Buscar en
Endocrinología, Diabetes y Nutrición (English ed.)
Toda la web
Inicio Endocrinología, Diabetes y Nutrición (English ed.) Endocrine sequelae in childhood cancer survivors
Journal Information
Vol. 64. Issue 9.
Pages 498-505 (November 2017)
Visits
2603
Vol. 64. Issue 9.
Pages 498-505 (November 2017)
Review article
Full text access
Endocrine sequelae in childhood cancer survivors
Secuelas endocrinológicas en supervivientes de cáncer infantil
Visits
2603
Paula Casano Sanchoa,b,c
a Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Universidad de Barcelona, Esplugues, Barcelona, Spain
b Unidad de Seguimiento del Superviviente de Cáncer Infantil, Servicio de Endocrinología Pediátrica, Hospital Sant Joan de Déu, Esplugues, Barcelona, Spain
c CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (2)
Table 1. Spanish Pediatric Tumor Registry (Registro Español de Tumores Infantiles [RETI-SEHOP]). Cases recorded according to diagnostic group, incident year cohorts, and microscopic verification (MV).
Table 2. Endocrine sequelae in childhood cancer survivors: risk according to treatments received.
Show moreShow less
Abstract

Thanks to the advances in cancer treatment, the five-year survival rate after childhood cancer has increased up to 80%. Therefore 1/500 young adults will be a survivor. Endocrine sequelae are most common, affecting 40–60% of survivors. The most frequent sequelae include growth failure and gonadal and thyroid diseases. Sequelae occur more frequently in survivors from central nervous system tumors, leukemia, and lymphoma. Their development will depend on the type of cancer, its location, age at diagnosis, and treatment administered. Treatments associated to more endocrine sequels are cranial radiotherapy and hematopoietic cell transplantation. Because of the high prevalence of endocrine sequelae, international guidelines recommend endocrinologists to prospectively evaluate the survivors. As some of these endocrine changes will not develop until adult life, transition programs should be implemented, and active investigation should be made to decrease the endocrine consequences of cancer treatment.

Keywords:
Endocrine sequelae
Childhood cancer survivor
Hypopituitarism
Gonadal dysfunction
Resumen

La evolución en los tratamientos oncológicos ha supuesto un aumento de la supervivencia del cáncer infantil cercana al 80% a 5 años, por lo que 1/500 adultos jóvenes será un superviviente. Las secuelas endocrinas son las más comunes y afectan al 40–60%, siendo las más frecuentes las alteraciones del crecimiento y la disfunción gonadal y tiroidea. Los pacientes con tumores del sistema nervioso central, leucemias y linfomas son los que presentan más secuelas, y estas dependen del tipo de cáncer, su localización, la edad de diagnóstico y el protocolo de tratamiento; las terapias de mayor riesgo son la radioterapia craneal y el trasplante de progenitores hematopoyéticos. Dado este elevado riesgo, las guías internacionales recomiendan a los endocrinólogos evaluar prospectivamente a los supervivientes. Algunas de las alteraciones endocrinas no se manifestarán hasta la vida adulta, por lo que debemos crear programas de transición, así como ser activos en la investigación para reducir las secuelas endocrinas de los tratamientos del cáncer.

Palabras clave:
Secuelas endocrinas
Superviviente cáncer infantil
Hipopituitarismo
Disfunción gonadal
Full Text
Introduction

The evolution of cancer treatments has resulted in improvements in the 5-year survival rate, reaching 80% for all cancers globally, and 90% in the case of leukemias and lymphomas. Approximately one out of every 285 children is diagnosed with cancer, and one out of every 530 young adults is a cancer survivor.1 According to the Spanish Pediatric Tumor Registry (Registro Español de Tumores Pediátricos),2 a total of 25,100 cases of malignant disease were recorded in patients between 0 and 14 years of age in the period between 1980 and 2015. This gives us an idea of the number of young adults in this country that survive childhood cancer. It has also been reported that 62% of all adult survivors experience some medical sequelae, and that 40–60% suffer endocrine disease. This implies a risk far greater than that found in the general population.3,4

The present review offers a global view of the follow-up of pediatric cancer survivors, focusing on current knowledge regarding endocrine sequelae, with the aim of optimizing follow-up and improving long-term survival.

New cancer treatment perspectives in children and adolescents

The most common cancers in the pediatric population are leukemias, lymphomas and tumors of the central nervous system (CNS) (Table 1). The sequelae among the survivors depend on the type of cancer, its location, patient age at the time of diagnosis, and the treatment protocol used (radiotherapy fractionation and dose; type and cumulative dose of chemotherapy). The changes in leukemia management include the identification of risk factors, the intensification of treatment and improvements in supportive measures, a lesser use of prophylactic radiotherapy, and the introduction of hematopoietic progenitor cell transplantation (HPCT) in cases of relapse, all of which have contributed to improve patient survival. In some diseases such as Hodgkin's lymphoma or Wilms tumor, where the survival rates are high, the current aim is to reduce their sequelae. Other diseases such as malignant astrocytomas or metastatic sarcomas continue to have a poor prognosis, and the aim in such cases is to develop new therapies capable of improving survival.5,6

Table 1.

