Next Article in Journal
Monitoring of Interfractional Proton Range Verification and Dosimetric Impact Based on Daily CBCT for Pediatric Patients with Pelvic Tumors
Previous Article in Journal
Real-World Outcomes of Nivolumab, Pembrolizumab, and Atezolizumab Treatment Efficacy in Korean Veterans with Stage IV Non-Small-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review

by
Monika Grubliauskaite
1,2,3,*,†,
M. E. Madeleine van der Perk
4,†,
Annelies M. E. Bos
4,5,
Annelot J. M. Meijer
4,
Zivile Gudleviciene
6,
Marry M. van den Heuvel-Eibrink
4,7,‡ and
Jelena Rascon
1,6,‡
1
Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Santariskiu Str. 4, LT-08406 Vilnius, Lithuania
2
Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
3
Department of Biobank, National Cancer Institute, Santariskiu Str. 1, LT-08406 Vilnius, Lithuania
4
Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
5
Department of Reproductive Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
6
Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21/27, LT-03101 Vilnius, Lithuania
7
Division of Child Health, UMCU-Wilhelmina Children’s Hospital, 3584 EA Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
These first authors contributed equally to this work.
These last authors contributed equally to this work.
Cancers 2023, 15(17), 4199; https://doi.org/10.3390/cancers15174199
Submission received: 18 July 2023 / Revised: 7 August 2023 / Accepted: 9 August 2023 / Published: 22 August 2023

Abstract

:

Simple Summary

Cryopreservation of ovarian tissue and transplantation are the only available fertility techniques to preserve fertility and endocrine function for prepubertal girls with cancer who require immediate cancer treatment. The primary objective of this systematic review was to identify and critically appraise the existing evidence regarding targeted minimal infiltrative disease detection in harvested ovarian tissues and identify markers that may be of value for assessment before autotransplantation, thereby facilitating fertility restoration in childhood cancer survivors. While the majority of malignancies were found to be at low risk of containing malignant cells in ovarian tissue, more studies are needed to ensure safe implementation of future fertility restoration in clinical practice.

Abstract

Background: Ovarian tissue cryopreservation and transplantation are the only available fertility techniques for prepubertal girls with cancer. Though autotransplantation carries a risk of reintroducing malignant cells, it can be avoided by identifying minimal infiltrative disease (MID) within ovarian tissue. Methods: A broad search for peer-reviewed articles in the PubMed database was conducted in accordance with PRISMA guidelines up to March 2023. Search terms included ‘minimal residual disease’, ‘cryopreservation’, ‘ovarian’, ‘cancer’ and synonyms. Results: Out of 542 identified records, 17 were included. Ovarian tissues of at least 115 girls were evaluated and categorized as: hematological malignancies (n = 56; 48.7%), solid tumors (n = 42; 36.5%) and tumors of the central nervous system (n = 17; 14.8%). In ovarian tissue of 25 patients (21.7%), MID was detected using RT-qPCR, FISH or multicolor flow cytometry: 16 of them (64%) being ALL (IgH rearrangements with/without TRG, BCL-ABL1, EA2-PBX1, TEL-AML1 fusion transcripts), 3 (12%) Ewing sarcoma (EWS-FLI1 fusion transcript, EWSR1 rearrangements), 3 (12%) CML (BCR-ABL1 fusion transcript, FLT3) and 3 (12%) AML (leukemia-associated immunophenotypes, BCR-ABL1 fusion transcript) patients. Conclusion: While the majority of malignancies were found to have a low risk of containing malignant cells in ovarian tissue, further studies are needed to ensure safe implementation of future fertility restoration in clinical practice.

1. Introduction

Substantial improvements in childhood cancer treatment, as well as enhanced stratification and refinement of supportive care, have resulted in a five-year survival rate of over 80% in high-income countries [1]. Hence, substantial growth in the childhood cancer survivor population is currently observed, which urges attention toward treatment-related late sequelae. Impaired fertility is one of the relevant long-term adverse effects of pediatric cancer treatment, which is mainly caused by gonadotoxic chemotherapy and/or radiotherapy. Alkylating agents are especially notorious for gonadotoxicity and can lead to delayed puberty, premature menopause, and ovarian insufficiency due to a substantial decrease in the follicle pool [2]. Nowadays, fertility issues among young cancer survivors are gaining increasing attention. The involvement of a multidisciplinary fertility specialist team is crucial to ensure a timely assessment of the gonadal damage risk and a proper fertility preservation decision process to improve the quality of life in remission [3].
Cryopreservation of ovarian tissue (OT) is one of the options to preserve fertility in prepubertal and adolescent female patients who require immediate cancer treatment [3]. To preserve follicles, ovarian tissue cryopreservation (OTC) is the only option since ovarian stimulation cannot be performed. OTC provides the opportunity to store ovarian tissue and to autotransplant it once the patient is disease-free and has a child-wish. According to recent data, 1019 OTC procedures have been performed in children and young adults with cancer (0–20.4 years), of whom 298 were younger than 13 years. Eighteen of them received an ovarian tissue (OT) transplant in adulthood, resulting in eleven pregnancies, of which nine resulted in a live birth [4,5]. In addition, hormonal function restoration was observed in 17 (94%) patients who had undergone OT transplantation [5]. In 2018, it was shown that 95% of 318 women who had OT transplantation worldwide had successful long-term restoration of ovarian and endocrine functions as adults [6]. Currently, alternatives to OT transplantation are being developed, such as in vitro activation and/or ex vivo growth and maturation of primordial follicles [7,8], construction of artificial ovaries [9] or developing oocytes from stem cells [10,11]. Unfortunately, the biological principles of follicular development are still not fully understood [8]. To give rise to high-quality oocytes, OT freezing should ideally be performed before treatment [12].
An important concern of OTC is the risk of reintroducing malignant cells that may be harbored within the OT [13,14,15]. Methods of identification of minimal infiltrative disease (MID) in cryopreserved OT are of crucial importance to prevent iatrogenic transplantation of cancer cells and subsequent relapse [6]. MID in cryopreserved OT can be identified using (a combination of) histology, immunohistochemistry (IHC) and currently available molecular technologies. Subsequently, purging strategies for residual cells in intact OT are being developed [16,17,18,19]. All of which aim to eliminate the risk of autotransplanting residual cancer cells.
This systematic review was pursued as part of The Twinning in Research and Education to Improve Survival in Childhood Solid Tumours in Lithuania (TREL) project, which is an EU Horizon 2020-funded project that aims to improve various aspects of childhood cancer care, including quality of survivorship, by the twinning of researchers from different institutions. We aimed to answer the following questions:
  • Which targets are currently being used to screen for MID in cryopreserved OTs of pediatric patients?
  • Which techniques were used for MID detection in OT in the relevant subgroups and may be used for assessment of the graft before autotransplantation?

2. Materials and Methods

This systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (www.prisma-statement.org/ (accessed on 18 July 2023)).

2.1. Search Strategy

Using the PubMed database, a broad search for peer-reviewed articles was carried out in February 2022 and updated on 7 March 2023. In this search, terms including ‘minimal residual disease’, ‘cryopreservation’, ‘ovarian’ and ‘cancer’ and their synonyms were used (Table A1). Papers addressing the presence or absence of malignant cells in human OT harvested for cryopreservation were included, as were all papers describing human–animal and human–human ovarian transplantation after malignant disease. The selection and filtering of relevant articles were performed by two independent authors (M.G. and M.E.M.v.d.P.) using the systematic review web application Rayyan (http://rayyan.qcri.org (accessed on 6 February 2022)) [20] (Figure 1). The reference list of identified articles was also screened to identify additional important articles.

2.2. Selection Criteria

For both aims, the inclusion and exclusion criteria can be found in Table A2. Briefly, all articles in English with fully available text on cryopreserved OT of female childhood (<18 years) cancer patients which included assessments of potential biomarkers for cancer cells in OT were included in this systematic review.
Excluded articles described adults or pediatric male patients, research solely including cancer cell lines or markers measured only in the origin tissue of the cancer, blood or serum, reviews, comments and guidelines.

2.3. Data Extraction

The information extracted from every identified article included the author and year of publication, study type, number and characteristics of patients (cancer diagnosis, age at the cryopreservation, received cancer treatment before OTC), MID markers and methods used to detect them and outcomes—presence/absence of MID.

2.4. Quality of Evidence Assessment

Two authors (M.G. and M.E.M.v.d.P.) evaluated the methodological quality of the included studies using the Quality in Prognosis Studies (QUIPS) tool criteria independently [22] and evaluated the overall quality of evidence in every study using the GRADE criteria (Table S1) [23]. The following six domains of the QUIPS tool were used: (1) study participation; (2) study attrition; (3) prognostic factor measurement; (4) outcome measurement; (5) study confounding; and (6) statistical analysis and reporting. Each domain was assessed as having a low, moderate or high risk of bias and subsequently an overall risk of bias was established. The most important domains for our systematic review were study participation, prognostic factor measurement, outcome measurement and study confounding since we aimed to evaluate which markers and methods can be used to detect MID in OT. The most important influential confounders for detection of MID included cancer treatment before OTC, no availability of original tumor material or markers and survival or disease recurrence. Disagreement between the authors was resolved by consensus.

