The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf ·...

14
Abstract. Reciprocal chromosomal translocations are recurrent features of many hematological malignancies. The cloning of the genes located at the breakpoints of chromosomal translocations in leukemia and lymphoma has led to the identification of new genes involved in carcinogenesis. Molecular studies of the breakpoint of several translocations involving chromosomal band 11q23 led to the cloning of a gene that was named MLL. Based on 7969 cases of acute myeloblastic leukemia (AML) and 1252 cases of acute lymphoblastic leukemia (ALL) taken from the literature, band 11q23 and/or the MLL gene was involved in 5.2% of AML and 22% of ALL. Differences in the frequency and the distribution of translocations were noted according to the type of acute leukemia and age of the patients. Seventy-five different rearrangements involving band 11q23 have so far been identified, 39 MLL partner genes having been cloned. The fusion of MLL and its partner gene leads to a gain of function of the MLL gene. The accumulating data suggests that the fusion protein affects the differentiation of the hematopoietic pluripotent stem cells or the lymphoid or myeloid committed stem cells by deregulating the HOX gene expression patterns. Reciprocal chromosomal translocations are recurrent features of many hematological malignancies. The cloning of the genes located at the breakpoints of chromosomal translocations in leukemia and lymphoma has led to the identification of new genes involved in carcinogenesis. These rearrangements mainly lead to the activation of a proto-oncogene by relocation near active regulatory sequences [t(11;14)(q13;q32) translocation in mantle cell lymphoma for example] or the generation of a new gene, called fusion gene [for example t(15;17)(q22;q21) leading to the PML-RAR· fusion gene] (1, 2). Most of these rearrangements are unique in that they are identified in specific leukemia and lymphoma subtypes and that they involve two specific genes. Molecular studies of the breakpoint of several translocations involving chromosomal band 11q23 led to the cloning of a gene that was named MLL (Mixed-Lineage Leukemia or Myeloid-Lymphoid Leukemia) (3-8). This gene is also known as ALL1, HTRX, HRX or TRX1. MLL abnormalities can be divided in two categories. The first category consists of MLL rearrangements, usually as translocations or insertions, some of them cryptic, leading to fusion genes with a large number of partners (9, 10). It will be the focus of this review. In addition, self-fusion of two parts of the MLL within the breakpoint cluster region (bcr), leading to internal rearrangements called partial tandem duplication (PTD), have also been described in several cases (11, 12). A second category of abnormalities is the amplification of the 11q23 region leading to the presence of multiple copies of the MLL gene, located either intrachromosomally as homo- geneously staining region (hsr), or extrachromosomally in double minutes (dmin) (13). Numerical abnormalities of chromosome 11, such as trisomies or tetrasomies, also result in additional copies of the MLL gene. Despite the large variety of rearrangements involving the MLL gene, the overall prognosis of acute myeloblastic leukemia (AML) with this abnormality is unfavorable (14, 1931 Presented at the 7th International Conference of Anticancer Research held in Corfu, Greece from October 25 - 30, 2004. Correspondence to: Pr. Marc De Braekeleer, Laboratoire de Cytogénétique, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 22, avenue Camille Desmoulins, CS 93837, F-29238 Brest cedex 3, France. Tel: + 33 (0)2 98 01 64 76, Fax: + 33 (0)2 98 01 81 89, e-mail: [email protected] Key Words: MLL gene, chromosomal band 11q23, acute leukemia. ANTICANCER RESEARCH 25: 1931-1944 (2005) Review The MLL Gene and Translocations Involving Chromosomal Band 11q23 in Acute Leukemia MARC DE BRAEKELEER 1,2 , FRÉDÉRIC MOREL 1,2 , MARIE-JOSÉE LE BRIS 2 , ANGÈLE HERRY 1 and NATHALIE DOUET-GUILBERT 1,2 1 Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest; 2 Service de Cytogénétique, Cytologie et Biologie de la Reproduction, CHU Morvan, Brest, France 0250-7005/2005 $2.00+.40

Transcript of The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf ·...

Page 1: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

Abstract. Reciprocal chromosomal translocations are recurrentfeatures of many hematological malignancies. The cloning of thegenes located at the breakpoints of chromosomal translocationsin leukemia and lymphoma has led to the identification of newgenes involved in carcinogenesis. Molecular studies of thebreakpoint of several translocations involving chromosomal band11q23 led to the cloning of a gene that was named MLL. Basedon 7969 cases of acute myeloblastic leukemia (AML) and 1252cases of acute lymphoblastic leukemia (ALL) taken from theliterature, band 11q23 and/or the MLL gene was involved in5.2% of AML and 22% of ALL. Differences in the frequencyand the distribution of translocations were noted according to thetype of acute leukemia and age of the patients. Seventy-fivedifferent rearrangements involving band 11q23 have so far beenidentified, 39 MLL partner genes having been cloned. The fusionof MLL and its partner gene leads to a gain of function of theMLL gene. The accumulating data suggests that the fusionprotein affects the differentiation of the hematopoietic pluripotentstem cells or the lymphoid or myeloid committed stem cells byderegulating the HOX gene expression patterns.

Reciprocal chromosomal translocations are recurrent

features of many hematological malignancies. The cloning

of the genes located at the breakpoints of chromosomal

translocations in leukemia and lymphoma has led to the

identification of new genes involved in carcinogenesis.

These rearrangements mainly lead to the activation of a

proto-oncogene by relocation near active regulatory

sequences [t(11;14)(q13;q32) translocation in mantle cell

lymphoma for example] or the generation of a new gene,called fusion gene [for example t(15;17)(q22;q21) leading to

the PML-RAR· fusion gene] (1, 2). Most of these

rearrangements are unique in that they are identified in

specific leukemia and lymphoma subtypes and that they

involve two specific genes.

Molecular studies of the breakpoint of several

translocations involving chromosomal band 11q23 led to the

cloning of a gene that was named MLL (Mixed-Lineage

Leukemia or Myeloid-Lymphoid Leukemia) (3-8). This

gene is also known as ALL1, HTRX, HRX or TRX1.

MLL abnormalities can be divided in two categories. The

first category consists of MLL rearrangements, usually as

translocations or insertions, some of them cryptic, leading to

fusion genes with a large number of partners (9, 10). It will be

the focus of this review. In addition, self-fusion of two parts

of the MLL within the breakpoint cluster region (bcr), leading

to internal rearrangements called partial tandem duplication

(PTD), have also been described in several cases (11, 12). A

second category of abnormalities is the amplification of the

11q23 region leading to the presence of multiple copies of the

MLL gene, located either intrachromosomally as homo-

geneously staining region (hsr), or extrachromosomally in

double minutes (dmin) (13). Numerical abnormalities of

chromosome 11, such as trisomies or tetrasomies, also result

in additional copies of the MLL gene.

Despite the large variety of rearrangements involving the

MLL gene, the overall prognosis of acute myeloblastic

leukemia (AML) with this abnormality is unfavorable (14,

1931

Presented at the 7th International Conference of Anticancer

Research held in Corfu, Greece from October 25 - 30, 2004.

Correspondence to: Pr. Marc De Braekeleer, Laboratoire de

Cytogénétique, Faculté de Médecine et des Sciences de la Santé,

Université de Bretagne Occidentale, 22, avenue Camille Desmoulins,

CS 93837, F-29238 Brest cedex 3, France. Tel: + 33 (0)2 98 01 64 76,

Fax: + 33 (0)2 98 01 81 89, e-mail: [email protected]

Key Words: MLL gene, chromosomal band 11q23, acute leukemia.

ANTICANCER RESEARCH 25: 1931-1944 (2005)

Review

The MLL Gene and Translocations Involving Chromosomal Band 11q23 in Acute Leukemia

MARC DE BRAEKELEER1,2, FRÉDÉRIC MOREL1,2, MARIE-JOSÉE LE BRIS2,

ANGÈLE HERRY1 and NATHALIE DOUET-GUILBERT1,2

1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest;

2Service de Cytogénétique, Cytologie et Biologie de la Reproduction, CHU Morvan, Brest, France

0250-7005/2005 $2.00+.40

Page 2: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

15). Therefore, detection of MLL disruption or

amplification is much needed for treatment decision.

Studying the wide variety of fusion genes involving MLLcould also lead to a better understanding of

leukemogenesis.

