high employment and relationship rates and an educational level comparable to matched controls. Thereby, most (88%) of the survivors reported to have an interest in sex.

Remarkably, the overall QoL in survivors of pediatric DTC seems to be better than the QoL of survivors diagnosed with DTC at older ages. HRQoL, fatigue, anxiety, and depression were reported as worse in both short- and long-term survivor studies in adult DTC compared with controls (10, 11). Additionally, survivors of (young) adult DTC seem to experience more severe thyroid cancer-specific complaints than do survivors of pediatric DTC (5, 13, 22).

Although overall QoL is normal in pediatric DTC survivors compared to controls, physical problems seemed to be relatively more prominent in the survivor group compared with controls. The impaired physical functioning observed in survivors of pediatric DTC has been described previously in other childhood cancer survivors (18, 19). Also in adult DTC survivors, the largest differences in scores between survivors and the general population were observed for physical complaints and complaints of fatigue (11).

Besides physical obstacles, survivors in the present study experienced mental constraints, in particular mental fatigue, entailing the inability to stay concentrated. Husson et al. (5) proposed that (mental) fatigue in survivors of (adult) DTC might be due to a suboptimal TSH level suppression target or could otherwise be explained by the presence of cancer-related fatigue (5, 31). The present data provided no support for a possible relationship between TSH levels and fatigue, which is consistent with findings in DTC survivors diagnosed at older ages (8, 10).

Normal anxiety and depression levels as we found in the survivors of pediatric DTC were similar to the reported similar levels of emotional distress found in survivors of other childhood cancers (e.g. acute lymphatic leukemia, renal tumor, non-Hodgkin lymphoma, and other types of cancer) and controls (32).

Survivors reported neuromuscular, throat/mouth, psychological, and sensory problems to be most present in the symptom scale items. Headache and feeling chilly were reported most frequently in the single items. Complaints most often reported

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

151

7

in survivors of adult DTC were sympathetic complaints, neuromuscular complaints, and complaints of fatigue (13). Physicians underestimate the incidence of several thyroid cancer-specific complaints in survivors (21). For example, although 40% of the survivors reported voice problems, for only 15% of the survivors, recurrent laryngeal nerve injury was reported in their medical records (Supplemental Table 1). Because only 3 survivors were diagnosed with a SMN after DTC (Supplemental Table 2), general conclusions regarding QoL in this specific group of survivors cannot be drawn.

QoL is more variable in survivors compared with controls, as shown by the wider distribution toward worse QoL in survivors [Figure 2(A-C)]. It would be of great value to be able to portend QoL (variability) based on predictors. Therefore, the present study evaluated whether long-term generic HRQoL, fatigue, anxiety, depression, and thyroid cancer-specific HRQoL were associated with survivor, tumor, treatment, or follow-up characteristics. Of all possible predictors of QoL, unemployment was most frequently associated with worse QoL on many domains. However, the retrospective design of the study does not allow us to make statements regarding the causality of this impairment. This means that employment could influence QoL or vice versa. Socioeconomic factors could play a role in the level of QoL, as has been described previously in studies evaluating QoL in survivors of adult DTC (21, 22). Additionally, we found evidence for a more impaired QoL among survivors undergoing more extensive treatment or survivors with a more extended disease. Survivors with a worse outcome may report more complaints due to their more active disease and treatment. Lower QoL in female survivors of DTC compared with male survivors has been reported by several authors (21, 22). However, owing to the limited number of male survivors in the current study, conclusions regarding associations between sex and QoL could not be drawn. Adverse late effects on physical functioning have been described after administration of 131I to survivors of adult DTC (33, 34). An association between worse QoL and higher doses of 131I in survivors of adult DTC has previously been described (35). The present data on survivors of pediatric DTC also show an association between higher cumulative doses of 131I and worse QoL. In contrast, QoL in survivors of pediatric DTC was shown to be independent of other tumor or treatment characteristics, which confirms findings of studies in survivors of adult DTC (10, 21). Survivors of adult DTC showed an increasing QoL >5 years after diagnosis (21). The normal QoL in the present survivors may be explained by their long-term follow-up, but present results do not confirm this hypothesis: a longer follow-up period was not associated with better QoL.

The present study has both strengths and limitations. This study considers the QoL of adult survivors of both pediatric and adolescent DTC. Moreover, survivors were compared to sex, age, and socioeconomic-matched controls. Because of the cross-sectional design of the study ≥5 years after diagnosis, short-term QoL was not evaluated. Because pediatric DTC is uncommon, gathering a larger group of survivors

CHAPTER 7

152

7

is difficult. owing to nonnormally distributed data and the limited number of survivors included in this study, multivariable regression or stratified analysis was not feasible. To make more conclusive statements, international collaboration in prospective studies assessing QoL in pediatric DTC survivors is urgently needed. Moreover, to identify characteristics that predict QoL, longitudinal prospective studies would be more appropriate.

Finally, the results of this study were based only on quantitative data. Combining quantitative and qualitative elements to assess QoL in survivors of pediatric DTC could help to evaluate perceived physical and mental limitations and parameters that could in turn predict QoL more clearly; such methodology has also been applied in studies of survivors of adult DTC (36).

To identify vulnerable childhood cancer survivors early and offer them the care they need, regular measurement of QoL parameters should be implemented during follow-up. This is in accordance with Dutch evidence-based guidelines for follow-up of childhood cancer survivors (37). Additionally, measurements at baseline and during treatment should also be performed to evaluate individual alterations in QoL. In our opinion, follow-up regarding monitoring QoL is necessary in survivors of pediatric DTC.

In conclusion, this study shows that, in general, long-term adult survivors of pediatric DTC function well in society, with relatively good HRQoL and levels of fatigue, anxiety, and depression that are similar to matched controls. Various thyroid cancer-specific complaints were present (e.g. sensory complaints, headache, and chilliness), but mostly in mild forms. However, physical problems and mental fatigue seemed to be relatively more prominent in the survivor group compared with controls. Thereby, scores on QoL in survivors are widespread, indicating individual differences between survivors. Factors most frequently associated with worse QoL were unemployment and more extensive disease or treatment characteristics. These predictors will have to be further explored in future studies. We recommend evaluating changes in the QoL of individual pediatric DTC patients from diagnosis to long-term follow-up, using a prospective screening program to be able to provide early and individualized care to prevent long-term alterations in QoL.

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

153

7

ACKNOWLEDGMENTSWe are grateful to our colleagues in The Netherlands for referring patients for this study. We thank the registration teams of the Comprehensive Cancer Centers for the collection of data for the Netherlands Cancer Registry, as well as the scientific staff of the Netherlands Cancer Registry. We also thank Heleen Maurice-Stam for sharing expert opinion with us during analysis of the Short Form 36.

FUNDINGThis work was supported by Stichting Kinderen Kankervrij (The Netherlands, Foundation Children Cancer-Free, Project 81). C.M.R. is supported by the Dutch Cancer Society.

CLINICAL TRIAL REGISTRYNetherlands Trial Registry 3448/NL3280.

DISCLOSURE SUMMARY The authors have nothing to disclose.

CHAPTER 7

154

7

REFERENCES1. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM,

Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L. 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. 2016;26(1):1–133.

2. Klein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-

Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Schmid KW, Smit JW, Williams

GR, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP. Pediatric differentiated thyroid carcinoma in

The Netherlands: a nationwide follow-up study. J Clin Endocrinol Metab. 2016;101(5): 2031–2039.

3. Vergamini LB, Frazier AL, Abrantes FL, Ribeiro KB, Rodriguez-Galindo C. Increase in the incidence of

differentiated thyroid carcinoma in children, adolescents, and young adults: a population-based study. J

Pediatr. 2014;164(6):1481–1485.

4. Siegel DA, King J, Tai E, Buchanan N, Ajani UA, Li J. Cancer incidence rates and trends among children and

adolescents in the United States, 2001–2009. Pediatrics. 2014;134(4):e945–e955.

5. Husson O, Nieuwlaat WA, Oranje WA, Haak HR, van de Poll-Franse LV, Mols F. Fatigue among short- and long-term

thyroid cancer survivors: results from the population-based PROFILES registry. Thyroid. 2013;23(10):1247–1255.

6. de Oliveira Chachamovitz DS, dos Santos Vigário P, Nogueira Cordeiro MF, de Castro CL, Vaisman M, dos Santos

Teixeira PdeF. Quality of life, muscle strength, and fatigue perception in patients on suppressive therapy with

levothyroxine for differentiated thyroid carcinoma. Am J Clin Oncol. 2013;36(4):354–361.

7. Sikjaer T, Moser E, Rolighed L, Underbjerg L, Bislev LS, Mosekilde L, Rejnmark L. Concurrent hypoparathyroidism

is associated with impaired physical function and quality of life in hypothyroidism. J Bone Miner

Res. 2016;31(7):1440–1448.

8. Oren A, Benoit MA, Murphy A, Schulte F, Hamilton J. Quality of life and anxiety in adolescents with differentiated

thyroid cancer. J Clin Endocrinol Metab. 2012;97(10):E1933–E1937.

9. Metallo M, Groza L, Brunaud L, Klein M, Weryha G, Feigerlova E. Long-term quality of life and pregnancy outcomes

of differentiated thyroid cancer survivors treated by total thyroidectomy and I131 during adolescence and young

adulthood. Int J Endocrinol. 2016; 2016:7586482.

10. Hoftijzer HC, Heemstra KA, Corssmit EP, van der Klaauw AA, Romijn JA, Smit JW. Quality of life in cured patients

with differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2008;93(1):200–203.

11. Gamper EM, Wintner LM, Rodrigues M, Buxbaum S, Nilica B, Singer S, Giesinger JM, Holzner B, Virgolini I. Persistent

quality of life impairments in differentiated thyroid cancer patients: results from a monitoring programme. Eur J

Nucl Med Mol Imaging. 2015;42(8):1179–1188.

12. Lee JI, Kim SH, Tan AH, Kim HK, Jang HW, Hur KY, Kim JH, Kim KW, Chung JH, Kim SW. Decreased health-related quality

of life in disease-free survivors of differentiated thyroid cancer in Korea. Health Qual Life Outcomes. 2010;8:101.

13. Husson O, Haak HR, Buffart LM, Nieuwlaat WA, Oranje WA, Mols F, Kuijpens JL, Coebergh JW, van de Poll-Franse

LV. Health-related quality of life and disease specific symptoms in long-term thyroid cancer survivors: a study from

the population-based PROFILES registry. Acta Oncol. 2013;52(2):249–258.

14. Sawka AM, Naeem A, Jones J, Lowe J, Segal P, Goguen J, Gilbert J, Zahedi A, Kelly C, Ezzat S. Persistent posttreatment

fatigue in thyroid cancer survivors: a scoping review. Endocrinol Metab Clin North Am. 2014;43(2):475–494.

15. Langeveld N, Ubbink M, Smets E, Dutch Late Effects Study Group. “I don’t have any energy”: the experience of

fatigue in young adult survivors of childhood cancer. Eur J Oncol Nurs. 2000;4(1):20–28.

16. van Santen HM, Aronson DC, Vulsma T, Tummers RF, Geenen MM, de Vijlder JJ, van den Bos C. Frequent adverse

events after treatment for childhood-onset differentiated thyroid carcinoma: a single institute experience. Eur J

Cancer. 2004;40(11):1743–1751.

17. Jóhannsdóttir IM, Hjermstad MJ, Moum T, Wesenberg F, Hjorth L, Schrøder H, Mört S, Jónmundsson G, Loge JH.

Increased prevalence of chronic fatigue among survivors of childhood cancers: a population-based study. Pediatr

Blood Cancer. 2012;58(3):415–420.

18. Rueegg CS, Gianinazzi ME, Rischewski J, Beck Popovic M, von der Weid NX, Michel G, Kuehni CE. Health-related

quality of life in survivors of childhood cancer: the role of chronic health problems. J Cancer Surviv. 2013;7(4):511–522.

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

155

7

19. Sundberg KK, Doukkali E, Lampic C, Eriksson LE, Arvidson J, Wettergren L. Long-term survivors of childhood cancer

report quality of life and health status in parity with a comparison group. Pediatr Blood Cancer. 2010;55(2):337–343.

20. Frederick NN, Kenney L, Vrooman L, Recklitis CJ. Fatigue in adolescent and adult survivors of non-CNS childhood

cancer: a report from project REACH. Support Care Cancer. 2016;24(9):3951–3959.

21. Aschebrook-Kilfoy B, James B, Nagar S, Kaplan S, Seng V, Ahsan H, Angelos P, Kaplan EL, GuerreroMA,

Kuo JH, Lee JA, Mitmaker EJ, Moalem J, Ruan DT, Shen WT, Grogan RH. Risk factors for decreased quality of

life in thyroid cancer survivors: initial findings from the North American thyroid cancer survivorship study.

Thyroid. 2015;25(12):1313–1321.

22. Goldfarb M, Casillas J. Thyroid cancer-specific quality of life and health-related quality of life in young adult

thyroid cancer survivors. Thyroid. 2016;26(7):923–932.

23. Ware JE, Jr, Sherbourne CD. The MOS 36-item short-form health survey (SF-36). I. Conceptual framework and item

selection. Med Care. 1992;30(6):473–483.

24. Aaronson NK, Muller M, Cohen PD, Essink-Bot ML, Fekkes M, Sanderman R, Sprangers MA, te Velde A, Verrips E.

Translation, validation, and norming of the Dutch language version of the SF-36 Health Survey in community and

chronic disease populations. J Clin Epidemiol. 1998;51(11):1055–1068.

25. Ware JE, Kosinski M, Keller SD. SF-36 Physical and Mental Health Summary Scales: A User’s Manual. Boston, MA:

Health Institute, New England Medical Center; 1994.

26. Smets EM, Garssen B, Bonke B, De Haes JC. The Multidimensional Fatigue Inventory (MFI) psychometric qualities

of an instrument to assess fatigue. J Psychosom Res. 1995;39(3):315–325.

27. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67(6):361–370.

28. Spinhoven P, Ormel J, Sloekers PP, Kempen GI, Speckens AE, Van Hemert AM. A validation study of the Hospital

Anxiety and Depression Scale (HADS) in different groups of Dutch subjects. Psychol Med. 1997;27(2):363–370.

29. Husson O, Haak HR, Mols F, Nieuwenhuijzen GA, Nieuwlaat WA, Reemst PH, Huysmans DA, Toorians AW, van de

Poll-Franse LV. Development of a disease-specific health-related quality of life questionnaire (THYCA-QoL) for

thyroid cancer survivors. Acta Oncol. 2013;52(2):447–454.

30. Klein Hesselink EN, Klein Hesselink MS, de Bock GH, Gansevoort RT, Bakker SJ, Vredeveld EJ, van der Horst-Schrivers

AN, van der Horst IC, Kamphuisen PW, Plukker JT, Links TP, Lefrandt JD. Long-term cardiovascular mortality in

patients with differentiated thyroid carcinoma: an observational study. J Clin Oncol. 2013; 31(32):4046–4053.

31. Berger AM, Abernethy AP, Atkinson A, Barsevick AM, Breitbart WS, Cella D, Cimprich B, Cleeland C, Eisenberger

MA, Escalante CP, Jacobsen PB, Kaldor P, Ligibel JA, Murphy BA, O’Connor T, Pirl WF, Rodler E, Rugo HS, Thomas J,

Wagner LI. Cancer-related fatigue. J Natl Compr Canc Netw. 2010;8(8):904–931.

32. van der Geest IM, van Dorp W, Hop WC, Neggers SJ, de Vries AC, Pieters R, Aarsen FK, van den Heuvel-Eibrink

MM. Emotional distress in 652 Dutch very long-term survivors of childhood cancer, using the hospital anxiety and

depression scale (HADS). J Pediatr Hematol Oncol. 2013;35(7):525–529.

33. Fard-Esfahani A, Emami-Ardekani A, Fallahi B, Fard-Esfahani P, Beiki D, Hassanzadeh-Rad A, Eftekhari

M. Adverse effects of radioactive iodine-131 treatment for differentiated thyroid carcinoma. Nucl Med

Commun. 2014;35(8):808–817.

34. Klein Hesselink EN, Links TP. Radioiodine treatment and thyroid hormone suppression therapy for differentiated

thyroid carcinoma: adverse effects support the trend toward less aggressive treatment for low-risk patients. Eur

Thyroid J. 2015;4(2):82–92.

35. Almeida JP, Vartanian JG, Kowalski LP. Clinical predictors of quality of life in patients with initial differentiated

thyroid cancers. Arch Otolaryngol Head Neck Surg. 2009;135(4):342–346.

36. Applewhite MK, James BC, Kaplan SP, Angelos P, Kaplan EL, Grogan RH, Aschebrook-Kilfoy B. Quality of life in

thyroid cancer is similar to that of other cancers with worse survival. World J Surg. 2016;40(3):551–561.

37. Dutch Childhood Oncology Group. Guidelines for follow-up in survivors of childhood cancer 5 years after

diagnosis. Den Haag/Amsterdam: SKION; 2010.

CHAPTER 7

156

7

SUPPLEMENTARY DATA

Supplemental Table 1. Tumor-, treatment-, and follow-up characteristics of adult survivors of pediatric

differentiated thyroid carcinoma included in the quality of life study compared to eligible, but not

included survivors (non-participants) and compared to survivors excluded for the quality of life study

Survivors in the nationwide study

n = 105

Survivors included for QoL

n = 75

Survivors excluded for QoL

n = 30

Participants

n = 67

Non-

participants

n = 8

P Value P Value

Gender, n (%) 0.3333 0.5443

Female 58 (86.6) 6 (75.0) 24 (80.0)

Male 9 (13.4) 2 (25.0) 6 (20.0)

Age at evaluation, years 0.837² <0.001*²

Median (range) 34.2 (18.8-61.7) 39.3 (12.2-51.4) 19.1 (10.6-39.5)

Age at diagnosis, years 0.066² 0.898²

Median (range) 15.8 (7.9-18.8) 13.8 (5.8-17.0) 15.9 (6.1-18.9)

Follow-up duration, years 0.595² <0.001*²

Median (range) 17.8 (5.0-44.7) 24.1 (6.4-34.5) 2.8 (1.1-21.5)

Histology, n (%) 1.0003 0.5341

Papillary 55 (82.1) 7 (87.5) 23 (76.7)

Follicular 12 (17.9) 1 (12.5) 7 (23.3)

TNM stage, n (%)T 0.6683 0.1401

T1-T2 40 (59.7) 7 (87.5) 18 (60.0)

T3-T4 13 (19.4) 1 (12.5) 12 (40.0)

Tx 14 (20.9) 0 (0) 0 (0)

N 0.7103 0.2591

N0 31 (46.3) 3 (37.5) 19 (63.3)

N1 30 (44.8) 5 (62.5) 11 (36.7)

Nx 6 (9.0) 0 (0) (0)

M 0.5563 0.1093

M0 54 (80.6) 6 (75.0) 22 (73.3)

M1 6 (9.0) 1 (12.5) 7 (23.3)

Mx 7 (10.4) 1 (12.5) 1 (3.3)

Surgery, n (%) n.a. n.a.

Total thyroidectomy 67 (100) 8 (100) 30 (100)

LND 1.000³ 0.1151

None 31 (46.3) 4 (50.0) 14 (46.7)

Central LND 4 (6.0) 0 (0) 6 (20.0)

LND incl. lateral levels 24 (35.8) 4 (50.0) 8 (26.7)

Unknown 8 (11.9) 0 (0) 2 (6.7)

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

157

7

Supplemental Table 1. (continued)

Survivors in the nationwide study

n = 105

Survivors included for QoL

n = 75

Survivors excluded for QoL

n = 30

Participants

n = 67

Non-

participants

n = 8

P Value P Value

Surgical complications, n (%)Permanent HP (yes) 17 (25.4) 1 (12.5) 0.6623 7 (23.3) 0.6421

RLN injury (yes) 10 (14.9) 0 (0) 0.573³ 3 (10.0) 0.5253

131I administration, n (%)131I administered (yes) 65 (97.0) 8 (100) 29 (96.7)

Cumulative 131I activity,

GBq

0.567² 0.1222

Median (range) 6.0 (1.2-35.2)a 5.6 (1.7-29.8)b 2.0 (0.7-17.1)c

Outcome, n (%) 1.000³ 0.1363

Remission 55 (82.1) 6 (75.0) 24 (80.0)

Recurrence 7 (10.4) 1 (12.5) 0 (0)

Persistent disease 5 (7.5) 0 (0) 4 (13.3)

Abbreviations: QoL, quality of life; TNM, tumor node metastasis; LND, lymph node dissection; HP, hypoparathyroidism;

RLN, recurrent laryngeal nerve; and 131I, radioactive iodine. a n=64 because 131I was not administered in two survivors

and 131I dose was unknown in one survivor. b n=29 because 131I dose was unknown in one non-participant. c n=8 because 131I was not administered to one non-QoL participant. Significant values (P <0.05) are marked with an asterisk (*).

¹ χ2 test. ² Mann-Whitney U test. ³ Fisher’s exact test.

CHAPTER 7

158

7

Supplemental Table 2. Quality of life in adult survivors of pediatric differentiated thyroid carcinoma

diagnosed with a secondary malignant neoplasm

Survivors

with no SMN

after DTC

n = 64

Survivors with SMN after DTC

High-

grade

CIN

n = 1

Breast

cancer

survivor 1

n = 1

Breast

cancer

survivor 2

n = 1

Short Form-361, median Physical functioning 95a 60 95 65

Role limitations due to physical problems 100a 0 100 100

Bodily pain 84a 12 84 84

Social functioning 88 0 100 88

Mental health 84a 68 92 84

Role limitations due to emotional problems 100a 0 100 100

General health perceptions 72a 35 92 72

Vitality 65a 30 75 50

Mental component summary scale 53b 31 56 54

Physical component summary scale 53b 31 55 47

Multidimensional Fatigue Index 202, medianGeneral fatigue 10 20 10 10

Physical fatigue 8 20 7 9

Reduced activity 8 17 5 5

Reduced motivation 6 5 6 4

Mental fatigue 9 18 6 9

Total 41 80 34 37

Hospital Anxiety and Depression Scale3, medianAnxiety 4 13 1 0

Depression 1 3 0 0

Total 6 16 1 0

THYCA-QoL scales4, median Neuromuscular 11 89 0 33

Voice 0 100 0 0

Concentration 0 67 17 0

Sympathetic 0 33 17 33

Throat/mouth 11 44 0 0

Psychological 8 83 8 0

Sensory 17 67 0 17

THYCA-QoL singe items5, n (%)Problems with scar (yes) 10 (15)a,c no no no

Felt chilly (yes) 26 (41)c very much a little no

Tingling hands/feet (yes) 22 (34)c quite a bit a little a little

Gained weight (yes) 18 (28)c very much a little no

Headache (yes) 29 (45)a,c a little a little a little

Interested in sex (yes) 57 (89)c quite a bit quite a bit no

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

159

7

Supplemental Table 2. (continued)

Abbreviations: SMN, second malignant neoplasm; DTC, differentiated thyroid carcinoma; CIN, cervical intraepithelial

neoplasia. 1 Higher scores indicate a better HRQoL, subscales range from 0-100. 2 Higher scores indicate more fatigue,

subscales range from 4-20, the total scale ranges from 20-100. 3 Higher scores indicate higher levels anxiety and/or

depression, subscales range from 0-21, the total scale ranges from 0-42. 4 A higher score indicates more symptoms,

subscales range from 0-100. 5 Not at all = no; a little, quite a bit and very much = yes. a n=63 and b n=62 because

one and two survivors, respectively, filled in the questionnaire erroneously. c For detailed a description of all scores,

see Table 2.

CHAPTER 7

160

7

Supplemental Table 3. Characteristics associated with quality of life in adult survivors of pediatric

differentiated thyroid carcinoma

Questionnaire - (sub)scale Survivor characteristics

Gender Marital status Employment/study Completed education Age at

evaluation

(years)

Females

n = 58

Median

Males

n = 9

P value Relationship

n = 43

Median

No

relationship

n = 24

P value Employed/student

n = 61

Median

Not employed

and no student

n = 6

P value Low level

n = 16

Median

Medium

level

n = 25

High

level

n = 26

P value

 

Corr.

coeff.