3. Results

A total of 515 hits were identified in the PubMed database and 14 additional articles were included from studies’ references or were recommended (Figure 1). The updated search on 7 March 2023 revealed 27 new hits. After the removal of duplicates and screening of titles and abstracts, 358 articles were excluded. Based on title–abstract screening, a full-text screen was performed on 195 articles and 17 articles dated from 2010–2022 were included in the qualitative synthesis.
The studies were grouped into three categories: (1) solid tumors including EWS, rhabdomyosarcoma, osteosarcoma, synovial sarcoma and clear cell sarcoma (n = 7); (2) hematological malignancies including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), juvenile myelomonocytic leukemia (JMML) and Burkitt’s lymphoma (n = 8); (3) tumors of the central nervous system (CNS) and brain tumors including astrocytoma, ependymoma, germinoma, glioblastoma, medulloblastoma and primitive neuroectodermal tumors (n = 2). No studies describing patients with Wilms tumors, hepatoblastomas, retinoblastomas or Hodgkin lymphomas were identified. Evaluation of the articles is shown in Table 1, Table 2, Table 3 and Table S2. The markers and methods described in the included articles are presented in Table 1, Table 2 and Table 3. In this systematic review, ovarian tissue from 115 to 122 girls (some studies recorded the age of patients in a range that included patients older than 18 years) was reported to be analyzed by histology, IHC, several types of polymerase chain reaction (PCR), fluorescence in situ hybridization (FISH) or multicolor flow cytometry (MFC). For 62 of the patients, additionally, OT was xenotransplanted in immunodeficient mice and evaluated for MID using histology, IHC, PCR or MFC methods [16,24,25,26,27,28,29]. The main cryopreservation technique used was slow freezing (used for 87 patients, 5 of them had additionally snap frozen OT pieces, while for the others the used technique was not reported). The risk of bias in most studies was moderate to high and was based primarily on study participation, inconsequent use of the methods to detect MID and insufficiently taking possible confounders into account. The overall quality of evidence was very low, primarily due to the study type (case reports and retrospective studies) and low number of participants.
Table 1. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with solid tumors.
Table 1. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with solid tumors.
Ref.Study TypePt (n)Type of CancerMarkers and Methods (1–5)Detected by Methods
(1–5)
Positive Patients, n (%)Potential BiasOverall Bias
Histology (1)IHC (2)FISH (3)PCR $ (4)Xeno (5)SBABMBDBSCSA*
[30]Retrospective study7Ewing sarcoman = 6CD99
n = 6
-EWS-FLI 1 FT
n = 5
-4method4: 1/5 (20%)Cancers 15 04199 i001Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002
[31]Retrospective study5Ewing sarcoman = 5-EWSR1 n = 5EWS-FLI 1 FT
n = 5
-ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i002
[32]Case report 1Ewing sarcoma---EWS-FLI 1 FT
n = 1
-ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i001
[15]Case reports2Ewing sarcoma-CD99
n = 2
EWSR1 n = 2--2, 3method2: 2/2 (100%)
method3: 2/2 (100%)
Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i001
[24]Retrospective study9Ewing sarcoma, synovial sarcoma, osteosarcoman = 9--EWS-FLI 1 FT
n = 6
n = 9ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002
[33]Retrospective study2–9 Ewing sarcoma, osteosarcoman = 5CD99
n = 2
-EWS-ERG FT
n = 2
-ND-Cancers 15 04199 i001Cancers 15 04199 i001Cancers 15 04199 i001Cancers 15 04199 i001Cancers 15 04199 i002Cancers 15 04199 i001Cancers 15 04199 i001
[34]Retrospective study16Ewing sarcoma,
PNET, clear cell
sarcoma, synovial sarcoma,
rhabdomyosarcoma
n = 16CD99, MDM2, myogenin, S100, MyoD 1, melanoma cocktail
n = 16
-EWS-FLI 1, EWS-ERG, PAX3-FOXO1, PAX7-FOXO1, SYT-SSX, EWS-ATF1 FTs, MyoD1
n = 12
-ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002
Grade assessment
Study designLowRetrospective cohort studies in 5/7; case reports in 2/7
Study limitations−2Important limitations: SB high in 7/7; AB low in 5/7, moderate in 1/7, high in 1/7; MB is low in 3/7, moderate in 3/7, high in 1/7; DB is low in 3/7, moderate in 3/7, high in 1/7; SC is low in 5/7, moderate in 2/7; SA is low in 4/7, moderate in 2/7, high in 1/7
Consistency0Little inconsistency: 6/7 (1/6 studies detected MID) studies used PCR and 4/7 IHC (1/4 studies detected MID) to detect MID; FISH detected MID in 1/2, and markers used were mostly the same across all the studies for a specific method)
Directness−1Though results are direct, population and outcomes cannot be generalized
Precision−1Low number of patients
Publication bias0Unlikely
Effect size0NA
Dose–response0NA
Plausible confounding+1Treatment as plausible confounder may seriously influence the effect
Quality of evidenceCancers 15 04199 i004Very low
Conclusion Detection of MID in ovarian tissues from solid tumor patients is unlikely despite various attempts to identify it but the results cannot be generalized (7 studies, 42–49 participants)
Cancers 15 04199 i003 = low risk of bias; Cancers 15 04199 i002 = moderate risk of bias; Cancers 15 04199 i001 = high risk of bias; Cancers 15 04199 i004 = very low quality of evidence. AB = attrition bias (the study data available (i.e., participants not lost to follow up) adequately represent the study sample), DB = detection bias (the outcome of interest is measured in a similar way for all participants), FISH = fluorescence in situ hybridization, FT = fusion transcript, IHC = immunohistochemistry, L = low risk, M = moderate risk, MB = measurement bias (the prognostic factor is measured in a similar way for all participants), ND = not detected, OT = ovarian tissue, OTC = ovarian tissue cryopreservation, PCR = polymerase chain reaction, PNET = primitive neuroectodermal tumor, Pt = patients, SA* = no statistical analysis performed, all primary outcomes are reported, SB = selection bias (the study sample adequately represents the population of interest), SC = study confounding (important potential confounding factors are appropriately accounted for), Xeno = xenotransplantation (and evaluation after). ‘$’ includes all types of PCR: PCR, RT-PCR, quantitative RT-PCR, digital droplet RT-PCR, nested PCR; ‘-’ means the method was not used in the specific study; OS: n = 7 (mean age 15, range 13–18 y) EW: n = 4 (mean 19, range 17–21 y), it is unclear how many patients were <18 years; letter to the editor about a case report.

3.1. Solid Tumors

Seven studies investigated MID in different solid tumors such as Ewing sarcoma, primitive neuroectodermal tumor (PNET), osteosarcoma, rhabdomyosarcoma, synovial sarcoma and clear cell sarcoma. In total, the OT of at least 42 patients with a mean age at the time of cryopreservation of 12.2 ± 3.6 years (median 13 years, range 1–17 years) was analyzed. Eleven patients had metastatic disease, twenty-four had localized disease and seven patients did not have metastasis status reported. Thirty patients had their OT frozen using slow freezing technique, five additionally had their OT snap frozen, while for twelve patients the used technique was not specified. EWS was the most common malignancy (27 out of 42 patients). The primary tumor or blood was used as a positive control to find MID markers in the OT, if possible. Histology, IHC, reverse transcription quantitative PCR (RT-qPCR), FISH and xenotransplantation were used for MID detection (Table 1). In summary, in 3 OT samples from 27 (11%) EWS patients, an infiltration of cancer cells was found while infiltrations were absent in the OT samples of patients with other solid tumors (n = 15). Twenty-nine patients had OTC performed before treatment initiation, ten after treatment initiation and for three patients it was not reported.
Ewing sarcoma—OTs of at least 27 pediatric EWS patients were analyzed and in 3 patients cancer cells were found by immunostaining or molecular testing (Table 1). Two out of three MID-positive patients did not have distant metastasis at the time of OTC reported [15] and for the third one the disease spread was not mentioned [30]. Seven of the twenty-four MID-negative patients had metastatic disease, for six patients it was not described and eleven patients had localized disease. Positive identification of MID included CD99-positive tumor islets, EWS-FLI1 fusion transcript and EWSR1 rearrangements detected by IHC, RT-qPCR and FISH analysis, accordingly. Histological evaluation or xenotransplantation did not detect infiltration of malignant cells. Interestingly, in peripheral blood of one EWS patient the result was positive for fusion transcript though OT evaluation showed the absence of MID [30].
PNET (Ewing family)—OTs of three patients with localized PNET of the Ewing family were found to be negative for MID [34]. OTs were analyzed by histology, IHC or RT-qPCR using CD99 and NSE or EWS-FLI1, accordingly.
Osteosarcoma—In OTs from osteosarcoma (OS) patients (n = 2–7 (girls were presented in an age range including patients >18 years; precise number is not known)), only histology was used to check for MID and no malignant cells were found [24,33]. Two patients were reported to have metastatic disease.
Soft tissue sarcomas—Malignant cells were absent in all ten patients, though two had metastatic disease (both deceased) and one had recurrence [34]. Detection of MID for rhabdomyosarcoma included myogenins, MyoD1 and PAX-FOXO1 fusion transcript, for synovial sarcoma—Bcl-2 protein and SYT-SSX fusion transcript, for clear cell sarcoma—S100 and melanoma cocktail and EWS-ATF1 fusion transcript by IHC and RT-qPCR [34]. Histological evaluation after xenotransplantation showed no MID infiltration [24].

3.2. Hematological Malignancies

Eight studies performed MID analysis in OT in different hematological malignancies including ALL, AML, CML, JMML and Burkitt’s lymphoma. In total, 56 harvested OTs were analyzed from patients with a mean age at the time of cryopreservation of 10.4 ± 5.0 years (median 13 years, range 1–17 years). All patients except one (not reported) had their OTs frozen using the slow freezing technique, in addition, 5 of those patients had their OT frozen by snap freezing technique as well. Bone marrow or a blood sample from each patient was used as a positive control and analyzed to find specific markers for MID detection, if possible (Table 2). Methods included histology, IHC, RT-PCR, MFC or xenotransplantation into immunodeficient mice. In summary, in 22 out of 56 (39%) analyzed patients, the infiltration of malignant cells was found. The OTC was performed at various time points, 10 patients showed positive MID before chemotherapy and 12 after the initiation of the treatment, while 15 out of 34 MID-negative patients had OTC performed before chemotherapy and 19 after having initiated the treatment.
Table 2. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with hematological malignancies.
Table 2. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with hematological malignancies.
Ref.Study TypePt
(n)
Type of CancerMarkers and Methods (1–5)Detected by Methods (1–5)Positive Patients, n (%)Potential BiasOverall Bias
Histology (1)IHC (2)PCR $ (3)Xeno (4)MFC (5)SBABMBDBSCSA*
[35]Case report1CMLn = 1Glycophorin A, MPO, CD34, CD68, LCA/DC45, Factor VIII
n = 1
BCR-ABL FT
n = 1
--31/1 (100%)Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i001
[25]Retrospective study8ALL, AMLn = 8-ETV6-RUNX1, BCR-ABL1 FTs, IGH, TCR, FLT3 rearrangements
n = 6
n = 8-1, 3, 4method1: 1/8 (12.5%)
method3: 5/6 (83%)
method4: 2/8 (25%)
Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i001
[36]Case report1ALL--BCR-ABL FT
n = 1
--ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i001
[37]Retrospective study14ALL, AML, CML, JMMLn = 14CD34, CD10, CD20, CD79a, CD3, TdT, CD117, MPO, CD68
n = 14
TEL-AML1, BCR-ABL b2a2, BCR-ABL b2a2, CBFB-MYH11 type A FTs
n = 5
--33/5 (60%)Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i001Cancers 15 04199 i001Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i001
[26]Retrospective study8B-ALL,
T-ALL, AML
---n = 6CD19, CD34, CD10-, negative for myeloid markers/CD45, HLA-DR2, CD10, CD19, CD22, CD33/CD19, CD10, CD22, CD38/CD45, CD10, CD19, CD22, CD34, HLA-DR2/CD2, cyCD3, CD5, CD7, CD10, CD33, CD34, CD45RA, CD123
n = 8
4, 5method4: 1/6 (12.5%)
method5: 2/8 (25%)
Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002
[38]Retrospective study9ALL, AML, Burkitt’s lymphoman = 9-E2A-PBX1, TEL-AML1, MLL-AF4, AML1-ETO FTs, IgK Kde, IgH, TCRD, TCRB
n = 9
--32/9 (22%)Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i002
[27]Retrospective study10ALL, CMLn = 10-BCR-ABL1, Ig and/or TCR-gama rearrangements
n = 8
n = 10-3, 4method3: 5/8 (62.5%)
method4: 4/10 (40%)
Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i002
[16]Retrospective study5ALLn = 5CD20, CD79a, CD3, TdT
n = 5
IgH rearrangements,
EA2-PBX1 FT n = 4
n = 5-34/4 (100%)Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i002
Grade assessment
Study designLowRetrospective cohort studies in 6/8; case reports in 2/8
Study limitations−1Some limitations: SB high in 8/8; AB low in 8/8; MB is low in 4/8, moderate in 3/8, high in 1/8; DB is low in 4/8, moderate in 3/8, high in 1/8; SC low in 5/8, moderate in 3/8; SA low in 8/8
Consistency −1Though MID was detected by PCR in 6/7 studies and by xenotransplantation in 3/4, IHC detected MID in 0/3 and histology in 1/6 studies. The markers analyzed were very different across the studies
Directness −1Though results are direct, population and outcomes cannot be generalized
Precision−1Low number of events
Publication bias0Unlikely
Effect size0NA
Dose–response0NA
Plausible confounding+1Treatment as plausible confounder may seriously influence the effect
Quality of evidenceCancers 15 04199 i004Very low
Conclusion Detection of MID in ovarian tissues from the patients of haematological malignancies is moderately likely, despite various attempts to identify it, the results cannot be generalized (8 studies, 56 participants)
Cancers 15 04199 i003 = low risk of bias; Cancers 15 04199 i002 = moderate risk of bias; Cancers 15 04199 i001 = high risk of bias; Cancers 15 04199 i004 = very low quality of evidence. AB = attrition bias (the study data available (i.e., participants not lost to follow up) adequately represent the study sample), ALL = acute lymphoblastic leukemia, AML = acute myeloid leukemia, CML = chronic myeloid leukemia, DB = detection bias (the outcome of interest is measured in a similar way for all participants), FISH = fluorescence in situ hybridization, FT = fusion transcript, IHC = immunohistochemistry, JMML = juvenile myelomonocytic leukemia, L = low risk, M = moderate risk, MB = measurement bias (the prognostic factor is measured in a similar way for all participants), MID = minimally infiltrative disease, MPO = myeloperoxidase, ND = not detected, OT = ovarian tissue, OTC = ovarian tissue cryopreservation, PCR = polymerase chain reaction, Pt = patients, SA* = no statistical analysis performed, all primary outcomes are reported, SB = selection bias (the study sample adequately represents the population of interest), SC = study confounding (important potential confounding factors are appropriately accounted for), Xeno = xenotransplantation (and evaluation after). ‘-’ means the method was not used in the specific study; ‘$’ includes all types of PCR: PCR, RT-PCR, quantitative RT-PCR, digital droplet RT-PCR, nested PCR.
ALL—OTs of 39 patients were analyzed in 7 studies, and in 16 of them (44%) leukemic cells were found (Table 2). Eleven out of sixteen MID-positive patients had started chemotherapy, while others (n = 5) received OTC before the start of the treatment. Fifteen patients tested negative for MID after starting chemotherapy, while eight patients before chemotherapy. PCR or RT-PCR were used for targeted immunoglobulin (Ig) gene rearrangements alone or in combination with T-cell receptor gamma, ETV6-RUNX1 [25,27], EA2-PBX1 [16,38], BCR-ABL1 [27] or TEL-AML1 fusion transcript [37,38] detection in MID-positive OTs. In half of those patients (n = 8), leukemic cells were detected in OTs prior to and after xenotransplantation as well [16,25,27].
While, in some cases, MID was detectable in OT prior to transplantation into mice, recovered grafts showed an absence of MID [25,27]. MFC targeting leukemia-associated immunophenotype (LAIP) did not detect MID in five patients who received chemotherapy prior to OTC [26]. Immunohistochemistry did not detect any markers in OT of seven patients, though for one of them RT-PCR was positive [37]. One childhood cancer survivor had her first autotransplantation in 2017 prior to receiving a negative result of BCR-ALB transcript in analyzed OT [36].
AML—MID was evaluated using IHC, RT-qPCR, PCR or MFC in OT before and after xenotransplantation in immunodeficient mice. Specific molecular markers identified MID in 3 out of 10 analyzed pediatric AML patients using RT-qPCR and MFC [25,26]. Two of them had started chemotherapy and one did not have treatment prior to OTC. BCR-ABL1 fusion transcript was found in one patient by RT-qPCR in the OT before but not after long-term xenotransplantation (22 weeks) in immunodeficient mice [25]. MFC by LAIP resulted in positive MID in ovarian cortical tissue of 2/3 patients (detected LAIPs: CD13, CD33, CD117, CD65, CD7, HLA DR2, CD34, CD38 or CD13, CD33, CD65, CD117, CD11c) before but not after xenotransplantation (24 weeks) in immunodeficient mice [26]. For the remaining 7/10 patients, PCR, RT-qPCR, IHC or xenotransplantation did not show the presence of cancer cells in OT, though only 1 of them had initiated the treatment before OTC.
CML—MID was analyzed in the OTs of five CML patients in three studies (Table 2). All had their OTs cryopreserved prior to chemotherapy. Malignant cells were identified by BCR-ABL fusion transcript using quantitative RT-PCR in three out of five patients (60%) [35,37]. IHC failed to detect CML cells though RT-PCR was positive. RT-PCR did not identify infiltration of CML cells in OT prior or after xenotransplantation in the remaining two patients [27].
JMML—IHC staining of CD3, CD4 and CD68 revealed no MID in OT of one JMML patient [37]. The patient had no chemotherapy prior to OTC.
Burkitt’s lymphoma—RT-qPCR showed the absence of IgH rearrangements in one patient, though the patient had started chemotherapy [38].

