Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Received: 1 October 2020 Revised: 27 October 2020 Accepted: 14 November 2020

DOI: 10.1002/pbc.28836 Pediatric


Blood &
Cancer The American Society of
Pediatric Hematology/Oncology
O N C O LO G Y: R E S E A R C H A RT I C L E

Clinical prognostic factors in pediatric adrenocortical tumors:


A meta-analysis

Elisa Zambaiti1 Miriam Duci1 Federica De Corti1 Piergiorgio Gamba1


Patrizia Dall’Igna2 Filippo Ghidini1 Calogero Virgone1

1
Department of Women’s and Children’s
Health, University Hospital of Padua, Padua, Abstract
Italy
Pediatric adrenocortical tumors (ACT) are rare and sometimes aggressive malignan-
2
Department of Emergencies and Organ
cies, but there is no consensus on the outcome predictors in children. A systematic
Transplantation, Azienda
Ospedaliero-Universitaria Consorziale search of MEDLINE, SCOPUS, Web of Science, and the Cochrane Library for studies
Ospedale Pediatrico Giovanni XXIII, Bari, Italy
from 1994 to 2020 about pediatric ACT was performed. In 42 studies, 1006 patients,
Correspondence aged 0-18 years, were included. The meta-analyses resulted in the following predictors
Calogero Virgone, Department of Women’s of better outcome: age <4 years (P < .00001), nonsecreting tumors (P = .004), complete
and Children’s Health, University Hospital of
Padua, Via Giustiniani 3, 35128 Padua, Italy. surgical resection (P < .00001), tumor volume (P < .0001), tumor weight (P < .00001),
Email: calogero.virgone@unipd.it tumor maximum diameter (P = .0009), and Stage I disease (P < .00001). Moreover,
patients affected by Cushing syndrome showed a worse outcome (P < .0001). Inter-
national prospective studies should be implemented to standardize clinical prognostic
factors evaluation, together with pathological scores, in the stratification of pediatric
ACT.

KEYWORDS
adrenocortical carcinoma, adrenocortical tumors, children, meta-analysis, prognosis study, prog-
nostic factors, systematic review

1 INTRODUCTION As a matter of fact, children with ACC seem to have a better out-
come when compared with adults, and some authors attributed this
Pediatric adrenocortical tumors (ACTs) are rare and sometimes aggres- finding to an overestimation of malignancy based on the use of adult
sive endocrine malignancies, frequently associated with Li-Fraumeni scores, which may be not fully applicable to the pediatric population.3,4
syndrome, a familial cancer predisposition disorder caused by germline The Wieneke index has been reported to have a prognostic value
mutations in the tumor suppressor gene TP53, and account for approx- more reliable in comparison with adult scores,5,6 and lately the five-
imately 0.38% of all childhood cancer cases.1 ACT develop from the item score7 was described to be useful when stratifying patients
adrenal cortex, with an estimated incidence of one per million each with ACT.
year, and comprise benign adrenocortical adenomas (ACA) and highly In this scenario, it is still difficult to establish which patients may
malignant adrenocortical carcinomas (ACC), whose pathogenesis is benefit from a more aggressive medical approach after surgery, and the
incompletely understood. Patients with ACC generally have a poor use of pediatric pathology scores could be a valid option if considered
clinical outcome, because there is no effective therapy for advanced together with clinical risk factors, as it happens in the management of
and metastatic forms, accounting for a 5-year overall survival of less other pediatric tumors as neuroblastoma or rhabdomyosarcoma.
than 40%.2 On the other hand, ACA is associated with excellent prog- In the past, various clinical features were variably reported to asso-
nosis, but only about 20% of pediatric ACTs are classified as ACA. ciate with a poor outcome: age >4 years, volume, weight, size, Cushing
syndrome, R1 resections, initial biopsy, and stage are the clinical fea-
Abbreviations: ACA, adrenocortical adenoma; ACC, adrenocortical carcinoma; ACT,
tures more commonly taken into account to define risk stratification.8,9
adrenocortical tumor; CI, confidence interval; RR, risk ratio

Pediatr Blood Cancer. 2021;68:e28836. wileyonlinelibrary.com/journal/pbc © 2020 Wiley Periodicals LLC 1 of 9


https://doi.org/10.1002/pbc.28836
2 of 9 ZAMBAITI ET AL .

