Therapy-related acute myeloid leukemia (t-AML) in most cases develops after chemotherapy of other malignancies and shows characteristic chromosome aberrations. Two general types of t-AML have previously been identified. One type is observed after therapy with alkylating agents and characteristically presents as therapy-related myelodysplasia with deletions or loss of the long arms of chromosomes 5 and 7 or loss of the whole chromosomes. The other type is observed after therapy with topoisomerase II inhibitors and characteristically presents as overt t-AML with recurrent balanced chromosome aberrations. Recent research suggests that these 2 general types of t-AML can now be subdivided into at least 8 genetic pathways with a different etiology and different biologic characteristics.

The genetic events leading to malignant transformation in most human tumors remain more or less concealed. A sequence of specific gene mutations has been identified in a few malignancies such as hereditary colon cancer and has in this disease been related to a stepwise transformation of normal epithelium to dysplastic cells, papillomas and, finally, localized or disseminated cancer.1 

Recent research indicates that myelodysplasia (MDS), often progressing to acute myeloid leukemia (AML), may represent a similar although more complicated model for leukemic transformation. Most cases of MDS and AML arise de novo without verified leukemogenic exposures. However, in a subset of 10% to 20% of the patients, the disease arises after previous therapy, most often chemotherapy, of other malignancies. Because the risk of therapy-related MDS (t-MDS) and therapy-related AML (t-AML) after intensive chemotherapy is often increased 100 times or more,2 99 of 100 cases of t-MDS and t-AML observed in this type of patient must be considered as excess cases directly induced by the cytostatic agents.

Approximately 50% to 60% of patients with MDS and AML de novo present chromosome abnormalities of unknown etiology,3,4 and in cases of overt AML there is most often no knowledge about a preleukemic phase of MDS. However, in t-MDS and t-AML, 85% to 90% of the patients show chromosome aberrations generally of the same types as observed in MDS and AML de novo,5-7 and the cytogenetic changes can be related to previous exposure to different chemically well-defined cytostatic agents with a known mechanism of action. In cases of overt t-AML a preleukemic phase of t-MDS, if present, is most often diagnosed at an early stage of the disease because of a regular follow-up with laboratory investigations of patients treated intensively with chemotherapy.

For 2 decades, deletions or loss of 5q and 7q or monosomy of these 2 chromosomes have been closely associated with previous therapy with alkylating agents and with presentation of the disease as t-MDS.5-7 The abnormalities of chromosomes 5 and 7 in t-MDS and t-AML have been classified together, and the abnormalities of chromosome 7, which are the most commonly observed, have for this reason sometimes been considered as the primary changes.7 

Many years later, another general type of t-AML was observed. Abnormalities of the long arm of chromosome 11 were initially observed in cases of overt t-AML after therapy with the epipodophyllotoxins.8-10 Subsequently, a variety of balanced translocations involving chromosome bands 11q23 or 21q22 in t-MDS and t-AML were shown to be significantly related to previous therapy with the whole group of topoisomerase II inhibitors.11,12 Based on this experience, 2 main groups of t-MDS and t-AML were defined,13 and their different genetic pathways were discussed.7 Much new information has now been obtained for both general types of t-MDS and t-AML, and for this reason we wish to propose a revised model for the genetic pathways of these diseases.

Pathway I in the revised model (Figure1) includes cases with deletions or loss of 7q or monosomy 7 but with normal chromosome 5. The abnormalities of chromosome 7 are the most commonly observed in t-MDS and t-AML following therapy with alkylating agents,5-7 but they are sometimes observed in only a subclone of cells or as evolutionary events during progression of the disease.14For this reason, other genetic abnormalities may be the important events in this pathway. Cases of t-MDS and t-AML with deletions or loss of 7q or monosomy 7 in many cases present additional chromosome aberrations, sometimes a balanced t(3;21)7 characteristic of t-MDS. Although the prognosis of patients belonging to pathway I is generally poor, some cases of t-MDS with monosomy 7 as the sole abnormality, only a modest cytopenia, and without an excess of blasts in the bone marrow may survive even for years on supportive therapy only.15 Mutations of the RASgenes16,17 and methylation of the of the p15gene promotor (D.H.C., J.P.-B., manuscript submitted) have been observed frequently in this pathway, but they probably do not represent primary genetic abnormalities in leukemic transformation. Mutations ofp53 are not very common.18,19 

Fig. 1.