3.3. Central Nervous System (CNS) Tumors

Two studies (Table 3) reported MID testing in OT of patients diagnosed with astrocytoma, ependymoma, germinoma, glioblastoma, medulloblastoma and PNET (primary CNS localization), with only one case being metastasized disease (a PNET patient). Ovarian tissues from 17 patients with a mean age at the time of cryopreservation of 8.2 ± 4.7 years (median 8 years, range 1–17 years) were analyzed. While OTC was performed for all 17 patients before chemotherapy, the slow freezing technique for OTC was reported only for 2 patients. All OTs analyzed were MID-negative by IHC or RT-ddPCR, with molecular markers including NSE and GFAP or GFAP and ENO2, or xenotransplantation [28,29]. However, we assume that patients described in [28] are included in a more recent article [29]. To avoid duplication of patients, only information on how the OTs were analyzed was included in Table 3 and Table S2.
Table 3. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with central nervous system tumors.
Table 3. Summary studies on minimal infiltrative disease in cryopreserved ovarian tissue of girls with central nervous system tumors.
Ref.Study TypePt
(n)
Type of CancerMarkers and Methods (1–5)Detected by Methods
(1–5)
Positive Patients, n (%)Potential BiasOverall Bias
Histology (1)IHC (2)PCR $ (3)Xeno (4)NGS (5)SBABMBDBSCSA*
[29]Prospective study17Astrocytoma, ependymoma, germinoma, glioblastomsa, medulloblastoma, PNETn = 17NSE, GFAP
n = 17
GFAP, ENO2
n = 14
n = 17-ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i002Cancers 15 04199 i003Cancers 15 04199 i002
[28]Case reports3PNETn = 3NSE, GFAP
n = 3
GFAP, ENO2
n = 3
n = 3n = 1ND-Cancers 15 04199 i001Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i003Cancers 15 04199 i001
Grade assessment
Study designLowProspective cohort study in 1/1 (case report is not incorporated in the body summary of evidence since the same patients are in the prospective study)
Study limitations−1Some limitations: SB moderate in 1/1; AB low in 1/1; MB is moderate in 1/1; DB is moderate in 1/2; SC moderate in 1/1; SA low in 1/1
ConsistencyNAOnly one study
Directness−1Though results are direct, population and outcomes cannot be generalized
Precision−1Low number of patients; only one study
Publication bias0Unlikely
Effect size0NA
Dose–response0NA
Plausible confounding+1Treatment as plausible confounder may seriously influence the effect
Quality of evidenceCancers 15 04199 i004Very low
Conclusion Detection of MID in ovarian tissues from central nervous system tumors patients is unlikely despite various attempts to identify it but the results cannot be generalized (1 study, 17 participants)
Cancers 15 04199 i003 = low risk of bias; Cancers 15 04199 i002 = moderate risk of bias; Cancers 15 04199 i001 = high risk of bias; Cancers 15 04199 i004 = very low quality of evidence. AB = attrition bias (the study data available (i.e., participants not lost to follow up) adequately represent the study sample), DB = detection bias (the outcome of interest is measured in a similar way for all participants), FISH = fluorescence in situ hybridization, IHC = immunohistochemistry, L = low risk, M = moderate risk, MB = measurement bias (the prognostic factor is measured in a similar way for all participants), MID = minimally infiltrative disease, ND = not detected, OT = ovarian tissue, OTC = ovarian tissue cryopreservation, PCR = polymerase chain reaction, PNET = primitive neuroectodermal tumor, Pt = patients, SA* = no statistical analysis performed, all primary outcomes are reported, SB = selection bias (the study sample adequately represents the population of interest), SC = study confounding (important potential confounding factors are appropriately accounted for), Xeno = xenotransplantation (and evaluation after). ‘-’ means the method was not used in the specific study; ‘$’ includes all types of PCR: PCR, RT-PCR, quantitative RT-PCR, digital droplet RT-PCR, nested PCR.

4. Discussion

In 17 studies, 25 patients tested positive for MID in the OT of at least 115 pediatric patients diagnosed with solid tumors (Table 1), hematological malignancies (Table 2) and CNS tumors (Table 3). The most commonly used methods for detection of MID in OT were histology, IHC and RT-qPCR. For 62 patients, additionally, OTs were xenotransplanted into immunodeficient mice and reevaluated for MID [16,24,25,26,27,28,29].
A substantial proportion of the included articles focused on EWS, since EWS treatment harbors a high risk of infertility due to the high cyclophosphamide equivalent dose score of the protocols used. Three patients who were identified as being MID-positive had their OT frozen before starting treatment and two were reported to have a localized disease. All reported patients with metastatic disease (n = 11) were found to be negative for MID, while most of them had OTC performed before starting chemotherapy. This is a very important finding, indicating that the initial stage of the disease in EWS is not a major predictive value for MID’s presence in cryopreserved OT and suggests that the diagnosis of disseminated EWS may not preclude offering OTC to the patients. Evaluating possible infiltration in hematological malignancies is another challenge. Since leukemic cells transported with blood flow are supposed to be omnipresent, presumably there is a much higher chance to detect MID prior to the initiation of chemotherapy. On the other hand, leukemic cells are extremely sensitive to chemotherapy and may start disappearing after initiation of treatment [39]. However, we observed that 12 out of 22 MID-positive patients had received chemotherapy prior to OTC and 10 patients had not. In summary, 71 patients had their OTs cryopreserved before treatment and 41 after treatment initiation. Though the moment when the OT was obtained—prior to the initiation of chemotherapy or during the treatment—is important, this confirms that treatment status is not the only confounder in the detection of MID.
Though CNS tumors have a 0.5–18% capacity to metastasize [29], to date only one case of a pediatric brain tumor with ovarian involvement has been reported [40] which makes metastasis relatively uncommon in children. In our systematic review of 17 CNS tumor patients, we identified that 100% of OT fragments were free of cancer cells, even in those who had experienced relapse or died during the treatment later, suggesting that CNS tumors may carry a low risk of ovarian infiltration [28], though larger-scale studies are needed.
The presence of malignant cells in OT is the subject of ongoing research and there are no widely established detection methods due to limited availability of ovarian tissue for analysis. Some general methods include conventional histology and IHC, however, their sensitivity and power are low with a detection limit of 1% or more tumor cells [41]. Targeted molecular-genetic approaches such as PCR or RT-qPCR with sensitivity of 10−3 to 10−6 seem to be better applicable and even broader and deeper screening methods such as RT-ddPCR or next generation sequencing with sensitivity of <10−5 to 10−6 are getting to the front lines in MID detection [42]. To achieve even higher sensitivity, sometimes methods are used in combination, however, it should be based on the disease context and sample availability while collaborating with experts to obtain the best detection power. Future studies to develop and validate the methods for MID are necessary to optimize methods and to enable required sensitivity. In addition, xenotransplantation of OT into immunodeficient animals may be used as a preclinical method to predict relapse [43]. However, xenotransplantation is a time-consuming method and the most important question is how results can be implemented in clinical care. Previous ex vivo experiments have shown that transplanted cryopreserved OT fragments in immunodeficient mice can vanish or decrease in size, thereby complicating analysis of MID [26,27,28]. It has also been reported that transplantation of a small number of cells may be insufficient to cause the disease, which means that the detected MID with highly sensitive methods might not have any clinical relevance, though a positive result should not be ignored [44,45]. Similar observations by other scientists were made when OTC was performed after induction of treatment or after achieving complete remission [45,46]. However, chemotherapy may not exclude malignant cells completely and the risk of them harboring within OT remains [26,27]. It has also been suggested that exploration by histology [24], blood blast count [25] or MCF and PCR-verified infiltrations into distant organs, such as bone marrow, spleen or lymph nodes [26], of xenotransplanted animals may be useful. MFC seems to be a very promising, adaptable technique for searching specific markers in leukemia patients, and it has already been used by some research groups on OT [47], testicular tissue [44], bone marrow or peripheral blood as a predictive tool of clinical outcome [48,49] with sensitivity of 10−3 to 10−4 [42].
This systematic review uncovered gaps of knowledge: no studies have been reported on ovarian involvement in children with renal tumors, hepatoblastomas, retinoblastomas or Hodgkin lymphomas (Table 4). These cancers carry specific molecular (driving) markers that can be used as candidates to identify MID [50]. In addition, the OTs of OS patients have not been tested using specific markers for MID detection or sensitive methods [24,33]. However, there was an attempt to identify molecular markers by FISH or RT-PCR in OS patients but without any success [34] and only later a valuable candidate RB1 as a prognostic factor was proposed [51]. Additionally, Dolmans et al. (2016) [34] claim that if no specific tumor markers are found, xenotransplantation into immunodeficient mice should be used for the detection of malignant cells before any reimplantation. The absence of specific markers at the time of diagnosis for some cancers underscores the importance of a comprehensive analysis of primary tumor tissues or finding new ways of detecting markers. Moreover, this systematic review indicated that the most used freezing technique for OTC was slow freezing. Only one study by Chaput et al., in 2019 [31], compared the impact on RNA yield after freezing OTs by slow freezing and snap freezing techniques. No differences between the two methods that could potentially have influenced MID detection by molecular methods were described/found.