Unfortunately, there is no consensus on which clinical features may be 2.3 Summary measures, synthesis of the results,
determinant to predict the outcome in pediatric ACT, as most of the and risk of bias
features described above did correlate with prognosis in a univariate
analysis but were not confirmed at the multivariate analysis.8 The clinical prognostic factors analyzed in this review were age, sex,
tumor secreting status, tumor volume, tumor weight, tumor maxi-
mum diameter, surgical results, and stage. Age greater than or less
2 METHODS
than 4 years was considered according to previous reports, which
showed that children under 4 years of age usually show a favorable
2.1 Protocol, eligibility criteria, information
outcome. Indeed, distant metastases are observed at a higher rate in
sources, and search
adolescents.7,8
Secretion pattern has been sporadically reported to associate with
This review was performed according to an a priori designed protocol
outcome: Cushing syndrome and nonsecreting tumors were reported
and recommended for systematic reviews and meta-analysis.10–12
to have worse prognosis.2
MEDLINE, SCOPUS, Web of Science, and the Cochrane Library,
Cut-off values for tumor volume (200 cm3 ), for tumor weight (100 or
including the Cochrane Database of Systematic Reviews (CDSR),
400 g), and for tumor maximum diameter (<5 cm, between 5 and 10 cm,
Database of Abstracts of Reviews of Effects (DARE), and the Cochrane
>10 cm) were established in previous literature data.1,8,9 Although size
Central Register of Controlled Trials (CENTRAL), were searched elec-
in the ACT literature has been described with different units of mea-
tronically in April 2020 utilizing combinations of relevant medical
surement, the evidence that larger tumors may present a dismal out-
subject heading (MeSH) terms, keywords, and word variants for
come at higher rates is widely recognized.
“adrenocortical tumors,” “adrenocortical adenoma,” “adrenocortical
Surgical results were defined as complete resection or incom-
carcinoma,” “prognostic factors,” and “children” (Supporting Informa-
plete resection (microscopic or macroscopic residuals) in patients who
tion S1). The search and selection criteria were restricted to English
underwent surgery on the primary tumor. A complete resection at
language. Reference lists of relevant articles and reviews were manu-
diagnosis is considered to be a major prognostic factor, since ACTs
ally searched for additional reports. The Preferred Reporting Items for
are localized in greater part. Stage was assessed on the basis of COG
Systematic Reviews and Meta-Analyses (PRISMA-P) guidelines13 were
staging system for ACT proposed by Sandrini et al15 and later mod-
followed.
ified by Michalkiewicz et al2 and Ribeiro et al,1 as it represents the
The study was registered with the PROSPERO database (registra-
most currently used worldwide: studies in which stage assessment
tion number CRD42020180970).
was performed with different systems were not included into this
analysis.
2.2 Study selection, data collection, and data The outcome analyzed in this systematic review was the reported
items patient’s status (alive or dead).

Studies were assessed according to the following criteria: population,


outcome, clinical features, and tumor features. Inclusion criteria
were: studies reporting pediatric cases of ACTs with available data 2.4 Statistical analysis
on treatment, histology, and follow up; publication span including
the last 25 years (1994-2020). Four authors, divided in two groups Overall, we evaluated separately the association between eight clinical
(Filippo Ghidini, Calogero Virgone and Elisa Zambaiti, Miriam Duci) prognostic factors and the outcome for a total of 17 separated meta-
reviewed all abstracts independently. Agreement about potential analyses.
relevance was reached by consensus, and full-text copies of those Data were analyzed by using Review Manager (RevMan, Version
articles were obtained. The same reviewers independently extracted 5.3, Cochrane Collaboration). For categorical variables risk ratio (RR)
relevant data regarding study characteristics and ACT features with 95% confidence intervals (CIs) were generated. We performed
and outcomes. Case reports or case series without outcome data descriptive statistics, and calculated the pooled RR and 95% CI for
and/or clinical data were excluded. Inconsistencies were discussed each prognostic factor that was studied in more than one different
by the reviewers and consensus reached. If more than one study was study. The RRs were pooled using the fixed-effects model. The hetero-
published for the same cohort with identical end points, the report geneity was assessed by the chi-square test and quantified using the I2
containing the most comprehensive information on the population was statistic. The I2 statistic represents the percentage of between-study
included to avoid overlapping populations. For those articles in which variation that is due to heterogeneity rather than chance: a value of
information was not reported but the methodology was such that 0% indicates no observed heterogeneity, whereas I2 values of ≥50%
this information would have been recorded initially, the authors were indicate a substantial level of heterogeneity. A fixed-effects model
contacted. Quality assessment of the included studies was performed was used if substantial statistical heterogeneity was not present.
using the Newcastle-Ottawa Scale for cohort studies (Supporting Publication bias was examined by funnel plot analysis. A P-value of less
Information S2).14 than .05 was considered statistically significant.
ZAMBAITI ET AL . 3 of 9

3.2 Sex

Thirty-six papers2,5,16–18,20–47,49,50 allowed the evaluation of gender


effect on outcome: females were found to have RR of 1.21, without
reaching a significance (P = .14; 95% CI 0.94-1.55; I2 0%).

3.3 Tumor secreting status

Three different meta-analyses were performed in order to deter-


mine the effect of secretion pattern on outcome. Firstly, the
patients affected by Cushing syndrome versus those without
Cushing syndrome were compared (Figure 3A). Twenty-seven
studies2,5,16–19,23,24,26,29–34,36–41,43,45,46,48,51 assessed the outcomes
between these two groups, and being affected by Cushing syndrome
resulted in a worse outcome (P < .0001; RR 1.88; 95% CI 1.38-2.58;
I2 6%). Secondly, 19 studies5,18,20,26,30–33,36–38,40,41,43,45,46,49,51 com-
pared the secreting tumors and nonsecreting masses, identifying
hormonal secretion as a negative prognostic factor (P = .004; RR
1.63; 95% CI 1.17-2.27; I2 0%). Finally, the comparison between
patients affected by Cushing syndrome and nonsecreting tumors was
performed in 17 studies5,18,20,26,30–33,37,38,40,41,43,45,46,49,51 and no
difference in outcome was found (P = .40; RR 1.19; 95% CI 0.79-1.79;
I2 0%) (Figure 3B).