Different genetic pathways in tMDS and tAML.

Fig. 1.

Different genetic pathways in tMDS and tAML.

Close modal

Pathway II includes cases of t-MDS and t-AML with deletions or loss of 5q or monosomy 5. These abnormalities have recently in most cases been unveiled by multicolor fluorescence in situ hybridization as unbalanced translocations to 5q.20,21 Defects of the long arm of chromosome 5 is the second most common cytogenetic abnormality of t-MDS and t-AML after therapy with alkylating agents. Abnormalities of chromosome 5 were in our series of patients always observed in all abnormal mitoses at diagnosis and were never seen as evolutionary events or in only a subclone.14 For this reason they were considered to represent more important changes in leukemogenesis than the defects of chromosome 7. Patients with abnormalities of chromosome 5 characteristically present many additional chromosome aberrations, including unidentified marker chromosomes, and sometimes deletions or loss of 7q or monosomy 7 also are observed. Mutations of the p53 gene are very common in this pathway. In 2 studies of 98 cases of t-MDS and t-AML, 20 of 26 patients with deletion or loss of 5q presented p53mutations, versus 7 of 72 patients with normal chromosome 518,19 (P < .001, χ2 test). The marker chromosomes in some cases have been identified as extensively deleted chromosome 5 or 720,21 and in other cases as derivative chromosomes with duplication or amplification of chromosome band 11q23, including the unrearranged MLLgene.22 Patients in pathway II have an extremely poor prognosis, particularly if p53 is mutated with loss of heterozygosity of the gene.19 

Pathway III comprises patients with balanced translocations to chromosome band 11q23 characteristically presenting as overt t-AML. These translocations are often observed without additional chromosome abnormalities and are the most frequently observed aberrations in t-AML following therapy with the epipodophyllotoxins.8-10 Many patients observed with these abnormalities are children.9Patients with balanced translocations to 11q23 often obtain a complete remission following intensive antileukemic therapy, but the prognosis, even in children, is poor due to relapse of t-AML.23 In this pathway the MLL gene at 11q23 is chimerically rearranged with one of its many partner genes. An interesting example is the MLL-CBP fusion of the t(11;16) predominantly observed in t-AML.24 Mutations of p53 are infrequent in this and in the following pathways.19,25 

Pathway IV in the former model was dominated by adult patients with overt t-AML and t(8;21).11,12 Subsequently, cases of t-AML with inv(16) also were observed related to therapy with topoisomerase II inhibitors,26-28 often an anthracycline. These patients present chimeric rearrangements of the core binding factor genesAML1(CBFA) at 21q22 and CBFB at 16q22, and they often have additional chromosome aberrations. Patients in this pathway show the best results following intensive antileukemic chemotherapy, and they often become long-term survivors.

Pathway V comprises cases of therapy-related acute promyelocytic leukemia with t(15;17) and chimeric rearrangement between thePML and the RARA genes. Such cases were first reported recurrently in Chinese patients treated with bimolane, a razoxane-related topoisomerase II inhibitor, for psoriasis.29 Subsequently, similar cases were observed after treatment with other topoisomerase II inhibitors, including anthracyclines, mitoxantrone,28,30-32 and radiotherapy only.33 Patients in this pathway often respond favorably to intensive antileukemic chemotherapy plus retinoic acid.

Pathway VI comprises more recently observed and rather rare cases of t-MDS and t-AML with different balanced translocations to chromosome band 11p15 and chimeric rearrangement between the NUP98 gene and its various partner genes.34-38 Almost half of the patients with these abnormalities developed leukemia after administration of topoisomerase II inhibitors, most often etoposide or anthracyclines or a combination of these drugs.

An important, still unsolved question is the extent to which each individual balanced chromosome aberration in pathways III to VI is associated with a specific topoisomerase II inhibitor. In a review of the literature of 442 cases of t-MDS and t-AML with balanced chromosome aberrations,33 it was shown that balanced translocations to 11q23, previous therapy with epipodophyllotoxins, and younger age were all significantly interrelated. When taking age into account in a multivariate analysis, there was no longer a significant association between previous therapy with epipodophyllotoxins and t-AML with translocations to 11q23. Thus, age and perhaps other factors such as race could explain the associations observed between specific drugs and specific cytogenetic subgroups of t-MDS and t-AML.

Pathway VII includes the subgroup of 10% to 15% of patients with t-MDS or t-AML and a normal karyotype, a subgroup well known for many years.5-7 As in AML de novo,39 it has been shown by multicolor fluorescence in situ hybridization that patients with t-MDS or t-AML and a normal karyotype only rarely present new cryptic cytogenetic rearrangements undetected by conventional G-banding.21 Unlike most patients with therapy-related leukemia and unbalanced chromosome aberrations, cases with a normal karyotype for unknown reasons predominantly present as overt t-AML.7 They have not been shown to be associated with any specific type of previous therapy, and they often respond to intensive antileukemic chemotherapy.7 Recently, we observed internal tandem duplications of the FLT3 or of the MLLgene in 4 of 6 patients with overt t-AML and a normal karyotype, whereas only 1 of another 76 patients with t-MDS or t-AML presented anMLL duplication.40 Both types of duplication were apparently not related to any specific type of previous therapy. Internal tandem duplications of the FLT3 and theMLL genes have previously been demonstrated mainly in cases of AML de novo with a normal karyotype,41-45 and theMLL duplications have been suggested to develop by endogenous recombinations at Alu repeats. In conclusion, patients with internal tandem duplications of FLT3 and MLL and perhaps also other patients in this pathway with a normal karyotype could represent sporadic and incidental cases of AML de novo unrelated to previous mutagenic exposures.

Pathway VIII includes patients with various chromosome aberrations uncharacteristic of t-MDS and t-AML. Some of these may likewise represent incidental cases of MDS and AML de novo; others may later turn out as recurrent but more rare cytogenetic abnormalities of t-MDS and t-AML.

Classification of t-MDS and t-AML in different genetic pathways may seem premature because major parts of leukemogenesis are still poorly understood. For instance, the specific genetic abnormalities directly cooperating with or predisposing to deletions or loss of the long arms of chromosomes 5 and 7 are still completely unknown. As far as the recurrent balanced chromosome aberrations in pathways III to V are concerned, recently developed mouse models support that their chimeric fusion genes are directly involved in leukemogenesis.46-56 It has been possible by various techniques to introduce these genes with appropriate promoters in mouse bone marrow precursors, and in many instances this resulted in the development of leukemia in a high percentage of the animals. Although the technique used has varied from study to study, and although caution must be taken in comparing these highly experimental models with human leukemia,57 at least 3 important observations have been made.

First, leukemias in mice develop after a latent period that sometimes is rather long, indicating that additional genetic events are required for leukemic transformation. Second, the phenotype of the leukemias observed in mice is sometimes very similar to its human counterpart. Thus, introduction of the PML-RARA fusion gene in mouse bone marrow progenitors resulted in the development of retinoic acid–sensitive promyelocytic leukemia,46-49 and introduction of 2 different MLL fusion genes resulted in the development of leukemias with monocytic differentiation or myelomonocytic leukemias.50,51 Finally, the potential to induce leukemias in mice seems to vary between the different chimeric genes. Thus, the AML1-MDS1-EVI1 fusion gene, primarily observed in the t(3;21) at blastic transformation of chronic myeloid leukemia and subsequently in t-MDS, often induced leukemia,52 whereas another 2 rearrangements of core binding factor genes, the AML1-ETO from the t(8;21)53,54 and the CBFB-MYH11 from the inv(16),55,56 so far have been shown only to induce defective and dysplastic hematopoiesis or impaired neutrophil maturation. The transforming potential of these transcripts has been suggested as more restricted,54 and additional exposure to a single sublethal dose of N-ethyl-N-nitrosourea was in one of these studies required for subsequent development of overt leukemia.56 The persistence of the AML1-ETOfusion transcript in bone marrow cells from most patients in long-term remission after intensive chemotherapy or even bone marrow transplantation of AML with t(8;21)58-60 further supports that the transcript, even if detected in vivo in human bone marrow cells, is not firmly associated with the development of overt leukemia.

The proposed model of genetic pathways in t-MDS and t-AML relates 3 types of etiology—alkylating agents, topoisomerase II inhibitors, and spontaneous endogenous recombinations unrelated to exogenous mutagenic exposures—to a complicated framework of cytogenetic and genetic abnormalities and clinical characteristics of these diseases. The diversity of cytogenetic and genetic abnormalities of t-MDS and t-AML is remarkable. Whereas most of the recurrent chromosome aberrations now must be considered as identified, many invisible genetic changes still remain to be discovered. Such new abnormalities could result in a further revision of the model. So far, methylation of the p15 promotor is the only abnormality observed in a high percentage of patients with t-MDS and t-AML and in their de novo counterparts. New techniques such as the microarray equipment for studies of gene expression may facilitate detection of additional and more general genetic abnormalities of MDS and AML.

Supported by grants from the Danish Cancer Society, HS Forskningspulje 1997, and Anders Hasselbalch's Foundation.

© 2002 by the American Society of Hematology

1
Kinzler
KW
Vogelstein
B
Lessons from hereditary colorectal cancer.
Cell.
87
1996
159
170
2
Pedersen-Bjergard
J
Philip
P
Larsen
SO
et al
Therapy-related myelodysplasia and acute myeloid leukemia: cytogenetic characteristics of 115 consecutive cases and risk in seven cohorts of patients treated intensively for malignant diseases in the Copenhagen series.
Leukemia.
7
1993
1975
1986
3
Greenberg
P
Cox
C
LeBeau
MM
et al
International scoring system for evaluating prognosis in myelodysplastic syndromes.
Blood.
89
1997
2079
2088
4
Grimwade
D
Walker
H
Oliver
F
et al
The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MCR AML 10 trial.
Blood.
92
1998
2322
2333
5
Le Beau
MM
Albain
KS
Larson
RA
et al
Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosomes no. 5 and 7.
J Clin Oncol.
4
1986
325
345
6
Johansson
B
Mertens
F
Heim
S
et al
Cytogenetics of secondary myelodysplasia (sMDS) and acute nonlymphocytic leukemia (sANLL).
Eur J Haematol.
47
1991
17
27
7
Pedersen-Bjergaard
J
Pedersen
M
Roulston
D
et al
Different genetic pathways in leukemogenesis for patients presenting with therapy-related myelodysplasia and therapy-related acute myeloid leukemia.
Blood.
86
1995
3542
3552
8
Ratain
MJ
Kaminer
LS
Bitran
JD
et al
Acute non-lymphocytic leukaemia following etoposide and cisplatin combination chemotherapy for advanced non-small cell carcinoma of the lung.
Blood.
70
1987
1412
1417
9
Pui
CH
Behm
FG
Raimondi
SC
et al
Secondary acute myeloid leukemia in children treated for acute lymphoid leukemia.
N Engl J Med.
321
1989
136
142
10
DeVore
R
Whitlock
J
Hainsworth
JD
et al
Therapy-related acute nonlymphocytic leukemia with monocytic features and rearrangement of chromosome 11q.
Ann Intern Med.
110
1989
740
742
11
Pedersen-Bjergaard
J
Philip
P
Balanced translocations involving chromosome bands 11q23 and 21q22 are highly characteristic of myelodysplasia and leukemia following therapy with cytostatic agents targeting at DNA-topoisomerase II.
Blood.
78
1991
1147
1148
12
Larson
RA
Le Beau
MM
Ratain
MJ
et al
Balanced translocations involving chromosome bands 11q23 and 21q22 in therapy-related leukemia.
Blood.
79
1992
1892
1893
13
Pedersen-Bjergaard
J
Rowley
JD
The balanced and the unbalanced chromosome aberrations of acute myeloid leukemia may develop in different ways and may contribute differently to malignant transformation.
Blood.
83
1994
2780
2786
14
Pedersen-Bjergaard
J
Philip
P
Larsen
SO
et al
Chromosome aberrations and prognostic factors in therapy-related myelodysplasia and acute nonlymphocytic leukemia.
Blood.
76
1990
1083
1091
15
Pedersen-Bjergaard
J
Philip
P
Pedersen
NT
et al
Acute nonlymphocytic leukemia, preleukemia, and acute myeloproliferative syndrome secondary to treatment of other malignant diseases.
Cancer.
54
1984
452
462
16
Stephenson
J
Lizhen
H
Mufti
GJ
Possible co-existence of ras activation and monosomy 7 in the leukaemic transformation of myelodysplastic syndromes.
Leuk Res.
19
1995
741
748
17
Side
L
Teel
K
Wang
P
et al
Activating RAS mutations in therapy-related myeloid disorders associated with deletions of chromosomes 5 and 7 [abstract].
Blood.
88(suppl 1)
1996
566a
18
Horiike
S
Misawa
S
Kaneko
H
et al
Distinct genetic involvement of the TP53 gene in therapy-related leukemia and myelodysplasia with chromosomal losses of nos 5 and/or 7 and its possible relationship to replication error phenotype.
Leukemia.
13
1999
1235
1242
19
Christiansen
DH
Andersen
MK
Pedersen-Bjergaard
J
Mutations with loss of heterozygosity of p53 are common in therapy-related MDS and AML following exposure to alkylating agents, and significantly associated with deletion or loss of 5q, a complex karyotype and a poor prognosis.
J Clin Oncol.
19
2001
1405
1413
20
Odero
MD
Carlson
KM
Calasanz
MJ
Rowley
JD
Further characterization of complex chromosomal rearrangements in myeloid malignancies: spectral karyotyping adds precision in defining abnormalities associated with poor prognosis.
Leukemia.
15
2001
1133
1145
21
Andersen
MK
Pedersen-Bjergaard
J
Multi-color FISH in 54 patients with therapy-related MDS/AML [abstract].
Blood.
98
2001
579a
22
Andersen
MK
Christiansen
DH
Kirchhoff
M
Pedersen-Bjergaard
J
Duplication or amplification of chromosome band 11q23, including the unrearranged MLL gene, is a recurrent abnormality in therapy-related MDS and AML, and is closely related to mutation of the TP53 gene and to previous therapy with alkylating agents.
Genes Chromosomes Cancer.
31
2001
33
41
23
Pui
C-H
Relling
MV
Rivera
GK
et al
Epipodophyllotoxin-related acute myeloid leukemia: a study of 35 cases.
Leukemia.
9
1995
1990
1996
24
Rowley
JD
Reshmi
S
Sobulo
S
et al
All patients with the t(11;16)(q23;p13.3) that involves MLL and CBP have treatment-related hematologic disorders.
Blood.
90
1997
535
541
25
Felix
CA
Hosler
MR
Provisor
D
et al
The p53 gene in pediatric therapy-related leukemia and myelodysplasia.
Blood.
87
1996
4376
4381
26
Quesnel
B
Kantarjian
H
Pedersen-Bjergaard
J
et al
Therapy-related acute myeloid leukemia with t(8;21), inv(16), and t(8;16): a report on 25 cases and review of the literature.
J Clin Oncol.
11
1993
2370
2379
27
Dissing
M
Le Beau
MM
Pedersen-Bjergaard
J
Inversion of chromosome 16 and uncommon rearrangements of the CBFB and MYH11 genes in therapy-related acute myeloid leukemia: rare events related to DNA-topoisomerase II inhibitors?