4.1. Strengths and Limitations

To our knowledge, this is the first systematic review on MID in cryopreserved OT of pediatric oncology patients that includes published markers and detection methods used for screening. We summarized 17 selected articles that we considered to be representative of the current state of the field. However, due to variations in techniques for detection, MID markers, metastasis status (in cases of solid and CNS tumors) and time of OTC (prior, during, after the treatment) and low sample sizes (ranging from 1 to 17 patients) in the studies, a meta-analysis was not feasible.

4.2. Implications for Clinical Practice and Research

Two recent reviews described the current status of strategies to safely use cryopreserved OT for fertility restoration in adult [53,54] and prepubertal (<12 years) [54] oncological patients, including in vitro maturation, construction of an artificial ovary, maturation of oocytes, stem cell-based oogenesis and purging. The reviews conclude that in pediatric oncology these methods are all still experimental procedures and require substantial additional research before clinical application. As OTC and subsequent OT transplantation remain the only option for fertility restoration for girls with cancer, this highlights the need for precise MID detection in OT. While encouraging results have been published for young girls with sarcomas and CNS malignancies, we can never exclude the possibility of the harboring of malignant cells within ovarian tissue. Table 5 shows proposed and potential markers and techniques to detect MID according to the markers used in ovarian tissue but also those used for disease detection and/or those of prognostic value. Although some of the proposed and potential markers are usually found in the original tumor tissue and less likely to be found in MID due to the small amount of DNA, with the newest methods such as NGS, even the smallest alterations could be found after careful accuracy and reliability validation. NGS panels after identification of disease-specific targets may be the best approach for MID detection. In this way, it would be possible to reach deeper sequencing coverage, use data streamlining and provide the best personalized medicine approach to the patients [55].

5. Conclusions

In conclusion, testing OTC harvests for MID is important for individual patients. If a patient expresses the desire for autotransplantation of harvested OT, it involves a preliminary MID-based risk assessment depending on their cancer type and molecular markers found at the diagnosis by an expert team. Applying the most reliable methods, as was shown in this systematic review, such as RT-qPCR, FISH or MFC, for determining the absence of MID, or alternatively, in the case of existing MID, purifying the OT from malignant cells to ensure oncologically safe future fertility restoration, is important for the clinical implementation of autotransplantation of cryopreserved OT material in girls with cancer.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15174199/s1, Table S1: GRADE criteria adapted for the systematic review for detection of minimal infiltrative disease in cryopreserved pediatric ovarian tissue; Table S2: Evidence tables of articles included in the systematic review [15,16,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38].

Author Contributions

Conceptualization, M.G., M.E.M.v.d.P., J.R. and M.M.v.d.H.-E.; methodology, M.G. and M.E.M.v.d.P.; formal analysis, M.G. and M.E.M.v.d.P.; investigation, M.G. and M.E.M.v.d.P.; writing—original draft preparation, M.G.; writing—review and editing, M.E.M.v.d.P., J.R., A.M.E.B., A.J.M.M., Z.G. and M.M.v.d.H.-E.; visualization, M.G.; supervision, J.R., A.M.E.B., A.J.M.M., Z.G. and M.M.v.d.H.-E.; project administration, J.R.; funding acquisition, J.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by TREL project, from European Union’s Horizon 2020 research and innovation program under grant number 952438.

Data Availability Statement

All data are presented in the manuscript and Supplementary Materials.

Acknowledgments

We are grateful to the staff from all research centers involved in this study and the TREL project and PubMed library for the support of the systematic review. The TREL project is a collaborative project supported by the Horizon 2020 initiative of the European Commission. Funded by H2020EU.4.b, grant agreement ID: 952438. Project partners are Vilnius University Hospital Santaros Klinikos, St Anna Kinderkrebsforschung Verein, Austria, Universitaetsklinikum Hamburg Eppendorf, Germany, Region Hovedstaden, Denmark, Prinses Máxima Centrum voor kinderoncologie, the Netherlands, Instituto Giannina Gaslini, Italy, Cineca Consorzio Interuniversitario, Italy, Institut Gustave Roussy, France, SIOP Europe ASBL, Belgium. Parts of the graphic abstract were drawn by using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (http://creativecommons.org/licecses/by/3.0/ (accessed on 27 May 2023)).

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Script with keywords and synonyms used for article identification in PubMed database.
Table A1. Script with keywords and synonyms used for article identification in PubMed database.
Script
(((((((((((((((((((((((((((((((((((((((minimal residual disease*[Title/Abstract]) OR (MRD[Title/Abstract])) OR (measurable residual disease*[Title/Abstract])) OR (residual tumor*[Title/Abstract])) OR (residual tumour*[Title/Abstract])) OR (residual minimal disease*[Title/Abstract])) OR (residual disease*[Title/Abstract])) OR (contaminat*[Title/Abstract])) OR (infiltrat*[Title/Abstract])) OR (detect*[Title/Abstract])) OR (molecular residual disease*[Title/Abstract]))) OR (aberration*[Title/Abstract])) OR (fusion transcript*[Title/Abstract])) OR (gene fusion*[Title/Abstract])) OR (translocation*[Title/Abstract])) OR (tumor-specific transcript*[Title/Abstract])) OR (PAX3[Title/Abstract])) OR (PHOX2B[Title/Abstract])) OR (CRMP1[Title/Abstract])) OR (GAP43[Title/Abstract])) OR (ISL1[Title/Abstract])) OR (PAX3- FKHR[Title/Abstract])) OR (PAX7-FKHR[Title/Abstract])) OR (FKHR[Title/Abstract])) OR (PAX7[Title/Abstract])) OR (FOXO1[Title/Abstract])) OR (RASSF1A[Title/Abstract])) OR (CTNNB1[Title/Abstract])) OR (onfFN[Title/Abstract])) OR (EWS-FLI1[Title/Abstract])) OR (EWSR1[Title/Abstract])) OR (FLI1[Title/Abstract])) OR (ETV1[Title/Abstract])) OR (ETV6[Title/Abstract])) OR (CHOP[Title/Abstract])) OR (GADD153[Title/Abstract])) OR (ATF1[Title/Abstract])) AND ((((((((((((((((((cancer*[Title/Abstract]) OR (cancer cell*[Title/Abstract]) OR (tumor*[Title/Abstract])) OR (tumour*[Title/Abstract])) OR (malignan*[Title/Abstract])) OR (neoplasm*[Title/Abstract])) OR (carcinoma*[Title/Abstract])) OR (oncolog*[Title/Abstract])) OR (sarcoma*[Title/Abstract])) OR (blastoma*[Title/Abstract]))) OR (osteosarcoma*[Title/Abstract])) OR (neuroblastoma*[Title/Abstract])) OR (rhabdomyosarcoma*[Title/Abstract])) OR (nephroblastoma*[Title/Abstract])) OR (retinoblastoma*[Title/Abstract])) OR (hepatoblastoma*[Title/Abstract])) OR (Wilms*[Title/Abstract])) OR (ewing[Title/Abstract]))) AND (((((ovarian[Title/Abstract]) OR (ovary[Title/Abstract])) OR (ovaries[Title/Abstract])) OR (gonad*[Title/Abstract])) AND ((((((cryopresev*[Title/Abstract]) OR (frozen[Title/Abstract])) OR (freeze[Title/Abstract])) OR (harvest*[Title/Abstract])) OR (frozen-thawed[Title/Abstract])) OR (thawed[Title/Abstract])))
Table A2. Study inclusion and exclusion criteria.
Table A2. Study inclusion and exclusion criteria.
InclusionExclusion
  • Articles in English
  • Articles on female childhood cancer patients
  • Articles on cryopreserved ovarian tissue
  • Articles on markers of metastasized cancer to ovarian tissue
  • Articles published any time
  • Case reports, case series, original research, communications
  • Articles not in English
  • Articles on male cancer patients
  • Articles on cryopreserved oocytes/embryos or other fertility preservation methods
  • Markers measured only serum or blood
  • Only cell lines
  • Reviews, systematic reviews, literature reviews, narrative reviews, comments, guidelines
  • A full text is not available

