You are on page 1of 9

Mini Review

HOR MONE Horm Res Paediatr Received: August 21, 2018


RESEARCH I N DOI: 10.1159/000493943 Accepted: September 20, 2018
Published online: November 7, 2018
PÆDIATRIC S

Childhood Cancer Treatments and Associated


Endocrine Late Effects: A Concise Guide for the
Pediatric Endocrinologist
Wassim Chemaitilly a, b Charles A. Sklar c
   

a Department
of Pediatric Medicine, Division of Endocrinology, St. Jude Children’s Research Hospital, Memphis, TN,
USA; b Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA;
 

c Department of Pediatrics, Memorial-Sloan Kettering Cancer Center, New York, NY, USA
 

Keywords crinologist in preparation for the clinical encounter with a


Childhood cancer survivor · Endocrine late-effects · CCS. A more detailed discussion of the management of spe-
Endocrine complications · Radiotherapy late-effects · cific endocrine late effects can be found in the other chap-
Chemotherapy late-effects ters in this series. © 2018 S. Karger AG, Basel

Abstract
Endocrine complications are frequently observed in child- Introduction
hood cancer survivors (CCS); in many instances, these com-
plications develop months to years after the completion of Progress in the treatment of cancer during childhood
cancer therapy. The estimated prevalence of endocrine late has resulted in 5-year survival rates exceeding 80%. The
effects is 50% among CCS; the main risk factors are external proportion of patients surviving pediatric cancer has
beam radiation that includes key endocrine organs (the hy- steadily increased over the last 4 decades; in the United
pothalamus/pituitary, thyroid and gonads) and/or alkylating States, the population of childhood cancer survivors
agents. Novel agents targeting tumor growth have increased (CCS) is poised to exceed 500,000 individuals by 2020 [1].
the options available to a small number of patients albeit Key to this success has been the use of multi-modal, in-
with the need for treatment over long periods of time. Some tensive treatment regimens combining surgery, radio-
of these agents, such as certain tyrosine kinase inhibitors and therapy, and/or chemotherapy coupled with effective,
immune system modulators have been shown to cause per- multi-disciplinary, supportive care strategies aimed at the
manent endocrine deficits. This chapter offers a brief sum- management of acute and frequently severe treatment
mary of the conventional treatment strategies for the most complications. Complications of cancer treatments may
common cancers of childhood and a brief overview of the also occur in a delayed fashion as chronic health condi-
endocrine late effects most commonly associated with these tions, developing months to years after the completion of
exposures. The impact of targeted therapies on the endo- cancer therapy; these are referred to as late effects [2]. En-
crine system will also be discussed. The aim of this chapter is docrine conditions are among the most commonly re-
to provide basic guidance to the consulting pediatric endo- ported late effects in CCS with a prevalence of 50% [3].
130.237.165.40 - 11/14/2018 1:17:10 PM