Distribution of rearrangements involving band11q23 and/or the MLL gene

Numerous studies have calculated the prevalence of

rearrangements of band 11q23 by banding cytogenetics and,

more recently, of the MLL gene by Southern blotting or

fluorescent in situ hybridization (FISH). Differences have

been noted between banding cytogenetics and the other

techniques. Indeed, Southern blotting and FISH are much

more sensitive and allow the recognition of rearrangements of

the MLL gene that are undetected by banding cytogenetics.

Furthermore, although the large majority of 11q23

translocations involves the MLL gene, a few cases of acute

leukemia not involving MLL have been described (16, 17).

A literature search allowed us to identify 7969 cases of

AML and 1252 cases of acute lymphoblastic leukemia (ALL)

that were studied for rearrangements of band 11q23 and/or

the MLL gene (References in Appendix 1). Overall, 5.2% of

AML and 22% of ALL have a translocation involving 11q23

or MLL. Their distribution by age groups shows 58.7% of

the AML and 67.6% of the ALL to have a rearrangement

among patients less than 1 year old (Figure 1). These

frequencies decline steadily among patients less or more

than 15 years old.

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1932

Figure 1. Distribution of 11q23 translocations by age groups and type ofacute leukemia (based on 7969 cases of AML and 1252 cases of ALLfrom the literature).

Figure 4. Distribution of 11q23 translocations in AML by FAB subtypes(based on 3360 cases from the literature).

Figure 3. Distribution of some specific 11q23 translocations by age groupsand type of acute leukemia (based on 7969 cases of AML and 1252 casesof ALL from the literature).

Figure 2. Distribution of some specific 11q23 translocations by type ofacute leukemia (based on 7969 cases of AML and 1252 cases of ALLfrom the literature).

Page 3: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

The t(4;11)(q21;q23) translocation is solely observed in

ALL patients and is present in 43.1% of the cases (Figure

2). The second most frequent translocation is

t(11;19)(q23;p13) in ALL (13%). In AML, t(9;11)(p21;q23)

is the most frequent translocation (18.6%). The

t(10;11)(p12;q23) is rare, but still present in 3.9% of AML

patients. A wider variety of other translocations than these

4 is observed in AML (14.7%) than in ALL (8.0%). The

distribution of translocations by type of acute leukemia and

age groups shows that the t(4;11) is almost exclusively

present in ALL infants (less than 1 year old) (Figure 3). In

AML, the frequency of t(9;11) increases with age, while

translocations other than t(4;11), t(9;11), t(10;11) and

t(11;19) are found in all age groups with similar frequencies.

Moreover, the distribution by FAB (French-American-

British) subtypes is available for 3360 of the 7969 AML

cases taken from the literature. Translocations involving

band 11q23 or the MLL gene are more likely to be found in

the M5 (24.9%) and M4 (8.8%) subtypes. No patient among

the 261 with M3 had a 11q23 rearrangement (Figure 4).

In 1998, the European Union Concerted Action

Workshop on 11q23 collected 550 cases of acute leukemia

and myelodysplastic syndromes with a rearrangement of

11q23 (18) (References in Appendix 2). This workshop

confirmed that the t(4;11) is the most frequent translocation

in ALL (68.1%), whereas the t(9;11) and other

translocations are mainly found in AML (41.2% and 14.8%,

respectively) (Figure 5).

De Braekeleer et al: MLL Gene and Acute Leukemia

1933

Figure 5. Distribution of some specific 11q23 translocations by type ofacute leukemia (based on 550 cases with an acquired abnormality of11q23 reported by the European Union Concerted Action Workshop on11q23 – Secker-Walker, 1998).

Figure 8. Distribution of some specific 11q23 translocations in FABsubtype AML-M5 (based on 550 cases with an acquired abnormality of11q23 reported by the European Union Concerted Action Workshop on11q23 – Secker-Walker, 1998).

Figure 6. Distribution of some specific 11q23 translocations by age groups(based on 550 cases with an acquired abnormality of 11q23 reported bythe European Union Concerted Action Workshop on 11q23 – Secker-Walker, 1998).

Figure 7. Distribution of some specific 11q23 translocations in AML byFAB subtypes (based on 550 cases with an acquired abnormality of 11q23reported by the European Union Concerted Action Workshop on 11q23 –Secker-Walker, 1998).

Page 4: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

The distribution of translocations by age groups also

confirms that the t(4;11) is more frequent in infants (less

than 1 year old) (Figure 6), the t(9;11) being fairly equally

distributed among the different age groups and the other

translocations being more frequent after 1 year old. These

differences are partly explained by the preferential

association of some translocations with specific types of

acute leukemia that occur at different ages.

The distribution of the several translocations shows that the

t(9;11) is well represented in the different AML subtypes

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1934

Figure 9. Example of translocation involving the MLL gene.

Figure 10. Exon structure of the MLL gene and location of the breakpoint cluster region (bcr).

Page 5: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

(Figure 7). However, this may be misleading; as an example,

there was only one patient with M7. Again, no patient with M3

was found among the 550 patients with 11q23 translocations.

It should also be noted that the patients with M5a are more

likely to have a t(9;11) than patients with M5b, while other

translocations are more frequent in M5b patients (Figure 8).

More recently, Bloomfield et al. published a paper from

an international workshop on balanced chromosome

aberrations in treatment-related myelodysplastic syndromes

and acute leukemia (19). They collected 511 cases, of which

162 had a rearrangement of band 11q23 (31.7%). The

t(9;11) translocation was the most frequent (47.3%),

followed by t(11;19) (21.6%).

All these studies indicate that band 11q23 and the MLLgene are frequently rearranged in de novo and therapy-

related acute leukemia. Furthermore, although some genes

are the preferential partners in most cases, translocations

involving other partner genes are not uncommon (20, 21).

The MLL gene: structure and function

The MLL gene consists of at least 37 exons spanning over

100kb. Sequence analysis of cDNA showed an open reading

frame of some 12kb, encoding for a protein of 3969 amino

acids localizing to nuclear structures (22). Its protein

structure includes several domains (3, 15, 22-30): i) 3 AT

hooks motifs binding to the minor groove of DNA and

influencing the chromatin structure; ii) 1 transcriptional

repression domain including a cysteine-rich region (CxxC)

of homology with DNA methyltransferase, which is involved

in the epigenetic regulation of transcription by methylation

and 2 zinc-finger domains (PHD – plant homology domain)

involved in protein-protein interaction; iii) a

serine/threonine rich region acting as a trans-activator; iv)

a SET domain, in the C-terminal region, the function of

which could be to recruit chromatin remodelling complexes

to specific chromosomal regions

The MLL gene shares 3 regions of homology with the

Drosophila trithorax gene (TRX); these are the 2 zinc-finger

domains and the SET domain. In fact, the MLL gene is the

human homologue of the TRX gene, a member of a highly

conserved family of genes, which regulates the homeotic

gene complex (HOX) by maintaining the transcriptional

states in the later developmental stages (31). The TRXprotein is part of the TAC1 complex that also includes a

histone acetyltransferase (dCBP) and a SET-binding factor

(SEBF1) (32).

Similarly, the MLL protein is part of a larger complex of

at least 27 proteins, whose components are involved in

epigenetic regulation, notably nucleosome remodelling and

De Braekeleer et al: MLL Gene and Acute Leukemia

1935

Figure 11. MLL-partner fusion gene structure.

Page 6: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

histone deacetylation and methylation (26, 28, 29). Through

direct physical interactions with DNA, MLL binds to

HOXA9 and HOXC8 promoters and regulates specific HOXtarget genes (28, 29, 33). The MLL gene is expressed at high

levels in differentiated myeloid cells, at low levels in stem

cells and lymphocytes, but not in erythrocytes (34, 35).

HOX genes are a major group of transcription factors,

playing a role in the early stages of development and

hematopoietic differentiation, as well as in the later stages

of hematopoietic differentiation with a specific pattern of

expression in different lineages at various differentiation

stages. They are down-regulated upon induction of terminal

differentiation, as the stem cell differentiates into mature

cells (36-39).

MLL fusion genes: structure, chromosome locationand function

Translocations involving band 11q23 usually lead to a

breakage in the MLL gene (Figure 9). The 5’ part of the

MLL gene is retained on the derivative chromosome 11

where it is fused with the 3’ part of the partner gene.

Therefore, the active fusion gene (5’ MLL-3’partner) is

almost always located in the der(11), except in rare cases of

insertion of the 5’ MLL in another chromosome (40).