References

  1. Corkum, K.S.; Rhee, D.S.; Wafford, Q.E.; Demeestere, I.; Dasgupta, R.; Baertschiger, R.; Malek, M.M.; Aldrink, J.H.; Heaton, T.E.; Weil, B.R.; et al. Fertility and hormone preservation and restoration for female children and adolescents receiving gonadotoxic cancer treatments: A systematic review. J. Pediatr. Surg. 2019, 54, 2200–2209. [Google Scholar] [CrossRef] [PubMed]
  2. Erdmann, F.; Frederiksen, L.E.; Bonaventure, A.; Mader, L.; Hasle, H.; Robison, L.L.; Winther, J.F. Childhood cancer: Survival, treatment modalities, late effects and improvements over time. Cancer Epidemiol. 2021, 71, 101733. [Google Scholar] [CrossRef] [PubMed]
  3. The ESHRE Guideline Group on Female Fertility Preservation; Anderson, R.A.; Amant, F.; Braat, D.; D’Angelo, A.; Lopes, S.M.C.d.S.; Demeestere, I.; Dwek, S.; Frith, L.; Lambertini, M.; et al. ESHRE guideline: Female fertility preservation. Hum. Reprod. Open 2020, 2020, hoaa052. [Google Scholar] [CrossRef] [PubMed]
  4. Marin, L.; Bedoschi, G.; Kawahara, T.; Oktay, K.H. History, Evolution and Current State of Ovarian Tissue Auto-Transplantation with Cryopreserved Tissue: A Successful Translational Research Journey from 1999 to 2020. Reprod. Sci. 2020, 27, 955–962. [Google Scholar] [CrossRef] [PubMed]
  5. van der Perk, M.M.; van der Kooi, A.-L.L.; Bos, A.M.; Broer, S.L.; Veening, M.A.; van Leeuwen, J.; van Santen, H.M.; van Dorp, W.; van den Heuvel-Eibrink, M.M. Oncofertility Perspectives for Girls with Cancer. J. Pediatr. Adolesc. Gynecol. 2022, 35, 523–526. [Google Scholar] [CrossRef]
  6. Gellert, S.E.; Pors, S.E.; Kristensen, S.G.; Bay-Bjørn, A.M.; Ernst, E.; Andersen, C.Y. Transplantation of frozen-thawed ovarian tissue: An update on worldwide activity published in peer-reviewed papers and on the Danish cohort. J. Assist. Reprod. Genet. 2018, 35, 561–570. [Google Scholar] [CrossRef]
  7. McLaughlin, M.; Albertini, D.F.; Wallace, W.H.B.; Anderson, R.A.; Telfer, E.E. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol. Hum. Reprod. 2018, 24, 135–142. [Google Scholar] [CrossRef]
  8. Telfer, E.E. Fertility Preservation: Progress and prospects for developing human immature oocytes in vitro. Reproduction 2019, 158, F45–F54. [Google Scholar] [CrossRef]
  9. Dolmans, M.-M.; Amorim, C.A. FERTILITY PRESERVATION: Construction and use of artificial ovaries. Reproduction 2019, 158, F15–F25. [Google Scholar] [CrossRef]
  10. Clarkson, Y.L.; McLaughlin, M.; Waterfall, M.; Dunlop, C.E.; Skehel, P.A.; Anderson, R.A.; Telfer, E.E. Initial characterisation of adult human ovarian cell populations isolated by DDX4 expression and aldehyde dehydrogenase activity. Sci. Rep. 2018, 8, 6953. [Google Scholar] [CrossRef]
  11. Silvestris, E.; Cafforio, P.; D’oronzo, S.; Felici, C.; Silvestris, F.; Loverro, G. In vitro differentiation of human oocyte-like cells from oogonial stem cells: Single-cell isolation and molecular characterization. Hum. Reprod. 2018, 33, 464–473. [Google Scholar] [CrossRef]
  12. Dolmans, M.-M.; Demylle, D.; Martinez-Madrid, B.; Donnez, J. Efficacy of in vitro fertilization after chemotherapy. Fertil. Steril. 2005, 83, 897–901. [Google Scholar] [CrossRef] [PubMed]
  13. Bastings, L.; Beerendonk, C.C.M.; Westphal, J.R.; Massuger, L.F.A.G.; Kaal, S.E.J.; van Leeuwen, F.E.; Braat, D.D.M.; Peek, R. Autotransplantation of cryopreserved ovarian tissue in cancer survivors and the risk of reintroducing malignancy: A systematic review. Hum. Reprod. Updat. 2013, 19, 483–506. [Google Scholar] [CrossRef]
  14. Dolmans, M.-M.; Luyckx, V.; Donnez, J.; Andersen, C.Y.; Greve, T. Risk of transferring malignant cells with transplanted frozen-thawed ovarian tissue. Fertil. Steril. 2013, 99, 1514–1522. [Google Scholar] [CrossRef] [PubMed]
  15. Schifflers, S.; Delbecque, K.; Galant, C.; Francotte, N.; Philippet, P.; Chantrain, C.F. Microscopic Infiltration of Cryopreserved Ovarian Tissue in 2 Patients with Ewing Sarcoma. J. Pediatr. Hematol. Oncol. 2018, 40, e167–e170. [Google Scholar] [CrossRef]
  16. Díaz-García, C.; Herraiz, S.; Such, E.; Andrés, M.D.M.; Villamón, E.; Mayordomo-Aranda, E.; Cervera, J.V.; Sanz, M.A.; Pellicer, A. Dexamethasone does not prevent malignant cell reintroduction in leukemia patients undergoing ovarian transplant: Risk assessment of leukemic cell transmission by a xenograft model. Hum. Reprod. 2019, 34, 1485–1493. [Google Scholar] [CrossRef]
  17. Mulder, C.L.; Eijkenboom, L.L.; Beerendonk, C.C.M.; Braat, D.D.M.; Peek, R. Enhancing the safety of ovarian cortex autotransplantation: Cancer cells are purged completely from human ovarian tissue fragments by pharmacological inhibition of YAP/TAZ oncoproteins. Hum. Reprod. 2019, 34, 506–518. [Google Scholar] [CrossRef]
  18. Eijkenboom, L.; Mulder, C.; van der Reijden, B.; van Mello, N.; van Leersum, J.; Koorenhof-Scheele, T.; Braat, D.; Beerendonk, C.; Peek, R. Purging human ovarian cortex of contaminating leukaemic cells by targeting the mitotic catastrophe signalling pathway. J. Assist. Reprod. Genet. 2021, 38, 1571–1588. [Google Scholar] [CrossRef]
  19. Moghassemi, S.; Dadashzadeh, A.; Camboni, A.; Feron, O.; Azevedo, R.B.; Amorim, C.A. Photodynamic therapy using OR141-loaded nanovesicles for eradication of leukemic cells from ovarian tissue. Photodiagnosis Photodyn. Ther. 2022, 40, 103139. [Google Scholar] [CrossRef]
  20. Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A web and mobile app for systematic reviews. Syst. Rev. 2016, 5, 210. [Google Scholar] [CrossRef]
  21. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, 71. [Google Scholar]
  22. Hayden, J.A.; Van Der Windt, D.A.; Cartwright, J.L.; Côté, P.; Bombardier, C. Assessing Bias in Studies of Prognostic Factors. Ann. Intern. Med. 2013, 158, 280–286. [Google Scholar] [CrossRef] [PubMed]
  23. Schünemann, H.J.; Oxman, A.D.; Brozek, J.; Glasziou, P.; Jaeschke, R.; Vist, G.E.; Williams, J.W., Jr.; Kunz, R.; Craig, J.; Montori, V.M.; et al. Grading quality of evidence and strength of recommendations for diagnostic tests and strategies. BMJ 2008, 336, 1106–1110. [Google Scholar] [CrossRef]
  24. Greve, T.; Wielenga, V.T.; Grauslund, M.; Sørensen, N.; Christiansen, D.B.; Rosendahl, M.; Andersen, C.Y. Ovarian tissue cryopreserved for fertility preservation from patients with Ewing or other sarcomas appear to have no tumour cell contamination. Eur. J. Cancer 2013, 49, 1932–1938. [Google Scholar] [CrossRef] [PubMed]
  25. Soares, M.; Saussoy, P.; Maskens, M.; Reul, H.; Amorim, C.A.; Donnez, J.; Dolmans, M.-M. Eliminating malignant cells from cryopreserved ovarian tissue is possible in leukaemia patients. Br. J. Haematol. 2017, 178, 231–239. [Google Scholar] [CrossRef]
  26. Zver, T.; Frontczak, S.; Poirot, C.; Rives-Feraille, A.; Leroy-Martin, B.; Koscinski, I.; Arbez-Gindre, F.; Garnache-Ottou, F.; Roux, C.; Amiot, C. Minimal residual disease detection by multicolor flow cytometry in cryopreserved ovarian tissue from leukemia patients. J. Ovarian Res. 2022, 15, 9. [Google Scholar] [CrossRef]
  27. Dolmans, M.-M.; Marinescu, C.; Saussoy, P.; Van Langendonckt, A.; Amorim, C.; Donnez, J. Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood 2010, 116, 2908–2914. [Google Scholar] [CrossRef]
  28. Nguyen, T.Y.T.; Camboni, A.; Masciangelo, R.; Donnez, J.; Dolmans, M.-M. Is Ovarian Tissue Transplantation Safe in Patients with Central Nervous System Primitive Neuroectodermal Tumors? J. Clin. Med. 2020, 9, 4101. [Google Scholar] [CrossRef]
  29. Nguyen, T.Y.T.; Cacciottola, L.; Camboni, A.; Ravau, J.; De Vos, M.; Demeestere, I.; Donnez, J.; Dolmans, M.-M. Ovarian tissue cryopreservation and transplantation in patients with central nervous system tumours. Hum. Reprod. 2021, 36, 1296–1309. [Google Scholar] [CrossRef]
  30. Abir, R.; Feinmesser, M.; Yaniv, I.; Fisch, B.; Cohen, I.J.; Ben-Haroush, A.; Meirow, D.; Felz, C.; Avigad, S. Occasional involvement of the ovary in Ewing sarcoma. Hum. Reprod. 2010, 25, 1708–1712. [Google Scholar] [CrossRef]
  31. Chaput, L.; Grèze, V.; Halle, P.; Radosevic-Robin, N.; Pereira, B.; Véronèse, L.; Lejeune, H.; Durand, P.; Martin, G.; Sanfilippo, S.; et al. Sensitive and Specific Detection of Ewing Sarcoma Minimal Residual Disease in Ovarian and Testicular Tissues in an In Vitro Model. Cancers 2019, 11, 1807. [Google Scholar] [CrossRef]
  32. Andersen, C.Y.; Ernst, E.; Bærentzen, S.; Birkebæk, N.H.; Clausen, N. No malignancy detected in surplus ovarian tissue from a former Ewing sarcoma patient who experienced relapse four years after being grafted with frozen/thawed ovarian tissue. J. Assist. Reprod. Genet. 2014, 31, 1567–1568. [Google Scholar] [CrossRef]
  33. Hoekman, E.J.; Smit, V.T.; Fleming, T.P.; Louwe, L.A.; Fleuren, G.J.; Hilders, C.G. Searching for metastases in ovarian tissue before autotransplantation: A tailor-made approach. Fertil. Steril. 2015, 103, 469–477. [Google Scholar] [CrossRef] [PubMed]
  34. Dolmans, M.; Iwahara, Y.; Donnez, J.; Soares, M.; Vaerman, J.; Amorim, C.; Poirel, H. Evaluation of minimal disseminated disease in cryopreserved ovarian tissue from bone and soft tissue sarcoma patients. Hum. Reprod. 2016, 31, 2292–2302. [Google Scholar] [CrossRef] [PubMed]
  35. Abir, R.; Aviram, A.; Feinmesser, M.; Stein, J.; Yaniv, I.; Parnes, D.; Ben-Haroush, A.; Meirow, D.; Rabizadeh, E.; Fisch, B. Ovarian minimal residual disease in chronic myeloid leukaemia. Reprod. Biomed. Online 2014, 28, 255–260. [Google Scholar] [CrossRef]
  36. Rodriguez-Wallberg, K.A.; Milenkovic, M.; Papaikonomou, K.; Keros, V.; Gustafsson, B.; Sergouniotis, F.; Wikander, I.; Perot, R.; Borgström, B.; Ljungman, P.; et al. Successful pregnancies after transplantation of ovarian tissue retrieved and cryopreserved at time of childhood acute lymphoblastic leukemia—A case report. Haematologica 2021, 106, 2783–2787. [Google Scholar] [CrossRef] [PubMed]
