TO THE EDITOR:

Pediatric acute lymphoblastic leukemia (ALL) is characterized by recurrent preleukemic chromosomal translocations that emerge frequently in utero.1  The most common translocation t(12;21) occurs in 25% of B-cell precursor ALL and results in the formation of the chimeric transcription factor ETV6-RUNX1. Secondary oncogenic hits acquired postnatally are necessary to develop overt leukemia.2,3  Therefore, the number of newborns harboring a preleukemic translocation is expected to equal or exceed the incidence of the corresponding leukemia.

In 2002, Mel Greaves’ group analyzed the frequency of ETV6-RUNX1 translocations in human newborns by independent reverse transcriptase (RT)–polymerase chain reaction (PCR) and quantitative PCR (qPCR) screens confirmed by multicolor fluorescence in situ hybridization. They observed that ≈1% of cord bloods from newborns (n = 6 of 567) contained ETV6-RUNX1+ cells.4  The translocation seemed to occur frequently during normal fetal development and to be 100-fold more common than the corresponding leukemia (1 in 10 000 children). This indicated a very low oncogenic potential of the ETV6-RUNX1 transcription factor and suggested that the usefulness of preventive screenings of ETV6-RUNX1–carrying newborns is limited because 99% would never develop the disease.

Since this initial investigation, several studies have carefully analyzed the incidence of the translocation in newborns, children, and adults using fresh or frozen cord blood or peripheral blood (summarized in supplemental Table 1, available on the Blood Web site).4-14  Remarkably, the suggested incidence ranged from 0.01% (equaling the corresponding leukemia rate)7-9  to ≈8% (exceeding it by a factor of 800).5  Because the investigated cohorts and specimens were similar, the variation was assumed to have technical explanations. Also, the frequency of preleukemic cells in healthy blood or cord blood was controversially discussed, ranging from <10−5 to 10−3.

All of the previous studies used RNA and RT-PCR for prevalence determination. A drawback of RT-PCR is the possible generation of false-positive results caused by contamination. These false-positive results cannot be distinguished from real ones, as for a given translocation the identical amplification products are created in different samples.9  In addition, low levels of positivity can be due to RNA instability, different processing of RNA, detection methods, and, possibly, low expression of ETV6-RUNX1 transcripts in cord blood.

To analyze the frequency of ETV6-RUNX1+ preleukemic cells in newborns, we therefore developed a DNA-based method termed genomic inverse PCR for exploration of ligated breakpoints (GIPFEL)15  because DNA, in contrast to RNA, is very stable. By DNA-based approaches, patient-specific amplification products of the translocation site that differ from one another in size and sequence are obtained and contaminations can be easily identified (Figure 1A). The advantages of DNA-based screening methods have led to an increased use in clinical applications (eg, DNA-based monitoring of minimal residual disease in BCR-ABL+ leukemia16 ).

Figure 1.