The breakpoints within the MLL gene cluster in the 8.5kb

region, called the breakpoint cluster region (bcr) located

between exons 5 and 11 (Figure 10) (24, 27, 41, 42). This

leads to a "hot spot" hypothesis supported by the

identification of a cluster of ALU repetitive elements,

recombinase signal sequences, a number of scaffold

attachment regions (SAR) and topoisomerase II consensus

binding sites (25, 27, 43 - 45). The ALU sequences are

located in the centromeric part of bcr, while the

topoisomerase II binding consensus sites are located in the

telomeric portion of bcr. These topoisomerase II binding

consensus sites include 11 sequences closely related to

topoisomerase II consensus binding sites and a perfect

consensus binding site in exon 9 (44, 46).

Acute leukemia and myelodysplastic syndromes secondary

to previous chemotherapy for a primary tumor or leukemia

have a higher frequency of MLL rearrangements if the

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1936

Figure 12. Distribution of the partners of 11q23 translocations or the MLL gene.

Page 7: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

De Braekeleer et al: MLL Gene and Acute Leukemia

1937

Table I. MLL fusion partners: chromosome location and functions.

Name Chromosome Function Other

location names

AF1p 1p32 ·-helical coils EPS15

Probable EGF receptor tyrosine kinase substrate MLLT5

Putative signal transduction

AF1q 1q21 Growth factor

LAF4 2q11-2q12 Putative transcription activator MLLT2

AF3p21 3p21 SH3-containing protein NCK1PSD

Signal transduction?

GMPS 3q24 Cell division

Guanosine monoP synthetase

LPP 3q27-3q28 Putative signal transduction

AF4 4q21 Transcription activator MLLT2

FEL

FLJ10849 4q21 ?

AF5 5q12 Dimerization protein

AF5q31 5q31 Transcription activator

GRAF 5q31 Negative regulator of RhoA KIAA0621

GTPase activating protein for Rho OPHN1L

AF6q21 6q21 Transcription factor FKHRL1

Forkhead DNA binding FOXO3A

AF6 6q27 Signal transduction MLLT4

·-helical coils

CDK6 7q21 Cyclin-dependent kinase 6

AF9 9p22 Transcription activator MLLT3

LTG9

AF9q34 9q34 Negative regulator of RAS proteins activity DAB2IP

DIP1/2

KIAA1743

FBP17 9q34 Telomere maintenance KIAA0554

AF10 10p12 Transcription activator MLLT10

Leucine zipper

ABI1 10p11.2 Cell growth inhibitor E3B1

Signal transduction SSH3BP1

LCX 10q21 Zinc-binding CXXC domain TET1

Methyltransferase domain KIAA1676

CALM 11q14-11q21 Role in integration of signals from different pathways PICALM

CBL 11q23-11q25 Negative regulatory activity in protein CBL2

tyrosine kinase-mediated signaling pathways

Page 8: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

therapeutic regimen contained topoisomerase II inhibitors

(epipodophyllotoxins such as etoposide and teniposide) (19,

47). The mechanism of action of the topoisomerase II

inhibitors (including bioflavonoids present in certain fruits

and vegetables, soybean, cocoa, tea, etc.) is still unclear.

Topoisomerase II is an ubiquitous enzyme that facilitates the

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1938

Table I. continued.

Name Chromosome Function Other

location names

ARHGEF12 11q23.3 Rho guanine nucleotide exchange factor LARG

KIAA0382

CIP29 12q13 DNA transcription

GPHN 14q23.3 Gly-receptor associated protein GPHRYN

KIAA1385

AF15q14 15q14 Growth repressor

MPFYVE 15q14 Signal transduction?

CBP 16p13.3 Transcription coactivator CREBBP

Histone acetyltransferase activity RTS

RSTS

GAS7 17p13 Growth-arrest specific protein KIAA0394

AF17 17q21 Transcription factor MLLT6

MSF 17q25 Cell cycle regulation? AF17q25

Signal transduction? MSF1

Cytoskeleton organization KIAA0991

Putative GTP binding domain PNUTL4

SEPT9

LASP1 17q11-17q21.3 ? MLN50

LIM

SH3 protein 1

ENL 19p13.3 Transcription activator LTG19

MLLT1

EEN 19p13.3 ·-helical coils SH3GL1

Signal transduction?

ELL 19p13.1 Transcription elongation factor MEN

Regulation of cell growth and survival

hCDCRel-1 22q11.2 Cytoskeleton organization PNUTL1

Putative GTP binding domain CDCRel

AF22

P300 22q13.2 Transcription coactivator E1A

Histone acetyltransferase activity EP300

Cell differentiation

AFX1 Xq13 Transcription factor MLLT7

Forkhead DNA binding FOXO4

Septin 6 Xq22 Cytoskeleton organization KIAA0128

Cytokinesis Septin 2

Page 9: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

unwinding of the DNA helix, allowing DNA replication and

transcription. The inhibitors may inhibit the ligase function

of the topoisomerase II enzyme, leaving DNA free ends that

can repair through non-homologous recombination between

the MLL and the partner genes (15, 44, 48-52).

The localization of the topoisomerase II binding

consensus sites in the telomeric portion of bcr may explain

why breakpoints in secondary leukemia and in infant

leukemia have a biased distribution towards the telomeric

portion of bcr, whereas breakpoints in adult de novoleukemia are randomly distributed within bcr (46). These

findings also raise the hypothesis that a maternal dietary

regime rich in bioflavonoids could be involved in

leukemogenesis in utero (51, 53, 54).

Fusion genes keep the AT hook domains and the

methyltransferase domain (transcriptional repression

domain) of MLL, but lack the activation domain and the

SET domain (Figure 11) (30, 42, 55-63). The fusion proteins

consist of the N-terminal portion of the MLL protein fused

to the C-terminal portion of a partner protein. Due to the

loss of the SET domain, histone methylation of the HOXA9and HOXC8 promoters cannot occur (28, 29). This induces

perturbations of HOX gene expression (64).

Partners of translocations involving band 11q23 or the

MLL gene have been localized on all the chromosomes

(Figure 12). A literature search till October 1st, 2004

enabled the recognition of 75 different rearrangements; 28

involving band 11q23 have not been proven to be associated

with a rearrangement of the MLL gene. In 8 different

translocations, fluorescent in situ hybridization showed split

signals of the MLL probe, signing a rearrangement of the

MLL gene. Thirty-nine MLL partner genes have been

identified (15, 25, 27, 30, 48, 53, 56, 65-69). They are

scattered in the whole karyotype. Different partner genes

are located in the same chromosomal band; for example,

AF9q34 and FBP17 are both located in band 9q34 and

ENL, EEN and ELL in band 19p13.

The function, sometimes still putative, of the partners

thus far identified is shown in Table I (42, 56-63, 66). Most

of the MLL partner proteins are distributed in two classes.

Some are transcription and transcription regulator factors,

while others participate in transduction signaling.

The fusion of MLL and its partner gene leads to a gain of

function of the MLL gene (53, 70-73). Two mechanisms have

been proposed. Fusion with nuclear partners having a

transcriptional activation activity induces an increased

transcriptional activation; these fusions are characterized by

a short latency and occur in infant and therapy-related acute

leukemia (for example: AF4, AF9, AF10, ENL). Fusion with

cytoplasmic partners can induce dimerization of MLL, which

results in an increased transcriptional activation; these

fusions are characterized by a longer latency and occur in

adult acute leukemia (for example: AF1p, AF6, EEN).

The MLL fusion genes usually occur in tumors of specific

hematological lineages, leading to the hypothesis that the

MLL partner plays a critical role in determining the disease

phenotype (for example: MLL-AFX1 in T-ALL, MLL-AF4

in B lineage ALL, MLL-EEN in AML, MLL-ENL in

ALL/AML) (66). This suggests that the fusion protein

affects the differentiation of the hematopoietic pluripotent

stem cells or the lymphoid or myeloid committed stem cells.

However, the HOX gene expression patterns appear to be

similarly deregulated, independently of the nature of the

fusion partner gene (64, 74-76).

Several functional consequences of MLL fusion proteins

are known (15, 34, 42, 68, 77-80). MLL fusion proteins can

interfere with cell survival regulation by immortalizing the

committed stem cells and/or by meddling in the apoptosis

process. They can also modify the function of specific

transcription factors controlling myeloid differentiation and

even stop the process. They can disrupt signaling pathways,

notably by interfering with chromatin remodeling.