  37. Rosendahl, M.; Andersen, M.T.; Ralfkiær, E.; Kjeldsen, L.; Andersen, M.K.; Andersen, C.Y. Evidence of residual disease in cryopreserved ovarian cortex from female patients with leukemia. Fertil. Steril. 2010, 94, 2186–2190. [Google Scholar] [CrossRef]
  38. Asadi-Azarbaijani, B.; Sheikhi, M.; Nurmio, M.; Tinkanen, H.; Juvonen, V.; Dunkel, L.; Hovatta, O.; Oskam, I.C.; Jahnukainen, K. Minimal residual disease of leukemia and the quality of cryopreserved human ovarian tissue in vitro. Leuk. Lymphoma 2016, 57, 700–707. [Google Scholar] [CrossRef]
  39. Lam, W.A.; Rosenbluth, M.J.; Fletcher, D.A. Chemotherapy exposure increases leukemia cell stiffness. Blood 2007, 109, 3505–3508. [Google Scholar] [CrossRef]
  40. Paterson, E. Distant metastases from medulloblastoma of the cerebellum. Brain 1961, 84, 301–309. [Google Scholar] [CrossRef]
  41. Uemura, S.; Ishida, T.; Thwin, K.K.M.; Yamamoto, N.; Tamura, A.; Kishimoto, K.; Hasegawa, D.; Kosaka, Y.; Nino, N.; Lin, K.S.; et al. Dynamics of Minimal Residual Disease in Neuroblastoma Patients. Front. Oncol. 2019, 9, 455. [Google Scholar] [CrossRef] [PubMed]
  42. Riva, G.; Nasillo, V.; Ottomano, A.M.; Bergonzini, G.; Paolini, A.; Forghieri, F.; Lusenti, B.; Barozzi, P.; Lagreca, I.; Fiorcari, S.; et al. Multiparametric Flow Cytometry for MRD Monitoring in Hematologic Malignancies: Clinical Applications and New Challenges. Cancers 2021, 13, 4582. [Google Scholar] [CrossRef] [PubMed]
  43. Rosendahl, M.; Greve, T.; Andersen, C.Y. The safety of transplanting cryopreserved ovarian tissue in cancer patients: A review of the literature. J. Assist. Reprod. Genet. 2013, 30, 11–24. [Google Scholar] [CrossRef] [PubMed]
  44. Hou, M.; Andersson, M.; Eksborg, S.; Soder, O.; Jahnukainen, K. Xenotransplantation of testicular tissue into nude mice can be used for detecting leukemic cell contamination. Hum. Reprod. 2007, 22, 1899–1906. [Google Scholar] [CrossRef]
  45. Greve, T.; Clasen-Linde, E.; Andersen, M.T.; Andersen, M.K.; Sørensen, S.D.; Rosendahl, M.; Ralfkiær, E.; Andersen, C.Y. Cryopreserved ovarian cortex from patients with leukemia in complete remission contains no apparent viable malignant cells. Blood 2012, 120, 4311–4316. [Google Scholar] [CrossRef]
  46. Chevillon, F.; Clappier, E.; Arfeuille, C.; Cayuela, J.-M.; Dalle, J.H.; Kim, R.; Caye-Eude, A.; Chalas, C.; Abdo, C.; Drouineaud, V.; et al. Minimal residual disease quantification in ovarian tissue collected from patients in complete remission of acute leukemia. Blood 2021, 137, 1697–1701. [Google Scholar] [CrossRef]
  47. Zver, T.; Mouloungui, E.; Berdin, A.; Roux, C.; Amiot, C. Validation of an automated technique for ovarian cortex dissociation: Isolation of viable ovarian cells and their qualification by multicolor flow cytometry. J. Ovarian Res. 2017, 10, 38. [Google Scholar] [CrossRef]
  48. Tembhare, P.R.; Narula, G.; Khanka, T.; Ghogale, S.; Chatterjee, G.; Patkar, N.V.; Prasad, M.; Badrinath, Y.; Deshpande, N.; Gudapati, P.; et al. Post-induction Measurable Residual Disease Using Multicolor Flow Cytometry Is Strongly Predictive of Inferior Clinical Outcome in the Real-Life Management of Childhood T-Cell Acute Lymphoblastic Leukemia: A Study of 256 Patients. Front. Oncol. 2020, 10, 577. [Google Scholar] [CrossRef]
  49. Wang, H.; Zhou, Y.; Huang, X.; Zhang, Y.; Qian, J.; Li, J.; Li, C.; Li, X.; Lou, Y.; Zhu, Q.; et al. Minimal residual disease level determined by flow cytometry provides reliable risk stratification in adults with T-cell acute lymphoblastic leukaemia. Br. J. Haematol. 2021, 193, 1096–1104. [Google Scholar] [CrossRef]
  50. Pfister, S.M.; Reyes-Múgica, M.; Chan, J.K.; Hasle, H.; Lazar, A.J.; Rossi, S.; Ferrari, A.; Jarzembowski, J.A.; Pritchard-Jones, K.; Hill, D.A.; et al. A Summary of the Inaugural WHO Classification of Pediatric Tumors: Transitioning from the Optical into the Molecular Era. Cancer Discov. 2022, 12, 331–355. [Google Scholar] [CrossRef]
  51. Ren, W.; Gu, G. Prognostic implications of RB1 tumour suppressor gene alterations in the clinical outcome of human osteosarcoma: A meta-analysis. Eur. J. Cancer Care 2017, 26, e12401. [Google Scholar] [CrossRef]
  52. Dolmans, M.-M.; Donnez, J. Fertility preservation in women for medical and social reasons: Oocytes vs ovarian tissue. Best Pract. Res. Clin. Obstet. Gynaecol. 2021, 70, 63–80. [Google Scholar] [CrossRef] [PubMed]
  53. Eijkenboom, L.; Saedt, E.; Zietse, C.; Braat, D.; Beerendonk, C.; Peek, R. Strategies to safely use cryopreserved ovarian tissue to restore fertility after cancer: A systematic review. Reprod. Biomed. Online 2022, 45, 763–778. [Google Scholar] [CrossRef]
  54. Kourta, D.; Kanbar, M.; Amorim, C.A.; Wyns, C. Cancer cell contamination and decontamination methods for ovaries and testes: Special focus on prepubertal gonads with a view to safe fertility restoration. Hum. Reprod. 2023, 38, 780–798. [Google Scholar] [CrossRef] [PubMed]
  55. Sánchez, R.; Ayala, R.; Martínez-López, J. Minimal Residual Disease Monitoring with Next-Generation Sequencing Methodologies in Hematological Malignancies. Int. J. Mol. Sci. 2019, 20, 2832. [Google Scholar] [CrossRef]
  56. Gough, S.M.; Slape, C.I.; Aplan, P.D. NUP98 gene fusions and hematopoietic malignancies: Common themes and new biologic insights. Blood 2011, 118, 6247–6257. [Google Scholar] [CrossRef] [PubMed]
  57. Ruteshouser, E.C.; Robinson, S.M.; Huff, V. Wilms tumor genetics: Mutations inWT1, WTX, andCTNNB1 account for only about one-third of tumors. Genes Chromosom. Cancer 2008, 47, 461–470. [Google Scholar] [CrossRef]
  58. Hesson, L.B.; Cooper, W.N.; Latif, F. The Role of RASSF1A Methylation in Cancer. Dis. Markers 2007, 23, 73–87. [Google Scholar] [CrossRef] [PubMed]
  59. Leroy, B.; Ballinger, M.L.; Baran-Marszak, F.; Bond, G.L.; Braithwaite, A.; Concin, N.; Donehower, L.A.; El-Deiry, W.S.; Fenaux, P.; Gaidano, G.; et al. Recommended Guidelines for Validation, Quality Control, and Reporting of TP53 Variants in Clinical Practice. Cancer Res. 2017, 77, 1250–1260. [Google Scholar] [CrossRef]
  60. Calandrini, C.; Schutgens, F.; Oka, R.; Margaritis, T.; Candelli, T.; Mathijsen, L.; Ammerlaan, C.; van Ineveld, R.L.; Derakhshan, S.; de Haan, S.; et al. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nat. Commun. 2020, 11, 1310. [Google Scholar] [CrossRef]
  61. South, A.P.; Cho, R.J.; Aster, J.C. The double-edged sword of Notch signaling in cancer. Semin. Cell Dev. Biol. 2012, 23, 458–464. [Google Scholar] [CrossRef] [PubMed]
  62. Mikhailova, E.; Illarionova, O.; Komkov, A.; Zerkalenkova, E.; Mamedov, I.; Shelikhova, L.; Olshanskaya, Y.; Miakova, N.; Novichkova, G.; Karachunskiy, A.; et al. Reliable Flow-Cytometric Approach for Minimal Residual Disease Monitoring in Patients with B-Cell Precursor Acute Lymphoblastic Leukemia after CD19-Targeted Therapy. Cancers 2022, 14, 5445. [Google Scholar] [CrossRef] [PubMed]
  63. Dondero, A.; Morini, M.; Cangelosi, D.; Mazzocco, K.; Serra, M.; Spaggiari, G.M.; Rotta, G.; Tondo, A.; Locatelli, F.; Castellano, A.; et al. Multiparametric flow cytometry highlights B7-H3 as a novel diagnostic/therapeutic target in GD2neg/low neuroblastoma variants. J. Immunother. Cancer 2021, 9, e002293. [Google Scholar] [CrossRef] [PubMed]
  64. Garcia, D.R.N.; Arancibia, A.M.; Ribeiro, R.C.; Land, M.G.P.; Silva, M.L.M. Intrachromosomal amplification of chromosome 21 (iAMP21) detected by ETV6/RUNX1 FISH screening in childhood acute lymphoblastic leukemia: A case report. Rev. Bras. Hematol. Hemoter. 2013, 35, 369–371. [Google Scholar] [CrossRef] [PubMed]
  65. Meyer, C.; Burmeister, T.; Gröger, D.; Tsaur, G.; Fechina, L.; Renneville, A.; Sutton, R.; Venn, N.C.; Emerenciano, M.; Pombo-De-Oliveira, M.S.; et al. The MLL recombinome of acute leukemias in 2017. Leukemia 2018, 32, 273–284. [Google Scholar] [CrossRef]
  66. Roberts, K.G.; Li, Y.; Payne-Turner, D.; Harvey, R.C.; Yang, Y.-L.; Pei, D.; McCastlain, K.; Ding, L.; Lu, C.; Song, G.; et al. Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2014, 371, 1005–1015. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, D.; Zhu, G.; Wang, N.; Zhou, X.; Yang, Y.; Zhou, S.; Xiong, J.; He, J.; Jiang, L.; Li, C.; et al. SIL-TAL1 Rearrangement is Related with Poor Outcome: A Study from a Chinese Institution. PLoS ONE 2013, 8, e73865. [Google Scholar] [CrossRef]
  68. Hurteau, G.J.; Broome, J.D.; Brock, G.J. Accurate detection of Asparagine Synthetase (ASNS) using quantitative real-time PCR (qRT-PCR), without requiring DNaseI treatment. Leukemia 2005, 19, 2368–2370. [Google Scholar] [CrossRef]
  69. Junmei, Z.; Fengkuan, Y.; Yongping, S.; Baijun, F.; Yuzhang, L.; Lina, L.; Qinglan, Z. Coexistence of P190 and P210 BCR/ABL transcripts in chronic myeloid leukemia blast crisis resistant to imatinib. Springerplus 2015, 4, 170. [Google Scholar] [CrossRef]
  70. Chatterjee, T.; Gupta, S.; Sharma, A.; Sharma, S.; Ganguli, P. Distribution of Different PML/RARα bcr Isoforms in Indian Acute Promyelocytic Leukemia (APL) Patients and Clinicohema-tological Correlation. Mediterr. J. Hematol. Infect. Dis. 2014, 6, e2014004. [Google Scholar] [CrossRef]
  71. Talami, A.; Bettelli, F.; Pioli, V.; Giusti, D.; Gilioli, A.; Colasante, C.; Galassi, L.; Giubbolini, R.; Catellani, H.; Donatelli, F.; et al. How to Improve Prognostication in Acute Myeloid Leukemia with CBFB-MYH11 Fusion Transcript: Focus on the Role of Molecular Measurable Residual Disease (MRD) Monitoring. Biomedicines 2021, 9, 953. [Google Scholar] [CrossRef]