3.4 Tumor volume


F I G U R E 1 Flowchart summarizing inclusion of studies in the
systematic review
Five papers reported tumor volume.5,18,20,30,31 The meta-analysis
(Supporting Information S3) showed a worse outcome for the tumors
3 RESULTS
more than 200 cm3 of volume (P < .0001; RR 3.37; 95% CI 1.93-5.88; I2
60%).
A total of 2233 articles were identified and 275 were assessed
with respect to their eligibility for inclusion. Forty-two studies were
included in the systematic review (Figure 1), accounting for a total of
3.5 Tumor weight
1006 pediatric patients with diagnosis of an ACT. The general charac-
teristics of the studies included in the systematic review are reported
Tumor weight was reported in 15 studies. Two meta-analyses were
in Table 1.
performed for the two different cutoffs. Either tumors weighing more
Quality assessment of the included studies was performed using
than 400 g17–21,27,31,33,35,37–40,42,52 or tumors weighing more than
Newcastle-Ottawa Scale for cohort studies. Almost all the included
100 g17,18,20,21,31,33–35,37–40,42,45,52 were considered as risk factor for
studies showed an overall good rate with regard to the selection and
a worse outcome (respectively, P < .00001; RR 4.29; 95% CI 3.19-5.78;
comparability of the study groups and to the ascertainment outcome
I2 0% and P < .00001; RR 5.46; 95% CI 3.24-9.20; I2 0%) (Figure 4A,B).
of interest (Supporting Information S2). The major weaknesses of these
studies were represented by their retrospective design, a lack of homo-
geneity of the clinical features evaluated, and the different thresholds
3.6 Tumor maximum diameter
adopted to define clinical features (as age, tumor volume, weight, max-
imum diameter, and stage).
Three different meta-analyses were performed, considering tumor
maximum diameter as a risk factor. The different groups were com-
3.1 Age pared as follows. First, tumors <5 cm versus tumors >5 and <10 cm
were included in 11 studies,5,25,29,32–35,37,42,45,52 showing a worse
Thirty-three papers entered this meta-analysis.2,5,16–45 An age of more outcome for the tumors >5 and <10 cm (P = .008; RR 2.85; 95%
than 4 years was found to have RR of 3.07 (P < .00001; 95% CI 2.35- CI 1.31-6.17; I2 0%). Second, tumors <5 cm versus tumors >5 cm
4.02; I2 0%) (Figure 2). were included in 15 studies,5,19,22,23,25,28,29,32–35,37,42,45,52 resulting
4 of 9 ZAMBAITI ET AL .