© 2018 S. Karger AG, Basel Wassim Chemaitilly, MD


Stockholm University Library

St. Jude Children’s Research Hospital, Division of Endocrinology


262 Danny Thomas Place MS 737
E-Mail karger@karger.com
Memphis, TN 38105 (USA)
www.karger.com/hrp
Downloaded by:

E-Mail wassim.chemaitilly @ stjude.org


The variable latency period between the time of diag- using 2-dimensional models to photon-based three-di-
nosis of cancer and the onset of late effects mandates re- mensional “conformal” radiotherapy [9] and intensity-
peated evaluations and lifelong follow-up in many in- modulated radiation therapy, a technique that allows the
stances [2]. Enhanced knowledge of the risk conferred by option to better take into account the shape of the tar-
a given cancer diagnosis and its treatment has allowed the geted tumor and reduce the amount of scatter to sur-
establishment of long-term follow-up guidelines, such as rounding structures [10]. Proton-based radiotherapy, a
those proposed by the Children’s Oncology Group – technique that seeks to exploit the decreased propensity
www.survivorshipguidelines.org. These guidelines assist of protons to travel beyond a targeted site in comparison
medical providers in determining which survivors are at to photons, has been recently introduced with the aim of
risk for specific late effects and how best to screen CCS decreasing even further the exposure of normal tissues
for various chronic health conditions. Nevertheless, re- located in proximity to the treated tumor [11]. Currently,
cent reports indicate that a relatively high proportion of only a limited number of institutions offer proton-based
at-risk CCS have undiagnosed and untreated endocrine radiotherapy and only a small number of studies, all with
conditions [2]. short follow-up durations, have reported data on endo-
A medical encounter with a CCS requires thorough crine late effects in children treated with proton-based
preparation by the consulting pediatric endocrinologist. radiotherapy, so the long-term risks remain unknown
CCS frequently have complex past medical histories and [12, 13].
multiple coexisting health conditions. It is essential to ob-
tain, whenever possible, detailed information on cancer Cranial Radiotherapy
history and treatment exposures, as these data are indis- Children are exposed to cranial radiotherapy most
pensable for accurate assessments and appropriate coun- commonly for the treatment of central nervous system
seling [4]. The present chapter seeks to provide basic (CNS) tumors. The most common childhood CNS tu-
guidance to the consulting pediatric endocrinologist by mors treated with radiotherapy include medulloblasto-
summarizing the treatments employed to treat the most ma, craniopharyngioma, ependymoma, high-grade glio-
common cancers in the pediatric age group and the en- ma, and low-grade glioma [5, 14]. Other childhood ma-
docrine complications associated with these therapeutic lignancies that may require cranial radiotherapy include
exposures. Treatment modalities most commonly associ- acute lymphoblastic leukemia (ALL), previously for pro-
ated with endocrine late-effects are summarized in Tables phylaxis and currently for treatment of CNS disease [15],
1 and 2. and malignant non-brain solid tumors of the head or
neck (e.g., nasopharyngeal carcinoma, retinoblastoma,
and rhabdomyosarcoma) [16].
The Effect of Radiotherapy
Linear Growth Impairment and
Endocrine late effects are most often observed in sur- Hypothalamic-Pituitary Dysfunction
vivors previously exposed to external beam radiation that In contrast to tumor-related or surgically induced HP
includes the hypothalamus-pituitary (HP), thyroid, or deficits, radiation-induced HP dysfunction is limited to
gonads. Skeletal growth, bone health, body composition, anterior pituitary disorders: growth hormone deficiency,
and metabolic homeostasis may be impaired as a conse- central precocious puberty, LH/FSH deficiency, TSH de-
quence of radiation-induced endocrine deficits or be- ficiency, ACTH deficiency, and hyperprolactinemia; cen-
cause of direct radiation-related toxicity. The effect of ra- tral diabetes insipidus has not been described as a com-
diotherapy on the various endocrine organs is dictated plication of radiation [17]. The different HP axes vary in
primarily by the total dose received and the duration their vulnerability to radiotherapy [18] (Fig. 1). Growth
since treatment such that the risk increases with increas- hormone deficiency, the most common, and frequently
ing doses and greater elapsed time between exposure and only, radiation-induced HP deficit has been associated
follow-up [5–7]. Additional considerations include frac- with radiotherapy doses to the HP region ≥18 Gy [5, 19].
tion size and number [8] and the method of delivery of Central precocious puberty may occur following treat-
the radiation [9–11]. ment with doses 18–50 Gy [20]. Deficiencies in LH/FSH,
Efforts to reduce the dose of radiation received by nor- TSH, and ACTH have been associated with HP radio-
mal tissue have included over the past 30 years moving therapy at doses ≥30 Gy and hyperprolactinemia with
from X-ray based radiotherapy and treatment planning even higher doses ≥40 Gy [17–19]. Efforts to improve the
130.237.165.40 - 11/14/2018 1:17:10 PM

2 Horm Res Paediatr Chemaitilly/Sklar


Stockholm University Library

DOI: 10.1159/000493943
Downloaded by:
Table 1. Radiotherapy-associated endocrine late effects

Radiotherapy Treated conditions Possible complications


– Field

Cranial CNS tumors Anterior pituitary disorders*


Includes: Acute lymphoblastic leukemia with CNS disease Obesity
– Whole brain Non brain solid tumors:
– Infratemporal Rhabdomyosarcoma
– Nasopharyngeal Nasopharyngeal carcinoma
– Orbital Retinoblastoma
– Waldeyer’s ring
Craniospinal Medulloblastoma/PNET Anterior pituitary disorders*
Skeletal dysplasia – short stature
Primary hypothyroidism
Hyperthyroidism
Thyroid cancer
Premature ovarian insufficiency
Total body irradiation Conditioning for hematopoietic stem cell transplantation Growth hormone deficiency
Acute leukemia Skeletal dysplasia – short stature
Neuroblastoma Primary hypothyroidism
Thyroid cancer
Premature ovarian insufficiency
Male germ cell failure
Decreased bone mineral density
Abnormal glucose metabolism
Metabolic syndrome
Neck, thorax, mediastinum Hodgkin lymphoma Primary hypothyroidism
Includes: Solid tumor located within field: Hyperthyroidism
– Neck Rhabdomyosarcoma Thyroid cancer
– Chest Neuroblastoma
– Lung Ewing sarcoma
– Mantle Nasopharyngeal carcinoma
– Nasopharyngeal
– Oropharyngeal
– Supraclavicular
Abdominal, pelvic, genitourinary Hodgkin lymphoma Premature ovarian insufficiency
Includes: Solid tumor located within field: Male germ cell failure
– Flank or hemi abdomen Rhabdomyosarcoma Leydig cell failure
– Whole abdomen Neuroblastoma Abnormal glucose metabolism
– Inverted Y Acute lymphoblastic leukemia with testicular relapse
– Bladder
– Vaginal (females)
– Prostate, testes (males)
– Iliac
– Inguinal, femoral (males)
131-I-MIBG
Neuroblastoma Primary hypothyroidism
Thyroid cancer
Premature ovarian insufficiency

*  Includes deficiencies in growth hormone, LH/FSH, TSH, and/or ACTH, central precocious puberty and hyperprolactinemia.
Adapted from the Children’s Oncology Group Long-Term Follow-Up Guidelines version 4.0 www.survivorshipguidelines.org.
CNS, central nervous system; PNET, primitive neuroectodermal tumors.
130.237.165.40 - 11/14/2018 1:17:10 PM

Childhood Cancer Treatments and Horm Res Paediatr 3


Stockholm University Library

Associated Endocrine Late Effects DOI: 10.1159/000493943


Downloaded by:
Table 2. Chemotherapeutic agents associated with endocrine late effects