  72. Cho, E.K.; Bang, S.M.; Ahn, J.Y.; Yoo, S.M.; Park, P.W.; Seo, Y.H.; Shin, D.B.; Lee, J.H. Prognostic Value of AML1/ETO Fusion Transcripts in Patients with Acute Myelogenous Leukemia. Korean J. Intern. Med. 2003, 18, 13–20. [Google Scholar] [CrossRef] [PubMed]
  73. Wilhelmsson, M.; Glosli, H.; Ifversen, M.; Abrahamsson, J.; Winiarski, J.; Jahnukainen, K.; Hasle, H.; Nordic Society of Pediatric Hematology and Oncology (NOPHO). Long-term health outcomes in survivors of childhood AML treated with allogeneic HSCT: A NOPHO–AML Study. Bone Marrow Transplant. 2019, 54, 726–736. [Google Scholar] [CrossRef] [PubMed]
  74. Noort, S.; Zimmermann, M.; Reinhardt, D.; Cuccuini, W.; Pigazzi, M.; Smith, J.; Ries, R.E.; Alonzo, T.A.; Hirsch, B.; Tomizawa, D.; et al. Prognostic impact of t(16;21)(p11;q22) and t(16;21)(q24;q22) in pediatric AML: A retrospective study by the I-BFM Study Group. Blood 2018, 132, 1584–1592. [Google Scholar] [CrossRef]
  75. Kim, H.J.; Baek, S.K.; Maeng, C.H.; Kim, S.-Y.; Park, T.S.; Han, J.J. Acute myeloid leukemia with t(11;19)(q23;p13.1) in a patient with a gastrointestinal stromal tumor undergoing imatinib therapy: A case report. World J. Clin. Cases 2020, 8, 1251–1256. [Google Scholar] [CrossRef]
  76. Dastugue, N.; Lafage-Pochitaloff, M.; Pagès, M.-P.; Radford, I.; Bastard, C.; Talmant, P.; Mozziconacci, M.J.; Léonard, C.; Bilhou-Nabéra, C.; Cabrol, C.; et al. Cytogenetic profile of childhood and adult megakaryoblastic leukemia (M7): A study of the Groupe Francais de Cytogenetique Hematologique (GFCH). Blood 2002, 100, 618–626. [Google Scholar] [CrossRef]
  77. Smith, M.L.; Cavenagh, J.D.; Lister, T.A.; Fitzgibbon, J. Mutation of CEBPA in Familial Acute Myeloid Leukemia. N. Engl. J. Med. 2004, 351, 2403–2407. [Google Scholar] [CrossRef]
  78. Noort, S.; van Oosterwijk, J.; Ma, J.; Garfinkle, E.A.; Nance, S.; Walsh, M.; Song, G.; Reinhardt, D.; Pigazzi, M.; Locatelli, F.; et al. Analysis of rare driving events in pediatric acute myeloid leukemia. Haematologica 2023, 108, 48–60. [Google Scholar] [CrossRef] [PubMed]
  79. Hitzler, J.K.; Cheung, J.; Li, Y.; Scherer, S.W.; Zipursky, A. GATA1 mutations in transient leukemia and acute megakaryoblastic leukemia of Down syndrome. Blood 2003, 101, 4301–4304. [Google Scholar] [CrossRef] [PubMed]
  80. Goemans, B.F.; Noort, S.; Blink, M.; Wang, Y.-D.; Peters, S.T.C.J.; van Wouwe, J.P.; Kaspers, G.; de Haas, V.; Kollen, W.J.; van der Velden, V.H.J.; et al. Sensitive GATA1 mutation screening reliably identifies neonates with Down syndrome at risk for myeloid leukemia. Leukemia 2021, 35, 2403–2406. [Google Scholar] [CrossRef]
  81. Damm-Welk, C.; Klapper, W.; Oschlies, I.; Gesk, S.; Röttgers, S.; Bradtke, J.; Siebert, R.; Reiter, A.; Woessmann, W. Distribution of NPM1-ALK and X-ALK fusion transcripts in paediatric anaplastic large cell lymphoma: A molecular-histological correlation. Br. J. Haematol. 2009, 146, 306–309. [Google Scholar] [CrossRef] [PubMed]
  82. Chuang, S.-S.; Lin, C.-N.; Shen, F.-C.; Liao, P.-J.; Liao, Y.-L.; Chang, J.H.; Tsai, Y.-C.; Cho, C.-Y.; Huang, W. Detecting Clonal Rearrangement in Non-Hodgkin’s Lymphomas in Taiwan by Polymerase Chain Reaction. Leuk. Lymphoma 2003, 44, 117–121. [Google Scholar] [CrossRef]
  83. Green, M.R.; Monti, S.; Rodig, S.J.; Juszczynski, P.; Currie, T.; O’Donnell, E.; Chapuy, B.; Takeyama, K.; Neuberg, D.; Golub, T.R.; et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010, 116, 3268–3277. [Google Scholar] [CrossRef] [PubMed]
  84. Almire, C.; Bertrand, P.; Ruminy, P.; Maingonnat, C.; Wlodarska, I.; Martín-Subero, J.I.; Siebert, R.; Tilly, H.; Bastard, C. PVRL2 is translocated to theTRA@ locus in t(14;19)(q11;q13)-positive peripheral T-cell lymphomas. Genes Chromosom. Cancer 2007, 46, 1011–1018. [Google Scholar] [CrossRef] [PubMed]
  85. Wegert, J.; Ishaque, N.; Vardapour, R.; Geörg, C.; Gu, Z.; Bieg, M.; Ziegler, B.; Bausenwein, S.; Nourkami, N.; Ludwig, N.; et al. Mutations in the SIX1/2 Pathway and the DROSHA/DGCR8 miRNA Microprocessor Complex Underlie High-Risk Blastemal Type Wilms Tumors. Cancer Cell 2015, 27, 298–311. [Google Scholar] [CrossRef] [PubMed]
  86. Williams, R.D.; Al-Saadi, R.; Chagtai, T.; Popov, S.; Messahel, B.; Sebire, N.; Gessler, M.; Wegert, J.; Graf, N.; Leuschner, I.; et al. Subtype-Specific FBXW7 Mutation and MYCN Copy Number Gain in Wilms’ Tumor. Clin. Cancer Res. 2010, 16, 2036–2045. [Google Scholar] [CrossRef]
  87. Hol, J.A.; Kuiper, R.P.; van Dijk, F.; Waanders, E.; van Peer, S.E.; Koudijs, M.J.; Bladergroen, R.; van Reijmersdal, S.V.; Morgado, L.M.; Bliek, J.; et al. Prevalence of (Epi)genetic Predisposing Factors in a 5-Year Unselected National Wilms Tumor Cohort: A Comprehensive Clinical and Genomic Characterization. J. Clin. Oncol. 2022, 40, 1892–1902. [Google Scholar] [CrossRef]
  88. Ooms, A.H.; Vujanić, G.M.; D’hooghe, E.; Collini, P.; L’herminé-Coulomb, A.; Vokuhl, C.; Graf, N.; Heuvel-Eibrink, M.M.v.D.; de Krijger, R.R. Renal Tumors of Childhood—A Histopathologic Pattern-Based Diagnostic Approach. Cancers 2020, 12, 729. [Google Scholar] [CrossRef]
  89. Roy, A.; Kumar, V.; Zorman, B.; Fang, E.; Haines, K.M.; Doddapaneni, H.; Hampton, O.A.; White, S.; Bavle, A.A.; Patel, N.R.; et al. Recurrent internal tandem duplications of BCOR in clear cell sarcoma of the kidney. Nat. Commun. 2015, 6, 8891. [Google Scholar] [CrossRef]
  90. Gooskens, S.L.; Gadd, S.; Auvil, J.M.G.; Gerhard, D.S.; Khan, J.; Patidar, R.; Meerzaman, D.; Chen, Q.-R.; Hsu, C.H.; Yan, C.; et al. TCF21 hypermethylation in genetically quiescent clear cell sarcoma of the kidney. Oncotarget 2015, 6, 15828–15841. [Google Scholar] [CrossRef]
  91. Xin, J.; Xu, R.; Lin, S.; Xin, M.; Cai, W.; Zhou, J.; Fu, C.; Zhen, G.; Lai, J.; Li, Y.; et al. Clinical potential of TCF21 methylation in the diagnosis of renal cell carcinoma. Oncol. Lett. 2016, 12, 1265–1270. [Google Scholar] [CrossRef] [PubMed]
  92. Li, J.; Zhang, W.; Hu, H.; Zhang, Y.; Wang, Y.; Gu, H.; Huang, D. Case Analysis of 14 Children with Malignant Rhabdoid Tumor of the Kidney. Cancer Manag. Res. 2021, 13, 4865–4872. [Google Scholar] [CrossRef] [PubMed]
  93. Zhanghuang, C.; Chen, S.; Li, L.; Yang, Z.; Xie, Y.; Li, J.; Tang, H.; He, X.; Dong, L.; Yan, B. Clinical and Molecular Differentiation Between Malignant Rhabdoid Tumor of the Kidney and Normal Tissue: A Two-Case Report. Front. Oncol. 2021, 11, 659709. [Google Scholar] [CrossRef]
  94. Argani, P.; Zhong, M.; Reuter, V.E.; Fallon, J.T.; Epstein, J.I.; Netto, G.J.; Antonescu, C.R. TFE3-Fusion Variant Analysis Defines Specific Clinicopathologic Associations Among Xp11 Translocation Cancers. Am. J. Surg. Pathol. 2016, 40, 723–737. [Google Scholar] [CrossRef]
  95. Cajaiba, M.M.; Dyer, L.M.; Geller, J.I.; Jennings, L.J.; George, D.; Kirschmann, D.; Rohan, S.M.; Cost, N.G.; Khanna, G.; Mullen, E.A.; et al. The classification of pediatric and young adult renal cell carcinomas registered on the children’s oncology group (COG) protocol AREN03B2 after focused genetic testing. Cancer 2018, 124, 3381–3389. [Google Scholar] [CrossRef]
  96. Cajaiba, M.M.; Jennings, L.J.; Rohan, S.M.; Perez-Atayde, A.R.; Marino-Enriquez, A.; Fletcher, J.A.; Geller, J.I.; Leuer, K.M.C.; Bridge, J.A.; Perlman, E.J. ALK-rearranged renal cell carcinomas in children. Genes Chromosom. Cancer 2016, 55, 442–451. [Google Scholar] [CrossRef] [PubMed]
  97. Somasundaram, D.B.; Aravindan, S.; Yu, Z.; Jayaraman, M.; Tran, N.T.B.; Li, S.; Herman, T.S.; Aravindan, N. Droplet digital PCR as an alternative to FISH for MYCN amplification detection in human neuroblastoma FFPE samples. BMC Cancer 2019, 19, 106. [Google Scholar] [CrossRef]
  98. Hung, Y.P.; Lee, J.P.; Bellizzi, A.M.; Hornick, J.L. PHOX2B reliably distinguishes neuroblastoma among small round blue cell tumours. Histopathology 2017, 71, 786–794. [Google Scholar] [CrossRef]
  99. Lee, N.H.; Son, M.H.; Choi, Y.B.; Yi, E.; Lee, J.W.; Yoo, K.H.; Sung, K.W.; Koo, H.H. Clinical Significance of Tyrosine Hydroxylase mRNA Transcripts in Peripheral Blood at Diagnosis in Patients with Neuroblastoma. Cancer Res. Treat. 2016, 48, 1399–1407. [Google Scholar] [CrossRef]
  100. van Zogchel, L.M.J.; Zappeij-Kannegieter, L.; Javadi, A.; Lugtigheid, M.; Gelineau, N.U.; Lak, N.S.M.; Zwijnenburg, D.A.; Koster, J.; Stutterheim, J.; van der Schoot, C.E.; et al. Specific and Sensitive Detection of Neuroblastoma mRNA Markers by Multiplex RT-qPCR. Cancers 2021, 13, 150. [Google Scholar] [CrossRef]
  101. Inomistova, M.V.; Svergun, N.M.; Khranovska, N.M.; Skachkova, O.V.; Gorbach, O.I.; Klymnyuk, G.I. Prognostic significance of MDM2 gene expression in childhood neuroblastoma. Exp. Oncol. 2015, 37, 111–115. [Google Scholar] [CrossRef]
  102. Cheung, N.-K.V.; Zhang, J.; Lu, C.; Parker, M.; Bahrami, A.; Tickoo, S.K.; Heguy, A.; Pappo, A.S.; Federico, S.; Dalton, J.; et al. Association of Age at Diagnosis and Genetic Mutations in Patients with Neuroblastoma. JAMA 2012, 307, 1062–1071. [Google Scholar] [CrossRef]
  103. Mossé, Y.P.; Laudenslager, M.; Longo, L.; Cole, K.A.; Wood, A.; Attiyeh, E.F.; Laquaglia, M.J.; Sennett, R.; Lynch, J.E.; Perri, P.; et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008, 455, 930–935. [Google Scholar] [CrossRef]
  104. Kenny, C.; Bausenwein, S.; Lazaro, A.; Furtwängler, R.; Gooskens, S.L.; van den Heuvel Eibrink, M.; Vokuhl, C.; Leuschner, I.; Graf, N.; Gessler, M.; et al. Mutually exclusive BCOR internal tandem duplications and YWHAE-NUTM2 fusions in clear cell sarcoma of kidney: Not the full story. J. Pathol. 2016, 238, 617–620. [Google Scholar] [CrossRef]
  105. Lak, N.S.; Voormanns, T.L.; Zappeij-Kannegieter, L.; van Zogchel, L.M.; Fiocco, M.; van Noesel, M.M.; Merks, J.H.; van der Schoot, C.E.; Tytgat, G.A.; Stutterheim, J. Improving Risk Stratification for Pediatric Patients with Rhabdomyosarcoma by Molecular Detection of Disseminated Disease. Clin. Cancer Res. 2021, 27, 5576–5585. [Google Scholar] [CrossRef]