TA B L E 1 General characteristics of the included studies

Mean age at Mean


Sample diagnosis Number of follow-up
Author, year (location) Study design size Period M;F (years) deaths years (range)
Federici, 1994 (Italy) Cohort 12 1976-1989 5;7 5.0 2 6.4 (1.5-14)
Damiani, 1995 (Brazil) Cohort 33 1975-1993 11;22 3.7 17 6.7 (0-20)
Bergadà, 1996 (Argentina) Cohort 20 1970-1991 5;15 7.1 2 10 (0-23)
Mendonca, 1996 (Brazil) Cohort 18 1980-1992 5;13 3.1 2 5.1 (0.5-9.5)
Michalkiewicz, 1997 (USA, Brazil, Cohort 20 1988-1994 18;1 2.0 1 2.5 (0.1-6.2)
Canada, Uruguay, Norway, Chile)
Mayer, 1997 (Canada) Cohort 11 1972-1996 3;8 7.0 1 5.6 (0.8-15)
Driver, 1998 (UK) Cohort 18 1954-1995 3;15 5.6 12 4.3 (0.1-26)
Teinturier, 1999 (France) Cohort 54 1973-1993 27;27 4.0 28 3.2 (0.1-20)
Wolthers, 1999 (UK) Cohort 30 1976-1996 7;23 4.9 4 3.8 (0.4-75)
Wilkin, 2000 (Canada) Cohort 10 - 2;8 5.7 1 7.5 (0.5-18)
Mishra, 2001 (India) Cohort 10 1990-1999 4;6 7.4 5 2.2 (0-7.6)
Misu, 2001 (Japan) Cohort 4 - 3;1 2.5 1 5.9 (3.0-12)
Ciftici, 2001 (Turkey) Cohort 30 1970-1999 11;19 6.7 11 2.5
Latronico, 2001 (Brazil) Cohort 18 - 5;13 2.2 1 5.3 (0.2-13)
McDonnell, 2003 (Australia) Cohort 12 1976-2001 6;6 2.5 3 10 (1.0-25.8)
Narasismhan, 2003 (India) Cohort 8 1989-2000 2;6 2.5 2 2.3 (0.5-5.0)
Sandrini, 2005 (Brazil) Cohort 21 - 7;14 3.7 4 8.6 (0.3-19)
Figuereido, 2005 (Brazil) Cohort 9 - 3;6 2.6 2 -
Barbosa, 2004 (Brazil) Cohort 7 - 4;3 5.1 1 5.8 (1.2-8.2)
Pinto, 2005 (Brazil) Cohort 16 - 6;10 4.4 1 4.4 (1.0-10)
Rosati, 2008 (Brazil) Cohort 12 - 3;9 3.6 5 6.6 (2.4-15.8)
Loncarevic, 2008 (Germany) Cohort 14 1998-2007 6;8 3.8 6 4.4 (4.3-9.8)
Lorea, 2012 (Brazil) Cohort 60 1991-2009 15;45 3.4 10 5.7 (0.7-14)
Waldmann, 2012 (Germany) Cohort 2 - 1;1 16 1 4.2 (1.9-6.5)
Fragoso, 2012 (Brazil) Cohort 24 1990-2010 7;17 3.8 5 6.7 (0.6-14)
Custodio, 2013 (Brazil) Cohort 19 2005-2010 4;15 1.5 4 3.8 (2.0-5.8)
Borges, 2013 (Brazil) Cohort 57 1991-2009 14;43 3.4 10 5.2 (0-14)
Wendt, 2014 (USA) Cohort 5 1975-2011 3;2 3.1 3 7.6 (0.5-32)
Nazli Gonç, 2014 (Turkey) Cohort 18 1999-2013 8;10 5.7 6 6.0 (1.0-11)
Kerkhofs, 2014 (The Netherlands) Cohort 12 1989-2013 3;9 4.1 5 6.4 (0.1-14.8)
Dall’Igna, 2014 (Italy) Cohort 58 1982-2011 19;39 5.1 12 5.9 (0.2-9.9)
Sakoda, 2014 (UK) Cohort 29 1987-2011 14;15 2.2 10 2.1 (0.1-15)
Chatterjee, 2015 (India) Cohort 13 2005-2014 4;9 2.9 3 2.2
Das, 2016 (India) Cohort 14 2005-2015 5;9 2.1 3 4.1 (0.5-5)
Bulzico, 2016 (Brazil) Cohort 27 1997-2015 8;19 5.1 10 2.3 (0.1-12.8)
Pinto, 2017 (USA) Cohort 59 2006-2013 17;42 3.3 14 4.0 (0-23)
Picard, 2018 (France) Cohort 95 2000-2018 28;67 5.0 16 5.3 (0.2-17)
Jehangir, 2019 (India, Australia) Cohort 22 2006-2016 12;10 6.3 2 6.0 (2.1-6.7)
Monteiro, 2019 (Brazil) Cohort 12 2004-2015 2;10 2.0 1 4.5 (0.4-11)
Parise, 2019 (Brazil) Cohort 48 - 18;30 2.8 15 7.0 (0.2-21)
Zekri, 2020 (Egypt) Cohort 18 2007-2016 6;12 4.0 8 3.2
Pinto, 2020 (USA) Cohort 11 - 3;8 4.5 3 -
ZAMBAITI ET AL . 5 of 9

F I G U R E 2 Forest plot showing risk ratio (RR) according to age (<4 years vs >4 years) for each study and pooled for all studies. Only first
author of each study is given

F I G U R E 3 Forest plots showing risk ratio (RR) (A) according to Cushing’s syndrome (Cushing vs non-Cushing); and (B) secretion status
(secreting vs nonsecreting tumors), for each study and pooled for all studies. Only first author of each study is given
6 of 9 ZAMBAITI ET AL .

F I G U R E 4 Forest plots showing risk ratio (RR) according to tumor weight for each study and pooled for all studies in (A) <100 g versus >100 g,
and (B) <400 g versus >400 g. Only first author of each study is given

F I G U R E 5 Forest plots showing risk ratio (RR) according to tumor size diameter for each study and pooled for all studies: (A) maximum size
<5 cm versus >5 cm, and in (B) maximum size <10 cm versus >10 cm. Only first author of each study is given

in a worse outcome for those >5 cm (P = .0009; RR 2.98; 95% CI I2 53%). Stage II was compared to Stage III in six studies,18,20,31,38,40,46
1.56-5.67; I2 0%) (Figure 5A). Finally, the last meta-analysis among 15 and no difference was found (P < .12; RR 1.93; 95% CI 0.84-4.47; I2
studies5,19,22,23,25,28,29,32–35,37,42,45,52 showed that tumors larger than 39%). Lower stages (Stage I and II) versus advanced disease (Stage III
10 cm had worse outcome (P < .00001; RR 3.87; 95% CI 2.51-5.96; I2 and IV) showed RR of 4.99 for advanced tumors (95% CI 3.57-6.98;
0%) (Figure 5B). P < .00001; I2 0%).7,18,20,31,36,38,40,46

