Abstract

The population of survivors of childhood leukemia who reach adulthood is growing due to improved therapy. However, survivors are at risk of long-term complications. Comprehensive follow-up programs play a key role in childhood leukemia survivor care. The major determinant of long-term complications is the therapeutic burden accumulated over time. Relapse chemotherapy, central nervous system irradiation, hematopoietic stem cell transplantation, and total body irradiation are associated with greater risk of long-term complications. Other parameters include clinical characteristics such as age and sex as well as environmental, genetic, and socioeconomic factors, which can help stratify the risk of long-term complications and organize follow-up program. Early diagnosis improves the management of several late complications such as anthracycline-related cardiomyopathy, secondary cancers, metabolic syndrome, development defects, and infertility. Total body irradiation is the treatment associated with worse long-term toxicity profile with a wide range of complications. Patients treated with chemotherapy alone are at a lower risk of long-term complications, although the optimal long-term follow-up remains unclear. Novel immunotherapies and targeted therapy are generally associated with a better short-term safety profile but still require careful long-term toxicity monitoring. Advances in understanding genetic susceptibility to long-term complications could enable tailored therapeutic strategies for leukemia treatment and optimized follow-up programs.

Improved antileukemia therapy and supportive care has led to a significant improvement in the prognosis of childhood acute leukemias. The cure rate now exceeds 90% for acute lymphoblastic leukemia (ALL), which is the most common type of pediatric leukemia. The population of survivors of childhood leukemia who reach adulthood is thus rapidly growing. However, survivors are at risk of long-term chronic health conditions due to the disease and therapy.1 Minimizing the long-term impacts of treatment is a significant challenge. The major determinant of long-term complications is therapeutic exposure. Other parameters include some clinical characteristics (eg, age and sex) as well as environmental, genetic, and socioeconomic factors (Figure 1). The objective of this document is to provide guidance on the risk-stratification, diagnosis, and clinical management principles of these long-term complications.

Figure 1.

Risk of long-term complications in survivors of childhood acute leukemia. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 1.

Risk of long-term complications in survivors of childhood acute leukemia. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Clinical case

A 19-year-old female patient consulted with the childhood leukemia survivorship program. She had a history of acute myeloid leukemia (AML) diagnosed at age 16 years and was treated with intensive chemotherapy without hematopoietic stem cell transplantation (HSCT). She has never relapsed from the disease. The patient weighed 89 kg and had a height of 163 cm and a body mass index of 33.5 kg/m2, vs 49 kg, 158 cm, and 19.6 kg/m2 at AML diagnosis. Her blood pressure was 147/85 mm Hg, and her abdominal circumference was 112 cm. Her fasting blood test revealed elevated levels of fasting triglycerides (3.4 mmol/L) and hyperglycemia (120 mg/dL; 6.7 mmol/L). The patient was diagnosed with metabolic syndrome. An ultrasound with transient elastography was performed and revealed hepatic steatosis without fibrosis. This patient was referred to an endocrinologist and was started on a nutritional and physical activity intervention to improve the metabolic parameters. The patient did not require pharmaceutical intervention for the metabolic parameters. Those parameters improved, and no overt cardiovascular complication was observed at the last evaluation.

This clinical case illustrates the importance of long-term follow-up programs in the early diagnosis of certain complications. Early diagnosis of long-term side effects is likely key to increase the chances of curing or preventing the complications of the side effects and can help preserve quality of life (Table 1). The management of metabolic syndrome, secondary malignant neoplasms and meningiomas, and anthracycline-related cardiomyopathy is significantly improved by early detection. Because the latter complication is specifically due to anthracycline exposure, it will be discussed in the dedicated section of the manuscript. Early diagnosis of fertility and developmental defects (growth retardation, neurodevelopmental defects, and pubertal abnormalities) are also essential. Because HSCT recipients are particularly exposed to these complications, they are discussed in the dedicated section of the manuscript.

Table 1.

Long-term complications of childhood leukemia that benefit from early diagnosis

Long-term complicationsScreening and diagnostic toolsEarly intervention
Secondary tumors
Thyroid cancer
Breast cancer
Skin cancer
Meningioma and other brain tumors 
Thyroid US
Mammography, US, MRI
Clinical screening
Brain MRI/CT as clinically indicated 
Early oncological management 
Cardiac and vascular risk
Anthracycline-related cardiomyopathy
Metabolic syndrome 
Calculation of anthracyclines cumulative dose (doxorubicin-equivalent dose), echocardiography (with global longitudinal strain)
Metabolic parameters (weight, abdominal circumference, fasting glycemia, insulin level and lipid profiles, HbA1c) 
Cardioprotective drugs (eg, angiotensin-converting enzyme inhibitors), cardiovascular risk
Nutritional and physical activity intervention, lipid-lowering and antidiabetic medications 
Development
Delayed puberty
Growth retardation
Neurodevelopment 
Puberty assessment (Tanner)
Growth chart analysis and follow-up
Endocrine evaluation
Focused clinical evaluation of neurodevelopment 
Induction of puberty
Hormone therapy
Educational support and interventions 
Iron overload Number of RBC transfusions, posttreatment ferritin levels, liver function tests, MRI liver iron quantification Phlebotomy, iron chelation 
Infertility Calculation of alkylating agent cumulative dose (cyclophosphamide-equivalent dose), AMH levels Early postcancer fertility counseling 
Long-term complicationsScreening and diagnostic toolsEarly intervention
Secondary tumors
Thyroid cancer
Breast cancer
Skin cancer
Meningioma and other brain tumors 
Thyroid US
Mammography, US, MRI
Clinical screening
Brain MRI/CT as clinically indicated 
Early oncological management 
Cardiac and vascular risk
Anthracycline-related cardiomyopathy
Metabolic syndrome 
Calculation of anthracyclines cumulative dose (doxorubicin-equivalent dose), echocardiography (with global longitudinal strain)
Metabolic parameters (weight, abdominal circumference, fasting glycemia, insulin level and lipid profiles, HbA1c) 
Cardioprotective drugs (eg, angiotensin-converting enzyme inhibitors), cardiovascular risk
Nutritional and physical activity intervention, lipid-lowering and antidiabetic medications 
Development
Delayed puberty
Growth retardation
Neurodevelopment 
Puberty assessment (Tanner)
Growth chart analysis and follow-up
Endocrine evaluation
Focused clinical evaluation of neurodevelopment 
Induction of puberty
Hormone therapy
Educational support and interventions 
Iron overload Number of RBC transfusions, posttreatment ferritin levels, liver function tests, MRI liver iron quantification Phlebotomy, iron chelation 
Infertility Calculation of alkylating agent cumulative dose (cyclophosphamide-equivalent dose), AMH levels Early postcancer fertility counseling 

AMH, anti-Mullerian hormone; CT, computed tomography; Hb, hemoglobin; MRI, magnetic resonance imaging; RBC, red blood cell; US, ultrasound.

Metabolic syndrome

Survivors of childhood acute leukemia have a cardiovascular mortality rate >4 times higher than their siblings or the general population.2,3 This excess mortality is partly associated with ischemic heart disease and stroke. These patients show early signs of atherosclerotic lesions.4 The natural history of atheromatous disease begins years before the onset of clinically relevant lesions. Metabolic syndrome, also called insulin resistance syndrome, is a cluster of conditions (abdominal obesity, dyslipidemia, glucose intolerance, and hypertension) associated with a high risk of cardiovascular events, including coronary heart disease, diabetes, and stroke.5 Hence, monitoring the metabolic syndrome in survivors of childhood leukemia is important, although recent data show a trend in declining cardiovascular toxicity among more recently treated patients with ALL.6 

Metabolic syndrome is more prevalent in young adult survivors of childhood leukemia than in the age- and sex-matched general population. Central nervous system (CNS) irradiation, HSCT, and total body irradiation (TBI) are key risk factors,7 although an increased risk has also been demonstrated for those who received chemotherapy alone (Table 2).8,9 Obesity, hyperlipidemia, and insulin resistance have been associated with constitutional variants in the glucocorticoid receptor gene NR3C1, the cadherin family gene CDH19, and genes involved in brain development (NALF1, SOX11, and GLRA3).14,15 Other studies have shown that genetic risk scores of the general population can be used to predict severe obesity in survivors of childhood leukemia.16 

Table 2.