Spanish Pediatric Tumor Registry (Registro Español de Tumores Infantiles [RETI-SEHOP]). Cases recorded according to diagnostic group, incident year cohorts, and microscopic verification (MV).

Diagnostic groups  Incident year cohortTotal 
  80–84  85–89  90–94  95–99  00–04  05–09  10–14  2015   
I Leukemias and myeloproliferative and myelodysplastic diseases  507  714  805  767  888  1236  1451  275  6643 
II Lymphomas and reticuloendothelial neoplasms  340  396  435  390  473  544  596  118  3292 
III CNS  421  527  574  660  686  1011  1188  204  5271 
IV SNS  260  340  298  316  361  400  435  80  2490 
V Retinoblastomas  67  92  66  101  104  144  166  37  777 
VI Renal  213  176  173  178  166  266  263  58  1493 
VII Hepatic  36    41  37  42  69  65  16  336 
VIII Bone  122  193  264  251  234  292  299  62  1717 
IX Soft tissue sarcomas  185  230  227  232  228  272  311  53  1738 
X Germinal cell, trophoblastic and gonadal cell tumors  60  89  86  103  110  155  162  28  793 
XI Other epithelial tumors and melanomas  38  46  58  79  59  88  94  22  484 
XII Others and not specified  18  11  66 
All tumors  2258  2851  3035  3122  3358  4482  5041  953  25,100 

0–14 years, 1980–2015. Excluded, not classifiable in ICCC-3.

Cases excluded because not classifiable in ICCC-3: 2040.

Total recorded cases, including those not classifiable in ICCC-3: 27,140.

Source: reproduced with permission from Peris et al.2
Long-term sequelae in childhood cancer survivors

The review conducted by the Childhood Oncology Group (COG) revealed shortcomings in survivor control: 68% of the patients treated between 1970 and 1986 had not undergone regular controls, and 39% had had no contact with the center that had treated them.3 The different cohorts studied to date involve different methodological approaches, though they report a comparable risk of medical problems.7 In order to adequately estimate excess risk, some cohorts such as the Childhood Cancer Survivor Study or the Adult Life after Childhood Cancer in Scandinavia survey have compared the incidence with a cohort of siblings of the survivors or with a comparable healthy population.4,8 As a result of these studies, in 2003 the COG published a clinical guide (COG-LTFU Guidelines) with concrete recommendations regarding the follow-up of survivors, and with a specific section addressing endocrine sequelae.9

In this regard, endocrine problems are among the most frequently described sequelae, along with cardiovascular disease. Survivors of tumors of the CNS, leukemias and Hodgkin's lymphoma are the patients that experience most sequelae. The Adult Life after Childhood Cancer in Scandinavia cohort presented a relative risk (RR) for endocrine disease of 4.8 (95%CI 4.6–5.0), with higher values among those under 20 years of age. The Childhood Cancer Survivor Study in turn described the relative risks regarding hypothyroidism (RR 14.3; 95%CI 9.7–21.0), growth hormone (GH) deficiency (RR=277.8; 95%CI 111.1–694.9), the need for the induction of puberty (RR=86.1; 95%CI 31.1–238.2) and osteoporosis (RR=24.7; 95%CI 9.9–61.4). Endocrine disorders moreover have implications in terms of long-term cardiometabolic morbidity in the adult, which affects 18% of all survivors.3,4 Few studies have described the prevalence of endocrine disease with prospective application of the COG-LTFU guidelines. One of the largest series describes 519 patients with non-CNS tumors evaluated an average of 7.2 years after diagnosis, with a mean patient age of 12.1 years. Endocrine sequelae were present in 57.6% of the cases.10 Following the introduction of the cancer survivor follow-up unit at our center, we reviewed the endocrine sequelae of 194 patients (including tumors of the CNS) with a mean age of 10 years, 5.2 years after treatment. Sixty-three percent of the patients had some endocrine disorder. Furthermore, on their first visit almost 30% of the survivors presented endocrine-metabolic disease not previously diagnosed and/or treated.11

Endocrine sequelae in childhood cancer survivors

A description of the endocrine sequelae is provided below. In order to facilitate description, the sequelae are analyzed according to the reported risk corresponding to the different treatments received (Table 2).12

Table 2.

Endocrine sequelae in childhood cancer survivors: risk according to treatments received.

  GH deficiency  Thyroid  Central adrenal insufficiency  Early puberty  Hypogonadism  Infertility  BMD  Obesity  Metabolic syndrome 
Cranial radiotherapya  √  Hypothyroidism
Nodules/cancer 
√  √  √  √    √  √ 
TBI π  √  Hypothyroidism
Nodules/cancer 
    √  √      √ 
Thyroid irradiationb    Hypothyroidism
Hyperthyroidism
Nodules/cancer 
             
Gonadal
irradiationc 
        √  √       
Alkylating agents          √  √       
Heavy metals          √  √    √Dyslipidemia   
Antimetabolites              √     
Corticosteroids
Hematopoietic progenitor cell transplantation 
            √     
New therapeutic targets (protein kinase inhibitors, immune modulators)  √  √  √        √     

BMD, bone mineral density; TBI, total body irradiation.

a

Cranial radiotherapy includes: cranial, orbital/ocular, ear/infratemporal and nasopharyngeal.

b

Thyroid irradiation includes: thyroid gland, neck, cervical spine, oropharyngeal, supraclavicular, mantle and mini-mantle.

c

Gonadal irradiation includes: lumbosacral spine, abdomen and pelvis (females), pelvis and testicles (males).