3.7 Surgery results 4 DISCUSSION

A meta-analysis (Supporting Information S3) including 13 4.1 Main findings


studies5,20,23,36,27,32,33,37,38,42,51,53,54 compared the outcome between
patients undergone complete resection and those undergone incom- The findings from this systematic review partially confirmed the previ-
plete resection. The first ones showed a better outcome (P < .00001; ous reports. Age <4 years, R0 resection, tumor diameter <5 cm, tumor
RR 3.50; 95% CI 2.51-4.88; I2 16%). volume <200 cm3 , tumor weight <100 g and <400 g, and lower stage
at diagnosis strongly correlated with good outcome. An association
with worse outcome was also found when patients presenting with
3.8 Stage Cushing syndrome were compared with patients without Cushingoid
features (secreting and nonsecreting together), but not when com-
Five different meta-analyses assessed the impact on the outcome of pared with nonsecreting tumors only. Secreting tumors showed a slight
the different stage of disease at diagnosis (Supporting Information S4). increase in the RR when compared with nonsecreting tumors. Lower
The stages were compared as follows. Stage I versus Stage II, III, and stages are associated with a better outcome independently from the
IV included eight studies.7,18,20,31,36,38,40,46 The meta-analysis showed inclusion of metastatic patients into the analysis.
a better outcome for Stage I (P < .00001; RR 6.94; 95% CI 3.35-
14.4; I2 12%). Then, Stage I versus Stage II and III included seven
studies.7,18,20,31,38,40,46 This resulted in a better outcome for patients 4.2 Limitations
with Stage I disease (P < .00001; RR 4.69; 95% CI 2.11-10.4; I2 28%).
Stage I versus Stage II, including six studies,7,18,20,31,38,40 showed a bet- Limitations and bias derived from the features of the studies are
ter outcome once again for Stage I (P < .002; RR 5.02; 95% CI 1.82-13.9; included in this review. The main weaknesses of these series were
ZAMBAITI ET AL . 7 of 9

represented by their retrospective design, small sample size, dif- or spillage60 and discouraged this technique.61,62 The definition of pre-
ferent thresholds or classifications used (such as to describe vol- operative clinical risk factors (eg, age >4 years, Cushing syndrome, or
ume and weight, or stage), and by the fact that most of the stud- nonsecreting tumor), aside from size and suspicious nodes or exten-
ies did not explore all the clinical features taken into account by sion into adjacent organ at imaging, could drive the surgeon’s decision
this systematic review. The variability in the thresholds limited the in order to avoid unnecessary hazards with potentially fatal outcomes.
evaluation of some clinical features, as it lowered the number of
patients who entered the single analysis, decreasing the rate of
events. In this scenario, it is plausible that the relationship between 4.4 Implications for research
a given clinical feature and the outcome may have been under- or
overestimated. Despite the presence of national registries and large studies, and a
considerable number of ACT patients reported in the literature, the
prognostic value to be attributed to a given clinical feature is currently
4.3 Implications for clinical practice extremely variable. For this reason, international cooperative studies
or registries prospectively aimed at defining a risk stratification for
The current pathological scores may not be sufficient to determine the pediatric ACT, ideally based both on clinical and histological features,
malignant behavior of localized ACTs, especially in case of a Stage II-III are needed. These studies should consider prospectively the predictive
disease. The Wieneke index seems to be the most reliable score in pedi- accuracy of the different features, alone and in combination. The opti-
atric age so far,5,6,27 but because it addresses a group of tumors with mal cutoffs of quantitative variables, such as size and volume, should
indeterminate malignancy, it still leaves partially unsolved the deci- be investigated in order to provide a precise estimation of the pre- and
sion on which may be the best postoperative treatment (chemotherapy postsurgical risk of localized ACT. Furthermore, such studies should
associated to mitotane or mitotane alone) for this subset of patients. provide standardization for qualitative variables in order to homoge-
In the past, various clinical features have been considered to be nize data collected in different countries (eg, staging or tumor scores).
associated with poor outcome, but they varied depending on the study:
age >4 years, size, Cushing syndrome, incomplete resections, and stage
have been more commonly reported. Additionally, initial biopsy is con- 5 CONCLUSIONS
sidered to be an unfavorable prognostic factor and it should be avoided
as much as possible.2,8,47,55,56 Moreover, few attempts have been made As expected, localized small tumors did show a better prognosis: a low
to overcome this variability of evidence before the present study, but stage, size <10 cm, and weight under 100 g seem to have lesser risk of
they retrospectively took into account a limited number of patients and death. Some clinical factors (age >4 years, Cushing syndrome) corre-
without reaching a wider consensus on a possible risk stratification. lated with a worse outcome and, despite the biological reasons being
A paper published by the European group (EXpERT) tried to define a far from clear, they could be considered when stratifying patients or
subset of “high-risk” tumors based on the presence of one of the fol- before planning the surgical approach at diagnosis. Notwithstanding its
lowing features: age at diagnosis, volume more than 200 cm3 , Cushing limitations, this systematic review strengthened in part the evidence
syndrome, initial biopsy (open or tru-cut), surgical excision with resid- coming from the previous literature, and it could be considered a step
uals or spillage, regional lymph node involvement, histologic vascular forward for risk stratification for pediatric ACT.
invasion, and distant metastases at diagnosis.8 An analysis performed
on 111 patients of the American National Cancer Data Base suggested ACKNOWLEDGMENTS
including in the risk stratification age, extension into adjacent organs, We thank Carlos Alberto Scrideli (Pediatrics Department, Ribeira õ
metastases, and incomplete resection.55 Preto Medical School, University of Saõ Paulo, Brazil) for his contribu-
Identifying some clinical features, in order to stratify patients who tions to this systematic review in terms of additional explanations on
are at major risk of relapse, could lead to identifying a subgroup of their published data and unpublished data supplied.
ACT which may be elected to a systemic treatment after surgery,
but also in which a mini-invasive approach at diagnosis should be CONFLICT OF INTEREST
avoided. The authors declare that there is no conflict of interest.
The use of adjuvant mitotane for 24-36 months in some Stage II
and III patients, alone or in combination with standard chemotherapy, DATA AVAILABILITY STATEMENT
has been found to be determinant56–59 to improve overall survival and The data that support the findings of this study are available on request
event-free survival. In these cases, a distinction between benign and from the corresponding author.
malignant forms may in turn avoid important and unnecessary side
effects, or represent a possible improvement in terms of survival. ORCID
Indeed, the use of a mini-invasive approach (laparoscopic or robotic- Elisa Zambaiti https://orcid.org/0000-0002-9610-1708
assisted) in ACT is also the object of debate: some authors suggested Patrizia Dall’Igna https://orcid.org/0000-0002-3822-3272
that it could be associated with a higher rate of incomplete resection Calogero Virgone https://orcid.org/0000-0002-3651-9416
8 of 9 ZAMBAITI ET AL .

