You are on page 1of 26

C H A P T E R 63

PATHOBIOLOGY OF ACUTE LYMPHOBLASTIC LEUKEMIA


Alejandro Gutierrez, Scott A. Armstrong, and A. Thomas Look

Normal lymphoid precursors undergo somatic recombination at their analysis, have been used to show that even rare ALL cases with two
immunoglobulin (Ig) or T- cell receptor (TCR) gene loci,1 and the completely different cytogenetic clones probably arise by clonal evo-
successful completion of V(D)J recombination, with the resultant lution from a single transformed progenitor.14
formation of a functional Ig or TCR, is required for the survival of
lymphocyte precursors. Positive and negative selection steps ensure
that only lymphocytes with Ig or TCRs that function appropriately LINEAGE-SPECIFIC FEATURES
within the context of an individuals immune microenvironment are OF LEUKEMIC LYMPHOBLASTS
allowed to proceed through the proliferation and differentiation steps
required for the development of mature lymphocytes. This develop- An important advance in the understanding and treatment of ALL
mental process generates a repertoire of lymphocytes with unique was the realization that malignant lymphoblasts share many of the
variations in the antigen-recognition portions of the Ig or TCR genes, features of normal lymphoid progenitors.15-17 Thus, ALL cells rear-
which form the foundation of a fully competent adaptive immune range their Ig and TCR genes and express components of antigen
system that can recognize a countless variety of foreign antigens. receptor molecules and other differentiation-linked cell-surface gly-
The acquisition of somatic genetic alterations involving oncogenes coproteins in ways that correspond to features of developing normal
or tumor suppressors can lead to the dysregulated proliferation and B and T lymphocytes. In many cases, leukemic cells appear to repre-
clonal expansion of lymphoid precursors that are characteristic of sent the clonal expansion of a lymphoid progenitor that has arrested
acute lymphoblastic leukemia (ALL). Many genes that are critical to its development at an early stage of B- or T-cell differentiation.18
leukemogenesis have been identified through the cloning and char- Furthermore, research continues with the goal of defining stem cell
acterization of genetic alterations induced by recurrent chromosomal populations in lymphoblastic leukemia, such as those that have been
translocations.2-5 Additionally, a number of translocations have prog- identified in acute myeloid leukemia (AML).19 However, with better
nostic significance and are used in modern ALL treatment protocols understanding of the normal patterns of antigen-independent lym-
to adjust the intensity of therapy. In many cases, the malignant phoid cell development, it has become clear that leukemic lympho-
transformation of lymphoid cells is the result of altered expression of blasts can show asynchronous gene expression with subtle variations
transcription factors that play critical roles in normal B- and T-cell in phenotype.20 Hence, it should not be surprising that in some cases
development, although it may also involve the aberrant expression of of ALL, the blast cell phenotypes differ from those of normal lym-
otherwise quiescent genes. More recent evidence has implicated phocyte progenitors, which is likely a result of aberrant regulation of
members of signal transduction pathways in leukemogenesis.6-8 gene expression. Still, the general concept that leukemic cells should
Although the incidence of specific genetic alterations in ALL varies be classified according to their normal developmental stage remains
according to patient age, evidence is accumulating that the pathogen- an important one, providing a basis for the study of immunophenotype-
esis underlying malignant transformation in similar genetic altera- specific genetic changes.
tions is similar across age groups.9,10

B-Cell Acute Lymphoblastic Leukemia


CLONAL ORIGIN OF LEUKEMIC LYMPHOID CELLS
The diagnosis of mature B-cell ALL is based on the detection of
Human ALL arises from a single progenitor cell that has acquired surface Ig on leukemic blasts. This rare phenotype accounts for only
somatic genetic changes, which lead to dysregulated proliferation and 2% to 3% of ALL cases, and the lymphoblasts generally have distinc-
differentiation arrest. The clonal origin of leukemia was first sug- tive morphology, with deeply basophilic cytoplasm containing promi-
gested by the identification of the Philadelphia chromosome in nent vacuoles; this morphologic pattern is designated L3 in the
chronic myeloid leukemia (CML).11 Subsequently, numerous lines of French-American-British (FAB) system.21-23 Prominent clinical fea-
evidence have provided additional support for this theory, which is tures include concomitant extramedullary lymphomatous masses
now generally accepted. Uniform structural and numerical chromo- in the abdomen or head and neck, frequent involvement of the
somal abnormalities are frequently demonstrated in all leukemic lym- central nervous system (CNS) and cranial nerves, and tumor
phoblasts from an individual patient. Identical rearrangements of Ig lysis syndrome, often complicated by acute renal failure from uric
or TCR genes, which are somatic in origin, have been demonstrated acid nephropathy.
in ALL cell populations.2,12 Additionally, identical patterns of Acute B-cell leukemia appears to be a disseminated form of
X-chromosome inactivation have been demonstrated within all cells Burkitt lymphoma because these conditions share common cytoge-
of individual patients with ALL by allelic analysis of the glucose-6- netic, molecular genetic, immunologic, cytologic, and clinical fea-
phosphate dehydrogenase gene on the X chromosome.13 Because tures.24 Acute B-cell leukemia does not respond well to chemotherapy
X-inactivation occurs in early embryogenesis before somatically traditionally used for childhood ALL. However, good outcomes have
acquired mutations begin to promote the transformed phenotype, been obtained with treatments designed for Burkitt lymphoma,
this is particularly strong evidence for the clonal origin of leukemia. which involve relatively brief but intensive regimens that emphasize
In addition, the methylation patterns of restriction fragment length cyclophosphamide and the rapid rotation of antimetabolites in high
polymorphisms in X-linked genes, as detected by Southern blot dosages.25-29 Thus, B-cell leukemia is the first form of ALL to be
935
936 Part VII Hematologic Malignancies

recognized as a distinct clinical entity based on immunophenotypic Lymphoblasts with a T-cell phenotype comprise approximately
and cytogenetic features and the first to be treated by separate pro- 15% of cases of ALL. These are often associated with distinctive
tocols designed specifically for the leukemias unique features. clinical features that include high circulating leukocyte counts, a male
predominance, CNS involvement, and a radiographically evident
thymic mass in many cases at presentation. Historically, patients
B-Precursor Acute Lymphoblastic Leukemia with T-cell ALL had an adverse prognosis compared with
patients with B-lineage ALL, but this gap has narrowed with the
Approximately 80% of ALL patients have lymphoblasts with pheno- intensification of therapy for these patients.46-48 In marked contrast to
types corresponding to those of B-cell progenitors.23,30 These cases can B-precursor ALL, in which the availability of a wide range of clinical
be identified on the basis of cell surface expression of CD19 and at and genetic prognostic markers has allowed the development of treat-
least one other recognized B lineageassociated antigen: CD20, ment protocols tailored to an individual patients risk of relapse,
CD24, CD22, CD21, or CD7923,30; most B-lineage ALL cases also robust pretreatment prognostic markers had been lacking in T-ALL49
express CD10 (common ALL antigen [CALLA]). The lymphoblasts until the recent identification of the early T-cell progenitor subtype
may also express nuclear terminal deoxynucleotidyl transferase (TdT) of T-ALL.
or CD34. About one-fourth of B-progenitor ALL cases express cyto-
plasmic Ig heavy-chain proteins and are designated preB-cell ALL.
Pre-B cases were originally shown to have a worse long-term response
to therapy compared with early pre-B cases, an observation that was Early T-Cell Progenitor Acute
later attributed to the presence of the t(1;19) translocation that forms Lymphoblastic Leukemia
the E2APBX1 fusion gene in about one-fourth of the pre-B cases.31
However, with the introduction of more intensive, risk-adjusted treat- Recent work has identified a very high-risk subset of T-ALL cases
ment regimens, the prognosis for patients with t(1;19)+ ALL has characterized by differentiation arrest at the earliest identifiable stages
improved significantly.32,33 of T-cell development, which are defined either by absence of biallelic
DNA rearrangement of Ig genes occurs before heavy-chain gene TCR deletion (ABD), indicating differentiation arrest before TCR
expression in B-cell development, providing a genetic marker of gene rearrangement, or by expression of a characteristic early T-cell
B-lymphocyte ontogeny. Korsmeyer and coworkers34,35 pioneered the precursor (ETP) phenotype.50,51 These cases comprise 8% to 15% of
use of heavy- and light-chain gene rearrangements to support an early pediatric T-ALL but appear to comprise a much higher fraction of
B-lineage origin of most ALL blasts. This work was extended to T-ALL in adults,52 thus providing one possible explanation for the
establish synchrony between Ig gene rearrangements and the expres- significantly inferior outcomes of adults versus children with T-ALL.
sion of B lineagerestricted cell surface antigens. However, Ig heavy- ABD/ETP T-ALL cases have recently been shown to harbor charac-
chain gene rearrangements have also been documented in about 15% teristic genetic alterations that include PTEN deletions and activating
of T-cell ALL cases and a similar percentage of AML cases.36-38 Thus, mutations of RAS, IL-7R, and FLT3, thus implicating these kinases,
caution must be exercised when assigning cell lineage on the basis of and the signal transduction pathways they regulate, as novel thera-
studies of Ig gene rearrangement. peutic targets in high-risk T-ALL.50,52,53 Moreover, ETP T-ALL patients
The identification of specific immunophenotypic, genetic, and also harbor oncogenic alterations that are characteristic of AML or
clinical features that predict response to therapy in patients with myelodysplastic syndrome (MDS), including DNMT3A, IDH1/2,
B-lineage ALL and the incorporation of these predictors into clinical and EZH2 mutations,52,53 suggesting that the cell of origin of ABD/
decision making are now widespread in modern ALL treatment pro- ETP T-ALL may be a multipotent hematopoietic stem or progenitor
tocols. This ability to predict outcome in these patients has been cell rather than a committed T-cell progenitor. Indeed, approximately
closely tied to the remarkable improvements in therapy for children 20% of relapses in T-ALL patients with biallelic TCR rearrange-
with this disease, which 50 years ago was universally fatal. However, ments at diagnosis are characterized by absence of biallelic TCR
many subgroups of pediatric and adult patients face a much poorer deletion (ABD) at relapse.50 It is implausible that a cell with a biallelic
prognosis, and much progress remains to be made. deletion of genomic DNA would reacquire the deleted sequences;
thus, these data further support the hypothesis that, at least in some
cases of high-risk T-ALL, the transformed leukemia-initiating clone
T-Cell Acute Lymphoblastic Leukemia is an immature progenitor characterized by differentiation arrest at
an earlier stage than that of the bulk lymphoblast population at the
Leukemias of T-cell precursors can be identified and classified accord- time of T-ALL diagnosis.
ing to the sequence of expression of T-cellassociated surface antigens
during normal thymocyte ontogeny.39,40 In this tightly regulated
process, the earliest T-cell precursors are characterized by the lack of Mixed-Lineage Leukemia
expression of CD4 and CD8 surface markers. These double-negative
(DN) thymocytes proceed through a series of differentiation stages Acute mixed-lineage leukemias are defined by blast cells that coex-
(DN1-DN4 in mice; pro-T to pre-T in humans) during which the press markers of both the lymphoid and myeloid lineages. Two dis-
cells lose CD34 expression, gain CD1a expression, and the TCR tinct forms of these leukemias are recognized: those with lymphoid
genes TCR-, TCR-, and TCR- become rearranged. The successful morphology that coexpress myeloid-associated antigen54,55 and those
rearrangement of TCR- drives the production of immature single- with myeloid morphology and reactivity to myeloperoxidase staining
positive cells (ISPs) with a surface CD4+, CD8, sCD3 phenotype that coexpress cell-surface antigens normally restricted to lymphoid
(CD4 CD8+ sCD3 in mice). These cells then differentiate into early cells. The origin of mixed-lineage leukemias has not been established.
double-positive (CD4+, CD8+) cells, at which point the TCR- gene One possibility is malignant transformation of pluripotent hemato-
rearrangement occurs. Subsequently, these double-positive progeni- poietic stem or progenitor cells that retain the ability to differentiate
tors differentiate into late cortical thymocytes showing a loss of CD1 in both the myeloid and lymphoid lineages; another is immortaliza-
and a gain of surface CD3 expression. This thymic T-cell develop- tion of rare progenitor cells that normally coexpress features of both
mental process ends when mature CD4+ or CD8+ single-positive cells lineages; and a third is aberrant gene expression caused by specific
emerge from the thymus,40,41 although further T-cell differentiation genetic alterations.56 There was initially considerable controversy over
occurs in the periphery upon antigen presentation. Numerous inves- whether patients with lymphoid leukemia with expression of one or
tigators, using a battery of monoclonal antibodies specific for T-cell more myeloid cell surface antigens (e.g., CD13, CD33, or CD14)
surface glycoproteins, have confirmed the close relationship between had an adverse prognosis. However, more recent data suggest that the
the recognizable patterns of surface antigen expression on leukemic coexpression of myeloid antigens in ALL does not carry an adverse
T cells and the normal stages of thymocyte development.42-45 prognosis in the setting of contemporary treatment regiments.57-59 The
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 937

expression of lymphoid markers in predominantly myeloid leukemias conventional cytogenetics, as well as lesions that are evident only by
is relatively rare, and although some studies reported an adverse effect molecular analysis. The ability to identify these changes and the
of T-lymphoid markers on outcomes,60 more recent analyses did not discovery that many of these can be used to predict response to
demonstrate a significant prognostic value to lymphoid antigen therapy have led to risk-adapted therapy for patients with ALL. It is
expression in AML.61 worth noting that the vast majority of patients with leukemia have
normal constitutional karyotypes, indicating that the genetic abnor-
malities in ALL lymphoblasts are usually acquired somatically and
GENETIC BASIS OF LYMPHOID LEUKEMIA thus are restricted to the malignant clone.
Chromosomal translocations are found in 75% of ALL patients
Multiple acquired genetic abnormalities are responsible for the (Fig. 63-1).22,62 They can be broadly classified as recurrent lineage-
aberrant proliferation and differentiation arrest characteristic of restricted abnormalities, which account for approximately two-thirds
ALL. These include chromosomal rearrangements detectable by of the translocations in ALL, or as random translocations, which

A
TCR translocations Random Hyperdiploid
7q35/TCR 25% 30%
14q11/TCR

Partner genes in
TCR translocations
TAL1 TAL2
LMO1 LMO2
LYL1 BHLHB1 3% 4% BCR-ABL
HOX11 MYC
HOX11L2 LCK 4% t(19;22)

MYC-lg translocations 2% 22% TEL-AML1


t(8;14), t(2;8), t(8;22) t(12;21)

6% MLL fusions
E2A-PBX1 5%
t(4;11), t(1;11), t(11, 19)
t(1;19) 1% 1%
E2A-HLF Low hypodiploid
t(17;19)

T cell Mature B cell Precursor B cell


Hyperdiploid
B Random
6%
41%
TCR translocations BCR-ABL
7q35/TCR 25% t(19;22)
14q11/TCR

Partner genes in
TCR translocations
2% TEL-AML1
TAL1 TAL2
LMO1 LMO2 2% t(12;21)
LYL1 BHLHB1 6%
HOX11 MYC 7% MLL fusions
HOX11L2 LCK
t(4;11), t(1;11), t(11, 19)

MYC-lg translocations 4% 4% Low hypodiploid

t(8;14), t(2;8), t(8;22) 3% E2A-PBX1


t(1;19)

T cell Mature B cell Precursor B cell


Figure 63-1 FREQUENCY OF THE MAJOR CHROMOSOMAL TRANSLOCATIONS IN PEDIATRIC (A)
AND ADULT (B) ACUTE LYMPHOBLASTIC LEUKEMIA (ALL). The genes affected by chromosomal transloca-
tion are shown in bold. T-cell receptor (TCR) translocations in T-ALL can activate a number of different proto-
oncogenes as shown in the insert, including TAL1, LMO1/2, HOX11, HOX11L2, and MYC.
938 Part VII Hematologic Malignancies

have been identified only in single or very small numbers of cases. control and coordinate the expression of large numbers of proteins
The characterization of genes that span the breakpoints of recurrent required for completion of lymphoid cell differentiation programs.
translocations has allowed the identification of genes that play critical Disruption of transcription factors in leukemic blasts occurs by at
roles in leukemogenesis.3-5 least two distinct mechanisms: the dysregulated expression of intact
genes and the creation of chimeric transcription factors.

Central Role of Transcription Factors


Developmental Biology of Oncogenic
Molecular studies on recurrent chromosomal translocations in ALL Transcription Factors
have demonstrated a central role for the aberrant expression of tran-
scription factors in the pathobiology of leukemia (Table 63-1). The A surprising connection has emerged from studies of oncogenic tran-
dysregulated expression of these transcription factors, which are often scription factors and the developmental proteins regulating segmenta-
functionally normal, leads to abnormal proliferation and differentia- tion in Drosophila because the DNA-binding domains of these
tion arrest of leukemic lymphoid and myeloid progenitors.3-5 Con- proteins often show striking homology. The proteins participating
served amino acid sequence motifs within the sequence-specific DNA in leukemogenesis interfere with a highly regulated network of
binding domains of these nuclear trans-activating proteins allow hematopoietic transcription factors, leading to arrested cell develop-
them to be grouped into families that, in many cases, appear to be ment at stages corresponding to those of early lymphoid or myeloid
involved in similar regulatory processes. Thus, the transcription progenitors. Recent evidence implicating the major HOX genes
factor genes in Table 63-1 are grouped according to shared structural in the control of hematopoiesis as well as embryogenesis, together
features of their DNA-binding domains: basic region helixloop with the established roles of Drosophila segmentation genes in
helix (bHLH), cysteine-rich (LIM), homeodomain (HOX), basic controlling HOM-C gene expression, suggests that the HOX loci
region-leucine zipper (bZIP), zinc-finger, A-T hook minor groove, may serve as proximal targets of a wide spectrum of hybrid and
ETS-like, or runt homology. Another important association is the dysregulated transcription factors in the acute leukemias.5 In this
lineage restriction of transcription factor genes affected by specific model, the oncogenic transcription factors inappropriately activate or
chromosomal translocations, suggesting that the proteins they encode suppress the expression of HOX genes, which in turn regulate gene
disrupt the differentiation of specific lymphoid progenitors. This programs with pleiotropic effects on normal hematopoietic cell
interpretation implicates a central role for transcription factors in the development.
initiation of leukemia and suggests that the normal developmental The mechanisms that lead to the activation of oncogenic tran-
programs of progenitor cells of different lineages are controlled by scription factors in ALL tend to vary according to the ALL subtype.
different regulatory programs. In mature B-cell ALL and in T-cell ALL, genetic lesions that lead to
Rabbitts63 has aptly described a key group of regulatory transcrip- the aberrant expression of structurally intact genes predominate, but
tion factors as the products of master genes. In his model, the the chromosomal translocations that often occur in B-precursor ALL
nuclear proteins encoded by these genes act positively to upregulate generally lead to the formation of chimeric proteins, which produced
critical target genes or negatively to interfere with normal regulatory as a result of the translocation-induced fusion of the coding sequence
pathways. The net effect is disruption of gene regulatory cascades that of two genes.

Table 63-1 Transcription Genes Affected by Chromosomal Breakpoints in the Acute Lymphoblastic Leukemias

Family* Translocation Affected Gene Disease


Basic helix-loop-helix (bHLH) proteins t(8;14)(q24;q32) MYC Burkitt lymphoma and
t(2;8)(p12;q24) MYC B-cell ALL
t(8;22)(q24;q11) MYC T-cell ALL
t(8;14)(q24;q11) MYC T-cell ALL
t(7;19)(q35;p13) LYL1 T-cell ALL
t(1;14)(p32;q11) TAL1 T-cell ALL
t(7;9)(q35;q34) TAL2 T-cell ALL
t(14;21)(q11.2;q22) BHLHB1
Cysteine-rich (LIM) proteins t(11;14)(p15;q11) LMO1 T-cell ALL
t(11;14)(p13;q11) LMO2 T-cell ALL
t(7;11)(q35;p13) LMO2 T-cell ALL
Homeodomain (HOX) proteins t(10;14)(q24;q11) HOX11 T-cell ALL
t(7;10)(q35;q24) HOX11 T-cell ALL
t(5;14)(q35;q32.2) HOX11L2 and BCL11B T-cell ALL
t(1;19)(q23;p13) E2A-PBX1 Pre-B-cell ALL
Basic-region/leucine-zipper (bZIP) proteins t(17;19)(q22;p13) E2A-HLF EPB ALL
A-T hook minor groove binding proteins
t(4;11)(q21;q23) MLL-AF4 EPB ALL
t(11;19)(q23;p13.3) MLL-ENL ALL or AML
ETS-like (TEL, ERG) proteins t(12;21)(p13;q22) TEL-AML1 ALL
Runt homology (AML1) t(12;21)(p13;q22) TEL-AML1 ALL

ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; EPB, early pre-B.
*Based on DNA-binding domain.