Adapted from: Nandagopal et al.12
Cranial radiotherapy

Cranial radiotherapy (CRT) produces a significant increase in the risk of sequelae secondary to hypothalamic–hypophyseal involvement. The impact depends on the dose, fractionation and the irradiation field. The degree of involvement has been correlated to the so-called biological effective dose, which allows for the comparison of therapeutic schemes and prediction of the impact upon hypophyseal function.13

According to some studies, patient age is another important factor, younger age being associated with greater hypophyseal involvement. Furthermore, the risk of hypophyseal dysfunction increases over time; follow-up therefore should be prolonged for several years. It has been suggested that the use of other techniques such as proton radiotherapy could reduce the risk of sequelae, though further studies are needed to determine whether they can reduce the long-term endocrine sequelae.14

Somatotropic axis (GH)

The most frequent and earliest manifesting type of hypophyseal involvement refers to GH. Alterations have been recorded after doses of 18Gy, and have been seen from as early as the first 12 months after treatment, likewise in relation to hypothalamic damage.15,16 In patients with a history of tumor disease of the CNS and CRT, a pathological test result is considered sufficient to establish the diagnosis.17 Studies have been made to determine whether there is an increased risk of recurrence in patients treated with recombinant growth hormone (rhGH), though the data do not appear to confirm this. Survivors are at an increased risk of suffering a second neoplasm independently of whether or not rhGH treatment is provided. Although some studies had reported an increased risk of second neoplasms (mainly meningiomas and gliomas), subsequent analyses appeared to correlate this risk to previous CRT.18,19 Despite the lack of studies, the consensus documents recommend a one year waiting period after the completion of cancer treatment, in order to ensure that there is no early recurrence.20

The protocolized follow-up of survivors has made it possible to optimize treatment with rhGH, with the observation of improvements in final body height, probably due to the early start of treatment, new therapeutic schemes, and the use of GnRH analogs in cases of early puberty.21 According to the literature, in only 60% of the adults treated with rhGH after childhood cancer is GH deficiency re-evaluated in the transition to adulthood.22 As has been pointed out by some authors, despite the demonstrated benefits of rhGH treatment in adults, the evidence of such benefits in the case of survivors may be limited by the negative effect of the treatments received. In this regard, prospective studies need to be expanded with endocrine-metabolic and quality of life assessments in adult life.23,24

Corticotropic axis

Adrenocorticotropic hormone (ACTH) deficiency is less frequent and manifests later in time. Follow-up periods of over 10 years are needed to detect deficiencies in a significant group of patients in the studied cohorts. In view of the characteristics of oncological patients, the use of insulin hypoglycemia tests is restricted, and this complicates full evaluation of the hypothalamic–pituitary–adrenal axis. However, some authors have reported an acceptable correlation between insulin hypoglycemia testing and glucagon stimulation; the latter therefore could be used as an alternative.25 Involvement of the corticotropic axis over the long term has been described in 19% of all patients.26

Thyrotropic axis

The hypothalamic–pituitary–thyroid axis appears to be the least affected axis. Nevertheless, according to some authors the 6% incidence of central hypothyroidism reported in some studies may be due to underestimation in cases characterized by normal free T4 levels. They consequently recommend complementing such assays with the study of nocturnal thyroid stimulating hormone (TSH) elevation and the thyrotropin releasing hormone (TRH) test, which together yield a 36% prevalence of such alterations.27

Gonadotropic axis

Cranial radiotherapy at doses of over 18Gy predisposes to early puberty. The female gender or irradiation at a younger age increases the risk. Doses of 40Gy can result in delayed puberty or central hypogonadism in 11–15% of the patients.28

Hyperprolactinemia

High doses (>40Gy) are likewise associated with hyperprolactinemia; prolactin measurement is therefore advised in the presence of suggestive clinical manifestations.

ChemotherapyGonadal involvement

Gonadal dysfunction is the main side effect of chemotherapy. High gonadotoxic potential fundamentally corresponds to the alkylating agents (mainly cyclophosphamide at doses of ≥7.5g/m2, ifosfamide ≥60mg/m2, the mechlorethamine, vincristine, procarbazine and prednisone (MOPP) protocol in ≥3 cycles or busulfan ≥600mg/m2).29 This risk is increased on adding radiotherapy at the gonadal or pelvic level, or total body irradiation (TBI). There appear to be no differences in the case of patients treated in prepubertal or pubertal stages.30 The testicles are particularly sensitive to the action of the alkylating agents. Specifically, the Sertoli cells and spermatogenesis are more often affected than the Leydig cells, which usually preserve their function. It has been reported that 70% of all patients treated for Hodgkin's lymphoma with the MOPP protocol, without radiotherapy, present azoospermia or oligospermia. This protocol has therefore been replaced by new and less toxic combinations.31 The evaluation of spermatogenesis therefore should be included in the follow-up programs. New biochemical markers such as inhibin B appear to have a closer correlation to spermatogenesis than follicle-stimulating hormone (FSH).32 Nevertheless, according to some authors the specificity of both markers is low. They consequently recommend testicular measurement and a seminogram among the survivors.33