  106. Butel, T.; Karanian, M.; Pierron, G.; Orbach, D.; Ranchere, D.; Cozic, N.; Galmiche, L.; Coulomb, A.; Corradini, N.; Lacour, B.; et al. Integrative clinical and biopathology analyses to understand the clinical heterogeneity of infantile rhabdomyosarcoma: A report from the French MMT committee. Cancer Med. 2020, 9, 2698–2709. [Google Scholar] [CrossRef]
  107. Arden, K.C.; Anderson, M.J.; Finckenstein, F.G.; Czekay, S.; Cavenee, W.K. Detection of the t(2;13) chromosomal translocation in alveolar rhabdomyosarcoma using the reverse transcriptase-polymerase chain reaction. Genes Chromosom. Cancer 1996, 16, 254–260. [Google Scholar] [CrossRef] [PubMed]
  108. Van Paemel, R.; De Koker, A.; Vandeputte, C.; van Zogchel, L.; Lammens, T.; Laureys, G.; Vandesompele, J.; Schleiermacher, G.; Chicard, M.; Van Roy, N.; et al. Minimally invasive classification of paediatric solid tumours using reduced representation bisulphite sequencing of cell-free DNA: A proof-of-principle study. Epigenetics 2021, 16, 196–208. [Google Scholar] [CrossRef]
  109. Orr, B.A. Pathology, diagnostics, and classification of medulloblastoma. Brain Pathol. 2020, 30, 664–678. [Google Scholar] [CrossRef] [PubMed]
  110. Timmerman, D.M.; Eleveld, T.F.; Sriram, S.; Dorssers, L.C.; Gillis, A.J.; Schmidtova, S.; Kalavska, K.; van de Werken, H.J.; Oing, C.; Honecker, F.; et al. Chromosome 3p25.3 Gain Is Associated with Cisplatin Resistance and Is an Independent Predictor of Poor Outcome in Male Malignant Germ Cell Tumors. J. Clin. Oncol. 2022, 40, 3077–3087. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram of the study selection process. The total number of full-text articles screened was 195; 17 of them were included in the systematic review and 178 were excluded. Assembled according to PRISMA guidelines [21].
Figure 1. Flow diagram of the study selection process. The total number of full-text articles screened was 195; 17 of them were included in the systematic review and 178 were excluded. Assembled according to PRISMA guidelines [21].
Cancers 15 04199 g001
Table 4. Reported MID in harvested ovarian tissue involvement in cancer patients. MID—minimal infiltrative disease.
Table 4. Reported MID in harvested ovarian tissue involvement in cancer patients. MID—minimal infiltrative disease.
Incidence RateTypes of Cancer in AdultsTypes of Cancer in Children
High risk
>11%
Leukemia, Burkitt’s lymphoma,
neuroblastoma
Ewing sarcoma, acute lymphoblastic leukemia, acute myeloid leukemia, chronic myeloid
leukemia
Moderate risk
0.2–11%
Cervical adenocarcinoma, breast cancer (infiltrating lobular subtype, stage IV), Ewing’s sarcoma, non-Hodgkin
lymphoma, colon cancer
Low risk
<0.2%
Remaining malignanciesOsteosarcoma, rhabdomyosarcoma, synovial sarcoma, clear cell sarcoma, Burkitt’s
lymphoma, juvenile myelomonocytic leukemia, central nervous system tumors
Classification of MID incidence in adults is from [52]; suggestion of classification of MID incidence rates in pediatric patients is according to the findings of this systematic review.
Table 5. Proposed and potential markers with techniques to detect minimal infiltrative disease in cryopreserved ovarian tissue of girls with specific malignancies.
Table 5. Proposed and potential markers with techniques to detect minimal infiltrative disease in cryopreserved ovarian tissue of girls with specific malignancies.
MalignancyProposed MarkersPotential TechniquesReferences
OverallABL class abnormalitiesRT-qPCR[25,27]
NUP98 gene fusion transcriptsRT-PCR[56]
WT1 mutationsPCR, qPCR[57]
RASSF1A hypermethylationQuantitative MSP[58]
P53 mutations, transcript levelsNGS, RNA sequencing[59,60]
Notch aberrationsSequencing[61]
CD19, CD20, CD22MFC, NGS, RT-qPCR[62]
GD2, B7H3 MFC[63]
Acute lymphoblastic leukemiaPhiladelphia chromosome-positive (Ph+; BCR-ABL1 fusion gene)PCR, RT-PCR[25,36]
(TCF3) E2A fusion genesRT-qPCR;RT-ddPCR[16,38]
CD34, CD10, CD20, CD79a, CD3, TdTIHC[37]
CD19, CD34, CD10-, negative for myeloid markers, CD45, HLA-DR2, CD10, CD19, CD22, CD33, CD38, CD2, cyCD3, CD5, CD7, CD33, CD45RA, CD123MFC[26]
Ig(H/K)/TCR targetsRT-PCR, RT-ddPCR[16,25,27,38]
RUNX1 (AML1)translocations, fusion genesRT-PCR, FISH, RT-qPCR, PCR[25,37,38,64]
MLL (KMT2A) rearrangements, patient-specific on DNART-qPCR, long-distance inverse PCR[38,65]
Intrachromosomal amplification of chromosome 21 (iAMP21)FISH[64]
IKZF1 mutations Sequencing[66]
SIL-TAL1 fusion genePCR[67]
Asparagine synthetase RT-qPCR[68]
Chronic myeloid leukemiaBCR-ABL fusion geneFISH, RT-PCR, PCR[27,37,69]
MPO, glycophorin A, CD34, CD68, CD117, LCA/CD45, Factor VIIIIHC[35,37]
Acute promyelocytic leukemiaPML-RARα brc isoformsRT-qPCR, FISH[70]
Acute myeloid leukemiaRUNX1 (AML1) fusion genesRT-PCR, RT-qPCR[38,71,72]
NPM1 mutationsRT-qPCR[38]
FLT3fusion transcriptsRT-PCR[25]
BCR-ABL1 fusion genesRT-PCR[25]
CD34, CD68, CD117, MPO, CD13, CD4IHC[37]
CD34, CD33, CD13, CD117, CD38, CD65, CD7, HLA-DR2, CD11cMFC[26]
CBFB-MYH11 fusion genesPCR, RT-qPCR[37,71]
NUP98 rearrangementsRT-qPCR, NGS[56,73]
FUS-ERG fusion geneFISH, PCR, RNA sequencing[74]
KMT2A rearrangementsRT-qPCR, FISH, RT-PCR; long-distance inverse PCR[65,75]
RBM15-MKL1 (OTT-MAL) fusion gene/transcriptRT-PCR[76]
CEBPA mutationsPCR, sequencing[77]
UBTF mutationsNGS[78]
Acute myeloid leukemia–Down
syndrome
GATA1 mutations, patient-specific on DNASequencing[79,80]
Anaplastic large cell lymphomaALK fusion genes, NPM1 mutationsRT-PCR, FISH, immunofluorescence[78,81]
Juvenile myelomonocytic leukemiaCD3, CD4, CD68IHC[37]
Lymphoma (Hodgkin and non-Hodgkin)Immunoglobulin (Ig) or T-cell receptor (TCR) gene rearrangementsRT-qPCR[38]
Multiplex PCR[82]
9p24 amplification/JAK2DNA copy number analysis, RT-qPCR[83]
14q11/TRA/DFISH[84]
Wilms tumorWT1, WTX mutationsPCR, qPCR [57]
CTNNB1 mutationsqPCR[57]
DROSHA/DGCR8 mutationsSequencing (DNA, RNA)[85]
SIX1/SIX2 mutationsSequencing (DNA, RNA)[85]
DICER1 mutationsSequencing (DNA, RNA)[85]
DIS3L2 mutationsSequencing (DNA, RNA)[85]
FBXW7 mutationsChromosome copy number profiling, sequencing[86]
DIS3L2 mutationsPCR[87]
TP53 mutationsNGS[88]
Clear cell sarcoma of the kidneyBCOR internal tandem duplicationsRNA sequencing, PCR, IHC, FISH[88,89]
YWHAE-NUTM2 fusion transcript RT-PCR[90]
TCF21 hypermethylationMethylation-based methods,
quantitative pyrosequencing methylation analysis
[90,91]
Malignant rhabdoid tumor of the kidneySMARCA4; SMARCB1IHC, WGS[92,93]
Renal cell carcinomaTFE3 (Xp11) translocations/fusion transcriptsFISH, WGS/NGS, IHC, RNA sequencing[60,88,94,95]
TFEB translocations/fusion transcriptsRNA sequencing, FISH, IHC[60,88,95]
ALK rearrangementsIHC, FISH, RT-PCR, NGS[96]
NeuroblastomaMYCN amplification/mutationsFISH, ddPCR, NGS[88,97]
PHOX2BIHC[98]
THRT-qPCR, multiplex RT-qPCR[99]
CHRNA3, DBH, GAP43, POSTN, PRRX1 and FMO3multiplex RT-qPCR[100]
MDM2RT-qPCR[101]
ATRX mutations WGS, IHC[102]
ALK mutationsSequencing, qPCR[103]
Synovial sarcomaBcl-2, SYT-SSX fusion geneIHC, RT-qPCR[34]
Clear cell sarcomaMelanoma cocktail/S100IHC[34]
EWS-ATF1 fusion geneRT-qPCR[34]
BROC mutationsRT-qPCR[104]
YWHAE-NUTM2 fusion transcriptRT-qPCR[104]
Ewing SarcomaEWS fusion transcriptsRT-qPCR[15,24,30,31,32,33,34]
t(X;22) EWS translocationsFISH[34,88]
CD99, INI1IHC[34,88]
OsteosarcomaRB1 deletionIHC, PCR, RT-PCR[51]
RhabdomyosarcomaPAX3/7-FOXO1 fusion geneRT-qPCR[34,105]
MYOD1IHC, RT-qPCR[34,105]
MYOGENINIHC, RT-qPCR[34,105]
VGLL2 fusion transcriptsRT-PCR, RNA sequencing[106]
NTRK fusion transcriptsRT-PCR, RNA sequencing[106]
(B)RAF fusion transcriptsRT-PCR, RNA sequencing[106]
t(2;13) translocation alveolarRT-PCR[107]
cfRRBSshWGS[108]
MedulloblastomaGFAP/NSEIHC[29]
Alterations in WNT and SHH pathways’ componentsFISH, sequencing, methylation-based methods and combinations of them[109]
AstrocytomaCD99/NSE
GFAP
IHC, RT-ddPCR[29]
EpendymomaGFAP/NSE
GFAP
IHC, RT-ddPCR[29]
GerminomaGFAP/NSE
GFAP
IHC, RT-ddPCR[29]
GlioblastomaGFAP/NSE
GFAP
IHC, RT-ddPCR[29]
Primitive neuroectodermal tumorGFAP/NSE
GFAP
IHC, RT-ddPCR[29]
Germ cell tumorsChromosome 3p gain, miRNANGS, RT-qPCR[110]
The markers and techniques in bold have been reported for testing in ovarian tissue (results from this review), the markers and techniques not in bold have not yet been described for detection in ovarian tissue but are used in the diagnostic setting. ddPCR—digital droplet PCR, FISH—fluorescence in situ hybridization, IHC—immunohistochemistry, MFC—multicolor flow cytometry, MSP—methylation-specific PCR, NGS—next generation sequencing, PCR—polymerase chain reaction, qPCR—quantitative PCR, RNA—ribonucleic acid, RT-PCR—real-time PCR, RT-qPCR—real-time quantitative PCR, shWGS—shallow WGS, SVS—synovial sarcoma, WGS—whole genome sequencing.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Grubliauskaite, M.; van der Perk, M.E.M.; Bos, A.M.E.; Meijer, A.J.M.; Gudleviciene, Z.; van den Heuvel-Eibrink, M.M.; Rascon, J. Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review. Cancers 2023, 15, 4199. https://doi.org/10.3390/cancers15174199

AMA Style

Grubliauskaite M, van der Perk MEM, Bos AME, Meijer AJM, Gudleviciene Z, van den Heuvel-Eibrink MM, Rascon J. Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review. Cancers. 2023; 15(17):4199. https://doi.org/10.3390/cancers15174199

Chicago/Turabian Style

Grubliauskaite, Monika, M. E. Madeleine van der Perk, Annelies M. E. Bos, Annelot J. M. Meijer, Zivile Gudleviciene, Marry M. van den Heuvel-Eibrink, and Jelena Rascon. 2023. "Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review" Cancers 15, no. 17: 4199. https://doi.org/10.3390/cancers15174199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop