Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2008

Open Access 01.12.2008 | Research

Persistence of TEL-AML1 fusion gene as minimal residual disease has no additive prognostic value in CD 10 positive B-acute lymphoblastic leukemia: a FISH study

verfasst von: Eman Mosad, Hosny B Hamed, Rania M Bakry, Azza M Ezz-Eldin, Nesrine M Khalifa

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2008

Abstract

Objectives

We have analyzed t(12;21)(p13:q22) in an attempt to evaluate the frequency and prognostic significance of TEL-AML1 fusion gene in patients with childhood CD 10 positive B-ALL by fluorescence in situ hybridization (FISH). Also, we have monitored the prognostic value of this gene as a minimal residual disease (MRD).

Methods

All bone marrow samples of eighty patients diagnosed as CD 10 positive B-ALL in South Egypt Cancer Institute were evaluated by fluorescence in situ hybridization (FISH) for t(12;21) in newly diagnosed cases and after morphological complete remission as a minimal residual disease (MRD). We determined the prognostic significance of TEL-AML1 fusion represented by disease course and survival.

Results

TEL-AML1 fusion gene was positive in (37.5%) in newly diagnosed patients. There was a significant correlation between TEL-AML1 fusion gene both at diagnosis (r = 0.5, P = 0.003) and as a MRD (r = 0.4, P = 0.01) with favorable course. Kaplan-Meier curve for the presence of TEL-AML1 fusion at the diagnosis was associated with a better probability of overall survival (OS); mean survival time was 47 ± 1 month, in contrast to 28 ± 5 month in its absence (P = 0.006). Also, the persistence at TEL-AML1 fusion as a MRD was not significantly associated with a better probability of OS; the mean survival time was 42 ± 2 months in the presence of MRD and it was 40 ± 1 months in its absence. So, persistence of TEL-AML1 fusion as a MRD had no additive prognostic value over its measurement at diagnosis in terms of predicting the probability of OS.

Conclusion

For most patients, the presence of TEL-AML1 fusion gene at diagnosis suggests a favorable prognosis. The present study suggests that persistence of TEL-AML1 fusion as MRD has no additive prognostic value.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-8722-1-17) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

EM participated in study design, conducted FISH technique, statistical analysis and wrote the manuscript. HB, RMB, AME-E participated in study design, in conducting FISH technique, critical manuscript revision. NMK participated in study concept and was responsible for the clinical aspect of the work as regards patients' clinical assessment, management and follow-up. All authors read and approved the manuscript.
Abkürzungen
ALL
Acute lymphoblastic leukemia
CD
cluster differentiation
CMML
chronic myelomonocytic leukemia
CR
Complete remission
FISH
fluorescence in situ hybridization
HLA
human leucocytic antigen
MRD
minimal residual disease
OS
overall survival
T
translocation.

Background

Acute lymphoblastic leukemia (ALL) is the most common malignancy of childhood. Cure of many of these children is difficult to predict and is considered an individual response of the patient to chemotherapy. It is likely that this clinical heterogeneity reflects a diverse pathogenesis of leukemia. The molecular basis of childhood ALL is largely unknown. Furthermore, it is likely that significant advance in the treatment of childhood ALL will be dependent on a better understanding of the molecular events that cause the disease [1, 2].
A recurrent t(12;21)(p13:q22) has been described in several human ALLs. In this translocation the TEL gene fuses to AML1; a gene previously cloned from translocation breakpoints in acute myeloid leukemia. These abnormalities consist of both translocations and deletions. The frequency of t(12;21) was estimated as to be 15–35% in childhood ALL. This translocation has been recognized as the most common chromosomal aberration in childhood ALL [24]. All (95–100%) of TEL-AML1 positive ALL patients found to has a consistent cell surface immunophenotype. (B lineage ALL based on the expression of HLA-DR, CD 10 and CD 19) [2, 4]. Thus, we raised a question if the opposite is true meaning that if CD 10 positive B-ALL immunophenotype will have a similarly high incidence of positive TEL-AML1 fusion gene?. Accordingly can we use this fusion gene as a minimal residual disease (MRD) in this specific subgroup of B-ALL.
It was also reported that patients with the TEL-AML1 fusion have a high sensitivity to chemotherapy [46]. Other investigators have reported that almost 10–28% of relapsed pediatric ALL patients express the TEL-AML1 fusion, but the relapse of patients with the TEL-AML1 fusion is not always associated with a poor prognosis [79]. However, some patients with the TEL-AML1 transcripts and additional molecular lesions had poor outcomes [10]. So, the prognostic significance of TEL-AML1 transcript remains controversial.
Patients with a poor treatment response by morphologic criteria have a high risk of relapse [11, 12]. But morphologic studies will only identify a minority of those children with ALL who eventually fail. Minimal residual disease (MRD) has been of prognostic value in children with ALL. Several studies have shown that children with a high leukemic cell burden at the end of induction therapy have an inferior outcome compared to children with a lower leukemic cell burden [1318]. The investigation of MRD using TEL-AML1 fusion gene as a marker has been carried out on a limited number of patients to date although it is a minor examination. The relation between relapse and the persistence of detectable MRD show heterogeneity [19, 20]. As this translocation is often difficult to detect by conventional G-banding analysis, in addition many patients with ALL were diagnosed as normal karyotype or could not examined for karyotype by classic cytogenetic analysis. In particular fluorescence in situ hybridization (FISH) analysis has been applied to hematopoietic malignancies with subtle or complex chromosomal aberrations which are difficult or impossible to detect by standard cytogenetic analysis [3]. Therefore, we conducted a retrospective study to determine the frequency and prognostic significance of TEL-AML1 fusion in CD 10 positive B-ALL, and to clarify whether the persistence of the TEL-AML1 fusion gene as a MRD has an additive value.