The hematopoietic pluripotent stem cell alone has the

capability of self-renewal. It gives rise to a multipotent

progenitor having both lymphoid and myeloid potential,

which, in turn, leads to progenitors of lymphoid cells and of

trilineage myeloid cells that are committed to differentiate

into the various blood cells.

Several hypotheses can be formulated (15, 81-85). The

MLL fusion gene is supposedly generated in the

hematopoietic pluripotent stem cell. It could immortalize an

early progenitor cell having both lymphoid and myeloid

potential. It could also induce commitment to a given pathway

(lymphoid or myeloid) followed by differentiation arrest.

Finally, it could stay silent until the transcription programs

that are normally regulated by the MLL gene become active at

a specific differentiation stage in a specific lineage.

In conclusion, much still needs to be learnt about the

function of the MLL gene and its fusion partner genes. This

endeavor will be a major step in understanding normal

hematopoietic development and leukemogenesis.

References

1 Cleary ML: Oncogenic conversion of transcription factors by

chromosomal translocations. Cell 66: 619-622, 1991.

2 Nichols J and Nimer SD: Transcription factors, translocations,

and leukemia. Blood 80: 2953-2963, 1992.

3 Tkachuk DC, Kohler S and Cleary ML: Involvement of a

homolog of Drosophila trithorax by 11q23 chromosomal

translocations in acute leukemias. Cell 71: 691-700, 1992.

4 Gu Y, Nakamura T, Alder H, Prasad R, Canaani O, Cimino G,

Croce CM and Canaani E: The t(4;11) chromosome translocation

of human acute leukemias fuses the ALL-1 gene, related to

Drosophilia trithorax, to the AF-4 gene. Cell 71: 701-708, 1992.

5 Djabali M, Selleri L, Parry P, Bower M, Young BD and Evans

GA: A trithorax-like gene is interrupted by chromosome 11q23

translocations in acute leukemias. Nature Genet 2: 113-118, 1992.

De Braekeleer et al: MLL Gene and Acute Leukemia

1939

Page 10: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

6 Ziemin-van der Poel S, McCabe NR, Gill HJ, Espinosa RI, Patel

Y, Harden A, Rubinelli P, Smith SD, LeBeau MM, Rowley JD

and Diaz MO: Identification of a gene, MLL, that spans the

breakpoint in 11q23 translocations associated with human

leukemias. Proc Natl Acad Sci USA 88: 10735-10739, 1991.

7 McCabe NR, Burnett RC, Gill HJ, Thirman MJ, Mbangkollo D,

Kipiniak M, van Melle E, Ziemin-van der Poel S, Rowley JD and

Diaz MO: Cloning of cDNAs of the MLL gene that detect DNA

rearrangements and altered RNA transcripts in human leukemic

cells with 11q23 translocations. Proc Natl Acad Sci USA 89:

11794-11798, 1992.

8 Cimino G, Moir DT, Canaani O, Williams K, Crist WM, Katzav

S, Cannizzaro L, Lange B, Nowell PC, Croce CM and Canaani E:

Cloning of ALL-1, the locus involved in leukemias with the

t(4;11)(q21;q23), t(9;11)(p22;q23), and t(11;19)(q23;p13)

chromosome translocations. Cancer Res 51: 6712-6714, 1991.

9 Mitelman F, Johansson B and Mertens F: Mitelman database of

chromosome aberrations in cancer. http://cgap nci nih

gov/Chromosomes/Mitelman 2003.

10 Watanabe N, Kobayashi H, Ochiji O, Yoshida MA, Kikuta A,

Komada Y, Sekine I, Ishida Y, Horiukoshi Y, Tsunematsu Y,

Yano M, Nakadate H and Kaneko Y: Cryptic insertion and

translocation or nondividing leukemic cells disclosed by FISH

analysis in infant acute leukemia with discrepant molecular and

cytogenetic findings. Leukemia 17: 876-882, 2003.

11 Caligiuri MA, Schichman SA, Strout MP, Mrozek K, Baer

MR, Frankel SR, Barcos M, Herzig GP, Croce CM and

Bloomfield CD: Molecular rearrangement of the ALL-1 gene

in acute myeloid leukemia without cytogenetic evidence of

11q23 chromosomal translocations. Cancer Res 54: 370-373,

1994.

12 Schnittger S, Kinkelin U, Schoch C, Heinecke A, Haase D,

Haferlach T, Buchner T, Wormann B, Hiddemann W and

Griesinger F: Screening for MLL tandem duplication in 387

unselected patients with AML identify a prognostically

unfavorable subset of AML. Leukemia 14: 796-804, 2000.

13 Cox MC, Panetta P, Venditti A, Del Poeta G, Maurillo L,

Tamburini A, Del Principe MI and Amadori S: Fluorescence insitu hybridization and conventional cytogenetics for the diagnosis

of 11q23+/MLL+ translocation in leukaemia. Br J Haematol 121:

952-959, 2003.

14 Schoch C, Schnittger S, Klaus M, Kern W, Hiddemann W and

Haferlach T: AML with 11q23/MLL abnormalities as defined by

the WHO classification: incidence, partner chromosomes, FAB

subtype, age distribution, and prognostic impact in an unselected

series of 1897 cytogenetically analyzed AML cases. Blood 102:

2395-2402, 2003.

15 DiMartino JF and Cleary ML: MLL rearrangements in

haematological malignancies: lessons from clinical and biological

studies. Br J Haematol 106: 614-626, 1999.

16 Ibrahim S, Estey EH, Pierce S, Glassman A, Keating M, O'Brien

S, Kantarjian HM and Albitar M: 11q23 abnormalities in patients

with acute myelogenous leukemia and myelodysplastic syndrome

as detected by molecular and cytogenetic analyses. Am J Clin

Pathol 114: 793-797, 2000.

17 Giugliano E, Rege Cambrin G, Scaravaglio P, Wlodarska I,

Emanuel B, Stul M, Serra A, Tonso A, Pini M, Saglio G and

Hagemeijer A: Two new translocations involving the 11q23 region

map outside the MLL locus in myeloid leukemias. Haematologica

87: 1014-1020, 2002.

18 Secker-Walker LM and on behalf of the European 11q23

Workshop participants: General report on the European Union

Concerted Action Workshop on 11q23, London, UK, May 1997.

Leukemia 12: 776-778, 1998.

19 Bloomfield CD, Archer KJ, Mrozek K, Lillington DM,

Kaneko Y, Head DR, Dal Cin P and Raimondi SC: 11q23

balanced chromosome aberrations in treatment-related

myelodysplastic syndromes and acute leukemia: report from

an international workshop. Genes Chromosomes Cancer 33:

362-378, 2002.

20 Douet-Guilbert N, Morel F, Le Bris MJ, Sassolas B, Giroux JD

and De Braekeleer M: Rearrangement of MLL in a patient with

congenital acute monoblastic leukemia and granulocytic

sarcoma associated with a t(1;11)(p36;q23) translocation.

Leukemia Lymphoma 46: 143-146, 2005.

21 Douet-Guilbert N, Morel F, Le Bris MJ, Herry A, Morice P,

Bourquard P, Banzakour S, Le Calvez G, Marion V, Berthou C

and De Braekeleer M: Rearrangement of the MLL gene in

acute myeloblastic leukemia: report of two rare translocations.

Cancer Genet Cytogenet 157: 169-174, 2005.

22 Butler LH, Slany R, Cui X, Cleary ML and Mason DY: The

HRX proto-oncogene product is widely expressed in human

tissues and localizes to nuclear structures. Blood 89: 3361-

3370, 1997.

23 Rasio D, Schichman SA, Negrini M, Canaani E and Croce CM:

Complete exon structure of the ALL1 gene. Cancer Res 56:

1766-1769, 1996.

24 Nilson I, Lochner K, Siegler G, Greil J, Beck JD, Fey GH and

Marschalek R: Exon/intron structure of the human ALL-1

(MLL) gene involved in translocations to chromosomal

region 11q23 and acute leukaemias. Br J Haematol 93: 966-

972, 1996.

25 Marschalek R, Nilson I, Lochner K, Greim R, Siegler G, Greil

J, Beck JD and Fey GH: The structure of the human ALL-

1/MLL/HRX gene. Leukemia Lymphoma 27: 417-428, 1997.

26 Xia ZB, Anderson M, Diaz MO and Zeleknik-Le NJ: MLL

repression domain interacts with histone deacetylases, the

polycomb group proteins HPC2 and BMI-1, and the

corepressor C-terminal-binding protein. Proc Natl Acad Sci

USA 100: 8342-8347, 2003.

27 Biondi A, Cimino G, Pieters R and Pui CH: Biological and

therapeutic aspects of infant leukemia. Blood 96: 24-33, 2000.

28 Milne TA, Briggs SD, Brock HW, Martin ME, Gibbs D, Allis

CD and Hess JL: MLL targets SET domain methyltransferase

activity to Hox gene promoters. Mol Cell 10: 1107-1117, 2002.

29 Nakamura T, Mori T, Tada S, Krajewski W, Rozovskaia T,

Wassell R, Dubois G, Mazo A, Croce CM and Canaani E: ALL-

1 is a histone methyltransferase that assembles a supercomplex

of proteins involved in transcriptional regulation. Mol Cell 10:

1119-1128, 2002.

30 Ayton PM and Cleary ML: Molecular mechanisms of

leukemogenesis mediated by MLL fusion proteins. Oncogene

20: 5695-5707, 2001.

31 Simon JA and Tamkun JW: Programming off and on states in

chromatin: mechanisms of Polycomb and trithorax group

complexes. Curr Opin Genetics Develop 12: 210-218, 2002.

32 Petruk S, Sedkov Y, Smith S, Tillib S, Kraevski V, Nakamura T,

Canaani E, Croce CM and Mazo A: Trithorax and dCBP acting

in a complex to maintain expression of a homeotic gene. Science

294: 1331-1334, 2001.

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1940

Page 11: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

33 Yu BD, Hanson RD, Hess JL, Horning SE and Korsmeyer SJ:

MLL, a mammalian trithorax-group gene, functions as a

transcriptional maintenance factor in morphogenesis. Proc Natl

Acad Sci USA 95: 10632-10636, 1998.

34 Hess JL, Yu BD, Li B, Hanson R and Korsmeyer SJ: Defects in

yolk sac hematopoiesis in MLL-null embryos. Blood 90: 1799-

1806, 1997.

35 Corral J, Lavenir I, Impey H, Warren AJ, Forster A, Larson TA,

Bell S, McKenzie AN, King G and Rabbitts TH: An MLL-AF9

fusion gene made by homologous recombination causes acute

leukemia in chimeric mice: a method to create fusion oncogenes.

Cell 85: 853-861, 1996.

36 Helgason CD, Sauvageau G, Lawrence HJ, Largman C and

Humphries RK: Overexpression of HOXB4 enhances the

hematopoietic potential of embryonic stem cells differentiated invitro. Blood 87: 2740-2749, 1996.

37 Sauvageau G, Lansdorp PM, Eaves CJ, Hogge DE, Dragowska

WH, Reid DS, Largman C, Lawrence HJ and Humphries RK:

Differential expression of homeobox genes in functionally distinct

CD34+ subpopulations of human bone marrow cells. Proc Natl

Acad Sci USA 91: 12223-12227, 1994.

38 Lawrence HJ, Sauvageau G, Humphries RK and Largman C: The

role of HOX homeobox genes in normal and leukemic

hematopoiesis. Stem Cells 14: 281-291, 1996.

39 Ayton PM and Cleary ML: Transformation of myeloid

progenitors by MLL oncoproteins is dependent on Hoxa7 and

Hoxa9. Genes Develop 17: 2298-2307, 2003.

40 Douet-Guilbert N, Arnaud B, Morel F, Le Bris MJ and De

Braekeleer M: Cryptic 5' MLL gene insertion in an X

chromosome in acute myeloblastic leukemia. Cancer Genet

Cytogenet 157: 178-180, 2005.

41 Gu Y, Alder H, Nakamura T, Schichman SA, Prasad R, Canaani

O, Saito H, Croce CM and Canaani E: Sequence analysis of the

breakpoint cluster region in the ALL-1 gene involved in acute

leukemia. Cancer Res 54: 2326-2330, 1994.

42 Rowley JD: The critical role of chromosome translocations in

human leukemias. Ann Rev Genet 32: 495-519, 1998.

43 Marschalek R, Greil J, Lochner K, Nilson I, Siegler G,

Zweckbronner I, Beck JD and Fey GH: Molecular analysis of the

chromosomal breakpoint and fusion transcripts in the acute

lymphoblastic SEM cell line with chromosomal translocation

t(4;11). Br J Haematol 90: 308-320, 1995.

44 Strissel Broeker PL, Gill Super H, Thirman MJ, Pomykala H,

Yonebayashi Y, Tanabe S, Zeleknik-Le N and Rowley JD:

Distribution of 11q23 breakpoints within the MLL breakpoint

cluster region in de novo acute leukemia and in treatment-related

acute myeloid leukemia: correlation with scaffold attachment

regions and topoisomerase II consensus binding sites. Blood 87:

1912-1922, 1996.

45 Echlin-Bell DR, Smith LL, Li L, Strissel PL, Strick R, Gupta

V, Banerjee J, Larson R, Relling MV, Raimondi SC, Hayashi

Y, Taki T, Zeleznik-Le N and Rowley JD: Polymorphisms in

the MLL breakpoint cluster region (BCR). Hum Genet 113: 80-

91, 2003.

46 Cimino G, Rapanotti MC, Biondi A, Elia L, Lo Coco F,

Price C, Rossi V, Rivolta A, Canaani E, Croce CM, Mandelli

F and Greaves M: Infant acute leukemias show the same

biased distribution of ALL1 gene breaks as topoisomerase II

related secondary acute leukemias. Cancer Res 57: 2879-

2883, 1997.

47 Gill Super HJ, MacCabe NR, Thirman MJ, Larson RA, Le Beau

MM, Pedersen-Bjergaard J, Philip P, Diaz MO and Rowley JD:

Rearrangements of the MLL in therapy-related acute myeloid

leukemia in patients previously treated with agents targeting

DNA-topoisomerase II. Blood 82: 3705-3711, 1993.

48 Hunger SP, Tkachuk DC, Amylon MD, Link MP, Carroll AJ,

Welborn JL, Willman CL and Cleary ML: HRX involvement in

de novo and secondary leukemias with diverse chromosome 11q23

abnormalities. Blood 81: 3197-3203, 1993.

49 Ross W, Rowe T, Glisson B, Yalowich J and Liu L: Role of

topoisomerase II in mediating epipodophyllotoxin-induced DNA

cleavage. Cancer Res 44: 5857-5860, 1984.

50 Dassonneville L and Bailly C: Translocations chromosomiques et

leucémies induites par les médicaments anticancéreux inhibiteurs

de la topo-isomérase II. Bull Cancer 85: 254-261, 1998.

51 Strick R, Strissel PL, Borgers S, Smith SL and Rowley JD: Dietary

bioflavonoids induce cleavage in the MLL gene and may

contribute to infant leukemia. Proc Natl Acad Sci USA 97: 4790-

4795, 2000.

52 Ross JA: Dietary bioflavonoids and the MLL gene: a pathway to

infant leukemia. Proc Natl Acad Sci USA 97: 4411-4413, 2000.

53 Cimino G, Rapanotti MC, Sprovieri T and Elia L: ALL1 gene

alterations in acute leukemia: biological and clinical aspects.

Haematologica 83: 350-357, 1998.

54 Ross JA, Davies SM, Potter JD and Robison LL: Epidemiology of

childhood leukemia, with a focus on infants. Epidem Rev 16: 243-

272, 1994.

55 Zeleznik-Le NJ, Harden AM and Rowley JD: 11q23

translocations split the "AT-hook" cruciform DNA-binding region

and the transcriptional repression domain from the activation

domain of the mixed-lineage leukemia (MLL) gene. Proc Natl

Acad Sci USA 91: 10610-10614, 1994.

56 Rubnitz JE, Behm FG and Downing JR: 11q23 rearrangements

in acute leukemia. Leukemia 10: 74-82, 1996.

57 Prasad R, Leshkowitz D, Gu Y, Alder H, Nakamura T, Saito H,

Huebner K, Berger R, Croce CM and Canaani E: Leucine-zipper

dimerization motif encoded by the AF17 gene fused to ALL-1

(MLL) in acute leukemia. Proc Natl Acad Sci USA 91: 8107-8111,

1994.

58 Hashii Y, Kim JY, Sawada A, Tokimasa S, Hiroyuki F, Ohta

H, Makiko K, Takihara Y, Ozono K and Hara J: A novel

partner gene CIP29 containing a SAP domain with MLLidentified in infantile myelomonocytic leukemia. Leukemia 18:

1546-1548, 2004.

59 Sano K: Structure of AF3p21, a new member of mixed lineage

leukemia (MLL) fusion partner proteins-implication for MLL-

induced leukemogenesis. Leukemia Lymphoma 42: 595-602,

2001.

60 Chaplin T, Bernard O, Beverloo HB, Saha V, Hagemeijer A,

Berger R and Young BD: The t(10;11) translocation in acute

myeloid leukemia (M5) consistently fuses the leucine zipper motif

of AF10 onto the HRX gene. Blood 86: 2073-2076, 1995.

61 DiMartino JF, Ayton PM, Chen EH, Naftzger CC, Young BD

and Cleary ML: The AF10 leucine zipper is required for leukemic

transformation of myeloid progenitors by MLL-AF10. Blood 99:

3780-3785, 2002.

62 Linder B, Newman R, Jones LK, Debernardi S, Young BD,

Freemont P, Verrijzer CP and Saha V: Biochemical analyses of

the AF10 protein: the extended LAP/PHD-finger mediates

oligomerisation. J Mol Biol 299: 369-378, 2000.

De Braekeleer et al: MLL Gene and Acute Leukemia

1941

Page 12: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

63 Saha V, Chaplin T, Gregorini A, Ayton P and Young BD: The

leukemia-associated-protein (LAP) domain, a cysteine-rich motif,

is present in a wide range of proteins, including MLL, AF10, and

MLLT6 proteins. Proc Natl Acad Sci USA 92: 9737-9741, 1995.

64 Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer

ML, Minden MD, Sallan SE, Lander ES, Golub TR and

Korsmeyer SJ: MLL translocations specify a distinct gene

expression profile that distinguishes a unique leukemia. Nature

Genet 30: 41-47, 2002.

65 Thirman MJ, Gill HJ, Burnett RC, Mbangkollo D, McCabe NR,

Kobayashi H, Ziemin-van der Poel S, Kaneko Y, Morgan R,

Sandberg AA, Chaganti RSK, Larson RA, Le Beau MM, Diaz

MO and Rowley JD: Rearrangement of the MLL gene in acute

lymphoblastic and acute myeloid leukemias with 11q23

chromosomal translocations. N Engl J Med 329: 909-914, 1993.

66 Collins F and Rabbitts TH: The promiscuous MLL gene links

chromosomal translocations to cellular differentiation and tumour

tropism. Trends Mol Med 8: 436-442, 2002.

67 Grimwade D: The clinical significance of cytogenetic

abnormalities in acute myeloid leukaemia. Best Pract Res Clin

Haematol 14: 497-529, 2001.

68 Rowley JD: The role of chromosome translocations in

leukemogenesis. Semin Hematol 36: 59-72, 1999.

69 Rowley JD: Molecular genetics in acute leukemia. Leukemia 14:

513-517, 2000.

70 Lavau C, Szilvassy SJ, Slany R and Cleary ML: Immortalization

and leukemic transformation of a myelomonocytic precursor by

retrovirally transduced HRX-ENL. EMBO J 16: 4226-4237, 1997.

71 Dobson CL, Warren AJ, Pannell R, Forster A, Lavenir I, Corral

J, Smith AJH and Rabbitts TH: The MLL-AF9 gene fusion in

mice controls myeloproliferation and specifies acute myeloid

leukemogenesis. EMBO J 18: 3564-3574, 1999.

72 DiMartino JF, Miller T, Ayton PM, Landewe T, Hess JL, Cleary

ML and Shilatifard A: A carboxy-terminal domain of ELL is

required and sufficient for immortalization of myeloid progenitors

by MLL-ELL. Blood 96: 3887-3893, 2000.

73 Martin ME, Milne TA, Bloyer S, Galoian K, Shen W, Gibbs D,

Brock HW, Slany R and Hess JL: Dimerization of MLL fusion

proteins immortalizes hematopoietic cells. Cancer Cell 4: 197-207,

2003.

74 Ferrando AA, Armstrong SA, Neuberg DS, Sallan SE, Silverman

LB, Korsmeyer SJ and Look AT: Gene expression signatures in

MLL-rearranged T-lineage and B-presursor acute leukemias:

dominance of HOX dysregulation. Blood 102: 262-268, 2003.

75 Poppe B, Vandesompele J, Schoch C, Lindvall C, Mrozek K,

Bloomfield CD, Beverloo HB, Michaux L, Dastugue N, Herens

C, Yigit N, de Paepe A, Hagemeijer A and Speleman F:

Expression analyses identify MLL as a prominent target of 11q23

amplification and support an etiologic role for MLL gain of

function in myeloid malignancies. Blood 103: 229-235, 2004.

76 Debernardi S, Lillington DM, Chaplin T, Tomlinson S, Amess J,

Rohatiner A, Lister TA and Young BD: Genome-wide analysis

of acute myeloid leukemia with normal karyotype reveals a unique

pattern of homeobox gene expression distinct from those with

translocation-mediated fusion events. Genes Chromosomes

Cancer 37: 149-158, 2003.

77 Alcalay M, Orleth A, Sebastiani C, Meani N, Chiaradonna F,

Casciari C, Sciurpa MT, Gelmetti V, Riganelli D, Minucci S,

Fagioli M and Pelicci PG: Common themes in the pathogenesis

of acute myeloid leukemia. Oncogene 20: 5680-5694, 2001.

78 Taki T, Sako M, Tsuchida M and Hayashi Y: The

t(11;16)(q23;p13) translocation in myelodysplastic syndrome fuses

the MLL gene to the CBP gene. Blood 89: 3945-3950, 1997.

79 Ida K, Kitabayashi I, Taki T, Taniwaki M, Noro K, Yamamoto M,

Ohki M and Hayashi Y: Adenoviral E1A-associated protein p300

is involved in acute myeloid leukemia with t(11;22)(q23;q13).

Blood 90: 4699-4704, 1997.

80 Fu JF, Hsu JJ, Tang TC and Shih LY: Identification of CBL, a

proto-oncogene at 11q23.3, as a novel MLL fusion partner in a

patient with de novo acute myeloid leukemia. Genes Chromosomes

Cancer 37: 214-219, 2003.

81 Daser A and Rabbitts TH: Extending the repertoire of the mixed-

lineage leukemia gene MLL in leukemogenesis. Genes Develop

18: 965-974, 2004.

82 Weissman IL, Anderson DJ and Gage F: Stem and progenitor

cells: origins, phenotypes, lineage commitments and

transdifferentiations. Annu Rev Cell Dev Biol 17: 387-403, 2001.

83 Bonnet D and Dick JE: Human acute myeloid leukemia is

organized as a hierarchy that originates from a primitive

hematopoietic cell. Nat Med 3: 730-737, 1997.

84 Miyamoto T, Weissman IL and Akashi K: AML1/ETO-expressing

nonleukemic stem cells in acute myelogenous leukemia with 8;21

chromosomal translocation. Proc Natl Acad Sci USA 97: 7521-

7526, 2000.

85 Armstrong SA, Golub TR and Korsmeyer SJ: MLL-rearranged

leukemias: insights from gene expression profiling. Semin

Hematol 40: 268-273, 2003.

Appendix 1

References used for the calculation of the prevalence of

rearrangements of band 11q23.

1 Abe R and Sandberg AA: Significance of abnormalities involving

chromosomal segment 11q22-q25 in acute leukemia. Cancer

Genet Cytogenet 13: 121-127, 1984.

2 Andreasson P, Hoglund M, Bekassy AN, Garwicz S, Heldrup J,

Mitelman F and Johansson B: Cytogenetic and FISH studies of

a single center consecutive series of 152 childhood acute

lymphoblastic leukemias. Eur J Haematol 65: 40-51, 2000.

3 Berger R, Flandrin G, Bernheim A, Le Coniat M, Vecchione

D, Pacot A, Derre J, Daniel MT, Valensi F, Sigaux F and

Ochoa-Noguera ME: Cytogenetic studies on 519 consecutive

de novo acute nonlymphocytic leukemias. Cancer Genet

Cytogenet 29: 9-21, 1987.

4 Bernstein R, Macdougall LG and Pinto MR: Chromosome

patterns in 26 South African children with acute nonlymphocytic

leukemia (ANLL). Cancer Genet Cytogenet 11: 199-214, 1984.

5 Bernstein R, Pinto MR, Morcom G, Macdougall LG, Bezwoda

W, Dukes I, Penfold G and Mendelow B: Karyotype analysis

in acute nonlymphocytic leukemia (ANLL): comparison with

ethnic group, age, morphology, and survival. Cancer Genet

Cytogenet 6: 187-199, 1982.

6 Bloomfield CD, Archer KJ, Mrozek K, Lillington DM, Kaneko

Y, Head DR, Dal Cin P and Raimondi SC: 11q23 balanced

chromosome aberrations in treatment-related myelodysplastic

syndromes and acute leukemia: report from an international

workshop. Genes Chromosomes Cancer 33: 362-378, 2002.

7 Borkhardt A, Wuchter C, Viehmann S, Pils S, Teigler-Schlegel

A, Stanulla M, Zimmermann M, Ludwig WD, Janka-Schaub C,

Schrappe M and Harbott J: Infant acute lymphoblastic

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1942

Page 13: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

leukemia – combined cytogenetic, immunophenotypical and

molecular analysis of 77 cases. Leukemia 16: 1685-1690, 2002.

8 Bower M, Parry P, Carter M, Lillington DM, Amess J, Lister

TA, Evans G and Young BD: Prevalence and clinical

correlations of MLL gene rearrangements in AML-M4/5. Blood

84: 3776-3780, 1994.

9 Cabrera ME, Campbell M, Quintana J, Undurraga MS, Ford

AA and Greaves MF: Clinical significance and frequency of

the 11q23/MLL genetic molecular alteration in Chilean infants

with acute leukemia. Rev Med Chile 129: 634-642, 2001.

10 Casasnovas RO, Campos L, Mugneret F, Charrin C, Bene MC,

Garand R, Favre M, Sartiaux C, Chaumarel I, Bernier M, Faure

G and Solary E: Immunophenotypic patterns and cytogenetic

anomalies in acute non-lymphoblastic leukemia subtypes: a

prospective study of 432 patients. Leukemia 12: 34-43, 1998.

11 Chen CS, Sorensen PHB, Domer PH, Reaman GH, Korsmeyer

SJ, Heerema NA, Hammond GD and Kersey JH: Molecular

rearrangements on chromosome 11q23 predominate in infant

acute lymphoblastic leukemia and are associated with specific

biologic variables and poor prognosis. Blood 81: 2386-2393, 1993.

12 Chillon MC, Garcia-Sanz R, Balanzategui A, Ramos F,

Fernandez-Calvo J, Rodriguez MJ, Rodriguez-Salazar MI,

Corrales A, Calmuntia MJ, Orfao A, Gonzalez M and San

Miguel JF: Molecular characterization of acute myeloblastic

leukemia according to the new WHO classification: a different

distribution in Central-West Spain. Haematologica 86: 162-

166, 2001.

13 Cimino G, Rapanotti MC, Elia L, Biondi A, Fizzotti M, Testi

AM, Tosti S, Croce CM, Canaani E, Mandelli F and Lo Coco

F: ALL-1 gene rearrangements in acute myeloid leukemia:

association with M4-M5 French-American-British classification

subtypes and young age. Cancer Res 55: 1625-1628, 1995.

14 Cox MC, Panetta P, Venditti A, Del Poeta G, Maurillo L,

Tamburini A, Del Principe MI and Amadori S: Fluorescence

in situ hybridization and conventional cytogenetics for the

diagnosis of 11q23+/MLL+ translocation in leukaemia. Br J

Haematol 121: 952-959, 2003.

15 Dastugue N, Payen C, Lafage-Pochitaloff M, Bernard P,

Leroux D, Huguet-Rigal F, Stoppa AM, Marit G, Molina L

and Michallet M: Prognostic significance of karyotype in denovo adult acute myeloid leukemia. The BGMT group.

Leukemia 9: 1491-1498, 1995.

16 Enjeti AK, Leng TS and Sivaswaren CR: Cytogenetic

abnormalities in de novo acute myeloid leukemia in adults:

relation to morphology, age, sex and ethnicity – a single center

study from Singapore. Hematol J 5: 419-425, 2004.

17 Frohling S, Skelin S, Liebisch C, Scholl C, Schlenk RF, Dohner

H, Dohner K and for the Acute Myeloid Leukemia Study

Group Ulm: Comparison of cytogenetic and molecular

cytogenetic detection of chromosome abnormalities in 240

consecutive adult patients with acute myeloid leukemia. J Clin

Oncol 20: 2480-2485, 2002.

18 Gill Super HJ, MacCabe NR, Thirman MJ, Larson RA, Le Beau

MM, Pedersen-Bjergaard J, Philip P, Diaz MO and Rowley JD:

Rearrangements of the MLL in therapy-related acute myeloid

leukemia in patients previously treated with agents targeting

DNA-topoisomerase II. Blood 82: 3705-3711, 1993.

19 Grimwade D, Walker H, Harrison G, Oliver F, Chatters S,

Harrison CJ, Wheatley K, Burnett AK, Goldstone AH and on

behalf of the Medical Research Council Adult Leukemia

Working Party: The predictive value of hierarchical cytogenetic

classificiation in older adults with acute myeloid leukemia

(AML): analysis of 1065 patients entered into the United

Kingdom Medical Research Council AML11 trial. Blood 98:

1312-1320, 2001.

20 Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C,

Harrison G, Rees J, Hann I, Stevens R, Burnett A and

Goldstone A: The importance of diagnostic cytogenetics on

outcome in AML: analysis of 1,612 patients entered into the

MRC AML 10 trial. Blood 92: 2322-2333, 1998.

21 Haferlach T, Schoch C, Schnittger S, Kern W, Loffler H and

Hiddemann W: Distinct genetic patterns can be identified in acute

monoblastic and acute monocytic leukaemia (FAB AML M5a and

M5b): a study of 124 patients. Br J Haematol 118: 426-431, 2002.

22 Hagemeijer A, van Zanen GE, Smit EM and Hahlen K: Bone

marrow karyotypes of children with nonlymphocytic leukemia.

Pediatr Res 13: 1247-1254, 1979.

23 Hilden JM, Smith FO, Frestedt JL, McGlennen R, Howells

WB, Sorensen PHB, Arthur DC, Woods WG, Buckley J,

Bernstein ID and Kersey JH: MLL gene rearrangement,

cytogenetic 11q23 abnormalities, and expresion of the NG2

molecule in infant acute myeloid leukemia. Blood 89: 3801-

3805, 1997.

24 Ishii E, Okamura J, Tsuchida M, Kobayashi M, Akiyama Y,

Nakahata T, Kojima S, Hanada R, Horibe K, Sato T, Komada Y,

Kimura K, Sakaguchi C, Hosoya R, Oda M, Tsunematsu Y, Kawa

K, Konishi S, Tsukimoto I, Matsuyama K and Ueda K: Infant

leukemia in Japan: clinical and biological analysis of 48 cases.

Med Pediatr Oncol 19: 28-32, 1991.

25 Kaneko Y, Rowley JD, Maurer HS, Variakojis D and Moohr JW:

Chromosome pattern in childhood acute nonlymphocytic

leukemia (ANLL). Blood 60: 389-399, 1982.

26 Kaneko Y, Shikano T, Maseki N, Sakurai M, Sakurai M, Takeda

T, Hiyoshi Y, Mimaya J and Fujimoto T: Clinical characteristics

of infant acute leukemia with or without 11q23 translocations.

Leukemia 2: 672-676, 1988.

27 Kim HJ, Cho HI, Kim EC, Ko EK, See CJ, Park SY and Lee DS:

A study on 289 consecutive Korean patients with acute

leukaemias revealed fluorescence in situ hybridization detects the

MLL translocation without cytogenetic evidence both initially and

during follow-up. Br J Haematol 119: 930-939, 2002.

28 Larson RA, Le Beau MM, Vardiman JW, Testa JR, Golomb HM

and Rowley JD: The predictive value of initial cytogenetic studies

in 148 adults with acute nonlymphocytic leukemia: a 12-year study

(1970-1982). Cancer Genet Cytogenet 10: 219-236, 1983.

29 Martinez-Climent JA, Lane NJ, Rubin CM, Morgan E, Johnstone

HS, Mick R, Murphy SB, Vardiman JW, Larson RA, Le Beau

MM and Rowley JD: Clinical and prognostic significance of

chromosomal abnormalities in childhood acute myeloid leukemia

de novo. Leukemia 9: 95-101, 1995.

30 Mitelman F, Nilsson PG, Brandt L, Alimena G, Gastaldi R and

Dallapiccola B: Chromosome pattern, occupation, and clinical

features in patients with acute nonlymphocytic leukemia. Cancer

Genet Cytogenet 4(3): 214, 1981.

31 Ohyashiki K: Nonrandom cytogenetic changes in human acute

leukemia and their clinical implications. Cancer Genet Cytogenet

11: 453-471, 1984.

32 Poirel H, Rack K, Delabesse E, Radford-Weiss I, Troussard X,

Debert C, Leboeuf D, Bastard C, Picard F, Veil-Buzyn A,

Flandrin G, Bernard O and MacIntyre E: Incidence and

De Braekeleer et al: MLL Gene and Acute Leukemia

1943

Page 14: The MLL Gene and Translocations Involving …ar.iiarjournals.org/content/25/3B/1931.full.pdf · 1Laboratoire d’Histologie, Embryologie et Cytogénétique, Faculté de Médecine

characterization of MLL gene (11q23) rearrangements in acute

myeloid leukemia M1 and M5. Blood 87: 2496-2505, 1996.

33 Prigogina EL, Fleischman EW, Puchkova GP, Kulagina OE,

Majakova SA, Balakirev SA, Frenkel MA, Khvatova NV and

Peterson IS: Chromosomes in acute leukemia. Hum Genet 53: 5-

16, 1979.

34 Pui CH, Behm FG, Downing JR, Hancock ML, Shurtleff SA,

Ribeiro RC, Head DR, Mahmoud HH, Sandlund JT, Furman

WL, Roberts WM, Crist WM and Raimondi SC: 11q23/MLL

rearrangement confers a poor prognosis in infants with acute

lymphoblastic leukemia. J Clin Oncol 12: 909-915, 1994.

35 Raimondi SC, Peiper SC, Kitchingman GR, Behm FG, Williams

DL, Hancock ML and Mirro J Jr: Childhood acute lymphoblastic

leukemia with chromosomal breakpoints at 11q23. Blood 73:

1627-1634, 1989.

36 Rowley JD and Potter D: Chromosomal banding patterns in acute

nonlymphocytic leukemia. Blood 47: 705-721, 1976.

37 Rubin CM, Arthur DC, Woods WG, Lange BJ, Nowell PC,

Rowley JD, Nachman J, Bostrom B, Baum ES, Suarez CR, Shah

NR, Morgan E, Maurer HS, McKenzie SE, Larson RA and Le

Beau MM: Therapy-related myelodysplastic syndrome and acute

myeloid leukemia in children: correlation between chromosomal

abnormalities and prior therapy. Blood 78: 2982-2988, 1991.

38 Rubnitz JE, Behm FG, Pui CH, Evans WE, Relling MV,

Raimondi SC, Harrison PL, Sandlund JT, Ribeiro RC, Grosveld

G and Downing JR: Genetic studies of childhood acute

lymphoblastic leukemia with emphasis on p16, MLL, and ETV6gene abnormalities: results of St Jude total therapy study XII.

Leukemia 11: 1201-1206, 1997.

39 Satake N, Maseki N, Nishiyama M, Kobayashi H, Sakurai M,

Inaba H, Katano N, Horikoshi Y, Eguchi H, Miyake M, Seto M

and Kaneko Y: Chromosome abnormalities and MLL

rearrangements in acute myeloid leukemia of infants. Leukemia

13: 1013-1017, 1999.

40 Schoch C, Schnittger S, Klaus M, Kern W, Hiddemann W and

Haferlach T: AML with 11q23/MLL abnormalities as defined by

the WHO classification: incidence, partner chromosomes, FAB

subtype, age distribution, and prognostic impact in an unselected

series of 1897 cytogenetically analyzed AML cases. Blood 102:

2395-2402, 2003.

41 Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS,

Mohamed A, Paietta E, Willman CL, Head DR, Rowe JM,

Forman SJ, Appelbaum FR and for the Southwest Oncology

Group and the Eastern Cooperative Oncology Group: Karyotypic

analysis predicts outcome of preremission and postremission

therapy in adult acute myeloid leukemia: a Southwest Oncology

Group/Eastern Cooperative Oncology Group study. Blood 96:

4075-4083, 2000.

42 Sorensen PHB, Chen CS, Smith FO, Arthur DC, Domer PH,

Bernstein ID, Korsmeyer SJ, Hammond GD and Kersey JH:

Molecular rearrangements of the MLL gene are present in most

cases of infant acute myeloid leukemia and are strongly correlated

with monocytic or myelomonocytic phenotypes. J Clin Invest 93:

429-437, 1994.

43 Takeuchi J, Ohshima T and Amaki I: Cytogenetic studies in adult

acute leukemias. Cancer Genet Cytogenet 4: 293-302, 1981.

44 Taki T, Ida K, Bessho F, Hanada R, Kikuchi A, Yamamoto K,

Sako M, Tsuchida M, Seto M, Ueda R and Hayashi Y: Frequency

and clinical significance of the MLL gene rearrangements in

infant acute leukemia. Leukemia 10: 1303-1307, 1996.

Appendix 2

References of the European Union Concerted Action Workshop on

11q23 used for the calculation of the prevalence of rearrangements

of band 11q23.

1 Bain BJ, Moorman AV, Johansson B, Mehta AB, Secker-Walker

LM and on behalf of the European 11q23 Workshop participants:

Myelodysplastic syndromes associated with 11q23 translocations.

Leukemia 12: 834-839, 1998.

2 Harbott J, Mancini M, Verellen-Dumoulin C, Moorman AV,

Secker-Walker LM and on behalf of the European 11q23

Workshop participants.: Hematological malignancies with a

deletion of 11q23: cytogenetic and clinical aspects. Leukemia 12:

823-827, 1998.

3 Harrison CJ, Cuneo A, Clark R, Johansson B, Lafage-Pochitaloff

M, Mugneret F, Moorman AV, Secker-Walker LM and on behalf

of the European 11q23 Workshop participants: ten novel 11q23

chromosomal partner sites. Leukemia 12: 811-822, 1998.

4 Johansson B, Moorman AV, Haas OA, Watmore AE, Cheung KL,

Swanton S, Secker-Walker LM and on behalf of the European

11q23 Workshop participants: Hematologic malignancies with

t(4;11)(q21;q23) – a cytogenetic, morphologic, immunophenotypic

and clinical study of 183 cases. Leukemia 12: 779-787, 1998.

5 Johansson B, Moorman AV, Secker-Walker LM and on behalf of

the European 11q23 Workshop participants: Derivative

chromosomes of 11q23-translocations in hematologic malignancies.

Leukemia 12: 828-833, 1998.

6 Lillington DM, Young BD, Berger R, Martineau M, Moorman

AV, Secker-Walker LM and on behalf of the European 11q23

Workshop participants: The t(10;11)(p12;q23) translocation in

acute leukaemia: a cytogenetic and clinical study of 20 patients.

Leukemia 12: 801-804, 1998.

7 Martineau M, Berger R, Lillington DM, Moorman AV, Secker-

Walker LM and on behalf of the EU Concerted Action 11q23

Workshop participants: The t(6;11)(q27;q23) translocation in

acute leukemia: a laboratory and clinical study of 30 cases.

Leukemia 12: 788-791, 1998.

8 Moorman AV, Hagemeijer A, Charrin C, Rieder H, Secker-

Walker LM and on behalf of the EU Concerted Action 11q23

Workshop participants: The translocations, t(11;19)(q23;p13.1)

and t(11;19)(q23;p13.3); a cytogenetic and clinical profile of 53

patients. Leukemia 12: 805-810, 1998.

9 Secker-Walker LM, Moorman AV, Bain BJ, Mehta AB and on

behalf of the EU Concerted Action 11q23 Workshop: Secondary

acute leukemia and myelodysplastic syndrome with 11q23

abnormalities. Leukemia 12: 840-844, 1998.

10 Secker-Walker LM and on behalf of the European 11q23

Workshop participants: General report on the European Union

Concerted Action Workshop on 11q23, London, UK, May 1997.

Leukemia 12: 776-778, 1998.

11 Swansbury GJ, Slater R, Bain BJ, Moorman AV, Secker-Walker

LM and on behalf of the European 11q23 Workshop participants:

Hematological malignancies with t(9;11)(p21-22;q23) – a laboratory

and clinical study of 125 cases. Leukemia 12: 792-800, 1998.

Received December 28, 2004Accepted February 23, 2005

ANTICANCER RESEARCH 25: 1931-1944 (2005)

1944