The ovaries appear to be less sensitive than testicular tissue, though the risk increases on adding radiotherapy and/or hematopoietic progenitor cell transplantation (HPCT). Ovarian involvement can affect puberty, with an increased risk of premature ovarian failure (POF) or infertility over the long term. The cumulative incidence of POF among survivors is 8% at 40 years of age.34 No studies have compared the different therapeutic schemes with respect to the individual risk of each drug. It was generally believed that the ovaries of patients under 10 years of age are more resistant to the effects of chemotherapy. However, some authors have found the risk to be no different between patients treated before or after puberty.35,36 Even in the presence of regular menstrual cycles, survivors are at risk of lesser ovarian reserve and therefore may have a shorter fertile period. In addition to the gonadotropins, new promising markers such as anti-Müllerian hormone have been proposed. Patients treated with the MOPP protocol show a decrease in anti-Müllerian hormone with respect to other cancer survivors.37 However, the correlation of this observation to future fertility/fecundity has not been clearly established, and further studies are needed in this regard. The usefulness of the antral follicle count via transvaginal ultrasound has not been firmly established in childhood cancer survivors, and prospective studies need to be carried out in this respect.38

Growth

Different studies in leukemia patients have described a decrease in growth during chemotherapy and a lack of recovering growth until the end of intensive treatment. A discrete alteration in final body height (decrease of −0.59 standard deviations [SD]) has been observed in series of leukemia patients treated without radiotherapy. This observation is not correlated to alterations in the GH tests, since GH deficiency has been described in only 0.9–1.2% of the cases. Its systematic screening is therefore not advised.39

Neck radiotherapy

The use of neck radiotherapy, mainly in cases of lymphomas, implies an increased risk of thyroid gland involvement. Hypothyroidism has been described at doses above 10Gy, with less frequent cases of hyperthyroidism. The reported incidence of hypothyroidism reaches 75% in patients with lymphomas. In addition, there is an increased risk of thyroid nodules and/or thyroid cancer at doses of 20–29Gy. This risk decreases with doses of over 30Gy, in accord with their cell destructing effect. The risk is greater in children under 5 years of age. Latency periods of 94 months have been described for the development of hypothyroidism, and of up to 10–20 years for the appearance of nodules. This indicates that long-term follow-up is needed.40,41 The follow-up of these patients is currently recommended due to the risk of thyroid cancer, but there is no consensus regarding the screening technique to be used. A recent literature review has stated that the detection of early stage papillary thyroid cancer in children is associated with lesser recurrence and mortality. However, in contrast to the situation found in adults, there is no evidence of lesser morbidity over the long term.12,42–45

Abdominal radiotherapy

The testicles are very sensitive to radiotherapy; in adults, alterations have been described at doses of 0.2–0.7Gy, with increased FSH and seminogram alterations.24 Azoospermia has been reported at doses of 6–10Gy, and Leydig cell insufficiency at ≥20Gy.46 Although scarce, the series of survivors treated with chemotherapy and radiotherapy at doses of 30–45Gy in childhood have shown 83% to suffer azoospermia. However, 17% are able to recover spermatogenesis 12–15 years after treatment; the authors therefore recommend serial controls.25 In the case of the ovaries, it has been reported that close to 40% of the patients with Hodgkin's lymphoma subjected to radiotherapy suffer POF. In these cases the recovery of gonadal function is exceptional. In addition, irradiated patients have been reported to suffer an increased risk of miscarriage, low birth weight and postpartum bleeding.47

Hematopoietic progenitor cell transplantation

Endocrine alterations are the most common sequelae following HPCT. Such alterations are of a multifactorial origin, secondary to rescue therapy added to the cumulative effects of chemotherapy, high-dose corticosteroids and the use of total body irradiation. These patients have a two- to three-fold increased risk of endocrine disease.4 It is very common to observe hypogonadism (83%), hypothyroidism (56%) and GH deficiency (50%). This frequency is even higher than that seen in patients subjected to prophylactic cranial radiotherapy, possibly because of lesser dose fractionation. HPCT is moreover a risk factor for growth both in patients with and without radiotherapeutic conditioning regimens. On the other hand, HPCT predisposes not only to overweight, but also to increased central adiposity and metabolic risk.48

New therapeutic targets

Novel therapies have also been introduced in recent years in pediatric oncology, though their impact in terms of endocrine sequelae requires further studies.49,50 Protein kinase inhibitors, interferon-alpha and monoclonal antibodies have been related to thyroid gland alterations (thyroiditis and hypothyroidism).51 Alterations of the corticotropic axis have been described (sometimes of a subclinical nature) with the use of protein kinase inhibitors; systematic controls of this axis are therefore advised.52 Ipilimumab has been reported to cause hypophysitis, resulting in panhypopituitarism.53 Decelerated growth has been described in patients treated with imatinib, this problem being more severe in smaller children.54 Likewise, there have been reports of calcium–phosphate metabolic alterations in adults treated with protein kinase inhibitors.55