Methods

Patients and Samples

Bone marrow (BM) samples were obtained from 80 CD 10 positive B-ALL patients aged from 3 to 11 years; mean age was 7.4 ± 2, diagnosed at our Institute between 2002 and 2004 and followed up till 2006. Diagnosis was performed according to the standard procedures; French American British (FAB) classification of lymphoblastic leukemia and determination of immunophenotypic markers. They were B precursor ALL patients diagnosed as common and preB-ALL by flowcytomety (expressing CD19, CD 10 and HLA-DR). Patients were considered in the standard risk category if they were aged 1–9 years, had white blood cell count < 50,000 per micro liter, or had central nervous system affection. The remaining patients were considered as high risk. Patients were treated according to modified Berlin-Frankfurt-Munster (BFM-90) ALL protocol.21 t(12;21) was evaluated by FISH in newly diagnosed cases (80 patients) and after morphological remission in patients who were positive for t(12;21) as a MRD (30 patients) and we determined the prognostic significance of TEL-AML1 fusion represented by disease course and survival and we clarified if the persistence of the TEL-AML1 fusion gene as MRD had an additive prognostic value. Five normal BM samples were taken as a control and the level of TEL-AML1 fusion by FISH estimated as 1 ± 0.2%. Therefore, the cut-off level used in this study was 1.2%. The study was approved by our faculty ethical committee and was adherent to the regulations of the declaration of Helsinski.

Response Criteria

Complete remission (CR) was defined as the complete disappearance of all tumor masses confirmed at clinical examination, or X-rays, and ultrasound studies; a normal BM examination and pathology; and no evidence of CNS disease by cerebrospinal fluid analysis.
The disease course was assessed by ranking patients according to their response to treatment into 4 categories; CR1, CR2, CR3, and resistance and/or death. CR1 patients were those who achieved first complete remission. Patients who received therapy for their first or second relapse and achieved <5% blasts in the marrow and had extramedullary sites of leukemia were considered to be in second or third remission (CR2 or CR3). Patients whose marrow showed >5% blasts with or without evidence of extramedullary disease were considered to be in relapse.

Detection Of t(12;21) By FISH Analysis In ALL Patients

In situ hybridization (ISH) is a technique that allows the visualization of a specific nucleic acid sequences within a cellular preparation. Specifically DNA FISH involves the precise annealing of a single standard fluorescently labeled DNA probe to complementary target sequences. The hybridization of the probe with the cellular DNA site is visible by direct detection using fluorescence microscopy.
After 24 hours of unstimulated culture, samples were fixed. Interphase cells were attached to glass slides using standard cytogenetic protocol. The resulting specimen DNA was denaturated to its single strand form and then allowed to hybridize with LSI TEL/AML1 ES Dual Color probe to detect t (12;21) 12p13 spectrum green/21q22 spectrum orange catalog 32-191005-Vysis. Following hybridization, the excess and unbound probe was removed by a series of washes and the chromosomes and nuclei were counter stained with DNA specific stain DAPI (4.6 diamidino-2-phenylindole) that fluoresces blue. The expected pattern in normal nucleus hybridized with TEL/AML1 probe is two orange, two green (2O2G). In the nucleus harboring the t(12;21), the probe hybridized to a nucleus containing the t (12;21) showing one green (native TEL), one large orange (native AML1), one smaller orange (ES) and one fused orange/green (20IGIF) signal pattern. The Microscopy and photography were conducted using a Zeiss Axiovert 200 fluorescence microscope fitted with a high resolution Leica CCD camera. Images were processed using Leica CW4000 imaging system and software (Leica, Germany).

Statistical Methods

The study cutoff time limit was September 2006. Overall survival (OS) was calculated from the first day of chemotherapy to the date of last follow up contact for patients who were alive. All data were analyzed using SPSS (Statistical Program for Social Sciences version 11 for windows, 2001, SPSS Inc., Chicago, IL, USA). Correlations are done using Pearson correlation test. Categorical variables were compared using chi-square test with Fisher's Exact correction. OS is estimated with the Kaplan-Meier method. A P value < 0.05 was considered to be significant.

Results

Eighty ALL patients were enrolled in the study at our Institute between 2002 and 2006. They males were (n = 56; 70%) and females were (n = 24; 30%), mean age 7.4 ± 2 years they were 44 patients L1 (55%) and 36 L2 (45%). They were all B-lineage ALL positive for CD 10 and CD 19 by immunophenotyping (common and pre B-ALL). Most of our patients were in the standard risk (n = 64; 80%), while (n = 16; 20%) were in the high risk category. The karyotypes: Seven metaphases were available for cytogenetic analysis and they were normal. A precise karyotype was not obtained from other patients because of poor morphology of metaphases.
TEL-AML1 fusion gene was evaluated by FISH which showed a fused yellow signal (Figure 1) on the der (21) chromosome in the metaphase and on the interphase nuclei of leukemic cells. It was measured in newly diagnosed cases (Table 1) and it was positive in 30/80 (37.5%) determining its frequency in B-lineage ALL positive for CD 10 and CD 19. The mean percent of TEL-AML1 fusion gene was 50 ± 22% estimated in 300 interphase cells. A control was performed using, five normal bone marrow samples and the cut-off level in this method was estimated to be 1.2%. There was a favorable significant correlation between TEL-AML1 fusion gene and disease course (r = 0.5, P = 0.003). Of particular interest was the observation that 10/50 (20%) of patients lacking the TEL-AML1 fusion had a very bad course (eight children did not achieve a complete remission after induction chemotherapy (resistant) and two achieved CNS relapse and died). No significant correlation was detected between the presence of TEL-AML1 fusion gene at diagnosis and peripheral WBC count, age, sex, organs, FAB classification, central nervous system disease, and risk category. We analyzed the patients who were positive for the presence of TEL-AML1 fusion at diagnosis (n = 30) to detect its persistence as a MRD in patients who entered in complete remission morphologically (Table 2). It was positive in (n = 15/30; 50%) patients. The mean percent of TEL-AML1 fusion gene was 7 ± 2% estimated in 300 interphase cells. The persistence of TEL-AML1 fusion gene as a MRD, was correlated with a favorable course (r = 0.4, P = 0.01). To be noticed that (n = 12/15; 80%) of MRD positivity were in CRI.
Table 1
Interpahse FISH results of the patients with the TEL-AML1 fusion gene at diagnosis
  
Clinical course
 
  
CR1
CR2
CR2, CNS relapse and death
Resistant and death
Total
P
T (12; 21) at Diagnosis
Yes
20
9
1
0
30
0.003
 
No
9
31
2
8
50
 
Total
 
29
40
3
8
80
 
FISH, fluorescence in situ hybridization; CR1, first complete remission; CR2, second complete remission; CNS, involvement of the central nervous system.
Table 2
Interpahse FISH results of the patients with the TEL-AML1 fusion after complete remission as MRD
  
Clinical course
  
CR1
CR2
CR2, CNS relapse and death
Resistant and death
Total
P
t (12; 21) at Remission (MRD)
Yes
12
2
1
-
15
0.01
 
No
6
9
-
-
15
 
Total
 
18
11
1
-
30
 
FISH was performed to the patients with a positive t(12;21) at the time of diagnosis who passed to remission; a total number of 30 patients. FISH, fluorescence in situ hybridization; CR1, first complete remission; CR2, second complete remission; CNS, involvement of the central nervous system
Kaplan-Meier curve for the presence of TEL-AML1 fusion at the diagnosis was associated with a better probability of OS (Figure 2); mean survival time was 47 ± 1 month, in contrast to 28 ± 5 month in its absence (P = 0.006). Also, the persistence at TEL-AML1 fusion as a MRD was not significantly associated with a better probability of OS (Figure 3); the mean survival time was 42 ± 2 months in the presence of MRD and it was 40 ± 1 months in its absence. So, persistence of TEL-AML1 fusion as a MRD had no additive prognostic value over its measurement at diagnosis in terms of predicting the probability of OS.

Discussion

The TEL gene encodes a member of the ETS family of transcription factors and is rearranged in a wide variety of hematological malignancies. In particular, TEL is fused to the platelet-derived growth factor receptor β in CMML, to the ABL tyrosine kinase in acute myeloid leukemia and ALL, and to the product of the MNI gene in myeloproliferative disorders. AML-I is the DNA-binding subunit of the transcription factor complex core binding factor (CBF-β). It is frequently rearranged in myeloid malignancy either through fusion to ETO as a result of t(8;21)(q22:q22) or to EVII, MDS1, or EAP as a result of t(3;21)(q26:q22).[2, 22] The frequent involvement of TEL and AML-I in chromosomal translocations suggests that these genes play important roles in the pathogenesis of human leukemia. In t(12;21) a high level of expression of the hybrid protein that contains the functional domains of AML-I under the transcriptional control of the TEL promoter may be involved in oncogenic transformation [2, 22]. In this study we demonstrated that the frequency of TEL-AML1 fusion in B-Lineage CD10 positive ALL was 37.5%. versus 30% in a previous study included multicentres and larger number of patients.23 In our data (22%)of patients with t(12;21) were CD34-positive, indicating that the leukemic cells originated from primitive hematopoietic cell similar to those of ALL patients with t(9;22) or 11q23 abnormalities [2]. We also, found that 67% of the t(12;21) positive patients were in (CR1), indicating a favorable course, as previously reported [1, 2].
The relationship between the TEL-AML1 fusion and a favorable prognosis represented by survival has already been described [2, 9]. Rubnitz et al [9] reported that the survival at five years follow-up of a group with the TEL-AML1 fusion was 91 ± 5%. These patients with positive TEL-AML1 fusion who achieved a favorable prognosis were found to be younger, without hyperleukocyosis, with the CD 10 positive B precursor ALL immunophenotyping and chemosensitive [2]. Also, a recent report studying the prognosis of relapsed patients showed an outcome consistent with ours. The median duration of remission of relapsed TEL-AML1-positive patients was reported to be 42.5 versus 27 months in those lacking the gene; P = 0.0001 [24]. Our study was consistent with the pervious studies as TEL-AML1 fusion at the diagnosis was associated with a better probability of overall survival [2, 8, 9]. On the other hand, other studies have reported that some patients with TEL-AML1 transcript had a poor outcome [25]. In many cases, TEL-AML1 transcripts detected by RT-PCR and Southern blotting in childhood ALL disappeared soon after the start of chemotherapy [6, 26]. Others reported that a patient with additional molecular lesions with p16 homozygous deletion in addition to TEL-AML1 transcript relapsed usually late, and the survival was ultimately favorable [8, 9]. An analysis of late or off-treatment relapse of TEL-AML1 positive ALL suggested that leukemic cells in relapse were not derived from the dominant clone at diagnosis. It represents a transformation of cells belonging to a persistent preleukemic clone that was generated by TEL-AML1 fusion in utero and survived chemotherapy [27].
The progress in treatment of ALL patients without conventional risk factors has been hampered by the inability to predict relapse after patients achieved a complete remission [19]. Whereas in large prospective studies on childhood ALL, residual disease is a powerful indicator of treatment outcome [20, 28]. In this study, MRD was detectable in 50% of patients of CD 10 positive B-ALL after 4 to 6 weeks of induction therapy. Whereas, in a prospective study on childhood ALL, MRD was detectable in 25% to 58% of patients after the same period of induction therapy [20]. It was reported that the frequency of MRD positivity is high after induction and decreases gradually during consolidation and maintenance phase being in some genes 88% during early induction to 13% at week 52 [19]. If MRD as a marker was detected, the general opinion is that it could become a risk factor for relapse [20, 28]. In contrast, MRD lasts among some patients in long-term remission in other forms of childhood acute leukemia like t(15;17) and t(8;21). [32,33] Cayuela et al [26], reported that one out of seven patients with the TEL-AML1 transcript, serially evaluated, exhibited persistence of detectable MRD over eight months, and that all the patients were in continuous complete remission. This study was consistent with that reported by others [26, 29, 30] that several patients were found to be positive for TEL-AML1 fusion, but the persistence of detectable MRD was not associated with a better probability of OS. Therefore, the relationship of the MRD level of TEL-AML1 fusion and prognosis shows heterogeneity and further investigation is required to evaluate their association and to design risk adapted therapeutic approaches.

Conclusion

TEL-AML1 fusion gene detected by FISH in newly diagnosed cases of CD 10 positive B-ALL is considered a favorable prognostic marker with a better course. The persistence of TEL-AML1 fusion gene as a MRD has no additive prognostic value. Considering the cost-benefit ratio TEL-AML1 fusion gene done once at diagnosis gives sufficient prognostic information. However, much research about the biologic and clinical significance of TEL-AML1 as MRD in CD 10 positive ALL is needed to determine how to best integrate TEL-AML1 testing into routine patient care.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

EM participated in study design, conducted FISH technique, statistical analysis and wrote the manuscript. HB, RMB, AME-E participated in study design, in conducting FISH technique, critical manuscript revision. NMK participated in study concept and was responsible for the clinical aspect of the work as regards patients' clinical assessment, management and follow-up. All authors read and approved the manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Endo C, Oda M, Nishiuchi R, Seino Y: Persistence of TEL-AML1 transcript in acute lymphoblastic leukemia in long-term remission. Pediatr Int. 2003, 45: 275-280. 10.1046/j.1442-200X.2003.01709.x.CrossRefPubMed Endo C, Oda M, Nishiuchi R, Seino Y: Persistence of TEL-AML1 transcript in acute lymphoblastic leukemia in long-term remission. Pediatr Int. 2003, 45: 275-280. 10.1046/j.1442-200X.2003.01709.x.CrossRefPubMed
2.
Zurück zum Zitat McLean TW, Ringold S, Neuberg D, Stegmaier K, Tantravahi R, Ritz J, Koeffler HP, Takeuchi S, Janssen JW, Seriu T, Bartram CR, Sallan SE, Gilliland DG, Golub TR: TEL-AML1 dimerizes and is associated with a favorable outcome in childhood acute lymphoblastic leukemia. Blood. 1996, 88: 4252-4258.PubMed McLean TW, Ringold S, Neuberg D, Stegmaier K, Tantravahi R, Ritz J, Koeffler HP, Takeuchi S, Janssen JW, Seriu T, Bartram CR, Sallan SE, Gilliland DG, Golub TR: TEL-AML1 dimerizes and is associated with a favorable outcome in childhood acute lymphoblastic leukemia. Blood. 1996, 88: 4252-4258.PubMed
3.
Zurück zum Zitat Eguchi-Ishimae M, Eguchi M, Tanaka K, Hamamoto K, Ohki M, Ueda K, Kamada N: Fluorescence in situ hybridization analysis of 12;21 translocation in Japanese childhood acute lymphoblastic leukemia. Jpn J Cancer Res. 1998, 89: 783-788.CrossRefPubMed Eguchi-Ishimae M, Eguchi M, Tanaka K, Hamamoto K, Ohki M, Ueda K, Kamada N: Fluorescence in situ hybridization analysis of 12;21 translocation in Japanese childhood acute lymphoblastic leukemia. Jpn J Cancer Res. 1998, 89: 783-788.CrossRefPubMed
4.
Zurück zum Zitat Loh ML, Silverman LB, Young ML, Neuberg D, Golub TR, Sallan SE, Gilliland DG: Incidence of TEL/AML1 fusion in children with relapsed acute lymphoblastic leukemia. Blood. 1998, 92: 4792-4797.PubMed Loh ML, Silverman LB, Young ML, Neuberg D, Golub TR, Sallan SE, Gilliland DG: Incidence of TEL/AML1 fusion in children with relapsed acute lymphoblastic leukemia. Blood. 1998, 92: 4792-4797.PubMed
5.
Zurück zum Zitat Shurtleff SA, Buijs A, Behm FG, Rubnitz JE, Raimondi SC, Hancock ML, Chan GC, Pui CH, Grosveld G, Downing JR: TEL-AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup of patients with an excellent prognosis. Leukemia. 1995, 9 (12): 985-989. Shurtleff SA, Buijs A, Behm FG, Rubnitz JE, Raimondi SC, Hancock ML, Chan GC, Pui CH, Grosveld G, Downing JR: TEL-AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup of patients with an excellent prognosis. Leukemia. 1995, 9 (12): 985-989.
6.
Zurück zum Zitat Satake N, Sakashita A, Kobayashi H, Maseki N, Sakurai M, Kaneko Yl: Minimal residual disease with TEL-AML1 fusion transcript in childhood acute lymphoblastic leukemia with t(12;21). Br J Haematol. 1997, 97: 607-611. 10.1046/j.1365-2141.1997.762712.x.CrossRefPubMed Satake N, Sakashita A, Kobayashi H, Maseki N, Sakurai M, Kaneko Yl: Minimal residual disease with TEL-AML1 fusion transcript in childhood acute lymphoblastic leukemia with t(12;21). Br J Haematol. 1997, 97: 607-611. 10.1046/j.1365-2141.1997.762712.x.CrossRefPubMed
7.
Zurück zum Zitat Rubnitz JE, Downing JR, Pui CH, Shurtleff SA, Raimondi SC, Evans WE, Head DR, Crist WM, Rivera GK, Hancock ML, Boyett JM, Buijs A, Grosveld G, Behm FG: TEL gene rearrangement in acute lymphoblastic leukemia: a new genetic marker with prognostic significance. J Clin Oncol. 1997, 15: 1150-1157.PubMed Rubnitz JE, Downing JR, Pui CH, Shurtleff SA, Raimondi SC, Evans WE, Head DR, Crist WM, Rivera GK, Hancock ML, Boyett JM, Buijs A, Grosveld G, Behm FG: TEL gene rearrangement in acute lymphoblastic leukemia: a new genetic marker with prognostic significance. J Clin Oncol. 1997, 15: 1150-1157.PubMed
8.
Zurück zum Zitat Harbott J, Viehmann S, Borkhardt A, Henze G, Lampert F: Incidence of TEL-AML1 fusion gene analyzed consecutively in children with acute lymphoblastic leukemia in relapse. Blood. 1997, 90: 4933-7.PubMed Harbott J, Viehmann S, Borkhardt A, Henze G, Lampert F: Incidence of TEL-AML1 fusion gene analyzed consecutively in children with acute lymphoblastic leukemia in relapse. Blood. 1997, 90: 4933-7.PubMed
9.
Zurück zum Zitat Rubnitz JE, Behm FG, Wichlan D, Ryan C, Sandlund JT, Ribeiro RC, Rivera GK, Hancock ML, Relling MV, Evans WE, Pui CH, Downing JR: Low frequency of TEL-AML1 in relapsed acute lymphoblastic leukemia supports a favorable prognosis for this genetic subgroup. Leukemia. 1999, 13: 19-21. 10.1038/sj/leu/2401257.CrossRefPubMed Rubnitz JE, Behm FG, Wichlan D, Ryan C, Sandlund JT, Ribeiro RC, Rivera GK, Hancock ML, Relling MV, Evans WE, Pui CH, Downing JR: Low frequency of TEL-AML1 in relapsed acute lymphoblastic leukemia supports a favorable prognosis for this genetic subgroup. Leukemia. 1999, 13: 19-21. 10.1038/sj/leu/2401257.CrossRefPubMed
10.
Zurück zum Zitat Anguita E, Gonzalez FA, Lopez J, Villegas A: TEL-AML1 transcript and p16 gene deletion in a patient with childhood acute lymphoblastic leukemia. Br J Haematol. 1997, 99 (1): 240-241.PubMed Anguita E, Gonzalez FA, Lopez J, Villegas A: TEL-AML1 transcript and p16 gene deletion in a patient with childhood acute lymphoblastic leukemia. Br J Haematol. 1997, 99 (1): 240-241.PubMed
11.
Zurück zum Zitat Gaynon PS, Bleyer WA, Steinherz PG, Finklestein JZ, Littman P, Miller DR, Reaman G, Sather H, Hammond GD: Day 7 marrow response and outcome for children with acute lymphoblastic leukemia and unfavorable presenting features. Med Pediatr Oncol. 1990, 18: 273-279. 10.1002/mpo.2950180403.CrossRefPubMed Gaynon PS, Bleyer WA, Steinherz PG, Finklestein JZ, Littman P, Miller DR, Reaman G, Sather H, Hammond GD: Day 7 marrow response and outcome for children with acute lymphoblastic leukemia and unfavorable presenting features. Med Pediatr Oncol. 1990, 18: 273-279. 10.1002/mpo.2950180403.CrossRefPubMed
12.
Zurück zum Zitat Miller DR, Coccia PF, Bleyer WA, Lukens JN, Siegel SE, Sather HN, Hammond GD: Early response to induction therapy as a predictor of disease-free survival and late recurrence of childhood acute lymphoblastic leukemia: a report from the Children's Cancer Study Group. J Clin Oncol. 1989, 7: 1807-1815.PubMed Miller DR, Coccia PF, Bleyer WA, Lukens JN, Siegel SE, Sather HN, Hammond GD: Early response to induction therapy as a predictor of disease-free survival and late recurrence of childhood acute lymphoblastic leukemia: a report from the Children's Cancer Study Group. J Clin Oncol. 1989, 7: 1807-1815.PubMed
13.
Zurück zum Zitat Kitchingman GR: Residual disease detection in multiple follow-up samples in children with acute lymphoblastic leukemia. Leukemia. 1994, 8: 395-401.PubMed Kitchingman GR: Residual disease detection in multiple follow-up samples in children with acute lymphoblastic leukemia. Leukemia. 1994, 8: 395-401.PubMed
14.
Zurück zum Zitat Biondi A, Yokota S, Hansen-Hagge TE, Rossi V, Giudici G, Maglia O, Basso G, Tell C, Masera G, Bartram CR: Minimal residual disease in childhood acute lymphoblastic leukemia: analysis of patients in continuous complete remission or with consecutive relapse. Leukemia. 1992, 6: 282-288.PubMed Biondi A, Yokota S, Hansen-Hagge TE, Rossi V, Giudici G, Maglia O, Basso G, Tell C, Masera G, Bartram CR: Minimal residual disease in childhood acute lymphoblastic leukemia: analysis of patients in continuous complete remission or with consecutive relapse. Leukemia. 1992, 6: 282-288.PubMed
15.
Zurück zum Zitat Potter MN, Steward CG, Oakhill A: The significance of detection of minimal residual disease in childhood acute lymphoblastic leukaemia. Br J Haematol. 1993, 83: 412-418. 10.1111/j.1365-2141.1993.tb04665.x.CrossRefPubMed Potter MN, Steward CG, Oakhill A: The significance of detection of minimal residual disease in childhood acute lymphoblastic leukaemia. Br J Haematol. 1993, 83: 412-418. 10.1111/j.1365-2141.1993.tb04665.x.CrossRefPubMed
16.
Zurück zum Zitat Brisco MJ, Condon J, Hughes E, Neoh SH, Sykes PJ, Seshadri R, Morley AA, Toogood I, Waters K, Tauro G, Ekert H: Outcome prediction in childhood acute lymphoblastic leukaemia by molecular quantification of residual disease at the end of induction. Lancet. 1994, 343: 196-200. 10.1016/S0140-6736(94)90988-1.CrossRefPubMed Brisco MJ, Condon J, Hughes E, Neoh SH, Sykes PJ, Seshadri R, Morley AA, Toogood I, Waters K, Tauro G, Ekert H: Outcome prediction in childhood acute lymphoblastic leukaemia by molecular quantification of residual disease at the end of induction. Lancet. 1994, 343: 196-200. 10.1016/S0140-6736(94)90988-1.CrossRefPubMed
17.
Zurück zum Zitat Neale GA, Menarguez J, Kitchingman GR, Fitzgerald TJ, Koehler M, Mirro J, Goorha RM: Detection of minimal residual disease in T-cell acute lymphoblastic leukemia using polymerase chain reaction. Blood. 1991, 78: 739-747.PubMed Neale GA, Menarguez J, Kitchingman GR, Fitzgerald TJ, Koehler M, Mirro J, Goorha RM: Detection of minimal residual disease in T-cell acute lymphoblastic leukemia using polymerase chain reaction. Blood. 1991, 78: 739-747.PubMed
18.
Zurück zum Zitat Yamada M, Wasserman R, Lange B, Reichard BA, Womer RB, Rovera G: Minimal residual disease in childhood B-lineage lymphoblastic leukemia: persistence of leukemic cells during the first 18 months of treatment. N Engl J Med. 1990, 323: 448-455.CrossRefPubMed Yamada M, Wasserman R, Lange B, Reichard BA, Womer RB, Rovera G: Minimal residual disease in childhood B-lineage lymphoblastic leukemia: persistence of leukemic cells during the first 18 months of treatment. N Engl J Med. 1990, 323: 448-455.CrossRefPubMed
19.
Zurück zum Zitat Brüggemann M, Raff T, Flohr T, Gökbuget N, Nakao M, Droese J, Lüschen S, Pott C, Ritgen M, Scheuring U, Horst HA, Thiel E, Hoelzer D, Bartram CR, Kneba M, German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia: Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006, 107: 1116-1123. 10.1182/blood-2005-07-2708.CrossRefPubMed Brüggemann M, Raff T, Flohr T, Gökbuget N, Nakao M, Droese J, Lüschen S, Pott C, Ritgen M, Scheuring U, Horst HA, Thiel E, Hoelzer D, Bartram CR, Kneba M, German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia: Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006, 107: 1116-1123. 10.1182/blood-2005-07-2708.CrossRefPubMed
20.
Zurück zum Zitat Nyvold C, Madsen HO, Ryder LP, Seyfarth J, Svejgaard A, Clausen N, Wesenberg F, Jonsson OG, Forestier E, Schmiegelow K, Nordic Society for Pediatric Hematology and Oncology: Precise quantification of minimal residual disease at day 29 allows identification of children with acute lymphoblastic leukemia and an excellent outcome. Blood. 2002, 99: 1253-1258. 10.1182/blood.V99.4.1253.CrossRefPubMed Nyvold C, Madsen HO, Ryder LP, Seyfarth J, Svejgaard A, Clausen N, Wesenberg F, Jonsson OG, Forestier E, Schmiegelow K, Nordic Society for Pediatric Hematology and Oncology: Precise quantification of minimal residual disease at day 29 allows identification of children with acute lymphoblastic leukemia and an excellent outcome. Blood. 2002, 99: 1253-1258. 10.1182/blood.V99.4.1253.CrossRefPubMed
21.
Zurück zum Zitat Aziz Z, Zahid M, Mahmood R, Maqbool S: Modified BFM protocol for childhood acute lymphoblastic leukemia: a retrospective analysis. Med Pediatr Oncol. 1997, 28: 48-2853. 10.1002/(SICI)1096-911X(199701)28:1<48::AID-MPO9>3.0.CO;2-T.CrossRefPubMed Aziz Z, Zahid M, Mahmood R, Maqbool S: Modified BFM protocol for childhood acute lymphoblastic leukemia: a retrospective analysis. Med Pediatr Oncol. 1997, 28: 48-2853. 10.1002/(SICI)1096-911X(199701)28:1<48::AID-MPO9>3.0.CO;2-T.CrossRefPubMed
22.
Zurück zum Zitat Raynaud S, Cave H, Baens M, Bastard C, Cacheux V, Grosgeorge J, Guidal-Giroux C, Guo C, Vilmer E, Marynen P, Grandchamp B: The 12;21 translocation involving TEL and deletion of the other TEL allele: two frequently associated alterations found in childhood acute lymphoblastic leukemia. Blood. 1996, 87: 2891-2899.PubMed Raynaud S, Cave H, Baens M, Bastard C, Cacheux V, Grosgeorge J, Guidal-Giroux C, Guo C, Vilmer E, Marynen P, Grandchamp B: The 12;21 translocation involving TEL and deletion of the other TEL allele: two frequently associated alterations found in childhood acute lymphoblastic leukemia. Blood. 1996, 87: 2891-2899.PubMed
23.