REFERENCES 20. Custodio G, Parise GA, Filho NK, et al. Impact of neonatal screen-
1. Ribeiro RC, Pinto EM, Zambetti GP, Rodriguez-Galindo C. The Inter- ing and surveillance for the TP53 R337H mutation on early detection
national Pediatric Adrenocortical Tumor Registry initiative: contri- of childhood adrenocortical tumors. J Clin Oncol. 2012;31(20):2619-
butions to clinical, biological, and treatment advances in pediatric 2626.
adrenocortical tumors. Mol Cell Endocrinol. 2012;351:37-43. 21. Damiani D, Della Manna T, Aquino LGW, et al. Proliferating cell nuclear
2. Michalkiewicz E, Sandrini R, Figueiredo B, et al. Clinical and outcome antigen immunoreaction in adrenal tumors. Tumori. 1995;81:273-277.
characteristics of children with adrenocortical tumors: a report from 22. Das S, Sengupta M, Islam N, et al. Weineke criteria, Ki-67 index and p53
the International Pediatric Adrenocortical Tumor Registry. J Clin Oncol. status to study pediatric adrenocortical tumors: is there a correlation?
2004;22:838-845. J Pediatr Surg. 2016;51(11):1795-1800.
3. Dehner LP. Pediatric adrenocortical neoplasms: on the road to some 23. Federici S, Galli G, Ceccarelli PL, et al. Adrenocortical tumors in chil-
clarity. Am J Surg Pathol. 2003;27(7):1005-1007. dren: a report of 12 cases. Eur J Pediatr Surg. 1994;4(1):21-25.
4. Dehner LP, Hill DA. Adrenal cortical neoplasms in children: why 24. Figueiredo BC, Cavalli LR, Pianovski MA, et al. Amplification of the
so many carcinomas and yet so many survivors. Pediatr Dev Pathol. steroidogenic factor 1 gene in childhood adrenocortical tumors. J Clin
2009;12:284-291. Endocrinol Metab. 2005;90(2):615-619.
5. Dall’Igna P, Virgone C, De Salvo GL, et al. Adrenocortical tumors 25. Fragoso MC, Almeida MQ, Mazzuco TL, et al. Combined expression of
in Italian children: analysis of clinical characteristics and P53 sta- BUB1B, DLGAP5, and PINK1 as predictors of poor outcome in adreno-
tus. Data from the national registries. J Pediatr Surg. 2014;49:1367- cortical tumors: validation in a Brazilian cohort of adult and pediatric
1371. patients. Eur J Endocrinol. 2012;166(1):61-67.
6. Wieneke JA, Thompson LD, Heffess CS. Adrenal cortical neoplasms in 26. NazliGönç E, Özön ZA, Çakir MD, Alikasifoglu A, Kandemir N. Need for
the pediatric population: a clinicopathologic and immunophenotypic comprehensive hormonal workup in the management of adrenocorti-
analysis of 83 patients. Am J Surg Pathol. 2003;27:867-881. cal tumors in children. J Clin Res Pediatr Endocrinol. 2014;6(2):68-73.
7. Picard C, Orbach D, Carton M, et al. Revisiting the role of the patholog- 27. Jehangir S, Nanjundaiah P, Sigamani E, et al. Pathological prognostica-
ical grading in pediatric adrenal cortical tumors: results from a national tion of paediatric adrenocortical tumours: is a gold standard emerging.
cohort study with pathological review. Mod Pathol. 2019;32:546-559. Pediatr Blood Cancer. 2019;66:e27567.
8. Cecchetto G, Ganarin A, Bien E, et al. Outcome and prognostic fac- 28. Kerkhofs TM, Ettaieb MH, Verhoeven RH, et al. Adrenocortical carci-
tors in high-risk childhood adrenocortical carcinomas: a report from noma in children: first population-based clinicopathological study with
the European Cooperative Study Group on Pediatric Rare Tumors long-term follow-up. Oncol Rep. 2014;32(6):2836-2844.
(EXPeRT). Pediatr Blood Cancer. 2017;64(6):e26368. 29. Latronico AC, Pinto EM, Domenice S, et al. An inherited mutation out-
9. Gulack BC, Rialon KL, Englum BR, et al. Factors associated with sur- side the highly conserved DNA-binding domain of the p53 tumor sup-
vival in pediatric adrenocortical carcinoma: an analysis of the National pressor protein in children and adults with sporadic adrenocortical
Cancer Data Base (NCDB). J Pediatr Surg. 2016;51:172-177. tumors. J Clin Endocrinol Metab. 2001;86(10):4970-4973.