Metabolic syndrome according to therapeutic exposure

Therapeutic exposureRelative risk8, Specific characteristics7,9 Pathophysiology10-12 Early detection12,13 Treatment specificities
Treatment-specificAll survivors
HSCT with TBI ×6.3 (×9.2 in females) Increased severity of metabolic syndrome
Lower incidence of obesity and lower abdominal circumference, higher triglycerides, and glucose level 
Radiation induced alteration of subcutaneous adipose tissue (preadipocyte differentiation)
Additional role of pancreatic radiation, testosterone, and growth hormone deficiency 
Low-grade chronic inflammation
Poor eating habits and reduced activity during prolonged periods
Genetic predisposition 
Regular monitoring:
Blood pressure
Abdominal circumference
Fasting glucose, triglyceride,
HDL- and LDL-cholesterol
Potential interest of early
biomarkers (adipokines)? 
Few specific data
Moderate effect of lifestyle modifications in the LEA experience 
HSCT without TBI ×2.2 Usually less severe than after HSCT with TBI Largely unknown
Role of testosterone deficiency 
Chemotherapy and CNS irradiation ×2.3 More frequent abdominal obesity
Low incidence of hypertension 
Important role of obesity
Leptin resistance and overproduction (damaged hypothalamic receptors)
Growth hormone deficiency 
Chemotherapy without CNS irradiation ×1.7 More frequent hypertension (compared to CNS irradiation) Largely unknown
Uncertain long-term role of steroid and asparaginase 
Therapeutic exposureRelative risk8, Specific characteristics7,9 Pathophysiology10-12 Early detection12,13 Treatment specificities
Treatment-specificAll survivors
HSCT with TBI ×6.3 (×9.2 in females) Increased severity of metabolic syndrome
Lower incidence of obesity and lower abdominal circumference, higher triglycerides, and glucose level 
Radiation induced alteration of subcutaneous adipose tissue (preadipocyte differentiation)
Additional role of pancreatic radiation, testosterone, and growth hormone deficiency 
Low-grade chronic inflammation
Poor eating habits and reduced activity during prolonged periods
Genetic predisposition 
Regular monitoring:
Blood pressure
Abdominal circumference
Fasting glucose, triglyceride,
HDL- and LDL-cholesterol
Potential interest of early
biomarkers (adipokines)? 
Few specific data
Moderate effect of lifestyle modifications in the LEA experience 
HSCT without TBI ×2.2 Usually less severe than after HSCT with TBI Largely unknown
Role of testosterone deficiency 
Chemotherapy and CNS irradiation ×2.3 More frequent abdominal obesity
Low incidence of hypertension 
Important role of obesity
Leptin resistance and overproduction (damaged hypothalamic receptors)
Growth hormone deficiency 
Chemotherapy without CNS irradiation ×1.7 More frequent hypertension (compared to CNS irradiation) Largely unknown
Uncertain long-term role of steroid and asparaginase 

CNS, central nervous system; HDL, high-density lipoprotein; LDL, low-density lipoprotein.

Compared with age- and sex-matched controls.

Regular monitoring of blood pressure, abdominal circumference, body mass index, as well as glycemic and lipid parameters should be included in long-term survivorship evaluations. In our experience, lifestyle modifications have a moderate effect on metabolic parameters in survivors of childhood leukemia diagnosed with metabolic syndrome. In these patients, endocrinology referral is required for evaluation and management. If nutritional and physical activity interventions do not correct the metabolic syndrome, appropriate medications should be prescribed to mitigate the cardiovascular and metabolic risks. Lipid-lowering medications include statins to prevent cardiovascular events. Antidiabetic medications primarily include metformin and glucagon-like-peptide 1 agonists, which are efficient therapies to treat diabetes associated with severe insulin resistance.

Secondary malignant neoplasm and meningioma

Secondary tumors are relatively rare (∼1%-2%) but severe long-term complications of childhood leukemia. These complications may occur rapidly after treatment (eg, secondary leukemia related to epipodophyllotoxin treatment associated with KMT2A rearrangement)17 or very late after the initial diagnosis. Continued patient follow-up suggests that the cumulative incidence of some secondary cancers such as meningioma, breast cancer, and other carcinomas does not plateau.18 The initial characteristics of primary leukemia are not related to the occurrence of secondary tumors. By contrast, therapeutic exposure is highly correlated with these complications. Patients treated with radiotherapy (TBI or CNS irradiation) have an increased risk of secondary tumors as described in the dedicated sections of this document. Chemotherapy with alkylating agents and epipodophyllotoxins are also risk factors of secondary cancers, primarily secondary leukemia. The prognosis of these secondary tumors varies. Some are associated with an adverse prognosis (eg, secondary myelodysplastic syndrome, AML, and nonmeningioma brain tumor). Other secondary tumors have a more favorable prognosis (eg, meningioma, Hodgkin lymphoma, thyroid carcinoma, and basal cell carcinoma), provided that the disease is diagnosed early. Long-term follow-up visits with clinical evaluation and screening tests for patients at high risk (eg, complete blood count monitoring, breast cancer screening, and thyroid ultrasound) may enable the detection of secondary tumors at an earlier stage. Other observations suggest that early oncological management may have a positive impact on the prognosis.19 Further research is needed to identify the molecular mechanisms and potential interventions to prevent or treat secondary cancers in survivors of pediatric leukemia.

Clinical case

The patient is a 44-year-old woman. She was treated for relapsed ALL at age 6 years and received combination chemotherapy and HSCT. The preparation included a 12 Gy TBI in 6 fractions over 3 days with 8 Gy lung shielding. This treatment cured the leukemia. Several long-term complications affecting her quality of life were diagnosed during the follow-up program. She had normal puberty and menstruation. However, she had a miscarriage after her first pregnancy at age 20 years, which may have been associated with radiation-induced uterus damage. She was diagnosed with bilateral posterior subcapsular cataracts that did not require surgery. She also had a growth hormone deficiency with growth retardation. Despite hormone replacement, she stopped growing at a height of 153 cm. The neck palpation was normal. However, a systematic thyroid ultrasound identified several suspect thyroid nodules at age 26 years. A secondary thyroid cancer was diagnosed and treated using thyroidectomy and radioactive iodine treatment.

This patient had several long-term complications directly associated with HSCT and TBI. This case illustrates that therapeutic exposure is the key determinant of long-term complications in survivors of childhood leukemia (Table 3).

Table 3.

Major complications according to therapeutic exposure

Therapeutic exposureLong-term complications
TBI Metabolic syndrome (lipodystrophy-like features), and overt metabolic and cardiovascular complications (myocardial infarction, stroke, diabetes), osteonecrosis, low bone mineral density, severe growth retardation, hypothyroidism, secondary malignant neoplasms and meningioma, ovarian/testicular hormone deficiency, reduced ovarian follicular pool, impaired spermatogenesis, uterine toxicity, cataract, chronic renal insufficiency 
High-dose alkylating agents (primarily busulfan-based conditioning) Complications overlap with TBI (notably fertility defects); less frequent metabolic syndrome, secondary malignant neoplasms and meningioma, growth retardation and cataracts; increased risk of persistent partial alopecia 
HSCT (any conditioning regimen) Bronchiolitis obliterans, chronic graft-versus-host disease 
CNS irradiation Metabolic syndrome, obesity, endocrine complications (central hypothyroidism, gonadotropin deficiency, precocious puberty), growth retardation, neurocognitive defects, secondary malignant neoplasms and meningioma, cataracts 
Anthracyclines and anthraquinones Anthracycline-related cardiomyopathy 
High-dose corticosteroids Osteonecrosis, cataracts 
Therapeutic exposureLong-term complications
TBI Metabolic syndrome (lipodystrophy-like features), and overt metabolic and cardiovascular complications (myocardial infarction, stroke, diabetes), osteonecrosis, low bone mineral density, severe growth retardation, hypothyroidism, secondary malignant neoplasms and meningioma, ovarian/testicular hormone deficiency, reduced ovarian follicular pool, impaired spermatogenesis, uterine toxicity, cataract, chronic renal insufficiency 
High-dose alkylating agents (primarily busulfan-based conditioning) Complications overlap with TBI (notably fertility defects); less frequent metabolic syndrome, secondary malignant neoplasms and meningioma, growth retardation and cataracts; increased risk of persistent partial alopecia 
HSCT (any conditioning regimen) Bronchiolitis obliterans, chronic graft-versus-host disease 
CNS irradiation Metabolic syndrome, obesity, endocrine complications (central hypothyroidism, gonadotropin deficiency, precocious puberty), growth retardation, neurocognitive defects, secondary malignant neoplasms and meningioma, cataracts 
Anthracyclines and anthraquinones Anthracycline-related cardiomyopathy 
High-dose corticosteroids Osteonecrosis, cataracts 

TBI

TBI has the most toxic profile of all antileukemia therapies. However, in children aged >4 years with ALL, TBI has been associated with improved overall survival and lower relapse risk than the chemotherapy-only conditioning.20 TBI is generally not used in children aged <2 to 4 years due to fear of major toxicity. Most patients now receive a fractionated schedule of 12 Gy over 3 days and 6 fractions with lung shielding at 8 Gy, but some patients treated during the 1980s may have received less fractionated schedules associated with increased toxicity. The long-term survivors of childhood leukemia treated with TBI require life-long follow-up, because they are at risk of various complications. Our recommendations for the surveillance of patients with childhood leukemia treated with TBI are described in Table 4. These recommendations are based on the data from the French LEA cohort and largely overlap with the Children Oncology Group and International Guideline Harmonization Group/PanCare guidelines.

Table 4.

Surveillance of patients treated with TBI

System or organ involvedComplications and screening method
Growth Failure to thrive: weight and height measurements compared against previous values and values of children of the same age using growth charts (yearly), pediatric endocrinology referral as clinically indicated 
Puberty, fertility Pubertal delay: Tanner stages including testicular volume evaluation in boys (yearly until sexually mature); ovarian/testicular hormone deficiency: FSH, LH, estradiol/testosterone levels at adolescence 
Brain Meningiomas and other CNS tumors: brain MRI (or CT) as clinically indicated; neurodevelopmental defects: referral for neurodevelopmental evaluation as clinically indicated 
Skin Skin cancer (basal cell carcinomas, melanomas): dermatological examination (yearly) 
Breast Breast cancer: education to breast autopalpation, mammogram and breast MRI (yearly after age 25 y) 
Heart Anthracycline-related cardiomyopathy: echocardiography with global longitudinal strain (every 2 y) 
Thyroid Thyroid cancer: neck palpation (yearly) and thyroid US (every 2 y), oncological referral in case of thyroid nodule; thyroid function: TSH, FT4 (yearly) 
Lungs Pulmonary function defects: pulmonary function tests including DLCO and spirometry (at entry into long-term follow-up, repeat as clinically indicated); lung cancer: pulmonary examination, consider lung spiral CT in high-risk patients (other risk factors such as smoking) 
Metabolic and cardiovascular risk Metabolic syndrome: blood pressure, waist circumference, body mass index, fasting triglyceride, total, LDL- and HDL-cholesterol, glucose, and insulin levels, HbA1c level (yearly from adulthood) 
Eyes Cataracts: ophthalmology referral for slit-lamp examination (every 2 y) 
Oral health Dental defects: dental care (yearly) 
Liver Liver toxicity: physical examination, AST, ALT, GGT, bilirubin (at entry into long-term follow-up, repeat as clinically indicated) 
Kidney Renal toxicity: creatinine, electrolytes including Ca, P, and Mg, urinary tests (at entry into long-term follow-up, repeat as clinically indicated) 
System or organ involvedComplications and screening method
Growth Failure to thrive: weight and height measurements compared against previous values and values of children of the same age using growth charts (yearly), pediatric endocrinology referral as clinically indicated 
Puberty, fertility Pubertal delay: Tanner stages including testicular volume evaluation in boys (yearly until sexually mature); ovarian/testicular hormone deficiency: FSH, LH, estradiol/testosterone levels at adolescence 
Brain Meningiomas and other CNS tumors: brain MRI (or CT) as clinically indicated; neurodevelopmental defects: referral for neurodevelopmental evaluation as clinically indicated 
Skin Skin cancer (basal cell carcinomas, melanomas): dermatological examination (yearly) 
Breast Breast cancer: education to breast autopalpation, mammogram and breast MRI (yearly after age 25 y) 
Heart Anthracycline-related cardiomyopathy: echocardiography with global longitudinal strain (every 2 y) 
Thyroid Thyroid cancer: neck palpation (yearly) and thyroid US (every 2 y), oncological referral in case of thyroid nodule; thyroid function: TSH, FT4 (yearly) 
Lungs Pulmonary function defects: pulmonary function tests including DLCO and spirometry (at entry into long-term follow-up, repeat as clinically indicated); lung cancer: pulmonary examination, consider lung spiral CT in high-risk patients (other risk factors such as smoking) 
Metabolic and cardiovascular risk Metabolic syndrome: blood pressure, waist circumference, body mass index, fasting triglyceride, total, LDL- and HDL-cholesterol, glucose, and insulin levels, HbA1c level (yearly from adulthood) 
Eyes Cataracts: ophthalmology referral for slit-lamp examination (every 2 y) 
Oral health Dental defects: dental care (yearly) 
Liver Liver toxicity: physical examination, AST, ALT, GGT, bilirubin (at entry into long-term follow-up, repeat as clinically indicated) 
Kidney Renal toxicity: creatinine, electrolytes including Ca, P, and Mg, urinary tests (at entry into long-term follow-up, repeat as clinically indicated) 

AMH, anti-Mullerian hormone; ALT, alanine aminotransferase; AST, aspartate aminotransferase; CT, computed tomography; DLCO, carbon monoxide diffusing capacity; FSH, follicle-stimulating hormone; FT4, thyroxin; GGT, gamma-glutamyl transferase; Hb, hemoglobin; HDL, high-density lipoprotein; LH, luteinizing hormone; TSH, thyroid-stimulating hormone; US, ultrasound.

Patients treated with TBI are at high risk of metabolic syndrome. Notably, many adults who have received TBI have dyslipidemia, insulin resistance, and ectopic fat accumulation including nonalcoholic fatty liver disease.8 These lipodystrophy-like features are associated with altered gene expression profiles of the subcutaneous adipose tissue and the alteration of preadipocyte differentiation.10 

The risk of secondary cancer (including thyroid papillary carcinomas, breast cancer, skin cancer, sarcoma, meningioma, and other brain tumors) is increased in patients who received TBI. The risk of breast cancer should be particularly considered in women.21 Breast cancer usually occurs very late (>20 years) after initial diagnosis with a relative risk of ∼20 compared with the general population.22 

Patients treated with TBI are at high risk of premature cataracts.23 High-dose corticosteroids and CNS irradiation are other demonstrated risk factors.24 Posterior subcapsular cataracts without clinically significant visual symptoms are the most frequent forms. However, cataracts can rarely manifest with decreased visual acuity, halos, or diplopia. Ophthalmologic consultation with slit-lamp examination is required for the diagnosis. Patients with severe visual discomfort should be treated with surgical replacement of the opaque lens with an intraocular implant.

Patients treated with TBI are at risk of hypothyroidism, especially when treated at a young age.25 Patients with hypothyroidism should be referred to endocrinology and appropriately supplemented.

Height growth after HSCT requires careful assessment. The risk of decreased growth velocity mainly depends on age, sex, and conditioning regimen. The median final height of adults included in the French LEA program who received TBI for ALL before age 9 years for girls and before 11 years for boys was 153 cm and 163 cm, respectively. After busulfan-based conditioning regimen, the median final height was 162 cm and 170 cm, respectively. Appropriate hormonal replacement therapy may improve the final height of patients with documented growth hormone deficiency after HSCT.26 Height growth and final height are usually little to unaffected in patients treated with chemotherapy alone.27 

Gonadal toxicity is very frequent after a myeloablative conditioning regimen and is influenced by age at HSCT and sex (Table 5). Infertility is clearly associated with premature ovarian insufficiency, with most risk factors shared between these 2 complications. However, some women who required puberty induction for apparent immediate premature ovarian insufficiency may experience spontaneous and sometimes undesired pregnancies.28 In addition to ovarian function, uterine volume and function are also determinants of fertility. Adult women who received HSCT during childhood often have a reduced uterus volume.29 The reduced uterus volume may be associated with significant pregnancy and delivery issues. Data from our group showed that less than half of spontaneous pregnancies result in live births due to a high rate of miscarriages and abortions.28 All pregnancies after HSCT should be considered at risk of unfavorable obstetric outcome and require adapted follow-up. Pre-HSCT ovarian cryopreservation should be discussed with patients according to age and after considering the potential increased risk of premature ovarian failure associated with ovariectomy.28 Two-thirds of men who received a 12 Gy fractionated TBI during childhood will have total or partial testosterone deficiency, with a higher risk for older boys at the time of HSCT. After a busulfan-based conditioning, more than half will retain normal testosterone production without any effect of age at the time of therapy.11 

Table 5.

Gonadal hormone function and uterine volume after HSCT according to the conditioning regimen

Sex/age at HSCTParameterOutcomeBusulfan–based conditioning regimenTBI–based conditioning regimen
Adult women who underwent transplantation before puberty Ovarian hormone function28  Immediate POI (pubertal induction required) 60% 
Spontaneous menarche with secondary POI during the 10 y after puberty 20% 
Spontaneous menarche without POI at 10 y after puberty 20% 
Favorable risk factor Younger age at HSCT 
Uterus volume29  Uterine volume (vs general population) 45% volume reduction 75% volume reduction 
Favorable risk factor Hormone level and younger age at HSCT None 
Adult men Leydig cell function11  Total testosterone deficiency§  25% 45% 
Partial testosterone deficiency 15% 15% 
No testosterone deficiency 60% 35% 
Favorable risk factor  Older at HSCT 
Sex/age at HSCTParameterOutcomeBusulfan–based conditioning regimenTBI–based conditioning regimen
Adult women who underwent transplantation before puberty Ovarian hormone function28  Immediate POI (pubertal induction required) 60% 
Spontaneous menarche with secondary POI during the 10 y after puberty 20% 
Spontaneous menarche without POI at 10 y after puberty 20% 
Favorable risk factor Younger age at HSCT 
Uterus volume29  Uterine volume (vs general population) 45% volume reduction 75% volume reduction 
Favorable risk factor Hormone level and younger age at HSCT None 
Adult men Leydig cell function11  Total testosterone deficiency§  25% 45% 
Partial testosterone deficiency 15% 15% 
No testosterone deficiency 60% 35% 
Favorable risk factor  Older at HSCT 

The data presented are from the LEA program.

Premature ovarian insufficiency (POI) is defined as the association of an oligo/amenorrhea for at least 4 months and an elevated follicle-stimulating hormone (FSH) level >25 IU/L on 2 occasions at 4 weeks apart before age 40 years of age.

Using MRI evaluation.

Either normal ovarian function or hormone replacement therapy.

§

Total testosterone deficiency was defined as a testosterone level <12 nmol/L or testosterone replacement therapy. Partial testosterone deficiency was defined as normal testosterone levels with elevated luteinizing hormone (LH) levels >10 IU/L. Leydig-cell function was considered normal when testosterone and LH levels were normal without hormone substitution.

HSCT

The range of complications after busulfan-based myeloablative conditioning regimen largely overlaps with those observed after TBI, particularly for the risk of gonadal toxicity. However, many late effects such as metabolic syndrome, secondary cancer, height growth impairment, and cataracts are less frequent, leading to an overall lower burden of health conditions instead of an increased risk of permanent partial alopecia.26,30 

Chronic graft-versus-host disease (GVHD) is a graft immune response against host organs primarily mediated by T lymphocytes. The severity of chronic GVHD can vary from mild defects to severe organ injury and can affect several organs or systems. Pediatric chronic GVHD tends to be less frequent and severe than in adults.31,32 However, extensive data on chronic GVHD in children are lacking, resulting in limited understanding of disease occurrence and the impact on childhood survivors. Based on data from the LEA cohort, chronic GVHD affects ∼25% of patients who received transplantation and persists long-term in ∼20% of patients.33 The most frequently affected organs include the eyes, skin, mouth, and lungs. Severe forms of long-term chronic GVHD represent ∼20% of cases. These severe forms include a lung defect characterized by airflow obstruction called bronchiolitis obliterans. Alloimmune reaction results in bronchioles inflammation, fibrosis, and irreversible constriction of the terminal air passages. Transplantation during adolescence and second transplantation constitute risk factors of chronic GVHD. Chronic GVHD significantly alters the long-term physical and psychological aspects of quality of life.

CNS irradiation

Historically, CNS irradiation has resulted in a major improvement in cure rates for childhood ALL by efficiently preventing CNS relapse. Cranial or craniospinal fields were considered with doses commonly ranging from 12 to 24 Gy. To minimize long-term sequelae, CNS irradiation has been nearly eliminated from most modern ALL protocols, although patients with CNS relapse still require this therapy. The main long-term complications after CNS radiation include metabolic syndrome, precocious puberty, height growth retardation, neurocognitive impairments, and secondary brain, skin, and thyroid tumors.34 Metabolic syndrome observed after CNS irradiation is usually associated with obesity, thus suggesting a unique pathophysiology.35 The neuropsychological effects of CNS radiation have been extensively studied. The most severe impairments are observed in survivors treated with higher radiation doses (ie, 24-28 Gy).36-38 CNS irradiation impairs height growth, depending on the radiation dose and field.39,40 

Steroids

Osteonecrosis is the main long-term complication caused by the administration of high-dose corticosteroids.41,42 This complication particularly affects adolescents. Female sex is also a risk factor for osteonecrosis. The diagnosis is confirmed using magnetic resonance imaging (MRI) centered on the affected structure. Osteonecrosis frequently affects weight-bearing joints and multiple sites. Specific orthopedic management is difficult and can range from simple monitoring to joint replacement.43 This complication generally occurs during or within several months after treatment. It is frequently responsible for long-term abnormal joint mobility, pain, activity limitations, and altered quality of life.44,45 Genetic studies have shown an association between osteonecrosis and constitutional variants near genes involved in bone metabolism, glutamate metabolism, endothelial cell function, angiogenesis, and cellular migration such as glutamate receptor genes and NWD2 gene.46,47 Screening for osteonecrosis using serial MRI during leukemia therapy may be investigated to detect actionable early signs of this complication.

Anthracycline-related cardiomyopathy

Anthracyclines and anthraquinones are antileukemia drugs used to treat almost all children with acute leukemia. Survivors treated with these drugs are at risk of developing cardiomyopathy.48-50 A strong dose-dependent relationship has been observed between anthracycline chemotherapy exposure and risk of cardiomyopathy. Anthracycline and anthraquinone doses are usually calculated as the doxorubicin-equivalent dose, applying cardiotoxicity conversion ratios (Table 6).51 The risk is markedly increased when the cumulative anthracycline dose is >250 mg/m2.58 Younger children are at higher risk of anthracycline-induced cardiomyopathy.52,53 The probable additive cardiotoxic effect of combined anthracycline and TBI exposure is still not well documented.54,55 Variants in genes involved in anthracycline transportation and metabolism, generation of reactive oxygen species, and cardiac diseases have been associated with cardiotoxicity, thus suggesting a genetic predisposition to this condition. Anthracycline-related cardiomyopathy is a progressive disorder with a natural history that frequently involves an asymptomatic phase. During this phase, the asymptomatic cardiomyopathy can be diagnosed using echocardiography, which is recommended every 2 to 5 years after anthracycline exposure.59 Echocardiographic monitoring should include global longitudinal strain, which is a more sensitive technique to diagnose cardiac dysfunction.60 Discontinuation of follow-up after a given monitoring time is not recommended because cardiopathy can occur many years after anthracycline exposure.61 Cardio-protectants such as dexrazoxane may minimize the long-term cardiac toxicity56 without affecting relapse or secondary cancer risk.57 Patients who plan to engage in high-intensity exercise or who have symptomatic or asymptomatic cardiomyopathy should consult a cardiologist.

Table 6.

Anthracycline-related cardiomyopathy

Risk factors, pathophysiology, prevention, management, and current research efforts
Main risk factor: anthracycline and anthraquinone exposure51  The risk for cardiac dysfunction increases with anthracycline dose
Cardiotoxicity conversion ratios: doxorubicin: 1 (reference), daunorubicin: 0.6, epirubicin: 0.8, idarubicin: 5, mitoxantrone: 10.5 
Other risk factors52-55  Younger age at anthracycline and anthraquinone exposure
Female sex
Chest radiation exposure (TBI?) 
Pathophysiology48-50  Single cell myocytolysis → patchy myocardial necrosis → focal myocardial fibrosis → multifocal myocardial fibrosis (clinically overt congestive heart failure)
Genetic predisposition 
Clinical classification48-50  Acute cardiotoxicity (withing weeks of anthracycline and anthraquinone exposure; impaired myocardial contractility)
Early-onset chronic cardiotoxicity (<1 y after leukemia therapy completion; dilated/hypokinetic cardiomyopathy)
Late-onset chronic cardiotoxicity (>1 y after leukemia therapy completion; dilated/hypokinetic cardiomyopathy) 
Cardioprotection56,57  Limiting anthracycline/anthraquinone cumulated dose
Use of cardioprotectant drug: dexrazoxane
Use of liposome-encapsulated versions of anthracyclines 
Interventions for cardiomyopathy48-50  Early use of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, or beta-blockers (may prevent severe contractile dysfunction and improve cardiac function) 
Current research efforts49  Early detection of anthracycline-related cardiomyopathy
Polygenic risk scores
Patient-specific hiPSC-CMs 
Risk factors, pathophysiology, prevention, management, and current research efforts
Main risk factor: anthracycline and anthraquinone exposure51  The risk for cardiac dysfunction increases with anthracycline dose
Cardiotoxicity conversion ratios: doxorubicin: 1 (reference), daunorubicin: 0.6, epirubicin: 0.8, idarubicin: 5, mitoxantrone: 10.5 
Other risk factors52-55  Younger age at anthracycline and anthraquinone exposure
Female sex
Chest radiation exposure (TBI?) 
Pathophysiology48-50  Single cell myocytolysis → patchy myocardial necrosis → focal myocardial fibrosis → multifocal myocardial fibrosis (clinically overt congestive heart failure)
Genetic predisposition 
Clinical classification48-50  Acute cardiotoxicity (withing weeks of anthracycline and anthraquinone exposure; impaired myocardial contractility)
Early-onset chronic cardiotoxicity (<1 y after leukemia therapy completion; dilated/hypokinetic cardiomyopathy)
Late-onset chronic cardiotoxicity (>1 y after leukemia therapy completion; dilated/hypokinetic cardiomyopathy) 
Cardioprotection56,57  Limiting anthracycline/anthraquinone cumulated dose
Use of cardioprotectant drug: dexrazoxane
Use of liposome-encapsulated versions of anthracyclines 
Interventions for cardiomyopathy48-50  Early use of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, or beta-blockers (may prevent severe contractile dysfunction and improve cardiac function) 
Current research efforts49  Early detection of anthracycline-related cardiomyopathy
Polygenic risk scores
Patient-specific hiPSC-CMs 

hiPSC-CMs, human induced pluripotent stem cell–derived cardiomyocytes.

Transfusions

Patients who underwent blood product transfusion before 1970 to 1995 (depending on the virus and the country) are susceptible to potential HIV, hepatitis C virus, or hepatitis B virus infection.62 Advancements in the safety measures implemented throughout the blood transfusion process have now nearly eliminated the risk of viral contamination.

Iron overload is a common complication of red blood cell transfusions in survivors of childhood leukemia. Prevalence rates are higher for those who undergo HSCT and those who receive TBI.63,64 Other factors that contribute to the risk include older age at the time of HSCT and intensity of chemotherapy (eg, AML or high-risk ALL treatment). The long-term impact of iron overload in survivors of childhood leukemia is unknown, but it may exacerbate cardiac, hepatic, and metabolic toxicities. When iron overload is suspected (>20 red blood cell transfusions, ferritin levels >1000 ng/mL, and HSCT), iron liver content should be evaluated using T2∗ MRI. Confirmed iron overload can be efficiently treated using phlebotomy after hematological recovery (eg, 10 mL/kg with a maximum of 500 mL every 3 weeks for 1 year). Oral iron chelators such as deferasirox constitute an alternative for patients with phlebotomy contraindication (eg, anemia, cardiovascular instability, and pregnancy) or poor peripheral venous access.

Clinical case

A 10-and-a-half-year-old girl was diagnosed with KMT2A-rearranged ALL. She had a relapse on therapy at the age of 11 treated with second-line chemotherapy and HSCT. Transplantation conditioning involved TBI. Ovarian cryopreservation was performed before HSCT. Long-term follow-up included monitoring by a pediatric endocrinologist. She developed a premature ovarian insufficiency, necessitating hormonal treatment for puberty induction. By the age of 20, despite 12 months of regular and unprotected sexual intercourse with her partner, she had not achieved pregnancy. She faced various social challenges, including unemployment, which delayed access to medically assisted reproduction. This case demonstrates the role of patient clinical characteristics and environmental factors in the occurrence and management of long-term complications.

Age and sex

Age at HSCT and sex are major determinants of the risk of decreased growth velocity after HSCT, as described in the previous section of the manuscript. For young children who have undergone transplantation, growth velocity is often near-normal until pubertal age, although the peripubertal growth spurt is very frequently impaired.65,66 

Age at HSCT and sex have a major impact on gonadal toxicity after HSCT; 65% of girls aged <5 years at the time of HSCT had spontaneous menarche, whereas 85% of prepubertal girls aged >10 years at the time of HSCT suffered premature ovarian insufficiency requiring hormone treatment to induce puberty.28 In patients who received HSCT after puberty, gonadal toxicity was even higher.67 Careful evaluation by a pediatric endocrinologist is crucial to manage the complex interaction between abnormalities of puberty, gonadal dysfunction, growth hormone deficiency, and peripubertal growth spurt.

Infants with leukemia face heightened vulnerability primarily due to the aggressive nature of the disease, with leukemia relapse being the predominant cause of mortality in this population.68 Those who achieve long-term survival are often expected to be at higher risk of severe long-term complications. However, patients who received chemotherapy or HSCT during the first years of life during infancy did not show a significantly increased risk of most late sequelae compared with older children.68,69 Children aged <6 months at the time of diagnosis probably require special consideration because of an increased risk of growth failure, underweight, and thyroid dysfunction.

Osteonecrosis particularly affects adolescents. This may be attributed to a specific bone vulnerability due to epiphyseal closure, the procoagulant effects of sex hormones, and increased bone metabolic activity secondary to the peak of growth hormone and insulin-like growth factor 1 during puberty.44 

Underlying diseases

When compared with other survivors of leukemia, patients with Down syndrome have a higher risk of cataracts, hypothyroidism, obesity, and bone mineral density abnormalities.70 Most observed complications are at least partly related to the constitutional disease itself rather than leukemia therapy.

Patients with constitutional telomere disease receiving HSCT for leukemia are at high risk of late life-threatening complications, including pulmonary complications, liver failure, and microangiopathy.71 

Patients with ataxia telangiectasia and Li-Fraumeni syndrome are predisposed to secondary malignant neoplasms.72,73 Constitutional (including synonymous) and somatic TP53 variants are enriched in survivors with secondary cancers who do not necessarily fulfill the classic Li-Fraumeni syndrome criteria.74 Constitutional TP53 variants may identify patients at high risk of secondary malignant cancers that would benefit from adapted surveillance program. There is currently insufficient data to support reduction of treatment intensity in these patients, notably regarding HSCT conditioning. However, unnecessary radiation should be avoided, and alternative approaches (such as immunotherapy) should be further evaluated.

Children with Fanconi anemia who survive leukemia therapy and/or allogeneic HSCT are at high risk of a wide range of secondary malignant neoplasms, including vulvar, vaginal, and head and neck squamous cell carcinoma.75 Frequent gynecologic examinations, as well as head and neck examinations (eg, every 6 months), including careful oral cavity evaluations and flexible laryngoscopy should be performed in these patients. To minimize the long-term toxicity of HSCT, these patients receive specific low-intensity fludarabine-based conditioning regimen.

Modifiable risk factors

Leukemia survivorship shares several medical risks with smoking, alcohol consumption, unhealthy diet, and physical inactivity. Shared risks include cardiovascular complications, pulmonary disorders, and cancer development, which the survivors treated with HSCT are particularly susceptible to. Survivors include a substantial proportion of consistent smokers.76 The survivors of childhood cancers with healthy lifestyle and no cardiovascular risk factors are at reduced risk of late mortality,6 although it remains unclear whether these modifiable risk factors carry more than additive risks. Preventing smoking and alcohol use, as well as implementing physical activity and nutritional intervention are important aspects of health promotion within this population.

Socioeconomic determinants

Socioeconomic and racial parameters have been increasingly recognized as determinants of long-term health and loss to follow-up of survivors of childhood leukemia. Among survivors of childhood cancer, loss to follow-up rates were higher among patients from ethnic minority groups and those who resided in areas associated with lower socioeconomic status at the time of diagnosis.77 This may jeopardize their long-term survival, treatment-related health conditions, and quality of life. Neighborhood deprivation was also independently associated with overweight and obesity in survivors of childhood leukemia, particularly those in the highest quartile of the area deprivation index.78 Collecting comprehensive information on social determinants of health is important to target interventions to improve long-term follow-up and health among disadvantaged survivors of childhood leukemia.

Surveillance of patients at lower risk of long-term complications

Patients at lower risk of long-term complications are those treated with chemotherapy alone, without intensive therapy for relapse, CNS irradiation, or HSCT. The cumulative dose of anthracyclines should also be taken into account, with lowest risk among those treated with a cumulative anthracycline dose of <100 mg/m2.58 The first 10 years after diagnosis is the period of greatest risk for complications.79 After 10 years postdiagnosis, cardiovascular risk factors and cardiotoxicity are the most frequent and severe long-term complications.79 However, the optimal modalities of long-term follow-up in these patients remain undetermined.

Organization of long-term follow-up and transition

Organization of long-term follow-up and transition practices are widely heterogeneous and largely depend on the community setting or country-wide health care systems.80 American and European guidelines have been developed (eg, Children Oncology Group and International Guideline Harmonization Group/PanCare guidelines) and inform risk stratification and follow-up. The French leukemia survivor follow-up program LEA consists of dedicated follow-up visits organized in pediatric centers. The visits start 1 year after leukemia therapy and repeat every 2 to 4 years thereafter. During the initial visit, the risk of long-term complications is stratified based on factors such as leukemia type, therapeutic exposure, and age at the time of evaluation. Every visit includes a clinical evaluation with appropriate laboratory analysis and imaging. Specific questionnaires are used to evaluate the patient’s quality of life as well as the socioeconomic factors of the patient and their family. Patients may be referred to pediatric or adult subspecialists for diagnosis and treatment of complications (eg, cataracts and cardiomyopathy). Patients at risk of multiple late complications (eg, patients who received irradiation or transplantation) greatly benefit from specialized follow-up centers that can provide the multiple expertise needed for their management. Transition to an adult-focused provider and environment is a standard in most settings.81 However, transition is challenging and several barriers have been identified,82 including social issues (eg, health insurance that does not cover long-term follow-up), the lack of adult providers familiar with survivorship care, and insufficient knowledge and empowerment among survivors.

Surveillance of investigational or recently approved therapies

Long-term follow-up of children and adolescents treated with investigational or recently approved therapies is crucial. However, the lack of long-term safety data may result in inconsistent screening practices. It is essential to establish a comprehensive follow-up program for these patients. Tyrosine kinase inhibitors, primarily imatinib, are administered to children and adolescents with chronic myeloid leukemia and Philadelphia-positive ALL. Several safety concerns regarding these drugs have been identified, including growth alterations and bone metabolism abnormalities.83 The impact on growth is most notable in children who initiate imatinib therapy before puberty.84 Recent reports have indicated that survivors with prolonged exposure to tyrosine kinase inhibitors may be at risk of specific pulmonary outcomes and pericardial effusion, which are typically reversible. There is currently no available long-term safety data for chimeric antigen receptor (CAR) T cells and T-cell engagers (eg, blinatumomab), which are increasingly used to treat children with ALL. It is crucial to monitor patients treated with immunotherapies, especially those who experience neurotoxicity. CD19-directed CAR T cells may be associated with prolonged B-cell aplasia with hypogammaglobulinemia requiring immunoglobulin replacement therapy. Recently, T-cell malignancies, including CAR-positive lymphoma, in patients treated with CD19-directed CAR T cells were reported to be under investigation.85 Treosulfan is an alkylating agent that is increasingly used in conditioning regimens for HSCT. Regarding long-term effects, data suggest that treosulfan-based conditioning may have a limited gonadotoxicity compared with busulfan.86 The follow-up program of patients treated with treosulfan should be similar to that proposed for patients who received other transplantations.

Because therapy and cure rates of childhood leukemia have made significant headway, health care objectives have expanded to improve long-term health and quality of life. The implementation of comprehensive follow-up programs plays a crucial role in childhood leukemia survivor care, enabling early detection and treatment of long-term complications. Risk-adapted therapies have reduced the use of HSCT, radiotherapy, and high-intensity chemotherapy, which are associated with increased long-term toxicity. However, these approaches remain essential to cure patients with aggressive disease. Innovative therapies (eg, antibody-drug conjugates and immunotherapy) are expected to have a better long-term safety profile, although diligent long-term toxicity monitoring is necessary. Advances in understanding genetic susceptibility to long-term complications hold promise regarding tailored therapeutic strategies for leukemia treatment and optimizing follow-up programs.

The authors thank Pascal Auquier, the LEA investigators, and the patients and families involved in the LEA survivorship program, Sophie Beliard and Blandine Courbière for insightful discussions on metabolic syndrome and gonadal toxicity, and Sandra Moore for revising the paper.

Contribution: P.S. and G.M. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Paul Saultier, Department of Pediatric Hematology, Immunology and Oncology, La Timone Children’s Hospital, APHM and Aix Marseille Université, 264 rue Saint-Pierre, 13385 Marseille, France; email: paul.saultier@ap-hm.fr.

1.
Oeffinger
KC
,
Mertens
AC
,
Sklar
CA
, et al
.
Chronic health conditions in adult survivors of childhood cancer
.
N Engl J Med
.
2006
;
355
(
15
):
1572
-
1582
.
2.
Mertens
AC
,
Liu
Q
,
Neglia
JP
, et al
.
Cause-specific late mortality among 5-year survivors of childhood cancer: the Childhood Cancer Survivor Study
.
J Natl Cancer Inst
.
2008
;
100
(
19
):
1368
-
1379
.
3.
Mody
R
,
Li
S
,
Dover
DC
, et al
.
Twenty-five–year follow-up among survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study
.
Blood
.
2008
;
111
(
12
):
5515
-
5523
.
4.
Gurney
JG
,
Ojha
RP
,
Ness
KK
, et al
.
Abdominal aortic calcification in young adult survivors of childhood acute lymphoblastic leukemia: results from the St. Jude Lifetime Cohort study
.
Pediatr Blood Cancer
.
2012
;
59
(
7
):
1307
-
1309
.
5.
Grundy
SM
,
Cleeman
JI
,
Daniels
SR
, et al
.
Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement
.
Circulation
.
2005
;
112
(
17
):
2735
-
2752
.
6.
Dixon
SB
,
Chen
Y
,
Yasui
Y
, et al
.
Reduced morbidity and mortality in survivors of childhood acute lymphoblastic leukemia: a report from the childhood cancer survivor study
.
J Clin Orthod
.
2020
;
38
(
29
):
3418
-
3429
.
7.
Oudin
C
,
Simeoni
M-C
,
Sirvent
N
, et al
.
Prevalence and risk factors of the metabolic syndrome in adult survivors of childhood leukemia
.
Blood
.
2011
;
117
(
17
):
4442
-
4448
.
8.
Oudin
C
,
Berbis
J
,
Bertrand
Y
, et al
.
Prevalence and characteristics of metabolic syndrome in adults from the French childhood leukemia survivors’ cohort: a comparison with controls from the French population
.
Haematologica
.
2018
;
103
(
4
):
645
-
654
.
9.
Saultier
P
,
Auquier
P
,
Bertrand
Y
, et al
.
Metabolic syndrome in long-term survivors of childhood acute leukemia treated without hematopoietic stem cell transplantation: an L.E.A. study
.
Haematologica
.
2016
;
101
(
12
):
1603
-
1610
.
10.
Visentin
S
,
Michel
G
,
Oudin
C
, et al
.
Lipodystrophy-like features after total body irradiation among survivors of childhood acute leukemia
.
Endocr Connect
.
2019
;
8
:
349
-
359
.
11.
Lopez
R
,
Plat
G
,
Bertrand
Y
, et al
.
Testosterone deficiency in men surviving childhood acute leukemia after treatment with hematopoietic stem cell transplantation or testicular radiation: an L.E.A. study
.
Bone Marrow Transplant
.
2021
;
56
(
6
):
1422
-
1425
.
12.
Romano
A
,
Del Vescovo
E
,
Rivetti
S
, et al
.
Biomarkers predictive of metabolic syndrome and cardiovascular disease in childhood cancer survivors
.
J Pers Med
.
2022
;
12
(
6
):
880
.
13.
Pluimakers
VG
,
van Santen
SS
,
Fiocco
M
, et al
.
Can biomarkers be used to improve diagnosis and prediction of metabolic syndrome in childhood cancer survivors? A systematic review
.
Obes Rev
.
2021
;
22
(
11
):
e13312
.
14.
Wilson
CL
,
Liu
W
,
Yang
JJ
, et al
.
Genetic and clinical factors associated with obesity among adult survivors of childhood cancer: a report from the St. Jude Lifetime Cohort
.
Cancer
.
2015
;
121
(
13
):
2262
-
2270
.
15.
Eipel
OT
,
Németh
K
,
Török
D
, et al
.
The glucocorticoid receptor gene polymorphism N363S predisposes to more severe toxic side effects during pediatric acute lymphoblastic leukemia (ALL) therapy
.
Int J Hematol
.
2013
;
97
(
2
):
216
-
222
.
16.
Sapkota
Y
,
Qiu
W
,
Dixon
SB
, et al
.
Genetic risk score enhances the risk prediction of severe obesity in adult survivors of childhood cancer
.
Nat Med
.
2022
;
28
(
8
):
1590
-
1598
.
17.
Whitlock
JA
,
Greer
JP
,
Lukens
JN
.
Epipodophyllotoxin-related leukemia. Identification of a new subset of secondary leukemia
.
Cancer
.
1991
;
68
(
3
):
600
-
604
.
18.
Yavvari
S
,
Makena
Y
,
Sukhavasi
S
,
Makena
MR
.
Large population analysis of secondary cancers in pediatric leukemia survivors
.
Children (Basel)
.
2019
;
6
(
12
):
130
.
19.
Schiffman
JD
,
Fisher
PG
,
Gibbs
P
.
Early detection of cancer: past, present, and future
.
Am Soc Clin Oncol Educ Book
.
2015
:
57
-
65
.
20.
Peters
C
,
Dalle
J-H
,
Locatelli
F
, et al
.
Total body irradiation or chemotherapy conditioning in childhood ALL: a multinational, randomized, noninferiority phase III study
.
J Clin Oncol
.
2021
;
39
(
4
):
295
-
307
.
21.
Friedman
DL
,
Rovo
A
,
Leisenring
W
, et al
.
Increased risk of breast cancer among survivors of allogeneic hematopoietic cell transplantation: a report from the FHCRC and the EBMT-Late Effect Working Party
.
Blood
.
2008
;
111
(
2
):
939
-
944
.
22.
Moskowitz
CS
,
Chou
JF
,
Wolden
SL
, et al
.
Breast cancer after chest radiation therapy for childhood cancer
.
J Clin Oncol
.
2014
;
32
(
21
):
2217
-
2223
.
23.
Horwitz
M
,
Auquier
P
,
Barlogis
V
, et al
.
Incidence and risk factors for cataract after haematopoietic stem cell transplantation for childhood leukaemia: an LEA study
.
Br J Haematol
.
2015
;
168
(
4
):
518
-
525
.
24.
Alloin
A-L
,
Barlogis
V
,
Auquier
P
, et al
.
Prevalence and risk factors of cataract after chemotherapy with or without central nervous system irradiation for childhood acute lymphoblastic leukaemia: an LEA study
.
Br J Haematol
.
2014
;
164
(
1
):
94
-
100
.
25.
Oudin
C
,
Auquier
P
,
Bertrand
Y
, et al
.
Late thyroid complications in survivors of childhood acute leukemia
.
An L.E.A. study. Haematologica
.
2016
;
101
(
6
):
747
-
756
.
26.
Giorgiani
G
,
Bozzola
M
,
Locatelli
F
, et al
.
Role of busulfan and total body irradiation on growth of prepubertal children receiving bone marrow transplantation and results of treatment with recombinant human growth hormone
.
Blood
.
1995
;
86
(
2
):
825
-
831
.
27.
Bruzzi
P
,
Predieri
B
,
Corrias
A
, et al
.
Final height and body mass index in adult survivors of childhood acute lymphoblastic leukemia treated without cranial radiotherapy: a retrospective longitudinal multicenter Italian study
.
BMC Pediatr
.
2014
;
14
:
236
.
28.
Chabut
M
,
Schneider
P
,
Courbiere
B
, et al
.
Ovarian function and spontaneous pregnancy after hematopoietic stem cell transplantation for leukemia before puberty: an L.E.A. cohort study
.
Transplant Cell Ther
.
2023
;
29
(
6
):
378.e1
-
378.e9
.
29.
Courbiere
B
,
Drikes
B
,
Grob
A
, et al
.
The uterine volume is dramatically decreased after hematopoietic stem cell transplantation during childhood regardless of the conditioning regimen
.
Fertil Steril
.
2023
;
119
(
4
):
663
-
672
.
30.
Bernard
F
,
Auquier
P
,
Herrmann
I
, et al
.
Health status of childhood leukemia survivors who received hematopoietic cell transplantation after BU or TBI: an LEA study
.
Bone Marrow Transplant
.
2014
;
49
(
5
):
709
-
716
.
31.
Inagaki
J
,
Moritake
H
,
Nishikawa
T
, et al
.
Long-term morbidity and mortality in children with chronic graft-versus-host disease classified by national institutes of health consensus criteria after allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2015
;
21
(
11
):
1973
-
1980
.
32.
Sobkowiak-Sobierajska
A
,
Lindemans
C
,
Sykora
T
, et al
.
Management of chronic Graft-vs.-Host disease in children and adolescents with ALL: present status and model for a personalised management plan
.
Front Pediatr
.
2022
;
10
:
808103
.
33.
Saultier
P
,
Michel
G
,
Sirvent
A
, et al
.
Chronic graft-versus-host disease in long-term survivors of childhood leukemia [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
3762
.
34.
Benadiba
J
,
Michel
G
,
Auquier
P
, et al
.
Health status and quality of life of long-term survivors of childhood acute leukemia: the impact of central nervous system irradiation
.
J Pediatr Hematol Oncol
.
2015
;
37
(
2
):
109
-
116
.
35.
Sklar
CA
,
Mertens
AC
,
Walter
A
, et al
.
Changes in body mass index and prevalence of overweight in survivors of childhood acute lymphoblastic leukemia: role of cranial irradiation
.
Med Pediatr Oncol
.
2000
;
35
(
2
):
91
-
95
.
36.
Waber
DP
,
Turek
J
,
Catania
L
, et al
.
Neuropsychological outcomes from a randomized trial of triple intrathecal chemotherapy compared with 18 Gy cranial radiation as CNS treatment in acute lymphoblastic leukemia: findings from Dana-Farber Cancer Institute ALL Consortium Protocol 95-01
.
J Clin Oncol
.
2007
;
25
(
31
):
4914
-
4921
.
37.
Mulhern
RK
,
Kovnar
E
,
Langston
J
, et al
.
Long-term survivors of leukemia treated in infancy: factors associated with neuropsychologic status
.
J Clin Oncol
.
1992
;
10
(
7
):
1095
-
1102
.
38.
Halberg
FE
,
Kramer
JH
,
Moore
IM
,
Wara
WM
,
Matthay
KK
,
Ablin
AR
.
Prophylactic cranial irradiation dose effects on late cognitive function in children treated for acute lymphoblastic leukemia
.
Int J Radiat Oncol Biol Phys
.
1992
;
22
(
1
):
13
-
16
.
39.
Hata
M
,
Ogino
I
,
Aida
N
, et al
.
Prophylactic cranial irradiation of acute lymphoblastic leukemia in childhood: outcomes of late effects on pituitary function and growth in long-term survivors
.
Int J Cancer
.
2001
;
96
(
suppl
):
117
-
124
.
40.
Sklar
C
,
Mertens
A
,
Walter
A
, et al
.
Final height after treatment for childhood acute lymphoblastic leukemia: comparison of no cranial irradiation with 1800 and 2400 centigrays of cranial irradiation
.
J Pediatr
.
1993
;
123
(
1
):
59
-
64
.
41.
Kadan-Lottick
NS
,
Dinu
I
,
Wasilewski-Masker
K
, et al
.
Osteonecrosis in adult survivors of childhood cancer: a report from the childhood cancer survivor study
.
J Clin Oncol
.
2008
;
26
(
18
):
3038
-
3045
.
42.
te Winkel
ML
,
Pieters
R
,
Hop
WCJ
, et al
.
Prospective study on incidence, risk factors, and long-term outcome of osteonecrosis in pediatric acute lymphoblastic leukemia
.
J Clin Oncol
.
2011
;
29
(
31
):
4143
-
4150
.
43.
Heneghan
MB
,
Rheingold
SR
,
Li
Y
, et al
.
Treatment of Osteonecrosis in Children and Adolescents With Acute Lymphoblastic Leukemia
.
Clin Lymphoma Myeloma Leuk
.
2016
;
16
(
4
):
223
-
229.e2
.
44.
Kunstreich
M
,
Kummer
S
,
Laws
H-J
,
Borkhardt
A
,
Kuhlen
M
.
Osteonecrosis in children with acute lymphoblastic leukemia
.
Haematologica
.
2016
;
101
(
11
):
1295
-
1305
.
45.
Huault
A
,
Michel
G
,
Charon
V
, et al
.
Symptomatic osteonecrosis in French survivors of childhood and adolescent leukemia: a clinical and MRI study of LEA cohort
.
Pediatr Hematol Oncol
.
2023
;
40
(
5
):
458
-
474
.
46.
Karol
SE
,
Yang
W
,
Van Driest
SL
, et al
.
Genetics of glucocorticoid-associated osteonecrosis in children with acute lymphoblastic leukemia
.
Blood
.
2015
;
126
(
15
):
1770
-
1776
.
47.
Yang
W
,
Devidas
M
,
Liu
Y
, et al
.
Genetics of osteonecrosis in pediatric acute lymphoblastic leukemia and general populations
.
Blood
.
2021
;
137
(
11
):
1550
-
1552
.
48.
Bychowski
J
,
Sobiczewski
W
.
Current perspectives of cardio-oncology: epidemiology, adverse effects, pre-treatment screening and prevention strategies
.
Cancer Med
.
2023
;
12
(
13
):
14545
-
14555
.
49.
Cejas
RB
,
Petrykey
K
,
Sapkota
Y
,
Burridge
PW
.
Anthracycline toxicity: light at the end of the tunnel?
.
Annu Rev Pharmacol Toxicol
.
2024
;
64
:
115
-
134
.
50.
Cardinale
D
,
Iacopo
F
,
Cipolla
CM
.
Cardiotoxicity of anthracyclines
.
Front Cardiovasc Med
.
2020
;
7
:
26
.
51.
Feijen
EAM
,
Leisenring
WM
,
Stratton
KL
, et al
.
Derivation of anthracycline and anthraquinone equivalence ratios to doxorubicin for late-onset cardiotoxicity
.
JAMA Oncol
.
2019
;
5
(
6
):
864
-
871
.
52.
Mulrooney
DA
,
Yeazel
MW
,
Kawashima
T
, et al
.
Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: retrospective analysis of the Childhood Cancer Survivor Study cohort
.
BMJ
.
2009
;
339
:
b4606
.
53.
van der Pal
HJ
,
van Dalen
EC
,
Hauptmann
M
, et al
.
Cardiac function in 5-year survivors of childhood cancer: a long-term follow-up study
.
Arch Intern Med
.
2010
;
170
(
14
):
1247
-
1255
.
54.
Uderzo
C
,
Pillon
M
,
Corti
P
, et al
.
Impact of cumulative anthracycline dose, preparative regimen and chronic graft-versus-host disease on pulmonary and cardiac function in children 5 years after allogeneic hematopoietic stem cell transplantation: a prospective evaluation on behalf of the EBMT Pediatric Diseases and Late Effects Working Parties
.
Bone Marrow Transplant
.
2007
;
39
(
11
):
667
-
675
.
55.
Barlogis
V
,
Auquier
P
,
Bertrand
Y
, et al
.
Late cardiomyopathy in childhood acute myeloid leukemia survivors: a study from the L.E.A. program
.
Haematologica
.
2015
;
100
(
5
):
e186
-
189
.
56.
Chow
EJ
,
Aggarwal
S
,
Doody
DR
, et al
.
Dexrazoxane and long-term heart function in survivors of childhood cancer
.
J Clin Oncol
.
2023
;
41
(
12
):
2248
-
2257
.
57.
Chow
EJ
,
Aplenc
R
,
Vrooman
LM
, et al
.
Late health outcomes after dexrazoxane treatment: a report from the Children’s Oncology Group
.
Cancer
.
2022
;
128
(
4
):
788
-
796
.
58.
Blanco
JG
,
Sun
C-L
,
Landier
W
, et al
.
Anthracycline-related cardiomyopathy after childhood cancer: role of polymorphisms in carbonyl reductase genes--a report from the Children’s Oncology Group
.
J Clin Oncol
.
2012
;
30
(
13
):
1415
-
1421
.
59.
Armenian
SH
,
Hudson
MM
,
Mulder
RL
, et al
.
Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group
.
Lancet Oncol
.
2015
;
16
(
3
):
e123
-
136
.
60.
Armstrong
GT
,
Joshi
VM
,
Ness
KK
, et al
.
Comprehensive echocardiographic detection of treatment-related cardiac dysfunction in adult survivors of childhood cancer: results from the St. Jude lifetime cohort study
.
J Am Coll Cardiol
.
2015
;
65
(
23
):
2511
-
2522
.
61.
Lipshultz
SE
,
Adams
MJ
,
Colan
SD
, et al
.
Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention, and research directions: a scientific statement from the American Heart Association
.
Circulation
.
2013
;
128
(
17
):
1927
-
1995
.
62.
Zou
S
,
Stramer
SL
,
Dodd
RY
.
Donor testing and risk: current prevalence, incidence, and residual risk of transfusion-transmissible agents in US allogeneic donations
.
Transfus Med Rev
.
2012
;
26
(
2
):
119
-
128
.
63.
Sirvent
A
,
Auquier
P
,
Oudin
C
, et al
.
Prevalence and risk factors of iron overload after hematopoietic stem cell transplantation for childhood acute leukemia: a LEA study
.
Bone Marrow Transplant
.
2017
;
52
(
1
):
80
-
87
.
64.
Nottage
K
,
Gurney
JG
,
Smeltzer
M
,
Castellanos
M
,
Hudson
MM
,
Hankins
JS
.
Trends in transfusion burden among long-term survivors of childhood hematological malignancies
.
Leuk Lymphoma
.
2013
;
54
(
8
):
1719
-
1723
.
65.
Bernard
F
,
Bordigoni
P
,
Simeoni
M-C
, et al
.
Height growth during adolescence and final height after haematopoietic SCT for childhood acute leukaemia: the impact of a conditioning regimen with BU or TBI
.
Bone Marrow Transplant
.
2009
;
43
(
8
):
637
-
642
.
66.
Chow
EJ
,
Friedman
DL
,
Yasui
Y
, et al
.
Decreased adult height in survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study
.
J Pediatr
.
2007
;
150
(
4
):
370
-
375.e1
.
67.
Borgmann-Staudt
A
,
Rendtorff
R
,
Reinmuth
S
, et al
.
Fertility after allogeneic haematopoietic stem cell transplantation in childhood and adolescence
.
Bone Marrow Transplant
.
2012
;
47
(
2
):
271
-
276
.
68.
Vrooman
LM
,
Millard
HR
,
Brazauskas
R
, et al
.
Survival and late effects after allogeneic hematopoietic cell transplantation for hematologic malignancy at less than three years of age
.
Biol Blood Marrow Transplant
.
2017
;
23
(
8
):
1327
-
1334
.
69.
Gandemer
V
,
Bonneau
J
,
Oudin
C
, et al
.
Late effects in survivors of infantile acute leukemia: a study of the L.E.A program
.
Blood Cancer J
.
2017
;
7
(
1
):
e518
.
70.
Goldsby
RE
,
Stratton
KL
,
Raber
S
, et al
.
Long-term sequelae in survivors of childhood leukemia with Down syndrome: a childhood cancer survivor study report
.
Cancer
.
2018
;
124
(
3
):
617
-
625
.
71.
Fioredda
F
,
Iacobelli
S
,
Korthof
ET
, et al
.
Outcome of haematopoietic stem cell transplantation in dyskeratosis congenita
.
Br J Haematol
.
2018
;
183
(
1
):
110
-
118
.
72.
Barnett
GC
,
West
CML
,
Dunning
AM
, et al
.
Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype
.
Nat Rev Cancer
.
2009
;
9
(
2
):
134
-
142
.
73.
Qian
M
,
Cao
X
,
Devidas
M
, et al
.
TP53 germline variations influence the predisposition and prognosis of B-cell acute lymphoblastic leukemia in children
.
J Clin Oncol
.
2018
;
36
(
6
):
591
-
599
.
74.
Sherborne
AL
,
Lavergne
V
,
Yu
K
, et al
.
Somatic and Germline TP53 alterations in second malignant neoplasms from pediatric cancer survivors
.
Clin Cancer Res
.
2017
;
23
(
7
):
1852
-
1861
.
75.
Kahn
JM
,
Brazauskas
R
,
Tecca
HR
, et al
.
Subsequent neoplasms and late mortality in children undergoing allogeneic transplantation for nonmalignant diseases
.
Blood Adv
.
2020
;
4
(
9
):
2084
-
2094
.
76.
Gibson
TM
,
Liu
W
,
Armstrong
GT
, et al
.
Longitudinal smoking patterns in survivors of childhood cancer: an update from the Childhood Cancer Survivor Study
.
Cancer
.
2015
;
121
(
22
):
4035
-
4043
.
77.
Puthenpura
V
,
Ji
L
,
Xu
X
, et al
.
Loss to follow-up of minorities, adolescents, and young adults on clinical trials: A report from the Children’s Oncology Group
.
Cancer
.
2023
;
129
(
10
):
1547
-
1556
.
78.
Oluyomi
A
,
Aldrich
KD
,
Foster
KL
, et al
.
Neighborhood deprivation index is associated with weight status among long-term survivors of childhood acute lymphoblastic leukemia
.
J Cancer Surviv
.
2021
;
15
(
5
):
767
-
775
.
79.
Saultier
P
,
El Riachy
N
,
Bertrand
Y
, et al
.
Cardiac and vascular risks are the essential parameters for long-term surveillance of lower risk childhood acute leukemia survivors [abstract]
.
Blood
.
2022
;
140
(
suppl 1
):
2187
-
2189
.
80.
Oeffinger
KC
,
Argenbright
KE
,
Levitt
GA
, et al
.
Models of cancer survivorship health care: moving forward
.
Am Soc Clin Oncol Educ Book
.
2014
:
205
-
213
.
81.
Freyer
DR
.
Transition of care for young adult survivors of childhood and adolescent cancer: rationale and approaches
.
J Clin Oncol
.
2010
;
28
(
32
):
4810
-
4818
.
82.
Otth
M
,
Denzler
S
,
Koenig
C
,
Koehler
H
,
Scheinemann
K
.
Transition from pediatric to adult follow-up care in childhood cancer survivors-a systematic review
.
J Cancer Surviv
.
2021
;
15
(
1
):
151
-
162
.
83.
Millot
F
,
Guilhot
J
,
Baruchel
A
, et al
.
Growth deceleration in children treated with imatinib for chronic myeloid leukaemia
.
Eur J Cancer
.
2014
;
50
(
18
):
3206
-
3211
.
84.
Shima
H
,
Tokuyama
M
,
Tanizawa
A
, et al
.
Distinct impact of imatinib on growth at prepubertal and pubertal ages of children with chronic myeloid leukemia
.
J Pediatr
.
2011
;
159
(
4
):
676
-
681
.
85.
FDA investigating serious risk of T-cell malignancy following BCMA-directed or CD19-directed autologous chimeric antigen receptor (CAR) T cell immunotherapies
. Accessed 31 December 2023. https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/fda-investigating-serious-risk-t-cell-malignancy-following-bcma-directed-or-cd19-directed-autologous.
86.
Faraci
M
,
Diesch
T
,
Labopin
M
, et al
.
Gonadal function after busulfan compared with treosulfan in children and adolescents undergoing allogeneic hematopoietic stem cell transplant
.
Biol Blood Marrow Transplant
.
2019
;
25
(
9
):
1786
-
1791
.
Sign in via your Institution