Zurück zum Zitat Borkhardt A, Viehmann S, Harbott J, Henze G, Lampert F: Incidence of TEL/AML1 fusion gene analyzed consecutively in children with acute lymphoblastic leukemia in relapse. Blood. 1997, 90 (12): 4933-4937.PubMed Borkhardt A, Viehmann S, Harbott J, Henze G, Lampert F: Incidence of TEL/AML1 fusion gene analyzed consecutively in children with acute lymphoblastic leukemia in relapse. Blood. 1997, 90 (12): 4933-4937.PubMed
24.
Zurück zum Zitat Seeger K, Adams HP, Buchwald D, Beyermann B, Kremens B, Niemeyer C, Ritter J, Schwabe D, Harms D, Schrappe M, Henze G: TEL-AML1 fusion transcript in relapsed childhood acute lymphoblastic leukemia. The Berlin-Frankfurt-Munster Study Group. Blood. 1998, 91: 1716-1722.PubMed Seeger K, Adams HP, Buchwald D, Beyermann B, Kremens B, Niemeyer C, Ritter J, Schwabe D, Harms D, Schrappe M, Henze G: TEL-AML1 fusion transcript in relapsed childhood acute lymphoblastic leukemia. The Berlin-Frankfurt-Munster Study Group. Blood. 1998, 91: 1716-1722.PubMed
25.
Zurück zum Zitat Lanza C, Volpe G, Basso G, Gottardi E, Barisone E, Spinelli M, Ricotti E, Cilli V, Perfetto F, Madon E, Saglio G: Outcome and lineage involvement in t(12;21) childhood acute lymphoblastic leukemia. Br J Haematol. 1997, 97: 460-462. 10.1046/j.1365-2141.1997.312676.x.CrossRefPubMed Lanza C, Volpe G, Basso G, Gottardi E, Barisone E, Spinelli M, Ricotti E, Cilli V, Perfetto F, Madon E, Saglio G: Outcome and lineage involvement in t(12;21) childhood acute lymphoblastic leukemia. Br J Haematol. 1997, 97: 460-462. 10.1046/j.1365-2141.1997.312676.x.CrossRefPubMed
26.
Zurück zum Zitat Cayuela JM, Baruchel A, Orange C, Madani A, Auclerc MF, Daniel MT, Schaison G, Sigaux F: TEL-AML1 fusion RNA as a new target to detect minimal residual disease in pediatric B-cell precursor acute lymphoblastic leukemia. Blood. 1996, 88: 302-308.PubMed Cayuela JM, Baruchel A, Orange C, Madani A, Auclerc MF, Daniel MT, Schaison G, Sigaux F: TEL-AML1 fusion RNA as a new target to detect minimal residual disease in pediatric B-cell precursor acute lymphoblastic leukemia. Blood. 1996, 88: 302-308.PubMed
27.
Zurück zum Zitat Ford AM, Fasching K, Panzer-Grümayer ER, Koenig M, Haas OA, Greaves MF: Origins of 'late' relapse in childhood acute lymphoblastic leukemia with TEL-AML1 fusion genes. Blood. 2001, 98: 558-564. 10.1182/blood.V98.3.558.CrossRefPubMed Ford AM, Fasching K, Panzer-Grümayer ER, Koenig M, Haas OA, Greaves MF: Origins of 'late' relapse in childhood acute lymphoblastic leukemia with TEL-AML1 fusion genes. Blood. 2001, 98: 558-564. 10.1182/blood.V98.3.558.CrossRefPubMed
28.
Zurück zum Zitat Lal A, Kwan E, Haber M, Norris MD, Marshall GM: Detection of minimal residual disease in peripheral blood prior to clinical relapse of childhood acute lymphoblastic leukaemia using PCR. Mol Cell Probes. 2001, 15: 99-103. 10.1006/mcpr.2001.0348.CrossRefPubMed Lal A, Kwan E, Haber M, Norris MD, Marshall GM: Detection of minimal residual disease in peripheral blood prior to clinical relapse of childhood acute lymphoblastic leukaemia using PCR. Mol Cell Probes. 2001, 15: 99-103. 10.1006/mcpr.2001.0348.CrossRefPubMed
29.
Zurück zum Zitat Radich JP: The use of PCR technology for detecting minimal residual disease in patients with leukemia. Rev Immunogenet. 1999, 1: 265-278.PubMed Radich JP: The use of PCR technology for detecting minimal residual disease in patients with leukemia. Rev Immunogenet. 1999, 1: 265-278.PubMed
30.
Zurück zum Zitat Kwong YL, Wong KF, Chan V, Chan CH: Persistence of AML1 rearrangement in peripheral blood cells in t(8;21). Cancer Genet Cytogenet. 1996, 88: 151-154. 10.1016/0165-4608(95)00282-0.CrossRefPubMed Kwong YL, Wong KF, Chan V, Chan CH: Persistence of AML1 rearrangement in peripheral blood cells in t(8;21). Cancer Genet Cytogenet. 1996, 88: 151-154. 10.1016/0165-4608(95)00282-0.CrossRefPubMed
Metadaten
Titel
Persistence of TEL-AML1 fusion gene as minimal residual disease has no additive prognostic value in CD 10 positive B-acute lymphoblastic leukemia: a FISH study
verfasst von
Eman Mosad
Hosny B Hamed
Rania M Bakry
Azza M Ezz-Eldin
Nesrine M Khalifa
Publikationsdatum
01.12.2008
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2008
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/1756-8722-1-17

Weitere Artikel der Ausgabe 1/2008

Journal of Hematology & Oncology 1/2008 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.