The incidence of ETV6-RUNX1 fusions in healthy newborns can be determined by DNA-based GIPFEL screening. (A) Only DNA- and not RNA-based ETV6-RUNX1 detection provides individual-specific breakpoints. The breakpoint cluster regions (BCRs; horizontal lines, exact regions are marked by dotted lines, sizes are given in base pairs) of ETV6 (top panel, intron 5) and RUNX1 (bottom panel, intron 1 and 2) are presented. Black boxes indicate exons and vertical black lines on top of the BCRs indicate reported patient-specific breakpoints.21-25  Asterisks mark the breakpoint present in the REH cell line. The breakpoints identified in the present study are indicated by orange (N424), blue (N726), red (N817), green (N823), and yellow (N890) arrows. Black arrows represent primers that are usually used for ETV6-RUNX1 screening by RT-PCR. They generate 1 of 2 PCR products for every possible breakpoint within the BCRs: exon 5 of ETV6 fused to either exon 2 or exon 3 of RUNX1. Only DNA-based techniques can differentiate between breakpoints that are specific for each individual patient and localized in intronic regions. (B-E) The workflow of cord blood screening using a modified GIPFEL technique15  is presented. (B) CD19+ B cells were enriched from newborn cord blood. DNA was isolated, fragmented using SacI, and purified. The DNA was ligated to achieve circularization and residual linear DNA was digested. DNA circles were purified and PCR (preamplification) and real-time PCR were used to detect the ligation joints produced by circularization. The protocol was carried out with each sample. (C) Optional continuation protocol that was carried out if the real-time PCR in panel B generated a putative positive result. Then a new real-time PCR was carried out with demultiplexed forward primers. If 1 of the forward primers produced a positive result, the PCR was repeated with this forward primer and demultiplexed reverse primers. The products were then analyzed on an agarose gel and Sanger sequenced. In case of negative results, no further validation steps were done. (D) Workflow of the transcriptional validation. RNA was reverse transcribed and subjected to real-time PCR. In case of a putative positive result, the PCR product was purified, cloned, and subjected to Sanger sequencing. (E) Further development of the original GIPFEL technique allowed the identification of patient-specific breakpoints with base-pair resolution. To this end, circularized DNA was amplified using specific primers that hybridized to the known ligation joint region and faced toward the unknown breakpoint. Amplification was carried out with Phi29 DNA polymerase, leading to linear products. With Phi29, amplification occurs at a constant temperature and new primers can constantly bind to the original and the amplified DNA, leading to further amplification. The amplified DNA was subsequently purified and Sanger sequenced. cDNA, complementary DNA; dNTP, deoxynucleotide triphosphate; Ex, exon; FAM, fluorescein amidite; Nuc, deoxynucleoside diphospate; P, phosphate.

Figure 1.

The incidence of ETV6-RUNX1 fusions in healthy newborns can be determined by DNA-based GIPFEL screening. (A) Only DNA- and not RNA-based ETV6-RUNX1 detection provides individual-specific breakpoints. The breakpoint cluster regions (BCRs; horizontal lines, exact regions are marked by dotted lines, sizes are given in base pairs) of ETV6 (top panel, intron 5) and RUNX1 (bottom panel, intron 1 and 2) are presented. Black boxes indicate exons and vertical black lines on top of the BCRs indicate reported patient-specific breakpoints.21-25  Asterisks mark the breakpoint present in the REH cell line. The breakpoints identified in the present study are indicated by orange (N424), blue (N726), red (N817), green (N823), and yellow (N890) arrows. Black arrows represent primers that are usually used for ETV6-RUNX1 screening by RT-PCR. They generate 1 of 2 PCR products for every possible breakpoint within the BCRs: exon 5 of ETV6 fused to either exon 2 or exon 3 of RUNX1. Only DNA-based techniques can differentiate between breakpoints that are specific for each individual patient and localized in intronic regions. (B-E) The workflow of cord blood screening using a modified GIPFEL technique15  is presented. (B) CD19+ B cells were enriched from newborn cord blood. DNA was isolated, fragmented using SacI, and purified. The DNA was ligated to achieve circularization and residual linear DNA was digested. DNA circles were purified and PCR (preamplification) and real-time PCR were used to detect the ligation joints produced by circularization. The protocol was carried out with each sample. (C) Optional continuation protocol that was carried out if the real-time PCR in panel B generated a putative positive result. Then a new real-time PCR was carried out with demultiplexed forward primers. If 1 of the forward primers produced a positive result, the PCR was repeated with this forward primer and demultiplexed reverse primers. The products were then analyzed on an agarose gel and Sanger sequenced. In case of negative results, no further validation steps were done. (D) Workflow of the transcriptional validation. RNA was reverse transcribed and subjected to real-time PCR. In case of a putative positive result, the PCR product was purified, cloned, and subjected to Sanger sequencing. (E) Further development of the original GIPFEL technique allowed the identification of patient-specific breakpoints with base-pair resolution. To this end, circularized DNA was amplified using specific primers that hybridized to the known ligation joint region and faced toward the unknown breakpoint. Amplification was carried out with Phi29 DNA polymerase, leading to linear products. With Phi29, amplification occurs at a constant temperature and new primers can constantly bind to the original and the amplified DNA, leading to further amplification. The amplified DNA was subsequently purified and Sanger sequenced. cDNA, complementary DNA; dNTP, deoxynucleotide triphosphate; Ex, exon; FAM, fluorescein amidite; Nuc, deoxynucleoside diphospate; P, phosphate.