J Clin Oncol.
16
1998
1890
1896
28
Pedersen-Bjergaard
J
Andersen
MK
Johansson
B
Balanced chromosome aberrations in leukemias following chemotherapy with DNA-topoisomerase II inhibitors.
J Clin Oncol.
16
1998
1897
1898
29
Xue
Y
Lu
D
Guo
Y
Lin
B
Specific chromosomal translocations and therapy-related leukemia induced by bimolane therapy for psoriasis.
Leuk Res.
16
1992
1113
1123
30
Detourmignies
L
Castaigne
S
Stoppa
AM
et al
Therapy-related acute promyelocytic leukemia: a report on 16 cases.
J Clin Oncol.
10
1992
1430
1435
31
Hoffmann
L
Möller
P
Pedersen-Bjergaard
J
et al
Therapy-related acute promyelocytic leukemia with t(15;17)(q22;q12) following chemotherapy with drugs targeting DNA topoisomerase II: a report of two cases and a review of the literature.
Ann Oncol.
6
1995
781
788
32
Carli
PM
Sgro
C
Parchin-Geneste
N
et al
Increase therapy-related leukemia secondary to breast cancer.
Leukemia.
14
2000
1014
1017
33
Andersen
MK
Johansson
B
Larsen
SO
Pedersen-Bjergaard
J
Chromosomal abnormalities in secondary MDS and AML. Relationship to drugs and radiation with specific emphasis on the balanced rearrangements.
Haematologica.
83
1998
483
488
34
Borrow
J
Shearman
AM
Stanton
VP
Jr
et al
The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nycleoporin NUP98 and class I homeoprotein HOXA9.
Nat Genet.
12
1996
159
167
35
Arai
Y
Hosoda
F
Kobayashi
H
et al
The inv(11)(p15q22) chromosome translocation of de novo and therapy-related myeloid malignancies results in fusion of the nucleoporin gene, NUP98, with the putative RNA helicase gene, DDX10.
Blood.
89
1997
3936
3944
36
Raza-Egilmez
SZ
Jani-Sait
SN
Grossi
M
Higgins
MJ
Shows
TB
Aplan
P
NUP98-HOXD13 gene fusion in therapy-related acute myelogenous leukemia.
Cancer Res.
58
1998
4269
4273
37
Nakamura
T
Yamazaki
Y
Hatano
Y
Miura
I
NUP98 is fused to PMXI homeobox gene in human acute myelogenous leukemia with chromosome translocation t(1;11)(q23;p15).
Blood.
94
1999
741
747
38
Ahuja
HG
Felix
CA
Aplan
PD
The t(11;20)(p15;q11) chromosomal translocation associated with therapy-related myelodysplastic syndrome results in an NUP98-TOP1 fusion.
Blood.
94
1999
3258
3261
39
Zhang
FF
Murata-Collins
JL
Gaytan
P
et al
Twenty-four-color spectral karyotyping reveals chromosome aberrations in cytogenetically normal acute myeloid leukemia.
Genes Chromosomes Cancer.
28
2000
318
328
40
Christiansen
DH
Pedersen-Bjergaard
J
Internal tandem duplications of the FLT3 and MLL genes are mainly observed in atypical cases of therapy-related acute myeloid leukemia with a normal karyotype and are unrelated to type of previous therapy.
Leukemia.
15
2001
1848
1851
41
Nakao
M
Yokota
S
Iwai
T
et al
Internal tandem duplication of the FLT3 gene found in acute myeloid leukemia.
Leukemia.
10
1996
1911
1918
42
Kiyoi
H
Naoe
T
Nakano
Y
et al
Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood.
93
1999
3074
3080
43
Schnittger
S
Schoch
C
Kern
W
et al
FLT3 length mutations in AML: correlation to cytogenetics, FAB-subtype, and prognosis in 652 patients [abstract].
Blood.
96(suppl 1)
2000
826a
44
Caligiuri
MA
Strout
MP
Lawrence
D
et al
Rearrangement of ALL1 (MLL) in acute myeloid leukemia with normal cytogenetics.
Cancer Res.
58
1998
55
59
45
Schnittger
S
Kinkelin
U
Schoch
C
et al
Screening for MLL tandem duplication in 387 unselected patients with AML identify a prognostically unfavourable subset of AML.
Leukemia.
14
2000
796
804
46
Grisolano
JL
Wesselschmidt
RL
Pélicci
PG
Ley
TJ
Altered myeloid development and acute leukaemia in transgenic mice expressing PML/RARA under control of cathepsin G regulatory sequences.
Blood.
89
1997
376
387
47
Brown
D
Kogan
S
Lagasse
E
et al
A PML/RARα transgene initiates murine acute promyelocytic leukemia.
Proc Natl Acad Sci U S A.
94
1997
2551
2556
48
He
L-Z
Tribioli
C
Rivi
R
et al
Acute leukemia with promyelocytic features in PML/RARα transgenetic mice.
Proc Natl Acad Sci U S A.
94
1997
5302
5307
49
Cheng
G-X
Zhu
X-H
Men
X-Q
et al
Distinct leukemia phenotypes in transgenetic mice and different corepressor interactions generated by promyelocytic leukemia variant fusion genes PLZF-RARα and PML-RARα.
Proc Natl Acad Sci U S A.
96
1999
6318
6323
50
Corral
J
Lavenir
I
Impey
H
et al
An MLL-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: a method to create fusion oncogenes.
Cell.
85
1996
853
861
51
Lavau
C
Szilvassy
SJ
Slany
R
et al
Immortalization and leukemic transformation of a myelomonocytic precursor by retrovirally transduced HRX-ENL.
EMBO J.
16
1997
4226
4237
52
Cuenco
GM
Nucifora
G
Re
R
Human AML1/MDS1/EVI1 fusion protein induces an acute myelogenous leukemia (AML) I mice: a model for human AML.
Proc Natl Acad Sci U S A.
97
2000
1760
1765
53
Okuda
T
Cai
Z
Yang
S
et al
Expression of a knocked-in AML1-ETO leukemia gene inhibits the establishment of normal definitive hematopoiesis and directly generates dysplastic hematopoietic progenitors.
Blood.
91
1998
3134
3143
54
Rhoades
KL
Hetherington
CJ
Harakawa
N
et al
Analysis of the role of AML1-ETO in leukemogenesis, using an inducible transgenic mouse model.
Blood.
96
2000
2108
2115
55
Kogan
SC
Lagasse
E
Atwater
S
et al
The PEBP2β/MYH11 fusion created by inv(16)(p13;q22) in myeloid leukemia impairs neutrophil maturation and contributes to granulocytic dysplasia.
Proc Natl Acad Sci U S A.
95
1998
11863
11868
56
Castilla
LH
Garrett
L
Adya
N
et al
The fusion gene CBFB-MYH11 blocks myeloid differentiation and predisposes mice to acute myelomonocytic leukaemia.
Nat Genet.
23
1999
144
146
57
Westervelt
P
Ley
TJ
Seed versus soil: the importance of the target cell for transgenic models of human leukemias.
Blood.
93
1999
2143
2148
58
Kusec
R
Laczika
K
Knöbl
P
et al
AML1/ETO fusion mRNA can be detected in remission blood samples of all patients with t(8;21) acute myeloid leukemia after chemotherapy or autologous bone marrow transplantation.
Leukemia.
8
1994
735
739
59
Miyamoto
T
Nagafuji
K
Akashi
K
et al
Persistence of multipotent progenitors expressing AML1/ETO transcripts in long-term remission patients with t(8;21) acute myelogenous leukemia.
Blood.
87
1996
4789
4796
60
Jurlander
J
Caligiuri
MA
Ruutu
T
et al
Persistence of the AML1/ETO fusion transcript in patients treated with allogeneic bone marrow transplantation for t(8;21) leukemia.
Blood.
88
1996
2183
2191

Author notes

Jens Pedersen-Bjergaard, Cytogenetic Laboratory, Section 4052, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.

Sign in via your Institution