10. Henderson LK, Craig JC, Willis NS, Tovey D, Webster AC. How to write 30. Loncarevic IF, Hering A, Posorski N, Linden T, Hoyer H, Bucky P. Num-
a Cochrane systematic review. Nephrology (Carlton). 2010;15(6):617- ber of genomic imbalances correlates with the overall survival for
624. adrenocortical cancer in childhood. Pediatr Blood Cancer. 2008;51:356-
11. NHS Centre for Reviews and Dissemination. Systematic Reviews. CRD’s 362.
Guidance for Undertaking Reviews in Health Care. York, UK: Centre for 31. Lorea CF, Moreno DA, Borges KS, et al. Expression profile of apoptosis-
Reviews and Dissemination, University of York; 2009 related genes in childhood adrenocortical tumors: low level of expres-
12. Leeflang MM, Deeks JJ, Gatsonis C, Bossuyt PM. Systematic reviews sion of BCL2 and TNF genes suggests a poor prognosis. Eur J Endocrinol.
of diagnostic test accuracy. Ann Intern Med. 2008;149(12):889-897. 2012;167(2):199-208.
13. Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for 32. Mayer SK, Oligny LL, Deal C, Yazbeck S, Gagné N, Blanchard
reporting systematic reviews and meta-analyses of studies that eval- H. Childhood adrenocortical tumors: case series and reevalua-
uate health care interventions: explanation and elaboration. PLoS Med. tion of prognosis–a 24-year experience. J Pediatr Surg. 1997;32
2009;6(7):e1000100. (6):911-915.
14. Wells GA, Shea B, O’Connell D, et al. The Newcastle-Ottawa Scale (NOS) 33. McDonnell CM, Zacharin MR. Adrenal cortical tumours: 25 years’
for Assessing the Quality if Nonrandomized Studies in Meta-Analyses. experience at the Royal Children’s Hospital, Melbourne. J Paediatr
Ottawa, Canada: Ottawa Health Research Institute; 1999. http: Child Health. 2003;39:682-685.
//www.ohri.ca/programs/clinical_epidemiology/oxford.asp. Accessed 34. Mendonca BB, Lucon AM, Menezes CA, et al. Clinical, hormonal and
June 28, 2020. pathological findings in a comparative study of adrenocortical neo-
15. Sandrini R, Ribeiro RC, DeLacerda L. Childhood adrenocortical tumors. plasms in childhood and adulthood. J Urol. 1995;154(6):2004-2009.
J Clin Endocrinol Metab. 1997;82:2027-2031. 35. Mishra A, Agarwal G, Misra AK, Agarwal A, Mishra SK. Functioning
16. Barbosa AS, Giacaglia LR, Martin RM, Mendonca BB, Lin CJ. Assess- adrenal tumours in children and adolescents: an institutional experi-
ment of the role of transcript for GATA-4 as a marker of unfavor- ence. ANZ J Surg. 2001;71:103-107.
able outcome in human adrenocortical neoplasms. BMC Endocr Disord. 36. Monteiro NML, Rodrigues KES, Vidigal PVT, Oliveira BM. Adrenal car-
2004;4(1):3. cinoma in children: longitudinal study in Minas Gerais, Brazil. Rev Paul
17. Bergadá I, Venara M, Maglio S, et al. Functional adrenal cortical Pediatr. 2019;37(1):20-26.
tumors in pediatric patients: a clinicopathologic and immunohisto- 37. Narasimhan KL, Samujh R, Bhansali A, et al. Adrenocortical tumors in
chemical study of a long term follow-up series. Cancer. 1996;77 childhood. Pediatr Surg Int. 2003;19:432-435.
(4):771-777. 38. Parise IZS, Parise GA, Noronha L, et al. The prognostic role of
18. Silva Borges K, Antunes Moreno D. Spindle assembly checkpoint gene CD8(+) T lymphocytes in childhood adrenocortical carcinomas com-
expression in childhood adrenocortical tumors (ACT): overexpression pared to Ki-67, PD-1, PD-L1, and the Weiss score. Cancers (Basel).
of Aurora kinases A and B is associated with a poor prognosis. Pediatr 2019;11(11):1730.
Blood Cancer. 2013;60:1809-1816. 39. Pinto EM, Billerbeck AEC, Villares Fragoso MCB, Mendonca B, Latron-
19. Chatterjee G, DasGupta S, Mukherjee G, et al. Usefulness of Wieneke ico AC. J Clin Endocrinol Metab. 2005;90(5):2976-2981.
criteria in assessing morphologic characteristics of adrenocortical 40. Pinto EM, Rodriguez-Galindo C, Pounds SB, et al. Identification of clin-
tumors in children. Pediatr Surg Int. 2015;31(6):563-571. ical and biologic correlates associated with outcome in children with
ZAMBAITI ET AL . 9 of 9

adrenocortical tumors without germline TP53 mutations: a St Jude 55. McAteer JP, Huaco JA, Gow KW. Predictors of survival in pedi-
Adrenocortical Tumor Registry and Children’s Oncology Group study. atric adrenocortical carcinoma: a Surveillance, Epidemiology, and
J Clin Oncol. 2017;35(35):3956-3963. End Results (SEER) program study. J Pediatr Surg. 2013;48(5):
41. Rosati R, Cerrato F, Doghman M, et al. High frequency of loss of het- 1025-1031.
erozygosity at 11p15 and IGF2 overexpression are not related to clini- 56. Picard C, Faure-Conter C, Leblond P, et al. Exploring heterogene-
cal outcome in childhood adrenocortical tutors positive for the R337H ity of adrenal cortical tumors in children: the French pediatric rare
TP53 mutation. Cancer Genet Cytogenet. 2008;186:19-24. tumor group (Fracture) experience. Pediatr Blood Cancer. 2020;67:
42. Sakoda A, Mushtaq I, Levitt G, Sebire NJ. Clinical and histopathological e28086.
features of adrenocortical neoplasms in children: retrospective review 57. Redlich A, Boxberger N, Strugala D, et al. Systemic treatment of
from a single specialist center. J Pediatr Surg. 2014;49(3):410-415. adrenocortical carcinoma in children: data from the German GPOH-
43. Sandrini F, Villani DP, Tucci S, Moreira AC, de Castro M, Elias LL. Inher- MET 97 trial. Klin Padiatr. 2012;224:366-371.
itance of R337H p53 gene mutation in children with sporadic adreno- 58. Fassnacht M, Terzolo M, Allolio B, et al. Combination chemotherapy
cortical tumor. Horm Metab Res. 2005;37(4):231-235. in advanced adrenocortical carcinoma. N Engl J Med. 2012;366:2189-
44. Wendt S, Shelso J, Wright K, Furman W. Neoplastic causes of abnormal 2197.
puberty. Pediatr Blood Cancer. 2014;61:664-671. 59. Terzolo M, Angeli A, Fassnacht M, et al. Adjuvant mitotane treat-
45. Wilkin F, Gagné N, Paquette J, Oligny LL, Cheri D. Pediatric adrenocor- ment for adrenocortical carcinoma. N Engl J Med. 2007;356:2372-
tical tumors: molecular events leading to insulin-like growth factor II 2380.
gene overexpression. J Clin Endocrinol Metab. 2000;85(5):2048-2056. 60. Miller BS, Gauger PG, Hammer GD, Doherty GM. Resection of adreno-
46. Zekri W, Hammad M, Rashed WM, et al. The outcome of childhood cortical carcinoma is less complete and local recurrence occurs sooner
adrenocortical carcinoma in Egypt: a model from developing countries. and more often after laparoscopic adrenalectomy than after open
Pediatr Hematol Oncol. 2020;37(3):198-210. adrenalectomy. Surgery. 2012;152(6):1150-1157.
47. Bulzico D, de Faria PA, de Paula MP, et al. Recurrence and mortality 61. Hubertus J, Boxberger N, Redlich A, von Schweinitz D, Vorwerk P.
prognostic factors in childhood adrenocortical tumors: analysis from Surgical aspects in the treatment of adrenocortical carcinomas in
the Brazilian National Institute of Cancer experience. Pediatr Hematol children: data of the GPOH-MET 97 trial. Klin Padiatr. 2012;224:
Oncol. 2016;33(4):248-258. 143-147.
48. Misu Y, Jiang S, Yukfumi Y, et al. Clinicopathological features of pedi- 62. Miller BS. Changes in the evaluation and management of adrenocorti-
atric functional adrenocortical carcinoma diagnosed by Weiss criteria; cal carcinoma. Ann Surg Oncol. 2018;25(12):3413-3415.
an analysis of four cases. Clin Pediatr Endocrinol. 2001;10(2):141-146.
49. Pinto EM, Faucz FR, Paza LZ, et al. Germline variants in phosphodi-
esterase genes and genetic predisposition to pediatric adrenocortical
tumors. Cancers (Basel). 2020;12(2):506. SUPPORTING INFORMATION
50. Waldmann J, Patsalis N, Fendrich V, et al. Clinical impact of TP53 Additional supporting information may be found online in the Support-
alterations in adrenocortical carcinomas. Langenbecks Arch Surg.
ing Information section at the end of the article.
2012;397(2):209-216.
51. Ciftci AO, Senocak ME, Tanyel FC, Büyükpamukçu N. Adrenocortical
tumors in children. J Pediatr Surg. 2001;36(4):549-554.
52. Wolthers OD, Cameron FJ, Scheimberg I, et al. Androgen secreting
adrenocortical tumours. Arch Dis Child. 1999;80(1):46-50. How to cite this article: Zambaiti E, Duci M, De Corti F, et al.
53. Driver CP, Birch J, Gough DC, Bruce J. Adrenal cortical tumors in child- Clinical prognostic factors in pediatric adrenocortical tumors:
hood. Pediatr Hematol Oncol. 1998;15(6):527-532. A meta-analysis. Pediatr Blood Cancer. 2021;68:e28836.
54. Teinturier C, Pauchard MS, Brugières L, Landais P, Chaussain JL,
https://doi.org/10.1002/pbc.28836
Bougnères PF. Clinical and prognostic aspects of adrenocortical neo-
plasms in childhood. Med Pediatr Oncol. 1999;32(2):106-111.

You might also like