Close modal

GIPFEL is capable of detecting gene fusions without prior knowledge of the exact breakpoint by exploiting the presence of genomic fragments that join material from known regions of 2 different chromosomes. These fragments can be digested and circularized by ligation, creating a junction across the restriction site whose sequence can be predicted from published genome data and used for the design of real-time PCRs.

For ETV6-RUNX1 fusion detection, genomic DNA is digested by SacI and circularized by intramolecular ligation (Figure 1B-E; supplemental Methods; supplemental Figure 1). A multiplexed, semi-nested real-time PCR is performed to quantify translocation-specific ligation products. Demultiplexed qPCR, gel electrophoresis, and sequencing are used for validation of the ligation product and to narrow down the breakpoint region. GIPFEL can detect 1 translocation-carrying cell within 10 000 normal cells (sensitivity ≈10−4) and is highly specific. The likelihood that GIPFEL produces false-positive signals is extremely low (<<10−10; supplemental Figure 2) and has so far never been observed (eg, >180 negative controls for ETV6-RUNX1).

Here, we applied GIPFEL to a retrospective screening of 1000 newborns. The study was approved by the Danish Data Protection Agency and the Danish Scientific Ethics Committee. Written informed consent was obtained. After B-cell enrichment, genomic DNA was prepared from cryopreserved cord bloods and GIPFEL was carried out. In 50 of 1000 samples, sequencing of the ligation joints validated a signal compatible with ETV6-RUNX1 translocation (Table 1; supplemental Figures 3 and 4). These data suggest that ≈5% of newborns harbor the ETV6-RUNX1 fusion at levels detectable by GIPFEL.

Table 1.

Fifty of 1000 cord bloods from healthy newborns were ETV6-RUNX1 translocation-positive in GIPFEL screening

Healthy newbornMultiplexed primer groupForward primerRUNX1 intronReverse primerETV6 intronEstimated frequency
N005 RUNX1-S6f ETV6-S1r-n 1 × 10−4 
N059 RUNX1-S13f ETV6-S2r-n 6 × 10−3 
N099 RUNX1-S2f ETV6-S2r-n 2 × 10−4 
N260 RUNX1-S14f ETV6-S2r-n 1.5 × 10−4 
N285 RUNX1-S11f ETV6-S1r-n 8 × 10−4 
N286 RUNX1-S8f ETV6-S1r-n 1 × 10−4 
N382 RUNX1-S11f ETV6-S3r-n 5/6 1 × 10−3 
N424* RUNX1-S23f ETV6-S1r-n 5 × 10−4 
N439 RUNX1-S23f ETV6-S1r-n 4 × 10−4 
N440 RUNX1-S18f ETV6-S2r-n 1 × 10−3 
N441 RUNX1-S22f ETV6-S2r-n 1 × 10−4 
N447 RUNX1-S28f ETV6-S3r-n 5/6 2 × 10−3 
N463 RUNX1-S10f ETV6-S2r-n 3 × 10−4 
N472 RUNX1-S21f ETV6-S3r-n 5/6 1 × 10−4 
N479 RUNX1-S13f ETV6-S2r-n 1 × 10−3 
N493 RUNX1-S18f ETV6-S2r-n 1 × 10−2 
N496 RUNX1-S5f ETV6-S1r-n 1.5 × 10−3 
N505 2 RUNX1-S11f 1 ETV6-S3r-n 5/6 4 × 103 
N505 4 RUNX1-S13f 1 ETV6-S1r-n 5 7 × 104 
N506 RUNX1-S28f ETV6-S1r-n 5 × 10−3 
N521 RUNX1-S4f ETV6-S2r-n 4 × 10−2 
N522 RUNX1-S10f ETV6-S2r-n 1 × 10−2 
N527 RUNX1-S21f ETV6-S1r-n 4 × 10−3 
N531 3 RUNX1-S29f 1 ETV6-S2r-n 5 5 × 103 
N531 4 RUNX1-S13f 1 ETV6-S2r-n 5 1.5 × 103 
N548 RUNX1-S21f ETV6-S2r-n 9 × 10−5 
N563 RUNX1-S11f ETV6-S2r-n 8 × 10−4 
N578 RUNX1-S24f ETV6-S3r-n 5/6 8 × 10−5 
N590 RUNX1-S26f ETV6-S1r-n 4 × 10−4 
N599 RUNX1-S10f ETV6-S3r-n 5/6 5 × 10−3 
N619 RUNX1-S5f ETV6-S3r-n 5/6 6 × 10−3 
N622 RUNX1-S26f ETV6-S2r-n 3 × 10−4 
N630 RUNX1-S12f ETV6-S2r-n 1 × 10−2 
N651 RUNX1-S4f ETV6-S2r-n 1 × 10−4 
N670 RUNX1-S12f ETV6-S3r-n 5/6 1 × 10−2 
N673 RUNX1-S8f ETV6-S2r-n 1 × 10−2 
N674 RUNX1-S28f ETV6-S3r-n 5/6 9 × 10−3 
N726* RUNX1-S22f ETV6-S1r-n 4 × 10−5 
N729 RUNX1-S23f ETV6-S3r-n 5/6 5 × 10−5 
N731 RUNX1-S4f ETV6-S3r-n 5/6 1.5 × 10−5 
N732 RUNX1-S10f ETV6-S2r-n 1 × 10−4 
N770 RUNX1-S13f ETV6-S3r-n 5/6 4 × 10−4 
N775 RUNX1-S14f ETV6-S2r-n 1 × 10−3 
N784 RUNX1-S28f ETV6-S2r-n 3 × 10−4 
N791 RUNX1-S6f ETV6-S2r-n 1 × 10−4 
N795 RUNX1-S24f ETV6-S3r-n 5/6 1 × 10−4 
N817* RUNX1-S25f ETV6-S1r-n 2 × 10−3 
N823* RUNX1-S13f ETV6-S1r-n 1 × 10−3 
N890* RUNX1-S23f ETV6-S3r-n 5/6 1 × 10−3 
N908 RUNX1-S15f ETV6-S3r-n 5/6 6 × 10−4 
N912 RUNX1-S24f ETV6-S2r-n 3 × 10−3 
N926 RUNX1-S5f ETV6-S3r-n 5/6 3 × 10−4 
Healthy newbornMultiplexed primer groupForward primerRUNX1 intronReverse primerETV6 intronEstimated frequency
N005 RUNX1-S6f ETV6-S1r-n 1 × 10−4 
N059 RUNX1-S13f ETV6-S2r-n 6 × 10−3 
N099 RUNX1-S2f ETV6-S2r-n 2 × 10−4 
N260 RUNX1-S14f ETV6-S2r-n 1.5 × 10−4 
N285 RUNX1-S11f ETV6-S1r-n 8 × 10−4 
N286 RUNX1-S8f ETV6-S1r-n 1 × 10−4 
N382 RUNX1-S11f ETV6-S3r-n 5/6 1 × 10−3 
N424* RUNX1-S23f ETV6-S1r-n 5 × 10−4 
N439 RUNX1-S23f ETV6-S1r-n 4 × 10−4 
N440 RUNX1-S18f ETV6-S2r-n 1 × 10−3 
N441 RUNX1-S22f ETV6-S2r-n 1 × 10−4 
N447 RUNX1-S28f ETV6-S3r-n 5/6 2 × 10−3 
N463 RUNX1-S10f ETV6-S2r-n 3 × 10−4 
N472 RUNX1-S21f ETV6-S3r-n 5/6 1 × 10−4 
N479 RUNX1-S13f ETV6-S2r-n 1 × 10−3 
N493 RUNX1-S18f ETV6-S2r-n 1 × 10−2 
N496 RUNX1-S5f ETV6-S1r-n 1.5 × 10−3 
N505 2 RUNX1-S11f 1 ETV6-S3r-n 5/6 4 × 103 
N505 4 RUNX1-S13f 1 ETV6-S1r-n 5 7 × 104 
N506 RUNX1-S28f ETV6-S1r-n 5 × 10−3 
N521 RUNX1-S4f ETV6-S2r-n 4 × 10−2 
N522 RUNX1-S10f ETV6-S2r-n 1 × 10−2 
N527 RUNX1-S21f ETV6-S1r-n 4 × 10−3 
N531 3 RUNX1-S29f 1 ETV6-S2r-n 5 5 × 103 
N531 4 RUNX1-S13f 1 ETV6-S2r-n 5 1.5 × 103 
N548 RUNX1-S21f ETV6-S2r-n 9 × 10−5 
N563 RUNX1-S11f ETV6-S2r-n 8 × 10−4 
N578 RUNX1-S24f ETV6-S3r-n 5/6 8 × 10−5 
N590 RUNX1-S26f ETV6-S1r-n 4 × 10−4 
N599 RUNX1-S10f ETV6-S3r-n 5/6 5 × 10−3 
N619 RUNX1-S5f ETV6-S3r-n 5/6 6 × 10−3 
N622 RUNX1-S26f ETV6-S2r-n 3 × 10−4 
N630 RUNX1-S12f ETV6-S2r-n 1 × 10−2 
N651 RUNX1-S4f ETV6-S2r-n 1 × 10−4 
N670 RUNX1-S12f ETV6-S3r-n 5/6 1 × 10−2 
N673 RUNX1-S8f ETV6-S2r-n 1 × 10−2 
N674 RUNX1-S28f ETV6-S3r-n 5/6 9 × 10−3 
N726* RUNX1-S22f ETV6-S1r-n 4 × 10−5 
N729 RUNX1-S23f ETV6-S3r-n 5/6 5 × 10−5 
N731 RUNX1-S4f ETV6-S3r-n 5/6 1.5 × 10−5 
N732 RUNX1-S10f ETV6-S2r-n 1 × 10−4 
N770 RUNX1-S13f ETV6-S3r-n 5/6 4 × 10−4 
N775 RUNX1-S14f ETV6-S2r-n 1 × 10−3 
N784 RUNX1-S28f ETV6-S2r-n 3 × 10−4 
N791 RUNX1-S6f ETV6-S2r-n 1 × 10−4 
N795 RUNX1-S24f ETV6-S3r-n 5/6 1 × 10−4 
N817* RUNX1-S25f ETV6-S1r-n 2 × 10−3 
N823* RUNX1-S13f ETV6-S1r-n 1 × 10−3 
N890* RUNX1-S23f ETV6-S3r-n 5/6 1 × 10−3 
N908 RUNX1-S15f ETV6-S3r-n 5/6 6 × 10−4 
N912 RUNX1-S24f ETV6-S2r-n 3 × 10−3 
N926 RUNX1-S5f ETV6-S3r-n 5/6 3 × 10−4 

Frequencies refer to B-cell population. Results for 2 newborns with concurrent fusions appear in bold.

*

In these cases, the translocation was confirmed by Sanger sequencing of the breakpoint.

GIPFEL screening also revealed 2 newborns (N505, N531) whose cord blood harbored 2 different ETV6-RUNX1 fusions each (Table 1; supplemental Figure 5). The estimated frequencies of the 2 fusions differed from one another (N505: 4 × 10−3 and 7 × 10−4, N531: 5 × 10−3 and 1.5 × 10−3), indicating 2 coexisting or overlapping clones.

To validate our screening results, we tested fusion transcript expression in GIPFEL+ and GIPFEL cord bloods by qPCR (Figure 1D; supplemental Figure 6; supplemental Table 2). Due to limited sample availability, we could only investigate 2 positive (N005 and N260) and 50 negative cord bloods. In the REH cell line, ETV6-RUNX1 expression was roughly in the same range as the ABL1 control gene. In GIPFEL+ cord blood samples, expression of ETV6-RUNX1 was detected at a lower level (10−4) in accord with the lower number of translocation-carrying cells (10−4) compared with the cell line. We confirmed the fusions on the RNA level by Sanger sequencing. None of the GIPFEL cord bloods showed transcription of ETV6-RUNX1. In addition, 9 translocation-negative cell lines were both GIPFEL and qPCR (supplemental Table 3).

To finally confirm positive GIPFEL results, we sequenced individual-specific breakpoints with base-pair resolution on the DNA level using targeted amplification of translocation-carrying circles and Sanger sequencing (Figure 1E). Due to limited sample availability, ETV6-RUNX1 breakpoints of 5 cord bloods (N424, N726, N817, N832, N890) have been identified so far (Figure 1A; supplemental Figure 7). The breakpoints were specific for each proband and never reported before.

Because of anonymized sample processing, no tracking of leukemia cases within the analyzed cohort could be carried out. However, the determined frequencies of ETV6-RUNX1 translocations (≈5% GIPFEL screen or ≈0.5% if taking into account only GIPFEL screen results validated by breakpoint sequencing) are both far above the leukemia frequency of ≈0.01%. The incidence of ETV6-RUNX1 fusions may even exceed ≈5% because complex rearrangements, rearrangements outside of the known breakpoint regions, and not-yet-expanded cell clones (<10−4) evade detection by GIPFEL.15  Thus, translocation-carrying clones (and potentially >1) are likely present in a high number of healthy individuals who will never develop leukemia. Similarly, other leukemia- or lymphoma-associated translocations have been detected in peripheral blood of healthy individuals (reviewed in Janz et al17 ). Illegitimate genetic recombination seems to occur frequently in hematopoietic precursors. For preleukemic clones to arise, the fusion has to occur in an early precursor with self-renewal capacity and must produce a functional oncoprotein that promotes clonal expansion.

In the case of ETV6-RUNX1, the leukemia-inducing potential of the fusion seems to be very low. This is in accordance with (1) the low concordance rate of ≈10% in identical twins,18  (2) the long postnatal latency phase (≈2-14 years),2  (3) the presence of recurrent secondary leukemia inducing genetic lesions (ETV6 deletions in 70%, extra copies of RUNX1 in 23%, or extra der(21)t(12;21) in 10% of cases),19  and (4) evidence from transgenic animal studies.20 

These results strengthen the importance of environmentally or spontaneously caused secondary hits in ETV6-RUNX1+ ALL. Future studies correlating GIPFEL screens with genetic and epidemiological data will provide closer insight into the pathogenesis of ETV6-RUNX1+ leukemia.

The online version of this article contains a data supplement.

The authors thank Sabine Hornhardt (Federal Office for Radiation Protection, Oberschleissheim, Germany) for advice and support. The authors thank Mel Greaves and Tony Ford (The Institute of Cancer Research, London, United Kingdom) and Jan Trka (Childhood Leukaemia Investigation Prague, Prague, Czech Republic) for valuable comments. The authors thank their colleague Cyrill Schipp for valuable support and Daniel Scholtyssik and Bianca Killing for excellent technical assistance.

This work was supported by the German Federal Office for Radiation Protection (grant 36 14 S 30034), by an intramural grant (2016-70) of the Research Commission of the Medical Faculty of Heinrich Heine University Düsseldorf, by the Katharina-Hardt-Stiftung, by the Danish Cancer Society, and by the Danish Childhood Cancer Foundation.

Contribution: D.S. performed laboratory work, designed research, analyzed data, and wrote the paper; D.L. designed research and critically reviewed the paper; M.S. provided clinical samples and analyzed clinical data; R.S. designed research; M.O. recruited the probands, analyzed clinical data, and designed and performed laboratory work; K.S. designed and supervised research; A.B. designed research and wrote the paper; U.F. designed research, analyzed data, and wrote the paper; and U.F. and A.B. were the principal investigators and take primary responsibility for the paper.

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

Correspondence: Ute Fischer, Department of Pediatric Oncology, Hematology, and Clinical Immunology, Center for Child and Adolescent Health, Heinrich Heine University, Moorenstr 5, 40225 Düsseldorf, Germany; e-mail: ute.fischer@med.uni-duesseldorf.de.

1.
Mullighan
CG
.
Molecular genetics of B-precursor acute lymphoblastic leukemia
.
J Clin Invest
.
2012
;
122
(
10
):
3407
-
3415
.
2.
Wiemels
JL
,
Ford
AM
,
Van Wering
ER
,
Postma
A
,
Greaves
M
.
Protracted and variable latency of acute lymphoblastic leukemia after TEL-AML1 gene fusion in utero
.
Blood
.
1999
;
94
(
3
):
1057
-
1062
.
3.
Papaemmanuil
E
,
Rapado
I
,
Li
Y
, et al
.
RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia
.
Nat Genet
.
2014
;
46
(
2
):
116
-
125
.
4.
Mori
H
,
Colman
SM
,
Xiao
Z
, et al
.
Chromosome translocations and covert leukemic clones are generated during normal fetal development
.
Proc Natl Acad Sci USA
.
2002
;
99
(
12
):
8242
-
8247
.
5.
Eguchi-Ishimae
M
,
Eguchi
M
,
Ishii
E
, et al
.
Breakage and fusion of the TEL (ETV6) gene in immature B lymphocytes induced by apoptogenic signals
.
Blood
.
2001
;
97
(
3
):
737
-
743
.
6.
Olsen
M
,
Madsen
HO
,
Hjalgrim
H
,
Gregers
J
,
Rostgaard
K
,
Schmiegelow
K
.
Preleukemic TEL-AML1-positive clones at cell level of 10(-3) to 10(-4) do not persist into adulthood
.
J Pediatr Hematol Oncol
.
2006
;
28
(
11
):
734
-
740
.
7.
Lausten-Thomsen
U
,
Hjalgrim
H
,
Marquart
H
,
Lutterodt
M
,
Petersen
BL
,
Schmiegelow
K
.
ETV6-RUNX1 transcript is not frequent in early human haematopoiesis
.
Eur J Haematol
.
2008
;
81
(
2
):
161
-
162
.
8.
Lausten-Thomsen
U
,
Madsen
HO
,
Vestergaard
TR
,
Hjalgrim
H
,
Lando
A
,
Schmiegelow
K
.
Increased risk of ALL among premature infants is not explained by increased prevalence of pre-leukemic cell clones
.
Blood Cells Mol Dis
.
2010
;
44
(
3
):
188
-
190
.
9.
Lausten-Thomsen
U
,
Madsen
HO
,
Vestergaard
TR
,
Hjalgrim
H
,
Nersting
J
,
Schmiegelow
K
.
Prevalence of t(12;21)[ETV6-RUNX1]-positive cells in healthy neonates
.
Blood
.
2011
;
117
(
1
):
186
-
189
.
10.
Zuna
J
,
Madzo
J
,
Krejci
O
, et al
. ETV6/RUNX1 (TEL/AML1) is a frequent prenatal first hit in childhood leukemia.
Blood
.
2011
;117(1):
368
-
369
; author reply 370-371.
11.
Olsen
M
,
Hjalgrim
H
,
Melbye
M
,
Madsen
HO
,
Schmiegelow
K
.
RT-PCR screening for ETV6-RUNX1-positive clones in cord blood from newborns in the Danish National Birth Cohort
.
J Pediatr Hematol Oncol
.
2012
;
34
(
4
):
301
-
303
.
12.
Škorvaga
M
,
Nikitina
E
,
Kubeš
M
, et al
.
Incidence of common preleukemic gene fusions in umbilical cord blood in Slovak population
.
PLoS One
.
2014
;
9
(
3
):
e91116
.
13.
Ornelles
DA
,
Gooding
LR
,
Garnett-Benson
C
.
Neonatal infection with species C adenoviruses confirmed in viable cord blood lymphocytes
.
PLoS One
.
2015
;
10
(
3
):
e0119256
.
14.
Kosik
P
,
Skorvaga
M
,
Durdik
M
, et al
.
Low numbers of pre-leukemic fusion genes are frequently present in umbilical cord blood without affecting DNA damage response
.
Oncotarget
.
2017
;
8
(
22
):
35824
-
35834
.
15.
Fueller
E
,
Schaefer
D
,
Fischer
U
, et al
.
Genomic inverse PCR for exploration of ligated breakpoints (GIPFEL), a new method to detect translocations in leukemia
.
PLoS One
.
2014
;
9
(
8
):
e104419
.
16.
Hovorkova
L
,
Zaliova
M
,
Venn
NC
, et al
.
Monitoring of childhood ALL using BCR-ABL1 genomic breakpoints identifies a subgroup with CML-like biology
.
Blood
.
2017
;
129
(
20
):
2771
-
2781
.
17.
Janz
S
,
Potter
M
,
Rabkin
CS
.
Lymphoma- and leukemia-associated chromosomal translocations in healthy individuals
.
Genes Chromosomes Cancer
.
2003
;
36
(
3
):
211
-
223
.
18.
Greaves
MF
,
Maia
AT
,
Wiemels
JL
,
Ford
AM
.
Leukemia in twins: lessons in natural history
.
Blood
.
2003
;
102
(
7
):
2321
-
2333
.
19.
Sun
C
,
Chang
L
,
Zhu
X
.
Pathogenesis of ETV6/RUNX1-positive childhood acute lymphoblastic leukemia and mechanisms underlying its relapse
.
Oncotarget
.
2017
;
8
(
21
):
35445
-
35459
.
20.
Hauer
J
,
Borkhardt
A
,
Sánchez-García
I
,
Cobaleda
C
.
Genetically engineered mouse models of human B-cell precursor leukemias
.
Cell Cycle
.
2014
;
13
(
18
):
2836
-
2846
.
21.
Wiemels
JL
,
Greaves
M
.
Structure and possible mechanisms of TEL-AML1 gene fusions in childhood acute lymphoblastic leukemia
.
Cancer Res
.
1999
;
59
(
16
):
4075
-
4082
.
22.
Wiemels
JL
,
Alexander
FE
,
Cazzaniga
G
,
Biondi
A
,
Mayer
SP
,
Greaves
M
.
Microclustering of TEL-AML1 translocation breakpoints in childhood acute lymphoblastic leukemia
.
Genes Chromosomes Cancer
.
2000
;
29
(
3
):
219
-
228
.
23.
Andersen
MT
,
Nordentoft
I
,
Hjalgrim
LL
, et al
.
Characterization of t(12;21) breakpoint junctions in acute lymphoblastic leukemia
.
Leukemia
.
2001
;
15
(
5
):
858
-
859
.
24.
Rodríguez-Hernández
G
,
Hauer
J
,
Martín-Lorenzo
A
, et al
.
Infection exposure promotes ETV6-RUNX1 precursor B-cell leukemia via impaired H3K4 demethylases
.
Cancer Res
.
2017
;
77
(
16
):
4365
-
4377
.
25.
Jin
Y
,
Wang
X
,
Hu
S
,
Tang
J
,
Li
B
,
Chai
Y
.
Determination of ETV6-RUNX1 genomic breakpoint by next-generation sequencing
.
Cancer Med
.
2016
;
5
(
2
):
337
-
351
.
Sign in via your Institution