Partial list of MLL fusions.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 939

transcriptional activation by MYCMAX complexes promotes prolif-


Dysregulated Expression of Structurally Intact Genes eration, binding by MADMAX and other MAX heterodimers pro-
duces opposite effects; for example, MAD inhibits MYC function
Activation of MYC in B-Cell Acute both by competing with MYC for binding to MAX and by directly
Lymphoblastic Leukemia inhibiting transcription. Moreover, in addition to its well-established
role as a transcriptional regulator, MYC is also an important regulator
The vast majority of cases of mature B-cell ALL and Burkitt lym- of microRNA expression and ribosome biogenesis,87-90 thus implicat-
phoma are characterized by a translocation that places one allele of ing additional post-transcriptional regulation of protein expression
MYC from chromosome 8 under the control of the regulatory ele- through which MYC exerts its potent biologic effects.
ments of an Ig gene, either the heavy-chain gene on chromosome
14q32 or the or light-chain genes on chromosomes 2 and 22. This
leads to the aberrant expression of MYC, a prototypical basic helix BHLH, LIM, and HOX Genes in T-Cell Acute
loophelix oncogenic transcription factor.64-72 In the predominant Lymphoblastic Leukemia
t(8;14) translocation, the involved MYC locus is translocated into the
heavy-chain gene on chromosome 14 adjacent to the coding sequences In leukemias with a T-cell phenotype, chromosomal breakpoints con-
of the Ig constant region.64-66 The coding sequences of the Ig variable sistently involve the TCR enhancer (7q34) or the TCRA/D enhancer
region generally are reciprocally translocated to the distal tip of chro- (14q11), both of which are highly active in committed T-cell progeni-
mosome 8. In variant translocations, the MYC gene remains on chro- tors and can cause dysregulated expression of transcription factor
mosome 8, and portions of the respective light-chain genes are genes located at the breakpoint on the reciprocal chromosome
translocated to that chromosome downstream of the MYC locus.67-71 involved in these phenotype-specific rearrangements.91 The affected
Although the MYC coding region is not structurally altered by transcription factors include (2) genes encoding bHLH family
translocation in most cases of B-cell ALL or Burkitt lymphoma, point members, such as TAL1,92,93 TAL2,94 LYL1,95 MYC,96-98 and BHLHB199;
mutations in the N-terminal phosphorylation domain of MYC com- (2) LIM-only domain (LMO) genes, such as LMO1 and LMO2100;
monly arise in these tumors at codons 58 or 62.73-75 These codons are and (3) the orphan homeobox genes HOX11 and HOL11L1.37,101-106
phosphorylation sites involved in the regulation of the activation and The observation that T-ALL oncogenes act as master transcriptional
degradation of the protein,76 and these mutations lead not only to regulators during the embryologic development of specific organ
the aberrant stabilization of MYC protein77-79 but also inhibit the systems suggests that their aberrant expression in T-cell precursors
ability of MYC to activate the proapoptotic BIM gene while its ability may contribute to the onset of leukemia by disrupting the mecha-
to stimulate proliferation remains intact.80 nisms that control cell proliferation, differentiation, and survival
The mechanisms by which MYC exerts its potent growth- during the discrete steps of normal T-cell development. Gene expres-
promoting effects are complex and only partially understood. MYC is sion profiling and mutational analyses have shown that cases of
estimated to regulate the expression of 15% of the genome81 and leads T-ALL can be separated into five subtypes based on the pattern of
to the transcriptional activation of a large number of genes involved multistep genetic abnormalities that occur (Fig. 63-2).
in cell division, growth, metabolism, adhesion, and motility.82 It also The best characterized of the oncogenic transcription factors
leads to the transcriptional repression of many other genes, such as the involved in T-ALL is TAL1 (also known as SCL), which is altered by
cell cycle inhibitors p27 and p21. MYC exerts its transcriptional activ- the t(1;14) or by site-specific deletions in approximately one-fourth of
ity via the formation of heterodimers with its DNA-binding partner childhood T-ALL cases.107-112 TAL1 is aberrantly expressed in the leu-
protein MAXMYCMAX heterodimers bind to canonical hexa- kemic cells of 60% of children and 45% of adults with T-ALL, and its
meric E-box DNA sequences (5-CACGTG-3) where they activate expression is required for maintenance of the leukemic phenotype in
transcription.83 MAX can also heterodimerize with other bHLHZip human cell culture studies.113 TAL1 acts as a master regulatory protein
proteins, including MAD,84 MXI-1 (MAD2),85 and MNT.86 Whereas during early hematopoietic development and is required for the

p14ARF H2AX,
1. NOTCH-1 + TAL1 + LMO1/2 + CyclinD3 + MYC + p16INK4A + ATM

5q gene H2AX,
2. NOTCH-1 + LYL1 + LMO2 + CyclinD2 + MYCN + 13q gene + ATM

p14ARF
3. NOTCH-1 + HOX11 (TLX1) + NUP214-ABL + p16INK4A

4. p14ARF
NOTCH-1 + HOX11L2 (TLX3) + NUP214-ABL + p16INK4A

5. NOTCH-1 + MLLENL + HOXA9, HOXA10, HOXC6, MEIS1

Figure 63-2 MULTISTEP ONCOGENIC PATHWAYS IN T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA


(ALL). Gene expression profiling and mutation analyses have revealed that five different multistep molecular pathways
can lead to the malignant transformation of developing thymocytes. NOTCH1 mutations are seen in samples from all
five T-ALL subtypes. HOX11, HOX11L2, and TAL1-overexpressing cases show high levels of MYC expression and share
the loss of the tumor suppressor genes p16/INK4A and p14/ARF on chromosome 9p. HOX11 and HOX11L2 are often
associated with a novel NUP214-ABL episomal fusion gene, which may render these T-ALLs sensitive to imatinib.
LYL1+ cases show high levels of expression of N-MYC and frequently have deletions affecting as yet unidentified loci
on chromosomal arms 5q and 13q. Finally, MLLENL+ cases have a clearly distinct gene expression profile character-
ized by low expression of MYC and other genes involved in cell growth and proliferation but high expression of HOXA9,
HOXA10, and HOXC6 in concert with the HOX gene regulator MEIS1. (Adapted from Armstrong SA, Look AT: Molecular
genetics of acute lymphoblastic leukemia. J Clin Oncol 23:6306, 2005, with permission.)
940 Part VII Hematologic Malignancies

generation of all blood cell lineages.114,115 However, it does not seem to HOX11 and HOX11L2 are closely related in structure and have
be required for the generation and function of hematopoietic stem a high degree of homology at the amino acid level, especially in the
cells (HSCs) during adult hematopoiesis.116 This class II bHLH tran- homeobox domain, where their sequences differ by only three amino
scription factor binds to DNA by forming heterodimers with class I acids. The high level of structural homology in their DNA-binding
bHLH factors such as E2A.117 Although TAL1 binding to promoter domains supports the hypothesis that HOX11 and HOX11L2 may
sites that are normally occupied by TAL1, E2A or HEB can lead to induce T-ALL through regulation of the same transcriptional targets.
either repression or activation of transcription.113 TAL1 appears to HOX11 expression has recently been shown to induce T-ALL in mice
exert its leukemogenic effects largely via the inhibition of E2A activ- and to disrupt the mitotic checkpoint via inhibition of CHK1, thus
ity.118 The observation that loss of E2A function induces T-cell leuke- accounting for the association of HOX11/HOX11L2 overexpression
mias in mice119,120 and that the DNA-binding domain of TAL1 is with aneuploidy, which is otherwise rare in human T-ALL.149
dispensable for transformation in transgenic mouse models121 gives Recently, the analysis of gene expression profiling using oligonu-
additional support to the notion that TAL1-mediated inhibition of cleotide microarrays has shown that the expression of different tran-
E2A plays a central role in its pathogenesis in T-ALL. scription factor oncogenes such as TAL1, LYL1, HOX11, and
The LIM-only domain genes, LMO1/RBTN1/TTG1 and LMO2/ HOX11L2 is associated with distinct gene expression profiles. These
RBTN2/TTG2,100,122,123 encode proteins that possess duplicated unique signatures resemble those of thymocytes blocked at discrete
cysteine-rich LIM domains involved in proteinprotein interactions. stages of T-cell development (Fig. 63-3),17 suggesting that transcrip-
LMO2 interacts with TAL1 in erythroid cells and in T-cell tion factor oncogenes contribute to the pathogenesis of T-ALL by
leukemias.124-126 Moreover, homozygous disruption of LMO2 in mice interfering with critical regulatory networks that control cell prolif-
causes the same phenotype as described earlier for TAL1 knock-outs, eration, survival, and differentiation during T-cell development.17
indicating that a multiprotein complex is required for normal hema- Based on the patterns of aberrant gene expression, human cases of
topoietic development that involves LMO2, TAL1, and other pro- T-ALL can be classified into five different subtypes (see Fig. 63-2).
teins such as GATA1.127-130 In addition, overexpression of LMO1 or
LMO2 in thymocytes of transgenic mice leads to T-cell lymphomas,
recapitulating human T-cell tumors,131-135 and accelerates the onset of HOXA Cluster Translocations in T-Cell Acute
leukemias in TAL1 transgenic mice lines.126 Lymphoblastic Leukemia
The homeodomain gene HOX11 (also known as TLX1), located
on chromosome 10, band 24, is one of the more interesting proteins A recurrent inversion on chromosome 7 that places the HOXA cluster
activated by translocation into the vicinity of the TCR loci.37,102,104-106 in the vicinity of the TCR gene regulatory elements and that leads
HOX11 is the founding member of a family of HOX genes that to activation of the entire HOXA cluster was recently discovered in
includes HOX11L1 and HOX11L2106 and whose family members are approximately 5% of cases of T-ALL.150,151 Many of these patients also
characterized by the presence of a threonine in the third helix of the carried cooperating oncogenic lesions consisting of NOTCH1 gene
homeodomain, which confers specific DNA binding properties. mutations and deletions of 9p21.152 This new translocation that
HOX11 was originally isolated from the recurrent t(10;14)(q24;q11) directly activates HOXA gene expression provides additional evidence
in T-ALL37,102,104,105 and is aberrantly expressed in 5% of pediatric and for the role of aberrant HOXA activation in leukemogenesis and in
up to 30% of adult T-ALL patients.101,136,137 The relatively low fre- the pathogenesis of MLL- and CALM-AF10rearranged leukemias.
quency translocations involving the HOX11 locus on 10q24, 2% to
5% in childhood T-ALL105 and 14% in adult cases,138 suggests that
alternative mechanisms leading to HOX11 overexpression account for
the activation of this transcription factor oncogene in a significant
fraction of T-ALL cases. CD8+
Similar to other HOX genes, HOX11 plays an important role in CD4-

embryonic development, and functions as a master transcriptional MLL-
regulator necessary for the genesis of the spleen.139,140 In the mouse ENL
embryo, Hox11 expression can be detected in the branchial arches, LYL1+
HOX11+ TAL1+ CD8-
restricted areas of the hindbrain, and the splenic primordium,141,142
where it is required for the survival of early splenic progenitors.140 The Late CD4+
CD4- CD8- Early
HOX11 oncogenic transcription factor is associated with T-cell leu- double negative cortical cortical
kemogenesis at the early cortical thymocyte stage and relies on spe- thymocytes Mature
cific homeodomain-DNA interactions for its leukemogenic effects.143 CD4+ CD8+ single positive
Additionally, HOX11 directly interacts with the catalytic subunits of double positive T cells
the serinethreonine phosphatases PP2A and PPA, which mediates thymocytes
disruption of the G2/M cell cycle checkpoint.144 Figure 63-3 THYMOCYTE DEVELOPMENT IS A TIGHTLY REGU-
A second HOX11 family member, HOX11L2, has also been impli- LATED PROCESS IN WHICH PROGENITOR CELLS UNDERGO
cated in the pathogenesis of human T-ALL through characterization SEQUENTIAL STAGES OF DIFFERENTIATION, PROLIFERATION,
of the t(5;14)(q35;q32), a cryptic chromosomal rearrangement LINEAGE COMMITMENT, AND SELECTION THAT RESULT IN
detectable only by fluorescence in situ hybridization or by chromo- THE PRODUCTION OF FUNCTIONALLY COMPETENT MATURE
some painting techniques.103 This translocation leads to the ectopic T CELLS. Microarray gene expression profiling shows that T-ALL lympho-
expression of HOX11L2, possibly by bringing it under the influence blasts expressing high levels of the LYL1 transcription factor oncogene
of regulatory elements in the CTIP2/BCL11B gene, which is highly undergo an early arrest at the double-negative thymocyte (CD4, CD8) stage
expressed during T-lymphoid differentiation. In contrast to the pre- of development. In T-ALL cases with aberrant expression of HOX11, the
dominance of HOX11 expression in adult T-ALL cases, both the leukemic cells show a developmental arrest at the double-positive (CD4+,
t(5;14) and expression of HOX11L2 can be detected in 20% to 25% CD8+) early cortical stage of thymocyte differentiation, but those with aber-
of children but in only 5% of adults with T-ALL.101,137,145-147 The role rant expression of TAL1 are arrested at the double positive late cortical stage.
of HOX11L2 as a master transcriptional regulator upstream of T-ALL cells with expression of the MLL-ENL fusion gene are characterized
important pathways involved in cell fate determination is supported by an early arrest of differentiation with a gene expression signature that
by its importance during embryonic development.148 In mice, Hox11l2 indicates commitment to the -delta lineage. (Adapted from Ferrando AA,
expression is essential for normal development of the ventral medul- Neuberg DS, Staunton J, etal: Gene expression signatures define novel oncogenic
lary respiratory center.148 Mice deficient in this protein die soon after pathways in T cell acute lymphoblastic leukemia. Cancer Cell 1:75, 2002, and Fer-
birth from respiratory failure that resembles congenital central rando AA, Look AT: Gene expression profiling in T-cell acute lymphoblastic leukemia.
hypoventilation syndrome in humans. Semin Hematol 40:274, 2003, with permission.)
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 941

Other Genes Activated by Translocation HLF and related proteins, no other PAR proteins are expressed in
hematopoietic cells, and the E2AHLF fusion binds DNA as a
Transcription factors are not the only genes activated by translocation homodimer in cells harboring the t(17;19). Similar to E2APBX1,
to the sites of the Ig or TCR genes. In cases of B-precursor ALL car- E2AHLF can transform NIH3T3 fibroblasts, a process that
rying the t(5;14), for example, the IL-3 gene is activated by juxtaposi- requires the HLF leucine zipper domain and the E2A transactivation
tion with the Ig heavy-chain locus.153,154 Similarly, relocation to the domains.182 E2AHLF can also induce lymphoid tumors in trans-
TCRB locus activates expression of the LCK tyrosine kinase genes in genic mice.183
T-ALL cases with the t(1;7).155-157 A major consequence of the activation of E2AHLF in lymphoid
precursors is dysregulation of the mechanisms that control pro-
grammed cell death in lymphoid progenitors. Expression of a
Chimeric Transcription Factor Genes dominant-negative form of E2AHLF in t(17;19)-carrying cell lines
blocks E2AHLF function and results in apoptosis.184 In normal
Formation of chimeric proteins whose functional domains come from pro-B lymphocytes, expression of E2AHLF reversed IL-3-depen-
two normally separate genes represents a second mechanism of aber- dent and p53-induced apoptosis. HLF is the mammalian homologue
rant transcription factor activation, which is more prevalent in of the nematode protein ces-2, which regulates the death of specific
B-precursor ALL. These chromosomal translocations result in the nerve cells in Caenorhabditis elegans.184-186 Ces-2 is necessary for the
production of a chimeric protein by fusing the DNA-binding, dimer- death of the sister cells of a specific pair of serotonergic neurons
ization, and trans-activation regions of discrete genes. This process is during worm development. Ces-2 induces apoptosis by inhibiting the
facilitated by the molecular structure of transcription factors in which expression of ces-1, a prosurvival gene that normally inhibits pro-
discrete coding regions of each gene encode particular functional grammed cell death by antagonizing the activity of the proapoptotic
domains; this molecular structure likely arose for evolutionary factor egl-1. This pathway, which is highly conserved through evolu-
reasons. tion, is disrupted by E2AHLF fusion. Thus, in contrast to the
proapoptotic role of ces-2 in the worm via repression of ces-1, E2A
HLF blocks apoptosis by inducing the expression of SLUG, a ces-1
E2A-PBX1 Fusion Genes in Pre-B Cell Acute homologue normally responsible for protecting hematopoietic pro-
Lymphoblastic Leukemia genitors from DNA-damageinduced apoptosis.187-190
The t(17;19) is seen in fewer than 1% of ALL patients, typically
A well-known example of a chimeric transcription factor with onco- occurs in adolescents, and is associated with disseminated intravascu-
genic potential is the E2A-PBX1 rearrangement, which results from lar coagulation and hypercalcemia at diagnosis. The t(17;19) seems
the t(1;19)(q23;p13) chromosomal translocation present in about to impart unfavorable prognosis because each of seven patients whose
5% of all B-lineage ALLs and in 25% of cases with a pre-B (cyto- blasts expressed E2AHLF died of leukemia despite aggressive
plasmic Ig-positive) phenotype.32,91,158 This translocation fuses the therapy.91 It seems likely that resistance to chemotherapy in these cases
transactivation domain of the E2A transcription factor on chromo- is mediated by the role of E2AHLF in driving the expression of
some 19 to a homeobox gene (PBX1) on chromosome 1, leading to SLUG and inhibiting apoptosis.190
the expression of several forms of hybrid E2A-PBX1 oncoproteins.159-161
PBX1 is related to the Drosophila exd gene, a homeobox gene that
plays a role in lymphocyte development.162,163 The hybrid proteins MLL Fusion Genes
resulting from the t(1;19) retain the amino-terminal trans-activation
domains of E2A (AD1 and AD2) but not the bHLH DNA-binding/ Translocations involving chromosome 11 band q23 occur in approxi-
protein interaction domain.160,161,164 The bHLH domain is replaced by mately 80% of infant ALL cases, 5% of AML cases, and 85% of
the homeobox DNA-binding domain of PBX1, enabling the fusion secondary AML cases that occur in patients treated with topoisom-
protein to function as a chimeric transcription factor.165-167 erase II inhibitors.91 The gene bisected by 11q23 translocations is
The transforming potential of E2APBX1 was first demonstrated designated MLL (also known as TRX1 or ALL-1), which is the human
by the rapid induction of AML in lethally irradiated mice repopulated ortholog of the trithorax Drosophila gene.191-194 The trithorax group
with bone marrow stem cells that had been infected with recombi- family of proteins are positive regulators of homeobox gene expres-
nant retroviruses containing E2APBX1 genes.168 The fusion has also sion and act antagonistically to the polycomb group of proteins.195
been shown to transform NIH-3T3 fibroblasts and induce T-cell Wild-type MLL positively regulates HOX gene expression and is
lymphomas in transgenic mice.169,170 Additional studies have shown required for both primitive and definitive hematopoiesis.196-198 The
that deletion of one of the E2A activation domains diminishes its MLL protein undergoes proteolytic processing by Taspase1, a special-
transforming activity, but deletion of the PBX1 homeodomain has ized protease that cleaves the MLL protein into N-terminal (MLLN)
no effect.170,171 However, the homeodomain and flanking sequences and C-terminal (MLLC) fragments that remain associated through
are required for interactions with other HOX proteins and for optimal intramolecular proteinprotein interaction domains.199-201 MLLN con-
binding of E2APBX1 to specific DNA sequences.172-176 It thus tains several DNA-binding domains, including AT-hook domains
appears that complex interactions between E2APBX1 and other that nonspecifically bind the minor groove of DNA, a methyltrans-
HOX proteins target the fusion protein to specific target genes whose ferase homology region (CxxC domain) that specifically binds
activation is critical to lymphoid cell transformation. The presence unmethylated DNA, four plant homeodomain zinc fingers with an
of the E2APBX1 translocation was originally associated with a poor embedded bromodomain, and a transcriptional repression domain.195
prognosis.31 However, this translocation no longer imparts an adverse The C-terminal fragment MLLC contains a transcriptional activation
prognosis in the setting of modern risk-adjusted protocols for child- domain that recruits the histone acetyltransferase cAMP response
hood ALL.32,177-179 element-binding protein (CBP) and a SET domain that is responsible
for its histone 3 lysine 4 (H3K4) methyltransferase activity.201 Recent
studies have revealed that wild-type MLL is a member of a large
E2AHLF Fusion Genes in Early Pre-B Acute multiprotein complex involved in chromatin modification and
Lymphoblastic Leukemia remodeling, together with histone deacetylases and members of the
Swi/Snf chromatin-remodeling complex.202
The t(17;19) is a rare recurrent chromosomal translocation that fuses MLL fusion oncogenes result from translocations whose break-
the amino-terminal transactivation domains of E2A to the C-terminal points cluster between exons 5 and 11 of MLL, and the resultant
DNA binding and dimerization domains of HLF,180,181 which belongs fusion proteins retain the N-terminal region of MLL, including the
to the PAR subfamily of bZIP transcription factors. Although E2A AT-hook and CxxC domains that bind DNA in a sequence-
HLF can bind DNA either as a homodimer or as a heterodimer with nonspecific manner.203 By contrast, the C-terminal domains that
942 Part VII Hematologic Malignancies

mediate the association of wild-type MLL with its endogenous chro- Both early B- and T-cell ALLs with MLL rearrangements showed a
matin modificationremodeling complex and its H3K4 methyltrans- characteristic upregulation of specific HOX genes, including HOXA9,
ferase activity are invariably lost from oncogenic MLL fusion proteins. HOXA10, and HOXC6 and the HOX gene regulator MEIS1.212-214
Instead, the C-terminus of MLL fusion oncoproteins is provided by These results, together with the demonstration that HOXA9 plays
one of more than 60 different translocation partners, with common important roles in the transformation of hematopoietic precursors by
translocations such as the t(4;11), t(9;11), and t(11;19)(q23;p13.3) MLL fusion oncogenes in murine leukemia models,215,216 emphasize
resulting in the in-frame fusion of MLL to AF4, AF9, and ENL, the central role of HOX gene dysregulation in the pathogenesis of
respectively. The unrelated t(11;19)(q23;p13.1) translocation results MLL-rearranged leukemias. Additionally, as discussed later in this
in fusion of MLL to ELL, the RNA polymerase II elongation chapter, overexpression or activating mutations of the FLT3 receptor
factor.204,205 tyrosine kinase are frequent in MLL-rearranged leukemias,212,213,217,218
Formal proof that MLL fusions play a critical role in the develop- and a model of the multistep pathogenesis of MLL-rearranged ALL
ment of leukemias has come from the generation of murine models is depicted in Fig. 63-4.
of MLL-induced leukemias. Chimeric mice harboring a MLLAF9 Until recently, the precise mechanisms mediating the oncogenic
fusion gene generated by homologous recombination developed leu- activity of MLL fusion oncoproteins was unclear because MLL trans-
kemias with a latency of 4 to 12 months.206 Retroviral transduction location partners have little sequence or functional similarity.
of MLLENL, MLLELL, and MLLCBP fusion genes in hemato- However, recent work has shown that a number of distinct MLL
poietic precursors induces transformation upon transplantation into translocation partners are functionally linked through their associa-
recipient mice.207-209 Similar results were recently obtained with a tion in protein complexes that mediate transcriptional elongation.219
model in which chromosomal translocations involving the Mll locus Oncogenic MLL-fusion proteins associate with at least three such
are induced by directed interchromosomal recombination in mice, a complexes, PAFc, DOT1L, and pTEFb (also known as SEC or AEP).
strategy that reproduces experimentally the initiating events in the PAFc, the polymerase-associated factor complex, regulates RNA poly-
pathogenesis of MLL-rearranged leukemias.210 Interestingly, the intro- merase II and associates with the amino-terminus of both wild-type
duction of MLLAF9 into committed granulocyte-macrophage pro- MLL and MLL fusions.220 The pTEFb complex (CDK9cyclin T)
genitors in the mouse leads to the reactivation of a subset of genes phosphorylates RNA polymerase II; interacts with several MLL
normally expressed only in HSCs and transforms these committed fusion partners, including ENL, ELL, AF4, and AF5; and co-purifies
precursors into AML leukemic stem cells by imparting the properties with several MLL fusion proteins.221,222 The DOT1L complex consists
of self-renewal,211 suggesting that the leukemogenic lesion in MLL- of DOT1L; a histone H3 lysine 79 (H3K79) methyltransferase; and
rearranged leukemia might occur in a committed progenitor rather multiple MLL fusion partners, including AF9, ENL, and AF10.223-228
than in a pluripotent HSC. Analysis of H3K79 methylation status has revealed increased
The analysis of gene expression profiles in MLL-rearranged methylation at genes overexpressed in MLL fusion-driven
B-lineage leukemias has shown that these tumors have a characteristic leukemias, including several direct targets of MLL fusions such as the
gene expression signature that includes the upregulation of several 5 HOXA cluster genes and MEIS1.229-231 Interestingly, whereas the
HOX genes and the expression of numerous myeloid markers.212-214 DOT1L methyltransferase is required for transformation of murine

Activated signaling
molecules (FLT3, others)