Category Drug Possible complication

Alkylating agents Busulfan Premature ovarian insufficiency


Carmustine Male germ cell failure
Chlorambucil Leydig cell dysfunction (most commonly subclinical)
Cyclophosphamide
Dacarbazine
Ifosfamide
Lomustine
Mechlorethamine
Melphalan
Procarbazine
Temozolomide
Thiotepa
Heavy metals Carboplatin Premature ovarian insufficiency
Cisplatin Male germ cell failure
Glucocorticoids Dexamethasone Obesity
Prednisone Decreased bone mineral density
Tyrosine kinase inhibitors Imatinib Impaired linear growth
Sorafenib Primary hypothyroidism
Sunitinib
Immunomodulator Interferon Auto-immune thyroid disease
Immune checkpoint inhibitor – Ipilimumab Immune hypophysitis:
Anti-CTLA4 monoclonal antibody Growth hormone deficiency
LH/FSH deficiency
TSH deficiency
ACTH deficiency
Auto-immune thyroid disease
Other Retinoic acid Skeletal dysplasia – short stature
Hedgehog pathway inhibitors

precision of the delivery of radiotherapy, as with the use was exposed to radiotherapy were also reported to have
of protons instead of photons, are likely to decrease scat- developed thyroid cancer [23].
ter to the HP region and may hopefully reduce or delay
the appearance of HP dysfunction especially in individu- Craniospinal Irradiation
als treated for tumors located outside the HP region [13]. Children may receive craniospinal irradiation (CSI)
for the treatment of CNS malignancies such as medullo-
Primary Thyroid Disorders blastoma or primitive neuroectodermal tumors. In addi-
Scatter from cranial radiation, especially in children tion to a standard dose delivered to the whole neuraxis,
exposed at a very young age has the potential to reach the patients may receive additional “boosts” to specific sites
thyroid gland. A history of prophylactic cranial irradia- of disease [14].
tion for ALL does not seem to be associated with a higher
risk of primary hypothyroidism [21]. However, thyroid Linear Growth Impairment and Skeletal Dysplasia
carcinoma has been reported in CCS treated with this In addition to the complications related to the cranial
modality [21] possibly as a result of exposure at a young component of CSI, exposed CCS may experience subop-
age and increased scatter due to older radiotherapy deliv- timal growth due to the direct injury of the vertebral body
ery techniques [22]. Survivors of various malignant non- growth plates and develop a skeletal dysplasia where spi-
brain solid tumors of the head and neck whose thyroid nal growth is more impaired than that of the legs [24].
130.237.165.40 - 11/14/2018 1:17:10 PM

4 Horm Res Paediatr Chemaitilly/Sklar


Stockholm University Library

DOI: 10.1159/000493943
Downloaded by:
own pre-harvested hematopoietic stem cells after treat-
■ GHD (n = 262) ment with myeloablative chemotherapy for a non-hema-
■ LH/FSHD (n = 21)
■ TSHD (n = 8)
tological cancer such as a solid tumor (e.g., medulloblas-
■ ACTHD (n = 1) toma or neuroblastoma) [14].
GHD
TSHD ■ ■ (n = 26) In patients treated for malignant hematological disor-
(n = 55)
(n = 336) ■ ■ (n = 13) ders, conditioning may involve TBI in addition to alkylat-
ACTHD ■ ■ (n = 3)
(n = 29) ing agent-based chemotherapy. The delivery of TBI has
■ ■ (n = 2)
■ ■ (n = 1) evolved over the past decades from single-dose to divided
LH/FSHD ■ ■ (n = 1) doses or fractions given via multiple sessions a day over
(n = 77) ■ ■ ■ (n = 10) 3–4 days in order to decrease treatment toxicity [8, 27].
■ ■ ■ (n = 2)
■ ■ ■ (n = 6) Other types of conditioning include non TBI-based-che-
■ ■ ■ (n = 1) motherapy-alone regimens as with fludarabine, melpha-
■ ■ ■■ (n = 14) lan, and thiotepa. Reduced intensity conditioning regi-
mens may use lower doses of fludarabine and busulfan or
fludarabine with low-dose TBI for the preparation of pa-
Fig. 1. Prevalence and overlap between anterior pituitary deficits
following cranial radiotherapy. Overlap among hypothalamic pi-
tients who are unable to tolerate traditional regimens
tuitary deficits after cranial radiotherapy in 748 adult survivors of [28].
childhood cancer survivors followed for a mean 27.3 years. Repro-
duced with permission [19]. Copyright © 2015 American Society Linear Growth Impairment and Hypothalamic
of Clinical Oncology. Pituitary Dysfunction
The risk of short stature is highest among CCS treat-
ed with HSCT who have a history of prior exposure to
cranial irradiation and conditioning with TBI [27, 29].
Primary Thyroid Disorders A subset of patients may not be able to experience an
The thyroid gland may be exposed to scatter radio- adequate pubertal growth spurt with lasting conse-
therapy after CSI. Thyroid complications observed after quences on adult height [29]. This is due, at least in part,
CSI include primary hypothyroidism, hyperthyroidism, to radiation-induced vertebral body growth plate dam-
and thyroid cancer [7, 25]. The use of proton-based ra- age [27]. HP dysfunction other than growth hormone
diotherapy for CSI holds promise in terms of decreasing deficiency is rare in HSCT recipients treated with TBI
the amount of scatter to the thyroid [13]. and who do not have a history of prior cranial radio-
therapy [29].
Primary Gonadal Disorders
Female survivors of childhood CNS tumors may expe- Primary Thyroid Disorders
rience premature ovarian insufficiency (POI) because of Primary hypothyroidism and thyroid cancer have
scatter from CSI [26]. The use of proton-based radiother- been reported following TBI-based conditioning for var-
apy for CSI holds promise in terms of decreasing the ious disorders in children and adolescents [30, 31].
amount of scatter to the ovaries [13]. The effect of scatter
from CSI to the testes seems to be minor and unlikely to Primary Gonadal Disorders
affect Leydig cell function, though it can potentially affect Nearly all female CCS treated with TBI after the age of
sperm production [26]. 10 develop POI, while a subset among those treated be-
fore the age of 10 years may experience a spontaneous
Total Body Irradiation onset for puberty or menarche [32]. Males exposed to TBI
Total body irradiation (TBI) is used for the condition- are at a great risk for impaired spermatogenesis and in-
ing of patients for hematopoietic stem cell transplanta- fertility [33]. Gonadal shielding has been reported to pos-
tion (HSCT). Allogeneic HSCT is used to treat different sibly protect the testes from the effects of TBI, but fertil-
disorders including hematological malignancies such as ity preservation via sperm banking prior to HSCT should
relapsed or high risk leukemia [15], nonmalignant blood be offered whenever feasible [34]. In contrast to germ cell
disorders such as Fanconi anemia as well as certain in- function, Leydig cell function (testosterone secretion) is
born errors of metabolism. Autologous HSCT is used to rarely affected in individuals without a history of prior
repopulate the bone marrow of an individual with her/his exposure to direct testicular irradiation [35].
130.237.165.40 - 11/14/2018 1:17:10 PM

Childhood Cancer Treatments and Horm Res Paediatr 5


Stockholm University Library

Associated Endocrine Late Effects DOI: 10.1159/000493943


Downloaded by:
Other Types of Exposure to Radiotherapy The Effect of Chemotherapy
Neck, Thoracic, and Mediastinal Radiotherapy Sites
The most common reason for the exposure of the Alkylating Agents
neck, thorax, or mediastinum to radiotherapy in pediatric Alkylating chemotherapy agents are a group of drugs
oncology is for the treatment of Hodgkin Lymphoma. that inhibit cell multiplication by adding an alkyl group
Treatments for pediatric Hodgkin lymphoma include ra- to DNA. Abnormally proliferating cells, such as cancer
diotherapy of involved regions and multi-agent chemo- cells are vulnerable to alkylating agents. Alkylating agents
therapy, with a major ongoing effort to limit the use of may also be toxic to normally multiplying nonmalignant
radiotherapy and adapt the intensity of chemotherapy to cells, which explains their gonadal toxicity and associa-
individual risk factors [36]. Treatment with radiotherapy tions with POI in females and germ cell dysfunction in
may also be necessary in children diagnosed with malig- males [40–42, 49–51]. Leydig cell failure may affect 10–
nant solid tumors of the neck or chest [37]. Survivors 57% of survivors of childhood ALL after treatment with
whose treatment involved neck irradiation are at high risk alkylating agents but the majority of cases are subclinical
for primary hypothyroidism, hyperthyroidism, and thy- or compensated with individuals experiencing with
roid cancer [6, 38, 39]. elevated LH but normal serum testosterone levels [52].
The most commonly used alkylating agents are listed in
Abdominal, Pelvic, and Genitourinary Radiotherapy Table 2.
Sites
The treatment of pediatric Hodgkin Lymphoma may Heavy Metals
require the exposure of the abdomen or pelvis to radio- Heavy metal chemotherapy drugs include cisplatin
therapy [36]. Individuals may also be exposed to abdom- and carboplatin and are used for the treatment of differ-
inal, pelvic, or genitourinary irradiation for the treatment ent solid tumors such as germ cell tumors of the CNS and
of solid tumors such as neuroblastoma and rhabdomyo- gonads as well as medulloblastoma. The use of heavy met-
sarcoma located within or near these regions [16]. Male als has been associated with a higher risk of primary go-
survivors of childhood ALL may require testicular radio- nadal injury in both males and females [49, 50].
therapy for the treatment of disease relapse in the testes
[35]. Prolonged Exposure to Systemic Glucocorticoids
Radiotherapy exposures involving the ovaries repre- Children may be exposed to high-dose systemic ste-
sent a major risk factor of POI in female CCS [40–42]. roids for many months to treat ALL [15], with potential-
Individuals whose testes have been exposed to radiother- ly lasting consequences on bone health [53] and metabo-
apy for the treatment of Hodgkin Lymphoma or solid tu- lism [54].
mors located nearby are at risk for germ cell and Leydig
cell failure [40, 43, 44]. Abdominal radiotherapy may also Novel Targeted Chemotherapy Agents
increase the risk of abnormal glucose metabolism, includ- In contrast to conventional cytotoxic chemotherapies,
ing insulin resistance and diabetes mellitus [45]. novel strategies for cancer treatment include targeted
agents designed to interfere with specific molecular path-
Treatment with (131)-I-Metaiodobenzylguanine ways to inhibit tumor growth. These agents may be used
Children diagnosed with neuroblastoma may require over long periods of time as maintenance therapy. De-
treatment with (131)-I-metaiodobenzylguanine (131–I- spite the intent for new treatment strategies to limit the
MIBG) and HSCT and have a high risk of developing pri- collateral damage to normal tissue by targeting tumor-
mary hypothyroidism [46]. Thyroid cancer has also been specific pathways, long-term endocrine and skeletal com-
reported after treatment with 131-I-MIBG for neuroblas- plications have been reported in individuals exposed to
toma without neck irradiation and despite prophylaxis such therapies.
with potassium iodide [47]. Treatment with 131-I-MIBG
for neuroblastoma has more recently been shown to pos- Tyrosine Kinase Inhibitors
sibly be associated with POI [48]. Imatinib mesylate, a tyrosine kinase inhibitor, has be-
come a frontline therapy option for children with chron-
ic myeloid leukemia, a myeloproliferative disorder with
an activated BCR-ABL tyrosine kinase fusion protein [55,
56]. Children may be treated with imatinib for other con-
130.237.165.40 - 11/14/2018 1:17:10 PM

6 Horm Res Paediatr Chemaitilly/Sklar


Stockholm University Library

DOI: 10.1159/000493943
Downloaded by:
ditions such as relapsed solid tumors [57]. Treatment cal trials to treat a subset of patients with malignancies
with imatinib has been shown to cause linear growth de- believed to be driven by the aberrant activation of this
celeration [56]. The mechanism of growth failure in chil- pathway, have recently been shown to also cause growth
dren treated with tyrosine kinase inhibitors such as ima- plate damage and premature epiphyseal fusions [60].
tinib is not well understood and may involve perturba-
tions of the GH-IGF-I axis and direct skeletal effects [55].
Primary hypothyroidism is also among the most com- Conclusion
monly reported side effects of treatment with tyrosine ki-
nase inhibitors, especially imatinib, sorafenib, and suni- Endocrine late effects are frequently observed in CCS,
tinib; the mechanism by which these agents cause hypo- primarily as a result of the therapies used to treat their
thyroidism is unknown and may involve multiple primary cancer. The optimal management of these pa-
pathways [57]. tients mandates obtaining detailed information regard-
ing the survivor’s therapeutic exposures such as radiation
Immune System Modulators fields, doses of radiation to key endocrine organs as well
Anti-CTLA-4 monoclonal antibodies including ipili- type and doses of chemotherapy. As endocrine complica-
mumab, which are increasingly used for the treatment of tions may develop months to years following a cancer di-
relapsed solid tumors, have been reported to cause im- agnosis, survivors at risk may require lifelong screening
mune-related hypophysitis and various HP deficits for various adverse endocrine outcomes. Changes and
(mostly central hypothyroidism and central adrenal in- improvements in cancer treatments necessitate a sus-
sufficiency); these were reported to be permanent in up tained effort in the investigation of long-term endocrine
to 50% of cases [58]. Patients treated with immunomodu- and general health outcomes in this population.
lators such as interferon and immune check point inhib-
itors including ipilimumab, nivolumab, tremelimumab,
and pembrolizumab may develop autoimmune thyroid Acknowledgment
disease with subsequent hypothyroidism or, more rarely
hyperthyroidism [58]. Wassim Chemaitilly, MD and Charles A. Sklar MD contrib-
uted to the drafting and review of the manuscript.

Others
Children treated with 13-cis-retinoic acid for neuro- Disclosure Statement
blastoma may have irreversible damage to their growth
plates and are at risk of significantly impaired final adult The authors have no conflicts of interest to disclose.
height [59]. Hedgehog pathway inhibitors, used in clini-

References
  1 Robison LL, Hudson MM. Survivors of child-  4 Chemaitilly W, Armstrong GT, Gajjar A,   7 Laughton SJ, Merchant TE, Sklar CA, Kun LE,
hood and adolescent cancer: life-long risks Hudson MM. Hypothalamic-Pituitary Axis Fouladi M, Broniscer A, et al. Endocrine out-
and responsibilities. Nat Rev Cancer. 2014 Dysfunction in Survivors of Childhood CNS comes for children with embryonal brain tu-
Jan;14(1):61–70. Tumors: Importance of Systematic Follow- mors after risk-adapted craniospinal and con-
 2 Hudson MM, Ness KK, Gurney JG, Mul- Up and Early Endocrine Consultation. J Clin formal primary-site irradiation and high-
rooney DA, Chemaitilly W, Krull KR, et al. Oncol. 2016 Dec;34(36):4315–9. dose chemotherapy with stem-cell rescue on
Clinical ascertainment of health outcomes  5 Merchant TE, Rose SR, Bosley C, Wu S, the SJMB-96 trial. J Clin Oncol. 2008 Mar;
among adults treated for childhood cancer. Xiong X, Lustig RH. Growth hormone se- 26(7):1112–8.
JAMA. 2013 Jun;309(22):2371–81. cretion after conformal radiation therapy in  8 Thomas BC, Stanhope R, Plowman PN,
 3 Brignardello E, Felicetti F, Castiglione A, pediatric patients with localized brain tu- Leiper AD. Growth following single fraction
Chiabotto P, Corrias A, Fagioli F, et al. Endo- mors. J Clin Oncol. 2011 Dec; 29(36): 4776– and fractionated total body irradiation for
crine health conditions in adult survivors of 80. bone marrow transplantation. Eur J Pediatr.
childhood cancer: the need for specialized  6 Sklar C, Whitton J, Mertens A, Stovall M, 1993 Nov;152(11):888–92.
adult-focused follow-up clinics. Eur J Endo- Green D, Marina N, et al. Abnormalities of   9 Fukunaga-Johnson N, Sandler HM, Marsh R,
crinol. 2013 Feb;168(3):465–72. the thyroid in survivors of Hodgkin’s disease: Martel MK. The use of 3D conformal radio-
data from the Childhood Cancer Survivor therapy (3D CRT) to spare the cochlea in pa-
Study. J Clin Endocrinol Metab. 2000 Sep; tients with medulloblastoma. Int J Radiat On-
85(9):3227–32. col Biol Phys. 1998 Apr;41(1):77–82.
130.237.165.40 - 11/14/2018 1:17:10 PM

Childhood Cancer Treatments and Horm Res Paediatr 7


Stockholm University Library

Associated Endocrine Late Effects DOI: 10.1159/000493943


Downloaded by:
10 Pai Panandiker A, Ning H, Likhacheva A, Ull- 22 Mazonakis M, Tzedakis A, Damilakis J, Varveris 35 Sklar CA, Robison LL, Nesbit ME, Sather HN,
man K, Arora B, Ondos J, et al. Craniospinal H, Kachris S, Gourtsoyiannis N. Scattered dose Meadows AT, Ortega JA, et al. Effects of ra-
irradiation with spinal IMRT to improve tar- to thyroid from prophylactic cranial irradiation diation on testicular function in long-term
get homogeneity. Int J Radiat Oncol Biol during childhood: a Monte Carlo study. Phys survivors of childhood acute lymphoblastic
Phys. 2007 Aug;68(5):1402–9. Med Biol. 2006 Apr;51(8):N139–45. leukemia: a report from the Children Cancer
11 Merchant TE, Farr JB. Proton beam therapy: 23 Tucker MA, Jones PH, Boice JD Jr, Robison Study Group. J Clin Oncol. 1990 Dec; 8(12):
a fad or a new standard of care. Curr Opin LL, Stone BJ, Stovall M, et al.; The Late Effects 1981–7.
Pediatr. 2014 Feb;26(1):3–8. Study Group. Therapeutic radiation at a 36 Mauz-Körholz C, Metzger ML, Kelly KM,
12 Bishop AJ, Greenfield B, Mahajan A, Paulino young age is linked to secondary thyroid can- Schwartz CL, Castellanos ME, Dieckmann K,
AC, Okcu MF, Allen PK, et al. Proton beam cer. Cancer Res. 1991 Jun;51(11):2885–8. et al. Pediatric Hodgkin Lymphoma. J Clin
therapy versus conformal photon radiation 24 Clayton PE, Shalet SM. The evolution of spi- Oncol. 2015 Sep;33(27):2975–85.
therapy for childhood craniopharyngioma: nal growth after irradiation. Clin Oncol (R 37 Punyko JA, Mertens AC, Gurney JG, Yasui Y,
multi-institutional analysis of outcomes, cyst Coll Radiol). 1991 Jul;3(4):220–2. Donaldson SS, Rodeberg DA, et al. Long-term
dynamics, and toxicity. Int J Radiat Oncol 25 Schmiegelow M, Feldt-Rasmussen U, Ras- medical effects of childhood and adolescent
Biol Phys. 2014 Oct;90(2):354–61. mussen AK, Poulsen HS, Müller J. A popula- rhabdomyosarcoma: a report from the child-
13 Eaton BR, Esiashvili N, Kim S, Patterson B, tion-based study of thyroid function after ra- hood cancer survivor study. Pediatr Blood
Weyman EA, Thornton LT, et al. Endocrine diotherapy and chemotherapy for a child- Cancer. 2005 Jun;44(7):643–53.
outcomes with proton and photon radiother- hood brain tumor. J Clin Endocrinol Metab. 38 Clement SC, Schoot RA, Slater O, Chisholm
apy for standard risk medulloblastoma. Neu- 2003 Jan;88(1):136–40. JC, Abela C, Balm AJ, et al. Endocrine disor-
ro-oncol. 2016 Jun;18(6):881–7. 26 Ahmed SR, Shalet SM, Campbell RH, Deakin ders among long-term survivors of childhood
14 Gajjar A, Packer RJ, Foreman NK, Cohen K, DP. Primary gonadal damage following treat- head and neck rhabdomyosarcoma. Eur J
Haas-Kogan D, Merchant TE; COG Brain ment of brain tumors in childhood. J Pediatr. Cancer. 2016 Feb;54:1–10.
Tumor Committee. Children’s Oncology 1983 Oct;103(4):562–5. 39 Mostoufi-Moab S, Seidel K, Leisenring WM,
Group’s 2013 blueprint for research: central 27 Chemaitilly W, Boulad F, Heller G, Kernan Armstrong GT, Oeffinger KC, Stovall M, et al.
nervous system tumors. Pediatr Blood Can- NA, Small TN, O’Reilly RJ, et al. Final height Endocrine Abnormalities in Aging Survivors
cer. 2013 Jun;60(6):1022–6. in pediatric patients after hyperfractionated of Childhood Cancer: A Report From the
15 Pui CH, Robison LL, Look AT. Acute lym- total body irradiation and stem cell transplan- Childhood Cancer Survivor Study. J Clin On-
phoblastic leukaemia. Lancet. 2008 Mar; tation. Bone Marrow Transplant. 2007 Jul; col. 2016 Sep;34(27):3240–7.
371(9617):1030–43. 40(1):29–35. 40 Papadakis V, Vlachopapadopoulou E, Van
16 Shalitin S, Laur E, Lebenthal Y, Ash S, Yaniv 28 Barfield RC, Kasow KA, Hale GA. Advances Syckle K, Ganshaw L, Kalmanti M, Tan C, et
I, Phillip M. Endocrine complications and in pediatric hematopoietic stem cell trans- al. Gonadal function in young patients suc-
components of the metabolic syndrome in plantation. Cancer Biol Ther. 2008 Oct;7(10): cessfully treated for Hodgkin disease. Med
survivors of childhood malignant non-brain 1533–9. Pediatr Oncol. 1999 May;32(5):366–72.
solid tumors. Horm Res Paediatr. 2014;81(1): 29 Clement-De Boers A, Oostdijk W, Van Weel- 41 Chemaitilly W, Li Z, Krasin MJ, Brooke RJ,
32–42. Sipman MH, Van den Broeck J, Wit JM, Vos- Wilson CL, Green DM, et al. Premature Ovar-
17 Clement SC, Schouten-van Meeteren AY, sen JM. Final height and hormonal function ian Insufficiency in Childhood Cancer Survi-
Boot AM, Claahsen-van der Grinten HL, after bone marrow transplantation in chil- vors: A Report From the St. Jude Lifetime
Granzen B, Sen Han K, et al. Prevalence and dren. J Pediatr. 1996 Oct;129(4):544–50. Cohort. J Clin Endocrinol Metab. 2017 Jul;
Risk Factors of Early Endocrine Disorders in 30 Boulad F, Bromley M, Black P, Heller G, Sara- 102(7):2242–50.
Childhood Brain Tumor Survivors: A Nation- foglou K, Gillio A, et al. Thyroid dysfunction 42 Sklar CA, Mertens AC, Mitby P, Whitton J,
wide, Multicenter Study. J Clin Oncol. 2016 following bone marrow transplantation using Stovall M, Kasper C, et al. Premature meno-
Dec;34(36):4362–70. hyperfractionated radiation. Bone Marrow pause in survivors of childhood cancer: a re-
18 Constine LS, Woolf PD, Cann D, Mick G, Mc- Transplant. 1995 Jan;15(1):71–6. port from the childhood cancer survivor
Cormick K, Raubertas RF, et al. Hypothalam- 31 Cohen A, Rovelli A, Merlo DF, van Lint MT, study. J Natl Cancer Inst. 2006 Jul; 98(13):
ic-pituitary dysfunction after radiation for Lanino E, Bresters D, et al. Risk for secondary 890–6.
brain tumors. N Engl J Med. 1993 Jan;328(2): thyroid carcinoma after hematopoietic stem- 43 Trahair TN, Vowels MR, Johnston K, Cohn
87–94. cell transplantation: an EBMT Late Effects RJ, Russell SJ, Neville KA, et al. Long-term
19 Chemaitilly W, Li Z, Huang S, Ness KK, Clark Working Party Study. J Clin Oncol. 2007 Jun; outcomes in children with high-risk neuro-
KL, Green DM, et al. Anterior hypopituita- 25(17):2449–54. blastoma treated with autologous stem cell
rism in adult survivors of childhood cancers 32 Sarafoglou K, Boulad F, Gillio A, Sklar C. Go- transplantation. Bone Marrow Transplant.
treated with cranial radiotherapy: a report nadal function after bone marrow transplan- 2007 Oct;40(8):741–6.
from the St Jude Lifetime Cohort study. J Clin tation for acute leukemia during childhood. J 44 Heikens J, Behrendt H, Adriaanse R, Bergh-
Oncol. 2015 Feb;33(5):492–500. Pediatr. 1997 Feb;130(2):210–6. out A. Irreversible gonadal damage in male
20 Chemaitilly W, Merchant TE, Li Z, Barnes N, 33 Rovó A, Tichelli A, Passweg JR, Heim D, Mey- survivors of pediatric Hodgkin’s disease. Can-
Armstrong GT, Ness KK, et al. Central Preco- er-Monard S, Holzgreve W, et al. Spermato- cer. 1996 Nov;78(9):2020–4.
cious Puberty following the Diagnosis and genesis in long-term survivors after allogene- 45 Meacham LR, Sklar CA, Li S, Liu Q, Gimpel
Treatment of Paediatric Cancer and Central ic hematopoietic stem cell transplantation is N, Yasui Y, et al. Diabetes mellitus in long-
Nervous System Tumours: Presentation and associated with age, time interval since trans- term survivors of childhood cancer. Increased
Long-term Outcomes. Clin Endocrinol (Oxf). plantation, and apparently absence of chronic risk associated with radiation therapy: a re-
2016 Mar;84(3):361-71. GvHD. Blood. 2006 Aug;108(3):1100–5. port for the childhood cancer survivor study.
21 Chow EJ, Friedman DL, Stovall M, Yasui Y, 34 Ishiguro H, Yasuda Y, Tomita Y, Shinagawa Arch Intern Med. 2009 Aug;169(15):1381–8.
Whitton JA, Robison LL, et al. Risk of thyroid T, Shimizu T, Morimoto T, et al. Gonadal 46 van Santen HM, de Kraker J, Vulsma T. En-
dysfunction and subsequent thyroid cancer shielding to irradiation is effective in protect- docrine late effects from multi-modality
among survivors of acute lymphoblastic leu- ing testicular growth and function in long- treatment of neuroblastoma. Eur J Cancer.
kemia: a report from the Childhood Cancer term survivors of bone marrow transplanta- 2005 Aug;41(12):1767–74.
Survivor Study. Pediatr Blood Cancer. 2009 tion during childhood or adolescence. Bone
Sep;53(3):432–7. Marrow Transplant. 2007 Apr;39(8):483–90.
130.237.165.40 - 11/14/2018 1:17:10 PM

8 Horm Res Paediatr Chemaitilly/Sklar


Stockholm University Library

DOI: 10.1159/000493943
Downloaded by:
47 van Santen HM, Tytgat GA, van de Wetering 51 Green DM, Liu W, Kutteh WH, Ke RW, Shel- 56 Samis J, Lee P, Zimmerman D, Arceci RJ, Sut-
MD, van Eck-Smit BL, Hopman SM, van der ton KC, Sklar CA, et al. Cumulative alkylating torp M, Hijiya N. Recognizing Endocrinopa-
Steeg AF, et al. Differentiated thyroid carci- agent exposure and semen parameters in thies Associated With Tyrosine Kinase Inhib-
noma after 131I-MIBG treatment for neuro- adult survivors of childhood cancer: a report itor Therapy in Children With Chronic My-
blastoma during childhood: description of the from the St Jude Lifetime Cohort Study. Lan- elogenous Leukemia. Pediatr Blood Cancer.
first two cases. Thyroid. 2012 Jun;22(6):643– cet Oncol. 2014 Oct;15(11):1215–23. 2016 Aug;63(8):1332–8.
6. 52 Howell SJ, Radford JA, Ryder WD, Shalet SM. 57 Lodish MB. Clinical review: kinase inhibitors:
48 Clement SC, Kraal KC, van Eck-Smit BL, van Testicular function after cytotoxic chemo- adverse effects related to the endocrine sys-
den Bos C, Kremer LC, Tytgat GA, et al. Pri- therapy: evidence of Leydig cell insufficiency. tem. J Clin Endocrinol Metab. 2013 Apr;
mary ovarian insufficiency in children after J Clin Oncol. 1999 May;17(5):1493–8. 98(4):1333–42.
treatment with 131I-metaiodobenzylguani- 53 Mostoufi-Moab S, Halton J. Bone morbidity 58 Corsello SM, Barnabei A, Marchetti P, De
dine for neuroblastoma: report of the first two in childhood leukemia: epidemiology, mech- Vecchis L, Salvatori R, Torino F. Endocrine
cases. J Clin Endocrinol Metab. 2014 Jan; anisms, diagnosis, and treatment. Curr Os- side effects induced by immune checkpoint
99(1):E112–6. teoporos Rep. 2014 Sep;12(3):300–12. inhibitors. J Clin Endocrinol Metab. 2013
49 Metzger ML, Meacham LR, Patterson B, 54 Zhang FF, Kelly MJ, Saltzman E, Must A, Apr;98(4):1361–75.
Casillas JS, Constine LS, Hijiya N, et al. Fe- Roberts SB, Parsons SK. Obesity in pediatric 59 Cohen LE, Gordon JH, Popovsky EY, Gu-
male reproductive health after childhood, ad- ALL survivors: a meta-analysis. Pediatrics. nawardene S, Duffey-Lind E, Lehmann LE, et
olescent, and young adult cancers: guidelines 2014 Mar;133(3):e704–15. al. Late effects in children treated with inten-
for the assessment and management of female 55 Narayanan KR, Bansal D, Walia R, Sachdeva sive multimodal therapy for high-risk neuro-
reproductive complications. J Clin Oncol. N, Bhansali A, Varma N, et al. Growth failure blastoma: high incidence of endocrine and
2013 Mar;31(9):1239–47. in children with chronic myeloid leukemia re- growth problems. Bone Marrow Transplant.
50 Kenney LB, Cohen LE, Shnorhavorian M, ceiving imatinib is due to disruption of GH/ 2014 Apr;49(4):502–8.
Metzger ML, Lockart B, Hijiya N, et al. Male IGF-1 axis. Pediatr Blood Cancer. 2013 Jul; 60 Robinson GW, Kaste SC, Chemaitilly W,
reproductive health after childhood, adoles- 60(7):1148–53. Bowers DC, Laughton S, Smith A, et al. Irre-
cent, and young adult cancers: a report from versible growth plate fusions in children with
the Children’s Oncology Group. J Clin Oncol. medulloblastoma treated with a targeted
2012 Sep;30(27):3408–16. hedgehog pathway inhibitor. Oncotarget.
2017 Sep;8(41):69295–302.

130.237.165.40 - 11/14/2018 1:17:10 PM

Childhood Cancer Treatments and Horm Res Paediatr 9


Stockholm University Library

Associated Endocrine Late Effects DOI: 10.1159/000493943


Downloaded by:

You might also like