P value

SF-361

Physical functioning 95 100 0.080 100 100 0.361 98 75 0.005* 88 100 100 0.028 - 0.185 0.137

Role physical 100 100 0.188 100 100 0.213 100 0 0.035 100 100 100 0.676 - 0.026 0.836

Bodily pain 84 92 0.638 84 84 0.276 84 52 0.049 84 84 84 0.956 0.169 0.175

Social functioning 94 88 0.445 88 88 0.922 88 81 0.326 94 88 100 0.550 - 0.017 0.888

Mental health 84 80 0.464 88 88 0.658 84 76 0.534 82 84 88 0.293 - 0.028 0.825

Role emotional 100 100 0.788 100 100 0.888 100 83 0.319 100 100 100 0.171 - 0.088 0.480

Vitality 65 75 0.221 70 70 0.583 68 50 0.376 65 60 70 0.250 0.197 0.112

General health 72 85 0.088 79 79 0.385 77 54 0.029 72 82 67 0.446 - 0.055 0.659

Physical component scale 53 57 0.082 57 57 0.142 55 41 0.003* 53 55 51 0.231 0.071 0.574

Mental component scale 54 51 0.563 54 54 0.858 54 52 0.956 53 49 55 0.113 0.039 0.755

MFI-202 General fatigue 11 8 0.056 9 9 0.172 10 17 0.009* 11 11 9 0.448 - 0.021 0.867

Physical fatigue 8 5 0.040 6 6 0.082 7 17 0.009* 8 8 8 0.821 0.095 0.442

Reduced activity 7 8 0.290 7 7 0.413 7 14 0.010 8 8 6 0.352 0.049 0.693

Reduced motivation 6 9 0.007* 6 6 0.613 6 8 0.129 7 8 6 0.225 0.024 0.845

Mental fatigue 9 11 0.477 9 9 0.655 8 16 0.045 11 10 8 0.186 0.066 0.596

Total fatigue 41 41 0.832 40 40 0.578 40 73 0.004* 45 48 38 0.282 0.021 0.863

HADS3 Anxiety 4 5 0.684 3 3 0.200 3 9 0.025 5 3 5 0.774 0.047 0.705

Depression 1 3 0.009* 1 1 0.615 1 3 0.395 0 2 1 0.351 0.036 0.772

Total HADS 5 10 0.119 6 6 0.345 5 13 0.073 5 6 6 0.962 0.031 0.803

THYCA-QOL scales4

Neuromuscular 11 0 0.124 11 11 0.209 11 33 0.161 17 11 11 0.369 0.095 0.446

Voice 0 0 0.891 0 0 0.736 0 17 0.305 8 0 0 0.725 0.030 0.809

Concentration 0 0 0.566 0 0 0.742 0 25 0.089 0 0 0 0.554 0.222 0.071

Sympathetic 17 0 0.055 0 8 0.615 0 8 0.676 8 0 8 0.523 0.106 0.392

Throat/mouth 11 0 0.039 11 6 0.535 11 11 0.513 11 11 11 0.947 0.017 0.894

Psychological 8 8 0.397 8 8 0.249 8 38 0.014 4 8 8 0.997 0.159 0.197

Sensory 17 17 0.596 17 17 0.125 17 17 0.724 17 17 17 0.558 0.017 0.890

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

161

7

Supplemental Table 3. Characteristics associated with quality of life in adult survivors of pediatric

differentiated thyroid carcinoma

Questionnaire - (sub)scale Survivor characteristics

Gender Marital status Employment/study Completed education Age at

evaluation

(years)

Females

n = 58

Median

Males

n = 9

P value Relationship

n = 43

Median

No

relationship

n = 24

P value Employed/student

n = 61

Median

Not employed

and no student

n = 6

P value Low level

n = 16

Median

Medium

level

n = 25

High

level

n = 26

P value

 

Corr.

coeff.

P value

SF-361

Physical functioning 95 100 0.080 100 100 0.361 98 75 0.005* 88 100 100 0.028 - 0.185 0.137

Role physical 100 100 0.188 100 100 0.213 100 0 0.035 100 100 100 0.676 - 0.026 0.836

Bodily pain 84 92 0.638 84 84 0.276 84 52 0.049 84 84 84 0.956 0.169 0.175

Social functioning 94 88 0.445 88 88 0.922 88 81 0.326 94 88 100 0.550 - 0.017 0.888

Mental health 84 80 0.464 88 88 0.658 84 76 0.534 82 84 88 0.293 - 0.028 0.825

Role emotional 100 100 0.788 100 100 0.888 100 83 0.319 100 100 100 0.171 - 0.088 0.480

Vitality 65 75 0.221 70 70 0.583 68 50 0.376 65 60 70 0.250 0.197 0.112

General health 72 85 0.088 79 79 0.385 77 54 0.029 72 82 67 0.446 - 0.055 0.659

Physical component scale 53 57 0.082 57 57 0.142 55 41 0.003* 53 55 51 0.231 0.071 0.574

Mental component scale 54 51 0.563 54 54 0.858 54 52 0.956 53 49 55 0.113 0.039 0.755

MFI-202 General fatigue 11 8 0.056 9 9 0.172 10 17 0.009* 11 11 9 0.448 - 0.021 0.867

Physical fatigue 8 5 0.040 6 6 0.082 7 17 0.009* 8 8 8 0.821 0.095 0.442

Reduced activity 7 8 0.290 7 7 0.413 7 14 0.010 8 8 6 0.352 0.049 0.693

Reduced motivation 6 9 0.007* 6 6 0.613 6 8 0.129 7 8 6 0.225 0.024 0.845

Mental fatigue 9 11 0.477 9 9 0.655 8 16 0.045 11 10 8 0.186 0.066 0.596

Total fatigue 41 41 0.832 40 40 0.578 40 73 0.004* 45 48 38 0.282 0.021 0.863

HADS3 Anxiety 4 5 0.684 3 3 0.200 3 9 0.025 5 3 5 0.774 0.047 0.705

Depression 1 3 0.009* 1 1 0.615 1 3 0.395 0 2 1 0.351 0.036 0.772

Total HADS 5 10 0.119 6 6 0.345 5 13 0.073 5 6 6 0.962 0.031 0.803

THYCA-QOL scales4

Neuromuscular 11 0 0.124 11 11 0.209 11 33 0.161 17 11 11 0.369 0.095 0.446

Voice 0 0 0.891 0 0 0.736 0 17 0.305 8 0 0 0.725 0.030 0.809

Concentration 0 0 0.566 0 0 0.742 0 25 0.089 0 0 0 0.554 0.222 0.071

Sympathetic 17 0 0.055 0 8 0.615 0 8 0.676 8 0 8 0.523 0.106 0.392

Throat/mouth 11 0 0.039 11 6 0.535 11 11 0.513 11 11 11 0.947 0.017 0.894

Psychological 8 8 0.397 8 8 0.249 8 38 0.014 4 8 8 0.997 0.159 0.197

Sensory 17 17 0.596 17 17 0.125 17 17 0.724 17 17 17 0.558 0.017 0.890

CHAPTER 7

162

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale

THYCA-QOL single items5

Survivor characteristics

Gender Marital status Employment/study Completed education Age at

evaluation

(years)

Females

n = 58

n (%)

Males

n = 9

P value Relationship

n = 43

n (%)

No

relationship

n = 24

P value Employed/student

n = 61

n (%)

Not employed

and no student

n = 6

P value Low level

n = 16

n (%)

Medium

level

n = 25

High

level

n = 26

P value Median P value

Problems with scar 0.130 1.000 0.580 0.154 0.497

yes 7 (12) 3 (33) 6 (14) 4 (17) 10 (16) 0 (0) 4 (25) 1 (4) 5 (19) 28.3

no 50 (86) 6 (67) 36 (84) 20 (83) 50 (82) 6 (100) 12 (75) 23 (92) 21 (81) 34.1

Felt chilly 0.069 0.616 0.668 0.593 0.387

yes 27 (47) 1 (11) 17 (40) 11 (46) 25 (41) 3 (50) 8 (50) 11 (44) 9 (35) 32.1

no 31 (53) 8 (89) 26 (60) 13 (54) 36 (59) 3 (50) 8 (50) 14 (56) 17 (65) 34.3

Tingling hands/feet 0.466 0.981 0.186 0.195 0.300

yes 23 (40) 2 (22) 16 (37) 9 (38) 21 (34) 4 (67) 8 (50) 6 (24) 11 (42) 36.0

no 35 (60) 7 (78) 27 (63) 15 (63) 40 (66) 2 (33) 8 (50) 19 (76) 15 (58) 32.1

Gained weight 0.260 0.228 0.060 0.624 0.132

yes 19 (33) 1 (11) 15 (35) 5 (21) 16 (26) 4 (67) 5 (31) 9 (36) 6 (23) 37.5

no 39 (67) 8 (89) 28 (65) 19 (79) 45 (74) 2 (33) 11 (69) 16 (64) 20 (77) 32.0

Headached 0.151 0.402 1.000 0.423 0.270

yes 30 (52) 2 (22) 22 (51) 10 (42) 29 (48) 3 (50) 6 (38) 11 (44) 15 (58) 28.8

no 27 (47) 7 (78) 20 (47) 14 (58) 31 (51) 3 (50) 10 (63) 13 (52) 11 (42) 35.9

Interest in sex 1.000 0.021 1.000 0.110 0.363

yes 51 (88) 8 (89) 41 (95) 18 (75) 54 (89) 5 (83) 12 (75) 22 (88) 25 (96) 32.3

no 7 (12) 1 (11) 2 (5) 6 (25) 7 (11) 1 (17) 4 (25) 3 (12) 1 (4) 42.3

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

163

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale

THYCA-QOL single items5

Survivor characteristics

Gender Marital status Employment/study Completed education Age at

evaluation

(years)

Females

n = 58

n (%)

Males

n = 9

P value Relationship

n = 43

n (%)

No

relationship

n = 24

P value Employed/student

n = 61

n (%)

Not employed

and no student

n = 6

P value Low level

n = 16

n (%)

Medium

level

n = 25

High

level

n = 26

P value Median P value

Problems with scar 0.130 1.000 0.580 0.154 0.497

yes 7 (12) 3 (33) 6 (14) 4 (17) 10 (16) 0 (0) 4 (25) 1 (4) 5 (19) 28.3

no 50 (86) 6 (67) 36 (84) 20 (83) 50 (82) 6 (100) 12 (75) 23 (92) 21 (81) 34.1

Felt chilly 0.069 0.616 0.668 0.593 0.387

yes 27 (47) 1 (11) 17 (40) 11 (46) 25 (41) 3 (50) 8 (50) 11 (44) 9 (35) 32.1

no 31 (53) 8 (89) 26 (60) 13 (54) 36 (59) 3 (50) 8 (50) 14 (56) 17 (65) 34.3

Tingling hands/feet 0.466 0.981 0.186 0.195 0.300

yes 23 (40) 2 (22) 16 (37) 9 (38) 21 (34) 4 (67) 8 (50) 6 (24) 11 (42) 36.0

no 35 (60) 7 (78) 27 (63) 15 (63) 40 (66) 2 (33) 8 (50) 19 (76) 15 (58) 32.1

Gained weight 0.260 0.228 0.060 0.624 0.132

yes 19 (33) 1 (11) 15 (35) 5 (21) 16 (26) 4 (67) 5 (31) 9 (36) 6 (23) 37.5

no 39 (67) 8 (89) 28 (65) 19 (79) 45 (74) 2 (33) 11 (69) 16 (64) 20 (77) 32.0

Headached 0.151 0.402 1.000 0.423 0.270

yes 30 (52) 2 (22) 22 (51) 10 (42) 29 (48) 3 (50) 6 (38) 11 (44) 15 (58) 28.8

no 27 (47) 7 (78) 20 (47) 14 (58) 31 (51) 3 (50) 10 (63) 13 (52) 11 (42) 35.9

Interest in sex 1.000 0.021 1.000 0.110 0.363

yes 51 (88) 8 (89) 41 (95) 18 (75) 54 (89) 5 (83) 12 (75) 22 (88) 25 (96) 32.3

no 7 (12) 1 (11) 2 (5) 6 (25) 7 (11) 1 (17) 4 (25) 3 (12) 1 (4) 42.3

CHAPTER 7

164

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Tumor characteristics

Age at diagnosis

(years)

T stage N stage M stage Histology

Corr. coeff. P value T1-2

n = 40

Median

T3-4

n = 13

P value N0

n = 31

Median

N1

n=30

P value M0

n = 54

Median

M1

n = 6

P value PTC

n = 55

Median

FTC

n = 12

P value

SF-361

Physical functioning -0.058 0.645 95 100 0.453 95 100 0.673 95 95 0.855 95 100 0.902

Role physical -0.147 0.238 100 100 0.376 100 100 0.701 100 50 0.153 100 100 0.728

Bodily pain 0.132 0.290 84 84 0.982 84 84 0.939 84 67 0.081 84 79 0.468

Social functioning -0.080 0.520 88 88 0.394 88 94 0.401 88 81 0.622 88 94 0.403

Mental health -0.159 0.202 84 88 0.154 84 84 0.976 88 74 0.153 84 86 0.738

Role emotional -0.196 0.115 100 100 0.115 100 100 0.428 100 50 0.193 100 100 0.500

Vitality -0.094 0.453 65 73 0.093 65 70 0.608 65 63 0.874 68 55 0.822

General health -0.116 0.355 72 77 0.913 72 72 0.711 72 75 0.893 71 76 0.164

Physical component scale 0.096 0.447 53 52 0.876 55 53 0.412 54 52 0.931 53 54 0.960

Mental component scale -0.222 0.075 52 54 0.214 52 54 0.704 54 44 0.118 54 53 0.879

MFI-202 General fatigue 0.035 0.777 11 10 0.138 11 10 0.347 10 11 0.727 10 11 0.883

Physical fatigue 0.119 0.339 8 8 0.248 8 7 0.228 8 7 0.588 8 6 0.489

Reduced activity 0.111 0.307 8 5 0.050 8 6 0.206 7 8 0.819 7 7 0.843

Reduced motivation 0.127 0.307 7 5 0.376 7 6 0.592 6 7 0.819 6 7 0.635

Mental fatigue 0.159 0.199 11 7 0.334 12 8 0.486 10 12 0.447 9 10 0.761

Total fatigue 0.119 0.339 48 37 0.102 48 39 0.356 41 44 0.674 41 36 0.612

HADS3 Anxiety 0.195 0.113 5 2 0.049 4 3 0.685 3 7 0.082 4 4 0.724

Depression 0.025 0.842 1 0 0.463 1 2 0.899 1 5 0.259 1 2 0.973

Total HADS 0.189 0.125 7 4 0.082 6 5 0.806 5 11 0.074 6 5 0.694

THYCA-QOL scales4

Neuromuscular 0.058 0.640 11 11 0.864 11 14 0.249 11 19 0.303 11 11 0.892

Voice 0.030 0.810 0 0 0.811 0 0 0.224 0 8 0.492 0 0 0.449

Concentration 0.089 0.476 0 0 0.582 0 0 0.573 0 33 0.055 0 0 0.937

Sympathetic 0.072 0.564 0 17 0.028 0 17 0.249 0 8 0.857 0 0 0.403

Throat/mouth 0.083 0.503 11 0 0.922 11 6 0.555 11 17 0.433 11 0 0.301

Psychological 0.306 0.012 8 0 0.141 8 8 0.637 8 17 0.572 8 4 0.416

Sensory 0.063 0.612 17 17 0.176 17 17 0.167 17 25 0.327 17 17 0.521

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

165

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Tumor characteristics

Age at diagnosis

(years)

T stage N stage M stage Histology

Corr. coeff. P value T1-2

n = 40

Median

T3-4

n = 13

P value N0

n = 31

Median

N1

n=30

P value M0

n = 54

Median

M1

n = 6

P value PTC

n = 55

Median

FTC

n = 12

P value

SF-361

Physical functioning -0.058 0.645 95 100 0.453 95 100 0.673 95 95 0.855 95 100 0.902

Role physical -0.147 0.238 100 100 0.376 100 100 0.701 100 50 0.153 100 100 0.728

Bodily pain 0.132 0.290 84 84 0.982 84 84 0.939 84 67 0.081 84 79 0.468

Social functioning -0.080 0.520 88 88 0.394 88 94 0.401 88 81 0.622 88 94 0.403

Mental health -0.159 0.202 84 88 0.154 84 84 0.976 88 74 0.153 84 86 0.738

Role emotional -0.196 0.115 100 100 0.115 100 100 0.428 100 50 0.193 100 100 0.500

Vitality -0.094 0.453 65 73 0.093 65 70 0.608 65 63 0.874 68 55 0.822

General health -0.116 0.355 72 77 0.913 72 72 0.711 72 75 0.893 71 76 0.164

Physical component scale 0.096 0.447 53 52 0.876 55 53 0.412 54 52 0.931 53 54 0.960

Mental component scale -0.222 0.075 52 54 0.214 52 54 0.704 54 44 0.118 54 53 0.879

MFI-202 General fatigue 0.035 0.777 11 10 0.138 11 10 0.347 10 11 0.727 10 11 0.883

Physical fatigue 0.119 0.339 8 8 0.248 8 7 0.228 8 7 0.588 8 6 0.489

Reduced activity 0.111 0.307 8 5 0.050 8 6 0.206 7 8 0.819 7 7 0.843

Reduced motivation 0.127 0.307 7 5 0.376 7 6 0.592 6 7 0.819 6 7 0.635

Mental fatigue 0.159 0.199 11 7 0.334 12 8 0.486 10 12 0.447 9 10 0.761

Total fatigue 0.119 0.339 48 37 0.102 48 39 0.356 41 44 0.674 41 36 0.612

HADS3 Anxiety 0.195 0.113 5 2 0.049 4 3 0.685 3 7 0.082 4 4 0.724

Depression 0.025 0.842 1 0 0.463 1 2 0.899 1 5 0.259 1 2 0.973

Total HADS 0.189 0.125 7 4 0.082 6 5 0.806 5 11 0.074 6 5 0.694

THYCA-QOL scales4

Neuromuscular 0.058 0.640 11 11 0.864 11 14 0.249 11 19 0.303 11 11 0.892

Voice 0.030 0.810 0 0 0.811 0 0 0.224 0 8 0.492 0 0 0.449

Concentration 0.089 0.476 0 0 0.582 0 0 0.573 0 33 0.055 0 0 0.937

Sympathetic 0.072 0.564 0 17 0.028 0 17 0.249 0 8 0.857 0 0 0.403

Throat/mouth 0.083 0.503 11 0 0.922 11 6 0.555 11 17 0.433 11 0 0.301

Psychological 0.306 0.012 8 0 0.141 8 8 0.637 8 17 0.572 8 4 0.416

Sensory 0.063 0.612 17 17 0.176 17 17 0.167 17 25 0.327 17 17 0.521

CHAPTER 7

166

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Tumor characteristics

Age at diagnosis

(years)

T stage N stage M stage Histology

Median P value T1-2

n = 40

n (%)

T3-4

n = 13

P value N0

n = 31

n (%)

N1

n=30

P value M0

n = 54

n (%)

M1

n = 6

P value PTC

n = 55

n (%)

FTC

n = 12

P value

THYCA-QOL single items5

Problems with scar 0.629 1.000 0.488 1.000 0.187

yes 16.4 5 (13) 2 (15) 4 (13) 6 (20) 8 (15) 1 (17) 10 (18) 0 (0)

no 15.7 34 (85) 11 (85) 26 (84) 24 (77) 45 (83) 5 (83) 44 (80) 12 (100)

Felt chilly 0.670 0.173 0.684 1.000 0.193

yes 16.0 21 (53) 4 (31) 14 (45) 12 (40) 22 (41) 2 (33) 25 (45) 3 (25)

no 15.6 19 (48) 9 (69) 17 (55) 18 (60) 32 (59) 4 (67) 30 (55) 9 (75)

Tingling hands/feet 0.337 0.579 0.529 0.179 0.751

yes 16.3 15 (38) 6 (46) 10 (32) 12 (40) 18 (33) 4 (67) 20 (36) 5 (42)

no 15.6 25 (63) 7 (54) 21 (68) 18 (60) 36 (67) 2 (33) 35 (64) 7 (58)

Gained weight 0.681 0.480 0.298 1.000 0.160

yes 15.5 10 (25) 5 (38) 11 (35) 7 (23) 15 (28) 2 (33) 14 (25) 6 (50)

no 16.6 30 (75) 8 (62) 20 (65) 23 (77) 39 (72) 4 (67) 41 (75) 6 (50)

Headached 0.509 0.262 0.073 0.092 0.246

yes 15.5 17 (43) 8 (62) 11 (35) 17 (57) 23 (43) 5 (83) 28 (51) 4 (33)

no 16.2 22 (55) 5 (38) 20 (65) 12 (40) 30 (56) 1 (17) 26 (47) 8 (67)

Interest in sex 0.817 1.000 0.707 0.541 0.147

yes 15.6 35 (88) 11 (85) 26 (84) 27 (90) 48 (89) 5 (83) 50 (91) 9 (75)

no 16.2 5 (13) 2 (15) 5 (16) 3 (10) 6 (11) 1 (17) 5 (9) 3 (25)

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

167

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Tumor characteristics

Age at diagnosis

(years)

T stage N stage M stage Histology

Median P value T1-2

n = 40

n (%)

T3-4

n = 13

P value N0

n = 31

n (%)

N1

n=30

P value M0

n = 54

n (%)

M1

n = 6

P value PTC

n = 55

n (%)

FTC

n = 12

P value

THYCA-QOL single items5

Problems with scar 0.629 1.000 0.488 1.000 0.187

yes 16.4 5 (13) 2 (15) 4 (13) 6 (20) 8 (15) 1 (17) 10 (18) 0 (0)

no 15.7 34 (85) 11 (85) 26 (84) 24 (77) 45 (83) 5 (83) 44 (80) 12 (100)

Felt chilly 0.670 0.173 0.684 1.000 0.193

yes 16.0 21 (53) 4 (31) 14 (45) 12 (40) 22 (41) 2 (33) 25 (45) 3 (25)

no 15.6 19 (48) 9 (69) 17 (55) 18 (60) 32 (59) 4 (67) 30 (55) 9 (75)

Tingling hands/feet 0.337 0.579 0.529 0.179 0.751

yes 16.3 15 (38) 6 (46) 10 (32) 12 (40) 18 (33) 4 (67) 20 (36) 5 (42)

no 15.6 25 (63) 7 (54) 21 (68) 18 (60) 36 (67) 2 (33) 35 (64) 7 (58)

Gained weight 0.681 0.480 0.298 1.000 0.160

yes 15.5 10 (25) 5 (38) 11 (35) 7 (23) 15 (28) 2 (33) 14 (25) 6 (50)

no 16.6 30 (75) 8 (62) 20 (65) 23 (77) 39 (72) 4 (67) 41 (75) 6 (50)

Headached 0.509 0.262 0.073 0.092 0.246

yes 15.5 17 (43) 8 (62) 11 (35) 17 (57) 23 (43) 5 (83) 28 (51) 4 (33)

no 16.2 22 (55) 5 (38) 20 (65) 12 (40) 30 (56) 1 (17) 26 (47) 8 (67)

Interest in sex 0.817 1.000 0.707 0.541 0.147

yes 15.6 35 (88) 11 (85) 26 (84) 27 (90) 48 (89) 5 (83) 50 (91) 9 (75)

no 16.2 5 (13) 2 (15) 5 (16) 3 (10) 6 (11) 1 (17) 5 (9) 3 (25)

CHAPTER 7

168

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Treatment characteristics

Permanent

hypoparathyroidism

RLN injury Cumulative 131I

dose (mCi)

no HP

n = 40

Median

HP

n = 17

P value no RLN

injury

n = 41

Median

RLN

injury

n = 10

P value Corr.

coeff.

P value

SF-361

Physical functioning 100 90 0.276 95 100 0.398 -0.161 0.197

Role physical 100 100 0.654 100 100 0.940 -0.137 0.272

Bodily pain 84 84 0.730 84 84 0.308 -0.120 0.336

Social functioning 88 100 0.101 88 94 0.541 -0.118 0.342

Mental health 84 88 0.829 84 82 0.372 -0.117 0.350

Role emotional 100 100 0.681 100 100 0.637 0.047 0.706

Vitality 65 75 0.037 65 78 0.079 -0.205 0.099

General health 77 72 0.681 72 71 0.865 -0.176 0.157

Physical component scale 54 53 0.913 54 55 0.346 -0.259 0.037

Mental component scale 52 55 0.105 52 52 0.747 -0.147 0.244

MFI-202 General fatigue 10 9 0.285 10 8 0.068 0.145 0.241

Physical fatigue 7 7 0.580 8 5 0.014 0.109 0.378

Reduced activity 8 5 0.091 8 5 0.389 -0.026 0.832

Reduced motivation 8 5 0.365 7 5 0.519 0.005 0.965

Mental fatigue 10 7 0.380 10 8 0.811 0.030 0.807

Total fatigue 43 34 0.171 41 35 0.132 0.087 0.483

HADS3 Anxiety 4 3 0.726 3 5 0.720 0.038 0.759

Depression 1 0 0.388 1 2 0.931 0.006 0.960

Total HADS 6 5 0.490 6 6 0.730 0.028 0.821

THYCA-QOL scales4

Neuromuscular 11 22 0.055 11 19 0.344 0.206 0.095

Voice 0 17 0.732 0 17 0.116 -0.040 0.747

Concentration 0 0 0.957 0 0 0.862 0.210 0.088

Sympathetic 0 17 0.089 0 17 0.134 0.159 0.197

Throat/mouth 11 11 0.861 11 11 0.313 -0.028 0.822

Psychological 8 8 0.906 8 8 0.739 0.037 0.764

Sensory 17 17 0.748 17 17 0.763 0.020 0.872

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

169

7

Supplemental Table 3. (continued)

Questionnaire - (sub)scale Treatment characteristics

Permanent

hypoparathyroidism

RLN injury Cumulative 131I

dose (mCi)

no HP

n = 40

n (%)

HP

n = 17

P value no RLN

injury

n = 41

n (%)

RLN

injury

n = 10

P value Median P value

THYCA-QOL single items5

Problems with scar 0.250 0.397 0.105

yes 8 (20) 1 (6) 7 (17) 3 (30) 347.0

no 31 (78) 16 (94) 33 (80) 7 (70) 155.5

Felt chilly 0.738 0.167 0.279

yes 16 (40) 6 (35) 19 (46) 2 (20) 200.0

no 24 (60) 11 (65) 22 (54) 8 (80) 151.0

Tingling hands/feet 0.297 0.722 0.037

yes 13 (33) 8 (47) 16 (39) 5 (50) 200.0

no 27 (68) 9 (53) 25 (61) 5 (50) 150.0

Gained weight 0.751 0.250 0.476

yes 10 (25) 5 (29) 13 (32) 1 (10) 179.5

no 30 (75) 12 (71) 28 (68) 9 (90) 157.0

Headached 0.150 0.480 0.006*

yes 21 (53) 5 (29) 17 (41) 6 (60) 262.5

no 19 (48) 11 (65) 23 (56) 4 (40) 101.4

Interest in sex 0.164 0.250 0.847

yes 34 (85) 17 (100) 38 (93) 8 (80) 159.0

no 6 (15) 0 (0) 3 (7) 2 (20) 155.5

CHAPTER 7

170

7

Supplemental Table 3. (continued)

Questionnaire -

(sub)scale

Follow-up characteristics

TSH levels

(mU/L)

Follow-up

duration

(years)

Outcome

Corr.

coeff.

P value Corr.

coeff.

P value Remission

n = 55

Median

Recurrence

n = 7

Persistent

disease

n = 5

P value

SF-361

Physical

functioning

-0.020 0.874 -0.186 0.134 98 85 95 0.082

Role physical -0.020 0.871 0.018 0.889 100 100 100 0.756

Bodily pain -0.057 0.651 0.161 0.197 84 84 84 1.000

Social

functioning

-0.093 0.453 0.004 0.972 100 88 88 0.242

Mental health -0.095 0.447 0.012 0.923 88 76 80 0.223

Role emotional -0.106 0.396 -0.022 0.860 100 100 100 0.589

Vitality 0.009 0.943 0.249 0.044 70 50 60 0.118

General health -0.016 0.901 -0.025 0.845 77 62 69 0.309

Physical

component scale

-0.015 0.906 0.050 0.695 54 52 52 0.586

Mental

component scale

0.026 0.837 0.118 0.350 54 50 52 0.180

MFI-202 General fatigue 0.072 0.560 -0.050 0.686 10 10 11 0.694

Physical fatigue 0.070 0.571 0.034 0.785 7 11 6 0.065

Reduced activity 0.169 0.173 -0.001 0.996 8 7 7 0.431

Reduced

motivation

0.026 0.834 0.006 0.962 6 7 8 0.542

Mental fatigue -0.040 0.751 0.025 0.839 8 13 12 0.330

Total fatigue 0.017 0.891 -0.022 0.860 36 41 41 0.396

HADS3 Anxiety 0.074 0.549 0.011 0.929 4 5 5 0.945

Depression -0.005 0.966 0.039 0.754 1 0 5 0.645

Total HADS 0.042 0.733 0.028 0.821 5 7 10 0.938

THYCA-QOL scales4

Neuromuscular -0.069 0.579 0.062 0.617 11 22 17 0.051

Voice -0.139 0.263 0.044 0.721 0 17 0 0.289

Concentration -0.087 0.484 0.192 0.120 0 33 33 0.043

Sympathetic -0.149 0.230 0.098 0.432 0 17 17 0.294

Throat/mouth -0.061 0.622 0.026 0.836 11 11 0 0.947

Psychological -0.073 0.555 0.041 0.744 8 17 8 0.369

Sensory -0.079 0.523 0.043 0.732 17 17 17 0.799

QUALITY OF LIFE AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

171

7

Supplemental Table 3. (continued)

Questionnaire -

(sub)scale

Follow-up characteristics

TSH levels

(mU/L)

Follow-up

duration

(years)

Outcome

Median P value Median P value Remission

n = 55

n (%)

Recurrence

n = 7

Persistent

disease

n = 5

P value

THYCA-QOL single items5

Problems with scar 0.002* 0.474 <0.001*

yes 0.06 12.8 4 (7) 1 (14) 5 (100)

no 0.20 18.2 50 (91) 6 (86) 0 (0)

Felt chilly 0.731 0.525 0.116

yes 0.18 17.6 20 (36) 4 (57) 4 (80)

no 0.17 18.2 35 (64) 3 (43) 1 (20)

Tingling hands/feet 0.525 0.650 0.257

yes 0.19 18.1 18 (33) 4 (57) 3 (60)

no 0.16 17.6 37 (67) 3 (43) 2 (40)

Gained weight 0.763 0.244 1.000

yes 0.20 21.5 17 (31) 2 (29) 1 (20)

no 0.16 17.4 38 (69) 5 (71) 4 (80)

Headached 0.663 0.275 0.251

yes 0.18 14.9 26 (47) 5 (71) 1 (20)

no 0.17 19.3 28 (51) 2 (29) 4 (80)

Interest in sex 0.699 0.428 0.191

yes 0.17 17.5 50 (91) 5 (71) 4 (80)

no 0.18 24.8 5 (9) 2 (29) 1 (20)

Significant values (P <0.01) are marked with an asterisk (*). Abbreviations: Corr. coeff., correlation coefficient; PTC,

papillary thyroid cancer; FTC, follicular thyroid cancer; HP, hypoparathyroidism; RLN, recurrent laryngeal nerve;

TSH, thyroid-stimulating hormone. 1 Short Form-36. Higher scores indicate a better health-related quality of life,

subscales range from 0-100. 2 Multidimensional Fatigue Inventory 20. Higher scores indicate more fatigue, subscales

range from 4-20, the total scale ranges from 20-100. 3 Hospital Anxiety and Depression Scale. Higher scores indicate

higher levels anxiety and/or depression, subscales range from 0-21, the total scale ranges from 0-42. 4 A higher score

indicates more symptoms, subscales range from 0-100. 5 Thyroid cancer-specific health-related quality of life. Not at

all and a little = no, quite a bit and very much = yes.

Chapter 8

Chapter 8

CHAPTER 8

Psychosocial development in survivors of

childhood differentiated thyroid carcinoma:

a cross-sectional study

Marloes Nies, Bernadette L. Dekker, Esther Sulkers, Gea A. Huizinga, Mariëlle S. Klein Hesselink, Heleen Maurice-Stam, Martha A. Grootenhuis, Adrienne H. Brouwers, Johannes G. M. Burgerhof, Eveline W.C.M. van Dam, Bas Havekes, Marry M. van den Heuvel-Eibrink,Eleonora P.M. Corssmit, Leontien C.M. Kremer, Romana T. Netea-Maier, Heleen J.H. van der Pal, Robin P. Peeters, John T.M. Plukker, Cécile M. Ronckers, Hanneke M. van Santen, Anouk N.A. van der Horst-Schrivers, Wim J.E. Tissing, Gianni Bocca, and Thera P. Links

Eur J Endocrinol. 2018;178(3):215–223.

CHAPTER 8

174

8

ABSTRACTObjectiveThe impact of childhood differentiated thyroid carcinoma (DTC) on psychosocial development has not yet been studied. The aim of this study was to evaluate the achievement of psychosocial developmental milestones in long-term survivors of childhood DTC.

Design and methodsSurvivors of childhood DTC diagnosed between 1970 and 2013 were included. Reasons for exclusion were age <18 or >35 years at follow-up, a follow-up period <5 years or diagnosis with DTC as a second malignant neoplasm. Survivors gathered peer controls of similar age and sex (n = 30). A comparison group non-affected with cancer (n = 508) and other childhood cancer survivors (CCS) were also used to compare psychosocial development. To assess the achievement of psychosocial milestones (social, autonomy, and psychosexual development), the course of life questionnaire (CoLQ) was used.

ResultsWe included 39 survivors of childhood DTC (response rate 83.0%, mean age at diagnosis 15.6 years, and mean age at evaluation 26.1 years). CoLQ scores did not significantly differ between survivors of childhood DTC and the two non-affected groups. CoLQ scores of childhood DTC survivors were compared to scores of other CCS diagnosed at similar ages (n = 76). DTC survivors scored significantly higher on social development than other CCS, but scores were similar on autonomy and psychosexual developmental scales.

ConclusionsSurvivors of childhood DTC showed similar development on social, autonomy, and psychosexual domains compared to non-affected individuals. Social development was slightly more favorable in DTC survivors than in other CCS, but was similar on autonomy and psychosexual domains.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

175

8

BACKGROUNDChildhood diagnosis of thyroid cancer is rare. Age-adjusted incidence rates of pediatric thyroid cancer are reported between 0.6 and 1.2 per 100 000 per year, but rates are increasing (1-3). Differentiated thyroid carcinoma (DTC) is the most frequently diagnosed histological subtype of this disease, accounting for over 90% of the thyroid cancers in children (1). The incidence of childhood DTC peaks during puberty and mainly affects girls (4, 5). The effect of DTC on the psychosocial development of children diagnosed with this disease has not been studied.

Childhood cancer arises in a period where several psychosocial developmental milestones are reached (6), also referred to as ‘the course of life’ (7). The psychosocial development from childhood toward adulthood involves increasing independence, growth of more symmetrical parent–adolescent relationships, maturation of personality and identity, and encountering and maintaining (first) psychosexual and social contacts (8). The diagnosis and treatment of childhood cancer may impair the psychosocial development as some childhood cancer survivors (CCS) have been shown to be hindered in their development. For example, CCS had fewer friends and tended to be older when they experienced their first sexual intimacy. However, data are inconclusive (7, 9-12). In survivors of various types of childhood cancer the biological behavior of the cancer and the impact of treatment interfere differently in the course of life: survivors of brain tumors seem most severely affected (9-11, 13).

Unfortunately, despite the excellent cure rates, most DTC survivors remain patients for the rest of their lives. Treatment of DTC consists of a thyroidectomy, and in case of lymph node metastases, it is extended with a lymph node dissection in the neck. In the Netherlands, about a third of the survivors of childhood DTC experience surgical complications: the most common complications are hypoparathyroidism and recurrent laryngeal nerve damage (5). Surgery is followed by one or more administrations of radioactive iodine (131I). During the 131I administrations, patients are withdrawn from thyroid hormone for several weeks; this causes deep hypothyroidism, involving changes in metabolism (14). Initial treatment usually takes from one to several years. Subsequently, lifelong thyroid hormone therapy is initiated. Especially for high-risk patients, relatively high levels of thyroid hormone are prescribed to induce subclinical hyperthyroidism, which decreases the risk of cancer recurrence (15). However, large fluctuations in thyroid hormone levels are known to have a great impact on well-being, behavior, and learning ability (16, 17). Childhood DTC has an excellent survival, with 5-year and 10-year survival rates >95% (4, 5). Furthermore, specific aspects of DTC survivorship include lifelong use of hormone medication and blood tests as well as the possibility of cancer recurrences even after decades of disease-free survival (18).

An explorative study was set up, ultimately to evaluate whether interventions for these survivors are necessary. Our aims were (I) to compare the developmental milestones reached by adult survivors of childhood DTC and by individuals non-

CHAPTER 8

176

8

affected with cancer; (II) to compare psychosocial development in adult survivors of childhood DTC and in survivors of other types of childhood cancer and (III) to assess whether the psychosocial development of survivors of childhood DTC is associated with medical characteristics.

SUBJECTS AND METHODSThis cross-sectional study was performed in the context of a Dutch nationwide study examining medical characteristics and long-term treatment effects (including a paper questionnaire) of childhood DTC from 2012 to 2014 (5). The Institutional Review Board of the University Medical Center Groningen approved the study on behalf of all participating institutions (ABR NL40572.042.12, file number 2012/183). This study was registered in the Netherlands Trial Registry (trial registration number 3448).

ParticipantsDTC survivorsFor the nationwide study, all patients diagnosed with DTC at age ≤18 years between 1970 and 2013 treated in the Netherlands were included (5). Exclusion criteria were, as previously reported: follow-up <5 years after diagnosis, attained age <18 years, diagnosis of DTC as a second malignant neoplasm, lack of understanding of the Dutch language, and 131I administration within three months before evaluation (19). For the current study, survivors aged >35 years during evaluation were also excluded because the questionnaire is validated only up to the age of 35 years (7).

Groups used for comparisonComparison to individuals unaffected by cancer

DTC survivors were compared to a group of persons unaffected by cancer from the survivors’ environment (peer controls) and compared to an unaffected group reflecting the general population (comparison group).

Peer controls: DTC survivors were asked to find one or two peers of the same sex and age (range plus or minus 5 years from the survivor’s age at evaluation, minimum age 18 years). Controls who had a medical history of malignancy were excluded. Although we preferred peers (e.g. friends, colleagues), we allowed inclusion of family members, provided they met the in/exclusion criteria (19). For the current substudy, peer controls aged >35 years were also excluded.

All DTC survivors and peer controls gave written informed consent before participating in the study.

Comparison group: A comparison group, gathered by general practitioners in a previous study, was used (7). For this comparison, group persons aged 18–30 years, with no history of cancer and with the ability to understand Dutch questionnaires

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

177

8

were included. This group consisted of 508 persons (consisting of 53% females and 47% males).

Comparison to survivors of other childhood cancers

Survivors of other childhood cancers: For this comparison, we used data of other CCS, gathered in a study by Stam and coworkers (7). These survivors were diagnosed with cancer at age <18 years, were at least 5 years in follow-up, aged 17–30 years during evaluation, and were able to understand Dutch questionnaires. The different cancer diagnoses were grouped as leukemia/lymphoma (e.g. acute lymphoblastic leukemia, (non-) Hodgkin lymphoma), solid tumors (e.g. rhabdomyosarcoma, osteosarcoma) and brain tumors (7). Three hundred and fifty-three CCS were eligible for comparison. Survivors of thyroid cancer (n = 3) were excluded from analyses since we only wanted to compare DTC survivors to other types of childhood cancer. Subsequently, 350 CCS of other types of cancer (hereafter referred to as CCS) were available for comparison.

MeasuresPsychosocial developmentPsychosocial development was assessed using the course of life questionnaire (CoLQ) (7), which measures achievement of developmental milestones on five domains: (1) social development, (2) autonomy development, (3) psychosexual development, (4) antisocial behavior, and (5) substance use and gambling. To restrict the length of the questionnaire, for the current study only domains 1–3 were evaluated. Possible scores for each item were scored 1 (milestone not (yet) achieved) or 2 (milestone achieved). Scores were added-up to form 3 scales: social development (12 items, range: 12–24), autonomy development (6 items, range: 6–12) and psychosexual development (4 items, range: 4–8). Higher scores indicate earlier achievement or achievement of more developmental milestones. The validity of the course of life scales is good. The internal consistency of the scales is satisfactory, except the autonomy scale, probably because the items refer to diverging aspects of autonomy (7). The Cronbach’s alphas in the populations under study were moderate to good: (1) social development (range: 12–24): DTC survivors 0.70, peer controls 0.76, comparison group 0.71, CCS 0.75; (2) autonomy development (range: 6–12): DTC survivors 0.52, peer controls 0.49, comparison group 0.49, CCS 0.43; (3) psychosexual development (range: 4–8): DTC survivors 0.91, peer controls 0.75, comparison group 0.71, CCS 0.74.

Sociodemographic and medical dataSociodemographics (marital status, educational level (low, medium and high) and employment status) and data regarding disease, treatment, and follow-up characteristics were retrieved from medical records and a self-administered questionnaire.

CHAPTER 8

178

8

Statistical methodsData are presented as median (interquartile range) unless otherwise specified. Characteristics and scores on the CoLQ (on scale level and item level) of survivors of childhood DTC were compared separately with the characteristics or scores of the peer controls, the comparison group and the CCS (total, and divided by type of diagnosis). Associations between medical characteristics and scores on developmental domains were evaluated.

χ 2 tests or Fisher’s exact tests (if >20% of the cells had an expected count of <5) were used for categorical variables. Mann–Whitney U and Kruskal–Wallis tests were performed for non-normally distributed continuous or ordinal variables. Spearman’s rank correlation coefficient was used to correlate two non-normally distributed continuous and/or ordinal variables. The tests performed are described in the table legends. Missing or unknown values were excluded from statistical testing (pairwise deletion). To compensate for multiple testing, we considered differences to be statistically significant at P < 0.01. All tests were two-sided. IBM SPSS Statistics for Windows, version 23 (IBM) was used for statistical analyses.

RESULTSI) Comparison to individuals unaffected with cancerSample characteristicsOne hundred and five survivors were included in the nationwide study (of 169 eligible subjects, response rate 62.1%) (5). Of the 47 survivors eligible for the current study, 39 (83.0%) participated (Supplementary Figure 1, see section on supplementary data given at the end of this article). The survivors gathered 59 peer controls, 30 of whom participated: 26 peer controls were excluded because of an age >35 years and 3 peer controls were excluded because they did not meet matching criteria.

Demographic characteristics appear in Table 1. Median age of the DTC survivors at evaluation was 26.2 years (range 18.8–35.7), compared to a median age of 25.8 years (range 19.4–34.4, P = 0.628) of peer controls and a median age of 23.8 years of the comparison group (range 18.0–31.0, P = 0.012). The group of DTC survivors had significantly more females than the comparison group (87% vs. 53%, P < 0.001). DTC survivors reported significantly more frequently higher levels of education than the comparison group (P = 0.002). Other characteristics did not significantly differ between DTC survivors and the other two groups.

All DTC survivors and peer controls had the Dutch nationality. Ninety-seven percent of the comparison group had the Dutch nationality; this was not significantly different from the DTC survivors (P = 0.558).

Psychosocial developmentBecause scores on psychosocial development did not differ between males and females in all evaluated groups (data not shown), no correction for sex was performed.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

179

8

Scale scores: Scores of the survivors of childhood DTC on all three developmental milestone scales (i.e. social development, autonomy development and psychosexual development) did not differ significantly from those of peer controls or of the comparison group (P = 0.592, P = 0.084, P = 0.841, P = 0.233, P = 0.241, and P = 0.556, respectively, Table 2).

Item scores: Item scores regarding social development, autonomy development and psychosexual development during or after secondary school did not differ between survivors of childhood DTC and the peer controls or the comparison group (Supplementary Table 1a, b, and c).

Table 1. Characteristics of survivors of childhood DTC vs. individuals non-affected with cancer

Characteristic DTC Survivors

n = 39

Peer controls

n = 30

P Value Comparison group

n = 508

P Value

Age at evaluation, years 26.2 (18.8-35.7) 25.8 (19.4-34.4) 0.628† 23.8 (18.0-31.0) 0.012†

Sex, n (%) 0.690‡ <0.001§

Female 34 (87) 28 (93) 269 (53)

Male 5 (13) 2 (7) 239 (47)

Employment, n (%) 0.576‡ 1.000‡

Employed and/or student 38 (97) 28 (93) 486 (96)

Not employed and

no student

1 (3) 2 (7) 21 (4)

Missing 0 (0) 0 (0) 1 (0)

Completed education, n (%) 0.035‡ 0.002§

Low level 7 (18) 0 (0) 143 (28)

Medium level 15 (39) 12 (40) 246 (48)

High level 17 (44) 18 (60) 97 (19)

Missing 0 (0) 0 (0) 22 (4)

Marital status‖, n (%) 0.303§ n.a.

Relationship 24 (62) 22 (73) -

No relationship 15 (38) 8 (27) -

Marital status‖, n (%) n.a. n.a.

Not married and not

living together

- - 299 (59)

Married or living together - - 192 (38)

Missing - - 17 (3)

†, Mann Whitney U test; ‡, Fisher’s Exact Test; §, Chi square test. ‖, answer options regarding marital status of two

different questionnaires were non-mergeable, therefore these are shown separately. Missing values were excluded

for statistical testing (pairwise deletion). Continuous variables are presented as median (range). P values in bold

indicate a statistically significant difference (P < 0.01). Abbreviations: DTC, differentiated thyroid carcinoma; n.a., not

applicable; CCS, childhood cancer survivors.

CHAPTER 8

180

8 II) Comparison to survivors of other types of childhood cancerSample characteristicsAge at diagnosis was significantly different between survivors of childhood DTC and

other CCS (15.6 vs. 6.3 years old, respectively, P < 0.001); the large majority (35 out of

39, 90%) of survivors of childhood DTC were diagnosed during secondary school. We

therefore chose to focus primarily on survivors (DTC and other CCS) diagnosed at age

≥12 years (the age at which Dutch children generally start secondary school).

Thirty-five of the DTC survivors were diagnosed at age ≥12 years (Supplementary

Figure 1). Of the 350 CCS non-affected with thyroid cancer 76 were diagnosed at age

≥12 years.

As shown in Table 3, the median age of these 35 DTC survivors at diagnosis was 15.8

years (range 12.0–18.7), and the median age at diagnosis of CCS was 14.0 years (range

12.0–17.0, P < 0.001). The group of DTC survivors included significantly more females

than the CCS (91% vs. 55%, P < 0.001). Median follow-up period for DTC survivors was

10.7 years (range 5.0–23.3), and for the CCS, it was 12.0 years (range 6.2–18.1, P = 0.088).

DTC survivors and CCS did not differ significantly on other characteristics. Ninety-

seven percent of the CCS group were Dutch; this was not significantly different from

the DTC survivors (P = 1.000).

Psychosocial developmentScale scores: DTC survivors scored higher on social development than did CCS (both

diagnosed at ≥12 years, 22 (21, 23) vs. 21 (19, 22) out of 24, respectively, P = 0.005). DTC

survivors also scored higher on social development compared to survivors of childhood

leukemia diagnosed at age ≥12 years (22 (21, 23) vs. 20 (18, 22) respectively, P = 0.001),

but their scores were not significantly different from those of survivors of solid tumors

Table 2. Scores on psychosocial developmental domains: comparison of survivors of childhood DTC with

individuals non-affected with cancer

Domain DTC

survivors

n = 39

Peer

controls

n = 30

P Value Comparison

group

n = 508

P Value

Social development † 22 (20.3, 23) 22 (18, 23) 0.592 21 (19, 23) 0.233

Autonomy development ‡ 9 (8, 10) 10 (9 ,11) 0.084 9 (8, 11) 0.241

Psycho-sexual development § 8 (6.5, 8) 8 (6, 8) 0.841 8 (7, 8) 0.556

Scores are shown as median (p25, p27). Comparisons between DTC survivors and other groups were performed

using Mann Whitney U tests. Higher scores indicate earlier achievement or achievement of more psychosocial

developmental milestones; †, scale ranges 12-24; ‡, scale ranges 6-12; §, scale ranges 4-8. Missing values were excluded

from statistical testing (pairwise deletion). P values in bold indicate a significant value (P <0.01). Abbreviations: DTC,

differentiated thyroid carcinoma; CCS, childhood cancer survivors.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

181

8

or brain tumors. Scale scores of CCS and subgroups of CCS, compared to DTC survivors, did not differ on autonomy development and psychosexual development (Table 4).

Item scores: Given the age at diagnosis of DTC survivors, only items that apply to the period during or after secondary school are discussed. Item scores regarding social development, autonomy development, and psychosexual development during or after secondary school did not differ between survivors of DTC and CCS diagnosed at ≥12 years old (Figure 1 and Supplemental Table 2a, b and c).

Characteristics and psychosocial development scale scores of DTC survivors and CCS diagnosed at all ages are shown in Supplemental Tables 3 and 4.

Table 3. Characteristics of survivors of childhood DTC vs. survivors of other childhood cancers (both

diagnosed age ≥12 years old)

Characteristic DTC Survivors

n = 35

Other CCS

n = 76

P Value

At diagnosisAge at diagnosis, years 15.8 (12.0-18.7) 14.0 (12.0-17.0) <0.001†

Sex, n (%) <0.001§

Female 32 (91) 42 (55)

Male 3 (9) 34 (45)

At follow-upAge at evaluation, years 26.3 (18.8-35.7) 26.2 (18.9-31.1) 0.741†

Follow-up period, years 10.7 (5.0-23.3) 12.0 (6.2-18.1) 0.088†

Employment, n (%) 1.000‡

Employed and/or student 34 (97) 72 (95)

Not employed and no student 1 (3) 3 (4)

Missing 0 (0) 1 (1)

Completed education, n (%) 0.237§

Low level 6 (17) 15 (20)

Medium level 13 (37) 36 (47)

High level 16 (46) 21 (28)

Missing 0 (0) 4 (5)

Marital status‖, n (%) n.a.

Relationship 23 (66) -

No relationship 12 (34) -

Marital status‖, n (%) n.a.

Not married and not living together - 31 (55)

Married or living together - 31 (41)

Missing - 3 (4)

†, Mann Whitney U test; ‡, Fisher’s Exact Test; §, Chi square test. ‖, answer options regarding marital status of two

different questionnaires were non-mergeable, therefore these are shown separately. Missing values were excluded

for statistical testing (pairwise deletion). Continuous variables are presented as median (range). P values in bold

indicate a statistically significant difference (P < 0.01). Abbreviations: DTC, differentiated thyroid carcinoma; n.a., not

applicable; CCS, childhood cancer survivors.

CHAPTER 8

182

8

Figure 1. Scores of survivors of childhood DTC (n = 35) and childhood cancer survivors (CCS, n = 76)

of other types of cancer, both diagnosed at age ≥12 years old, on individual items of the CoLQ. Scores

indicate the percentage of the group that has reached the developmental milestone. Dark bars: DTC

survivors, light bars: other CCS. An asterisk (*) indicates a P value <0.01.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

183

8

Tab

le 4

. Sco

res

on

psyc

hoso

cial

dev

elo

pmen

tal d

om

ains

: co

mpa

riso

n o

f sur

vivo

rs o

f chi

ldho

od

DTC

wit

h su

rviv

ors

of o

ther

chi

ldho

od

canc

ers

(bo

th d

iagn

ose

d

age

≥12

year

s o

ld)

Do

mai

nD

TC

surv

ivo

rs

n =

35

Ch

ildh

oo

d C

ance

r Su

rviv

ors

(C

CS)

Tota

l CC

S

n =

76

P V

alue

Leuk

emia

/

lym

ph

om

a

n =

45

P V

alue

Solid

tum

ors

n =

23

P V

alue

Bra

in t

umo

rs

n =

8

P V

alue

Soci

al d

evel

op

men

t †

22 (

21, 2

3)21

(19

, 22)

0.0

05

20 (

18, 2

2)0.

00

122

(19

, 23)

0.16

421

.5 (

20.3

, 22.

8)0

.391

Aut

on

om

y d

evel

op

men

t ‡

9 (8

,10)

9 (8

, 10

)0

.688

10 (

8, 10

)0

.753

9 (8

, 10

.5)

0.6

779

(7.5

, 11)

0.8

90

Psyc

ho

-sex

ual d

evel

op

men

t §

8 (7

, 8)

7 (6

, 8)

0.0

207

(6, 8

)0

.040

7 (6

, 8)

0.0

835.

5 (4

.3, 8

)0

.097

Sco

res a

re sh

ow

n as

med

ian

(p25

, p27

). C

om

pari

sons

bet

wee

n D

TC su

rviv

ors

and

oth

er g

roup

s wer

e p

erfo

rmed

usi

ng M

ann

Whi

tney

U te

sts.

Hig

her s

core

s ind

icat

e ea

rlie

r ach

ieve

men

t

or

achi

evem

ent

of m

ore

psy

cho

soci

al d

evel

opm

enta

l mile

sto

nes;

†, sc

ale

rang

es 1

2-24

; ‡, s

cale

ran

ges

6-12

; §, s

cale

ran

ges

4-8.

Mis

sing

val

ues

wer

e ex

clud

ed fr

om

sta

tist

ical

tes

ting

(pai

rwis

e d

elet

ion)

. P v

alue

s in

bo

ld in

dic

ate

a si

gnifi

cant

val

ue (

P <0

.01)

. Abb

revi

atio

ns: D

TC, d

iffer

enti

ated

thy

roid

car

cino

ma;

CC

S, c

hild

hoo

d c

ance

r su

rviv

ors

.

CHAPTER 8

184

8

Possible determinants of psychosocial developmentDisease- treatment and, follow-up characteristics were not significantly associated with scores on domains of psychosocial development (Supplemental Table 5).

DISCUSSIONThe current study shows similar achievement of psychosocial developmental milestones in long-term survivors of childhood DTC as compared to non-affected groups. A slightly better social development in DTC survivors was observed compared to other CCS; differences with leukemia survivors were the most pronounced. Medical characteristics were not associated with a better or worse psychosocial development. Altogether, this indicates that even though the diagnosis of DTC during childhood is a life-altering event, the disease does not seem to have consequences related to altered psychosocial development. Current results align with the overall normal QoL reported in a previous study describing the same cohort (19).

Current results imply that survivors of childhood DTC may be less restricted in their psychosocial development than other CCS; previous studies report hampered psychosocial development in other CCS compared to a non-affected comparison group of similar sex and age (7). This indicates that the degree to which a child is hindered in his or her development depends on the type of cancer (13). For instance, survivors of brain tumors and CCS treated with neurotoxic treatment (i.e. cranial irradiation, intrathecal chemotherapy and specific intravenous chemotherapies) were found to be most vulnerable to impairment of psychosexual and social development as compared to other CCS (10, 11, 13, 20). However, this finding was not confirmed in the current study. This could be due to the fact that results in the current study were based on brain tumor survivors diagnosed during secondary school: brain tumor survivors diagnosed at younger ages were not represented while younger age at diagnosis of a brain tumor is associated with more developmental impairment (21).

Current results allow for conclusions regarding the majority of survivors of childhood DTC that was diagnosed during secondary school. Diagnosis of childhood DTC before secondary school occurs less frequently, therefore, concise conclusions cannot be made.

Factors that could interfere with psychosocial development (e.g. sex, age at diagnosis, age at follow-up or follow-up duration: Supplemental Table 5) were evaluated, but surprisingly showed no significant associations with psychosocial development. It may be that other, not investigated, factors may have played a role.

A first possible explanation for the normal psychosocial development of survivors of childhood DTC could lie in the excellent prognosis of the disease. In addition, the indolent course of DTC allows for flexibility in treatment, thus accommodating school schedules. In general, attending school benefits social development. The more favorable social development in DTC survivors compared to other CCS emphasizes

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

185

8

this explanation. However, this statement remains speculative since we did not study school attendance. DTC treatment modalities differ from those of other types of cancer, using chemotherapeutics administered over longer periods of time. Another explanation could be the age upon diagnosis of most children with DTC. Relatively older upon diagnosis, these children as well as their parents have already experienced considerable developmental progression. For example, the foundations for social interactions (friendly as well as romantic) have already been formed. Lastly, fluctuations in thyroid hormone levels during periods of thyroid hormone withdrawal or long-term treatment effects of DTC, such as a weakened voice or the need for medication monitoring, may have less impact on psychosocial development than the aforementioned neurotoxicity and other physical or mental sequelae involved in other childhood cancer treatments (9, 10, 12, 22). It is common practice to substitute DTC survivors with levothyroxine after initial treatment and only use triiodothyronine in preparation for treatment with 131I (23). As a result, current results suggest that neurological development of childhood DTC survivors is not affected by this approach to treatment.

Strengths and limitationsThere is a great lack of knowledge regarding the long-term impact of childhood thyroid cancer on psychosocial domains (23). A strength of the current study is that it is the first to evaluate psychosocial development in a, though relatively small, unique cohort of survivors of childhood DTC. Using various groups for comparison allowed psychosocial development in these survivors to be placed in different perspectives. However, when interpreting the results, one must keep in mind the limitations of the study. One cannot use the results of this cross-sectional design in long-term survivors to elaborate about psychosocial development in the first 5 years after diagnosis, but eventual long-term results are promising. Not all predictors of psychosocial development were evaluated; for instance, we did not include the dependency of autonomy development on parenting behavior. Moreover, since most DTC survivors were diagnosed during secondary school, the items regarding elementary school were less relevant for the current population. However, the CoLQ is only validated containing all items (7).

Clinical implicationsCurrent results may be reassuring for children newly diagnosed with DTC, and for their families and caregivers, regarding the possible psychosocial impact of their disease. However, the results do not imply that physicians need not monitor problems with psychosocial development in these survivors. This study presents data of a group of survivors, but individual differences should not be overlooked. Presenting patients with thyroid cancer as a good cancer makes them feel that physicians are downplaying their cancer experiences (24).

CHAPTER 8

186

8

In conclusion, the current study aimed to evaluate the achievement of psychosocial developmental milestones in survivors of childhood DTC and found no delay in autonomy, social, and psychosexual domains after diagnosis compared to individuals non-affected with cancer. It did find a slightly more favorable development in DTC survivors compared to other CCS. However, before drawing definite conclusions, current findings need to be confirmed in subsequent studies.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

187

8

ACKNOWLEDGEMENTSThe authors are grateful to their colleagues in the Netherlands for referring patients for this study. We thank the registration teams of the Comprehensive Cancer Centers for the collection of data for the Netherlands Cancer Registry, as well as the scientific staff of the Netherlands Cancer Registry.

FUNDINGThis work was supported by Stichting Kinderen Kankervrij (the Netherlands, Foundation Children Cancer-Free, Project 81). C M R is supported by the Dutch Cancer Society.

DECLARATION OF INTERESTThe authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

CHAPTER 8

188

8

REFERENCES1. Dermody S, Walls A, Harley, EH, Jr. Pediatric thyroid cancer: an update from the SEER database 2007–2012. Int J

Pediatr Otorhinolaryngol. 2016;89:121–126.

2. Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I, Parkin DM. Thyroid cancer incidence and survival

among European children and adolescents (1978–1997): report from the Automated Childhood Cancer Information

System project. Eur J Cancer. 2006;42(13):2150–2169.

3. Siegel DA, King J, Tai E, Buchanan N, Ajani UA, Li J. Cancer incidence rates and trends among children and

adolescents in the United States, 2001–2009. Pediatrics. 2014;134(4):945.

4. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167–172.

5. Klein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-

Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Schmid KW, Smit JW, Williams

GR, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP. Pediatric differentiated thyroid carcinoma in

the Netherlands: a nationwide follow-up study. J Clin Endocrinol Metab. 2016;101(5):2031–2039.

6. Hay WJ, Levin M, Deterding R, Abzug M. Current Diagnosis and Treatment Pediatrics 23rd ed. New York, NY:

McGraw-Hill; 2016.

7. Stam H, Grootenhuis MA, Last BF. The course of life of survivors of childhood cancer. Psycho-Oncology. 2005;14(3):227–238.

8. Meeus W. Adolescent psychosocial development: a review of longitudinal models and research. Dev

Psych. 2016;52(12):1969–1993.

9. Font-Gonzalez A, Feijen EL, Sieswerda E, van Dulmen-den Broeder E, Grootenhuis M, Maurice-Stam H, Caron H,

Essink-Bot ML, van der Pal H, Geskus R, Kremer L. Social outcomes in adult survivors of childhood cancer compared

to the general population: linkage of a cohort with population registers. Psycho-Oncology. 2016;25(8):933–941.

10. Lehmann V, Tuinman MA, Keim MC, Winning AM, Olshefski RS, Bajwa RPS, Hagedoorn M Gerhardt CA. Psychosexual

development and satisfaction in long-term survivors of childhood cancer: neurotoxic treatment intensity as a risk

indicator. Cancer. 2017;123(10):1869–1876.

11. Gurney JG, Krull KR, Kadan-Lottick N, Nicholson HS, Nathan PC, Zebrack B, Tersak JM, Ness KK. Social outcomes

in the Childhood Cancer Survivor Study cohort. J Clin Oncol. 2009;27(14):2390–2395.

12. Frederick NN, Recklitis CJ, Blackmon JE, Bober S. Sexual dysfunction in young adult survivors of childhood cancer.

Pediatr Blood Cancer. 2016;63(9):1622–1628.

13. Maurice-Stam H, Grootenhuis MA, Caron HN, Last BF. Course of life of survivors of childhood cancer is related to

quality of life in young adulthood. J Psychosoc Oncol. 2007;25(3):43–58.

14. Maxon HR 3rd, Smith HS. Radioiodine-131 in the diagnosis and treatment of metastatic well differentiated thyroid

cancer. Endocrinol Metab Clin North Am. 1990;19(3):685–718.

15. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA, Udelsman R. The treatment of

differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocr

Rev. 2011;32(6):798–826.

16. Schroeder PR, Haugen BR, Pacini F, Reiners C, Schlumberger M, Sherman SI, Cooper DS, Schuff KG, Braverman

LE, Skarulis MC, Davies TF, Mazzaferri EL, Daniels GH, Ross DS, Luster M, Samuels MH, Weintraub BD, Ridgway

EC, Ladenson PW. A comparison of short-term changes in health-related quality of life in thyroid carcinoma

patients undergoing diagnostic evaluation with recombinant human thyrotropin compared with thyroid hormone

withdrawal. J Clin Endocrinol Metab. 2006;91(3):878–884.

17. Bauer AJ. Approach to the pediatric patient with Graves’ disease: when is definitive therapy warranted? J Clin

Endocrinol Metab. 2011;96(3):580–588.

18. Mazzaferri EL, Kloos RT. Clinical review 128: current approaches to primary therapy for papillary and follicular

thyroid cancer. J Clin Endocrinol Metab. 2001;86(4):1447–1463.

19. Nies M, Klein Hesselink MS, Huizinga GA, Sulkers E, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van

den Heuvel-Eibrink MM, Corssmit EP, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker JTM,

Rockers CM, van Santen HM, Tissing WJE, Linka TP, Bocca G. Long-term quality of life in adult survivors of pediatric

differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2016;102(4):1218–1226.

20. Howard AF, Tan de Bibiana J, Smillie K, Goddard K, Pritchard S, Olson R, Kazanjian A. Trajectories of social isolation

in adult survivors of childhood cancer. J Cancer Surviv. 2014;8(1):80–93.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

189

8

21. Reimers TS, Ehrenfels S, Mortensen EL, Schmiegelow M, Sonderkaer S, Carstensen H, Schmiegelow K, Muller J.

Cognitive deficits in long-term survivors of childhood brain tumors: Identification of predictive factors. Medi

Pediatr Oncol. 2003;40(1):26–34.

22. Wengenroth L, Rueegg CS, Michel G, Gianinazzi ME, Essig S, von der Weid NX, Grotzer M, Kuehni CE, Swiss

Paediatric Oncology Group. Concentration, working speed and memory: cognitive problems in young childhood

cancer survivors and their siblings. Pediatr Blood Cancer. 2015;62(5):875–882.

23. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management guidelines for children with thyroid nodules and differentiated thyroid cancer.

Thyroid. 2015;25(7):716–759.

24. Easley J, Miedema B, Robinson L. It’s the ‘good’ cancer, so who cares? Perceived lack of support among young

thyroid cancer survivors. Oncol Nurs Forum. 2013;40(6):596–600.

CHAPTER 8

190

8

SUPPLEMENTARY DATA

Supplemental Figure 1. Flowchart of inclusion of long-term survivors of childhood differentiated thyroid

carcinoma in the current study evaluating achievement of psychosocial developmental milestones. 1 Klein

Hesselink MS et al. J Clin Endocrinol Metab. 2016;101(5):2031-2039.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

191

8

Supplemental Table 1a. Social development in survivors of childhood DTC versus peer controls and

comparison group on item level

Item DTC

Survivors

n = 39

Peer

controls

n = 30

P Value Comparison

group

n = 508

P Value

At least one year competitive sports,

elementary school, n (%)

.4401 0.1752

Yes 36 (92) 24 (80) 427 (84)

No 3 (8) 4 (13) 80 (16)

Missing 0 (0) 2 (7) 1 (0)

Number of friends in kindergarten

through third grade, elementary

school, n (%)

.0342 0.0072

Less than 4 6 (15) 11 (37) 187 (37)

4 or more 33 (85) 18 (60) 319 (63)

Missing 0 (0) 1 (3) 2 (0)

Number of friends in fourth-sixth

grade, elementary school, n (%)

0.5342 0.2032

Less than 4 8 (21) 8 (27) 156 (31)

4 or more 30 (77) 21 (70) 349 (69)

Missing 1 (3) 1 (3) 3 (1)

Best friend, elementary school, n (%) 0.1561 0.0112

Yes 36 (92) 23 (77) 377 (74)

No 3 (8) 6 (20) 131 (26)

Missing 0 (0) 1 (3) 0 (0)

Most of time playing with …

elementary school, n (%)

0.0662 1.0001

Friends 34 (87) 20 (67) 436 (86)

Brothers and/or sisters, parents, on

your own

5 (13) 9 (30) 62 (12)

Missing 0 (0) 1 (3) 10 (2)

At least one year competitive sports,

middle and/or high school, n (%)

0.8002 0.8092

Yes 28 (72) 20 (67) 373 (73)

No 11 (28) 9 (30) 134 (26)

Missing 0 (0) 1 (3) 1 (0)

Number of friends, middle and/or high

school, n (%)

0.547² 0.9652

Less than 4 12 (31) 7 (23) 154 (30)

4 or more 27 (69) 22 (73) 352 (69)

Missing 0 (0) 1 (3) 2 (0)

Best friend, middle and/or high school,

n (%)

0.853² 0.7402

Yes 27 (69) 20 (67) 372 (73)

No 11 (28) 9 (30) 134 (26)

Missing 1 (3) 1 (3) 2 (0)

CHAPTER 8

192

8

Supplemental Table 1a. (continued)

Item DTC

Survivors

n = 39

Peer

controls

n = 30

P Value Comparison

group

n = 508

P Value

Belonging to a group of friends, middle

and/or high school, n (%)

0.5741 0.0102

Yes 37 (95) 27 (90) 403 (79)

No 1 (3) 2 (7) 97 (19)

Missing 1 (3) 1 (3) 8 (2)

Leisure time, mainly with … middle

and/or high school, n (%)

0.7471 0.6022

Friends 32 (82) 25 (83) 430 (85)

Brothers and/or sisters, parents, on

your own

7 (18) 4 (13) 75 (15)

Missing 0 (0) 1 (3) 3 (1)

Going to a bar or disco, middle and/or

high school, n (%)

0.310² 0.1662

Sometimes / often 29 (74) 25 (83) 430 (85)

Never 9 (23) 4 (13) 77 (15)

Missing 1 (3) 1 (3) 1 (0)

At least one year competitive sports,

after high school, n (%)

0.197² 0.0812

Yes 13 (33) 14 (47) 243 (48)

No 25 (64) 14 (47) 254 (50)

Missing 1 (3) 2 (7) 11 (2)

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

193

8

Supplemental Table 1b. Autonomy development in survivors of childhood DTC versus peer controls and

comparison group on item level

Item DTC

Survivors

n = 39

Peer

controls

n = 30

P Value Comparison

group

n = 508

P Value

Regular chores/tasks in your family,

elementary school, n (%)

0.1702 0.0132

Yes 10 (26) 12 (40) 233 (46)

No 29 (74) 17 (57) 273 (54)

Missing 0 (0) 1 (3) 2 (0)

Paid jobs, elementary school, n (%) 0.2782 0.3092

Yes 10 (26) 11 (37) 170 (33)

No 29 (74) 18 (60) 336 (66)

Missing 0 (0) 1 (3) 2 (0)

Regular chores/tasks in your family,

middle and/or high school, n (%)

0.0382 0.0422

Yes 17 (44) 20 (67) 304 (60)

No 22 (56) 9 (30) 201 (40)

Missing 0 (0) 1 (3) 3 (1)

Paid jobs, middle and/or high school, n (%) 0.7151 0.8051

At the age of 18 or younger 35 (90) 25 (83) 443 (87)

At the age of 19 or older / never 4 (10) 4 (13) 64 (13)

Missing 0 (0) 1 (3) 1 (0)

First time vacation without adults, n (%) 0.2132 0.0962

At the age of 17 or younger 26 (67) 15 (50) 268 (53)

At the age of 18 or older / never 13 (33) 14 (47) 239 (47)

Missing 0 (0) 1 (3) 1 (0)

Leaving parents’ home, n (%) 0.1562 0.3622

Not living with parents 28 (72) 25 (83) 328 (64)

Still living with parents 11 (28) 4 (13) 180 (35)

Missing 0 (0) 1 (3) 0 (0)

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

CHAPTER 8

194

8

Supplemental Table 1c. Psychosexual development in survivors of childhood DTC versus peer controls

and comparison group on item level

Item DTC

Survivors

n = 39

Peer

controls

n = 30

P Value Comparison

group

n = 508

P Value

First girlfriend / boyfriend, n (%) 0.9072 0.3162

At the age of 17 or younger 28 (72) 21 (70) 407 (80)

At the age of 18 or older / never 10 (26) 8 (27) 99 (19)

Missing 1 (3) 1 (3) 2 (0)

First time falling in love, n (%) 0.2241 0.3621

At the age of 18 or younger 33 (85) 28 (93) 462 (91)

At the age of 19 or older / never 5 (13) 1 (3) 42 (8)

Missing 1 (3) 1 (3) 4 (1)

First time sexual intimacy, n (%) 0.9862 0.5342

At the age of 18 or younger 31 (80) 23 (77) 421 (83)

At the age of 19 or older / never 8 (21) 6 (20) 84 (17)

Missing 0 (0) 1 (3) 4 (1)

First time sexual intercourse, n (%) 0.7462 0.1892

At the age of 18 or younger 27 (69) 19 (63) 296 (58)

At the age of 19 or older / never 12 (31) 10 (33) 210 (41)

Missing 0 (0) 1 (3) 2 (0)

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

195

8

Supplemental Table 2a. Social development in survivors of childhood DTC versus other childhood cancer

survivors (diagnosed at age ≥12 years) on item level

Item DTC Survivors

n = 35

Childhood Cancer

Survivors

n = 76

P Value

At least one year competitive sports, elementary

school, n (%)

    0.0592

Yes 32 (91) 58 (76)  

No 3 (9) 18 (24)  

Missing 0 (0) 0 (0)  

Number of friends in kindergarten through third

grade, elementary school, n (%)

    0.0052

Less than 4 3 (9) 25 (33)  

4 or more 32 (91) 49 (65)  

Missing 0 (0) 2 (3)  

Number of friends in fourth-sixth grade,

elementary school, n (%)

    0.0542

Less than 4 5 (14) 24 (32)  

4 or more 30 (86) 52 (68)  

Missing 0 (0) 0 (0)  

Best friend, elementary school, n (%)     0.0082

Yes 33 (94) 55 (72)  

No 2 (6) 21 (28)  

Missing 0 (0) 0 (0)  

Most of time playing with ….., elementary school,

n (%)

    0.3341

Friends 32 (91) 61 (80)  

Brothers and/or sisters, parents, on your own 3 (9) 11 (14)  

Missing 0 (0) 4 (5)  

At least one year competitive sports, middle and/

or high school, n (%)

    0.3452

Yes 24 (69) 45 (59)

No 11 (31) 31 (41)

Missing 0 (0) 0 (0)  

Number of friends, middle and/or high school, n (%)     0.3942

Less than 4 10 (29) 28 (37)  

4 or more 25 (71) 48 (63)  

Missing 0 (0) 0 (0)  

Best friend, middle and/or high school, n (%)     0.9862

Yes 25 (71) 56 (74)  

No 9 (26) 20 (26)  

Missing 1 (3) 0 (0)  

Belonging to a group of friends, middle and/or

high school, n (%)

    0.0112

Yes 33 (94) 59 (78)  

No 1 (3) 17 (22)  

CHAPTER 8

196

8

Supplemental Table 2a. (continued)

Item DTC Survivors

n = 35

Childhood Cancer

Survivors

n = 76

P Value

Missing 1 (3) 0 (0)

Leisure time, mainly with ….., middle and/or high

school, n (%)

    0.0262

Friends 31 (89) 51 (67)  

Brothers and/or sisters, parents, on your own 4 (11) 23 (30)  

Missing 0 (0) 2 (3)  

Going to a bar or disco, middle and/or high school,

n (%)

    0.7972

Sometimes / often 28 (80) 61 (80)  

Never 6 (17) 15 (20)  

Missing 1 (3) 0 (0)  

At least one year competitive sports, after high

school, n (%)

    0.9202

Yes 11 (31) 25 (33)  

No 23 (66) 50 (66)  

Missing 1 (3) 1 (3)  

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

197

8

Supplemental Table 2b. Autonomy development in survivors of childhood DTC versus other childhood

cancer survivors (diagnosed at age ≥12 years) on item level

Item DTC Survivors

n = 35

Childhood Cancer

Survivors

n = 76

P Value

Regular chores/tasks in your family, elementary

school, n (%)

    0.0332

Yes 8 (23) 33 (43)  

No 27 (77) 42 (55)  

Missing 0 (0) 1 (1)  

Paid jobs, elementary school, n (%)     0.4772

Yes 8 (23) 22 (29)  

No 27 (77) 53 (70)  

Missing 0 (0) 1 (1)  

Regular chores/tasks in your family, middle and/

or high school, n (%)

    0.3492

Yes 16 (46) 42 (55)

No 19 (54) 34 (45)  

Missing 0 (0) 0 (0)  

Paid jobs, middle and/or high school, n (%)     0.0972

At the age of 18 or younger 32 (91) 59 (78)  

At the age of 19 or older / never 3 (9) 17 (22)  

Missing 0 (0) 0 (0)  

First time vacation without adults, n (%)     0.1462

At the age of 17 or younger 24 (69) 41 (54)

At the age of 18 or older / never 11 (31) 35 (46)  

Missing 0 (0) 0 (0)  

Leaving parents’ home, n (%)     0.9362

Not living with parents 26 (74) 57 (75)

Still living with parents 9 (26) 19 (25)  

Missing 0 (0) 0 (0)  

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

CHAPTER 8

198

8

Supplemental Table 2c. Psychosexual development in survivors of childhood DTC versus other childhood

cancer survivors (diagnosed at age ≥12 years) on item level

Item DTC Survivors

n = 35

Childhood Cancer

Survivors

n = 76

P Value

First girlfriend / boyfriend, n (%)     0.0802

At the age of 17 or younger 26 (74) 45 (59)

At the age of 18 or older / never 8 (23) 31 (41)  

Missing 1 (3) 0 (0)  

First time falling in love, n (%)     0.6961

At the age of 18 or younger 30 (86) 68 (90)  

At the age of 19 or older / never 4 (11) 7 (9)  

Missing 1 (3) 1 (1)  

First time sexual intimacy, n (%)     0.2562

At the age of 18 or younger 30 (86) 58 (76)

At the age of 19 or older / never 5 (14) 18 (24)  

Missing 0 (0) 0 (0)  

First time sexual intercourse, n (%)     0.0102

At the age of 18 or younger 27 (77) 39 (51)

At the age of 19 or older / never 8 (23) 37 (49)  

Missing 0 (0) 0 (0)  

¹ Fisher’s Exact test ² Chi squares test. P Values in bold are P values <0.01

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

199

8

Supplemental Table 3. Characteristics of survivors of childhood DTC versus survivors of other childhood

cancers (diagnosed at all ages)

Characteristic DTC Survivors

n = 39

Childhood Cancer

Survivors

n = 350‖

P Value

At diagnosis Age at diagnosis, years 15.6 (8.7-18.7) 6.3 (0.0-17.0) 0.001§

Sex, n (%) <0.001‡

Female 34 (87) 176 (50)

Male 5 (13) 175 (50)

At follow-upAge at evaluation, years 26.2 (18.8-35.7) 24.4 (17.7-31.1) 0.021§

Follow-up period, years 10.7 (5.0-25.3) 16.9 (6.2-30.7) <0.001§

Employment, n (%) 0.338†

Employed and/or student 38 (97) 318 (91)

Not employed and no student 1 (3) 27 (8)

Missing 0 (0) 5 (1)

Completed education, n (%) <0.001‡

Low level 7 (18) 113 (32)

Medium level 15 (39) 167 (48)

High level 17 (44) 54 (15)

Missing 0 (0) 16 (5)

Marital status¶, n (%) n.a.

Relationship 24 (62) -

No relationship 15 (38) -

Marital status¶, n (%) n.a.

Not married and not living together - 234 (67)

Married or living together - 104 (30)

Missing - 12 (3)

† Fisher’s Exact Test ‡ Chi square test § Mann Whitney U test. ‖ n = 350, three childhood cancer survivors diagnosed with

thyroid cancer were removed for analyses (two had thyroid carcinoma as primary malignancy, one had thyroid cancer

as second malignant neoplasm); ¶, answer options regarding marital status of two different questionnaires were

non-mergeable, therefore these are shown separately. Missing values were excluded for statistical testing (pairwise

deletion). Continuous variables are presented as median (range). P Values in bold indicate a significant difference (P

< 0.01). Abbreviations: DTC, differentiated thyroid carcinoma; n.a., not applicable.

CHAPTER 8

200

8

Sup

ple

men

tal T

able

4. S

core

s on

psyc

hoso

cial

dev

elop

men

tal d

omai

ns o

f sur

vivo

rs o

f chi

ldho

od

DTC

ver

sus s

urvi

vors

of o

ther

chi

ldho

od

canc

ers (

diag

nose

d at

all

ages

)

Do

mai

nD

TC s

urvi

vors

n =

39

Ch

ildh

oo

d C

ance

r Su

rviv

ors

(C

CS)

Tota

l CC

S

n =

350

P V

alue

Leuk

emia

/

lym

ph

om

a

n =

175

P V

alue

Solid

tum

ors

n =

150

P V

alue

Bra

in t

umo

rs

n =

25

P V

alue

Soci

al d

evel

op

men

t †

22 (

20.3

, 23)

21 (

18, 2

2)0.

00

820

(18

, 22)

0.0

04

21 (

19, 2

3)0

.068

20 (

17, 2

2)0.

00

3A

uto

no

my

dev

elo

pm

ent

‡9

(8, 1

0)

9 (8

, 10

)0

.734

9 (8

, 10

)0

.693

9 (8

, 10

)0

.974

9 (8

, 10

)0

.278

Psyc

ho

sexu

al d

evel

op

men

t §

8 (6

.5, 8

)7

(5, 8

)0

.036

7 (5

, 8)

0.0

427

(6, 8

)0

.097

5 (4

, 8)

0.0

07

Sco

res

are

sho

wn

as m

edia

n (p

25, p

27).

Co

mpa

riso

ns b

etw

een

DTC

sur

vivo

rs a

nd o

ther

gro

ups

wer

e p

erfo

rmed

usi

ng M

ann

Whi

tney

U te

sts.

Hig

her

sco

re in

dic

ates

ach

ieve

men

t o

f

earl

ier

or

mo

re p

sych

oso

cial

dev

elo

pmen

tal m

ilest

one

s; †,

scal

e ra

nges

12-

24; ‡

, sca

le r

ange

s 6-

12; §,

sca

le r

ange

s 4-

8. M

issi

ng v

alue

s w

ere

excl

uded

for

stat

isti

cal t

esti

ng. P

val

ues

in

bo

ld in

dic

ate

a si

gnifi

cant

val

ue (

P <0

.01)

. Abb

revi

atio

n: D

TC, d

iffer

enti

ated

thy

roid

car

cino

ma;

CC

S, c

hild

hoo

d c

ance

r su

rviv

ors

.

PSYCHOSOCIAL DEVELOPMENT AFTER CHILDHOOD DIFFERENTIATED THYROID CARCINOMA

201

8

Supplemental Table 5. Medical characteristics associated with psychosocial development scales in survivors

of childhood DTC

Characteristic Social

development

P Value Autonomy

development

P Value Psychosexual

development

P Value

Survivor characteristics            

Sex, n (%) 0.790† 0.107† 0.949†

Females (n = 34) 22 9 8

Males (n = 5) 22 8 8

Age at evaluation,

years

0.946‡ 0.043‡ 0.713‡

Correlation

coefficient

-0.012 0.325 0.063

Tumor characteristics            

Age at diagnosis,

years

0.691‡ 0.816‡ 0.140‡

Correlation

coefficient

0.069 0.038 0.248

T stage, n (%) 0.676† 0.578† 0.224†

T1-2 (n = 27) 22 9 8

T3-4 (n = 10) 23 10 6

N stage, n (%) 0.539† 0.989† 0.772†

N0 (n = 16) 23 9 8

N1 (n = 23) 22 9 8

M stage, n (%) 0.292† 0.474† 0.350†

M0 (n = 34) 22 9 8

M1 (n = 5) 21 9 6

Histology, n (%) 0.450† 0.488† 0.293†

PTC (n = 35) 22 9 8

FTC (n = 4) 23 9 8

Treatment

characteristics

           

Permanent

hypoparathyroidism,

n (%)

0.428† 0.832† 0.428†

Yes (n = 10) 21 10 8

No (n = 28) 22 10 8

RLN injury, n (%) 0.955† 0.450† 0.565†

Yes (n = 4) 22 9 7

No (n = 32) 22 9 8

Cumulative 131I dose,

MBq

0.511‡ 0.114‡ 0.946‡

Correlation

coefficient

0.113 0.257 0.012

CHAPTER 8

202

8

Supplemental Table 5. (continued)

Characteristic Social

development

P Value Autonomy

development

P Value Psychosexual

development

P Value

Follow-up

characteristics

           

TSH suppression, mU/l 0.448‡ 0.858‡ 0.396‡

Correlation

coefficient

0.129 0.035 0.142

Follow-up duration,

years

0.615‡ 0.038‡ 0.700‡

Correlation

coefficient

-0.087 0.334 -0.065

Outcome, n (%) 0.043§ 0.539§ 0.789§

Remission (n = 32) 22 9 8

Recurrence (n = 3) 23 12 8

Persistent disease

(n = 4)

24   10   8  

Scores are presented as medians, except when otherwise specified. † Mann-Whitney U test ‡ Spearman’s rank

correlation coefficient § Kruskal-Wallis test. P Values in bold are P values <0.01. Abbreviations: DTC, differentiated

thyroid carcinoma; TNM stage (according to 7th ed. Of American Joint Committeea), tumor node metastases stage;

RLN, recurrent laryngeal nerve; 131I, radioactive iodine; MBq, megabecquerel; TSH, thyroid-stimulating hormone;

mU/l, milliunits per liter.

Chapter 9

Chapter 9

CHAPTER 9

Summary and general discussion

SUMMARY AND GENERAL DISCUSSION

207

9

SUMMARYClinical course and outcome of childhood differentiated thyroid cancerIn Chapter 2, we evaluated the clinical course of patients diagnosed with differentiated thyroid carcinoma (DTC) during childhood (i.e. diagnosed before the age of 19 years). Disease-specific survival of 170 patients was 100% after a median follow-up period of 13.5 years. One patient had died from a cause other than DTC. From 105 survivors, we obtained informed consent to review their medical record. After surgical treatment, almost one in three patients suffered from permanent complications such as hypoparathyroidism or recurrent laryngeal nerve injury. Patients with more advanced primary tumors, lymph node or distant metastases (DM) were treated with a higher number of radioactive iodine (131I) administrations and a higher cumulative dose of 131I. The majority of the patients remained in remission during follow-up, but 9% had persistent disease and 8% experienced disease recurrence. Having a higher tumor, node or metastases stage was associated with having persistent disease.

A minority of patients with childhood DTC develops distant metastatic disease and no large scale studies have been conducted regarding long-term outcomes of these patients. When patients with an aggressive form of DTC do not respond to standard treatment, molecular targeted therapy may be beneficial. However, further development of targeted therapy requires better definition of the mutational landscape in patients with childhood DTC. In Chapter 3 we therefore described the clinical course and mutational landscape of patients diagnosed with DM from childhood DTC in a cohort of patients treated at the MD Anderson Cancer Center in the United States. We showed that distant spread of childhood DTC is persistent in nature, but mild in its course. The median overall survival was 50.7 years, with a median disease-specific survival of 52.8 years. Patients were treated with surgery and 131I, and – in exceptional cases – with systemic therapy. Most patients with an identified oncogenic driver had a fusion involving the rearranged during transfection (RET) gene. The presence of fusions involving the neurotrophic tyrosine kinase receptor (NTRK) gene and v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations was less pronounced. No clear genotype-phenotype associations were discovered.

Late effects of the administration of radioactive iodineIn Chapter 4, we evaluated female fertility in the Dutch nationwide cohort, using a fertility-focused questionnaire and measuring serum Anti-Müllerian Hormone (AMH, a marker of ovarian reserve), luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estradiol (E2). The numbers of live births and of couples reporting problems with conceiving were comparable to the general population. AMH levels evaluated after a median follow-up period of 15.4 years did not differ significantly from the comparison group consisting of women not treated for cancer, even after adjusting for age. LH, FSH, and E2 levels were within reference ranges. Based on this

CHAPTER 9

208

9

broad evaluation of female fertility, we concluded that there is no evidence to suggest that long-term female fertility is impaired after the treatment of childhood DTC.

In the nationwide childhood DTC study, 17 males participated, four of whom agreed to participate in semen analysis. Because of this low number, and to obtain a representative number of males, we decided to evaluate male fertility in survivors of adult DTC, as shown in Chapter 5. We examined 51 males who had had a follow-up period of at least two years after their last administration of 131I. Depending on the cut-off for ‘low semen quality’, a minority of participants had low semen quality compared to a general population (1). However, reproductive characteristics and evaluation of LH, FSH, and Testosterone showed no overt disturbances.

Late effects of thyroid-stimulating hormone suppression therapy A first evaluation of cardiac function in the current cohort of survivors of childhood

DTC revealed the presence of diastolic dysfunction in one out of five survivors after a median follow-up of 17 years after diagnosis (2). When compared to age- and sex-matched controls, the median (left ventricular) diastolic function was lower in survivors. Systolic function was unaffected and no atrial fibrillation was observed. As diastolic dysfunction is associated with accelerated cardiac aging and an increased risk of cardiovascular disease, we re-evaluated 47 of the aforementioned survivors five years after their first cardiac evaluation, as described in Chapter 6. Paired analyses showed a significant decrease in diastolic function over these five years. Moreover, compared to the results of the first evaluation, the number of survivors with diastolic dysfunction had increased. When the decline in diastolic function of the current survivors was compared to the average decline based on age-categorized reference ranges, the decline among survivors was greater. Thereby, the significant increase of the N-terminal pro–B-type natriuretic peptide (NT-proBNP) between the two evaluations also indicated an increase in cardiac damage. In a multivariate linear regression analysis, body mass index and attained age were significantly associated with diastolic function, but thyroid-stimulating hormone (TSH) levels during follow-up were not.

Late effects on well-being and psychosocial developmentIn Chapter 7, survivors of childhood DTC were compared to age- and sex-matched controls regarding health-related quality of life (HRQoL) and the presence of fatigue, anxiety, and depression. Survivors’ thyroid cancer-specific complaints were also evaluated. The overall well-being of survivors did not differ from their controls, except for a slightly higher but statistically significant rate of physical problems and mental fatigue. The majority of the survivors reported none or few thyroid cancer-specific complaints. Extensive disease and treatment and unemployment were associated with worse well-being.

SUMMARY AND GENERAL DISCUSSION

209

9

To evaluate whether and to what extent the course of life was influenced by the diagnosis and treatment of childhood DTC, in Chapter 8 we evaluated the achievement of psychosocial developmental milestones of these survivors. Their psychosocial development was compared to age- and sex-matched controls and to survivors of other childhood cancers. Social, autonomy, and psychosexual development in childhood DTC survivors and controls was similar. Compared to survivors of other childhood cancers, social development in childhood DTC survivors was better. Development of autonomy and psychosexual development were similar in both groups. Overall, the diagnosis and treatment of childhood DTC did not seem to affect psychosocial development.

CHAPTER 9

210

9

DISCUSSIONThe aim of this thesis was to evaluate the clinical course and late effects of childhood DTC. The ultimate aim of the studies was to provide an outlook for newly diagnosed patients, but also to provide relevant information for patients previously treated for childhood DTC. Finally, we aimed to contribute knowledge for developing future management guidelines to optimize care for patients diagnosed with childhood DTC.

Clinical course and outcome of childhood differentiated thyroid cancerDeaths caused by childhood DTC have been consistently low, with survival rates higher than 95% after decades (3, 4). This high survival rate is confirmed in Chapters 2 and 3. Even in children presenting with advanced disease, the survival of children with DTC remains excellent (Chapter 3). This is unlike in adults, were the presence of local or distant metastases increases the risk of thyroid cancer-related deaths (5). Despite the high survival rate, long-term follow-up of childhood DTC patients is recommended because recurrence of the disease can occur decades after initial treatment (6). Follow-up is also required because thyroid hormone levels must be regularly evaluated, as life-long substitution of levothyroxine is necessary due to iatrogenic hypothyroidism after a thyroidectomy.

Because of the low mortality rate from childhood DTC, optimal treatment for these children involves balancing between reducing the chance of recurrent or persistent disease and limiting the risk of the negative consequences of treatment.

Optimization of treatment of childhood DTC starts with adequate and timely diagnosis of the disease. The Dutch pediatric guidelines indicate which diagnostic steps are appropriate at the right level of care: rare diagnoses are treated in referral centers to provide more experienced and multidisciplinary treatment (7). Initial thyroid surgery, consisting of a total thyroidectomy for the majority of children diagnosed with DTC, also has to be performed in dedicated centers, as advised by the pediatric management guidelines of the American Thyroid Association (ATA) (8).

Increasing evidence supports that a hemithyroidectomy may also be sufficient for childhood DTC patients who have a small, unifocal tumor, no extrathyroidal extension, and no lymph node metastases, or only a small number of lymph node metastases (9, 10). However, most children present with T3 or T4 tumors and locoregional lymph node metastases (3, 11) and require more aggressive surgery, as advanced disease is associated with a higher chance of recurrence or persistent disease (12, 13). Unfortunately, extensive surgery is correlated with the occurrence of more surgical complications. These occur to a lesser extent, however, when children are treated by high-volume surgeons (14-16). Minimizing surgical complications decreases long-term disease burden, such as voice problems caused by vocal cord paralysis. Other problems include parathyroid damage, which creates a need for daily medication, frequent laboratory controls, and additional clinical evaluation. Children

SUMMARY AND GENERAL DISCUSSION

211

9

with hypoparathyroidism, for instance, are at risk of nephrocalcinosis (17). To meet a higher quality standard, (surgical) treatment of children with DTC in the Netherlands has in recent years been centralized (7), which will benefit patient outcomes (18). However, to optimize treatment, not only surgery but all disciplines involved with childhood DTC management (e.g. endocrinology, nuclear medicine, pathology, and radiology) require a centralized experienced multidisciplinary management team to offer patient-tailored advice in individual situations.

Along with the disease burden caused by initial thyroid surgery, additional negative effects induced by treatment of childhood DTC may also arise. Long-term adverse consequences of cancer treatment are called late effects.

Late effects of the administration of radioactive iodineFemale fertility after radioactive iodineChapter 4 presents an elaborate assessment of reproductive characteristics and ovarian reserve of female survivors of childhood DTC; the assessment showed no long-term impairment.

Scarce evidence obtained from previous studies performed in survivors of childhood DTC presumed similar indications, but the small number of patients and lack of well-defined outcomes hindered drawing firm conclusions (19, 20).

131I administration in women treated for adult DTC caused a decrease in levels of AMH particularly pronounced in women aged 35 years and older (21-23), but after long-term follow-up normal AMH levels were found (24, 25). The clinical consequences of these studies solely reporting on AMH levels may be limited, since the assessment of AMH levels does not provide a complete evaluation of all aspects of (onco)fertility. Reviews and large-scale studies concluded that transient disturbances of fertility may occur after treatment with 131I in women treated for adult DTC, but long-term fertility is not affected because gonadal function, the ability to conceive, birthrates, and pregnancy outcomes were not permanently impaired (26-29).

Two factors may explain the normal fertility in the survivors of childhood DTC: duration of follow-up and age at treatment. First, an increased follow-up duration may allow primordial follicles to develop into secondary and early antral follicles, resulting over time in a rise of AMH levels (30). The temporary disruption of the menstrual cycle seen up to one year in women treated with 131I could reflect damage to the secondary and early antral follicles only (26). Secondly, the larger ovarian reserve in children will be relatively less impacted than the smaller number of oocytes present in adult women, manifested by a more pronounced decrease in AMH levels in women treated at ages 35 and older (21, 23, 28).

The research in Chapter 4 combined with extrapolated results from studies of adult DTC do not indicate that long-term female fertility is impaired after treatment of childhood DTC.

CHAPTER 9

212

9

Male fertility after radioactive iodineTo date, because of the small number of boys diagnosed with the disease, no studies have been performed to evaluate male fertility after childhood DTC. Thereby, earlier studies on male fertility after 131I administrations for adult DTC were limited by a short-term follow-up, a limited study size, and an incomplete evaluation of fertility parameters (31-36). Chapter 5 presents an assessment of male fertility after adult DTC after long-term follow-up. All males were treated with a relatively high cumulative dose of 131I (100 mCi or more). Semen quality, reproductive hormones, and reproductive characteristics were evaluated.

The normal long-term semen quality of the majority of the participants in Chapter 5 may be a result of the long follow-up period in the study. Radiation may initially impair semen quality, but spermatogenesis continues as long as spermatogonial stem cells are unaffected (37). In earlier studies in adult males treated with 131I for DTC, initial damage and some recovery of male fertility was seen after 131I treatment, but patients were followed up to a maximum of only 18 months after 131I administration (31-36). Unlike female fertility, male fertility is not dependent on age, and spermatogenesis takes place up to the last decades of life (38). The long-term follow-up described in Chapter 5 may have provided time for recovery of the semen quality in the majority of male DTC patients.

Next to the comparison with adult DTC survivors, an indication of the possible damage to fertility in boys treated for DTC can be derived from other childhood cancer survivors treated with radiotherapy. The radiation dose to the testes in other childhood cancer radiotherapies is up to 10 or 100 times higher than the testicular dose from 131I therapy (39-41), but even after high doses of radiotherapy, semen quality can recover (42). No clear radiation-induced threshold for permanent azoospermia is defined, but a higher dose of radiation leads to an increased recovery period. Studies in survivors of other childhood cancers showed that the effects of radiotherapy on gonadal tissue are similar in children and adults (43, 44). Compared to other (childhood) cancer patients receiving radiation to the testes, the damage to male fertility caused by 131I radiation is probably of a smaller order.

Of course, characteristics of each cancer and its treatment need to be recognized, since consequences for male fertility may differ. For instance, it is unknown whether the expression of the sodium iodine symporter (NIS) in germinal and Leydig cells facilitates the uptake of 131I in the testes (45). Additionally, treatment of DTC is accompanied by periods of hyper- and hypothyroidism which also have a transient negative effect on semen quality (46, 47).

Although future studies face logistical challenges in acquiring a representative number of participants, research regarding male fertility after childhood DTC is essential for drawing ultimate conclusions. To gather an adequate sample size, (inter)national collaboration is needed. Until then, patients should be informed about

SUMMARY AND GENERAL DISCUSSION

213

9

the low risk of poor long-term semen quality. Since we do not know which patients are at risk, fertility preservation should be discussed and offered as a precaution to all postpubertal boys and to adults receiving doses of 131I equal to or higher than 100 mCi. In prepubertal boys, performing testicular sperm extraction or testicular tissue freezing is too great an intervention for the possible minimal effect of 131I administration on fertility. When semen is cryopreserved, it is highly recommended that the patient is euthyroid upon preservation, since we found that hypothyroid patients did not preserve semen of optimal quality.

Short-term fertility after 131I administrationShort-term disruption of female and male fertility is observed after the administration of 131I. A disruption of the menstrual cycle or decrease in semen quality may transiently reduce the chance of conceiving. For females, the tendency in most studies is that 131I does not affect pregnancy outcomes or the health of children (26, 27), but a single study observed an increase in miscarriages the first year after 131I administration (48). For males, it is unknown if the increased DNA fragmentation and chromosomal abnormalities that are observed in spermatozoa up to one year after 131I administration (32, 49) have clinical consequences or impair the health of conceived children. However, no evidence exists that their children experience health problems (19, 50). Guidelines cautiously advise to avoid (conceiving) pregnancies 6 to 12 months after 131I administration (8), but to date, there is no convincing evidence to assume that 131I impairs pregnancy (outcomes). Males with a short-term desire to have children may benefit from cryopreserving their semen.

Salivary gland function after radioactive iodineSalivary gland function was also evaluated in our cross-sectional nationwide study by performing sialometry, sialochemistry, and analysis of the completed xerostomia inventory. Almost half of the survivors of childhood DTC had impaired salivary gland function, and one in three survivors reported complaints of xerostomia (51). The extent of damage to the stimulated salivary secretion and the occurrence of complaints of xerostomia increased with a higher cumulative dose of 131I. Salivary gland dysfunction (SGD) can be explained by the presence of the sodium iodine symporter in the salivary glands, which facilitates uptake of (radioactive) iodine. In adults, salivary gland function also decreases after treatment with 131I (52, 53). SGD can cause a range of difficulties such as problems with swallowing, chewing or speaking. SGD is also associated with a higher risk of dental caries, halitosis, and oral candidiasis (54). Xerostomia or SGD can be treated (symptomatically), but prevention of SGD should be the main focus. Stimulation of the salivary glands during 131I treatment, such as eating sour candy or drinking lemon juice, has been advised to prevent SGD, but no specific studies have been performed to evaluate the effectiveness of these precautions in children (8).

CHAPTER 9

214

9

Secondary primary malignancies after radioactive iodineChildren or young adults who have been treated for DTC may have a slightly higher risk of developing SPM (55, 56). One study found that survivors of childhood DTC who died predominantly died of SPM (56). 131I is also associated with a slightly higher incidence of SPM in survivors of adult DTC (57-59). The SPM found in patients treated with 131I mainly concerns rare cancers, such as salivary gland carcinomas (55, 59). It is the question if this increase represents a likely threat to DTC patients (60). Moreover, it could be that DTC patients have a genetic predisposition for the development of cancer(s). In the cohort of 105 patients described in Chapter 2, three patients were diagnosed with a SPM. However, studies to evaluate SPM require large cohorts, emphasizing the need for international collaboration.

Pulmonary fibrosis after radioactive iodinePulmonary fibrosis is a rare but serious side effect of 131I administration in patients with lung metastases (61). In Chapter 3 we described one patient who died from pulmonary fibrosis as a complication of 131I administration. The ATA pediatric guidelines advise adjusting the dose of 131I when significant (diffuse) lung uptake is present on the diagnostic whole-body scan in patients with lung metastases (8).

Bone marrow suppression after radioactive iodine131I has a suppressing effect on the bone marrow (62, 63). Moreover, the concentration of 131I in bone metastases can also cause bone marrow toxicity. However, long-term bone marrow suppression after administration of 131I is rare (8). No studies have been performed in children with DTC to evaluate long-term bone marrow suppression following 131I administration. In the Dutch cohort, no long-term bone marrow suppression was observed (unpublished results).

Considerations regarding 131I administration in childhood DTCTo improve the recurrence free survival of children with DTC, 131I was routinely administered to those having residual disease after their initial thyroid surgery (64). In line with this, 131I administration in addition to initial thyroid surgery is considered beneficial in high risk patients with (inoperable) lymph node metastases or with distant metastases (8). However, large-scale studies have shown conflicting results regarding the potential decrease in recurrences ascribed to administration of 131I (56, 65, 66). Some studies show that recurrence rates are not changed by administration of 131I, while others report that 131I does decrease the chance of recurrence.

Therefore, in children with DTC who have (inoperable) local or distant metastases, administration of 131I is more easily justified than in patients with small initial tumors, no thyroid remnant after surgery, and no metastases. For the last group of patients,

SUMMARY AND GENERAL DISCUSSION

215

9

refraining from administration of 131I can be considered (8). Further, when giving 131I, the size of the dose or the number of doses of 131I can be adjusted per patient.

SGD is a late effect of 131I administration that is the most prominent. Possible long-term damage to fertility is not an argument for administering less, or no 131I. It should be discussed whether the increased risk of late effects is acceptable when administering (more) 131I. Whenever 131I is administered, children and their caregivers must be able to make an informed decision.

Late effects of TSH suppression therapy Cardiac function after TSH suppression therapyThe decreased diastolic function initially found in childhood DTC survivors (2) further deteriorated in the five years following the initial evaluation, as shown in Chapter 6. The first evaluation did not identify arrhythmias or systolic function defects. The substantial incidence of diastolic dysfunction in this relatively young group of survivors with no pronounced cardiovascular risk factors requires follow-up, since the presence of diastolic dysfunction is associated with a higher risk of accelerated cardiac aging.

No explanation has yet been found for the cardiac damage in these survivors of childhood DTC. Diastolic dysfunction is also seen in other (childhood) cancer survivors, but the etiology in these patients lies within the cardiotoxic effects of chemotherapy or damage caused by the irradiation (67-69). The most plausible cause of the diastolic dysfunction in survivors of childhood DTC is the TSH suppression therapy, since TSH suppression therapy is strongly associated with (reversible) diastolic dysfunction in survivors of adult DTC (70-73): prolonged subclinical hyperthyroidism is associated with impaired diastolic function due to slowed myocardial relaxation (80). Survivors of adult DTC have an increased risk of cardiovascular disease and cardiovascular mortality when they are being treated with TSH suppression therapy (74). However, in contrast with results in survivors of adult DTC, in the first and second evaluations of the described survivors of childhood DTC, TSH levels were no significant predictor of diastolic function. This could be attributed to the limited availability of TSH measurements during follow-up, resulting in an incomplete representation of actual TSH levels and suppression in this retrospective study. The median of the available TSH values during the total follow-up period of 23 years was not in the suppressed range. Hypothetically, TSH suppression during the first years after initial treatment may have initiated cardiac damage. Prospective research is needed to identify if and when suppressed TSH levels initiate diastolic dysfunction in survivors of childhood DTC. Although survivors did not have a known cardiovascular disease, our findings seem clinically relevant seen the substantial impairment of diastolic function at this relatively young age. Thereby, this prospective research can also assess the possible effects on cardiac functioning of hypothyroidism induced during the treatment

CHAPTER 9

216

9

of children, as hypothyroidism also effects cardiac functioning in adults (75, 76). In adults, it is advised to limit thyroid hormone withdrawal because of its risk of cardiac impairment (75). As the effects of the administration of 131I on cardiac function have not been well-established or evaluated, they require attention in future research (77).

It is important to assess if or when diastolic dysfunction becomes clinically relevant by repeating the cardiac evaluation of these survivors. Thereby, the pathophysiological mechanism causing the diastolic dysfunction needs to be understood. Future research should assess whether treatment of childhood DTC should be adjusted. Cardiac screening seems required, with the intensity to be determined by future research.

Bone mineral density after TSH suppression therapyIn adult patients with DTC, TSH suppression therapy is associated with lower bone mineral density: exogenous subclinical hyperthyroid state induced by TSH suppression therapy is associated with increased bone resorption and low bone density (78). This effect is seen especially in postmenopausal women (79, 80). In survivors of childhood DTC, cross-sectional studies have shown no impairment of BMD (81, 82). However, due to large inter-individual differences in BMD, cross-sectional studies do not adequately assess individual changes in BMD. Currently, a longitudinal assessment of BMD is being performed in the childhood DTC cohort.

Considerations regarding TSH suppression therapy in childhood DTCBased on expert opinions, the ATA pediatric guidelines recommend aiming for a more suppressed TSH in childhood DTC patients who are considered to be at higher risk of recurrence (8) because in adult DTC series, a suppressed TSH was associated with a lower risk of recurrence (71, 83). This finding can be explained by the fact that TSH has a proliferative effect on the thyroid tissue. However, the actual data proving the added value of TSH suppression therapy in children is lacking. Therefore, it is necessary to evaluate its benefits, especially since this unsubstantiated therapy may cause adverse effects. Caregivers must be aware of the emergence of possible diastolic dysfunction in survivors of childhood DTC.

Molecular landscape of childhood differentiated thyroid carcinomaKnowledge about the molecular landscape in childhood DTC may provide additional tools to understand differences in characteristics between patients, although univocal associations between mutations and clinical characteristics were found neither in the study described in Chapter 3, nor previously (84-86). This might be due to the small sample sizes of individual studies, illustrating the need for meta-analyses or collaborations to obtain larger study sizes.

Molecular targeted therapy is successfully administered in exceptional cases in which patients with widespread, aggressive DTC do not respond to standard treatment

SUMMARY AND GENERAL DISCUSSION

217

9

(87-89). In the coming years, the role of this therapy will be further identified. It should be evaluated whether these treatments should remain applicable in patients with advanced disease, or whether these systemic therapies should be favored above standard therapy for their outcomes and late effects.

Late effects on well-being and psychosocial developmentQuality of life (QoL), levels of fatigue, and feelings of anxiety or depression are not impaired in adult survivors of childhood DTC. Most survivors will not be hindered in reaching social, autonomic, and psychosexual developmental milestones during their journey into adulthood. Thyroid cancer-specific complaints were evaluated and showed no abnormalities (Chapters 7 and 8).

Although this generally mild course of DTC provides some leeway in treatment and prognosis, it does not mean all patients will experience this in a similar way. Caregivers of DTC patients have been found to misjudge the impact of DTC on patients’ lives (90, 91). As physicians focus on the positive outlook of the disease, some patients have conflicting emotions about their own experiences, and report that they are not fully understood (90-92).

Childhood DTC may not change the achievement of developmental milestones, but the effects on these survivors may be more subtle, as we found slightly more specific complaints of physical problems and mental fatigue. These results could indicate that childhood DTC does not greatly impact well-being in the long-term, but they could also mean that we are not taking into account some specific issues that arise in this group of survivors. The members of the focus group participating in the development of the thyroid cancer-specific questionnaire were all adults upon DTC diagnosis (93). Generally, it seems that a diagnosis of DTC at an older age has a more negative effect on well-being than diagnosis at a younger age (94-96).

Thereby, although overall well-being was similar between survivors and controls, QoL was more variable in survivors, as they had a wider distribution toward worse QoL. This could indicate that a subgroup of survivors is more affected in their well-being. Future research regarding well-being in survivors of childhood DTC should address relevant topics and identify those survivors most at risk of having poor QoL. We further elaborate on this subject in the paragraph ‘future perspectives’.

Children are not small adultsDTC in children requires a different approach than DTC in adults. A younger age upon DTC diagnosis is associated with more aggressive or more advanced disease (97, 98). Thereby, the distribution of different somatic mutations varies between childhood and adult DTC (99-109). Moreover, after treatment, more recurrence or persistent disease is seen in children, but deaths are less common (5, 56, 60, 110). Late effects of treatment may be caused by similar mechanisms in children and adults,

CHAPTER 9

218

9

but consequences may differ depending on the stage of life in which the patient is treated. Some of the differences between children and adults in the presentation and clinical course might be explained by the different oncogenic drivers of adult and childhood DTC.

A gap exists in knowledge concerning the disease characteristics of patients in the transition phase from childhood to adulthood. There is no biological or psychological cut-off point after which a patient with DTC should be considered an adult. Some young adults diagnosed with DTC may have disease with the same characteristics as childhood DTC (i.e. advanced disease with a mild disease course and molecular profile in line with other childhood DTC patients), but will be treated as an adult. To prevent mismanagement, the recognition of patterns of disease can only be provided by experienced caregivers, resulting in optimal treatment

Future perspectivesIn all studies that are performed in patients with childhood DTC, achieving an adequate sample size is a challenge because of the rarity of the disease. Therefore, national and international collaborations are essential for future studies. Herein lies a challenge, because collaborations require that cultural, logistical or financial differences between physicians, hospitals or countries are overcome. Examples of (inter)national collaborations are the European Reference Network on Rare Endocrine Conditions, European Thyroid Association Taskforce for Children with Thyroid Nodules and Thyroid Cancer, and the North American Child and Adolescent Thyroid Consortium.

Regarding late effects after childhood DTC, the increase in diastolic dysfunction in this relatively young group of survivors requires attention. Longitudinal research is needed to evaluate the clinical significance of this finding, but should also focus on preventive and therapeutic interventions. It is also important to evaluate possibilities for preventing and managing SGD in these survivors.

With regard to well-being, patient meetings may serve multiple purposes. First, gatherings can function as a place where patients are informed about various aspects of their rare disease. Secondly, they can (anonymously) address their thoughts regarding subjects for future research on late effects. Third, patient information meetings could also offer an opportunity for patients to meet other patients with this rare disease. Lastly, provision of patient information regarding current knowledge is crucial to ensure that patients in general understand the importance of participating in studies. Of course, structured focus groups to assess problems arising in patients and survivors of childhood DTC can also be instituted.

Other future research should focus on optimizing treatment outcomes with minimal late effects. The role of 131I administration and TSH suppression therapy in childhood patients of all risk factor categories should be re-established, by comparing long-term outcomes between treatments. Prospective studies are most desirable

SUMMARY AND GENERAL DISCUSSION

219

9

for assessing these endpoints. Shared (inter)national databases with well-defined definitions can overcome the sample size issues. Databases can serve prospective and retrospective research, where retrospective research could reveal risk factors for late effects of childhood DTC treatment. By adding data from (young) adult DTC patients to the databases, more insight can be gained into the structural differences in disease characteristics and outcomes between age groups.

Survivors of childhood DTC may benefit from care for late effects in specialized late effects outpatient clinic, as developed for other childhood cancer survivors in the Netherlands and many other countries.

CONCLUSIONAs most children with DTC will survive their disease, it is important to keep late effects in mind during treatment or follow-up. A multidisciplinary team should balance the disease outcome and the burden of the disease and its treatment, requiring individual decisions for every patient. Regarding late effects of treatment, our research showed that survivors of childhood DTC are at risk for diastolic dysfunction, requiring follow-up to assess its consequences. Although subtle or transient changes may occur, no overt permanent damage of male or female fertility was observed. In general, QoL and psychosocial development do not seem to be harmed by childhood DTC or its treatment.

CHAPTER 9

220

9

REFERENCES1. Cooper TG, Noonan E, von Eckardstein S, Auger J, Baker HW, Behre HM, Haugen TB, Kruger T, Wang C, Mbizvo

MT, Vogelsong KM. World health organization reference values for human semen characteristics. Hum Reprod

Update. 2010;16(3):231-245.

2. Klein Hesselink MS, Bocca G, Hummel YM, Brouwers AH, Burgerhof JGM, van Dam EWCM, Gietema JA, Havekes

B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker

JTM, Ronckers CM, van Santen HM, van der Meer P, Links TP, Tissing WJE. Diastolic dysfunction is common in

survivors of pediatric differentiated thyroid carcinoma. Thyroid. 2017;27(12):1481-1489.

3. Hay ID, Johnson TR, Kaggal S, Reinalda MS, Iniguez-Ariza NM, Grant CS, Pittock ST, Thompson GB. Papillary

thyroid carcinoma (PTC) in children and adults: comparison of initial presentation and long-term postoperative

outcome in 4432 patients consecutively treated at the Mayo Clinic during eight decades (1936-2015). World J

Surg. 2018;42(2):329-342.

4. Kowalski LP, Filho JG, Pinto CAL, Carvalho AL, de Camargo B. Long-term survival rates in young patients with

thyroid carcinoma. Arch Otolaryngol Head Neck Surg. 2003;129(7):746-749.

5. Eustatia-Rutten CFA, Corssmit EPM, Biermasz NR, Pereira AM, Romijn JA, Smit JW. Survival and death causes in

differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2006;91(1):313-319.

6. Welch Dinauer CA, Tuttle RM, Robie DK, McClellan DR, Svec RL, Adair C, Francis GL. Clinical features associated

with metastasis and recurrence of differentiated thyroid cancer in children, adolescents and young adults. Clin

Endocrinol (Oxf). 1998;49(5):619-628.

7. Lebbink CA, Dekker BL, Bocca G, Braat AJ, Derikx JP, Dierselhuis MP, de Keizer B, Kruijff S, Kwast AB, van

Nederveen FH, Nieveen van Dijkum E, Nievelstein RA, Peeters RP, Terwisscha van Scheltinga CE, Tissing WJ, van

der Tuin K, Vriens MR, Zsiros J, van Trotsenburg AS, Links TP, van Santen HM. New national recommendations for

the treatment of pediatric differentiated thyroid carcinoma in the Netherlands. Eur J Endocrinol. 2020;183(4):11-18.

8. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management guidelines for children with thyroid nodules and differentiated thyroid cancer.

Thyroid. 2015;25(7):716-759.

9. Sugino K, Nagahama M, Kitagawa W, Ohkuwa K, Uruno T, Matsuzu K, Suzuki A, Tomoda C, Hames KY, Akaishi J,

Masaki C, Ito K. Risk stratification of pediatric patients with differentiated thyroid cancer: Is total thyroidectomy

necessary for patients at any risk? Thyroid. 2020;30(4):548-556.

10. Spinelli C, Rossi L, Piscioneri J, Strambi S, Antonelli A, Ferrari A, Massimino M, Miccoli P. Pediatric differentiated

thyroid cancer: When to perform conservative and radical surgery. Curr Pediatr Rev. 2016;12(4):247-252.

11. Park S, Jeong JS, Ryu HR, Lee C-R, Park JH, Kang S-W, Jeong JJ, Nam K-H, Chung WY, Park CS. Differentiated

thyroid carcinoma of children and adolescents: 27-year experience in the Yonsei university health system. J

Korean Med Sci. 2013;28(5):693-699.

12. Medas F, Canu GL, Boi F, Lai ML, Erdas E, Calò PG. Predictive factors of recurrence in patients with differentiated

thyroid carcinoma: a retrospective analysis on 579 patients. Cancers (Basel). 2019;11(9):1230.

13. Sung TY, Jeon MJ, Lee YH, Lee Y-m, Kwon H, Yoon JH, Chung K-W, Kim WG, Song DE, Hong SJ. Initial and dynamic risk

stratification of pediatric patients with differentiated thyroid cancer. J Clin Endocrinol Metab. 2016;102(3):793-800.

14. Sosa JA, Tuggle CT, Wang TS, Thomas DC, Boudourakis L, Rivkees S, Roman SA. Clinical and economic outcomes

of thyroid and parathyroid surgery in children. J Clin Endocrinol Metab. 2008;93(8):3058-3065.

15. Baumgarten HD, Bauer AJ, Isaza A, Mostoufi-Moab S, Kazahaya K, Adzick NS. Surgical management of pediatric

thyroid disease: Complication rates after thyroidectomy at the children’s hospital of philadelphia high-volume

pediatric thyroid center. J Pediatr Surg. 2019;54(10):1969-1975.

16. Tuggle CT, Roman SA, Wang TS, Boudourakis L, Thomas DC, Udelsman R, Ann Sosa J. Pediatric endocrine surgery:

Who is operating on our children? Surgery. 2008;144(6):869-877.

17. Levy I, Licht C, Daneman A, Sochett E, Harrington J. The impact of hypoparathyroidism treatment on the kidney in

children: long-term retrospective follow-up study. J Clin Endocrinol Metab. 2015;100(11):4106-4113.

18. Youngwirth LM, Adam MA, Thomas SM, Roman SA, Sosa JA, Scheri RP. Pediatric thyroid cancer patients referred

to high-volume facilities have improved short-term outcomes. Surgery. 2018;163(2):361-366.

SUMMARY AND GENERAL DISCUSSION

221

9

19. Sarkar SD, Beierwaltes WH, Gill SP, Cowley BJ. Subsequent fertility and birth histories of children and adolescents

treated with 131i for thyroid cancer. J Nucl Med. 1976;17(6):460-464.

20. Albano D, Bertagna F, Panarotto MB, Giubbini R. Early and late adverse effects of radioiodine for pediatric

differentiated thyroid cancer. Pediatr Blood Cancer. 2017;64(11):e26595.

21. van Velsen EFS, Visser WE, van den Berg SAA, Kam BLR, van Ginhoven TM, Massolt ET, Peeters RP. Longitudinal

analysis of the effect of radioiodine therapy on ovarian reserve in females with differentiated thyroid cancer.

Thyroid. 2020;30(4):580-587.

22. Evranos B, Faki S, Polat SB, Bestepe N, Ersoy R, Cakir B. Effects of radioactive iodine therapy on ovarian reserve:

A prospective pilot study. Thyroid. 2018;28(12):1702-1707.

23. Yaish I, Azem F, Gutfeld O, Silman Z, Serebro M, Sharon O, Shefer G, Limor R, Stern N, Tordjman KM. A single

radioactive iodine treatment has a deleterious effect on ovarian reserve in women with thyroid cancer: results of

a prospective pilot study. Thyroid. 2018;28(4):522-527.

24. Giusti M, Mittica M, Comite P, Campana C, Gay S, Mussap M. Anti-müllerian hormone in pre-menopausal females

after ablative radioiodine treatment for differentiated thyroid cancer. Endocrine. 2018;60(3):516-523.

25. Mittica M, Dotto A, Comina M, Teliti M, Monti E, Giusti M. Cross-sectional and prospective study on anti-Müllerian

hormone changes in a cohort of pre-menopausal women with a history of differentiated thyroid cancer. Thyroid

Res. 2020;13:1.

26. Sawka AM, Lakra DC, Lea J, Alshehri B, Tsang RW, Brierley JD, Straus S, Thabane L, Gafni A, Ezzat S, George SR,

Goldstein DP. A systematic review examining the effects of therapeutic radioactive iodine on ovarian function and

future pregnancy in female thyroid cancer survivors. Clin Endocrinol (Oxf). 2008;69(3):479-490.

27. Sioka C, Fotopoulos A. Effects of I-131 therapy on gonads and pregnancy outcome in patients with thyroid cancer.

Fertil Steril. 2011;95(5):1552-1559.

28. Wu JX, Young S, Ro K, Li N, Leung AM, Chiu HK, Harari A, Yeh MW. Reproductive outcomes and nononcologic

complications after radioactive iodine ablation for well-differentiated thyroid cancer. Thyroid. 2015;25(1):133-138.

29. Anderson C, Engel SM, Weaver MA, Zevallos JP, Nichols HB. Birth rates after radioactive iodine treatment for

differentiated thyroid cancer. Int J Cancer. 2017;141(11):2291-2295.

30. Andersen CY, Schmidt KT, Kristensen SG, Rosendahl M, Byskov AG, Ernst E. Concentrations of AMH and Inhibin-B

in relation to follicular diameter in normal human small antral follicles. Hum Reprod. 2010;25(5):1282-1287.

31. Wichers M, Benz E, Palmedo H, Biersack HJ, Grunwald F, Klingmuller D. Testicular function after radioiodine

therapy for thyroid carcinoma. Eur J Nucl Med. 2000;27(5):503-507.

32. Bourcigaux N, Rubino C, Berthaud I, Toubert ME, Donadille B, Leenhardt L, Petrot-Keller I, Brailly-Tabard S,

Fromigue J, de Vathaire F, Simon T, Siffroi JP, Schlumberger M, Bouchard P, Christin-Maitre S. Impact on testicular

function of a single ablative activity of 3.7 GBq radioactive iodine for differentiated thyroid carcinoma. Hum

Reprod. 2018;33(8):1408-1416.

33. Rosario PW, Barroso AL, Rezende LL, Padrao EL, Borges MA, Guimaraes VC, Purisch S. Testicular function after

radioiodine therapy in patients with thyroid cancer. Thyroid. 2006;16(7):667-670.

34. Pacini F, Gasperi M, Fugazzola L, Ceccarelli C, Lippi F, Centoni R, Martino E, Pinchera A. Testicular function in

patients with differentiated thyroid carcinoma treated with radioiodine. J Nucl Med. 1994;35(9):1418-1422.

35. Esfahani AF, Eftekhari M, Zenooz N, Saghari M. Gonadal function in patients with differentiated thyroid cancer

treated with (131)I. Hell J Nucl Med. 2004;7(1):52-55.

36. Canale D, Ceccarelli C, Caglieresi C, Moscatelli A, Gavioli S, Santini P, Elisei R, Vitti P. Effects of radioiodine

treatment for differentiated thyroid cancer on testis function. Clin Endocrinol (Oxf). 2015;82(2):295-299.

37. De Felice F, Marchetti C, Marampon F, Cascialli G, Muzii L, Tombolini V. Radiation effects on male fertility.

Andrology. 2019;7(1):2-7.

38. Eskenazi B, Wyrobek AJ, Sloter E, Kidd SA, Moore L, Young S, Moore D. The association of age and semen quality

in healthy men. Hum Reprod. 2003;18(2):447-454.

39. Garsi JP, Schlumberger M, Ricard M, Labbe M, Ceccarelli C, Schvartz C, Henry-Amar M, Bardet S, Rubino C, De

Vathaire F. Health outcomes of children fathered by patients treated with radioiodine for thyroid cancer. Clin

Endocrinol (Oxf). 2009;71(6):880-883.

40. Thomson AB, Campbell AJ, Irvine DS, Anderson RA, Kelnar CJ, Wallace WHB. Semen quality and spermatozoal DNA

integrity in survivors of childhood cancer: a case-control study. The Lancet. 2002;360(9330):361-367.

CHAPTER 9

222

9

41. Ceccarelli C, Battisti P, Gasperi M, Fantuzzi E, Pacini F, Gualdrini G, Pierantoni MC, Luciani A, Djokich D, Pinchera

A. Radiation dose to the testes after 131I therapy for ablation of postsurgical thyroid remnants in patients with

differentiated thyroid cancer. J Nucl Med. 1999;40(10):1716-1721.

42. Okada K, Fujisawa M. Recovery of spermatogenesis following cancer treatment with cytotoxic chemotherapy and

radiotherapy. World J Mens Health. 2019;37(2):166-174.

43. Sy Ortin TT, Shostak CA, Donaldson SS. Gonadal status and reproductive function following treatment for

hodgkin’s disease in childhood: The stanford experience. Int J Radiat Oncol Biol Phys. 1990;19(4):873-880.

44. Meistrich ML. Male gonadal toxicity. Pediatr Blood Cancer. 2009;53(2):261-266.

45. Russo D, Scipioni A, Durante C, Ferretti E, Gandini L, Maggisano V, Paoli D, Verrienti A, Costante G, Lenzi A, Filetti S.

Expression and localization of the sodium/iodide symporter (NIS) in testicular cells. Endocrine. 2011;40(1):35-40.

46. Krassas GE, Papadopoulou F, Tziomalos K, Zeginiadou T, Pontikides N. Hypothyroidism has an adverse effect on

human spermatogenesis: a prospective, controlled study. Thyroid. 2008;18(12):1255-1259.

47. Krassas GE, Pontikides N, Deligianni V, Miras K. A prospective controlled study of the impact of hyperthyroidism

on reproductive function in males. J Clin Endocrinol Metab. 2002;87(8):3667-3671.

48. Schlumberger M, De Vathaire F, Ceccarelli C, Delisle MJ, Francese C, Couette JE, Pinchera A, Parmentier C.

Exposure to radioactive Iodine-131 for scintigraphy or therapy does not preclude pregnancy in thyroid cancer

patients. J Nucl Med. 1996;37(4):606-612.

49. Esquerre-Lamare C, Isus F, Moinard N, Bujan L. Sperm DNA fragmentation after radioiodine treatment for

differentiated thyroid cancer. Basic Clin Androl. 2015;25:1.

50. Hyer S, Vini L, O’Connell M, Pratt B, Harmer C. Testicular dose and fertility in men following i(131) therapy for

thyroid cancer. Clin Endocrinol (Oxf). 2002;56(6):755-758.

51. Selvakumar T, Nies M, Klein Hesselink MS, Brouwers AH, van der Horst-Schrivers ANA, Klein Hesselink EN, Tissing

WJE, Vissink A, Links TP, Dutch Pediatric Thyroid Cancer study consortium. Long-term effects of radioiodine

treatment on salivary gland function in adult survivors of pediatric differentiated thyroid carcinoma. J Nucl

Med. 2018;60(2):172-177.

52. Klein Hesselink EN, Brouwers AH, de Jong JR, van der Horst-Schrivers AN, Coppes RP, Lefrandt JD, Jager PL,

Vissink A, Links TP. Effects of radioiodine treatment on salivary gland function in patients with differentiated

thyroid carcinoma: a prospective study. J Nucl Med. 2016;57(11):1685-1691.

53. Jeong SY, Kim HW, Lee SW, Ahn BC, Lee J. Salivary gland function 5 years after radioactive iodine ablation in

patients with differentiated thyroid cancer: Direct comparison of pre- and postablation scintigraphies and their

relation to xerostomia symptoms. Thyroid. 2013;23(5):609-616.

54. Millsop JW, Wang EA, Fazel N. Etiology, evaluation, and management of xerostomia. Clin Dermatol. 2017;35(5):468-476.

55. Marti JL, Jain KS, Morris LGT. Increased risk of second primary malignancy in pediatric and young adult patients

treated with radioactive iodine for differentiated thyroid cancer. Thyroid. 2015;25(6):681-687.

56. Hay ID, Gonzalez-Losada T, Reinalda MS, Honetschlager JA, Richards ML, Thompson GB. Long-term outcome

in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J

Surg. 2010;34(6):1192-1202.

57. Sawka AM, Thabane L, Parlea L, Ibrahim-Zada I, Tsang RW, Brierley JD, Straus S, Ezzat S, Goldstein DP. Second

primary malignancy risk after radioactive iodine treatment for thyroid cancer: A systematic review and meta-

analysis. Thyroid. 2009;19(5):451-457.

58. Molenaar RJ, Sidana S, Radivoyevitch T, Advani AS, Gerds AT, Carraway HE, Angelini D, Kalaycio M, Nazha A,

Adelstein DJ, Nasr C, Maciejewski JP, Majhail NS, Sekeres MA, Mukherjee S. Risk of hematologic malignancies after

radioiodine treatment of well-differentiated thyroid cancer. J Clin Oncol. 2018;36(18):1831-1839.

59. Rubino C, de Vathaire F, Dottorini ME, Hall P, Schvartz C, Couette JE, Dondon MG, Abbas MT, Langlois C,

Schlumberger M. Second primary malignancies in thyroid cancer patients. Br J Cancer. 2003;89(9):1638-1644.

60. Links TP, Tol KMv, Jager PL, Plukker JTM, Piers DA, Boezen HM, Dullaart RPF, Vries EGEd, Sluiter WJ. Life expectancy

in differentiated thyroid cancer: a novel approach to survival analysis. Endocr Relat Cancer. 2005;12(2):273.

61. Chen L, Shen Y, Luo Q, Yu Y, Lu H, Zhu R. Pulmonary fibrosis following radioiodine therapy of pulmonary metastases

from differentiated thyroid carcinoma. Thyroid. 2010;20(3):337-340.

62. Prinsen HT, Klein Hesselink EN, Brouwers AH, Plukker JT, Sluiter WJ, van der Horst-Schrivers AN, van Imhoff

GW, Links TP. Bone marrow function after (131)I therapy in patients with differentiated thyroid carcinoma. J Clin

Endocrinol Metab. 2015;100(10):3911-3917.

SUMMARY AND GENERAL DISCUSSION

223

9

63. Dong P, Li L, Huang R, Qu Y. Acute and long-term bone marrow suppression of radioiodine therapy for pediatric

patients with differentiated thyroid carcinoma. J Nucl Med. 2018;59(supplement 1):311.

64. Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Mang O, Lau WH. Differentiated thyroid carcinoma in childhood

and adolescence-clinical course and role of radioiodine. Pediatr Blood Cancer. 2004;42(2):176-183.

65. Jarzab B, Handkiewicz-Junak D, Wloch J. Juvenile differentiated thyroid carcinoma and the role of radioiodine in

its treatment: A qualitative review. Endocr Relat Cancer. 2005;12(4):773-803.

66. Newman KD, Black T, Heller G, Azizkhan RG, Holcomb GW, 3rd, Sklar C, Vlamis V, Haase GM, La Quaglia MP.

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. Ann Surg. 1998;227(4):533-541.

67. Heidenreich PA, Hancock SL, Vagelos RH, Lee BK, Schnittger I. Diastolic dysfunction after mediastinal irradiation.

Am Heart J. 2005;150(5):977-982.

68. Brouwer CA, Postma A, Vonk JM, Zwart N, van den Berg MP, Bink-Boelkens MTE, Dolsma WV, Smit AJ, de

Vries EG, Tissing W. Systolic and diastolic dysfunction in long-term adult survivors of childhood cancer. Eur J

Cancer. 2011;47(16):2453-2462.

69. Calabrese V, Menna P, Annibali O, Armento G, Carpino A, Cerchiara E, Greco C, Marchesi F, Spallarossa P, Toglia

G, Reggiardo G, Minotti G. Early diastolic dysfunction after cancer chemotherapy: Primary endpoint results of

a multicenter cardio-oncology study. Chemotherapy. 2018;63(2):55-63.

70. Biondi B, Fazio S, Palmieri EA, Carella C, Panza N, Cittadini A, Bonè F, Lombardi G, Saccà L. Left ventricular diastolic

dysfunction in patients with subclinical hypothyroidism. J Clin Endocrinol Metab. 1999;84(6):2064-2067.

71. Biondi B, Cooper DS. Benefits of thyrotropin suppression versus the risks of adverse effects in differentiated

thyroid cancer. Thyroid. 2010;20(2):135-146.

72. Biondi B, Fazio S, Carella C, Amato G, Cittadini A, Lupoli G, Saccà L, Bellastella A, Lombardi G. Cardiac effects of

long term thyrotropin-suppressive therapy with levothyroxine. J Clin Endocrinol Metab. 1993;77(2):334-338.

73. Smit JW, Eustatia-Rutten CF, Corssmit EP, Pereira AM, Frölich M, Bleeker GB, Holman ER, van der Wall EE, Romijn

JA, Bax JJ. Reversible diastolic dysfunction after long-term exogenous subclinical hyperthyroidism: A randomized,

placebo-controlled study. J Clin Endocrinol Metab. 2005;90(11):6041-6047.

74. Klein Hesselink EN, Klein Hesselink MS, de Bock GH, Gansevoort RT, Bakker SJ, Vredeveld EJ, van der Horst-Schrivers

AN, van der Horst IC, Kamphuisen PW, Plukker JT, Links TP, Lefrandt JD. Long-term cardiovascular mortality in

patients with differentiated thyroid carcinoma: An observational study. J Clin Oncol. 2013;31(32):4046-4053.

75. Botella-Carretero JI, Gómez-Bueno M, Barrios V, Caballero C, García-Robles R, Sancho J, Escobar-Morreale

HcF. Chronic thyrotropin-suppressive therapy with levothyroxine and short-term overt hypothyroidism after

thyroxine withdrawal are associated with undesirable cardiovascular effects in patients with differentiated thyroid

carcinoma. 2004;11(2):345.

76. Hoftijzer HC, Bax JJ, Heemstra KA, Bleeker GB, Delgado V, Van der Klaauw AA, Romijn JA, Smit J, Corssmit E. Short‖

term overt hypothyroidism induces discrete diastolic dysfunction in patients treated for differentiated thyroid

carcinoma. Eur J Clin Invest. 2009;39(3):204-210.

77. Ryödi E, Metso S, Huhtala H, Välimäki M, Auvinen A, Jaatinen P. Cardiovascular morbidity and mortality after

treatment of hyperthyroidism with either radioactive iodine or thyroidectomy. Thyroid. 2018;28(9):1111-1120.

78. Dhanwal DK. Thyroid disorders and bone mineral metabolism. Indian J Endocrinol Metab. 2011;15(Suppl 2):S107-S112.

79. Wang LY, Smith AW, Palmer FL, Tuttle RM, Mahrous A, Nixon IJ, Patel SG, Ganly I, Fagin JA, Boucai L. Thyrotropin

suppression increases the risk of osteoporosis without decreasing recurrence in ata low- and intermediate-risk

patients with differentiated thyroid carcinoma. Thyroid. 2015;25(3):300-307.

80. Sugitani I, Fujimoto Y. Effect of postoperative thyrotropin suppressive therapy on bone mineral density in patients

with papillary thyroid carcinoma: a prospective controlled study. Surgery. 2011;150(6):1250-1257.

81. Leonova TA, Drozd VM, Saenko VA, Mine M, Biko J, Rogounovitch TI, Takamura N, Reiners C, Yamashita S. Bone

mineral density in treated at a young age for differentiated thyroid cancer after Chernobyl female patients on

TSH-suppressive therapy receiving or not Calcium-D3 supplementation. Endocr J. 2015;62(2):173-182.

82. Mendonca Monteiro de Barros G, Madeira M, Vieira Neto L, de Paula Paranhos Neto F, Carvalho Mendonca

LM, Correa Barbosa Lima I, Corbo R, Fleiuss Farias ML. Bone mineral density and bone microarchitecture after

long-term suppressive levothyroxine treatment of differentiated thyroid carcinoma in young adult patients. J

Bone Miner Metab. 2016;34(4):417-421.

CHAPTER 9

224

9

83. McGriff NJ, Csako G, Gourgiotis L, Lori CG, Pucino F, Sarlis NJ. Effects of thyroid hormone suppression therapy on

adverse clinical outcomes in thyroid cancer. Ann Med. 2002;34(7-8):554-564.

84. Nikita ME, Jiang W, Cheng SM, Hantash FM, McPhaul MJ, Newbury RO, Phillips SA, Reitz RE, Waldman FM, Newfield

RS. Mutational analysis in pediatric thyroid cancer and correlations with age, ethnicity, and clinical presentation.

Thyroid. 2016;26(2):227-234.

85. Alzahrani AS, Alswailem M, Moria Y, Almutairi R, Alotaibi M, Murugan AK, Qasem E, Alghamdi B, Al-Hindi H.

Lung metastasis in pediatric thyroid cancer: Radiological pattern, molecular genetics, response to therapy, and

outcome. J Clin Endocrinol Metab. 2018;104(1):103-110.

86. Cordioli MI, Moraes L, Cury AN, Cerutti JM. Are we really at the dawn of understanding sporadic pediatric thyroid

carcinoma? Endocr Relat Cancer. 2015;22(6):R311-324.

87. Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, Nathenson M, Doebele RC, Farago AF, Pappo

AS, Turpin B, Dowlati A, Brose MS, Mascarenhas L, Federman N, Berlin J, El-Deiry WS, Baik C, Deeken J, Boni V,

Nagasubramanian R, Taylor M, Rudzinski ER, Meric-Bernstam F, Sohal DPS, Ma PC, Raez LE, Hechtman JF, Benayed

R, Ladanyi M, Tuch BB, Ebata K, Cruickshank S, Ku NC, Cox MC, Hawkins DS, Hong DS, Hyman DM. Efficacy of

larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731-739.

88. Mahajan P, Dawrant J, Kheradpour A, Quintanilla NM, Lopez ME, Orth RC, Athanassaki I, Venkatramani R. Response

to lenvatinib in children with papillary thyroid carcinoma. Thyroid. 2018;28(11):1450-1454.

89. Waguespack SG, Sherman SI, Williams MD, Clayman GL, Herzog CE. The successful use of sorafenib to treat

pediatric papillary thyroid carcinoma. Thyroid. 2009;19(4):407-412.

90. Easley J, Miedema B, Robinson L. It’s the “good” cancer, so who cares? Perceived lack of support among young

thyroid cancer survivors. Oncol Nurs Forum. 2013;40(6):596-600.

91. James BC, Aschebrook-Kilfoy B, White MG, Applewhite MK, Kaplan SP, Angelos P, Kaplan EL, Grogan RH. Quality of

life in thyroid cancer-assessment of physician perceptions. J Surg Res. 2018;226:94-99.

92. Randle RW, Bushman NM, Orne J, Balentine CJ, Wendt E, Saucke M, Pitt SC, Macdonald CL, Connor NP, Sippel RS.

Papillary thyroid cancer: The good and bad of the “good cancer”. Thyroid. 2017;27(7):902-907.

93. Husson O, Haak HR, Mols F, Nieuwenhuijzen GA, Nieuwlaat WA, Reemst PH, Huysmans DA, Toorians AW, van de

Poll-Franse LV. Development of a disease-specific health-related quality of life questionnaire (THYCA-QOL) for

thyroid cancer survivors. Acta Oncologica (Stockholm, Sweden). 2013;52(2):447-454.

94. Goldfarb M, Casillas J. Thyroid cancer specific quality of life and health related quality of life in young adult thyroid

cancer survivors. Thyroid. 2016;26(7):923-932.

95. Oren A, Benoit MA, Murphy A, Schulte F, Hamilton J. Quality of life and anxiety in adolescents with differentiated

thyroid cancer. J Clin Endocrinol Metab. 2012;97(10):1933.

96. Hoftijzer HC, Heemstra KA, Corssmit EP, van der Klaauw AA, Romijn JA, Smit JW. Quality of life in cured patients

with differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2008;93(1):200-203.

97. Jarzab B, Handkiewicz-Junak D. Differentiated thyroid cancer in children and adults: Same or distinct disease?

Hormones (Athens, Greece). 2007;6(3):200-209.

98. Lazar L, Lebenthal Y, Steinmetz A, Yackobovitch-Gavan M, Phillip M. Differentiated thyroid carcinoma in pediatric patients:

Comparison of presentation and course between pre-pubertal children and adolescents. J Pediatr. 2009;154(5):708-714.

99. Yamashita S, Saenko V. Mechanisms of disease: Molecular genetics of childhood thyroid cancers. Nat Clin Pract

Endocrinol Metab. 2007;3(5):422-429.

100. Fenton C, Anderson J, Lukes Y, Dinauer CA, Tuttle RM, Francis GL. Ras mutations are uncommon in sporadic

thyroid cancer in children and young adults. J Endocrinol Invest. 1999;22(10):781-789.

101. Fenton CL, Lukes Y, Nicholson D, Dinauer CA, Francis GL, Tuttle RM. The ret/ptc mutations are common in

sporadic papillary thyroid carcinoma of children and young adults. J Clin Endocrinol Metab. 2000;85(3):1170-1175.

102. Cordioli MI, Moraes L, Bastos AU, Besson P, Alves MT, Delcelo R, Monte O, Longui C, Cury AN, Cerutti JM.

Fusion oncogenes are the main genetic events found in sporadic papillary thyroid carcinomas from children.

Thyroid. 2017;27(2):182-188.

103. Diesen DL, Skinner MA. Pediatric thyroid cancer. Semin Pediatr Surg. 2012;21(1):44-50.

104. Sisdelli L, Cordioli M, Vaisman F, Moraes L, Colozza-Gama GA, Alves PAG, Jr., Araujo ML, Jr., Alves MTS, Monte O,

Longui CA, Cury AN, Carvalheira G, Cerutti JM. AGK-BRAF is associated with distant metastasis and younger age in

pediatric papillary thyroid carcinoma. Pediatr Blood Cancer. 2019;66(7):e27707.

SUMMARY AND GENERAL DISCUSSION

225

9

105. Galuppini F, Vianello F, Censi S, Barollo S, Bertazza L, Carducci S, Colato C, Manso J, Rugge M, Iacobone M,

Watutantrige Fernando S, Pennelli G, Mian C. Differentiated thyroid carcinoma in pediatric age: genetic and

clinical scenario. Front Endocrinol (Lausanne). 2019;10:552.

106. Faggiano A, Coulot J, Bellon N, Talbot M, Caillou B, Ricard M, Bidart JM, Schlumberger M. Age-dependent variation

of follicular size and expression of iodine transporters in human thyroid tissue. J Nucl Med. 2004;45(2):232-237.

107. Pekova B, Dvorakova S, Sykorova V, Vacinova G, Vaclavikova E, Moravcova J, Katra R, Vlcek P, Sykorova P, Kodetova

D, Vcelak J, Bendlova B. Somatic genetic alterations in a large cohort of pediatric thyroid nodules. Endocr

Connect. 2019;8(6):796-805.

108. Alzahrani AS, Murugan AK, Qasem E, Alswailem M, Al-Hindi H, Shi Y. Single point mutations in pediatric

differentiated thyroid cancer. Thyroid. 2017;27(2):189-196.

109. Mostoufi-Moab S, Labourier E, Sullivan L, LiVolsi V, Li Y, Xiao R, Beaudenon-Huibregtse S, Kazahaya K,

Adzick NS, Baloch Z, Bauer AJ. Molecular testing for oncogenic gene alterations in pediatric thyroid lesions.

Thyroid. 2018;28(1):60-67.

110. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA, Udelsman R. The treatment of

differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocr

Rev. 2011;32(6):798-826.

CHAPTER 10

Appendices

APPENDICES

229

10

NEDERLANDSE SAMENVATTINGGedifferentieerd schildkliercarcinoom bij kinderenSchildkliercarcinoom bij kinderen is een zeldzame aandoening, waarbij meer dan 90% van de patiënten een papillaire of folliculaire variant van deze ziekte heeft (1). Deze twee varianten van schildkliercarcinoom worden gedifferentieerd schildkliercarcinoom (DTC) genoemd. De ziekte komt overwegend voor bij het vrouwelijke geslacht. De incidentie van DTC op de kinderleeftijd neemt toe, waarbij een deel van deze stijging kan worden verklaard door een stijging in het aantal toevalsbevindingen (2, 3).

De meeste kinderen met DTC presenteren zich met een asymptomatische schildkliernodus of met een solitaire massa in de nek (4). In uitzonderlijke gevallen ontstaat er dyspneu, stemproblematiek of een verstikkingsgevoel door de tumor. Het weefsel in de nodus of massa kan worden onderzocht door middel van een dunne-naald biopt. De kans dat het weefsel van maligne origine is, wordt ingeschat aan de hand van het Bethesda-classificatiesysteem (5).

Wanneer er uit de Bethesda-classificatie blijkt dat er een grote kans is op een maligniteit, wordt er bij kinderen in de meerderheid van de gevallen een totale thyreoidectomie uitgevoerd (6, 7). Indien er ook bewijs is van of een sterke verdenking op lokale lymfekliermetastasen, wordt er tevens een halsklierdissectie uitgevoerd. Na de chirurgische ingreep, kan de vervolgbehandeling bestaan uit het toedienen van radioactief jodium (131I). 131I vernietigt schildklierweefsel, waardoor het onder andere wordt gebruikt om resterend schildklier(tumor)weefsel, kleine inoperabele lymfekliermetastasen of metastasen op afstand te behandelen (8, 9). Na een (totale) thyreoidectomie is levenslange suppletie met levothyroxine noodzakelijk. Omdat het thyreoïdstimulerend hormoon (TSH) een proliferatief effect heeft op schildklierweefsel, geeft men na de totale thyreoidectomie een TSH-suppressieve dosis levothyroxine. Hierdoor wordt de ontwikkeling van (maligne) schildklierweefsel gelimiteerd.

De overleving van patiënten met DTC op de kinderleeftijd is uitstekend: 99% van de kinderen is 30 jaar na hun initiële diagnose nog in leven (10, 11). Ondanks deze goede overleving krijgt 10 tot 30% van de kinderen een recidief van hun ziekte. In sommige gevallen is er sprake van persisterende ziekte, waarbij de tumor(marker) aantoonbaar blijft (12, 13).

Doordat veel kinderen met DTC hun ziekte overleven, is er in toenemende mate aandacht voor langetermijneffecten van DTC. De zeldzame aard van DTC bij kinderen zorgt er voor dat er weinig wetenschappelijk onderzoek wordt gedaan naar deze ziekte. Veel aspecten van de behandeling van kinderen met DTC zijn gebaseerd op resultaten die zijn verkregen bij patiënten die op de volwassen leeftijd DTC kregen.

CHAPTER 10

230

10

Het klinische beloop van gedifferentieerd schildkliercarcinoom bij kinderen Hoofdstuk 1 betreft een algemene introductie op dit proefschrift en een beschrijving van de onderzoeksdoelen van dit proefschrift. Het doel van dit proefschrift is het in kaart brengen van het klinische beloop en de uitkomsten van DTC bij kinderen. Daarnaast richten de onderzoeken zich op diverse langetermijneffecten van de ziekte en de behandeling.

Het klinische beloop en de uitkomsten van kinderen met DTC in Nederland worden besproken in Hoofdstuk 2. De overleving in dit Nederlandse cohort was 100% na een mediane periode van 14 jaar na diagnose. Uit de medische dossiers van 105 deelnemers bleek dat er bij één op de drie patiënten sprake was van een chirurgische complicatie met permanente schade (bv. hypoparathyreoïdie of nervus recurrens letsel). De meeste patiënten kwamen in remissie, 9% van de patiënten had persisterende ziekte en bij 8% was er sprake van een recidief van de ziekte. Kinderen die zich presenteerden met gevorderde ziekte en kinderen met persisterende ziekte werden intensiever behandeld.

Een klein deel van de kinderen met DTC ontwikkelt metastasen op afstand, waarbij longmetastasen het meest frequent voorkomen. Onderzoeken die deze specifieke groep patiënten evalueren, worden vaak beperkt door het kleine aantal patiënten in hun onderzoeksgroep. Hoofdstuk 3 beschrijft de resultaten van een samenwerking met het MD Anderson Cancer Center in de Verenigde Staten, waarbij we het klinische beloop en de somatische mutaties van 148 kinderen met DTC en afstandsmetastasen in kaart brachten. Uit dit onderzoek bleek dat een aanzienlijk deel van de kinderen met metastasen op afstand persisterende ziekte houdt na hun initiële behandeling. Ziekte-gerelateerde sterfte trad echter zelden op. In uitzonderlijke gevallen werden kinderen behandeld met systemische therapie. Deze systemische therapie grijpt in op de biologische effecten van de gevonden somatische mutatie. Bij de meeste patiënten met een bewezen somatische mutatie was er sprake van een fusie in het RET proto-oncogen. In mindere mate werden NTRK fusies of BRAF mutaties gevonden. We vonden geen duidelijke associaties tussen de verschillende somatische mutaties en de ziektekarakteristieken.

Langetermijneffecten van de behandeling met radioactief jodiumDe behandeling met 131I veroorzaakt soms een tijdelijke verstoring van de vrouwelijke fertiliteit. Het onderzoek dat wordt beschreven in Hoofdstuk 4 beoordeelde vruchtbaarheid van 56 vrouwen uit het Nederlandse cohort op de lange termijn. De evaluatie bestond uit een vragenlijst over fertiliteit en bepalingen van het anti-Müller-hormoon (AMH, een maat voor ovariële reserve), luteïniserend hormoon (LH), follikelstimulerend hormoon (FSH) en oestradiol (E2). AMH niveaus van deelnemers werden uitgezet tegen een vergelijkingsgroep bestaande uit vrouwen die niet waren

APPENDICES

231

10

behandeld voor kanker. Uit de resultaten bleek dat er geen permanente schade van fertiliteit optrad bij deze vrouwen die op de kinderleeftijd waren behandeld voor DTC.

Omdat er weinig jongens worden gediagnosticeerd met DTC, kozen we er voor om mannelijke fertiliteit na de behandeling met 131I te onderzoeken bij patiënten die op de volwassen leeftijd waren behandeld (Hoofdstuk 5). Op deze manier konden we een representatieve groep deelnemers includeren. We onderzochten mannen die minimaal twee jaar geleden hun laatste behandeling met 131I hadden gehad. Daarnaast moesten ze zijn behandeld met een minimale cumulatieve dosis van 100 mCi. Deelname bestond uit een semenanalyse, het meten van LH, FSH en testosteron en een fertiliteitsgeoriënteerde vragenlijst. De kwaliteit van het semen van deelnemers werd vergeleken met referentiewaarden van een algemene populatie, gepubliceerd door de Wereldgezondheidsorganisatie (14). De semenkwaliteit van het 5e, 10e en 25ste percentiel van deze algemene populatie werd gebruikt als afkappunt. Het aantal deelnemers met een semenkwaliteit die lager lag dan het afkappunt gebaseerd op het 5e percentiel, was klinisch en statistisch significant groter dan de vijf procent in de algemene populatie. Vergeleken met de twee andere afkappunten was de proportie deelnemers met een lage semenkwaliteit gelijk aan het aantal mannen met een lage semenkwaliteit in de algemene populatie. De antwoorden op de vragenlijst over vruchtbaarheid en de hormoonwaarden lieten geen klinisch relevante afwijkingen zien.

De kans is klein dat mannen die zijn behandeld met 131I voor DTC op lange termijn een verminderde semenkwaliteit hebben. Omdat we echter niet weten welke mannen een verhoogd risico hebben op een lage semenkwaliteit, verdient het de voorkeur om semenpreservatie te bespreken en aan te bieden aan alle mannen die een dosis van 100 mCi of meer toegediend zullen krijgen.

Langetermijneffecten van TSH suppressie therapieVijf jaar geleden werd de cardiale functie van het Nederlandse cohort voor het eerst onderzocht. Een op de vijf deelnemers bleek diastolische dysfunctie te hebben (15). De systolische functie was niet aangedaan. Daarnaast werd er bij geen van de deelnemers atriumfibrilleren geobserveerd.

Omdat diastolische dysfunctie is geassocieerd met een versnelde cardiale veroudering, wilden we het klinische beloop van deze bevinding vervolgen middels een vijfjarig vervolgonderzoek. De resultaten van deze studie worden besproken in Hoofdstuk 6. Gepaarde analyses lieten zien dat de initiële diastolische dysfunctie verder toenam. Een significante stijging in het N-terminal pro-B-type natriureticpeptide ondersteunde de bevinding dat er hartschade optrad. Mogelijk wordt deze cardiale schade bij patiënten die op de kinderleeftijd zijn behandeld voor DTC in de toekomst klinisch relevant. De bevinding vereist een vervolg in de toekomst, waarbij cardiale screening van kinderen mogelijk van toepassing is.

CHAPTER 10

232

10

Langetermijneffecten op kwaliteit van leven en psychosociale ontwikkelingIn Hoofdstuk 7 beschrijven we een studie waarin we gezondheidsgerelateerde kwaliteit van leven, vermoeidheid, angst, depressie, en schildklierkanker specifieke klachten onderzochten bij volwassenen die op de kinderleeftijd waren behandeld voor DTC. De deelnemers werden vergeleken met controlepersonen die waren gematcht op leeftijd en geslacht. Het welzijn van de deelnemers was over het algemeen gelijk aan dat van de controlepersonen. Deelnemers rapporteerden net iets vaker fysieke problemen of mentale vermoeidheid.

De resultaten in Hoofdstuk 8 beschrijven het behalen van psychosociale ontwikkelingsmijlpalen van deze kinderen. In vergelijking met controlepersonen van een vergelijkbare leeftijd en hetzelfde geslacht, waren de ontwikkeling op het gebied van autonomie, de sociale ontwikkeling en de psychoseksuele ontwikkeling vergelijkbaar. In vergelijking met patiënten die voor een andere soort kinderkanker waren behandeld, bleek dat de sociale ontwikkeling bij kinderen met DTC beter was. Ontwikkeling van autonomie en psychoseksuele ontwikkeling waren vergelijkbaar in beide groepen patiënten. We concludeerden dat de diagnose en behandeling van DTC bij kinderen hun psychosociale levensloop niet beïnvloedt.

Over het algemeen lijken het welzijn en de psychosociale ontwikkeling van kinderen met DTC niet te zijn aangedaan. Omdat dit de eerste onderzoeken zijn die deze uitkomsten in kaart brachten, verdient het de voorkeur om deze resultaten te bevestigen in toekomstig onderzoek.

APPENDICES

233

10

REFERENTIES1. NKR Cijfers [Internet]. Integraal Kankercentrum Nederland. 2020 [cited April 2020]. Available from:

https://www.iknl.nl/nkr-cijfers.

2. Siegel DA, King J, Tai E, Buchanan N, Ajani UA, Li J. Cancer incidence rates and trends among children and

adolescents in the United States, 2001-2009. Pediatrics. 2014;134(4):945.

3. Steliarova-Foucher E, Stiller CA, Pukkala E, Lacour B, Plesko I, Parkin DM. Thyroid cancer incidence and survival

among European children and adolescents (1978-1997): report from the Automated Childhood Cancer Information

System project. Eur J Cancer. 2006;42(13):2150-2169.

4. Rivkees SA, Mazzaferri EL, Verburg FA, Reiners C, Luster M, Breuer CK, Dinauer CA, Udelsman R. The treatment of

differentiated thyroid cancer in children: emphasis on surgical approach and radioactive iodine therapy. Endocr

Rev. 2011;32(6):798-826.

5. Cibas ES, Ali SZ. The 2017 Bethesda System for reporting thyroid cytopathology. Thyroid. 2017;27(11):1341-1346.

6. Francis GL, Waguespack SG, Bauer AJ, Angelos P, Benvenga S, Cerutti JM, Dinauer CA, Hamilton J, Hay ID,

Luster M, Parisi MT, Rachmiel M, Thompson GB, Yamashita S, American Thyroid Association Guidelines

Task Force. Management Guidelines for Children with Thyroid Nodules and Differentiated Thyroid Cancer.

Thyroid. 2015;25(7):716-759.

7. Lebbink CA, Dekker BL, Bocca G, Braat AJ, Derikx JP, Dierselhuis MP, de Keizer B, Kruijff S, Kwast AB, van Nederveen

FH, Nieveen van Dijkum E, Nievelstein RA, Peeters RP, Terwisscha van Scheltinga CE, Tissing WJ, van der Tuin K, Vriens

MR, Zsiros J, van Trotsenburg AS, Links TP, van Santen HM. New national recommendations for the treatment of

pediatric differentiated thyroid carcinoma in the Netherlands. Eur J Endocrinol. 2020;183(4):P11-P18.

8. Jarzab B, Handkiewicz-Junak D, Wloch J. Juvenile differentiated thyroid carcinoma and the role of radioiodine in

its treatment: a qualitative review. Endocr Relat Cancer. 2005;12(4):773-803.

9. Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Mang O, Lau WH. Differentiated thyroid carcinoma in childhood

and adolescence-clinical course and role of radioiodine. Pediatr Blood Cancer. 2004;42(2):176-183.

10. Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in

1753 patients. J Surg Res. 2009;156(1):167-172.

11. Hay ID, Johnson TR, Kaggal S, Reinalda MS, Iniguez-Ariza NM, Grant CS, Pittock ST, Thompson GB. Papillary

thyroid carcinoma (PTC) in Children and Adults: comparison of initial presentation and long-term postoperative

outcome in 4432 patients consecutively treated at the Mayo Clinic during eight decades (1936-2015). World J

Surg. 2018;42(2):329-342.

12. Russo M, Malandrino P, Moleti M, Vermiglio F, D’Angelo A, La Rosa G, Sapuppo G, Calaciura F, Regalbuto C, Belfiore

A, Vigneri R, Pellegriti G. Differentiated thyroid cancer in children: Heterogeneity of predictive risk factors. Pediatr

Blood Cancer. 2018;65(9):e27226.

13. Verburg FA, de Keizer B, Lam MGEH, de Klerk JMH, Lips CJM, Borel-rinkes IHM, van Isselt JW. Persistent Disease

in Patients with Papillary Thyroid Carcinoma and Lymph Node Metastases After Surgery and Iodine-131 Ablation.

World J Surg. 2007;31(12):2309-2314.

14. Cooper TG, Noonan E, von Eckardstein S, Auger J, Baker HW, Behre HM, Haugen TB, Kruger T, Wang C, Mbizvo

MT, Vogelsong KM. World Health Organization reference values for human semen characteristics. Hum Reprod

Update. 2010;16(3):231-245.

15. Klein Hesselink MS, Bocca G, Hummel YM, Brouwers AH, Burgerhof JGM, van Dam EWCM, Gietema JA, Havekes

B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker

JTM, Ronckers CM, van Santen HM, van der Meer P, Links TP, Tissing WJE. Diastolic dysfunction is common in

survivors of pediatric differentiated thyroid carcinoma. Thyroid. 2017;27(12):1481-1489.

APPENDICES

235

10

NEDERLANDSE SAMENVATTING VOOR LEKENDe schildklierDe schildklier bevindt zich in de hals, voor de luchtpijp. De schildklier is een orgaan dat hormonen maakt. De hormonen die de schildklier worden gemaakt zijn belangrijk voor de stofwisseling van het lichaam. Schildklierhormonen zijn ook van belang voor de groei van nieuwe cellen.

SchildklierkankerEr zijn verschillende soorten kanker van de schildklier. Meer dan 90% van de schildklierkankerpatiënten heeft gedifferentieerde schildklierkanker. Deze vorm van schildklierkanker is meestal niet agressief. Bij kinderen is deze ziekte heel zeldzaam. In 2019 kregen ongeveer 15 kinderen in Nederland deze diagnose. Bij volwassenen werd deze diagnose in dit jaar bij meer dan 750 patiënten gesteld. De ziekte komt vaker voor bij vrouwen dan bij mannen. Dit proefschrift gaat over de gedifferentieerde vorm van schildlierkanker, en zal verder “schildklierkanker” worden genoemd.

Schildklierkanker bij kinderen wordt meestal ontdekt doordat het kind een bult in de nek heeft. Vaak heeft het kind hier geen last van. Deze bult wordt ook wel een tumor genoemd. Met een naald kunnen er cellen uit deze tumor worden gehaald. Een patholoog bekijkt deze cellen onder de microscoop onderzoekt of dit schildklierkankercellen zijn.

Als er schildklierkankercellen worden gevonden, wordt er een operatie uitgevoerd. Meestal wordt de hele schildklier verwijderd. Als de kanker zich heeft verspreid naar de lymfeklieren in de hals, worden deze ook tijdens de operatie verwijderd. Na de operatie wordt de patiënt meestal behandeld met radioactief jodium. Radioactief jodium vernietigt achtergebleven schildklier(kanker)weefsel of uitzaaiingen. Als de hele schildklier is verwijderd, moet deze persoon levenslang tabletten met schildklierhormoon slikken. Door een net iets te hoge dosis schildklierhormoon te geven, wordt de groei van schildkliercellen onderdrukt. Dit voorkomt ook dat achtergebleven schildklierkankercellen groeien. Deze behandeling wordt onderdrukkingstherapie genoemd (schildklier stimulerend hormoon-suppressie therapie).

De overlevingskans van patiënten met schildklierkanker op de kinderleeftijd is heel goed: 99% van de kinderen is 30 jaar na hun diagnose in leven. Ondanks deze goede overleving komt de ziekte bij 10 tot 30% van de kinderen terug. In sommige gevallen blijft de ziekte aantoonbaar in het lichaam, dit wordt blijvende ziekte genoemd.

Doordat de meeste kinderen met schildklierkanker hun ziekte overleven, is er steeds meer aandacht voor de langetermijneffecten van schildklierkanker. Langetermijneffecten zijn nadelige gevolgen van een ziekte of de behandeling ervan. Omdat schildklierkanker bij kinderen zeldzaam is, is er tot nu toe weinig wetenschappelijk

CHAPTER 10

236

10

onderzoek gedaan naar deze ziekte en is er over de langetermijneffecten niet veel bekend.

Dit proefschrift Het verloop van schildklierkanker bij kinderen Hoofdstuk 1 beschrijft het doel van dit proefschrift. Met dit proefschrift willen we in kaart brengen hoe het met kinderen na hun diagnose met schildklierkanker gaat. We onderzoeken het verloop van de ziekte, de overleving en de langetermijneffecten van de ziekte.

Het verloop van de ziekte en de overleving van kinderen met schildklierkanker in Nederland worden beschreven in Hoofdstuk 2. Geen van de 170 patiënten met de diagnose schildklierkanker op de kinderleeftijd was na een periode van 14 jaar overleden aan deze kanker. Na uitgebreid onderzoek van medische dossiers van 105 deelnemers bleek dat er bij één op de drie patiënten sprake was van een complicatie door de operatie met blijvende schade. Het grootste deel van deze 105 patiënten was genezen na hun behandeling. Negen procent van de patiënten had blijvende ziekte en bij acht procent van de kinderen kwam de kanker terug. Kinderen met uitzaaiingen of agressievere ziekte kregen meer behandelingen of werden uitgebreider behandeld. Zo werden ze bijvoorbeeld behandeld met een hogere dosis radioactief jodium.

Bij een klein deel van de kinderen met schildklierkanker zaait de kanker uit naar ander deel van het lichaam (uitzaaiingen op afstand). Uitzaaiingen naar de longen komen het meeste voor. Hoofdstuk 3 beschrijft de resultaten van een samenwerking met het MD Anderson Cancer Center in de Verenigde Staten. We onderzochten 148 kinderen met schildklierkanker en uitzaaiingen op afstand. Het onderzoek bekeek het verloop van de ziekte en de aanwezigheid van veranderingen in het erfelijk materiaal van de schildkliertumor (somatische mutaties). Bij de onderzochte kinderen kwam sterfte door schildklierkanker bijna niet voor. Dit is opvallend, omdat een groot deel van de kinderen met uitzaaiingen op afstand na de behandeling niet geneest, maar blijvende ziekte houdt. Bij kinderen worden er andere veranderingen in het erfelijk materiaal van de schildkliertumor gevonden dan bij volwassenen.

Langetermijneffecten van de behandeling met radioactief jodiumVrouwelijke vruchtbaarheidDe behandeling met radioactief jodium veroorzaakt soms tijdelijke verstoring van de vrouwelijke of mannelijke vruchtbaarheid. Het onderzoek dat wordt beschreven in Hoofdstuk 4 bestudeerde de vruchtbaarheid van 56 vrouwen uit de Nederlandse onderzoeksgroep uit hoofdstuk 2 op de lange termijn. Het onderzoek bestond onder andere uit het meten van een vruchtbaarheidshormoon, het anti-Müller-hormoon (AMH). Dit hormoon geeft een schatting van het aantal eicellen bij de vrouw. Hoe hoger het niveau van dit hormoon, hoe groter de eicelvoorraad bij deze vrouw.

APPENDICES

237

10

Het niveau van het vruchtbaarheidshormoon bij de patiënten die op kinderleeftijd behandeld waren voor schildklierkanker verschilde niet van het niveau van dit hormoon bij gezonde vrouwen.

Het onderzoek bestond daarnaast uit een vragenlijst over vruchtbaarheid. We vroegen de vrouwen bijvoorbeeld of het lukte om zwanger te worden en of hun geboren kinderen gezond waren. Ook uit de antwoorden op de vragenlijst bleek dat hun vruchtbaarheid op lange termijn normaal was.

Mannelijke vruchtbaarheidOmdat er weinig jongens op de kinderleeftijd schildklierkanker krijgen, hebben we vruchtbaarheid na schildklierkanker onderzocht bij patiënten die op de volwassen leeftijd waren behandeld (Hoofdstuk 5). Op deze manier kon er een grotere groep deelnemers meedoen.

We onderzochten mannen die minimaal twee jaar geleden hun laatste behandeling met radioactief jodium kregen. Daarnaast waren ze behandeld met een hoge dosis radioactief jodium.

Deelname bestond uit een spermaonderzoek, het meten van hormonen in het bloed en een vragenlijst over vruchtbaarheid. De kwaliteit van het sperma van de deelnemers werd vergeleken met de spermakwaliteit van de algemene bevolking. We onderzochten of een lage spermakwaliteit vaker voorkwam bij patiënten die waren behandeld voor schildklierkanker dan in de algemene bevolking. We vergeleken de spermakwaliteit van de patiënten met de spermakwaliteit van de laagste 5, 10 en 25 procent in de algemene bevolking.

Er waren meer patiënten met schildklierkanker met een slechte spermakwaliteit in deze laagste 5-procent groep, dan in de algemene bevolking. Vergeleken met de laagste 10 en 25 procent, was de spermakwaliteit van patiënten gelijk aan de spermakwaliteit in de algemene bevolking. De antwoorden op de vragenlijst over vruchtbaarheid (bijvoorbeeld de verwekte zwangerschappen en de afloop van de zwangerschappen) en de hormoonwaarden lieten geen belangrijke schade aan de mannelijke vruchtbaarheid zien.

De kans is klein dat mannen die zijn behandeld met radioactief jodium voor schildklierkanker op lange termijn een slechte spermakwaliteit hebben. We weten alleen niet welke mannen op lange termijn te maken krijgen met een slechte spermakwaliteit. We adviseren daarom dat het invriezen (cryopreserveren) van sperma moet worden besproken met elke man die wordt behandeld met een hoge dosis radioactief jodium. Per patiënt kan er besloten worden om het sperma wel of niet in te vriezen.

CHAPTER 10

238

10

Langetermijneffecten van een hoge dosis schildklierhormoonHartfunctieVijf jaar geleden werd de hartfunctie van de onderzoeksgroep voor het eerst onderzocht. Eén op de vijf deelnemers had een vorm van hartschade waarbij de hartspier zich niet (meer) goed ontspant (diastolische dysfunctie). Een hart dat minder goed ontspant, kan zich minder goed vullen met bloed. De uitpompfunctie van het hart (systolische functie) was niet beschadigd. Ritmestoornissen van het hart kwamen niet voor bij de deelnemers. Van patiënten die op de volwassen leeftijd zijn behandeld voor schildklierkanker weten we dat een verminderde ontspanningsfunctie kan worden veroorzaakt door een hoge dosis schildklierhormoon.

Omdat een verminderde ontspanningsfunctie is gekoppeld aan een versnelde veroudering van het hart, onderzochten we de hartfunctie van deze deelnemers na vijf jaar nog een keer. Deze resultaten worden besproken in Hoofdstuk 6.

De ontspanningsfunctie van het hart nam in deze vijf jaar verder af. Er ontstond dus meer hartschade. Het meten van een eiwit in het bloed dat wordt gemaakt door het hart, bevestigde dat er hartschade ontstond.

Op dit moment is de hartschade nog zo minimaal dat de patiënt hier niets van merkt. We weten alleen niet of deze hartschade bij de schildklierkankerpatiënten toeneemt, of zij in de toekomst klachten krijgen of ziek worden. Dit willen we onderzoeken door het onderzoek over een paar jaar nog een keer te herhalen.

Langetermijneffecten op kwaliteit van leven en psychosociale ontwikkelingKwaliteit van levenIn Hoofdstuk 7 beschrijven we een studie waarin we de kwaliteit van leven, vermoeidheid, angst, depressie, en schildklierkanker specifieke klachten onderzochten bij volwassenen die op de kinderleeftijd waren behandeld voor schildklierkanker. De antwoorden van de deelnemers werden vergeleken met de antwoorden van controlepersonen met een vergelijkbare leeftijd en hetzelfde geslacht. Het welzijn van de deelnemers was over het algemeen hetzelfde in beide groepen. Patiënten met schildklierkanker antwoordden net iets vaker dat zij lichamelijke problemen of geestelijke vermoeidheid hadden.

Psychosociale ontwikkelingDe resultaten in Hoofdstuk 8 beschrijven het behalen van ontwikkelingsmijlpalen van dezelfde groep patiënten die op de kinderleeftijd schildklierkanker heeft gehad. Vergeleken met de controlepersonen waren de ontwikkeling op het gebied van zelfstandigheid (autonomie), sociale ontwikkeling en psychoseksuele ontwikkeling hetzelfde. In vergelijking met patiënten die voor een andere soort kinderkanker waren behandeld, was de sociale ontwikkeling bij kinderen met schildklierkanker beter. De ontwikkeling van zelfstandigheid en psychoseksuele ontwikkeling waren

APPENDICES

239

10

hetzelfde in beide groepen patiënten. Uit dit onderzoek blijkt dat de diagnose en behandeling van schildklierkanker bij kinderen hun ontwikkeling op deze gebieden niet beschadigd.

Over het algemeen lijken het welzijn en de psychosociale ontwikkeling van kinderen met schildklierkanker normaal. Omdat dit de eerste onderzoeken zijn over dit onderwerp, willen we dit verder onderzoeken in de toekomst.

APPENDICES

241

10

DANKWOORDDat ik dit dankwoord mag schrijven is een goed teken: mijn proefschrift is af! Dit proefschrift stoelt op de zichtbare en onzichtbare bijdragen van de mensen die ik (gelukkig) om mij heen heb. Er zijn veel mensen die ik dankbaar ben voor hun bijdrage aan dit proefschrift.

Allereerst wil ik de deelnemers en patiënten bedanken voor hun deelname aan de onderzoeken. Ik ben onder de indruk van jullie openheid en bereidheid om anderen te helpen. Dank jullie wel!

Na een maandagochtendbespreking stapte ik vrolijk(er) het kantoor van Thera uit: het overleg met mijn (co)promotoren deed me altijd goed.

Prof. dr. Thera P. Links, beste Thera, je gedrevenheid en enthousiasme werken erg aanstekelijk. Ik wil je bedanken voor alle levenslessen op het gebied van wetenschappelijk onderzoek, endocrinologie en samenwerkingen, maar ook zeker op persoonlijk gebied. Ik ga het missen om zo intensief samen te werken.

Prof. dr. Wim. J.E. Tissing, beste Wim, door je vanzelfsprekende vertrouwen in een goede uitkomst leerde ik hier ook op vertrouwen. Je leerde me dat beslissingen maken niet altijd moeilijk hoeft te zijn. Bedankt voor je fijne begeleiding.

Dr. Gianni Bocca, beste Gianni, met je nuchtere kijk op zaken zorgde je voor rust tijdens mijn promotietraject. Ik vond het erg gezellig om met je samen te werken. Dank je wel voor je nauwe betrokkenheid en razendsnelle feedback.

Ik wil de leescommissie, prof. dr. R. Sanderman, prof. dr. A. Hoek, en prof. dr. C.M. Zwaan, bedanken voor de tijd en moeite die zij hebben genomen om mijn proefschrift te beoordelen.

Professor Steven. G. Waguespack, dear Steven, I am grateful that I have the opportunity of working with you. The compassion with which you treat your patients is inspiring. Thank you for your persistent preciseness to make our work perfect. I promise that I will not forget about Endocrinology.

I want to thank my colleagues and collaborators from the MD Anderson for working with me. In particular I want to thank Marilyne, Sasan, Sara, Pamm, Londa, and Eduardo for making my stay so enjoyable!

Beste Carla, dank je wel voor alle gezellige en vrolijke koffiemomentjes. Het was fijn om onze ervaringen te delen en tijd met elkaar door te brengen.

Dr. A.N.A. van der Horst-Schrivers, beste Anouk, bedankt voor je begeleiding en betrokkenheid bij mijn promotietraject. Ik ben blij dat onze wegen elkaar kruisten. Ik heb veel aan je adviezen gehad!

CHAPTER 10

242

10

Dr. ir. Eus G.J.M. Arts en dr. Astrid E.P. Cantineau, beste Eus en Astrid, ik heb veel geleerd van jullie kennis en ervaring in de voortplantingsgeneeskunde. Ik wil jullie bedanken voor deze fijne samenwerking.

Dr. Gea Huizinga en dr. Esther Sulkers, bedankt dat jullie me introduceerden in het kwaliteit van leven onderzoek. Door de combinatie van jullie theoretische en praktische kennis werd het schrijven van de artikelen een stuk eenvoudiger.

Hans Burgerhof, beste Hans, ik vond het prettig om met je samen te werken. Bedankt voor je precisie en het concreet maken van onze soms vage plannen.

Ik wil de hoofdonderzoekers van de andere centra, de leden van het Nederlands onderzoeksconsortium voor schildklierkanker bij kinderen, en andere coauteurs bedanken. Dankzij jullie hebben we de multicenter studies succesvol kunnen opzetten en afronden. De geschreven en ongeschreven regels maakten het werk niet altijd makkelijk, maar ik ben jullie dankbaar voor al jullie inspanningen. Bedankt voor al jullie waardevolle bijdragen aan de studies en de daaropvolgende artikelen.

Aan de medewerkers en collega’s van de interne geneeskunde, cardiologie, nucleaire geneeskunde, voortplantingsgeneeskunde en laboratoriumgeneeskunde van de onderzoekscentra: bedankt voor jullie hulp en ondersteuning!

Mijn begeleiders en de medewerkers van het Isala fertiliteitscentrum, tijdens mijn semi-artsstage leerde ik door jullie over de dagelijkse klinische praktijk van de voortplantingsgeneeskunde. Met veel plezier heb ik samen met jullie gewerkt!

Mariëlle, jouw werk legde de basis voor dit proefschrift. Doordat je het onderzoek met zoveel precisie uitvoerde, was het een feest om met deze data verder te werken. Wat is het leuk dat we de projecten samen kunnen afsluiten.

Lammie, bedankt voor je inspanningen voor de studies. Het was erg fijn om met je samen te werken.

Marga, dank je wel voor al je ondersteunende werkzaamheden.

Tharsana en Antoinette, wat was het leuk om jullie te mogen begeleiden, en met zo’n mooi eindresultaat! Bedankt voor al jullie hulp!

Mijn (oud-)kamergenoten en mede-onderzoekers, Sandra, Sarah, Edward, Jorien, Karin, Robert en Trynke, bedankt voor de gezelligheid, tips en adviezen. Tom, Annet, Hanneke, Charlotte en Rita, de digitale vrijdagmiddagborrels maakten het

APPENDICES

243

10

thuiswerken een stuk leuker! Bernadette, Yvonne en Dineke, ik heb het gemist om bij jullie op de onderzoekskamer te werken de afgelopen maanden. Bedankt voor alle gezelligheid, steun, gesprekken over belangrijke (en vaak ook minder belangrijke) dingen en de motiverende ballonnen en taart!

Ellen, Lara, Romy, Lotte en Eline, wat gaan we al een tijd mee. Ondanks de fysieke afstand voelen jullie nooit écht ver weg. Bedankt voor jullie steun, de fantastische chaos als we bij elkaar zijn en deze onvoorwaardelijke basis.

Judith, Jelger, Joren en Pascalle, wie had er gedacht dat jullie door mijn promotietraject op vakanties op stapelbedden en slaapbanken moesten slapen. Hier ben ik jullie nog flink wat kopjes koffie verschuldigd. Bedankt voor de afleiding, gezelligheid en harde grappen.

Larissa, Koen, Annet, Tim, Miron, Klaske en Hilde, jullie waren niet altijd bijdragend aan mijn productiviteit en dat is maar goed ook. Bedankt voor alle ruimte voor (onderzoeks)klaagjes, etentjes en andere gezelligheid. Zonder jullie was het allemaal een stuk minder leuk geweest. Tim en Annet, ik voel me vereerd dat jullie tijdens mijn verdediging naast me willen staan als paranimfen. Bedankt voor jullie ondersteuning in de vorm van een slaapplek, (soms eindeloze) hardloop- of fietsrondjes en relativerende gesprekken.

Jan en Corry, als ik ergens leer dat doorzetten wordt beloond, is dat bij jullie thuis. Bedankt voor het warme nest dat jullie bieden.

Christine, Gerard en Jacco-Pieter, bedankt voor jullie betrokkenheid en vrolijke kleurplaten.

Pap en mam, bedankt voor jullie steun en de rust die jullie mij bieden. Ik ben jullie dankbaar voor het vertrouwen dat jullie in me hebben.

Jan, Joanne, Jur en Lisa (en baby!), wat heb ik een geluk met zo’n fijne familie. Het is heel leuk om naast grafiekjes ook zoveel andere dingen in het leven met jullie te kunnen delen.

Lieve Richard, ik ben blij dat jij er bent om alles met me mee te maken. Dank je wel voor het zijn van mijn persoonlijke mascotte, je steun en het meevieren van de successen. Mijn werk is leuk, maar het leukste aan mijn werkdag is thuiskomen bij jou.

CHAPTER 10

244

10

ABOUT THE AUTHORMarloes Nies was born on May 9th, 1991 in Enschede, the Netherlands. Growing up in Zwolle, she graduated from the Carolus Clusius College in 2009. Hereafter, she studied Human Movement Sciences at the University of Groningen, obtaining her Bachelor’s degree in 2012. A year later, she started the pre-master Medicine at the same university and enrolled in the Master’s program. Marloes performed her research internship at the department of Endocrinology of the University Medical Center Groningen (UMCG), evaluating the clinical course of differentiated thyroid cancer (DTC) during childhood in the Netherlands. Her application for a MD/PhD-student position, expanding on this research, was approved after which she alternated her medical training and PhD research. In 2019, she obtained her Master’s degree in Medicine. The PhD research involved setting up and completing two multicenter studies in the Netherlands that evaluated several late effects of treatment of childhood DTC. During the last year of her PhD training, she performed research at the department of Endocrine Neoplasia and Hormonal Disorders of the University of Texas MD Anderson Cancer Center, evaluating children with distant metastases from childhood DTC. In September of 2020, she finished her PhD. Since October of 2020, Marloes works at the department of Radiotherapy of the UMCG.

APPENDICES

245

10

LIST OF PUBLICATIONSKlein Hesselink MS, Nies M, Bocca G, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Schmid KW, Smit JW, Williams GR, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP. Pediatric differentiated thyroid carcinoma in the Netherlands: a nationwide follow-up study. J Clin Endocrinol Metab. 2016;101(5):2031–2039.

Nies M*, Klein Hesselink MS*, Huizinga GA, Sulkers E, Brouwers AH, Burgerhof JG, van Dam EW, Havekes B, van den Heuvel-Eibrink MM, Corssmit EP, Kremer LC, Netea-Maier RT, van der Pal HJ, Peeters RP, Plukker JT, Ronckers CM, van Santen HM, Tissing WJ, Links TP, Bocca G. Long-term quality of life in adult survivors of pediatric differentiated thyroid carcinoma. J Clin Endocrinol Metab. 2017;102(4):1218-1226

Nies M, Dekker BL, Sulkers E, Huizinga GA, Klein Hesselink MS, Maurice-Stam H, Grootenhuis MA, Brouwers AH, Burgerhof JGM, van Dam EWCM, Havekes B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker JTM, Ronckers CM, van Santen HM, van der Horst-Schrivers ANA, Tissing WJE, Bocca G, Links TP. Psychosocial development in survivors of childhood differentiated thyroid carcinoma: a cross-sectional study. Eur J Endocrinol. 2018;178(3):215-223.

Selvakumar T, Nies M*, Klein Hesselink MS*, Brouwers AH, van der Horst-Schrivers ANA, Klein Hesselink EN, Bocca G, Burgerhof JGM, van Dam EWCM, Havekes B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Smit JWA, Plukker JTM, Ronckers CM, van Santen HM, Tissing WJE, Vissink A, Links TP. Long-term effects of radioiodine treatment on salivary gland function in adult survivors of pediatric differentiated thyroid carcinoma. J Nucl Med. 2019;60(2):172-177.

Nies M, Cantineau AEP*, Arts EGJM*, van den Berg MH, van Leeuwen FE, Muller Kobold AC, Klein Hesselink MS, Burgerhof JGM, Brouwers AH, van Dam EWCM, Havekes B, van den Heuvel-Eibrink MM, Corssmit EPM, Kremer LCM, Netea-Maier RT, van der Pal HJH, Peeters RP, Plukker JTM, Ronckers CM, van Santen HM, van der Horst-Schrivers ANA, Tissing WJE, Bocca G, van Dulmen-den Broeder E**, Links TP**. Long-term effects of radioiodine treatment on female fertility in survivors of childhood differentiated thyroid carcinoma. Thyroid. 2020;30(8):1169-1176.

Nies M, Arts EGJM, van Velsen EFS, Burgerhof JGM, Muller Kobold AC, Corssmit EPM, Netea-Maier RT, Peeters RP, van der Horst-Schrivers ANA, Cantineau AEP**, Links TP**. Long-term evaluation of male fertility after treatment with radioactive iodine for differentiated thyroid carcinoma. Submitted.

CHAPTER 10

246

10

Reichert AD*, Nies M*, Tissing WJE, Muller Kobold AC, Klein Hesselink MS, Brouwers AH, Havekes B, van den Heuvel-Eibrink MM, van der Pal HJH, Plukker JTM, van Santen HM, Burgerhof JGM, van der Meer P, Corssmit EPM, Netea-Maier RT, Peeters RP, van Dam EWCM, Bocca G, Links TP. Cardiac dysfunction in survivors of pediatric differentiated thyroid carcinoma. Submitted.

Nies M, Vassilopoulou-Sellin R, Bassett RL, Yedururi S, Zafereo ME, Cabanillas ME, Sherman SI, Links TP, Waguespack SG. Distant metastases from childhood differentiated thyroid carcinoma: clinical course and mutational landscape. Submitted.

* and **: both authors contributed equally.

Recommended