Program of
self-renewal
Cytoplasm

Survival or
proliferation HOX genes

Meis 1
Fusion
MLL
Nucleus

Figure 63-4 MULTISTEP PATHOGENESIS OF MLL-REARRANGED ACUTE LYMPHOBLASTIC LEUKE-


MIA (ALL). MLL translocations induce self-renewal in hematopoietic progenitors as a first step in leukemogenesis.
The presence of FLT3 mutations in MLL-rearranged ALLs support activation of FLT3 or other kinases as cooperating
events in this disease. Clinical trials designed to assess the efficacy of FLT3 inhibitors in MLL-rearranged ALL are
being developed. (Adapted from Armstrong SA, Look AT: Molecular genetics of acute lymphoblastic leukemia. J Clin Oncol
23:6306, 2005, with permission.)
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 943

bone marrow progenitors by MLL fusion oncoproteins, DOT1L the inactivation of TEL in adult mice leads to the selective loss of
plays a less prominent role in normal hematopoiesis, thus suggesting HSCs from adult bone marrow, but hematopoiesis is sustained by
a therapeutic window for therapeutic targeting of DOT1L in MLL- committed precursors.249 Most TELAML1 leukemias show loss of
driven leukemias.227,232-235 Indeed, a small molecule inhibitor of the normal TEL allele, suggesting that the leukemogenic effect of
DOT1L has recently been developed and demonstrates promising TEL-AML1 may be mediated in part by loss of function of the TEL
activity against MLL-positive leukemia cells in vitro and in vivo, with gene.250-253
little toxicity in murine models.236 AML1 is the DNA-binding component of the AML1CBF tran-
The presence of MLL rearrangements is associated with dismal scription factor complex disrupted by the t(8;21), t(3;21), and
outcomes despite aggressive chemotherapy in most cases of ALL.23,237- inv(16) in AML, making it the most common target of chromosomal
239
Additionally, bone marrow transplantation (BMT) in first remis- translocations in leukemia. AML1 is a transcription factor that has
sion in these patients is not clearly beneficial and may actually be been shown to be required for the expression of several hematopoietic
associated with a decrease in event-free and overall survival compared genes involved in myeloid and lymphoid development, including
with chemotherapy alone.239,240 However, the recent data implicating PU.1 and IL-3, although it can also act as a transcriptional repressor
DOT1L as a promising therapeutic target in MLL-driven leukemias in some settings.254 Homozygous disruption of the murine AML1
have generated considerable enthusiasm for the development of clini- gene or CBFB gene results in the lack of definitive hematopoiesis,
cal DOT1L inhibitors for the treatment of MLL-rearranged indicating that genes regulated by AML1 are essential for normal
leukemias. hematopoietic development.255,256 Additionally, rare familial muta-
tions in the AML1 DNA-binding domain lead to the familial platelet
disorder and predisposition to myeloid malignancy syndrome.257
CALMAF10 Fusion Gene in T-Cell Acute However, the molecular pathways mediating TELAML1-induced
Lymphoblastic Leukemia leukemogenesis remain incompletely understood.
The presence of the TELAML1 translocation is associated with
The t(10;11)(p13;q14) detected in approximately 3% to 10% of an excellent prognosis, with event-free survival rates of approximately
T-ALL patients and in occasional AML patients and results in the 90% in a variety of studies.91,258,259 However, TELAML1 may not
fusion of CALM, encoding a protein with high homology to the represent an independent predictor of prognosis when age and white
murine clathrin assembly protein ap3, with AF10, a gene identified blood cell count at the time of diagnosis are taken into account in
as an MLL partner in the MLLAF10 fusion resulting from the multivariate analysis.259 Nevertheless, this translocation identifies a
t(10;11)(p13;q23).241 Although the mechanism of action of CALM large subset of children with B-precursor ALL who appear to repre-
AF10 remains incompletely understood, the expression of this fusion sent good candidates for less intensive therapy. The TELAML1
transcript has been associated with early arrest in T-cell development translocation is associated with lower expression of the MDR-1 mul-
and to differentiation into the -lineage in T-ALL.242 Additionally, tidrug resistance gene260 and of genes involved in purine metabo-
recent evidence suggests that aberrant upregulation of HOX gene lism,261 which might account for the particular efficacy of these cases
expression appears to be involved in CALMAF10-mediated leuke- to current combination chemotherapy regimens that rely heavily on
mogenesis, at least in AML cells that carry this translocation.243 Inter- methotrexate and mercaptopurine, agents that inhibit de novo purine
estingly, analysis of a mouse model of CALMAF10-induced AML synthesis.
suggests that the leukemia stem cell in this model has lymphoid
characteristics, and cells from human patients with AML can be
identified that have similar characteristics to the disease-propagating
cell in this animal model.244 Tyrosine Kinase Genes
BCRABL in B-Precursor Acute
TELAML1 (ETV6-RUNX1) Fusion Gene in Early Lymphoblastic Leukemia
Pre-B Acute Lymphoblastic Leukemia
The 22q chromosomal marker, often called the Philadelphia (Ph)
Although most t(12;21) translocations are not detectable by standard chromosome, which arises from the t(9;22)(q34;q11), was originally
cytogenetic analysis, this translocation is detectable by molecular identified in patients with CML, although it is also found in about
techniques in approximately 25% of childhood B-lineage ALL, which 4% of childhood cases and 25% of adult cases of ALL,91 which are
makes this the most common genetic lesion in pediatric ALL.91 The almost always of the B-precursor subtype. The t(9;22) generates a
TELAML1 translocation often arises prenatally and is probably the BCRABL fusion gene, consisting of 5 (upstream) sequences
initiating mutation in ALL, as evidenced by the identification of from BCR and 3 (downstream) sequences of ABL. The t(9;22) break-
identical TELAML1 translocations in identical twins with concor- points on the distal tip of the long arm of chromosome 9 are scattered
dant ALL245 and in retrospectively analyzed neonatal blood specimens over a distance of nearly 200kb within the first intron of the ABL
of children diagnosed with ALL.246 However, TELAML1 is insuffi- proto-oncogene, upstream of the tyrosine kinase domain.262-264 The
cient in causing leukemia because the incidence of detectable TEL breakpoints in the BCR gene on chromosome 22 cluster in two sepa-
AML1 fusions in the blood of normal newborns is about 100-fold rate regions of that gene, known as the major breakpoint cluster
greater than the incidence of leukemia.247 region (M-bcr) or minor breakpoint cluster region (m-bcr). In two-
The molecular mechanisms mediating TELAML1-induced leu- thirds of cases of Ph-positive ALL, the breakpoint in the BCR gene
kemogenesis remain poorly understood. This fusion gene encodes a occurs in the minor breakpoint cluster region (m-bcr), but in all cases
chimeric protein that contains the helixloophelix (HLH) domain of CML and about one third of cases of ALL, the breaks occur in the
of TEL fused to nearly all of AML1 (also known as RUNX1 or major breakpoint cluster region (M-bcr).265 The fusion transcript
CBFA2), including both the transactivation domain and the DNA- more commonly present in ALL (m-bcr) encodes a 190-kd protein
and protein-binding Runt homology domain. Both of these genes (p190), but the transcripts found in CML and some cases of ALL
are found in other leukemia-related translocations, and both are (M-bcr breakpoint) encodes a 210-kd hybrid protein (p210).266-269
essential for normal hematopoiesis. TEL was first identified in the Both types of fusions generate chimeric oncoproteins that are acti-
t(5;12) in chronic myelomonocytic leukemia, where it is fused to the vated as a tyrosine-specific protein kinase, similar to the v-abl
platelet derived growth factor receptor gene (PDGFRB), and is also protein.270-272
fused to ABL, MN1, and EVI1 in AML and to JAK2 in T-ALL.248 The ABL tyrosine kinase is localized both in the nucleus and in
The role of TEL in hematopoiesis has been demonstrated with a the cytoplasm of proliferating cells. It is normally activated by DNA
conditional mouse model in which the absence of TEL in early damage downstream of ATM and appears to promote p53-mediated
development leads to the absence of fetal hematopoiesis. Interestingly, growth arrest.273-276 Mice deficient in ABL develop a wasting syndrome
944 Part VII Hematologic Malignancies

and die soon after birth.277,278 In contrast to the nuclear and cytoplas- NUP214 protein is a component of the nuclear pore complex and
mic distribution of normal ABL, the BCRABL fusion oncoprotein may contribute oligomerization motifs to the NUP214ABL1 fusion
has a cytoplasmic location and shows increased tyrosine kinase activ- oncogene. The NUP214-ABL1 fusion protein has constitutively acti-
ity.279,280 When expressed in murine hematopoietic precursors, both vated ABL1 tyrosine kinase activity, which is inhibited by the BCR
p190 and p210 transform hematopoietic cells in vitro and induce a ABL kinase inhibitor imatinib.311 The therapeutic potential of
syndrome similar to CML in mice.281-284 Transformation by the BCR imatinib or second-generation tyrosine kinase inhibitors for NUP214
ABL oncoprotein involves activation of the RASMAPK (mitogen- ABL1-positive T-cell ALL is of considerable interest.
activated protein kinase) pathway, PI-3 and JUN kinases, c-CBL and
CRKL, JAKSTAT, NFB (nuclear factor kappa-B), Src, and cyclin
D1.285-292 Among these targets, the RAS, JUNkinase, and PI-3 kinase FLT3 in MLL-Rearranged Acute
pathways are involved in the induction of cell proliferation.293 The Lymphoblastic Leukemia
BCRABL oncoprotein affects multiple aspects of cell homeostasis,
including apoptosis, differentiation, and cell adhesion. An important FLT3 encodes a receptor tyrosine kinase that is highly expressed in
cellular effect of BCRABL is the induction of cellular resistance to early hematopoietic precursors, where it plays important functional
DNA damage agents such as cytostatic drugs and irradiation. After roles.312,313 Multiple studies have shown that activating mutations of
DNA damage, BCRABL extends the duration of the G2/M cell FLT3, which lead to constitutive receptor tyrosine kinase activity even
cycle checkpoint and facilitates DNA repair. It also upregulates the in the absence of ligand, are common in leukemic myeloblasts in
antiapoptotic BCLXL gene, contributing to the suppression of apop- patients with AML but are rare in adults with ALL.314-316 However,
totic cell death.294 gene expression studies demonstrated high expression of FLT3 in
The presence of the Philadelphia chromosome has historically most cases of ALL that involve MLL gene rearrangements or hyper-
been associated with an extremely poor prognosis in ALL patients diploidy.212,213,217 Additionally, activating mutations were identified in
despite treatment with intensified chemotherapeutic regimens.295-297 18% of infants with MLL-rearranged ALL,218 in 21% to 24% of
However, these patients have been shown to have particularly good hyperdiploid ALL cases,7,218 and in all three cases of the prothymic
responses to allogeneic BMT in first remission, whether from matched CD117/KIT+ subtype of T-cell ALL in adults who were
sibling or from unrelated donors.298-302 Moreover, the development of examined.317
imatinib mesylate, a pharmacologic tyrosine kinase inhibitor target- In the absence of FLT3 ligand, wild-type FLT3 receptors are inac-
ing the BCRABL oncoprotein, has opened novel therapeutic oppor- tive because of autoinhibition mediated by the juxtamembrane
tunities for the management of Ph-positive ALL. The utility of domain of the receptor. Upon binding of FLT3 ligand, normal FLT3
imatinib as a single agent for BCRABL ALL is limited by the rapid receptors homodimerize, become activated by phosphorylation, and
development of drug resistance.303,304 However, the combination of lead to the activation of signal transduction pathways that promote
imatinib with conventional chemotherapy has led to remarkable proliferation and cell survival.318 Activating mutations of FLT3 found
improvements in outcome for patients with BCRABL ALL, with in leukemias occur in two separate regions of the gene. In-frame
3-year survival rates in children with this disease rivaling those tandem duplications in the juxtamembrane domain lead to loss of
obtained with BMT.305 the autoinhibiton mediated by this domain, with subsequent dimer-
Despite its activity, imatinib resistance remains a barrier to further ization and receptor activation in the absence of FLT3 ligand.319
therapeutic improvements in BCRABL ALL. Resistance most com- Alternatively, point mutations or insertions in the second tyrosine
monly emerges because of point mutations in the kinase domain of kinase domain of the FLT3 receptor lead to autophosphorylation and
BCRABL, and a series of novel BCRABL1 kinase inhibitors have activation of downstream signaling in the absence of FLT3 ligand.6,316,320
been developed that retain activity against many of these mutant Small molecule inhibitors of the FLT3 kinase lead to apoptosis in
oncoproteins.306-308 Although not all BCRABL mutations that confer AML cell lines in vitro and are currently undergoing phase I and II
resistance can be overcome with newer agents, the potential of these testing in adults with AML and MDS.321 These small molecule inhibi-
newer BCRABL inhibitors with broader specificity to further tors are also active against MLL-rearranged ALL cell lines217,322 and
improve outcomes is actively being investigated. Additionally, a novel hold promise as targeted therapies for cases of ALL that rely on aber-
pathway mediating resistance to BCRABL inhibition has recently rant activation of FLT3.
been uncovered in BCRABL ALL lymphoblasts. Upon BCRABL
inhibition, these cells markedly upregulate expression of the BCL6
transcription factor, a well-known oncogene that is often translocated CRLF2 and JAK2 Mutations in B-Precursor
in diffuse large B-cell lymphomas.309 BCL6 then blocks activation of Acute Lymphoblastic Leukemia
the p53 pathway via transcriptional repression of ARF and thus
blocks the therapeutic efficacy of monotherapy with imatinib in The CRLF2 cytokine receptor binds its ligand, thymic stromal lym-
BCRABL ALL cells. Importantly, inhibition of BCL6 activity gener- phopoietin (TSLP), as a heterodimeric complex with the IL-7 recep-
ically or using a peptide inhibitor of BCL6 demonstrated marked tor subunit (IL-7R). TSLPCRLF signaling plays physiologic roles
activity in patient-derived BCRABL ALL cells grown in immuno- during normal B-cell development and in inflammation. Genomic
deficient mice, highlighting the therapeutic relevance of these analyses of B-precursor ALL patient samples revealed recurrent dele-
findings.310 tions within the pseudoautosomal region of Xp22.3/Yp11.3 that
result in overexpression of the entire coding region of CRLF2 under
control of gene regulatory elements of P2RY8, a purinergic receptor
NUP214ABL1 in T-Cell Acute that is expressed at high levels in ALL cells.323-325 In some cases, such
Lymphoblastic Leukemia rearrangements are also accompanied by a Phe232Cys point muta-
tion in CRLF2 that promotes constitutive dimerization and cytokine-
Although the BCRABL1 translocation is rare in T-cell ALL, ampli- independent growth.324,325 CRLF2 rearrangements occur with
fied episomes containing NUP214ABL1 fusion genes have recently especially high frequency in patients with Down syndrome323-325 and
been described in approximately 6% of children and adults have been found to portend a poor prognosis in adults.324 Interest-
with T-cell ALL.311 These episomes appear to arise via a mechanism ingly, aberrant CRLF2 expression very frequently co-occurs with
in which the genomic region of chromosome 9q34, which contains JAK2 activating mutations, and these two genetic lesions collaborate
both the NUP214 and ABL1 genes, is circularized in a manner to induce ligand-independent activation of downstream signal trans-
that leads to the fusion of these two genes. The breakpoint in the duction pathways, suggesting that CRLF2 may act as a scaffold that
ABL1 gene in all of these cases occurs in intron 1, which is the same is required for activation of oncogenic JAK-STAT signaling by JAK2
breakpoint observed in Philadelphia-positive CML and B-precursor mutations in B-precursor ALL.323-325 A number of JAK2 inhibitors
ALL, but the NUP214 breakpoints are variable. The wild-type have been developed for the treatment of myeloproliferative
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 945

syndromes,326 and these findings have thus generated considerable


excitement to the application of such inhibitors to CRLF/JAK2- NOTCH1 in T-Cell Acute Lymphoblastic Leukemia
mutant B-precursor ALL.
The NOTCH1 gene was discovered as a partner gene in a t(7;9)
chromosomal translocation that is found in exceedingly rare cases of
Interleukin-7 Receptor Mutations in T-Cell T-ALL in which NOTCH1 is truncated and placed under the control
Acute Lymphoblastic Leukemia of the TCR locus.332 NOTCH1 plays several critical roles in promot-
ing T-cell development40,333-335 and is highly leukemogenic when
The IL-7R is required for normal T-cell development, with inacti- expressed in murine T-cell precursors.336 Despite the rarity of trans-
vating IL7R mutations leading to severe combined immunodefi- locations involving this gene, a search for activating NOTCH1 muta-
ciency.327 The IL-7R protein can heterodimerize with either IL2R, tions demonstrated that these are present in more than 50% of cases
resulting in the receptor for IL-7, or with CRLF2 to form the receptor of T-ALL, and these occur in all molecular subtypes of T-ALL.8
for thymic stromal lymphopoietin.328,329 Recently, point mutations in NOTCH1 is a transmembrane protein that is proteolytically
the IL-7R have been described in approximately 10% of T-ALL processed during its transit to the cell surface, where it exists as a
cases.53,330,331 These point mutations often introduce a novel cysteine heterodimer consisting of extracellular and transmembrane subunits
residue in the transmembrane domain of the protein that induces (Fig. 63-5). Upon ligand binding, the transmembrane subunit
ligand-independent receptor dimerization and activation of down- undergoes additional proteolytic cleavage within the plasma mem-
stream oncogenic signal transduction pathways, including JAK brane, which leads to the release of its intracellular domain, known
STAT and PI3KAKT. IL-7R mutations occur in approximately 10% as ICN1 (intracellular domain of NOTCH1), into the cytosol. ICN1
of T-ALL cases and are enriched in the TLX3-rearranged and HOXA- subsequently translocates into the nucleus, where it is active as
overexpressing cases, thus suggesting the therapeutic utility of target- a transcription factor. Activating NOTCH1 mutations in T-ALL
ing this signal transduction pathway in T-ALL. can occur as either missense mutations in the heterodimerization
domain, which allow constitutive proteolytic activation of the ICN1
domain,8,337 or as frameshift mutations or stop codons that lead to
Mutated Genes truncation of the PEST domain.8,338 PEST domains are protein
sequences rich in proline (P), glutamate (E), serine (S), and threonine
The activation of cellular proto-oncogenes by point mutation is dif- (T) that regulate the proteasomal degradation of proteins with short
ficult to detect because such lesions lack the cytogenetic abnormalities half-lives, thus these PEST-inactivating mutations impair the degra-
that signal other forms of transforming alterations. Genes of this type dation of ICN1. Mutations in both regions are often found on the
must first be identified in experimental systems so that investigators same allele in cases of T-ALL, and in experimental systems, mutations
know in advance the types of activating point mutations that are in both domains of the same gene indeed are synergistic.8 Recent
likely to occur in human tumors. The prototypic genes of this class work has demonstrated that MYC is an important transcriptional
are NOTCH1 and members of the RAS family, with mutations that target of NOTCH1, and it mediates many of the leukemogenic prop-
affect defined functional domains of the corresponding proteins. erties of MYC in human T-ALL cell lines.339-341

Nicastrin

Receiving cell

Signaling
cell -Secretase

Cleavage 2
Nucleus

NOTCH ICN

DSL MAM1
ICN
CSL
Cleavage 1

Metalloprotease

Figure 63-5 ACTIVATION OF NOTCH SIGNALING VIA EXTRACELLULAR AND INTRACELLULAR PROTEO-
LYTIC CLEAVAGE AND NUCLEAR TRANSLOCATION OF THE INTRACELLULAR NOTCH DOMAIN (ICN).
Interaction with serrate ligand (DSL) stimulates proteolytic cleavage of NOTCH by metalloproteases and -secretase. This
leads to the release of the intracellular ICN domain, which translocates to the nucleus, where it acts as a transcription factor
to regulate gene expression. (Adapted from Armstrong SA, Look AT: Molecular genetics of acute lymphoblastic leukemia. J Clin Oncol
23:6306, 2005, with permission.)
946 Part VII Hematologic Malignancies

The proteolytic activation of the ICN domain of NOTCH1 upon neuroblastoma; such amplification has been linked to an advanced
ligand binding is mediated by -secretase, and -secretase inhibitors stage of disease and a poor prognosis.367 However, the cytogenetic
have previously been developed because of the role of this enzyme in hallmarks of gene amplificationdouble-minute chromatin bodies
the pathogenesis of Alzheimer disease. These inhibitors inhibit the and homogeneously staining regionsare rarely found in karyotypes
growth of many T-ALL cell lines,8,342 and trials of -secretase inhibitors of human leukemia cells, making it unlikely that high-level amplifica-
in patients with T-ALL are currently underway. tion of cellular proto-oncogenes is widespread in ALL. However, as
noted earlier, the NUP214ABL1 fusion oncogene is amplified as a
small, cytogenetically undetectable episome in T-ALL.
RAS Gene Mutations
Human tumor DNAs were initially found to contain activated homo- MYB Duplication in T-Cell Acute
logues of either HRAS or KRAS,343,344 proto-oncogenes that were iden- Lymphoblastic Leukemia
tified on the basis of their homology with viral oncogenes. Gene
transfer methods identified an additional member of the RAS gene
family, called NRAS,345,346 that had not been observed as a component A duplication of the c-MYB gene was recently discovered in cases of
of a transforming retrovirus. Proto-oncogenes of the RAS family T-ALL,368-370a and a chromosomal translocation that juxtaposes c-MYB
(HRAS, KRAS, and NRAS) encode 21-kDa proteins that are associ- to the TCR gene regulatory elements was also identified in a small
ated with the inner surface of the cytoplasmic membrane and are number of patients.369 The discovery of this gene duplication involv-
involved in growth factor receptor signaling.347 The RAS proto- ing a small region of the chromosome was made possible by recent
oncogenes are activated to the status of transforming oncogenes by technologic advances that now allow the high-resolution detection of
somatic mutations that alter the amino acids specified by codons 12, small regions of focal amplifications and deletions. The MYB tran-
13, or 61. Mutated RAS genes lose their intrinsic GTPase activity and scription factor is the cellular counterpart of the avian v-MYB avian
become insensitive to GAP proteins, thus accumulating in their myeloblastosis virus, which causes a rapidly fatal monoblastic leuke-
active, GTP-bound conformation even in the absence of growth mia in chickens and is essential for normal hematopoiesis, including
factor binding to surface receptors. Aberrant RAS-mediated signaling T-cell development. The MYB duplication is mediated by homolo-
contributes to transformation through activation of the PI3K (phos- gous recombination between identical sequence regions that flank the
phoinositide-3-kinase) and MAPK (mitogen-activated protein kinase) gene,370a and its expression appears to be important in preventing
pathways.347 T-lymphoblast differentiation because knock-down of MYB expres-
Activated NRAS genes appear to be preferentially involved in sion in T-ALL cell lines induces T-cell differentiation.368
hematopoietic malignancies. They have been detected in myeloid cell
lines,348,349 in fresh leukemic cell samples from patients with AML or
CML,350-352 and in patients with MDS.353 In ALL, mutations of codons Tumor Suppressor Genes
12, 13, or 61 of NRAS have been found in approximately 10% of
patients, and KRAS mutations have been identified in 5% to 10% Much attention has been focused on tumor suppressors, whose loss
of patients.354,355 RAS mutations are particularly common in cases of of function via deletion or mutational inactivation leads to malignant
MLL-rearranged B-precursor ALL, in which 40% of cases have KRAS transformation. Knudson first proposed that inactivation of both
mutations and an additional 10% have NRAS mutations.355 Addition- alleles of a single locus is needed to initiate the development of reti-
ally, RAS mutations have also been associated with high hyperdip- noblastoma, basing his hypothesis on the observed frequencies of
loidy.356 The presence of RAS mutations does not appear to have hereditary and sporadic forms of this disease.370b Allelic loss of defined
prognostic significance in the setting of contemporary treatment regions of many different chromosomes has been linked to specific
regimens.354 types of human tumors. By analogy with the findings in retinoblas-
toma, a reasonable hypothesis is that each of these regions harbors a
tumor suppressor gene whose product is uniquely involved in the
PTENPI3KAKT Mutations in T-Cell Acute inhibition of cell cycle progression and promotion of terminal dif-
Lymphoblastic Leukemia ferentiation of the normal cells that give rise to these different types
of tumors. Tumor suppressor genes that play an important role in
The PI3KAKT signal transduction pathway, which is negatively ALL include p53 and the p16 locus.
regulated by the PTEN tumor suppressor, induces cellular growth
and proliferation while inhibiting apoptosis and is aberrantly acti-
vated in a range of human cancers.357-359 In T-ALL, recent work has The p53 Tumor Suppressor
identified a very high frequency of mutational activation of oncogenic
signaling through this pathway, most often via deletions or truncating p53, located on chromosome 17, band p13, is mutated or lost
mutations of PTEN, but activating mutations of PI3K and AKT through chromosomal deletion in a wide variety of human tumors,371
genes also occur.360-363 Moreover, deletions of PTEN have been found including colon cancer, lung cancer, breast cancer, and osteosarcoma.
to predict treatment resistance in clinical specimens,53,360 and PTEN Heritable cancer-associated changes of the p53 tumor suppressor gene
inactivation has been shown to induce resistance to inhibition of occur in families with Li-Fraumeni syndrome, an unusual aggregation
MYC and of NOTCH1 in zebrafish and murine models of T-ALL, of sarcomas, brain tumors, leukemias, adrenocortical carcinomas, and
further implicating PTEN loss in T-ALL treatment resistance.362,364,365 premenopausal breast cancers.372-375 p53 encodes a 53-kDa transcrip-
Taken together, these findings thus suggest the potential clinical tion factor that functions as a cell cycle and apoptosis checkpoint reg-
utility of PI3K-AKT pathway inhibitors in high-risk T-ALL, many ulator.371,376-380 The p53 protein is increased by DNA damage, blocks
of which are currently in human clinical trials.366 cell division at G1 to allow DNA repair, and activates apoptosis in
cells that have sustained DNA damage.381-386 The mechanism of p53
activation is triggered by the loss of activity of MDM2 after DNA
Gene Amplification damage (via ATM) or oncogenic stress (via p14/ARF). As a negative
regulator of p53, MDM2 induces the ubiqitination of p53 and its
Gene amplification at the DNA level provides the cell a means to degradation by the proteasome. Hence, when MDM2 activity is abol-
increase expression of critical genes whose products are ordinarily ished, p53 accumulates and certain cell cycle regulatory genes such as
tightly controlled. Clinically important examples of proto-oncogene p21(WAF1/CIP1/SDI1/CAP20) and proapoptotic factor genes such
amplification have been documented in solid tumors of both adults as BAX, PUMA, and NOXA are transcriptionally activated.
and children. The MYCN gene, for example, is amplified from 10- to p53 is also inactivated in a variety of hematopoietic malignancies,
300-fold in tumor cells from about one-third of cases of childhood including B-cell ALL and Burkitt lymphoma, but is mutated or
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 947

deleted in fewer than 3% of pediatric B-precursor or T-cell ALL cases PAX5 and Other B-Cell Developmental
at diagnosis.387-389 It thus appears to play a limited role in the etiology Gene Alterations in B-Precursor Acute
of pediatric leukemia. However, p53 mutations are seen in approxi-
mately 25% of relapsed T-cell ALL cases, suggesting a role for p53 Lymphoblastic Leukemia
inactivation in the development of resistant disease.387,388 In addition,
p53 mutations were detected in three of 10 ALL patients who failed A recent high-resolution genome-wide analysis of B-precursor ALL
on induction therapy or suffered early relapse, further supporting a cases using single nucleotide polymorphism arrays identified copy
role for p53 inactivation in disease progression.390,391 number alterations in a number of genes that play important roles in
B-cell development.418 Genes involved in B-cell development were
found to be altered by deletion, amplification, mutation, or rear-
The Cyclin-Dependent Kinase Inhibitors rangement in 40% of cases of B-precursor ALL. The most common
abnormalities identified were deletions of PAX5, and, upon further
The cyclin-dependent kinase (CDK) inhibitors, which include p15 analysis in other cases, other mechanisms that led to inactivation of
(INK4B/MTS2), p16 (INK4A/MTS1/CDKN2), p18 (INK4C), p19 PAX5 were identified. These included a number of translocations that
(INK4D), p21 (WAF1/CIP1/SDI1/CAP20), p27 (KIP1), and p57 led to fusion proteins that maintained the ability to bind to PAX5
(KIP2)m constitute a family of tumor suppressors that negatively transcriptional targets but lost regulatory ability, thus having domi-
regulate the cell cycle by inhibiting CDK phosphorylation of pRB.392 nant negative activity, and inactivating point mutations that altered
The INK4A locus, located on the short arm of chromosome band the transcriptional activity of PAX5. Deletions were also detected in
9q21, contains two different tumor suppressor genes, p16INK4A the TCF3, EBF1, LEF1, IKZF1, and IKZF3 genes, all of which play
and p14ARF (p19ARF in mice),393,394 each with a distinct promoter important roles in B-cell development.
and first exon but common second and third exons. Despite this
close relationship at the genomic level, p16 and p14 have totally
unrelated amino acid sequences because they use different reading
frames in their common second and third exons.395 A third tumor Ikaros Mutations in High-Risk B-Precursor Acute
suppressor gene, the CDK inhibitor p15INK4B, also resides in this Lymphoblastic Leukemia
region.396,397 p16INK4A and p15INK4B directly inhibit cyclin
DCDK4/6 complexes and interfere with cell cycle progression. Ikaros is a DNA-binding transcription factor that is required for the
Cyclin DCDK4/6 complexes promote entry into S phase through development of all lymphoid lineages, and expression of a dominant
phosphorylation of the retinoblastoma protein, PRB, leading to negative Ikaros mutation in mice was shown to lead to T-cell lym-
the release of transcription factors, such as E2F, that promote entry phomas.419,420 However, the role of Ikaros in human leukemias was
into S phase. By contrast, p14ARF lacks a direct effect on the cell not appreciated until genomic analyses of B-precursor ALL patient
cycle machinery, acting instead to stabilize and upregulate p53 samples revealed Ikaros (IKZF1) deletions in 29% of pediatric
through the inhibition of MDM2.396-400 The role of the INK4a samples.418 Ikaros deletions are strongly associated with BCRABL-
locus in tumorigenesis was substantiated when mice with targeted positive ALL, and these are characteristically acquired at transforma-
disruption of exon 2 in p16 developed tumors (primarily lymphomas tion of CML to ALL (lymphoid blast crisis).421 Ikaros deletions
and fibrosarcomas) whose induction was enhanced by the topical predict a very high risk of treatment failure that appears to be inde-
application of carcinogens and ultraviolet light.401,402 The phenotype pendent of BCRABL, with patients with Ikaros-deleted, BCRABL
of the p16/p19/ mice could be duplicated by selective mutation of negative ALL faring similarly poorly to those with BCRABL-
the p19ARF gene (by targeting the first exon),393 although mice defi- positive ALL.422 Interestingly, recent work has implicated mutational
cient in p16INK4A with intact p19ARF also showed increased sus- activation of CRLF2-JAK2 signaling in Ikaros-deleted, BCRABL-
ceptibility to cancer.401,402 negative ALL.423 Given the remarkable clinical activity of imatinib in
The short arm of chromosome 9 is the most frequent target of high-risk BCRABL ALL, these findings thus suggest the need for
chromosomal alterations in human cancer. In particular, the human clinical trials of small molecule inhibitors of JAK-STAT signaling in
leukemias and lymphomas show a high frequency of 9p21 deletions this high-risk subtype of B-precursor ALL.
involving both the p16INK4A/p14ARF and the p15INK4B loci. Epi-
genetic silencing of these tumor suppressor genes through hyper-
methylation of their promoter sequences represents an alternative
mechanism of gene inactivation. Although p16INK4A/p14ARF and Inactivation of LEF1 in T-Cell Acute
p15INK4B are homozygously deleted in 20% to 30% of B-precursor Lymphoblastic Leukemia
ALL cases and in 70% to 80% of T-cell ALL cases, epigenetic silenc-
ing of the p15INK4B promoter has been observed in 44% of primary The LEF1 transcription factor, a member of the LEF/TCF (lymphoid
B-lineage ALLs.403-415 enhancer binding factor/T-cell specific transcription factor) family of
DNA-binding transcription factors, is best known for its role as a
positive mediator -catenin transcriptional activity, a well-established
FBW7 in T-Cell Acute Lymphoblastic Leukemia oncogene.424 Recent genomic analyses have identified mono- and
biallelic deletions and truncating mutations of LEF1 in 18% of
FBW7 is an E3 ubiquitin ligase that targets the transcriptionally primary T-ALL patient samples, unexpectedly implicating LEF1 as a
active intracellular form of NOTCH (ICN), MYC, and cyclin E for T-ALL tumor suppressor.425 LEF1 inactivation is associated with a
degradation, and this gene is inactivated by mutation or deletion in very young age at diagnosis, T-cell differentiation arrest at an early
approximately 10% of T-ALL cases.416 In T-ALL cell lines, FBW7 cortical stage of T-cell development, and activating NOTCH1 muta-
mutation or homozygous deletion leads to resistance of NOTCH tions. Moreover, T-ALL cases with LEF1 inactivation are character-
pathway inhibition by -secretase inhibitor therapy, likely because ized by the highest expression of MYC mRNA of any T-ALL subtype,
intracellular NOTCH protein levels remain high in the absence of a surprising finding given that LEF/TCF transcription factors trans-
FBW7-mediated degradation despite inhibition of -secretase activ- activate MYC expression when bound to -catenin.426,427 In addition
ity. Additionally, tumor-derived FBW7 mutations maintain their to their well-established functions as transcriptional activators, LEF/
ability to bind MYC but do not lead to its degradation, and these TCF transcription factors can also act as transcriptional repressors
may act as dominant negative mutants that protect MYC from deg- when bound to Groucho/TLE family members in the absence of
radation.416 More recent work has shown that FBW7 also targets the -catenin activation.428 Although the precise mechanisms responsible
antiapoptotic protein MCL1 for proteasomal degradation in T-ALL, for the pathogenic effect of LEF1 inactivation in T-ALL have not yet
providing an additional oncogenic consequence resulting from inac- been established, these findings thus suggest the intriguing possibility
tivation of the FBW7 tumor suppressor.417 that LEF1 may actively repress MYC in T-ALL cells lacking -catenin
948 Part VII Hematologic Malignancies

activation, and that LEF1 inactivation in this context may promote Conversely, hypodiploidy (<45 chromosomes) carries an extremely
maximal MYC overexpression downstream of other oncogenic lesions poor prognosis.91,258,446 Overall, adults with ALL have significantly
that drive MYC overexpression, such as NOTCH1 mutations. fewer numeric chromosomal abnormalities than do children; however,
abnormalities of ploidy do also appear to have prognostic significance
in adults because one large series demonstrated a favorable prognostic
BCL11B Inactivation in T-Cell Acute impact of hyperdiploidy, but adults with hypodiploidy had extremely
Lymphoblastic Leukemia poor outcomes.33

The BCL11B transcription factor is required for normal T-cell devel-


opment. In murine T-cell progenitors, inactivation of BCL11B leads CHILDHOOD ACUTE LYMPHOBLASTIC LEUKEMIA:
to developmental arrest at DN stages, acquisition of NK-like features, A MODEL FOR GENE-BASED RISK ASSESSMENT
and aberrant self-renewal activity.429-432 Monoallelic BCL11B deletions
or point mutations have recently been identified in 9% to 16% of Many of the chromosomal and molecular genetic abnormalities in
primary T-ALL patient samples.149,433 The point mutations identified the leukemic blasts of patients with B-precursor ALL are important
typically occur within the DNA-binding domains of BCL11B and predictors of response to currently available chemotherapy. Lympho-
are predicted to disrupt their ability to bind DNA. Moreover, previ- blasts from each new case of childhood ALL should be examined for
ous work in murine models has shown that BCL11B suppresses molecular prognostic markers, including leukemia cell ploidy, MLL
T-lymphoblastic malignancies induced by p53 haploinsufficiency, gene rearrangements, and the presence of BCRABL and TELAML1
radiation, or the BCRABL oncogene,434,435 and BCL11B inactivation fusion transcripts. Additional clinical criteria that have prognostic
is a particularly common cooperating lesion in murine T-ALL significance, such as the age of the patient, the peripheral blood white
induced by the TLX1 oncogene or by ATM deficiency.149,436 In human blood cell count at diagnosis, and the presence of CNS involvement,
T-ALL, recurrent cryptic t(5;14)(q35;q32) translocations juxtaposing are used by modern pediatric ALL treatment regimens to further
BCL11B and TLX3 have been described, which result in BCL11B adjust the intensity of therapy to an individual patients risk of
gene regulatory elements driving overexpression of TLX3.103,437,438 relapse. It is also worth noting that B-precursor ALL in patients
These translocations were long thought to be pathogenic because of younger than 1 year of age is associated with an extremely poor
the resultant overexpression of the TLX3 oncogene. However, these prognosis secondary to an exceedingly high rate of relapse, which is
recent findings indicate that both BCL11B inactivation and TLX at least partly attributable to the very high frequency of MLL trans-
oncogene overexpression are important pathogenic consequences of locations in this population, and these patients are typically treated
this translocation, thus representing two oncogenic events from a with unique treatment regimens designed specifically for infant ALL.
single genomic lesion. In T-ALL, the paucity of clinically useful prognostic markers has
greatly hindered progress toward personalized therapy, but the recent
identification of differentiation arrest at the earliest stages of T-cell
PHF6 Mutations in T-Cell Acute development as a marker of very poor prognosis,50,51 together with the
Lymphoblastic Leukemia identification of mutations and gene expression signatures that closely
resemble those of AML and HSCs,52,53 has generated considerable
T-ALL has a conspicuous male predominance, suggesting the poten- excitement for the application of intensified, AML-like treatment
tial involvement of tumor suppressors on the X-chromosome. Tar- regimens to this high-risk subset of T-ALL. Finally, response to treat-
geted mutational analysis of the X chromosome revealed one such ment has independent prognostic significance. Induction failure,
candidate tumor suppressor, PHF6, which encodes a nucleolar often defined in pediatric protocols as any morphologic evidence of
protein of unknown function whose germline mutation leads to the leukemia at the end of the initial month of induction chemotherapy,
Borjeson-Forssman-Lehmann syndrome (OMIM 301900), which is represents a very poor prognostic factor and is an indication for BMT
associated with severe mental retardation and facial dysmorphisms after remission is achieved in most centers. Additionally, sensitive
but is not known to be associated with T-ALL. Somatic PHF6 muta- flow cytometric and polymerase chain reactionbased methodologies
tions were identified in 16% of pediatric and in 38% of adult T-ALL for the detection of minimal residual disease (MRD) are now avail-
patient samples, were most often nonsense or frameshift mutations, able, and the level of MRD after induction chemotherapy has recently
and were associated with overexpression of the TLX1 or TLX3 onco- been shown to provide an additional means of identifying patients at
genic transcription factors.439 The pathogenic consequences of PHF6 low or high risk of relapse.447,448 With the availability of these prog-
inactivation in T-ALL are presently unknown but are a matter of nostic indicators, there remains little justification for uniform treat-
active investigation. ment of newly diagnosed cases of ALL. Rather, modern trials
emphasize risk-based therapy to reduce toxicity in patients likely to
become long-term responders and to intensify therapy for those at
Abnormalities of Leukemia Cell Ploidy high risk of relapse.
A risk classification scheme based on a patients clinical features
Abnormalities of chromosome number, which generally occur in the and on the genetic features of leukemic blasts has been proposed for
absence of specific chromosomal translocations, have important prog- patients with B-precursor or T-cell ALL who are 1 year of age or older
nostic implications in childhood ALL. Found in 25% to 30% of (Table 63-2). Patients with TELAML1 positivity or with simultane-
childhood ALL cases, hyperdiploidy of more than 50 chromosomes ous trisomies of chromosomes 4, 10, and 17 constitute the lower risk
in the leukemic clone is one of the most powerful means of identify- group and are candidates for reduced-intensity treatment regimens.
ing patients with a very good prognosis.91,258 In particular, trisomies The intermediate-risk group includes patients between the ages of 1
of chromosomes 4, 10, and 17 impart a particularly good prognosis, and 9 years with standard-risk age and leukocyte count, as defined
and hyperdiploidy in the absence of these trisomies is less of a favor- by criteria of the National Cancer Institute, and whose leukemic
able prognostic factor.440 As mentioned previously, activating muta- lymphoblasts lack prognostically important genetic features.
tions of FLT3, a receptor tyrosine kinase, are common in these The high-risk group includes patients who meet any of the following
patients. Patients with hyperdiploidy typically present with favorable criteria: age 10 years or older, high leukocyte counts at diagnosis,
prognostic indicators, such as age between 2 and 10 years, a low white a T-cell immunophenotype, or a slow early response to therapy.
blood cell count, and an early pre-B immunophenotype, and can Patients in the very high-risk group, defined by extreme hypodip-
expect cure rates that approach 90%.441-443 The mechanism accounting loidy, MLL gene rearrangements, the BCRABL translocation, high
for the favorable outcome of patients with hyperdiploid ALL MRD at the end of induction chemotherapy, or T-ALL blasts harbor-
remains elusive but may reflect an increased sensitivity to antimetabo- ing the high-risk early T-cell progenitor (ETP) or absence of biallelic
lite therapy444 and a greater propensity to undergo apoptosis.445 TCR deletion (ABD) markers are eligible for BMT in first remis-
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 949

Table 63-2 Clinical Risk Assignment in Childhood Acute are assigned to treatment according to their initial clinical features
Lymphoblastic Leukemia and, increasingly, the genetic and biologic properties of their leuke-
mic cells. We are now in a position to view ALL as a group of het-
Risk Group Features Recommended Therapy erogeneous diseases defined by discrete molecular lesions. As these
lesions have been systematically analyzed in larger numbers of
Low risk Simultaneous trisomies of Less intensive
patients, it has been possible to devise new classification schemes for
chromosomes 4, 10, 17 antimetabolite-based
ALL that reflect prognosis with exquisite precision. The development
TELAML1 fusion chemotherapy
of new drugs based on the molecular biology of ALL, whose promise
Intermediate Standard-risk age or Intermediate is highlighted by the early success of imatinib in BCRABLpositive
risk leukocyte count without antimetabolite-based ALL, is clearly a priority for the future and will likely take the form
other genetic risk features chemotherapy of compounds developed to specifically interfere with oncoproteins
High risk Presence of any of the Intensified multiagent expressed by each patients leukemic blasts. Additionally, the discov-
following: chemotherapy with ery that several kinases play important roles in ALL pathogenesis has
Age 10 years or older cranial radiation provided new opportunities for targeted drug development. The
T-cell immunophenotype opportunity is now at hand to improve therapy through randomized
WBC count >50,000/mm3 trials coordinated on a nationwide or even worldwide scale that focus
CNS leukemia on key subsets of acute leukemia patients whose lymphoblasts harbor
specific genetic abnormalities.
Very high BCRABL translocation* Allogeneic bone
risk MLL translocations marrow
Hypodiploidy <45 transplantation in
chromosomes first remission SUGGESTED READINGS
Induction failure or Comprehensive Reviews
elevated MRD at the end
of induction chemotherapy Armstrong SA, Look AT: Molecular genetics of acute lymphoblastic leukemia.
T-ALLs with early T-cell J Clin Oncol 23:6306, 2005.
progenitor (ETP) or Bernt KM, Armstrong SA: Targeting epigenetic programs in MLL-rearranged
absence of biallelic TCR leukemias. Hematology Am Soc Hematol Educ Program 2011:354,
deletion (ABD) markers 2011.
Pui CH, Robison LL, Look AT: Acute lymphoblastic leukaemia. Lancet
ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia;
CNS, central nervous system; MRD, minimal residual disease; TCR, T-cell
371:1030, 2008.
receptor; WBC, white blood cell.
*Recent data also indicate excellent 3-year survival rates with the use of
imatinib, a targeted BCR-ABL inhibitor, in combination with conventional Specific Genetic Lesions in Acute
chemotherapy. Lymphoblastic Leukemia

Current evidence suggests that bone marrow transplantation in first remission
may not improve outcomes for children with MLL-rearranged ALL. Bernt KM, Zhu N, Sinha AU, et al: MLL-rearranged leukemia is

Although MLL translocations are almost always associated with very poor
outcomes, patients with T-cell ALL who have the t(11;19) MLL-ENL dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell
translocation appear to have excellent outcomes with conventional 20:66, 2011.
chemotherapy. De Keersmaecker K, Real PJ, Gatta GD, et al: The TLX1 oncogene drives
aneuploidy in T cell transformation. Nat Med 16:1321, 2010.
Gutierrez A, Sanda T, Grebliunaite R, et al: High frequency of PTEN, PI3K,
and AKT abnormalities in T-cell acute lymphoblastic leukemia. Blood
114:647, 2009.
Gutierrez A, Sanda T, Ma W, et al: Inactivation of LEF1 in T-cell acute
sion. It is worth noting that the evidence supporting a
lymphoblastic leukemia. Blood 115:2845, 2010.
benefit for BMT in first remission is especially strong in the presence
Mullighan CG, Collins-Underwood JR, Phillips LA, et al: Rearrangement of
of the BCRABL translocation, although its benefit in MLL-
CRLF2 in B-progenitor- and Down syndrome-associated acute lympho-
rearranged ALL remains unclear. Moreover, recent data in the setting
blastic leukemia. Nat Genet 41:1243, 2009.
of BCRABL ALL indicate that the combination of imatinib (a
Mullighan CG, Goorha S, Radtke I, et al: Genome-wide analysis of
BCRABL inhibitor) with conventional chemotherapy leads to
genetic alterations in acute lymphoblastic leukaemia. Nature 446:758,
excellent short-term (3-year) survival rates, although longer follow-up
2007.
will be required to ensure that long-term outcomes are comparable.
Weng AP, Ferrando AA, Lee W, et al: Activating mutations of NOTCH1
Among several exceptions to this risk stratification criteria are T-ALL
in human T cell acute lymphoblastic leukemia. Science 306:269,
patients with the t(11;19) and expression of MLL-ENL, who
2004.
have a favorable prognosis,17,449 and infants younger than 1 year of
Yoda A, Yoda Y, Chiaretti S, et al: Functional screening identifies CRLF2 in
age, in whom separate risk stratification criteria have been
precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A
devised. The incorporation of highly predictive risk stratification
107:252, 2010.
methods into clinical trials, together with the development of
Zhang J, Ding L, Holmfeldt L, et al: The genetic basis of early T-cell precursor
novel molecularly tailored therapies, should eliminate the still signifi-
acute lymphoblastic leukaemia. Nature 481:157, 2012.
cant risk of relapse in patients with newly diagnosed ALL, ensuring
permanent cure without the hazard of acute or chronic
complications. Clinical Implications of Acute Lymphoblastic
Leukemia Genetics
FUTURE DIRECTIONS Armstrong SA, Kung AL, Mabon ME, et al: Inhibition of FLT3 in MLL:
Validation of a therapeutic target identified by gene expression based clas-
The immediate applications of the emerging molecular information sification. Cancer Cell 3:173, 2003.
include a redefinition of risk classification schemes to emphasize the Coustan-Smith E, Mullighan CG, Onciu M, et al: Early T-cell precursor
roles of somatically acquired genetic abnormalities that carry a defined leukaemia: A subtype of very high-risk acute lymphoblastic leukaemia.
prognosis and likelihood of therapeutic failure. Currently, patients Lancet Oncol 10:147, 2009.
950 Part VII Hematologic Malignancies

Gutierrez A, Dahlberg SE, Neuberg DS, et al: Absence of biallelic TCRgamma Schultz KR, Bowman WP, Aledo A, et al: Improved early event-free survival
deletion predicts early treatment failure in pediatric T-cell acute lympho- with imatinib in Philadelphia chromosome-positive acute lymphoblastic
blastic leukemia. J Clin Oncol 28:3816, 2010. leukemia: A childrens oncology group study. J Clin Oncol 27:5175, 2009.
Mullighan CG, Su X, Zhang J, et al: Deletion of IKZF1 and prognosis in
acute lymphoblastic leukemia. N Engl J Med 360:470, 2009. For complete list of references log on to www.expertconsult.com.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 950.e1

Burkitts lymphoma and B-cell acute lymphoblastic leukemia. J Clin


REFERENCES Oncol 9:133, 1991.
1. Market E, Papavasiliou FN: V(D)J recombination and the evolution of 27. Reiter A, Schrappe M, Ludwig WD, et al: Favorable outcome of B-cell
the adaptive immune system. PLoS Biol 1:E16, 2003. acute lymphoblastic leukemia in childhood: A report of three consecu-
2. Arnold A, Cossman J, Bakhshi A, et al: Immunoglobulin-gene rear- tive studies of the BFM group. Blood 80:2471, 1992.
rangements as unique clonal markers in human lymphoid neoplasms. 28. Cairo MS, Sposto R, Perkins SL, et al: Burkitts and Burkitt-like lym-
N Engl J Med 309:1593, 1983. phoma in children and adolescents: A review of the Childrens Cancer
3. Rabbitts TH: Chromosomal translocations in human cancer. Nature Group experience. Br J Haematol 120:660, 2003.
372:143, 1994. 29. Thomas DA, Faderl S, OBrien S, et al: Chemoimmunotherapy with
4. Cleary ML: Oncogenic conversion of transcription factors by chromo- hyper-CVAD plus rituximab for the treatment of adult Burkitt and
somal translocations. Cell 66:619, 1991. Burkitt-type lymphoma or acute lymphoblastic leukemia. Cancer
5. Look AT: Oncogenic transcription factors in the human acute leuke- 106:1569, 2006.
mias. Science 278:1059, 1997. 30. Pui CH, Behm FG, Crist WM: Clinical and biologic relevance of
6. Armstrong SA, Kung AL, Mabon ME, et al: Inhibition of FLT3 in immunologic marker studies in childhood acute lymphoblastic leuke-
MLL: Validation of a therapeutic target identified by gene expression mia. Blood 82:343, 1993.
based classification. Cancer Cell 3:173, 2003. 31. Crist WM, Carroll AJ, Shuster JJ, et al: Poor prognosis of children with
7. Armstrong SA, Mabon ME, Silverman LB, et al: FLT3 mutations in pre-B acute lymphoblastic leukemia is associated with the t(1;19)
childhood acute lymphoblastic leukemia. Blood 103:3544, 2004. (q23;p13): A Pediatric Oncology Group study. Blood 76:117, 1990.
8. Weng AP, Ferrando AA, Lee W, et al: Activating mutations of NOTCH1 32. Raimondi SC, Behm FG, Roberson PK, et al: Cytogenetics of pre-B-cell
in human T cell acute lymphoblastic leukemia. Science 306:269, 2004. acute lymphoblastic leukemia with emphasis on prognostic implica-
9. Kohlmann A, Schoch C, Schnittger S, et al: Pediatric acute lympho- tions of the t(1;19). J Clin Oncol 8:1380, 1990.
blastic leukemia (ALL) gene expression signatures classify an indepen- 33. Secker-Walker LM, Prentice HG, Durrant J, et al: Cytogenetics adds
dent cohort of adult ALL patients. Leukemia 18:63, 2004. independent prognostic information in adults with acute lymphoblastic
10. Armstrong SA, Look AT: Molecular genetics of acute lymphoblastic leukaemia on MRC trial UKALL XA: MRC Adult Leukaemia Working
leukemia. J Clin Oncol 23:6306, 2005. Party. Br J Haematol 96:601, 1997.
11. Nowell PC, Hungerford DA: Chromosome studies in human leukemia. 34. Korsmeyer SJ, Arnold A, Bakhshi A, et al: Immunoglobulin gene rear-
II: Chronic granulocytic leukemia. J Natl Cancer Inst 27:1013, 1961. rangement and cell surface antigen expression in acute lymphocytic
12. Waldmann TA, Davis MM, Bongiovanni KF, et al: Rearrangements of leukemias of T cell and B cell precursor origins. J Clin Invest 71:301,
genes for the antigen receptor on T cells as markers of lineage and 1983.
clonality in human lymphoid neoplasms. N Engl J Med 313:776, 1985. 35. Korsmeyer SJ, Hieter PA, Ravetch JV, et al: Developmental hierarchy
13. Dow LW, Martin P, Moohr J, et al: Evidence for clonal development of immunoglobulin gene rearrangements in human leukemic pre-B-
of childhood acute lymphoblastic leukemia. Blood 66:902, 1985. cells. Proc Natl Acad Sci U S A 78:7096, 1981.
14. Pui CH, Raskind WH, Kitchingman GR, et al: Clonal analysis of 36. Kitchingman GR, Rovigatti U, Mauer AM, et al: Rearrangement of
childhood acute lymphoblastic leukemia with cytogenetically indepen- immunoglobulin heavy chain genes in T cell acute lymphoblastic leu-
dent cell populations. J Clin Invest 83:1971, 1989. kemia. Blood 65:725, 1985.
15. Kersey J, Nesbit M, Hallgren H, et al: Evidence for origin of certain 37. Hatano M, Roberts CW, Minden M, et al: Deregulation of a homeobox
childhood acute lymphoblastic leukemias and lymphomas in thymus- gene, HOX11, by the t(10;14) in T cell leukemia. Science 253:79, 1991.
derived lymphocytes. Cancer 36:1348, 1975. 38. Ford AM, Molgaard HV, Greaves MF, et al: Immunoglobulin gene
16. Sen L, Borella L: Clinical importance of lymphoblasts with T markers organisation and expression in haemopoietic stem cell leukaemia. Embo
in childhood acute leukemia. N Engl J Med 292:828, 1975. J 2:997, 1983.
17. Ferrando AA, Neuberg DS, Staunton J, et al: Gene expression signa- 39. Reinherz EL, Schlossman SF: Current concepts in immunology: Regu-
tures define novel oncogenic pathways in T cell acute lymphoblastic lation of the immune responseinducer and suppressor T-lymphocyte
leukemia. Cancer Cell 1:75, 2002. subsets in human beings. N Engl J Med 303:370, 1980.
18. Greaves MF: Differentiation-linked leukemogenesis in lymphocytes. 40. Rothenberg EV, Taghon T: Molecular genetics of T cell development.
Science 234:697, 1986. Annu Rev Immunol 23:601, 2005.
19. Bonnet D, Dick JE: Human acute myeloid leukemia is organized as a 41. Staal FJ, Weerkamp F, Langerak AW, et al: Transcriptional control of t
hierarchy that originates from a primitive hematopoietic cell. Nat Med lymphocyte differentiation. Stem Cells 19:165, 2001.
3:730, 1997. 42. Reinherz EL, Nadler LM, Sallan SE, et al: Subset derivation of T-cell
20. Hurwitz CA, Loken MR, Graham ML, et al: Asynchronous antigen acute lymphoblastic leukemia in man. J Clin Invest 64:392, 1979.
expression in B lineage acute lymphoblastic leukemia. Blood 72:299, 43. Nadler LM, Reinherz EL, Weinstein HJ, et al: Heterogeneity of T-cell
1988. lymphoblastic malignancies. Blood 55:806, 1980.
21. Bennett JM, Catovsky D, Daniel MT, et al: The morphological clas- 44. Bernard A, Boumsell L, Reinherz EL, et al: Cell surface characterization
sification of acute lymphoblastic leukaemia: Concordance among of malignant T cells from lymphoblastic lymphoma using monoclonal
observers and clinical correlations. Br J Haematol 47:553, 1981. antibodies: Evidence for phenotypic differences between malignant T
22. Pui CH, Crist WM, Look AT: Biology and clinical significance of cells from patients with acute lymphoblastic leukemia and lymphoblas-
cytogenetic abnormalities in childhood acute lymphoblastic leukemia. tic lymphoma. Blood 57:1105, 1981.
Blood 76:1449, 1990. 45. Roper M, Crist WM, Metzgar R, et al: Monoclonal antibody charac-
23. Pui CH: Childhood leukemias. N Engl J Med 332:1618, 1995. terization of surface antigens in childhood T-cell lymphoid malignan-
24. Murphy SB, Bowman WP, Abromowitch M, et al: Results of treatment cies. Blood 61:830, 1983.
of advanced-stage Burkitts lymphoma and B cell (SIg+) acute lympho- 46. Uckun FM, Steinherz PG, Sather H, et al: CD2 antigen expression on
blastic leukemia with high-dose fractionated cyclophosphamide and leukemic cells as a predictor of event-free survival after chemotherapy
coordinated high-dose methotrexate and cytarabine. J Clin Oncol for T-lineage acute lymphoblastic leukemia: A Childrens Cancer Group
4:1732, 1986. study. Blood 88:4288, 1996.
25. Patte C, Philip T, Rodary C, et al: High survival rate in advanced-stage 47. Czuczman MS, Dodge RK, Stewart CC, et al: Value of immunophe-
B-cell lymphomas and leukemias without CNS involvement with a notype in intensively treated adult acute lymphoblastic leukemia:
short intensive polychemotherapy: Results from the French Pediatric Cancer and leukemia Group B study 8364. Blood 93:3931, 1999.
Oncology Society of a randomized trial of 216 children. J Clin Oncol 48. Goldberg JM, Silverman LB, Levy DE, et al: Childhood T-cell acute
9:123, 1991. lymphoblastic leukemia: The Dana-Farber Cancer Institute acute lym-
26. Schwenn MR, Blattner SR, Lynch E, et al: HiC-COM: A 2-month phoblastic leukemia consortium experience. J Clin Oncol 21:3616,
intensive chemotherapy regimen for children with stage III and IV 2003.
950.e2 Part VII Hematologic Malignancies

49. Pullen J, Shuster JJ, Link M, et al: Significance of commonly used three Burkitt lymphoma lines with t(2;8) translocations. EMBO J
prognostic factors differs for children with T cell acute lymphocytic 3:2951, 1984.
leukemia (ALL), as compared to those with B-precursor ALL: A Pedi- 72. Taub R, Kelly K, Battey J, et al: A novel alteration in the structure of
atric Oncology Group (POG) study. Leukemia 13:1696, 1999. an activated c-myc gene in a variant t(2;8) Burkitt lymphoma. Cell
50. Gutierrez A, Dahlberg SE, Neuberg DS, et al: Absence of biallelic 37:511, 1984.
TCRgamma deletion predicts early treatment failure in pediatric T-cell 73. Bhatia K, Huppi K, Spangler G, et al: Point mutations in the c-Myc
acute lymphoblastic leukemia. J Clin Oncol 28:3816, 2010. transactivation domain are common in Burkitts lymphoma and mouse
51. Coustan-Smith E, Mullighan CG, Onciu M, et al: Early T-cell precur- plasmacytomas. Nat Genet 5:56, 1993.
sor leukaemia: A subtype of very high-risk acute lymphoblastic leukae- 74. Albert T, Urlbauer B, Kohlhuber F, et al: Ongoing mutations in the
mia. Lancet Oncol 10:147, 2009. N-terminal domain of c-Myc affect transactivation in Burkitts lym-
52. Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, et al: ETV6 phoma cell lines. Oncogene 9:759, 1994.
mutations in early immature human T cell leukemias. J Exp Med 75. Yano T, Sander CA, Clark HM, et al: Clustered mutations in the second
208:2571, 2011. exon of the MYC gene in sporadic Burkitts lymphoma. Oncogene
53. Zhang J, Ding L, Holmfeldt L, et al: The genetic basis of early T-cell 8:2741, 1993.
precursor acute lymphoblastic leukaemia. Nature 481:157, 2012. 76. Sears RC: The life cycle of C-myc: From synthesis to degradation. Cell
54. Smith LJ, Curtis JE, Messner HA, et al: Lineage infidelity in acute Cycle 3:1133, 2004.
leukemia. Blood 61:1138, 1983. 77. Gregory MA, Hann SR: c-Myc proteolysis by the ubiquitin-proteasome
55. Mirro J, Zipf TF, Pui CH, et al: Acute mixed lineage leukemia: Clini- pathway: Stabilization of c-Myc in Burkitts lymphoma cells. Mol Cell
copathologic correlations and prognostic significance. Blood 66:1115, Biol 20:2423, 2000.
1985. 78. Salghetti SE, Kim SY, Tansey WP: Destruction of Myc by ubiquitin-
56. Greaves MF, Chan LC, Furley AJ, et al: Lineage promiscuity in hemo- mediated proteolysis: Cancer-associated and transforming mutations
poietic differentiation and leukemia. Blood 67:1, 1986. stabilize Myc. Embo J 18:717, 1999.
57. Pui CH, Rubnitz JE, Hancock ML, et al: Reappraisal of the clinical 79. Bahram F, von der Lehr N, Cetinkaya C, et al: c-Myc hot spot muta-
and biologic significance of myeloid-associated antigen expression in tions in lymphomas result in inefficient ubiquitination and decreased
childhood acute lymphoblastic leukemia. J Clin Oncol 16:3768, 1998. proteasome-mediated turnover. Blood 95:2104, 2000.
58. Pui CH, Behm FG, Singh B, et al: Myeloid-associated antigen expres- 80. Hemann MT, Bric A, Teruya-Feldstein J, et al: Evasion of the p53
sion lacks prognostic value in childhood acute lymphoblastic leukemia tumour surveillance network by tumour-derived MYC mutants. Nature
treated with intensive multiagent chemotherapy. Blood 75:198, 1990. 436:807, 2005.
59. Uckun FM, Sather HN, Gaynon PS, et al: Clinical features and treat- 81. Dang CV, ODonnell KA, Zeller KI, et al: The c-Myc target gene
ment outcome of children with myeloid antigen positive acute lympho- network. Semin Cancer Biol 16:253, 2006.
blastic leukemia: A report from the Childrens Cancer Group. Blood 82. Coller HA, Grandori C, Tamayo P, et al: Expression analysis with oli-
90:28, 1997. gonucleotide microarrays reveals that MYC regulates genes involved in
60. Cross AH, Goorha RM, Nuss R, et al: Acute myeloid leukemia with growth, cell cycle, signaling, and adhesion. Proc Natl Acad Sci U S A
T-lymphoid features: A distinct biologic and clinical entity. Blood 97:3260, 2000.
72:579, 1988. 83. Grandori C, Mac J, Siebelt F, et al: Myc-Max heterodimers activate a
61. Casasnovas RO, Slimane FK, Garand R, et al: Immunological classifica- DEAD box gene and interact with multiple E box-related sites in vivo.
tion of acute myeloblastic leukemias: Relevance to patient outcome. EMBO J 15:4344, 1996.
Leukemia 17:515, 2003. 84. Ayer DE, Kretzner L, Eisenman RN: Mad: A heterodimeric partner
62. Williams DL, Look AT, Melvin SL, et al: New chromosomal transloca- for Max that antagonizes Myc transcriptional activity. Cell 72:211,
tions correlate with specific immunophenotypes of childhood acute 1993.
lymphoblastic leukemia. Cell 36:101, 1984. 85. Foley KP, Eisenman RN: Two MAD tails: What the recent knockouts
63. Rabbitts TH: Translocations, master genes, and differences between the of Mad1 and Mxi1 tell us about the MYC/MAX/MAD network.
origins of acute and chronic leukemias. Cell 67:641, 1991. Biochim Biophys Acta 1423:M37, 1999.
64. Dalla-Favera R, Bregni M, Erikson J, et al: Human c-myc onc gene is 86. Hurlin PJ, Queva C, Eisenman RN: Mnt: A novel Max-interacting
located on the region of chromosome 8 that is translocated in Burkitt protein and Myc antagonist. Curr Top Microbiol Immunol 224:115,
lymphoma cells. Proc Natl Acad Sci U S A 79:7824, 1982. 1997.
65. Taub R, Kirsch I, Morton C, et al: Translocation of the c-myc gene into 87. van Riggelen J, Yetil A, Felsher DW: MYC as a regulator of ribosome
the immunoglobulin heavy chain locus in human Burkitt lymphoma biogenesis and protein synthesis. Nat Rev Cancer 10:301, 2010.
and murine plasmacytoma cells. Proc Natl Acad Sci U S A 79:7837, 88. Barna M, Pusic A, Zollo O, et al: Suppression of Myc oncogenic activity
1982. by ribosomal protein haploinsufficiency. Nature 456:971, 2008.
66. Adams JM, Gerondakis S, Webb E, et al: Cellular myc oncogene is 89. Bui TV, Mendell JT: Myc: Maestro of MicroRNAs. Genes Cancer 1:568,
altered by chromosome translocation to an immunoglobulin locus in 2010.
murine plasmacytomas and is rearranged similarly in human Burkitt 90. ODonnell KA, Wentzel EA, Zeller KI, et al: c-Myc-regulated microR-
lymphomas. Proc Natl Acad Sci U S A 80:1982, 1983. NAs modulate E2F1 expression. Nature 435:839, 2005.
67. Erikson J, Nishikura K, ar-Rushdi A, et al: Translocation of an immu- 91. Ferrando AA, Look AT: Clinical implications of recurring chromosomal
noglobulin kappa locus to a region 3 of an unrearranged c-myc onco- and associated molecular abnormalities in acute lymphoblastic leuke-
gene enhances c-myc transcription. Proc Natl Acad Sci U S A 80:7581, mia. Semin Hematol 37:381, 2000.
1983. 92. Chen Q, Cheng JT, Tasi LH, et al: The tal gene undergoes chromosome
68. Croce CM, Thierfelder W, Erikson J, et al: Transcriptional activation translocation in T cell leukemia and potentially encodes a helix-loop-
of an unrearranged and untranslocated c-myc oncogene by transloca- helix protein. EMBO J 9:415, 1990.
tion of a C lambda locus in Burkitt. Proc Natl Acad Sci U S A 80:6922, 93. Begley CG, Aplan PD, Davey MP, et al: Chromosomal translocation
1983. in a human leukemic stem-cell line disrupts the T-cell antigen receptor
69. Emanuel BS, Selden JR, Chaganti RS, et al: The 2p breakpoint of a delta-chain diversity region and results in a previously unreported
2;8 translocation in Burkitt lymphoma interrupts the V kappa locus. fusion transcript. Proc Natl Acad Sci U S A 86:2031, 1989.
Proc Natl Acad Sci U S A 81:2444, 1984. 94. Xia Y, Brown L, Yang CY, et al: TAL2, a helix-loop-helix gene activated
70. Hollis GF, Mitchell KF, Battey J, et al: A variant translocation places by the (7;9)(q34;q32) translocation in human T-cell leukemia. Proc
the lambda immunoglobulin genes 3 to the c-myc oncogene in Burkitts Natl Acad Sci U S A 88:11416, 1991.
lymphoma. Nature 307:752, 1984. 95. Mellentin JD, Smith SD, Cleary ML: Lyl-1, a novel gene altered by
71. Rappold GA, Hameister H, Cremer T, et al: c-myc and immunoglobu- chromosomal translocation in T cell leukemia, codes for a protein with
lin kappa light chain constant genes are on the 8q+ chromosome of a helix-loop-helix DNA binding motif. Cell 58:77, 1989.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 950.e3

96. Finger LR, Harvey RC, Moore RC, et al: A common mechanism of 117. Baer R: TAL1, TAL2 and LYL1: A family of basic helix-loop-helix
chromosomal translocation in T- and B-cell neoplasia. Science 234:982, proteins implicated in T cell acute leukaemia. Semin Cancer Biol 4:341,
1986. 1993.
97. McKeithan TW, Shima EA, Le Beau MM, et al: Molecular cloning of 118. ONeil J, Shank J, Cusson N, et al: TAL1/SCL induces leukemia by
the breakpoint junction of a human chromosomal 8;14 translocation inhibiting the transcriptional activity of E47/HEB. Cancer Cell 5:587,
involving the T-cell receptor alpha-chain gene and sequences on the 3 2004.
side of MYC. Proc Natl Acad Sci U S A 83:6636, 1986. 119. Bain G, Engel I, Robanus Maandag EC, et al: E2A deficiency leads to
98. Shima EA, Le Beau MM, McKeithan TW, et al: Gene encoding the abnormalities in alphabeta T-cell development and to rapid develop-
alpha chain of the T-cell receptor is moved immediately downstream of ment of T-cell lymphomas. Mol Cell Biol 17:4782, 1997.
c-myc in a chromosomal 8;14 translocation in a cell line from a human 120. Yan W, Young AZ, Soares VC, et al: High incidence of T-cell tumors
T-cell leukemia. Proc Natl Acad Sci U S A 83:3439, 1986. in E2A-null mice and E2A/Id1 double-knockout mice. Mol Cell Biol
99. Wang J, Jani-Sait SN, Escalon EA, et al: The t(14;21)(q11.2;q22) 17:7317, 1997.
chromosomal translocation associated with T-cell acute lymphoblastic 121. Aplan PD, Jones CA, Chervinsky DS, et al: An scl gene product lacking
leukemia activates the BHLHB1 gene. Proc Natl Acad Sci U S A the transactivation domain induces bony abnormalities and cooperates
97:3497, 2000. with LMO1 to generate T-cell malignancies in transgenic mice. EMBO
100. McGuire EA, Hockett RD, Pollock KM, et al: The t(11;14)(p15;q11) J 16:2408, 1997.
in a T-cell acute lymphoblastic leukemia cell line activates multiple 122. Boehm T, Foroni L, Kaneko Y, et al: The rhombotin family of cysteine-
transcripts, including Ttg-1, a gene encoding a potential zinc finger rich LIM-domain oncogenes: Distinct members are involved in T-cell
protein. Mol Cell Biol 9:2124, 1989. translocations to human chromosomes 11p15 and 11p13. Proc Natl
101. Ferrando AA, Look AT: Gene expression profiling in T-cell acute lym- Acad Sci U S A 88:4367, 1991.
phoblastic leukemia. Semin Hematol 40:274, 2003. 123. Royer-Pokora B, Loos U, Ludwig WD: TTG-2, a new gene encoding
102. Kennedy MA, Gonzalez-Sarmiento R, Kees UR, et al: HOX11, a a cysteine-rich protein with the LIM motif, is overexpressed in acute
homeobox-containing T-cell oncogene on human chromosome 10q24. T-cell leukaemia with the t(11;14)(p13;q11). Oncogene 6:1887, 1991.
Proc Natl Acad Sci U S A 88:8900, 1991. 124. Wadman I, Li J, Bash RO, et al: Specific in vivo association between
103. Bernard OA, Busson-LeConiat M, Ballerini P, et al: A new recurrent the bHLH and LIM proteins implicated in human T cell leukemia.
and specific cryptic translocation, t(5;14)(q35;q32), is associated with EMBO J 13:4831, 1994.
expression of the Hox11L2 gene in T acute lymphoblastic leukemia. 125. Valge-Archer VE, Osada H, Warren AJ, et al: The LIM protein RBTN2
Leukemia 15:1495, 2001. and the basic helix-loop-helix protein TAL1 are present in a complex
104. Lu M, Gong ZY, Shen WF, et al: The tcl-3 proto-oncogene altered by in erythroid cells. Proc Natl Acad Sci U S A 91:8617, 1994.
chromosomal translocation in T-cell leukemia codes for a homeobox 126. Larson RC, Lavenir I, Larson TA, et al: Protein dimerization between
protein. EMBO J 10:2905, 1991. Lmo2 (Rbtn2) and Tal1 alters thymocyte development and potentiates
105. Dube ID, Kamel-Reid S, Yuan CC, et al: A novel human homeobox T cell tumorigenesis in transgenic mice. EMBO J 15:1021, 1996.
gene lies at the chromosome 10 breakpoint in lymphoid neoplasias with 127. Porcher C, Swat W, Rockwell K, et al: The T cell leukemia oncoprotein
chromosomal translocation t(10;14). Blood 78:2996, 1991. SCL/tal-1 is essential for development of all hematopoietic lineages.
106. Dear TN, Sanchez-Garcia I, Rabbitts TH: The HOX11 gene encodes Cell 86:47, 1996.
a DNA-binding nuclear transcription factor belonging to a distinct 128. Warren AJ, Colledge WH, Carlton MB, et al: The oncogenic cysteine-
family of homeobox genes. Proc Natl Acad Sci U S A 90:4431, rich LIM domain protein rbtn2 is essential for erythroid development.
1993. Cell 78:45, 1994.
107. Brown L, Cheng JT, Chen Q, et al: Site-specific recombination of the 129. Osada H, Grutz GG, Axelson H, et al: LIM-only protein Lmo2 forms
tal-1 gene is a common occurrence in human T cell leukemia. EMBO a protein complex with erythroid transcription factor GATA-1. Leuke-
J 9:3343, 1990. mia 11:307, 1997.
108. Aplan PD, Lombardi DP, Kirsch IR: Structural characterization of SIL, 130. Lahlil R, Lecuyer E, Herblot S, et al: SCL assembles a multifactorial
a gene frequently disrupted in T-cell acute lymphoblastic leukemia. Mol complex that determines glycophorin A expression. Mol Cell Biol
Cell Biol 11:5462, 1991. 24:1439, 2004.
109. Bernard O, Lecointe N, Jonveaux P, et al: Two site-specific 131. McGuire EA, Rintoul CE, Sclar GM, et al: Thymic overexpression of
deletions and t(1;14) translocation restricted to human T-cell Ttg-1 in transgenic mice results in T-cell acute lymphoblastic leukemia/
acute leukemias disrupt the 5 part of the tal-1 gene. Oncogene 6:1477, lymphoma. Mol Cell Biol 12:4186, 1992.
1991. 132. Fisch P, Boehm T, Lavenir I, et al: T-cell acute lymphoblastic lymphoma
110. Aplan PD, Lombardi DP, Reaman GH, et al: Involvement of the puta- induced in transgenic mice by the RBTN1 and RBTN2 LIM-domain
tive hematopoietic transcription factor SCL in T-cell acute lymphoblas- genes. Oncogene 7:2389, 1992.
tic leukemia. Blood 79:1327, 1992. 133. Larson RC, Fisch P, Larson TA, et al: T cell tumours of disparate phe-
111. Breit TM, Mol EJ, Wolvers-Tettero IL, et al: Site-specific deletions notype in mice transgenic for Rbtn-2. Oncogene 9:3675, 1994.
involving the tal-1 and sil genes are restricted to cells of the T cell 134. Larson RC, Osada H, Larson TA, et al: The oncogenic LIM protein
receptor alpha/beta lineage: T cell receptor delta gene deletion mecha- Rbtn2 causes thymic developmental aberrations that precede malig-
nism affects multiple genes. J Exp Med 177:965, 1993. nancy in transgenic mice. Oncogene 11:853, 1995.
112. Bash RO, Hall S, Timmons CF, et al: Does activation of the TAL1 gene 135. Neale GA, Rehg JE, Goorha RM: Ectopic expression of rhombotin-2
occur in a majority of patients with T-cell acute lymphoblastic leuke- causes selective expansion of CD4-CD8 lymphocytes in the thymus
mia? A pediatric oncology group study. Blood 86:666, 1995. and T-cell tumors in transgenic mice. Blood 86:3060, 1995.
113. Palomero T, Odom DT, ONeil J, et al: Transcriptional regulatory 136. Kees UR, Heerema NA, Kumar R, et al: Expression of HOX11 in
networks downstream of TAL1/SCL in T-cell acute lymphoblastic leu- childhood T-lineage acute lymphoblastic leukaemia can occur in the
kemia. Blood 108:986, 2006. absence of cytogenetic aberration at 10q24: A study from the Childrens
114. Shivdasani RA, Mayer EL, Orkin SH: Absence of blood formation in Cancer Group (CCG). Leukemia 17:887, 2003.
mice lacking the T-cell leukaemia oncoprotein tal-1/SCL. Nature 137. Berger R, Dastugue N, Busson M, et al: t(5;14)/HOX11L2-positive
373:432, 1995. T-cell acute lymphoblastic leukemia. A collaborative study of the
115. Robb L, Lyons I, Li R, et al: Absence of yolk sac hematopoiesis from Groupe Francais de Cytogenetique Hematologique (GFCH). Leukemia
mice with a targeted disruption of the scl gene. Proc Natl Acad Sci U S 17:1851, 2003.
A 92:7075, 1995. 138. Cytogenetic abnormalities in adult acute lymphoblastic leukemia: Cor-
116. Mikkola HK, Klintman J, Yang H, et al: Haematopoietic stem cells relations with hematologic findings outcome. A Collaborative Study of
retain long-term repopulating activity and multipotency in the absence the Group Francais de Cytogenetique Hematologique. Blood 87:3135,
of stem-cell leukaemia SCL/tal-1 gene. Nature 421:547, 2003. 1996.
950.e4 Part VII Hematologic Malignancies

139. Roberts CW, Shutter JR, Korsmeyer SJ: Hox11 controls the genesis of 163. Murre C, McCaw PS, Vaessin H, et al: Interactions between heterolo-
the spleen. Nature 368:747, 1994. gous helix-loop-helix proteins generate complexes that bind specifically
140. Dear TN, Colledge WH, Carlton MB, et al: The Hox11 gene is essen- to a common DNA sequence. Cell 58:537, 1989.
tial for cell survival during spleen development. Development 121:2909, 164. Mellentin JD, Nourse J, Hunger SP, et al: Molecular analysis of the
1995. t(1;19) breakpoint cluster region in pre-B cell acute lymphoblastic
141. Raju K, Tang S, Dube ID, et al: Characterization and developmental leukemias. Genes Chromosomes Cancer 2:239, 1990.
expression of Tlx-1, the murine homolog of HOX11. Mech Dev 44:51, 165. Van Dijk MA, Voorhoeve PM, Murre C: Pbx1 is converted into a
1993. transcriptional activator upon acquiring the N-terminal region of E2A
142. Roberts CW, Sonder AM, Lumsden A, et al: Development expression in pre-B-cell acute lymphoblastoid leukemia. Proc Natl Acad Sci U S A
of Hox11 and specification of splenic cell fate. Am J Pathol 146:1089, 90:6061, 1993.
1995. 166. LeBrun DP, Cleary ML: Fusion with E2A alters the transcriptional
143. Owens BM, Hawley TS, Spain LM, et al: TLX1/HOX11-mediated properties of the homeodomain protein PBX1 in t(1;19) leukemias.
disruption of primary thymocyte differentiation prior to the CD4+CD8+ Oncogene 9:1641, 1994.
double-positive stage. Br J Haematol 132:216, 2006. 167. Lu Q, Wright DD, Kamps MP: Fusion with E2A converts the Pbx1
144. Kawabe T, Muslin AJ, Korsmeyer SJ: HOX11 interacts with protein homeodomain protein into a constitutive transcriptional activator in
phosphatases PP2A and PP1 and disrupts a G2/M cell-cycle check- human leukemias carrying the t(1;19) translocation. Mol Cell Biol
point. Nature 385:454, 1997. 14:3938, 1994.
145. Ballerini P, Blaise A, Busson-Le Coniat M, et al: HOX11L2 expression 168. Kamps MP, Baltimore D: E2A-Pbx1, the t(1;19) translocation protein
defines a clinical subtype of pediatric T-ALL associated with poor prog- of human pre-B-cell acute lymphocytic leukemia, causes acute myeloid
nosis. Blood 100:991, 2002. leukemia in mice. Mol Cell Biol 13:351, 1993.
146. Mauvieux L, Leymarie V, Helias C, et al: High incidence of Hox11L2 169. Dedera DA, Waller EK, LeBrun DP, et al: Chimeric homeobox gene
expression in children with T-ALL. Leukemia 16:2417, 2002. E2A-PBX1 induces proliferation, apoptosis, and malignant lymphomas
147. Cave H, Suciu S, Preudhomme C, et al: Clinical significance of in transgenic mice. Cell 74:833, 1993.
HOX11L2 expression linked to t(5;14)(q35;q32), of HOX11 expres- 170. Monica K, LeBrun DP, Dedera DA, et al: Transformation properties of
sion, and of SIL-TAL fusion in childhood T-cell malignancies: Results the E2a-Pbx1 chimeric oncoprotein: Fusion with E2a is essential, but
of EORTC studies 58881 and 58951. Blood 103:442, 2004. the Pbx1 homeodomain is dispensable. Mol Cell Biol 14:8304, 1994.
148. Shirasawa S, Arata A, Onimaru H, et al: Rnx deficiency results in 171. Kamps MP, Wright DD, Lu Q: DNA-binding by oncoprotein E2a-
congenital central hypoventilation. Nat Genet 24:287, 2000. Pbx1 is important for blocking differentiation but dispensable for fibro-
149. De Keersmaecker K, Real PJ, Gatta GD, et al: The TLX1 oncogene blast transformation. Oncogene 12:19, 1996.
drives aneuploidy in T cell transformation. Nat Med 16:1321, 2010. 172. Chang CP, Shen WF, Rozenfeld S, et al: Pbx proteins display
150. Speleman F, Cauwelier B, Dastugue N, et al: A new recurrent inversion, hexapeptide-dependent cooperative DNA binding with a subset of Hox
inv(7)(p15q34), leads to transcriptional activation of HOXA10 and proteins. Genes Dev 9:663, 1995.
HOXA11 in a subset of T-cell acute lymphoblastic leukemias. Leukemia 173. van Dijk MA, Peltenburg LT, Murre C: Hox gene products modulate
19:358, 2005. the DNA binding activity of Pbx1 and Pbx2. Mech Dev 52:99, 1995.
151. Soulier J, Clappier E, Cayuela JM, et al: HOXA genes are included in 174. Lu Q, Knoepfler PS, Scheele J, et al: Both Pbx1 and E2A-Pbx1 bind
genetic and biologic networks defining human acute T-cell leukemia the DNA motif ATCAATCAA cooperatively with the products of mul-
(T-ALL). Blood 106:274, 2005. tiple murine Hox genes, some of which are themselves oncogenes. Mol
152. Cauwelier B, Cave H, Gervais C, et al: Clinical, cytogenetic and molec- Cell Biol 15:3786, 1995.
ular characteristics of 14 T-ALL patients carrying the TCRbeta-HOXA 175. Knoepfler PS, Kamps MP: The pentapeptide motif of Hox proteins is
rearrangement: A study of the Groupe Francophone de Cytogenetique required for cooperative DNA binding with Pbx1, physically contacts
Hematologique. Leukemia 21:121, 2007. Pbx1, and enhances DNA binding by Pbx1. Mol Cell Biol 15:5811,
153. Meeker TC, Hardy D, Willman C, et al: Activation of the interleukin-3 1995.
gene by chromosome translocation in acute lymphocytic leukemia with 176. Chang CP, de Vivo I, Cleary ML: The Hox cooperativity motif of the
eosinophilia. Blood 76:285, 1990. chimeric oncoprotein E2a-Pbx1 is necessary and sufficient for oncogen-
154. Grimaldi JC, Meeker TC: The t(5;14) chromosomal translocation in a esis. Mol Cell Biol 17:81, 1997.
case of acute lymphocytic leukemia joins the interleukin-3 gene to the 177. Pui CH, Crist WM: Cytogenetic abnormalities in childhood acute
immunoglobulin heavy chain gene. Blood 73:2081, 1989. lymphoblastic leukemia correlates with clinical features and treatment
155. Tycko B, Smith SD, Sklar J: Chromosomal translocations joining LCK outcome. Leuk Lymphoma 7:259, 1992.
and TCRB loci in human T cell leukemia. J Exp Med 174:867, 1991. 178. Rivera GK, Raimondi SC, Hancock ML, et al: Improved outcome in
156. Burnett RC, David JC, Harden AM, et al: The LCK gene is involved childhood acute lymphoblastic leukaemia with reinforced early
in the t(1;7)(p34;q34) in the T-cell acute lymphoblastic leukemia treatment and rotational combination chemotherapy. Lancet 337:61,
derived cell line, HSB-2. Genes Chromosomes Cancer 3:461, 1991. 1991.
157. Wright DD, Sefton BM, Kamps MP: Oncogenic activation of the Lck 179. Uckun FM, Sensel MG, Sather HN, et al: Clinical significance of
protein accompanies translocation of the LCK gene in the human translocation t(1;19) in childhood acute lymphoblastic leukemia in the
HSB2 T-cell leukemia. Mol Cell Biol 14:2429, 1994. context of contemporary therapies: A report from the Childrens Cancer
158. Kamps MP, Look AT, Baltimore D: The human t(1;19) translocation Group. J Clin Oncol 16:527, 1998.
in pre-B ALL produces multiple nuclear E2A-Pbx1 fusion proteins with 180. Hunger SP, Ohyashiki K, Toyama K, et al: Hlf, a novel hepatic bZIP
differing transforming potentials. Genes Dev 5:358, 1991. protein, shows altered DNA-binding properties following fusion to
159. Mellentin JD, Murre C, Donlon TA, et al: The gene for enhancer E2A in t(17;19) acute lymphoblastic leukemia. Genes Dev 6:1608,
binding proteins E12/E47 lies at the t(1;19) breakpoint in acute leuke- 1992.
mias. Science 246:379, 1989. 181. Inaba T, Roberts WM, Shapiro LH, et al: Fusion of the leucine zipper
160. Nourse J, Mellentin JD, Galili N, et al: Chromosomal translocation gene HLF to the E2A gene in human acute B-lineage leukemia. Science
t(1;19) results in synthesis of a homeobox fusion mRNA that codes for 257:531, 1992.
a potential chimeric transcription factor. Cell 60:535, 1990. 182. Yoshihara T, Inaba T, Shapiro LH, et al: E2A-HLF-mediated cell trans-
161. Kamps MP, Murre C, Sun XH, et al: A new homeobox gene contributes formation requires both the trans-activation domains of E2A and the
the DNA binding domain of the t(1;19) translocation protein in pre-B leucine zipper dimerization domain of HLF. Mol Cell Biol 15:3247,
ALL. Cell 60:547, 1990. 1995.
162. Sigvardsson M, ORiordan M, Grosschedl R: EBF and E47 collaborate 183. Hunger SP: Chromosomal translocations involving the E2A gene in
to induce expression of the endogenous immunoglobulin surrogate acute lymphoblastic leukemia: Clinical features and molecular patho-
light chain genes. Immunity 7:25, 1997. genesis. Blood 87:1211, 1996.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 950.e5

184. Inaba T, Inukai T, Yoshihara T, et al: Reversal of apoptosis by the 208. Lavau C, Luo RT, Du C, et al: Retrovirus-mediated gene transfer
leukaemia-associated E2A-HLF chimaeric transcription factor. Nature of MLL-ELL transforms primary myeloid progenitors and causes
382:541, 1996. acute myeloid leukemias in mice. Proc Natl Acad Sci U S A 97:10984,
185. Metzstein MM, Hengartner MO, Tsung N, et al: Transcriptional regu- 2000.
lator of programmed cell death encoded by Caenorhabditis elegans gene 209. Lavau C, Du C, Thirman M, et al: Chromatin-related properties of
ces-2. Nature 382:545, 1996. CBP fused to MLL generate a myelodysplastic-like syndrome that
186. Thompson CB: Transcription. A fate worse than death. Nature 382:492, evolves into myeloid leukemia. EMBO J 19:4655, 2000.
1996. 210. Forster A, Pannell R, Drynan LF, et al: Engineering de novo reciprocal
187. Inukai T, Inoue A, Kurosawa H, et al: SLUG, a ces-1-related zinc finger chromosomal translocations associated with Mll to replicate primary
transcription factor gene with antiapoptotic activity, is a downstream events of human cancer. Cancer Cell 3:449, 2003.
target of the E2A-HLF oncoprotein. Mol Cell 4:343, 1999. 211. Krivtsov AV, Twomey D, Feng Z, et al: Transformation from committed
188. Metzstein MM, Horvitz HR: The C. elegans cell death specification progenitor to leukaemia stem cell initiated by MLL-AF9. Nature
gene ces-1 encodes a snail family zinc finger protein. Mol Cell 4:309, 442:818, 2006.
1999. 212. Armstrong SA, Staunton JE, Silverman LB, et al: MLL translocations
189. Inoue A, Seidel MG, Wu W, et al: Slug, a highly conserved zinc finger specify a distinct gene expression profile that distinguishes a unique
transcriptional repressor, protects hematopoietic progenitor cells from leukemia. Nat Genet 30:41, 2002.
radiation-induced apoptosis in vivo. Cancer Cell 2:279, 2002. 213. Yeoh EJ, Ross ME, Shurtleff SA, et al: Classification, subtype discovery,
190. Wu WS, Heinrichs S, Xu D, et al: Slug antagonizes p53-mediated and prediction of outcome in pediatric acute lymphoblastic leukemia
apoptosis of hematopoietic progenitors by repressing puma. Cell by gene expression profiling. Cancer Cell 1:133, 2002.
123:641, 2005. 214. Ferrando AA, Armstrong SA, Neuberg DS, et al: Gene expression sig-
191. Ziemin-van der Poel S, McCabe NR, Gill HJ, et al: Identification of a natures in MLL-rearranged T-lineage and B-precursor acute leukemias:
gene, MLL, that spans the breakpoint in 11q23 translocations associ- Dominance of HOX dysregulation. Blood 102:262, 2003.
ated with human leukemias. Proc Natl Acad Sci U S A 88:10735, 1991. 215. Ayton PM, Cleary ML: Transformation of myeloid progenitors by MLL
192. Tkachuk DC, Kohler S, Cleary ML: Involvement of a homolog of oncoproteins is dependent on Hoxa7 and Hoxa9. Genes Dev 17:2298,
Drosophila trithorax by 11q23 chromosomal translocations in acute 2003.
leukemias. Cell 71:691, 1992. 216. Kumar AR, Hudson WA, Chen W, et al: Hoxa9 influences the pheno-
193. Gu Y, Nakamura T, Alder H, et al: The t(4;11) chromosome transloca- type but not the incidence of Mll-AF9 fusion gene leukemia. Blood
tion of human acute leukemias fuses the ALL-1 gene, related to Dro- 103:1823, 2004.
sophila trithorax, to the AF-4 gene. Cell 71:701, 1992. 217. Stam RW, den Boer ML, Schneider P, et al: Targeting FLT3 in primary
194. Djabali M, Selleri L, Parry P, et al: A trithorax-like gene is interrupted MLL-gene-rearranged infant acute lymphoblastic leukemia. Blood
by chromosome 11q23 translocations in acute leukaemias. Nat Genet 106:2484, 2005.
2:113, 1992. 218. Taketani T, Taki T, Sugita K, et al: FLT3 mutations in the activation
195. Muntean AG, Hess JL: The pathogenesis of mixed-lineage leukemia. loop of tyrosine kinase domain are frequently found in infant ALL with
Annu Rev Pathol 7:283, 2012. MLL rearrangements and pediatric ALL with hyperdiploidy. Blood
196. Jude CD, Climer L, Xu D, et al: Unique and independent roles for 103:1085, 2004.
MLL in adult hematopoietic stem cells and progenitors. Cell Stem Cell 219. Mohan M, Lin C, Guest E, et al: Licensed to elongate: A molecular
1:324, 2007. mechanism for MLL-based leukaemogenesis. Nat Rev Cancer 10:721,
197. Ernst P, Fisher JK, Avery W, et al: Definitive hematopoiesis requires the 2010.
mixed-lineage leukemia gene. Dev Cell 6:437, 2004. 220. Muntean AG, Tan J, Sitwala K, et al: The PAF complex synergizes with
198. Ernst P, Mabon M, Davidson AJ, et al: An Mll-dependent Hox program MLL fusion proteins at HOX loci to promote leukemogenesis. Cancer
drives hematopoietic progenitor expansion. Curr Biol 14:2063, Cell 17:609, 2010.
2004. 221. Lin C, Smith ER, Takahashi H, et al: AFF4, a component of the
199. Hsieh JJ, Cheng EH, Korsmeyer SJ: Taspase1: A threonine aspartase ELL/P-TEFb elongation complex and a shared subunit of MLL chime-
required for cleavage of MLL and proper HOX gene expression. Cell ras, can link transcription elongation to leukemia. Mol Cell 37:429,
115:293, 2003. 2010.
200. Yokoyama A, Kitabayashi I, Ayton PM, et al: Leukemia proto- 222. Yokoyama A, Lin M, Naresh A, et al: A higher-order complex contain-
oncoprotein MLL is proteolytically processed into 2 fragments with ing AF4 and ENL family proteins with P-TEFb facilitates oncogenic
opposite transcriptional properties. in Blood 3710, 2002. and physiologic MLL-dependent transcription. Cancer Cell 17:198,
201. Hsieh JJ, Ernst P, Erdjument-Bromage H, et al: Proteolytic cleavage of 2010.
MLL generates a complex of N- and C-terminal fragments that confers 223. Bitoun E, Oliver PL, Davies KE: The mixed-lineage leukemia fusion
protein stability and subnuclear localization. Mol Cell Biol 23:186, partner AF4 stimulates RNA polymerase II transcriptional elongation
2003. and mediates coordinated chromatin remodeling. Hum Mol Genet
202. Nakamura T, Mori T, Tada S, et al: ALL-1 is a histone methyltransferase 16:92, 2007.
that assembles a supercomplex of proteins involved in transcriptional 224. Mohan M, Herz HM, Takahashi YH, et al: Linking H3K79 trimeth-
regulation. Mol Cell 10:1119, 2002. ylation to Wnt signaling through a novel Dot1-containing complex
203. Somervaille TC, Cleary ML: Grist for the MLL: How do MLL onco- (DotCom). Genes Dev 24:574, 2010.
genic fusion proteins generate leukemia stem cells? Int J Hematol 225. Mueller D, Garcia-Cuellar MP, Bach C, et al: Misguided transcriptional
91:735, 2010. elongation causes mixed lineage leukemia. PLoS Biol 7:e1000249,
204. Thirman MJ, Levitan DA, Kobayashi H, et al: Cloning of ELL, a gene 2009.
that fuses to MLL in a t(11;19)(q23;p13.1) in acute myeloid leukemia. 226. Mueller D, Bach C, Zeisig D, et al: A role for the MLL fusion partner
Proc Natl Acad Sci U S A 91:12110, 1994. ENL in transcriptional elongation and chromatin modification. Blood
205. Mitani K, Kanda Y, Ogawa S, et al: Cloning of several species of MLL/ 110:4445, 2007.
MEN chimeric cDNAs in myeloid leukemia with t(11;19)(q23;p13.1) 227. Okada Y, Feng Q, Lin Y, et al: hDOT1L links histone methylation to
translocation. Blood 85:2017, 1995. leukemogenesis. Cell 121:167, 2005.
206. Corral J, Lavenir I, Impey H, et al: An Mll-AF9 fusion gene made by 228. Zhang W, Xia X, Reisenauer MR, et al: Dot1a-AF9 complex mediates
homologous recombination causes acute leukemia in chimeric mice: A histone H3 Lys-79 hypermethylation and repression of ENaCalpha in
method to create fusion oncogenes. Cell 85:853, 1996. an aldosterone-sensitive manner. J Biol Chem 281:18059, 2006.
207. Zeisig BB, Garcia-Cuellar MP, Winkler TH, et al: The oncoprotein 229. Guenther MG, Lawton LN, Rozovskaia T, et al: Aberrant chromatin at
MLL-ENL disturbs hematopoietic lineage determination and trans- genes encoding stem cell regulators in human mixed-lineage leukemia.
forms a biphenotypic lymphoid/myeloid cell. Oncogene 22:1629, 2003. Genes Dev 22:3403, 2008.
950.e6 Part VII Hematologic Malignancies

230. Krivtsov AV, Feng Z, Lemieux ME, et al: H3K79 methylation profiles 253. Takeuchi S, Seriu T, Bartram CR, et al: TEL is one of the targets for
define murine and human MLL-AF4 leukemias. Cancer Cell 14:355, deletion on 12p in many cases of childhood B-lineage acute lympho-
2008. blastic leukemia. Leukemia 11:1220, 1997.
231. Krivtsov AV, Armstrong SA: MLL translocations, histone modifications 254. Zelent A, Greaves M, Enver T: Role of the TEL-AML1 fusion gene in
and leukaemia stem-cell development. Nat Rev Cancer 7:823, the molecular pathogenesis of childhood acute lymphoblastic leukae-
2007. mia. Oncogene 23:4275, 2004.
232. Chang MJ, Wu H, Achille NJ, et al: Histone H3 lysine 79 methyltrans- 255. Okuda T, van Deursen J, Hiebert SW, et al: AML1, the target of mul-
ferase Dot1 is required for immortalization by MLL oncogenes. Cancer tiple chromosomal translocations in human leukemia, is essential for
Res 70:10234, 2010. normal fetal liver hematopoiesis. Cell 84:321, 1996.
233. Nguyen AT, Taranova O, He J, et al: DOT1L, the H3K79 methyltrans- 256. Wang Q, Stacy T, Binder M, et al: Disruption of the Cbfa2 gene causes
ferase, is required for MLL-AF9-mediated leukemogenesis. Blood necrosis and hemorrhaging in the central nervous system and blocks
117:6912, 2011. definitive hematopoiesis. Proc Natl Acad Sci U S A 93:3444, 1996.
234. Jo SY, Granowicz EM, Maillard I, et al: Requirement for Dot1l in 257. Minelli A, Maserati E, Rossi G, et al: Familial platelet disorder with
murine postnatal hematopoiesis and leukemogenesis by MLL transloca- propensity to acute myelogenous leukemia: Genetic heterogeneity and
tion. Blood 117:4759, 2011. progression to leukemia via acquisition of clonal chromosome anoma-
235. Bernt KM, Zhu N, Sinha AU, et al: MLL-rearranged leukemia is lies. Genes Chromosomes Cancer 40:165, 2004.
dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 258. Schultz KR, Pullen DJ, Sather HN, et al: Risk- and response-based
20:66, 2011. classification of childhood B-precursor acute lymphoblastic leukemia:
236. Daigle SR, Olhava EJ, Therkelsen CA, et al: Selective killing of mixed A combined analysis of prognostic markers from the Pediatric Oncology
lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Group (POG) and Childrens Cancer Group (CCG). Blood 109:926,
Cancer Cell 20:53, 2011. 2007.
237. Greaves MF: Infant leukaemia biology, aetiology and treatment. Leuke- 259. Loh ML, Goldwasser MA, Silverman LB, et al: Prospective analysis of
mia 10:372, 1996. TEL/AML1-positive patients treated on Dana-Farber Cancer Institute
238. Biondi A, Cimino G, Pieters R, et al: Biological and therapeutic aspects Consortium Protocol 95-01. Blood 107:4508, 2006.
of infant leukemia. Blood 96:24, 2000. 260. Asakura K, Uchida H, Miyachi H, et al: TEL/AML1 overcomes drug
239. Pui CH, Gaynon PS, Boyett JM, et al: Outcome of treatment in child- resistance through transcriptional repression of multidrug resistance-1
hood acute lymphoblastic leukaemia with rearrangements of the 11q23 gene expression. Mol Cancer Res 2:339, 2004.
chromosomal region. Lancet 359:1909, 2002. 261. Zaza G, Yang W, Kager L, et al: Acute lymphoblastic leukemia
240. Chessells JM, Eden OB, Bailey CC, et al: Acute lymphoblastic leukae- with TEL-AML1 fusion has lower expression of genes involved in
mia in infancy: Experience in MRC UKALL trials. Report from the purine metabolism and lower de novo purine synthesis. Blood 104:1435,
Medical Research Council Working Party on Childhood Leukaemia. 2004.
Leukemia 8:1275, 1994. 262. Heisterkamp N, Stephenson JR, Groffen J, et al: Localization of the
241. Dreyling MH, Martinez-Climent JA, Zheng M, et al: The t(10;11) c-ab1 oncogene adjacent to a translocation break point in chronic
(p13;q14) in the U937 cell line results in the fusion of the AF10 gene myelocytic leukaemia. Nature 306:239, 1983.
and CALM, encoding a new member of the AP-3 clathrin assembly 263. Leibowitz D, Schaefer-Rego K, Popenoe DW, et al: Variable break-
protein family. Proc Natl Acad Sci U S A 93:4804, 1996. points on the Philadelphia chromosome in chronic myelogenous leu-
242. Asnafi V, Radford-Weiss I, Dastugue N, et al: CALM-AF10 is a kemia. Blood 66:243, 1985.
common fusion transcript in T-ALL and is specific to the TCRgam- 264. Grosveld G, Verwoerd T, van Agthoven T, et al: The chronic myelocytic
madelta lineage. Blood 102:1000, 2003. cell line K562 contains a breakpoint in bcr and produces a chimeric
243. Okada Y, Jiang Q, Lemieux M, et al: Leukaemic transformation by bcr/c-abl transcript. Mol Cell Biol 6:607, 1986.
CALM-AF10 involves upregulation of Hoxa5 by hDOT1L. Nat Cell 265. Deininger MW, Goldman JM, Melo JV: The molecular biology of
Biol 8:1017, 2006. chronic myeloid leukemia. Blood 96:3343, 2000.
244. Deshpande AJ, Cusan M, Rawat VP, et al: Acute myeloid leukemia is 266. Chan LC, Karhi KK, Rayter SI, et al: A novel abl protein expressed in
propagated by a leukemic stem cell with lymphoid characteristics in a Philadelphia chromosome positive acute lymphoblastic leukaemia.
mouse model of CALM/AF10-positive leukemia. Cancer Cell 10:363, Nature 325:635, 1987.
2006. 267. Clark SS, McLaughlin J, Crist WM, et al: Unique forms of the abl
245. Greaves MF, Maia AT, Wiemels JL, et al: Leukemia in twins: Lessons tyrosine kinase distinguish Ph1-positive CML from Ph1-positive ALL.
in natural history. Blood 102:2321, 2003. Science 235:85, 1987.
246. Greaves MF, Wiemels J: Origins of chromosome translocations in child- 268. Kurzrock R, Shtalrid M, Romero P, et al: A novel c-abl protein product
hood leukaemia. Nat Rev Cancer 3:639, 2003. in Philadelphia-positive acute lymphoblastic leukaemia. Nature
247. Mori H, Colman SM, Xiao Z, et al: Chromosome translocations and 325:631, 1987.
covert leukemic clones are generated during normal fetal development. 269. Konopka JB, Watanabe SM, Singer JW, et al: Cell lines and clinical
Proc Natl Acad Sci U S A 99:8242, 2002. isolates derived from Ph1-positive chronic myelogenous leukemia
248. Golub TR, Barker GF, Stegmaier K, et al: The TEL gene contributes patients express c-abl proteins with a common structural alteration. Proc
to the pathogenesis of myeloid and lymphoid leukemias by diverse Natl Acad Sci U S A 82:1810, 1985.
molecular genetic mechanisms. Curr Top Microbiol Immunol 220:67, 270. Konopka JB, Watanabe SM, Witte ON: An alteration of the human
1997. c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase
249. Hock H, Meade E, Medeiros S, et al: Tel/Etv6 is an essential and selec- activity. Cell 37:1035, 1984.
tive regulator of adult hematopoietic stem cell survival. Genes Dev 271. Kloetzer W, Kurzrock R, Smith L, et al: The human cellular abl gene
18:2336, 2004. product in the chronic myelogenous leukemia cell line K562 has an
250. Cave H, Cacheux V, Raynaud S, et al: ETV6 is the target of chromo- associated tyrosine protein kinase activity. Virology 140:230, 1985.
some 12p deletions in t(12;21) childhood acute lymphocytic leukemia. 272. Naldini L, Stacchini A, Cirillo DM, et al: Phosphotyrosine antibodies
Leukemia 11:1459, 1997. identify the p210c-abl tyrosine kinase and proteins phosphorylated on
251. Jousset C, Carron C, Boureux A, et al: A domain of TEL conserved in tyrosine in human chronic myelogenous leukemia cells. Mol Cell Biol
a subset of ETS proteins defines a specific oligomerization interface 6:1803, 1986.
essential to the mitogenic properties of the TEL-PDGFR beta oncop- 273. Kharbanda S, Ren R, Pandey P, et al: Activation of the c-Abl tyrosine
rotein. EMBO J 16:69, 1997. kinase in the stress response to DNA-damaging agents. Nature 376:785,
252. McLean TW, Ringold S, Neuberg D, et al: TEL/AML-1 dimerizes and 1995.
is associated with a favorable outcome in childhood acute lymphoblastic 274. Sawyers CL, McLaughlin J, Goga A, et al: The nuclear tyrosine kinase
leukemia. Blood 88:4252, 1996. c-Abl negatively regulates cell growth. Cell 77:121, 1994.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 950.e7

275. Mattioni T, Jackson PK, Bchini-Hooft van Huijsduijnen O, et al: Cell 298. Arico M, Valsecchi MG, Camitta B, et al: Outcome of treatment in
cycle arrest by tyrosine kinase Abl involves altered early mitogenic children with Philadelphia chromosome-positive acute lymphoblastic
response. Oncogene 10:1325, 1995. leukemia. N Engl J Med 342:998, 2000.
276. Goga A, Liu X, Hambuch TM, et al: p53 dependent growth suppres- 299. Balduzzi A, Valsecchi MG, Uderzo C, et al: Chemotherapy versus
sion by the c-Abl nuclear tyrosine kinase. Oncogene 11:791, 1995. allogeneic transplantation for very-high-risk childhood acute lympho-
277. Tybulewicz VL, Crawford CE, Jackson PK, et al: Neonatal lethality and blastic leukaemia in first complete remission: Comparison by genetic
lymphopenia in mice with a homozygous disruption of the c-abl proto- randomisation in an international prospective study. Lancet 366:635,
oncogene. Cell 65:1153, 1991. 2005.
278. Schwartzberg PL, Stall AM, Hardin JD, et al: Mice homozygous for 300. Satwani P, Sather H, Ozkaynak F, et al: Allogeneic bone marrow trans-
the ablm1 mutation show poor viability and depletion of selected B plantation in first remission for children with ultra-high-risk features of
and T cell populations. Cell 65:1165, 1991. acute lymphoblastic leukemia: A childrens oncology group study
279. Van Etten RA, Jackson P, Baltimore D: The mouse type IV c-abl gene report. Biol Blood Marrow Transplant 13:218, 2007.
product is a nuclear protein, and activation of transforming ability is 301. Kiehl MG, Kraut L, Schwerdtfeger R, et al: Outcome of allogeneic
associated with cytoplasmic localization. Cell 58:669, 1989. hematopoietic stem-cell transplantation in adult patients with acute
280. Lugo TG, Pendergast AM, Muller AJ, et al: Tyrosine kinase activity and lymphoblastic leukemia: No difference in related compared with unre-
transformation potency of bcr-abl oncogene products. Science 247:1079, lated transplant in first complete remission. J Clin Oncol 22:2816,
1990. 2004.
281. Daley GQ, Baltimore D: Transformation of an interleukin 3-dependent 302. Talano JM, Casper JT, Camitta BM, et al: Alternative donor bone
hematopoietic cell line by the chronic myelogenous leukemia-specific marrow transplant for children with Philadelphia chromosome ALL.
P210bcr/abl protein. Proc Natl Acad Sci U S A 85:9312, 1988. Bone Marrow Transplant 37:135, 2006.
282. Elefanty AG, Hariharan IK, Cory S: Bcr-abl, the hallmark of chronic 303. von Bubnoff N, Peschel C, Duyster J: Resistance of Philadelphia-
myeloid leukaemia in man, induces multiple haemopoietic neoplasms chromosome positive leukemia towards the kinase inhibitor imatinib
in mice. EMBO J 9:1069, 1990. (STI571, Glivec): A targeted oncoprotein strikes back. Leukemia
283. Gishizky ML, Johnson-White J, Witte ON: Efficient transplantation 17:829, 2003.
of BCR-ABL-induced chronic myelogenous leukemia-like syndrome in 304. Gorre ME, Sawyers CL: Molecular mechanisms of resistance to STI571
mice. Proc Natl Acad Sci U S A 90:3755, 1993. in chronic myeloid leukemia. Curr Opin Hematol 9:303, 2002.
284. Kelliher M, Knott A, McLaughlin J, et al: Differences in oncogenic 305. Schultz KR, Bowman WP, Aledo A, et al: Improved early event-free
potency but not target cell specificity distinguish the two forms of the survival with imatinib in Philadelphia chromosome-positive acute lym-
BCR/ABL oncogene. Mol Cell Biol 11:4710, 1991. phoblastic leukemia: A childrens oncology group study. J Clin Oncol
285. Cortez D, Reuther G, Pendergast AM: The Bcr-Abl tyrosine kinase 27:5175, 2009.
activates mitogenic signaling pathways and stimulates G1-to-S phase 306. Quintas-Cardama A, Kantarjian H, Cortes J: Imatinib and beyond
transition in hematopoietic cells. Oncogene 15:2333, 1997. exploring the full potential of targeted therapy for CML. Nat Rev Clin
286. Varticovski L, Daley GQ, Jackson P, et al: Activation of phosphati- Oncol 6:535, 2009.
dylinositol 3-kinase in cells expressing abl oncogene variants. Mol Cell 307. Druker BJ: Circumventing resistance to kinase-inhibitor therapy.
Biol 11:1107, 1991. N Engl J Med 354:2594, 2006.
287. Reuther JY, Reuther GW, Cortez D, et al: A requirement for NF-kappaB 308. OHare T, Corbin AS, Druker BJ: Targeted CML therapy: Controlling
activation in Bcr-Abl-mediated transformation. Genes Dev 12:968, drug resistance, seeking cure. Curr Opin Genet Dev 16:92, 2006.
1998. 309. Saito M, Gao J, Basso K, et al: A signaling pathway mediating down-
288. Carlesso N, Frank DA, Griffin JD: Tyrosyl phosphorylation and DNA regulation of BCL6 in germinal center B cells is blocked by BCL6 gene
binding activity of signal transducers and activators of transcription alterations in B cell lymphoma. Cancer Cell 12:280, 2007.
(STAT) proteins in hematopoietic cell lines transformed by Bcr/Abl. 310. Duy C, Hurtz C, Shojaee S, et al: BCL6 enables Ph+ acute lympho-
J Exp Med 183:811, 1996. blastic leukaemia cells to survive BCR-ABL1 kinase inhibition. Nature
289. Raitano AB, Halpern JR, Hambuch TM, et al: The Bcr-Abl leukemia 473:384, 2011.
oncogene activates Jun kinase and requires Jun for transformation. Proc 311. Graux C, Cools J, Melotte C, et al: Fusion of NUP214 to ABL1 on
Natl Acad Sci U S A 92:11746, 1995. amplified episomes in T-cell acute lymphoblastic leukemia. Nat Genet
290. Sawyers CL, McLaughlin J, Witte ON: Genetic requirement for Ras in 36:1084, 2004.
the transformation of fibroblasts and hematopoietic cells by the Bcr-Abl 312. Mackarehtschian K, Hardin JD, Moore KA, et al: Targeted disruption
oncogene. J Exp Med 181:307, 1995. of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic
291. Skorski T, Kanakaraj P, Nieborowska-Skorska M, et al: progenitors. Immunity 3:147, 1995.
Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is 313. Gilliland DG, Griffin JD: The roles of FLT3 in hematopoiesis and
required for the growth of Philadelphia chromosome-positive cells. leukemia. Blood 100:1532, 2002.
Blood 86:726, 1995. 314. Gilliland DG: Molecular genetics of human leukemias: New insights
292. Skorski T, Bellacosa A, Nieborowska-Skorska M, et al: Transformation into therapy. Semin Hematol 39:6, 2002.
of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt- 315. Nakao M, Yokota S, Iwai T, et al: Internal tandem duplication of
dependent pathway. EMBO J 16:6151, 1997. the flt3 gene found in acute myeloid leukemia. Leukemia 10:1911,
293. Sattler M, Griffin JD: Molecular mechanisms of transformation by the 1996.
BCR-ABL oncogene. Semin Hematol 40:4, 2003. 316. Yamamoto Y, Kiyoi H, Nakano Y, et al: Activating mutation of D835
294. Skorski T: BCR/ABL regulates response to DNA damage: The role in within the activation loop of FLT3 in human hematologic malignan-
resistance to genotoxic treatment and in genomic instability. Oncogene cies. Blood 97:2434, 2001.
21:8591, 2002. 317. Paietta E, Ferrando AA, Neuberg D, et al: Activating FLT3 mutations
295. Ribeiro RC, Abromowitch M, Raimondi SC, et al: Clinical and biologic in CD117/KIT(+) T-cell acute lymphoblastic leukemias. Blood 104:558,
hallmarks of the Philadelphia chromosome in childhood acute lympho- 2004.
blastic leukemia. Blood 70:948, 1987. 318. Griffith J, Black J, Faerman C, et al: The structural basis for autoinhibi-
296. Crist W, Carroll A, Shuster J, et al: Philadelphia chromosome positive tion of FLT3 by the juxtamembrane domain. Mol Cell 13:169,
childhood acute lymphoblastic leukemia: Clinical and cytogenetic char- 2004.
acteristics and treatment outcome. A Pediatric Oncology Group study. 319. Kiyoi H, Ohno R, Ueda R, et al: Mechanism of constitutive activation
Blood 76:489, 1990. of FLT3 with internal tandem duplication in the juxtamembrane
297. Fletcher JA, Lynch EA, Kimball VM, et al: Translocation (9;22) is domain. Oncogene 21:2555, 2002.
associated with extremely poor prognosis in intensively treated children 320. Stirewalt DL, Radich JP: The role of FLT3 in haematopoietic malignan-
with acute lymphoblastic leukemia. Blood 77:435, 1991. cies. Nat Rev Cancer 3:650, 2003.
950.e8 Part VII Hematologic Malignancies

321. Markovic A, MacKenzie KL, Lock RB: FLT-3: A new focus in the 344. Parada LF, Tabin CJ, Shih C, et al: Human EJ bladder carcinoma
understanding of acute leukemia. Int J Biochem Cell Biol 37:1168, oncogene is homologue of Harvey sarcoma virus ras gene. Nature
2005. 297:474, 1982.
322. Brown P, Levis M, Shurtleff S, et al: FLT3 inhibition selectively kills 345. Shimizu K, Goldfarb M, Perucho M, et al: Isolation and preliminary
childhood acute lymphoblastic leukemia cells with high levels of FLT3 characterization of the transforming gene of a human neuroblastoma
expression. Blood 105:812, 2005. cell line. Proc Natl Acad Sci U S A 80:383, 1983.
323. Mullighan CG, Collins-Underwood JR, Phillips LA, et al: Rearrange- 346. Shimizu K, Goldfarb M, Suard Y, et al: Three human transforming
ment of CRLF2 in B-progenitor- and Down syndrome-associated acute genes are related to the viral ras oncogenes. Proc Natl Acad Sci U S A
lymphoblastic leukemia. Nat Genet 41:1243, 2009. 80:2112, 1983.
324. Yoda A, Yoda Y, Chiaretti S, et al: Functional screening identifies 347. Schubbert S, Shannon K, Bollag G: Hyperactive Ras in developmental
CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl disorders and cancer. Nat Rev Cancer 7:295, 2007.
Acad Sci U S A 107:252, 2010. 348. Murray MJ, Cunningham JM, Parada LF, et al: The HL-60 transform-
325. Hertzberg L, Vendramini E, Ganmore I, et al: Down syndrome acute ing sequence: A ras oncogene coexisting with altered myc genes in
lymphoblastic leukemia, a highly heterogeneous disease in which aber- hematopoietic tumors. Cell 33:749, 1983.
rant expression of CRLF2 is associated with mutated JAK2: A report 349. Janssen JW, Steenvoorden AC, Collard JG, et al: Oncogene activation
from the International BFM Study Group. Blood 115:1006, 2010. in human myeloid leukemia. Cancer Res 45:3262, 1985.
326. Tefferi A, Pardanani A: JAK inhibitors in myeloproliferative 350. Bos JL, Toksoz D, Marshall CJ, et al: Amino-acid substitutions at codon
neoplasms: Rationale, current data and perspective. Blood Rev 25:229, 13 of the N-ras oncogene in human acute myeloid leukaemia. Nature
2011. 315:726, 1985.
327. Puel A, Ziegler SF, Buckley RH, et al: Defective IL7R expression in 351. Gambke C, Signer E, Moroni C: Activation of N-ras gene in bone
T(-)B(+)NK(+) severe combined immunodeficiency. Nat Genet 20:394, marrow cells from a patient with acute myeloblastic leukaemia. Nature
1998. 307:476, 1984.
328. Noguchi M, Nakamura Y, Russell SM, et al: Interleukin-2 receptor 352. Hirai H, Tanaka S, Azuma M, et al: Transforming genes in human
gamma chain: A functional component of the interleukin-7 receptor. leukemia cells. Blood 66:1371, 1985.
Science 262:1877, 1993. 353. Padua RA, Carter G, Hughes D, et al: RAS mutations in myelodysplasia
329. Liu YJ, Soumelis V, Watanabe N, et al: TSLP: An epithelial cell cytokine detected by amplification, oligonucleotide hybridization, and transfor-
that regulates T cell differentiation by conditioning dendritic cell matu- mation. Leukemia 2:503, 1988.
ration. Annu Rev Immunol 25:193, 2007. 354. Perentesis JP, Bhatia S, Boyle E, et al: RAS oncogene mutations and
330. Zenatti PP, Ribeiro D, Li W, et al: Oncogenic IL7R gain-of-function outcome of therapy for childhood acute lymphoblastic leukemia. Leu-
mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet kemia 18:685, 2004.
43:932, 2011. 355. Liang DC, Shih LY, Fu JF, et al: K-Ras mutations and N-Ras mutations
331. Shochat C, Tal N, Bandapalli OR, et al: Gain-of-function mutations in childhood acute leukemias with or without mixed-lineage leukemia
in interleukin-7 receptor-alpha (IL7R) in childhood acute lymphoblas- gene rearrangements. Cancer 106:950, 2006.
tic leukemias. J Exp Med 208:901, 2011. 356. Wiemels JL, Zhang Y, Chang J, et al: RAS mutation is associated with
332. Ellisen LW, Bird J, West DC, et al: TAN-1, the human homolog of the hyperdiploidy and parental characteristics in pediatric acute lympho-
Drosophila notch gene, is broken by chromosomal translocations in T blastic leukemia. Leukemia 19:415, 2005.
lymphoblastic neoplasms. Cell 66:649, 1991. 357. Manning BD, Cantley LC: AKT/PKB signaling: Navigating down-
333. Maillard I, Fang T, Pear WS: Regulation of lymphoid development, stream. Cell 129:1261, 2007.
differentiation, and function by the Notch pathway. Annu Rev Immunol 358. Chalhoub N, Baker SJ: PTEN and the PI3-kinase pathway in cancer.
23:945, 2005. Annu Rev Pathol 4:127, 2009.
334. Radtke F, Wilson A, Mancini SJ, et al: Notch regulation of lymphocyte 359. Cully M, You H, Levine AJ, et al: Beyond PTEN mutations: The PI3K
development and function. Nat Immunol 5:247, 2004. pathway as an integrator of multiple inputs during tumorigenesis. Nat
335. Grabher C, von Boehmer H, Look AT: Notch 1 activation in the Rev Cancer 6:184, 2006.
molecular pathogenesis of T-cell acute lymphoblastic leukaemia. Nat 360. Gutierrez A, Sanda T, Grebliunaite R, et al: High frequency of PTEN,
Rev Cancer 6:347, 2006. PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia.
336. Zweidler-McKay PA, Pear WS: Notch and T cell malignancy. Semin Blood 114:647, 2009.
Cancer Biol 14:329, 2004. 361. Maser RS, Choudhury B, Campbell PJ, et al: Chromosomally unstable
337. Malecki MJ, Sanchez-Irizarry C, Mitchell JL, et al: Leukemia-associated mouse tumours have genomic alterations similar to diverse human
mutations within the NOTCH1 heterodimerization domain fall into cancers. Nature 447:966, 2007.
at least two distinct mechanistic classes. Mol Cell Biol 26:4642, 2006. 362. Palomero T, Sulis ML, Cortina M, et al: Mutational loss of PTEN
338. Chiang MY, Xu ML, Histen G, et al: Identification of a conserved induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med
negative regulatory sequence that influences the leukemogenic activity 13:1203, 2007.
of NOTCH1. Mol Cell Biol 26:6261, 2006. 363. Larson Gedman A, Chen Q, Kugel Desmoulin S, et al: The impact
339. Weng AP, Millholland JM, Yashiro-Ohtani Y, et al: c-Myc is an impor- of NOTCH1, FBW7 and PTEN mutations on prognosis and
tant direct target of Notch1 in T-cell acute lymphoblastic leukemia/ downstream signaling in pediatric T-cell acute lymphoblastic leukemia:
lymphoma. Genes Dev 20:2096, 2006. A report from the Childrens Oncology Group. Leukemia 23:1417,
340. Palomero T, Lim WK, Odom DT, et al: NOTCH1 directly regulates 2009.
c-MYC and activates a feed-forward-loop transcriptional network pro- 364. Gutierrez A, Grebliunaite R, Feng H, et al: Pten mediates Myc onco-
moting leukemic cell growth. Proc Natl Acad Sci U S A 103:18261, gene dependence in a conditional zebrafish model of T cell acute lym-
2006. phoblastic leukemia. J Exp Med 208:1595, 2011.
341. Sharma VM, Calvo JA, Draheim KM, et al: Notch1 contributes to 365. Gutierrez A, Look AT: NOTCH and PI3K-AKT pathways intertwined.
mouse T-cell leukemia by directly inducing the expression of c-myc. Cancer Cell 12:411, 2007.
Mol Cell Biol 26:8022, 2006. 366. Liu P, Cheng H, Roberts TM, et al: Targeting the phosphoinositide
342. Aster JC: Deregulated NOTCH signaling in acute T-cell lymphoblastic 3-kinase pathway in cancer. Nat Rev Drug Discov 8:627, 2009.
leukemia/lymphoma: New insights, questions, and opportunities. Int J 367. Seeger RC, Brodeur GM, Sather H, et al: Association of multiple copies
Hematol 82:295, 2005. of the N-myc oncogene with rapid progression of neuroblastomas.
343. Der CJ, Krontiris TG, Cooper GM: Transforming genes of human N Engl J Med 313:1111, 1985.
bladder and lung carcinoma cell lines are homologous to the ras genes 368. Lahortiga I, De Keersmaecker K, Van Vlierberghe P, et al: Duplication
of Harvey and Kirsten sarcoma viruses. Proc Natl Acad Sci U S A of the MYB oncogene in T cell acute lymphoblastic leukemia. Nat
79:3637, 1982. Genet 39:593, 2007.
Chapter 63 Pathobiology of Acute Lymphoblastic Leukemia 950.e9

369. Clappier E, Cuccuini W, Kalota A, et al: The C-MYB locus is involved 394. Quelle DE, Zindy F, Ashmun RA, et al: Alternative reading frames of
in chromosomal translocation and genomic duplications in human the INK4a tumor suppressor gene encode two unrelated proteins
T-cell acute leukemia (T-ALL)the translocation defining a new capable of inducing cell cycle arrest. Cell 83:993, 1995.
T-ALL subtype in very young children. Blood 110:1251, 2007. 395. Sidransky D: Two tracks but one race? Cancer genetics. Curr Biol 6:523,
370a. ONeil JE, Tchinda J, Gutierrez A, et al: Alu Elements Mediate MYB 1996.
Gene Tandem Duplication in Human T-ALL. J Exp Med 204:3059, 396. Kamb A, Gruis NA, Weaver-Feldhaus J, et al: A cell cycle regulator
2007. potentially involved in genesis of many tumor types. Science 264:436,
370b. Knudson AG: Mutation and cancer: Statistical study of retinoblas- 1994.
toma. Proc Natl Acad Sci U S A 68:820, 1971. 397. Nobori T, Miura K, Wu DJ, et al: Deletions of the cyclin-dependent
371. Nigro JM, Baker SJ, Preisinger AC, et al: Mutations in the p53 gene kinase-4 inhibitor gene in multiple human cancers. Nature 368:753,
occur in diverse human tumour types. Nature 342:705, 1989. 1994.
372. Li FP, Fraumeni JF, Jr, Mulvihill JJ, et al: A cancer family syndrome in 398. Zhang Y, Xiong Y, Yarbrough WG: ARF promotes MDM2 degradation
twenty-four kindreds. Cancer Res 48:5358, 1988. and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and
373. Malkin D, Li FP, Strong LC, et al: Germ line p53 mutations in a p53 tumor suppression pathways. Cell 92:725, 1998.
familial syndrome of breast cancer, sarcomas, and other neoplasms. 399. Pomerantz J, Schreiber-Agus N, Liegeois NJ, et al: The Ink4a tumor
Science 250:1233, 1990. suppressor gene product, p19Arf, interacts with MDM2 and neutralizes
374. Srivastava S, Zou ZQ, Pirollo K, et al: Germ-line transmission of a MDM2s inhibition of p53. Cell 92:713, 1998.
mutated p53 gene in a cancer-prone family with Li-Fraumeni syn- 400. Ashcroft M, Vousden KH: Regulation of p53 stability. Oncogene
drome. Nature 348:747, 1990. 18:7637, 1999.
375. Frebourg T, Friend SH: Cancer risks from germline p53 mutations. 401. Sharpless NE, Bardeesy N, Lee KH, et al: Loss of p16Ink4a with
J Clin Invest 90:1637, 1992. retention of p19Arf predisposes mice to tumorigenesis. Nature 413:86,
376. Levine AJ: p53, the cellular gatekeeper for growth and division. Cell 2001.
88:323, 1997. 402. Krimpenfort P, Quon KC, Mooi WJ, et al: Loss of p16Ink4a confers
377. Matlashewski G, Lamb P, Pim D, et al: Isolation and characterization susceptibility to metastatic melanoma in mice. Nature 413:83, 2001.
of a human p53 cDNA clone: Expression of the human p53 gene. 403. Hebert J, Cayuela JM, Berkeley J, et al: Candidate tumor-suppressor
EMBO J 3:3257, 1984. genes MTS1 (p16INK4A) and MTS2 (p15INK4B) display frequent
378. Zakut-Houri R, Bienz-Tadmor B, Givol D, et al: Human p53 cellular homozygous deletions in primary cells from T- but not from B-cell
tumor antigen: cDNA sequence and expression in COS cells. EMBO J lineage acute lymphoblastic leukemias. Blood 84:4038, 1994.
4:1251, 1985. 404. Quesnel B, Preudhomme C, Philippe N, et al: p16 gene homozygous
379. Baker SJ, Fearon ER, Nigro JM, et al: Chromosome 17 deletions deletions in acute lymphoblastic leukemia. Blood 85:657, 1995.
and p53 gene mutations in colorectal carcinomas. Science 244:217, 405. Haidar MA, Cao XB, Manshouri T, et al: p16INK4A and p15INK4B
1989. gene deletions in primary leukemias. Blood 86:311, 1995.
380. Baker SJ, Markowitz S, Fearon ER, et al: Suppression of human 406. Fizzotti M, Cimino G, Pisegna S, et al: Detection of homozygous dele-
colorectal carcinoma cell growth by wild-type p53. Science 249:912, tions of the cyclin-dependent kinase 4 inhibitor (p16) gene in acute
1990. lymphoblastic leukemia and association with adverse prognostic fea-
381. Funk WD, Pak DT, Karas RH, et al: A transcriptionally active DNA- tures. Blood 85:2685, 1995.
binding site for human p53 protein complexes. Mol Cell Biol 12:2866, 407. Rasool O, Heyman M, Brandter LB, et al: p15ink4B and p16ink4 gene
1992. inactivation in acute lymphocytic leukemia. Blood 85:3431, 1995.
382. el-Deiry WS, Kern SE, Pietenpol JA, et al: Definition of a consensus 408. Okuda T, Shurtleff SA, Valentine MB, et al: Frequent deletion of
binding site for p53. Nat Genet 1:45, 1992. p16INK4a/MTS1 and p15INK4b/MTS2 in pediatric acute lympho-
383. Farmer G, Bargonetti J, Zhu H, et al: Wild-type p53 activates transcrip- blastic leukemia. Blood 85:2321, 1995.
tion in vitro. Nature 358:83, 1992. 409. Hirama T, Koeffler HP: Role of the cyclin-dependent kinase inhibitors
384. Fields S, Jang SK: Presence of a potent transcription activating sequence in the development of cancer. Blood 86:841, 1995.
in the p53 protein. Science 249:1046, 1990. 410. Iolascon A, Faienza MF, Coppola B, et al: Homozygous deletions of
385. Kern SE, Kinzler KW, Bruskin A, et al: Identification of p53 as a cyclin-dependent kinase inhibitor genes, p16(INK4A) and p18, in
sequence-specific DNA-binding protein. Science 252:1708, 1991. childhood T cell lineage acute lymphoblastic leukemias. Leukemia
386. Kastan MB, Onyekwere O, Sidransky D, et al: Participation of p53 10:255, 1996.
protein in the cellular response to DNA damage. Cancer Res 51:6304, 411. Nakao M, Yokota S, Kaneko H, et al: Alterations of CDKN2 gene
1991. structure in childhood acute lymphoblastic leukemia: Mutations of
387. Hsiao MH, Yu AL, Yeargin J, et al: Nonhereditary p53 mutations in CDKN2 are observed preferentially in T lineage. Leukemia 10:249,
T-cell acute lymphoblastic leukemia are associated with the relapse 1996.
phase. Blood 83:2922, 1994. 412. Cayuela JM, Madani A, Sanhes L, et al: Multiple tumor-suppressor
388. Diccianni MB, Yu J, Hsiao M, et al: Clinical significance of p53 muta- gene 1 inactivation is the most frequent genetic alteration in T-cell acute
tions in relapsed T-cell acute lymphoblastic leukemia. Blood 84:3105, lymphoblastic leukemia. Blood 87:2180, 1996.
1994. 413. Takeuchi S, Bartram CR, Seriu T, et al: Analysis of a family of cyclin-
389. Wada M, Bartram CR, Nakamura H, et al: Analysis of p53 mutations dependent kinase inhibitors: p15/MTS2/INK4B, p16/MTS1/INK4A,
in a large series of lymphoid hematologic malignancies of childhood. and p18 genes in acute lymphoblastic leukemia of childhood. Blood
Blood 82:3163, 1993. 86:755, 1995.
390. Marks DI, Kurz BW, Link MP, et al: High incidence of potential p53 414. Heyman M, Rasool O, Borgonovo Brandter L, et al: Prognostic impor-
inactivation in poor outcome childhood acute lymphoblastic leukemia tance of p15INK4B and p16INK4 gene inactivation in childhood acute
at diagnosis. Blood 87:1155, 1996. lymphocytic leukemia. J Clin Oncol 14:1512, 1996.
391. Marks DI, Kurz BW, Link MP, et al: Altered expression of p53 and 415. Drexler HG: Review of alterations of the cyclin-dependent kinase
mdm-2 proteins at diagnosis is associated with early treatment failure inhibitor INK4 family genes p15, p16, p18 and p19 in human
in childhood acute lymphoblastic leukemia. J Clin Oncol 15:1158, leukemia-lymphoma cells. Leukemia 12:845, 1998.
1997. 416. ONeil J, Grim J, Strack P, et al: FBW7 Mutations in Leukemic Cells
392. Sherr CJ, Roberts JM: Inhibitors of mammalian G1 cyclin-dependent Mediate NOTCH Pathway Activation and Resistance to Gamma-
kinases. Genes Dev 9:1149, 1995. Secretase Inhibitors. J Exp Med 204:1813, 2007.
393. Kamijo T, Zindy F, Roussel MF, et al: Tumor suppression at the mouse 417. Inuzuka H, Shaik S, Onoyama I, et al: SCF(FBW7) regulates cellular
INK4a locus mediated by the alternative reading frame product apoptosis by targeting MCL1 for ubiquitylation and destruction.
p19ARF. Cell 91:649, 1997. Nature 471:104, 2011.
950.e10 Part VII Hematologic Malignancies

418. Mullighan CG, Goorha S, Radtke I, et al: Genome-wide analysis of myelogenous leukemia in a transgenic mouse model. Cancer Sci
genetic alterations in acute lymphoblastic leukaemia. Nature 446:758, 100:1219, 2009.
2007. 436. Zha S, Bassing CH, Sanda T, et al: ATM-deficient thymic lymphoma
419. Winandy S, Wu P, Georgopoulos K: A dominant mutation in the Ikaros is associated with aberrant tcrd rearrangement and gene amplification.
gene leads to rapid development of leukemia and lymphoma. Cell J Exp Med 207:1369, 2010.
83:289, 1995. 437. Su XY, Della-Valle V, Andre-Schmutz I, et al: HOX11L2/TLX3 is
420. Georgopoulos K, Bigby M, Wang JH, et al: The Ikaros gene is transcriptionally activated through T-cell regulatory elements down-
required for the development of all lymphoid lineages. Cell 79:143, stream of BCL11B as a result of the t(5;14)(q35;q32). Blood 108:4198,
1994. 2006.
421. Mullighan CG, Miller CB, Radtke I, et al: BCR-ABL1 lymphoblastic 438. Nagel S, Scherr M, Kel A, et al: Activation of TLX3 and NKX2-5 in
leukaemia is characterized by the deletion of Ikaros. Nature 453:110, t(5;14)(q35;q32) T-cell acute lymphoblastic leukemia by remote 3-
2008. BCL11B enhancers and coregulation by PU.1 and HMGA1. Cancer
422. Mullighan CG, Su X, Zhang J, et al: Deletion of IKZF1 and prognosis Res 67:1461, 2007.
in acute lymphoblastic leukemia. N Engl J Med 360:470, 2009. 439. Van Vlierberghe P, Palomero T, Khiabanian H, et al: PHF6 mutations
423. Harvey RC, Mullighan CG, Chen IM, et al: Rearrangement of CRLF2 in T-cell acute lymphoblastic leukemia. Nat Genet 42:338, 2010.
is associated with mutation of JAK kinases, alteration of IKZF1, 440. Sutcliffe MJ, Shuster JJ, Sather HN, et al: High concordance from
Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor independent studies by the Childrens Cancer Group (CCG) and Pedi-
acute lymphoblastic leukemia. Blood 115:5312, 2010. atric Oncology Group (POG) associating favorable prognosis with
424. van Noort M, Clevers H: TCF transcription factors, mediators of Wnt- combined trisomies 4, 10, and 17 in children with NCI Standard-Risk
signaling in development and cancer. Dev Biol 244:1, 2002. B-precursor Acute Lymphoblastic Leukemia: A Childrens Oncology
425. Gutierrez A, Sanda T, Ma W, et al: Inactivation of LEF1 in T-cell acute Group (COG) initiative. Leukemia 19:734, 2005.
lymphoblastic leukemia. Blood 115:2845, 2010. 441. Trueworthy R, Shuster J, Look T, et al: Ploidy of lymphoblasts is the
426. Yochum GS, Cleland R, Goodman RH: A genome-wide screen for strongest predictor of treatment outcome in B-progenitor cell acute
beta-catenin binding sites identifies a downstream enhancer element lymphoblastic leukemia of childhood: A Pediatric Oncology Group
that controls c-Myc gene expression. Mol Cell Biol 28:7368, 2008. study. J Clin Oncol 10:606, 1992.
427. He TC, Sparks AB, Rago C, et al: Identification of c-MYC as a target 442. Raimondi SC, Pui CH, Hancock ML, et al: Heterogeneity of hyper-
of the APC pathway. Science 281:1509, 1998. diploid (5167) childhood acute lymphoblastic leukemia. Leukemia
428. Brantjes H, Roose J, van De Wetering M, et al: All Tcf HMG box 10:213, 1996.
transcription factors interact with Groucho-related co-repressors. 443. Martinez-Climent JA: Molecular cytogenetics of childhood hemato-
Nucleic Acids Res 29:1410, 2001. logical malignancies. Leukemia 11:1999, 1997.
429. Li L, Leid M, Rothenberg EV: An early T cell lineage commitment 444. Belkov VM, Krynetski EY, Schuetz JD, et al: Reduced folate carrier
checkpoint dependent on the transcription factor Bcl11b. Science expression in acute lymphoblastic leukemia: A mechanism for ploidy
329:89, 2010. but not lineage differences in methotrexate accumulation. Blood
430. Wakabayashi Y, Watanabe H, Inoue J, et al: Bcl11b is required for dif- 93:1643, 1999.
ferentiation and survival of alphabeta T lymphocytes. Nat Immunol 445. Ito C, Kumagai M, Manabe A, et al: Hyperdiploid acute lymphoblastic
4:533, 2003. leukemia with 51 to 65 chromosomes: A distinct biological entity with
431. Ikawa T, Hirose S, Masuda K, et al: An essential developmental a marked propensity to undergo apoptosis. Blood 93:315, 1999.
checkpoint for production of the T cell lineage. Science 329:93, 446. Heerema NA, Nachman JB, Sather HN, et al: Hypodiploidy with less
2010. than 45 chromosomes confers adverse risk in childhood acute lympho-
432. Li P, Burke S, Wang J, et al: Reprogramming of T cells to natural killer- blastic leukemia: A report from the childrens cancer group. Blood
like cells upon Bcl11b deletion. Science 329:85, 2010. 94:4036, 1999.
433. Gutierrez A, Kentsis A, Sanda T, et al: The BCL11B tumor suppressor 447. Pui CH, Evans WE: Treatment of acute lymphoblastic leukemia. N Engl
is mutated across the major molecular subtypes of T-cell acute lympho- J Med 354:166, 2006.
blastic leukemia. Blood 118:4169, 2011. 448. Pui CH, Campana D, Evans WE: Childhood acute lymphoblastic
434. Kamimura K, Ohi H, Kubota T, et al: Haploinsufficiency of Bcl11b leukaemiacurrent status and future perspectives. Lancet Oncol 2:597,
for suppression of lymphomagenesis and thymocyte development. 2001.
Biochem Biophys Res Commun 355:538, 2007. 449. Rubnitz JE, Behm FG, Curcio-Brint AM, et al: Molecular analysis of
435. Nagamachi A, Yamasaki N, Miyazaki K, et al: Haploinsufficiency and t(11;19) breakpoints in childhood acute leukemias. Blood 87:4804,
acquired loss of Bcl11b and H2AX induces blast crisis of chronic 1996.

You might also like