Combination therapies—multifactorial sequelaeCalcium–phosphate metabolism

A decrease in bone mineral density (BMD) has been observed in childhood cancer survivors, the underlying cause being of a multifactorial nature. An increased risk has been associated with treatment with hematopoietic progenitor cells, methotrexate or prolonged corticosteroid therapy, and with older patients. Since the best approach for measuring BMD in pediatric patients is subject to debate, the screening method of choice is controversial. The COG-LTFU guidelines recommend densitometry or quantitative computed axial tomography in high risk patients upon admission to the follow-up program, and the repetition of such measures if clinically indicated. The Z-score associated with increased fracture risk in the pediatric population likewise has not been defined. As a result, there is no consensus regarding the values indicating the treatment in such cases.4 In terms of efficacy in reducing fractures over the long term, some authors recommend the promotion of physical activity, with the optimization of calcium intake and vitamin D levels among the survivors.56,57

Obesity

Some authors do not include obesity among the sequelae, due to its multifactorial nature and the difficulty of establishing a cause–effect relationship with the treatments. The prevalence of obesity is variable; some studies document an overweight/obesity rate of 25%/18%, which is even lower than that of the general pediatric population in the same geographical setting. The risk is greater in patients subjected to cranial radiotherapy at doses of >20Gy, in women, and in irradiated patients <4 years of age. However, an increase in metabolic risk has been observed, 26% of the patients presenting insulin resistance and 23% suffering from dyslipidemia in relation to an increase in abdominal adiposity or certain therapeutic modalities such as HPCT and/or total body irradiation.58,59

Conclusion

In the last decade progress has been made in improving the health of childhood cancer survivors. In this regard, the creation of international survivor registries has been an inflexion point. There is still a long way to go, however. In effect, we need more multidisciplinary units for the care of childhood cancer survivors; we must be proactive in our approach to patient follow-up and in the recording of sequelae; and we must ensure continuity between pediatric and adult units, since many of the sequelae will not become apparent until the patients reach adulthood. Scientific bodies have a crucial role to play here, by promoting the follow-up of survivors, encouraging ties between pediatric and adult units, and contributing to general strategies designed to encourage a healthy lifestyle. In other words, our goal is no longer limited to just healing the patient.

Conflicts of interest

The author states that she has no conflicts of interest.

Acknowledgments

Thanks are due to the Hematological Oncology team of Hospital Sant Joan de Déu for its support, and particularly to Dr. Ofelia Cruz, Dr. Albert Catalá and Dr. Ibañez for their valuable contributions to this review.

References
[1]
A. Jemal, R. Siegel, J. Xu, E. Ward.
Cancer statistics, 2010.
CA Cancer J Clin, 60 (2010), pp. 277-300
[2]
R. Peris, E. Pardo, A. Muñoz, N. Sayas, S. Valero.
Cáncer infantil en España. Estadísticas 1980–2015.
Registro Español de Tumores Infantiles (RETI-SEHOP), Universitat de València, (2016),
(Edición revisada, CD-Rom)
[3]
M.M. Hudson, K.K. Ness, J.G. Gurney, D.A. Mulrooney, W. Chemaitilly, K.R. Krull, et al.
Clinical ascertainment of health outcomes among adults treated for childhood cancer.
JAMA, 309 (2013), pp. 2371-2381
[4]
K.C. Oeffinger, A. Mertens, C.A. Sklar, T. Kawashima, M.M. Hudson, A.T. Meadows, for the Childhood Cancer Survivor Study*, et al.
Chronic health conditions in adult survivors of childhood cancer.
N Engl J Med, 355 (2006), pp. 1572-2158
[5]
G.T. Armstrong, Y. Chen, Y. Yasui, W. Leisenring, T.M. Gibson, A.C. Mertens, et al.
Reduction in late mortality among 5-year survivors of childhood cancer.
N Engl J Med, 374 (2016), pp. 833-842
[6]
G. Tallen, A. Resch, G. Calaminus, A. Wiener, U. Leiss, T. Pletschko, German Paediatric Brain Tumour Consortium (HIT Network), et al.
Strategies to improve the quality of survival for childhood brain tumour survivors.
Eur J Paediatr Neurol, 19 (2015), pp. 619-639
[7]
K.C. Oeffinger, C.A. Sklar.
Childhood cancer, endocrine disorders, and cohort studies.
Lancet, 383 (2014), pp. 1950-1952
[8]
S. De Fine Licht, J.F. Winther, T. Gudmundsdottir, A.S. Holmqvist, T.G. Bonnesen, P.H. Asdahl, ALiCCS study group, et al.
Hospital contacts for endocrine disorders in Adult Life after Childhood Cancer in Scandinavia (ALiCCS): a population-based cohort study.
Lancet, 383 (2014), pp. 1981-1989
[9]
Children's Oncology Group Long-Term Follow-Up Guidelines. Version 4.0 survivorshipguidelines.org. Available from: http://www.survivorshipguidelines.org/pdf/COG_LTFU Guidelines.
[10]
B.C. Patterson, K. Wasilewski-Masker, A.B. Ryerson, A. Mertens, L. Meacham.
Endocrine health problems detected in 519 patients evaluated in a pediatric cancer survivor program.
J Clin Endocrinol Metab, 97 (2012), pp. 810-818
[11]
P. Casano-Sancho, F. Carvallo, S. Rives, A. Catala, O. Cruz, A. Parareda.
Implementation of endocrine follow up in a Childhood Cancer Survivor Unit.
Horm Res Paediatr, 76 (2011), pp. 116
[12]
A. Nandagopal, C. Laverdière, D. Mulrooney, M.M. Hudson, L. Meacham.
Endocrine late effects of childhood cancer therapy: a report from the Children's Oncology Group.
Horm Res, 69 (2008), pp. 65-74
[13]
M. Schmiegelow, S. Lassen, H.S. Poulsen, U. Feldt-Rasmussen, K. Schmiegelow, H. Hertz.
Growth hormone response to a growth hormone-releasing hormone stimulation test in a population-based study following cranial irradiation of childhood brain tumors.
Horm Res, 54 (2000), pp. 53-59
[14]
V. Viswanathan, K.R. Pradhan, E.A. Eugster.
Pituitary hormone dysfunction after proton beam radiation therapy in children with brain tumors.
Endocr Pract, 17 (2011), pp. 891-896
[15]
P.K. Duffner, M.E. Cohen, M.L. Voorhess, M.H. MacGillivray, M.L. Brecher, A. Panahon, et al.
Long-term effects of cranial irradiation on endocrine function in children with brain tumors. A prospective study.
Cancer, 56 (1985), pp. 2189-2193
[16]
T.E. Merchant, S.R. Rose, C. Bosley, S. Wu, X. Xiong, R.H. Lustig.
Growth hormone secretion after conformal radiation therapy in pediatric patients with localized brain tumors.
J Clin Oncol, 29 (2011), pp. 4776-4780
[17]
Growth Hormone Research Society.
Consensus guidelines for the diagnosis and treatment of growth hormone (GH) deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society.
J Clin Endocrinol Metab, 85 (2000), pp. 3990-3993
[18]
B.C. Patterson, Y. Chen, C.A. Sklar, J. Neglia, Y. Yasui, A. Mertens, et al.
Growth hormone exposure as a risk factor for the development of subsequent neoplasms of the central nervous system: a report from the Childhood Cancer Survivor Study.
J Clin Endocrinol Metab, 99 (2014), pp. 2030-2037
[19]
J.P. Neglia, L.L. Robison, M. Stovall, Y. Liu, R.J. Packer, S. Hammond, et al.
New primary neoplasms of the central nervous system in survivors of childhood cancer: a report from the Childhood Cancer Survivor Study.
J Natl Cancer Inst, 98 (2006), pp. 1528-1537
[20]
T.A. Wilson, S.R. Rose, P. Cohen, A.D. Rogol, P. Backeljauw, R. Brown, Lawson Wilkins Pediatric Endocrinology Society Drug and Therapeutics Committee, et al.
Update of guidelines for the use of growth hormone in children: the Lawson Wilkins Pediatric Endocrinology Society Drug and Therapeutics Committee.
J Pediatr, 143 (2003), pp. 415-421
[21]
H.K. Gleeson, R. Stoeter, A.L. Ogilvy-Stuart, H.R. Gattamaneni, B.M. Brennan, S.M. Shalet.
Improvements in final height over 25 years in growth hormone (GH)-deficient childhood survivors of brain tumors receiving GH replacement.
J Clin Endocrinol Metab, 88 (2003), pp. 3682-3689
[22]
H.K. Gleeson, H.R. Gattamaneni, L. Smethurst, B.M. Brennan, S.M. Shalet.
Reassessment of growth hormone status is required at final height in children treated with growth hormone replacement after radiation therapy.
J Clin Endocrinol Metab, 89 (2004), pp. 662-666
[23]
M.L. Reed, G.R. Merriam, A.Y. Kargi.
Adult growth hormone deficiency – benefits, side effects, and risks of growth hormone replacement.
Front Endocrinol, 4 (2013), pp. 64
[24]
M. Steffens, V. Beauloye, B. Brichard, A. Robert, O. Alexopoulou, Ch. Vermylen, et al.
Endocrine health conditions in adult survivors of childhood cancer: the need for specialized adult-focused follow-up clinics.
Eur J Endocrinol, 168 (2013), pp. 465-472
[25]
K.S. Leong, A.B. Walker, I. Martin, D. Wile, J. Wilding, I.A. MacFarlane.
An audit of 500 subcutaneous glucagon stimulation test to assess growth hormone and ACTH secretion in patients with hypothalamic–pituitary disease.
Clin Endocrinol, 54 (2001), pp. 463-468
[26]
M. Schmiegelow, U. Feldt-Rasmussen, A.K. Rasmussen, M. Lange, H.S. Poulsen, J. Muller.
Assessment of the hypothalamo–pituitary–adrenal axis in patients treated with radiotherapy and chemotherapy for childhood brain tumor.
J Clin Endocrinol Metab, 88 (2003), pp. 3149-3154
[27]
S.R. Rose, R.H. Lustig, P. Pitukcheewanont, D.C. Broome, G.A. Burghen, H. Li, et al.
Diagnosis of hidden central hypothyroidism in survivors of childhood cancer.
J Clin Endocrinol Metab, 84 (1999), pp. 4472-4479
[28]
G.T. Armstrong, E.J. Chow, C.A. Sklar.
Alterations in pubertal timing following therapy for childhood malignancies.
Endocr Dev, 15 (2009), pp. 25-39
Review
[29]
L.B. Kenney, L.E. Cohen, M. Shnorhavorian, M.L. Metzger, B. Lockart, N. Hijiya, et al.
Male reproductive health after childhood, adolescent, and young adult cancers: a report from the Children's Oncology Group.
J Clin Oncol, 30 (2012), pp. 3408-3416
[30]
F.H. Pierik, J.T. Vreeburg, T. Stijnen, F.H. de Jong, R.F. Weber.
Serum inhibin B as a marker of spermatogenesis.
J Clin Endocrinol Metab, 83 (1998), pp. 3110-3114
[31]
M.W. Ben Arush, I. Solt, A. Lightman, S. Linn, A. Kuten.
Male gonadal function in survivors of childhood Hodgkin and non-Hodgkin lymphoma.
Pediatr Hematol Oncol, 17 (2000), pp. 239-245
[32]
R.D. Van Beek, M. Smit, M.M. van den Heuvel-Eibrink, F.H. de Jong, F.G. Hakvoort-Cammel, C. van den Bos, et al.
Inhibin B is superior to FSH as a serum marker for spermatogenesis in men treated for Hodgkin's lymphoma with chemotherapy during childhood.
Hum Reprod, 22 (2007), pp. 3215-3222
[33]
D.M. Green, L. Zhu, N. Zhang, C.A. Sklar, R.W. Ke, W.H. Kutteh, et al.
Lack of specificity of plasma concentrations of inhibin B and follicle-stimulating hormone for identification of azoospermic survivors of childhood cancer: a report from the St Jude lifetime cohort study.
J Clin Oncol, 31 (2013), pp. 1324-1328
[34]
T.T. Ortin, C.A. Shostak, S.S. Donaldson.
Gonadal status and reproductive function following treatment for Hodgkin's disease in childhood: the Stanford experience.
Int J Radiat Oncol Biol Phys, 19 (1990), pp. 873-880
[35]
S. Lie Fong, J.S. Laven, F.G. Hakvoort-Cammel, I. Schipper, J.A. Visser, A.P. Themmen, et al.
Assessment of ovarian reserve in adult childhood cancer survivors using anti-Müllerian hormone.
Hum Reprod, 24 (2009), pp. 982-990
[36]
S. Lie Fong, P.J. Lugtenburg, I. Schipper, A.P. Themmen, F.H. de Jong, P. Sonneveld, et al.
Anti-Müllerian hormone as a marker of ovarian function in women after chemotherapy and radiotherapy for haematological malignancies.
Hum Reprod, 23 (2008), pp. 674
[37]
A.A. Gupta, A. Lee Chong, C. Deveault, J. Traubici, A.M. Maloney, Knight, et al.
Anti-Müllerian hormone in female adolescent cancer patients before, during, and after completion of therapy: a pilot feasibility study.
J Pediatr Adolesc Gynecol, 29 (2016), pp. 599-603
[38]
W. Van Dorp, R.L. Mulder, L.C. Kremer, M.M. Hudson, M.M. van den Heuvel-Eibrink, M.H. van den Berg, et al.
Recommendations for premature ovarian insufficiency surveillance for female survivors of childhood, adolescent, and young adult cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCareSurFup consortium.
J Clin Oncol, 34 (2016), pp. 3440-3450
[39]
E. Vandecruys, C. Dhooge, M. Craen, Y. Benoit, J. de Schepper.
Longitudinal linear growth and final height is impaired in childhood acute lymphoblastic leukemia survivors after treatment without cranial irradiation.
J Pediatr, 163 (2013), pp. 268-273
[40]
A.J. Taylor, A.P. Croft, A.M. Palace, D.L. Winter, R.C. Reulen, C.A. Stiller, et al.
Risk of thyroid cancer in survivors of childhood cancer: results from the British Childhood Cancer Survivor Study.
Int J Cancer, 125 (2009), pp. 2400-2405
[41]
E. Brignardello, F. Felicetti, A. Castiglione, M. Gallo, F. Maletta, G. Isolato, et al.
Ultrasound surveillance for radiation-induced thyroid carcinoma in adult survivors of childhood cancer.
Eur J Cancer, 55 (2016), pp. 74-80
[42]
M. Goldfarb, D.R. Freyer.
Comparison of secondary and primary thyroid cancer in adolescents and young adults.
Cancer, 120 (2014), pp. 1155-1161
[43]
S.C. Clement, L.C. Kremer, T.P. Links, R.L. Mulder, C.M. Ronckers, B.L. van Eck-Smit, et al.
Is outcome of differentiated thyroid carcinoma influenced by tumor stage at diagnosis?.
Cancer Treat Rev, 41 (2015), pp. 9-16
[44]
H. Gharib, E. Papini, R. Paschke, D.S. Duick, R. Valcavi, L. Hegedüs, AACE/AME/ETA Task Force on Thyroid Nodules, et al.
American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and European Thyroid Association Medical guidelines for clinical practice for the diagnosis and management of thyroid nodules: executive summary of recommendations.
Endocr Pract, 16 (2010), pp. 468-475
[45]
American Thyroid Association (ATA) Guidelines Taskforce on thyroid nodules and differentiated thyroid cancer, D.S. Cooper, G.M. Doherty, B.R. Haugen, R.T. Kloos, S.L. Lee, S.J. Mandel, et al.
Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer.
Thyroid, 19 (2009), pp. 1167-1214
[46]
L.B. Kenney, L.E. Cohen, M. Shnorhavorian, M.L. Metzger, B. Lockart, N. Hijiya, et al.
Male reproductive health after childhood, adolescent, and young adult cancers: a report from the Children's Oncology Group.
J Clin Oncol, 20 (2012), pp. 3408-3416
[47]
M.M. Hudson, C. Greenwald, E. Thompson, J. Wilimas, N. Marina, D. Fairclough, et al.
Efficacy and toxicity of multiagent chemotherapy and low-dose involved-field radiotherapy in children and adolescents with Hodgkin's disease.
J Clin Oncol, 11 (1993), pp. 100-108
[48]
E.J. Chow, L. Anderson, K.S. Baker, S. Bhatia, G.M. Guilcher, J.T. Huang, et al.
Late effect surveillance recommendations among survivors of childhood hematopoietic cell transplantation: a Children's Oncology Group Report.
Biol Blood Marrow Transplant, 22 (2016), pp. 782-795
[49]
S.M. Corsello, A. Barnabei, P. Marchetti, L. de Vecchis, R. Salvatori, F. Torino.
Endocrine side effects induced by immune checkpoint inhibitors.
J Clin Endocrinol Metab, 98 (2013), pp. 1361-1375
[50]
M.B. Lodish.
Clinical review: kinase inhibitors: adverse effects related to the endocrine system.
J Clin Endocrinol Metab, 98 (2013), pp. 1333-1342
[51]
F. Torino, A. Barnabei, R. Paragliola, R. Baldelli, M. Appetecchia, S.M. Corsello.
Thyroid dysfunction as an unintended side effect of anticancer drugs.
Thyroid, 23 (2013), pp. 1345-1366
[52]
O. Bilgir, L. Kebapcilar, F. Bilgir, I. Sari, P. Oner, B. Karaca, et al.
Is there any relationship between imatinib mesylate medication and hypothalamic–pituitary–adrenal axis dysfunction?.
Int J Clin Pract, 64 (2010), pp. 45-50
[53]
A. Juszczak, A. Gupta, N. Karavitaki, M.R. Middleton, A.B. Grossman.
Ipilimumab: a novel immunomodulating therapy causing autoimmune hypophysitis: a case report and review.
Eur J Endocrinol, 167 (2012), pp. 1-5
[54]
K.R. Narayanan, D. Bansal, R. Walia, N. Sachdeva, A. Bhansali, N. Varma, et al.
Growth failure in children with chronic myeloid leukemia receiving imatinib is due to disruption of GH/IGF-1 axis.
Pediatr Blood Cancer, 60 (2013), pp. 1148-1153
[55]
S. O'Sullivan, J.M. Lin, M. Watson, K. Callon, P.C. Tong, D. Naot, et al.
The skeletal effects of the tyrosine kinase inhibitor nilotinib.
[56]
K. Wasilewski-Masker, S.C. Kaste, M.M. Hudson, N. Esiashvili, L.A. Mattano, L.R. Meacham.
Bone mineral density deficits in survivors of childhood cancer: long-term follow-up guidelines and review of the literature.
Pediatrics, 121 (2008), pp. e705-e713
[57]
J.G. Gurney, S.C. Kaste, W. Liu, D.K. Srivastava, W. Chemaitilly, K.K. Ness, et al.
Bone mineral density among long-term survivors of childhood acute lymphoblastic leukemia: results from the St. Jude Lifetime Cohort Study.
Pediatr Blood Cancer, 61 (2014), pp. 1270-1276
[58]
E.G. Garmey, Q. Liu, C.A. Sklar, L.R. Meacham, A.C. Mertens, M.A. Stovall, et al.
Longitudinal changes in obesity and body mass index among adult survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study.
J Clin Oncol, 26 (2008), pp. 4639-4645
[59]
S. Casco, E. Soto-Vega.
Development of metabolic syndrome associated to cancer therapy: review.
Horm Cancer, 7 (2016), pp. 289-295

Please cite this article as: Casano Sancho P. Secuelas endocrinológicas en supervivientes de cáncer infantil. Endocrinol Diabetes Nutr. 2017;64:498–505.

Copyright © 2017. SEEN and SED
Article options
Tools
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos