Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

SYMPOSIUM ON ONCOLOGY PRACTICE: HEMATOLOGICAL MALIGNANCIES

CHRONIC MYELOID LEUKEMIA

Chronic Myeloid Leukemia: Diagnosis and Treatment

ALFONSO QUINTÁS-CARDAMA, MD, AND JORGE E. CORTES, MD

Chronic myeloid leukemia (CML) has become a model in research targeted therapies in cancer medicine. In this review, we
and management among malignant disorders. Since the discovery
of the presence of a unique and constant chromosomal abnormal- summarize the current knowledge of the molecular biology
ity slightly more than 40 years ago, substantial progress has been of CML and discuss the current treatment modalities, in-
made in the understanding of the biology of the disease. This cluding the important historical role of recombinant inter-
progress has translated into significant improvement in the long-
term prognosis of patients with this disease. This change came feron alfa, the development of imatinib mesylate and the
first with the use of stem cell transplantation and interferon alfa, new tyrosine kinase inhibitors (TKIs), stem cell transplan-
but recently it has opened the era of molecularly targeted thera- tation (SCT), and other novel therapeutic approaches still
pies. Imatinib, a potent and selective tyrosine kinase inhibitor,
may be the best example of our attempts to identify molecular in preclinical or early clinical development.
abnormalities and develop drugs directed specifically at them.
Furthermore, the understanding of at least some of the mecha-
nisms of resistance to imatinib has led to rapid development of EPIDEMIOLOGY AND ETIOLOGY
new agents that may overcome this resistance. The outlook today
for patients with CML is much brighter than just a few years ago. Chronic myeloid leukemia is a rare disease worldwide.
It is our hope that this fascinating journey in CML can be repli- Approximately 4600 new cases of CML were diagnosed in
cated in other malignancies. In this article, we review our current
understanding of this disease. 2004 in the United States, among 33,400 new cases of
leukemia. Hence, CML represents 14% of all leukemias
Mayo Clin Proc. 2006;81(7):973-988
and 20% of adult leukemias. The annual incidence is 1.6
ALL = acute lymphoblastic leukemia; AML = acute myeloid leukemia; cases per 100,000 adults,8 with a slight male preponderance
AP = accelerated phase; ATP = adenosine triphosphate; BP = blast (male-female ratio, 1.4:1). The median age at diagnosis
phase; CCGR = complete cytogenetic response; CHR = complete hema-
tologic response; CML = chronic myeloid leukemia; CP = chronic phase; is 65 years. In contrast, CML is exceedingly rare in chil-
Grb2 = growth factor receptor–bound protein 2; IC50 = inhibitory concen- dren, and its incidence increases with age.9 There are no
tration that results in a 50% reduction in proliferation; MCGR= major
cytogenetic response; MMR = major molecular response; MUD = known hereditary, familial, geographic, ethnic, or eco-
matched unrelated donor; Ph = Philadelphia; PI3K = phosphatidylinositol nomic associations with CML; therefore, the disease is nei-
3-kinase; RT-PCR = reverse transcriptase–polymerase chain reaction;
SCT = stem cell transplantation; STAT = signal transducer and activator ther preventable nor inherited. In most patients, the factor
of transcription; TKI = tyrosine kinase inhibitor; WBC = white blood cell responsible for the induction of the Ph chromosome is un-
known, although CML has been observed with increased
frequency in individuals exposed to the atom bomb explo-

C hronic myeloid leukemia (CML) is a clonal myelopro-


liferative disorder of a pluripotent stem cell1 first de-
scribed by John Hughes Bennett in 1845 at The Royal
sions in Japan in 1945, in radiologists, and in patients with
ankylosing spondylitis treated with radiation therapy.10

Infirmary of Edinburgh.2 It has been classified as a myelo-


CLINICAL PRESENTATION AND NATURAL HISTORY
proliferative disorder along with agnogenic myeloid meta-
plasia, polycythemia vera, and essential thrombocythe- Classically, 3 phases of disease progression are recognized
mia.3 Chronic myeloid leukemia was the first malignancy in CML: chronic phase (CP), accelerated phase (AP), and
that had a specific chromosomal abnormality uniquely blast phase (BP) (Table 1). Currently, nearly 90% of pa-
linked to it after the discovery of a minute chromosome tients with CML have their conditions diagnosed in the CP.
now known as the Philadelphia (Ph) chromosome,4 later Frequently, the diagnosis is made incidentally after a rou-
defined to result from a t(9;22) reciprocal chromosomal
translocation.5 Of critical importance was the demonstra- From the Department of Leukemia, The University of Texas, M. D. Anderson
tion that this translocation involved the ABL1 (Abelson) Cancer Center, Houston, Tex.
protooncogene in chromosome 9 and the BCR (breakpoint Dr Cortes has research grant support from Novartis, BMS, The Vaccine Com-
pany, ChemGenex Pharmaceuticals, Ltd, and Breakthrough Therapeutics.
cluster region) gene in chromosome 22.6,7 Since then, a
Address correspondence to Jorge E. Cortes, MD, Department of Leukemia,
constant stream of clinical and basic advances has made The University of Texas, M. D. Anderson Cancer Center, 1515 Holcombe Blvd,
CML one of the most extensively studied human malignan- Unit 428, Houston, TX 77030 (e-mail: jcortes@mdanderson.org). Individual
reprints of this article and a bound booklet of the entire Symposium on
cies. The introduction of rationally designed small mol- Oncology Practice: Hematological Malignancies will be available for purchase
ecules that target the tyrosine kinase activity of Bcr-Abl in from our Web site www.mayoclinicproceedings.com.
the treatment of CML has pioneered the development of © 2006 Mayo Foundation for Medical Education and Research

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 973

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

TABLE 1. Criteria for Accelerated Phase According to 3


Frequently Used Classifications*

MDACC11 IBMTR12 WHO13


Blasts (%) ≥15 ≥10 10-19†
Blasts and
promyelocytes (%) ≥30 ≥20 NA
Basophils (%) ≥20 ≥20‡ ≥20
Platelets (× 109/L) <100 Unresponsive increase or <100 or >1000 unresponsive
persistent decrease to treatment
Cytogenetics CE CE CE not at the time of diagnosis
White blood cell NA Difficult to control or NA
doubling in <5 d
Anemia NA Unresponsive NA
Splenomegaly NA Increasing NA
Other NA Chloromas, myelofibrosis Megakaryocyte proliferation,
fibrosis
*CE = clonal evolution; IBMTR = International Bone Marrow Transplant Registry; MDACC = M. D. Ander-
son Cancer Center; NA = not applicable; WHO = World Health Organization.
†Blast phase of 20% or more blasts (≥30% for MDACC and IBMTR).
‡Basophils and eosinophils.

tine complete blood cell count is performed for unrelated from CML-CP to CML-AP is usually subclinical, and labo-
reasons. This is due to the fact that patients with CML-CP ratory monitoring is necessary for detection of disease
have a competent immune system and may remain asymp- progression. The median survival for patients with CML-
tomatic for prolonged periods. When symptoms appear, they AP is 1 to 2 years.19 Most patients’ CML will remain in AP
are generally related to the expansion of CML cells and for 4 to 6 months before progressing to BP.20,21 Classic
consist typically of malaise, weight loss, and discomfort criteria define CML-BP by the demonstration of at least
caused by splenomegaly.14 Leukocytosis is a common fea- 30% of blasts in the peripheral blood or the bone marrow or
ture of CP, and the white blood cell (WBC) count can be as the presence of extramedullary blastic foci. The World
high as 1000 × 109/L, leading in rare instances to signs and Health Organization classification proposes a reduction
symptoms of hyperviscosity, such as retinal hemorrhage, from 30% or more to 20% or more blasts for the diagnosis
priapism, cerebrovascular accidents, tinnitus, confusion, of CML-BP. Clinically, most patients with CML-BP expe-
and stupor. After a median of 3 to 5 years, untreated pa- rience signs and symptoms related to increasing tumor
tients with CML-CP inevitably progress to CML-BP, an burden, including inability to control WBC counts with
aggressive form of acute leukemia highly refractory to previously stable doses of medication, marked constitu-
chemotherapy that is rapidly fatal.15 The risk of transforma- tional symptoms (fever, night sweats, anorexia, malaise,
tion to CML-BP is estimated at 3% to 4% per year.15,16 weight loss), splenic infarcts due to massive splenomegaly,
Chronic myeloid leukemia-AP is characterized by an in- bone pain, and increased risk of infections and bleeding.
creasing arrest of maturation that usually heralds transfor- The presence of cytogenetic abnormalities in addition to
mation to CML-BP. the Ph chromosome (ie, clonal evolution) involves most
Different criteria have been used to define CML-AP. frequently trisomy 8, isochromosome 17, and duplicate Ph
One commonly used classification defines AP as the pres- chromosome and has been considered a criterion of CML-
ence of at least 1 of the following hematologic features: AP. However, patients who are classified as having CML-
15% or more blasts, 30% or more blasts plus promyelo- AP based solely on the presence of clonal evolution appear
cytes, 20% or more basophils, or platelet counts lower than to fare better than those whose conditions were diagnosed
100 × 109/L unrelated to therapy.17 Of note, most classifica- on the basis of other clinical features. Immunopheno-
tion systems for CML-AP and CML-BP proposed through- typically, CML-BP can be characterized by myeloid or
out the years in the literature were developed before the lymphoid phenotype. In rare instances, blast cells can be
introduction of imatinib, and some, such as the recent biphenotypic with a mixed lymphoblastic-myeloblastic
World Health Organization proposal,13 have not been vali- lineage.19,22 Lymphoid CML-BP occurs in 20% to 30%
dated in clinical studies and therefore may lack clinical of patients, myeloid in 50%, and undifferentiated in
importance.18 Most discrepancies among different classi- 25%.22,23 Median survival of patients with lymphoid CML-
fications relate to the use of slightly different cutoffs for BP is 3 to 6 months, with patients with lymphoid CML-BP
hematologic values and other additional minor criteria. having a slightly better prognosis than those with myeloid
Although patients may become symptomatic, the transition phenotype.

974 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

LABORATORY AND PATHOLOGICAL FEATURES lymphoid CML-BP. Rare cases of megakaryoblastic,33


erythroblastic,34 and mastoblastic35 transformation have
Laboratory findings in CML include leukocytosis with a been reported.
remarkable left shift, basophilia, and eosinophilia. Platelet
count may be either high or low, and mild anemia is com-
PROGNOSTIC SYSTEMS
monly observed. The leukocyte alkaline phosphatase ac-
tivity is reduced, although phagocytic function remains Because of the difference in clinical aggressiveness be-
essentially normal.24 Low values may also be observed in tween the initial CP and advanced phases of CML and
agnogenic myeloid metaplasia, whereas activity may in- because the prognosis in CML can vary significantly even
crease with infection, clinical remission, or at the onset of among patients in the same phase of the disease, several
BP. The increase in the size of the WBC pool is reflected by risk stratification strategies have been developed in an
marked elevation of serum B12 levels and unsaturated B12- attempt to prognosticate and guide patient management.
binding capacity. During transformation to CML-BP, cir- The system most extensively used is the Sokal score, which
culating basophil levels, histamine levels, or both often was derived from 813 patients with CML collected from 6
increase, but the causative factors and importance of these European and American series in the 1960s and 1970s.36 In
phenomena remain unknown.25 this system, the hazard ratio for death is calculated from
The bone marrow of patients with CML is notoriously baseline patient and disease characteristics using the fol-
hypercellular and devoid of fat.26 In fact, it can be hypoth- lowing formula: λi(+)/λo(t) = Exp 0.0116 (age – 43.4) +
esized that the primary biologic defect in CML is not 0.0345 (spleen – 7.51) + 0.188 [(platelets/700)2 – 0.563] +
unregulated proliferation of leukemic stem cells but rather 0.0887 (blasts – 2.10). This scoring system establishes 3
discordant maturation, wherein a slight delay in cell matu- prognostic groups with hazard ratios of less than 0.8, 0.8 to
ration within the myeloid compartment results in increased 1.2, and more than 1.2 and median survivals of approxi-
myeloid mass.27 All stages of myeloid maturation are mately 2.5, 3.5, and 4.5 years, respectively.36 Of note, most
present, with predominance of myelocytes. In CP, the sum of the patients from whom the Sokal score was derived
of myeloblasts and promyelocytes usually accounts for less were treated with therapies currently not in use, such as
than 10% of the marrow cellularity. A decrease in apopto- busulfan, intensive chemotherapy, and splenectomy. How-
sis of myeloid cells contributes to the expansion of this ever, several studies performed in the 1990s have demon-
compartment and defective adherence of immature CML strated the applicability of this system in more modern
cells to marrow stromal cells in vitro,28 with subsequent series. Of 625 patients with CML-CP treated with chemo-
early release into the circulation. Megakaryocytes may be therapy in one study, 168 (27%) were high risk and had a 2-
increased, and Gaucher-like cells can be observed in 10% of year actuarial survival of 70% and a subsequent probability
cases.29 As CML progresses, varying degrees of reticulin of death of approximately 30% per year. In contrast, the
fibrosis may be seen,30 likely a result of the interaction low-risk group had a 2-year actuarial survival of 91% and a
between the proliferative clone of megakaryocytes and subsequent risk of death of approximately 17% per year.37
cytokines, such as platelet-derived growth factor, transform- The Sokal system has several limitations. First, in asymp-
ing growth factor β, and basic fibroblastic growth factor, tomatic patients whose disease is detectable only by mo-
whose plasma levels are significantly increased in CML.31 lecular testing, lead-time bias can be substantial relative to
Of note, these cytokines play an important role modulating those whose conditions are diagnosed after presenting with
angiogenesis, and characteristically the bone marrow from symptoms. Second, the use of more refined and potent
patients with CML shows the highest number of blood ves- therapeutic modalities in CML, such as imatinib, has been
sels and largest vascular area of all leukemias.31 associated with the loss of predictive value of some prog-
Blastic cells from patients with lymphoid CML-BP ex- nostic factors.38 Still, the Sokal score identifies patients
press terminal deoxynucleotidyl transferase, an enzyme with significantly different probabilities of response to
that catalyzes the polymerization of deoxynucleoside tri- imatinib, although patients with high-risk scores now have
phosphates and is mainly found in poorly differentiated B a much better outcome.
and T cells.32 In most cases of lymphoid CML-BP, blasts Other scoring systems have been proposed that may be
exhibit a B-cell immunophenotype, expressing CD10, more applicable to patients treated with interferon alfa. The
CD19, and CD22. In contrast, myeloid CML-BP closely best-characterized and most widely used is the Hasford
resembles acute myeloid leukemia (AML), and blasts stain score,39 which considers patient age, platelet count, periph-
with myeloperoxidase and express myeloid markers such eral blast count, spleen size, eosinophils, and basophils to
as CD13, CD33, and CD117. Frequently, myeloid markers determine risk. Some analyses have suggested that this
such as CD13 and CD33 may be expressed in patients with scoring system is less predictive among patients treated

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 975

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

with imatinib, but others have found it still useful. Finally, velopment of these molecular features and disease progres-
the Gratwohl score system has been developed specifically sion has not been established yet. In addition, other mo-
for patients undergoing SCT.40 lecular events, such as deletion or inactivation of p53,53
p16,54 and the retinoblastoma gene product55 have been
associated with disease transformation, but like overex-
CYTOGENETICS
pression of EVI-1,56 these are relatively infrequent and
A conclusive diagnosis of CML relies on cytogenetic and therefore not specific to CML-BP.57 Recently, it was sug-
molecular testing to identify the t(9;22)(q34.1;q11.21) and/ gested that progression of CML to BP probably involves
or the BCR-ABL1 hybrid gene, which is pathognomonic of several events in primitive progenitors, including BCR-
this disease.4,5 The Ph chromosome results from the bal- ABL1 amplification, acquisition of resistance to apoptosis,
anced translocation of cytogenetic material after a break on genomic instability, escape from innate and adaptive im-
chromosome 9 at band q34.1 that transposes the 3′ segment mune responses, and activation of β-catenin in granulo-
of the ABL1 gene to the 5′ segment of the BCR gene on cyte-macrophage progenitors, resulting in the acquisition
chromosome 22 at band q11.21.5 The Ph chromosome is of self-renewal capacity.58 Gene methylation has also been
detected in 95% of patients with CML and in 5% of chil- associated with progression in CML.59-61 The Pa promoter
dren and 15% to 30% of adults with acute lymphoblastic of ABL1 and the p15 promoter are hypermethylated in 81%
leukemia (ALL). In addition, approximately 2% of patients and 24% of patients, respectively. Interestingly, hyper-
with newly diagnosed AML can present with this cytoge- methylation of p15, but not of the Pa promoter, is associ-
netic abnormality.41 Occasionally, translocations that in- ated with disease progression. Methylation of the cadherin-
volve 3 or more chromosomes occur and are termed variant 13 gene occurs at an early stage in CML and is associated
Ph chromosome translocations.42,43 Although initially with high-risk features and lower probability of response to
thought to confer poor prognosis,44 recent data in patients interferon alfa.62
treated with imatinib have demonstrated that variant trans- Cytogenetic aberrancies have been reported occasion-
locations are associated with a similar prognosis compared ally in Ph chromosome–negative cells during treatment
with patients with the classic Ph chromosome.45 In 5% of with interferon alfa and, more recently, with imatinib.
patients with typical CML phenotype, the Ph chromosome These aberrancies do not represent clonal evolution be-
is not detectable but harbors the BCR-ABL1 rearrange- cause they occur in cells without the Ph chromosome and
ment.46 These patients have the same biology and outcome should not be considered as part of the definition of AP.
as those who express the Ph chromosome. Among the 5% The importance of these events is currently unknown.63 The
to 10% of patients with CML clinical features who do not most common such cytogenetic abnormalities include tri-
express the Ph chromosome, 30% to 50% have molecular somy 8, monosomy 5 or 7, and deletion 20q.64-66 These
evidence of the rearrangement involving BCR and ABL1. abnormalities frequently regress spontaneously, and it has
Those who do not express this molecular abnormality (ie, been suggested that patients with these abnormalities have
true Ph chromosome negative) are called atypical CML and similar long-term outcome as patients without such abnor-
have a different prognosis and treatment.47 The characteris- malities. However, occasional cases of progression to
tics and management of atypical CML are not covered any AML or myelodysplastic syndrome have been reported.
further in this article.
Little is known about the mechanisms involved in trans-
MOLECULAR BIOLOGY OF BCR-ABL1
formation to CML-BP, but the acquisition of additional
cytogenetic abnormalities, referred to as clonal evolution, The ABL1 gene is the human homologue of the v-abl
in Ph chromosome–positive cells has been thought to play oncogene harbored by the Abelson murine leukemia vi-
a prime role in CML progression. The most frequent sec- rus,67 and it encodes a nonreceptor tyrosine kinase (Figure
ondary cytogenetic abnormalities encountered in patients 1).68 In turn, BCR encodes a protein with serine–threonine
with clonal evolution are trisomy 8, isochromosome 17, kinase activity. The fusion of these 2 genes results in the
and duplicate Ph chromosome, which have been linked activation of the c-ABL protooncogene to its oncogenic
to c-Myc overexpression, loss of 17p, and BCR-ABL1 form.69,70 The breakpoints within the ABL1 gene at 9q34
overexpression, respectively.48-50 Other cytogenetic aber- map to an area spanning more than 300 kilobase (kb) at its
rancies, such as trisomy 19, trisomy 21, trisomy 17, and 5′ end occur upstream of exon Ib, downstream of exon Ia,
deletion 7, have been implicated in less than 10% of cases or, more frequently, between the two.71 Alternatively,
of clonal evolution.51,52 These genetic lesions are more breakpoints within BCR localize to 3 main breakpoint clus-
frequently associated with myeloid than with lymphoid ter regions. In most patients with CML and a third of those
CML-BP. Nonetheless, a clear correlation between the de- with Ph chromosome–positive ALL, the break occurs

976 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

FIGURE 1. Activation of signal transduction pathways by BCR/ABL. AKT = protein kinase B; ERK =
extracellular signal-regulated kinase; JAK = Janus kinase; MEK = mitogen-activated protein kinase
kinase; PI3K = phosphatidylinositol 3-kinase; SAPK = stress-activated protein kinase; STAT =
signal transducer and activator of transcription.

within a 5.8-kb area spanning BCR exons e12-e16 (for- esis to explain this phenomenon is that BCR-ABL1 might not
merly called b1-b5), referred to as the major breakpoint be the sole genetic lesion necessary for the development of
cluster region (M-bcr). Because of alternative splicing, CML. JunB is a component of the activator protein 1 family
a fusion transcript with either b2a2 or b3a2 junctions of transcription factors79 Modifications of JunB expression
can result and will give rise to a 210-kd hybrid protein may have a role in the pathogenesis of CML by modulating
(p210BCR-ABL).72,73 The clinical features and response to the initiation, progression, and maintenance of the myeloid
treatment are apparently similar for both transcripts. In differentiation program.80 A recent report in which trans-
two thirds of patients with Ph chromosome–positive ALL genic mice specifically lacking JunB in the myeloid lineage
and rarely in CML, the breakpoints within BCR localize (JunB–/–Ubi-JunB mice) develop a myeloproliferative disor-
to an area of 54.4 kb between exons e2′ and e2, termed the der that resembles human CML, including progression to
minor breakpoint cluster region (m-bcr), giving rise to an BP, seems to support the latter hypothesis.81
e1a2 messenger RNA that is translated to a 190-kd pro- Several biological models, such as BCR-ABL1–express-
tein (p190BCR-ABL). Patients with CML who carry this tran- ing CD34+ cells in culture82,83 or retrovirally transduced
script can present with marked monocytosis and may BCR-ABL1+ mouse cells,84 have demonstrated that BCR-
have a worse prognosis than those with the typical p210.74 ABL1 is an oncogene that promotes CML pathogenesis.
A third breakpoint cluster region (µ-bcr) downstream of Numerous pathways are activated in BCR-ABL1–express-
exon 19 has been identified, giving rise to a 230-kd fusion ing cells,85 and phosphorylation at the Y177 site of BCR is
protein (p230BCR-ABL) linked to cases of chronic neutrophilic essential for BCR-ABL1 leukemogenesis.86,87 This residue
leukemia.75 Although all 3 transcripts can induce a CML-like constitutes a high-affinity docking site for the SH2 domain
disorder in mice, p230BCR-ABL has reduced tyrosine kinase of growth factor receptor–bound protein 2 (Grb2). In turn,
activity relative to p190BCR-ABL76 and confers only partial Grb2 recruits SOS (a guanine-nucleotide exchanger of
growth factor independence, which parallels the milder clini- RAS) and the scaffold adapter Grb2-associated binding
cal course of chronic neutrophilic leukemia.75 All the protein 2 (GAB2) through its SH3 domain. Phosphoryla-
isoforms of Bcr-Abl have an active and an inactive configu- tion of GAB2 leads to recruitment of phosphatidylinositol
ration. Interestingly, using reverse transcriptase–polymerase 3-kinase (PI3K) and SHP2 (also known as PTPN11),
chain reaction (RT-PCR) with a sensitivity of approximately whereas SOS activates RAS.88 Mutation of Y177 to phenyl-
10–8, the BCR-ABL1 chimerism has been identified in up to alanine (Y177F) largely abolishes Grb2 binding and abro-
25% to 30% of presumably healthy adults.77,78 One hypoth- gates Bcr-Abl–induced RAS activation. Therefore, despite

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 977

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

preservation of the kinase activity of Abl, the Y177F mutant The achievement of a CCGR is associated with improved
impedes the transformation of primary bone marrow cul- survival, with 78% of patients who achieved a CCGR alive
tures.86 In an SCT model of CML, BCR-ABL1Y177F has a at 10 years, establishing the achievement of a CCGR as the
greatly reduced ability to induce a myeloproliferative dis- therapeutic goal in the interferon alfa era.106 Furthermore,
order in mice.89 Of note, SHP2 is required for normal approximately 30% of patients in CCGR also had undetect-
activation of the RAS extracellular signal-regulated ki- able disease by PCR (ie, complete molecular remission);
nase pathway that most receptor tyrosine kinases signal none of them has relapsed after a median follow-up of 10
through.88 Bcr-Abl also phosphorylates the Src kinases years and are therefore probably cured.106,107 Interestingly,
Lyn, Hck, and Fgr. Once phosphorylated, Hck activates 40% to 60% of those in CCGR with the presence of mini-
signal transducer and activator of transcription (STAT) mal residual disease at the molecular level have not re-
5.90,91 Importantly, overexpression of Hck/Lyn has been lapsed after 10 years. This has been attributed to interferon
linked with disease progression, particularly with a lym- alfa–induced immune modulation, such as the presence of
phoid phenotype.92 Therefore, Bcr-Abl promotes hemato- cytotoxic T lymphocytes specific for PR1, a peptide de-
poietic cell transformation mainly through RAS, PI3K, and rived from proteinase 3, which is overexpressed in CML
STAT 5 activation. All these elements have been shown to cells. This phenomenon has been demonstrated in patients
up-regulate the expression of cyclin D1, thus inducing cell- in complete remission after interferon alfa therapy and SCT
cycle progression from G1 to S phase.93,94 When BCR- but not in those who fail to achieve CCGR or those treated
ABL1–positive K562 cells were induced to express domi- with chemotherapy.108
nant negative forms of RAS, PI3K, or STAT 5, marked
apoptosis was observed in cells coexpressing 2 of 3 domi- STEM CELL TRANSPLANTATION
nant negative mutants in any combination.95 Notably, Bcr- Stem cell transplantation remains an important treatment
Abl also enhances SET expression, particularly during pro- option for patients with CML, particularly younger indi-
gression to BP.96 SET is a nucleus/cytoplasm-localized viduals with HLA-identical siblings. Initially, SCT was
phosphoprotein that potently inhibits the tumor suppressor thought to render the best outcomes when performed
protein phosphatase 2A (PP2A). In turn, PP2A is a phos- within 12 months from CML diagnosis. It has been re-
phatase that regulates cell proliferation, survival, and dif- ported that SCT performed within the first 24 months, and
ferentiation.97 Some of the elements involved in Bcr-Abl in some reports within the first 36 months, from diagnosis
signal transduction have been sought as therapeutic targets may have similar outcomes.40 Although it was initially
in CML. suggested that prior interferon alfa therapy adversely af-
fected the outcome after SCT,109 subsequent trials demon-
strated that this adverse effect was nonexistent,110-113 and
TREATMENT
current evidence suggests that prior exposure to imatinib
The treatment of CML has experienced a great degree of does not affect the outcome either.114,115 Current registry
refinement during the past few years. Until recently, inter- data revealed that the post-SCT mortality within the first
feron alfa and SCT were the only therapeutic options for year is 10% to 20% even in patients who underwent trans-
patients with CML. Currently, SCT still remains a valid plantation in optimal conditions (ie, HLA-matched sibling,
option, particularly for patients who do not respond appro- first CP <1 year from diagnosis, age <40 years). In addi-
priately to TKIs, and still remains a potentially curative tion, only one third to one half of patients may have a
therapeutic modality. However, the explosion of the field of suitable HLA-matched sibling, and stringent inclusion cri-
molecularly targeted TKIs in recent years led by imatinib teria, such as age, adequate organ function, and perfor-
mesylate has brought about a change in the natural history of mance status, may preclude the possibility of SCT in a
the disease and in the therapeutic approach to CML. large proportion of patients. A strategy to overcome the
lack of suitable donors is the use of matched unrelated
INTERFERON donors (MUDs).116 Unfortunately, the survival rate of pa-
Recombinant human interferon alfa has shown significant tients undergoing MUD SCT is still lower when compared
antitumor and immunomodulatory activity in the treatment with those receiving grafts from HLA-matched siblings.
of CML.98,99 Major cytogenetic responses (MCGRs) (<35% The outcome of MUD SCT may be improved by careful
Ph chromosome–positive cells) have been reported in up to molecular typing, particularly for HLA-A, HLA-B, and
40% of patients and complete cytogenetic responses HLA-DRB1.116 However, the use of stringent typing crite-
(CCGRs) (0% Ph chromosome–positive cells) in up to ria is inevitably linked to a lower probability of finding an
25%.100 The addition of cytarabine resulted in a significantly adequate donor. In addition, SCT is fraught with risks such
higher probability of a CCGR in up to 35% of patients.101-105 as graft-vs-host disease, veno-occlusive disease, life-

978 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

threatening infections, risk of secondary malignancy, and sion in up to 70% of patients who relapse in the CP,
poorer overall quality of life.117 Also, the risk of late relapse particularly when administered at the time of molecular
is being increasingly recognized. A recent series analyzed relapse. Alternatively, imatinib can be used in the post-
the long-term follow-up of 89 patients who underwent SCT relapse setting and induces CCGR in more than 40%
transplantation at a single institution.118 After more than 10 of patients treated in the CP.124
years of follow-up, 28 patients (31%) were alive. The mean
time to hematologic or cytogenetic relapse was 7.7 years, IMATINIB MESYLATE
with 5 patients relapsing more than 10 years after SCT. In a Imatinib mesylate (Gleevec) is an orally bioavailable 2-
recent study by the Seattle group, 131 patients with early phenylaminopyrimidine with targeted inhibitory activity
CML-CP with a median age of 43 years (range, 14-66 against the constitutively active tyrosine kinase of the Bcr-
years) received targeted busulfan and cyclophosphamide as Abl chimeric fusion protein. In addition, imatinib inhibits
a conditioning regimen. At 3 years, the projected non- other kinases, such as c-Kit, platelet-derived growth factor
relapse mortality rate was 14%, and 78% were projected to α and β, and Abl-related gene (ARG).125,126 Imatinib has
be alive and free of disease. However, 60% of survivors become the standard therapy for CML because of its re-
had extensive chronic graft-vs-host disease 1 year after markable activity and mild toxicity profile. In a phase 1
transplantation, although only 10% had a Karnofsky score dose-finding study in patients who were intolerant to or in
less than 80%. Notably, 11% of patients had minimal re- whom interferon alfa–based therapy failed, daily doses of
sidual disease documented by PCR but had not relapsed at 25 to 1000 mg were investigated. Responses among pa-
the time of the report.119 In contrast, patients with advanced tients receiving doses lower than 250 mg/d were rare, in
phase CML who undergo SCT have a 5-year survival prob- contrast to 98% of patients receiving a dose of at least 300
ability of 40% to 60% in the AP and 10% to 20% in the BP. mg/d who achieved a complete hematologic response
Notably, the long-term outcome of patients with CML-BP (CHR), including 54% who attained a cytogenetic re-
who achieve a second CP and who are undergoing SCT sponse. Of note, no dose-limiting toxicity was identified,
appears to be similar to that of patients with CML-AP who and the maximum tolerated dose was not reached.127 A
undergo transplantation. dosage of 400 mg/d was selected for a subsequent phase 2
The long-term disease-free survival rate after MUD study, including 454 patients with late CP disease who
SCT for young patients in the early CP stage of disease is were intolerant to or in whom interferon alfa therapy
57% compared with 67% in patients receiving grafts from failed.128 Recently updated data showed that 96% of pa-
HLA-matched siblings.120 The incorporation of molecular tients had achieved a CHR and 55% a CCGR. After a
matching in recent years has decreased the rate of graft-vs- median follow-up of 5 years, the overall survival rate was
host disease and improved the probability of long-term 79%.128 This rate is in keeping with results from a recent
survival in patients undergoing MUD SCT. single-institution study in which among 261 patients
Reduced-intensity conditioning regimens have been treated, 73% obtained a MCGR and 63% a CCGR.129 After
used as a means to make SCT available to a broader num- a median follow-up of 45 months, 86% of patients are
ber of patients who otherwise would be ineligible for stan- alive, of whom 80% are free of progression. More than
dard SCT. This approach takes advantage of the ability of 90% of patients who achieved a CCGR maintained a major
the T cells harbored in the graft to eliminate the CML cells response. The BCR-ABL1/ABL1 ratio by nested PCR was
(graft-vs-leukemia effect) as opposed to obtaining this ef- less than 0.05% in 31% and undetectable in 15% of pa-
fect by using ablative cytotoxic chemotherapy. This strat- tients.129 In the prospective, multicenter phase 3 Interna-
egy increases tolerability and decreases morbidity and tional Randomized Study of Interferon and STI571, the
mortality significantly. Long-term results with reduced- efficacy of imatinib was compared with the combination of
intensity conditioning regimens are not yet available, but interferon alfa and low-dose cytarabine in patients with
early results in small series (frequently including still newly diagnosed CML-CP. 130 Patients treated with
mostly young patients) report disease-free survival as high imatinib (n=553) or interferon alfa and low-dose cytarabine
as 85% at 70 months.121 In contrast, a recent study from the (n=553) were allowed to cross over to the alternative group
European Bone Marrow Transplant Registry reports a 3- if treatment failure or intolerance was demonstrated.131
year overall survival rate of 58% and a progression-free Imatinib therapy was superior to interferon alfa plus low-
survival rate of 37%.122 Patients with minimal residual dose cytarabine regarding hematologic and cytogenetic re-
disease by PCR 12 months after SCT have a risk of relapse sponses, tolerability, and transformation to CML-AP. After
of 30% to 40% compared with less than 5% for those with a median follow-up of 54 months, the CHR, MCGR, and
negative PCR results.123 However, relapse can be fre- CCGR rates were 98%, 92%, and 86%, respectively.132
quently managed successfully with donor lymphocyte infu- Progression-free survival is estimated to be 84%, and only

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 979

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

1.0

Year Total Dead


95%
Imatinib 230 7
0.8 1900-2000 960 334
1982-1989 365 265
1975-1981 132 127
Proportion surviving

1965-1974 123 122


0.6

0.4

0.2

0.0
0 2 4 6 8 10 12 14 16
Time from referral (y)

FIGURE 2. Survival of patients with early chronic phase chronic myeloid leukemia treated at M. D.
Anderson Cancer Center in different eras compared with those treated with imatinib.

6% of patients have progressed to AP or BP. The overall study was based primarily on the excellent responses ob-
annual progression rate has declined to 1.5% in the fourth tained when doses higher than 300 mg/d were used, it has
year of therapy, compared with 4.8% and 7.5% in the been suggested that imatinib at higher dosages (600-800
previous 2 years, suggesting that disease progression may mg/d) might result in improved outcomes (Table 2). In one
plateau in the ensuing years. The depth of the response after study, 36 patients with CML-CP in whom interferon alfa
12 months of imatinib therapy has important implications therapy failed were treated with imatinib 400 mg twice
regarding disease progression and relapse. In fact, in 97% daily, resulting in a CCGR rate of 90%, compared with
of patients who had a 3-log or greater reduction in BCR- 48% in historical matched controls treated with standard-
ABL transcript levels (ie, major molecular response dose therapy.141 In addition, 56% of patients had an MMR,
[MMR]) at 12 months, the probability of remaining free of including 41% with undetectable levels. The toxicity pro-
progression to AP or BP was 97% at 54 months compared file was similar to that of imatinib, 400 mg, although there
with 89% for patients in CCGR but not in MMR and 72% was a higher rate of grade 3 or higher myelosuppression.
for patients who did not achieve a CCGR at 12 months Several single-arm, phase 2 studies have used high-dose
(P=.001).132,133 In addition, the median BCR-ABL levels imatinib as the starting dose for patients with previously
continue to decrease, with a mean log reduction at 48 untreated CML-CP. These studies have documented a
months of 3.4 compared with 3.0 at 12 months.134 Although higher rate of CCGR of up to 95% and higher rates of
this study did not document a survival advantage compared molecular responses, particularly responses at the level of a
with the interferon alfa arm because of the crossover de- 4-log or greater reduction in transcript levels. In addition,
sign, studies comparing survival of patients treated with responses occur significantly earlier with high-dose com-
imatinib with historical cohorts treated with interferon pared with standard-dose therapy.137-139 One study reported
alfa–based therapy demonstrate the anticipated survival that by multivariate analysis, only treatment with high-dose
advantage135,136 (Figure 2). These results suggest that a trial imatinib (P=.02) was associated with achievement of an
of imatinib must be pursued in every patient before consid- MMR and that achieving an MMR within the first 12
ering SCT. months of therapy is predictive of a durable cytogenetic
The results from the International Randomized Study of remission.142 Similarly, patients receiving the highest dose
Interferon and STI571 have established imatinib 400 mg/d, intensity during the first year of therapy have the highest
as the standard therapy for CML. However, because the probability of achieving a major molecular remission at 24
imatinib dosage selected after the dose-finding phase 1 months (89%), whereas those receiving a median daily

980 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

dosage less than 600 mg had a significantly lower probabil- TABLE 2. Summary of Studies Using High-Dose Imatinib
as Frontline Therapy for Early Chronic Phase
ity both at 12 and at 24 months.143 Thus, these studies Chronic Myeloid Leukemia*
suggest that high-dose imatinib may provide more and
Dosage No. of CGCR MMR CMR
better remissions, but the results from ongoing randomized Study (mg/d) patients (%) (%) (%)
studies will determine whether high-dose imatinib provides
MDACC137 800 175 90 61 32
only faster responses or whether these translate into pro- TIDEL138 600→800 41 90 44 25
longed progression-free and/or overall survival. RIGHT139 800 114 85 64 44
Duration of Imatinib Therapy. Currently, no evidence GIMEMA140 800 100 88 60 NA
exists to support the belief that patients taking imatinib can *CGCR = complete cytogenetic remission; CMR = complete molecular
safely discontinue therapy once they achieve a complete remission (BCR-ABL1 undetectable); GIMEMA = Gruppo Italiano
Malattie Ematologiche dell’Adulto; MDACC = M. D. Anderson Cancer
molecular response. Most patients who have discontinued Center; MMR = major molecular response (BCR-ABL1/ABL1 <0.05% or
imatinib therapy have rapidly experienced both molecular 3-log reduction); RIGHT = Rationale and Insight on Gleevec [imatinib]
and cytogenetic relapse, even when some had sustained un- High-Dose Therapy; TIDEL = Therapeutic Intensification in de novo
leukemia.
detectable levels of BCR-ABL for significant periods.144-146
Recently, Rousselot et al147 described 8 patients who dis-
continued use of imatinib after maintaining undetectable monitoring for most patients. However, the lack of consis-
BCR-ABL levels for 24 months, 4 of whom still tested tency in reporting BCR-ABL transcript levels has been a
PCR negative after a follow-up of 8, 9, 12, and 13 months, source of intense debate. A recent proposal aimed at the
respectively. All these patients had been treated with inter- standardization of RT-PCR techniques and result reporting
feron alfa, suggesting that interferon alfa immunomodula- will greatly help this objective. According to this proposal,
tory effects may account for sustained and prolonged mo- results will be converted by comparing analysis of standard-
lecular responses observed on therapy discontinuation. ized reference samples with a value of 0.1% corresponding
It has been suggested that the most primitive quiescent to MMR in all laboratories.155 After the patient has achieved
leukemic progenitors are insensitive to imatinib in vitro a complete cytogenetic remission, RT-PCR should be per-
and are responsible for CML relapse once the inhibitory formed every 3 months and a routine cytogenetic analysis
pressure of imatinib is removed.148 Emerging data suggest every 12 months.155 Fluorescence in situ hybridization in
that resistance of quiescent cells to imatinib is indeed a peripheral blood specimens can be used to monitor the cyto-
Bcr-Abl–dependent phenomenon. 149 Other potential genetic responses among cytogenetic analyses. In addition,
mechanisms implicated in quiescent CML progenitors in- approximately 5% of patients who obtain a cytogenetic re-
clude expression of drug transport proteins such as PgP and sponse develop clonal karyotypic abnormalities in Ph chro-
Abcg2, atypical kinase domain mutations,150 and BCR- mosome–negative cells, primarily consistent with those en-
ABL overexpression.151 countered in myelodysplastic syndromes, although only rare
Monitoring of Imatinib Therapy and Minimal Re- cases of myelodysplastic syndrome or AML have been re-
sidual Disease. The significant MMR rate observed with ported.63 Although the long-term implications of these ab-
imatinib and its prognostic implications152,153 have led to the normalities are unknown, the risk of transformation to AML
redefinition of the therapeutic goals in CML. Rather than warrants careful follow-up of these patients.
aiming solely at achieving a CCGR,106,154 modern CML Imatinib Resistance. A subset of patients with CML
therapy should aim for the achievement of molecular re- will exhibit either primary or secondary resistance to
sponses. The long-term importance of molecular monitor- imatinib. Primary resistance refers to patients never re-
ing in CML has been best exemplified by patients under- sponding to imatinib, whereas secondary resistance occurs
going SCT, in whom detection of BCR-ABL transcripts, when a patient who had an initial response to imatinib
particularly at high levels and 6 months after transplanta- eventually loses the response. Among patients treated in
tion, confer a significantly higher risk of relapse. In these CP, the rate of resistance has been estimated to be less than
instances, prompt intervention (eg, donor lymphocyte infu- 2% per year. Several mechanisms of resistance have been
sion) has been effective. As mentioned earlier, among pa- reported, many of them mostly in vitro or in selected pa-
tients who achieve a CCGR with interferon alfa, the risk of tient samples, and their individual contribution to this phe-
relapse is minimal for those with low transcript levels.107 nomenon has not been completely defined. The most fre-
Patients who achieve undetectable BCR-ABL transcripts quently identified mechanism of resistance is the develop-
by nested PCR have all had sustained cytogenetic remis- ment of mutations in the Abl tyrosine kinase domain. More
sions beyond 10 years.106 In the imatinib setting, once a than 40 different BCR-ABL point mutations have been
CCGR is attained, quantitative RT-PCR in peripheral reported thus far, and new ones continue to be described.156
blood samples at 3-month intervals is the mainstay of The region of the Abl kinase where mutations occur is

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 981

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

TABLE 3. Results With New Tyrosine Kinase Inhibitors After Failure of Imatinib
in All Phases of CML*
Hematologic response Cytogenetic response
(%) (%)
No. of
Agent and CML phase patients Overall Complete Overall Complete
Phase 1
170
Nilotinib
CP 17 92 92 53 35
CE 10 100 100 90 20
AP 46 72 46 48 13
MyBP 24 42 8 29 4
LyBP 9 33 … 22 11
Dasatinib171,172
CP 40 93 93 63 35
AP 11 81 45 36 18
MyBP 23 61 35 52 26
LyBP 10 80 70 90 30
Phase 2
Dasatinib
CP173 186 90 90 45 33
AP174 107 59 33 36 21
MyBP175 74 51 24 42 27
LyBP and Ph+ ALL176 78 40 26 54 46
*AP = accelerated phase; CE = clonal evolution; CML = chronic myeloid leukemia; CP = chronic
phase; LyBP = lymphoid blast phase; MyBP = myeloid blast phase; Ph+ ALL = Philadelphia
chromosome–positive acute lymphoblastic leukemia.

critical in their ability to confer varying degrees of insensi- tations have been described mapping to the catalytic do-
tivity to imatinib. In fact, ranges of 220 to more than 5000 main and the activation loop of Abl, impairing the ability of
nM/L in the inhibitory concentration that results in a 50% the kinase to adopt a close (inactive) spatial conformation
reduction in proliferation (IC50) have been described in the necessary to bind to imatinib.167
various Bcr-Abl mutants.157 The most frequent mutations Despite the poor prognosis conferred by the T315I mu-
are those that map to the P-loop region of the kinase do- tant, in vitro and clinical data suggest that other mutations
main.157-159 The P-loop region is a glycine-rich structure that may be clinically relevant.168 Furthermore, other mecha-
serves as a docking site for phosphate groups of adenosine nisms of resistance, such as overexpression or amplification
triphosphate (ATP). G250E, Q252H, and the highly of the BCR-ABL message and/or protein, may influence the
imatinib-insensitive Y253F and E255K mutations locate in outcome of patients treated with imatinib. Recently, it was
this region. Despite not being directly involved in imatinib shown that activation of Src kinases and NF-κB may play a
binding, these residues have been linked to poor prognosis role in imatinib resistance in some patients.92,169
in some series, with a median survival of 4 to 5 months.160-163
Other series have refuted this notion.164 The difference in TREATMENT STRATEGIES FOR THE FUTURE
these series may be due to several factors, including patient New Bcr-Abl Inhibitors. Several strategies are cur-
selection and the criteria used to select patients for screen- rently being developed to overcome imatinib resistance
ing for mutations. In addition, the first studies that sug- (Table 3). New Abl TKIs have been developed with
gested a poor outcome for patients with P-loop mutations enhanced potency and less stringent binding requirements
did not address the management of patients after develop- and, in some cases, with inhibitory activity against other
ing mutations, and it is now clear that several treatment kinases involved in mechanisms of resistance to imatinib.
strategies can overcome resistance through mutations.165 In addition, new agents that block Bcr-Abl in a non–ATP-
Also, considering that individual mutations have different competitive manner may represent an alternative for
levels of resistance to imatinib and other agents, it is more patients who develop imatinib-insensitive Bcr-Abl kinase
appropriate to refer to individual mutations rather than mutations.
grouping them.155 The imatinib-insensitive T315I mutation Nilotinib (AMN107). Nilotinib is a phenylamino-py-
occurs in a highly conserved “gatekeeper” threonine resi- rimidine derivative with a 20- to 30-fold increased potency
due near the Bcr-Abl catalytic domain, thus leading to compared with imatinib against Bcr-Abl. Crystallographic
steric interference with imatinib binding.161,162,166 Other mu- models suggest that this increased potency is a result of

982 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

nilotinib representing a better topographical fit for the Abl nal toxic effects seen in some patients. Pleural effusions
kinase pocket. However, nilotinib, like imatinib, only binds have also been reported in 10% to 15% of patients, particu-
the Abl protein in its inactive conformation. Nilotinib larly those in BP, and it is usually grade 1 or 2. Despite the
inhibits the tyrosine kinase activity of most clinically impressive results achieved with dasatinib, it may not
important Bcr-Abl mutants, with the exception of T315I.157 eradicate all quiescent leukemic stem cells, although it may
A phase 1/2 study of nilotinib included 106 patients with be more effective at this level than imatinib.149 Another
imatinib-resistant CML in all phases and 13 patients with Ph dual Abl/Src kinase inhibitor termed SKI-606179 is current-
chromosome–positive ALL.170 Patients received nilotinib at ly being evaluated in phase 1 studies, and others, such as
dosages ranging from 50 to 1200 mg/d. In CML, hematolog- NS-187 (INNO406),180 AZD0530,181 PD166326,182 and
ic response rates ranged from 44% to 89%, and cytogenetic PD180970,183 may soon follow.
responses ranged from 22% in lymphoid and myeloid BP to Non–ATP-Competitive Bcr-Abl Inhibitors. Imatinib,
29% in AP and 50% in CP. Among patients with Bcr-Abl nilotinib, and dasatinib are ATP-competitive inhibitors and
mutations before nilotinib therapy, 60% had a hematologic therefore amenable to being affected in their ability to bind
response, and 41% had a cytogenetic response. Nilotinib to the Bcr-Abl kinase by the T315I mutation, which is
dosage was escalated up to 1200 mg/d with good tolerance, considered the “gatekeeper” of the kinase domain. Com-
and the maximum tolerated dose was estimated at 600 mg pounds that target binding sites unrelated to the ATP-
twice daily. The most common drug-related adverse events kinase domain may overcome this problem. ON012380
were gastrointestinal, rash, and hematologic. Importantly, targets the substrate-binding site of Bcr-Abl, competing
greater exposure to the drug and inhibition of Bcr-Abl with natural substrates such as Crkl but not with ATP.184 In
phosphorylation were observed with twice-daily dosing fact, ON012380 causes regression of leukemia induced by
schedules compared with equivalent total doses admin- intravenous injection of 32DcI3 cells that express the Bcr-
istered on a once-daily schedule.170 Abl mutant T315I.184 BIRB796, a p38 mitogen-activated
Dasatinib (BMS-354825). Because it is possible that protein kinase inhibitor, binds with excellent affinity to the
inhibition of Bcr-Abl alone may not be sufficient to eradi- Bcr-Abl T315I mutant (Kd 40 nM), although high concen-
cate all CML cells, particularly the leukemic imatinib- trations of this compound are necessary to inhibit
insensitive quiescent stem cells, agents with additional in- autophosphorylation of this mutant in Ba/F3 cells (IC50, 1-2
hibitory capacity against other kinases have been investi- µM).159 In this regard, VX680 (MK-0457), a multikinase
gated. Since Src kinases have been implicated in CML inhibitor that inhibits, among others, aurora A and B ki-
pathogenesis and progression, dual Abl/Src inhibitors may nase, seems to have a more favorable profile against T315I,
have enhanced activity compared with imatinib. Among although remarkably less affinity for wild-type Bcr-Abl
this new class of compounds, dasatinib has reached the (IC50, >10 µM).159
furthest level of clinical development. Dasatinib is an ATP- Other Therapeutic Options. Other approaches are be-
competitive, dual-specific Src- and Abl-kinase inhibitor ing developed for patients who develop resistance to
with 100- to 300-fold higher potency against Bcr-Abl com- imatinib. Farnesyl transferase inhibitors, such as lonafarnib
pared with imatinib and significant activity against c-kit and tipifarnib, have demonstrated activity both as single
(IC50, 5 nM), platelet-derived growth factor receptor β agents and in combination with imatinib.185-188 This ap-
(IC50, 28 nM), and Hck, Fyn, Src, and Lck kinases (IC50, proach has rendered anecdotal responses in patients har-
approximately 0.5 nM).177 However, dasatinib does not in- boring the T315I mutant. Homoharringtonine is a Cephalo-
hibit the T315I mutant, even in the presence of micromolar taxus alkaloid that represented the best therapeutic option
concentrations.157 In phase 2 studies, dasatinib was admin- in patients in whom interferon alfa–based therapies failed
istered at 70 mg twice daily to patients with CML in all before the introduction of imatinib in clinical practice.189 Its
phases after imatinib failure. A CHR was achieved by 87%, potential as salvage therapy in imatinib-resistant CML has
66%, 55%, and 46% of patients in CP, AP, myeloid BP, rekindled interest in this drug. Another avenue that has
and lymphoid BP/Ph chromosome–positive ALL, respec- been pursued in recent years is the use of hypomethylating
tively. Among patients in CP, 44% had a CCGR, whereas agents190,191 and histone deacetylase inhibitors.192-194 In a
46% to 66% of patients with advanced-phase CML had phase 2 study, decitabine administered at 15 mg/m2, 5 days
MCGRs.173-176 In a dose-escalating study, dasatinib ren- a week for 2 consecutive weeks, rendered a CHR in 34%
dered 2-log or greater reductions in Bcr-Abl transcripts in and a CCGR in 46% of patients.190,191 These and other
43% of patients in advanced phase, and 37% of patients in agents with different targets are currently being developed
CP achieved 2-log or greater reductions, including 4 pa- in an attempt to prevent or overcome resistance to imatinib.
tients in each group who had an MMR.178 Overall, dasatinib Immune-mediated events play an important role in the
is well tolerated, with myelosuppression and gastrointesti- suppression of the CML clone, as evidenced by a subset of

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 983

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

patients who maintained a durable CHR after discontinua- patients who develop the Bcr-Abl T315I mutant, under-
tion of interferon alfa therapy195 and after allogeneic-SCT standing the mechanism of resistance of patients without
due to a graft-vs-leukemia effect.196 These observations detectable mutations, the emergence of new mechanisms of
have sparked interest in developing immunologic ap- resistance such as new mutations to the new TKI and other
proaches to treating CML. One such approach is the use of agents, developing novel strategies for the management of
vaccines to elicit specific immune responses directed to- minimal residual disease, and delineating the current role
ward CML-restricted tumor antigens. Several vaccines are of SCT, will be the subject of arduous investigation in
currently being developed in clinical studies, which en- future years. Despite all these uncertainties, the treatment
compass the use of BCR-ABL junction-spanning se- prospects of patients with CML have never been brighter.
quences,197-199 the nonapeptide PR1 derived from protein-
ase 3,200 leukocyte-derived HSP70-peptide complexes,201
REFERENCES
and granulocyte-macrophage colony-stimulating factor se- 1. Abramson S, Miller RG, Phillips RA. The identification in adult bone
creting vaccines.202 The Bcr-Abl breakpoint fusion peptide marrow of pluripotent and restricted stem cells of the myeloid and lymphoid
systems. J Exp Med. 1977;145:1567-1579.
vaccine has been already tested in phase 2 clinical stud- 2. Bennett J. Case of hypertrophy of the spleen and liver in which death
ies.198,199 Bocchia et al199 have reported on 16 patients with took place from suppuration of the blood. Edinb Med Surg J. 1845;64:413-423.
3. Dameshek W. Some speculations on the myeloproliferative syndromes.
stable residual disease after more than 12 months of imatinib Blood. 1951;6:372-375.
therapy (n=10) or 24 months of interferon alfa therapy (n=6) 4. Nowell PC, Hungerford DA. A minute chromosome in human chronic
granulocytic leukemia. Science. 1960;132:1497.
who received 6 vaccinations with a peptide vaccine derived 5. Rowley JD. Letter: A new consistent chromosomal abnormality in
from the sequence p210-b3a2. Five of 9 patients taking chronic myelogenous leukaemia identified by quinacrine fluorescence and
imatinib obtained a CCGR after vaccination, and 3 had Giemsa staining. Nature. 1973;243:290-293.
6. Bartram CR, de Klein A, Hagemeijer A, et al. Translocation of c-ab1
undetectable levels of b3a2 transcripts by RT-PCR. Among oncogene correlates with the presence of a Philadelphia chromosome in
the patients treated with interferon alfa, 2 achieved a CCGR, chronic myelocytic leukaemia. Nature. 1983;306:277-280.
7. Groffen J, Stephenson JR, Heisterkamp N, de Klein A, Bartram CR,
and 3 had improvement in the percentage of Ph chromo- Grosveld G. Philadelphia chromosomal breakpoints are clustered within a
some–positive metaphases. Overall, these data suggest that limited region, bcr, on chromosome 22. Cell. 1984;36:93-99.
8. Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer
this peptide vaccine may have a role in the management of J Clin. 2004;54:8-29.
patients with CML and minimal residual disease. 9. Deininger MW, Goldman JM, Melo JV. The molecular biology of
chronic myeloid leukemia. Blood. 2000;96:3343-3356.
10. Bizzozero OJ Jr, Johnson KG, Ciocco A, Kawasaki S, Toyoda S.
Radiation-related leukemia in Hiroshima and Nagasaki 1946-1964: II. Ann
SUMMARY Intern Med. 1967;66:522-530.
11. Kantarjian HM, Keating MJ, Smith TL, Talpaz M, McCredie KB.
Consensus exists that imatinib therapy is the standard ap- Proposal for a simple synthesis prognostic staging system in chronic myelog-
enous leukemia. Am J Med. 1990;88:1-8.
proach to the treatment of CML, and this modality will be 12. Speck B, Bortin MM, Champlin R, et al. Allogeneic bone-marrow
measured in the future against all new therapies. The recent transplantation for chronic myelogenous leukaemia. Lancet. 1984;1:665-668.
13. Vardiman JW, Harris NL, Brunning RD. The World Health Organization
addition of the highly potent TKIs nilotinib and dasatinib (WHO) classification of the myeloid neoplasms. Blood. 2002;100:2292-2302.
has further improved the armamentarium against CML but 14. Kantarjian HM, Smith TL, McCredie KB, et al. Chronic myelogenous
leukemia: a multivariate analysis of the associations of patient characteristics
also posed new challenges. First, the role of these new and therapy with survival. Blood. 1985;66:1326-1335.
compounds in modern algorithms of CML therapy is still 15. Sokal JE, Baccarani M, Russo D, Tura S. Staging and prognosis in
chronic myelogenous leukemia. Semin Hematol. 1988;25:49-61.
unknown and must be defined. Despite having shown im- 16. Sokal JE, Baccarani M, Tura S, et al. Prognostic discrimination among
pressive response rates in patients after imatinib failure or younger patients with chronic granulocytic leukemia: relevance to bone mar-
row transplantation. Blood. 1985;66:1352-1357.
intolerance, the duration of these responses will be deter- 17. Kantarjian HM, Dixon D, Keating MJ, et al. Characteristics of acceler-
mined with longer follow-up. Their role in frontline ated disease in chronic myelogenous leukemia. Cancer. 1988;61:1441-1446.
18. Cortes JE, Talpaz M, O’Brien S, et al. Staging of chronic myeloid
therapy for CML will also have to be defined. Second, as leukemia in the imatinib era: an evaluation of the World Health Organization
our resources for controlling CML improve, the refinement proposal. Cancer. 2006;106:1306-1315.
19. Cortes J, Kantarjian H. Advanced-phase chronic myeloid leukemia.
in molecular monitoring must improve in parallel. In this Semin Hematol. 2003;40:79-86.
regard, the lack of consistency among different laboratories 20. Karanas A, Silver RT. Characteristics of the terminal phase of chronic
granulocytic leukemia. Blood. 1968;32:445-459.
in BCR-ABL transcript results by current PCR technolo- 21. Griesshammer M, Heinze B, Hellmann A, et al. Chronic myelogenous
gies is still an ongoing problem. Future improvements in leukemia in blast crisis: retrospective analysis of prognostic factors in 90
molecular techniques and their standardization by using a patients. Ann Hematol. 1996;73:225-230.
22. Kantarjian HM, Keating MJ, Talpaz M, et al. Chronic myelogenous
laboratory-specific conversion factor will be crucial to fur- leukemia in blast crisis: analysis of 242 patients. Am J Med. 1987;83:445-
ther our understanding of the dynamics of molecular re- 454.
23. Cortes JE, Talpaz M, Kantarjian H. Chronic myelogenous leukemia: a
sponse to TKIs, providing a uniform method and definition review. Am J Med. 1996;100:555-570.
of MMR and defining the concept of molecular relapse. 24. Cramer E, Auclair C, Hakim J, et al. Metabolic activity of phagocy-
tosing granulocytes in chronic granulocytic leukemia: ultrastructural observa-
Other challenges, such as defining the best treatment for tion of a degranulation defect. Blood. 1977;50:93-106.

984 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

25. Denburg JA, Wilson WE, Bienenstock J. Basophil production in myelo- 51. Mitelman F, Levan G, Nilsson PG, Brandt L. Non-random karyotypic
proliferative disorders: increases during acute blastic transformation of chronic evolution in chronic myeloid leukemia. Int J Cancer. 1976;18:24-30.
myeloid leukemia. Blood. 1982;60:113-120. 52. Lawler SD, O’Malley F, Lobb DS. Chromosome banding studies in
26. Silver RT. Morphology Of The Blood And Marrow In Clinical Practice. Philadelphia chromosome positive myeloid leukaemia. Scand J Haematol.
New York, NY: Grune & Stratton; 1970:84. 1976;17:17-28.
27. Strife A, Clarkson B. Biology of chronic myelogenous leukemia: is 53. Feinstein E, Cimino G, Gale RP, et al. p53 in chronic myelogenous
discordant maturation the primary defect? Semin Hematol. 1988;25:1-19. leukemia in acute phase. Proc Natl Acad Sci U S A. 1991;88:6293-6297.
28. Gordon MY, Dowding CR, Riley GP, Goldman JM, Greaves MF. 54. Sill H, Goldman JM, Cross NC. Homozygous deletions of the p16
Altered adhesive interactions with marrow stroma of haematopoietic progeni- tumor-suppressor gene are associated with lymphoid transformation of chronic
tor cells in chronic myeloid leukaemia. Nature. 1987;328:342-344. myeloid leukemia. Blood. 1995;85:2013-2016.
29. Dosik H, Rosner F, Sawitsky A. Acquired lipidosis: Gaucher-like cells 55. Towatari M, Adachi K, Kato H, Saito H. Absence of the human retino-
and “blue cells” in chronic granulocytic leukemia. Semin Hematol. 1972;9: blastoma gene product in the megakaryoblastic crisis of chronic myelogenous
309-316. leukemia. Blood. 1991;78:2178-2181.
30. Castro-Malaspina H, Moore MA. Pathophysiological mechanisms op- 56. Ogawa S, Mitani K, Kurokawa M, et al. Abnormal expression of Evi-1
erating in the development of myelofibrosis: role of megakaryocytes. Nouv Rev gene in human leukemias. Hum Cell. 1996;9:323-332.
Fr Hematol. 1982;24:221-226. 57. Ahuja H, Bar-Eli M, Arlin Z, et al. The spectrum of molecular alter-
31. Aguayo A, Kantarjian H, Manshouri T, et al. Angiogenesis in acute and ations in the evolution of chronic myelocytic leukemia. J Clin Invest. 1991;
chronic leukemias and myelodysplastic syndromes. Blood. 2000;96:2240- 87:2042-2047.
2245. 58. Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocyte-macrophage
32. Derderian PM, Kantarjian HM, Talpaz M, et al. Chronic myelogenous progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J
leukemia in the lymphoid blastic phase: characteristics, treatment response, Med. 2004;351:657-667.
and prognosis. Am J Med. 1993;94:69-74. 59. Zion M, Ben-Yehuda D, Avraham A, et al. Progressive de novo DNA
33. Bain B, Catovsky D, O’Brien M, Spiers AS, Richards GH. methylation at the bcr-abl locus in the course of chronic myelogenous leuke-
Megakaryoblastic transformation of chronic granulocytic leukaemia. An elec- mia. Proc Natl Acad Sci U S A. 1994;91:10722-10726.
tron microscopy and cytochemial study. J Clin Pathol. 1977;30:235-242. 60. Issa JP, Kantarjian H, Mohan A, et al. Methylation of the ABL1 pro-
34. Rosenthal S, Cancellos GP, Gralnick HP. Erythroblastic transformation moter in chronic myelogenous leukemia: lack of prognostic significance.
of chronic granulocytic leukemia. Am J Med. 1977;63:116-124. Blood. 1999;93:2075-2080.
35. Soler J, O’Brien M, de Castro JT, et al. Blast crisis of chronic granulo- 61. Asimakopoulos FA, Shteper PJ, Krichevsky S, et al. ABL1 methylation
cytic leukemia with mast cell and basophilic precursors. Am J Clin Pathol. is a distinct molecular event associated with clonal evolution of chronic my-
1985;83:254-259. eloid leukemia. Blood. 1999;94:2452-2460.
36. Sokal JE, Cox EB, Baccarani M, et al. Prognostic discrimination in 62. Roman-Gomez J, Castillejo JA, Jimenez A, et al. Cadherin-13, a media-
“good-risk” chronic granulocytic leukemia. Blood. 1984;63:789-799. tor of calcium-dependent cell-cell adhesion, is silenced by methylation in
37. Silver RT, Peterson BL, Szatrowski TP, et al. Treatment of the chronic chronic myeloid leukemia and correlates with pretreatment risk profile and
phase of chronic myeloid leukemia with an intermittent schedule of recombi- cytogenetic response to interferon alfa. J Clin Oncol. 2003;21:1472-1479.
nant interferon alfa-2b and cytarabine: results from CALGB study 9013. Leuk 63. Loriaux M, Deininger M. Clonal cytogenetic abnormalities in Philadel-
Lymphoma. 2003;44:39-48. phia chromosome negative cells in chronic myeloid leukemia patients treated
38. Quintas-Cardama A, Kantarjian H, Talpaz M, et al. Imatinib mesylate with imatinib. Leuk Lymphoma. 2004;45:2197-2203.
therapy may overcome the poor prognostic significance of deletions of deriva- 64. Fayad L, Kantarjian H, O’Brien S, et al. Emergence of new clonal
tive chromosome 9 in patients with chronic myelogenous leukemia. Blood. abnormalities following interferon-alpha induced complete cytogenetic re-
2005;105:2281-2286. sponse in patients with chronic myeloid leukemia: report of three cases. Leuke-
39. Hasford J, Pfirrmann M, Hehlmann R, et al, Writing Committee for the mia. 1997;11:767-771.
Collaborative CML Prognostic Factors Project Group. A new prognostic score 65. Medina J, Kantarjian H, Talpaz M, et al. Chromosomal abnormalities in
for survival of patients with chronic myeloid leukemia treated with interferon Philadelphia chromosome-negative metaphases appearing during imatinib
alfa. J Natl Cancer Inst. 1998;90:850-858. mesylate therapy in patients with Philadelphia chromosome-positive chronic
40. Gratwohl A, Hermans J, Niederwieser D, et al, Chronic Leukemia myelogenous leukemia in chronic phase. Cancer. 2003;98:1905-1911.
Working Party of the European Group for Bone Marrow Transplantation. Bone 66. Bumm T, Muller C, Al-Ali HK, et al. Emergence of clonal cytogenetic
marrow transplantation for chronic myeloid leukemia: long-term results. Bone abnormalities in Ph-cells in some CML patients in cytogenetic remission to
Marrow Transplant. 1993;12:509-516. imatinib but restoration of polyclonal hematopoiesis in the majority. Blood.
41. Kurzrock R, Gutterman JU, Talpaz M. The molecular genetics of Phila- 2003;101:1941-1949.
delphia chromosome-positive leukemias. N Engl J Med. 1988;319:990-998. 67. Abelson HT, Rabstein LS. Lymphosarcoma: virus-induced thymic-
42. Huret JL. Complex translocations, simple variant translocations and Ph- independent disease in mice. Cancer Res. 1970;30:2213-2222.
negative cases in chronic myelogenous leukaemia. Hum Genet. 1990;85:565- 68. Laneuville P. Abl tyrosine protein kinase. Semin Immunol. 1995;7:255-
568. 266.
43. De Braekeleer M. Variant Philadelphia translocations in chronic my- 69. Bernards A, Rubin CM, Westbrook CA, Paskind M, Baltimore D. The
eloid leukemia. Cytogenet Cell Genet. 1987;44:215-222. first intron in the human c-abl gene is at least 200 kilobases long and is a target
44. Albano F, Specchia G, Pannunzio A, et al. Variant t(9;22) translocation for translocations in chronic myelogenous leukemia. Mol Cell Biol.
may identify a subgroup of chronic myeloid leukemia patients with poor 1987;7:3231-3236.
prognosis [abstract]. Blood. 2001;98:321a. 70. Heisterkamp N, Jenster G, ten Hoeve J, Zovich D, Pattengale PK,
45. El-Zimaity MM, Kantarjian H, Talpaz M, et al. Results of imatinib Groffen J. Acute leukaemia in bcr/abl transgenic mice. Nature. 1990;344:251-
mesylate therapy in chronic myelogenous leukaemia with variant Philadelphia 253.
chromosome. Br J Haematol. 2004;125:187-195. 71. Melo JV. The diversity of BCR-ABL fusion proteins and their relation-
46. Shtalrid M, Talpaz M, Blick M, et al. Philadelphia-negative chronic ship to leukemia phenotype. Blood. 1996;88:2375-2384.
myelogenous leukemia with breakpoint cluster region rearrangement: molecu- 72. Faderl S, Talpaz M, Estrov Z, O’Brien S, Kurzrock R, Kantarjian HM.
lar analysis, clinical characteristics, and response to therapy. J Clin Oncol. The biology of chronic myeloid leukemia. N Engl J Med. 1999;341:164-172.
1988;6:1569-1575. 73. Sawyers CL. Chronic myeloid leukemia. N Engl J Med. 1999;340:
47. Kurzrock R, Kantarjian HM, Shtalrid M, Gutterman JU, Talpaz M. 1330-1340.
Philadelphia chromosome-negative chronic myelogenous leukemia without 74. Ravandi F, Cortes J, Albitar M, et al. Chronic myelogenous leukaemia
breakpoint cluster region rearrangement: a chronic myeloid leukemia with a with p185(BCR/ABL) expression: characteristics and clinical significance. Br
distinct clinical course. Blood. 1990;75:445-452. J Haematol. 1999;107:581-586.
48. Jennings BA, Mills KI. c-myc locus amplification and the acquisition of 75. Pane F, Frigeri F, Sindona M, et al. Neutrophilic-chronic myeloid leu-
trisomy 8 in the evolution of chronic myeloid leukaemia. Leuk Res. 1998; kemia: a distinct disease with a specific molecular marker (BCR/ABL with C3/
22:899-903. A2 junction). Blood. 1996;88:2410-2414.
49. Calabretta B, Perrotti D. The biology of CML blast crisis. Blood. 2004; 76. Lugo TG, Pendergast AM, Muller AJ, Witte ON. Tyrosine kinase
103:4010-4022. activity and transformation potency of bcr-abl oncogene products. Science.
50. Gaiger A, Henn T, Horth E, et al. Increase of bcr-abl chimeric mRNA 1990;247:1079-1082.
expression in tumor cells of patients with chronic myeloid leukemia precedes 77. Bose S, Deininger M, Gora-Tybor J, Goldman JM, Melo JV. The
disease progression. Blood. 1995;86:2371-2378. presence of typical and atypical BCR-ABL fusion genes in leukocytes of

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 985

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

normal individuals: biologic significance and implications for the assessment 102. O’Brien S, Talpaz M, Cortes J, et al. Simultaneous homoharringtonine
of minimal residual disease. Blood. 1998;92:3362-3367. and interferon-alpha in the treatment of patients with chronic-phase chronic
78. Biernaux C, Sels A, Huez G, Stryckmans P. Very low level of major myelogenous leukemia. Cancer. 2002;94:2024-2032.
BCR-ABL expression in blood of some healthy individuals. Bone Marrow 103. Baccarani M, Rosti G, de Vivo A, et al. A randomized study of inter-
Transplant. 1996;17(suppl 3):S45-S47. feron-alpha versus interferon-alpha and low-dose arabinosyl cytosine in
79. Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell- chronic myeloid leukemia. Blood. 2002;99:1527-1535.
proliferation and transformation. Biochim Biophys Acta. 1991;1072:129-157. 104. Kantarjian HM, O’Brien S, Smith TL, et al. Treatment of Philadelphia
80. Liebermann DA, Gregory B, Hoffman B. AP-1 (Fos/Jun) transcription chromosome-positive early chronic phase chronic myelogenous leukemia with
factors in hematopoietic differentiation and apoptosis. Int J Oncol. 1998;12: daily doses of interferon alpha and low-dose cytarabine. J Clin Oncol. 1999;17:
685-700. 284-292.
81. Passegue E, Jochum W, Schorpp-Kistner M, Mohle-Steinlein U, 105. Guilhot F, Chastang C, Michallet M, et al, French Chronic Myeloid
Wagner EF. Chronic myeloid leukemia with increased granulocyte progenitors Leukemia Study Group. Interferon alfa-2b combined with cytarabine versus
in mice lacking junB expression in the myeloid lineage. Cell. 2001;104:21-32. interferon alone in chronic myelogenous leukemia. N Engl J Med. 1997;337:
82. Ramaraj P, Singh H, Niu N, et al. Effect of mutational inactivation of 223-229.
tyrosine kinase activity on BCR/ABL-induced abnormalities in cell growth and 106. Kantarjian HM, O’Brien S, Cortes JE, et al. Complete cytogenetic and
adhesion in human hematopoietic progenitors. Cancer Res. 2004;64:5322- molecular responses to interferon-alpha-based therapy for chronic myelog-
5331. enous leukemia are associated with excellent long-term prognosis. Cancer.
83. Zhao RC, Jiang Y, Verfaillie CM. A model of human p210(bcr/ABL)- 2003;97:1033-1041.
mediated chronic myelogenous leukemia by transduction of primary normal 107. Hochhaus A, Reiter A, Saussele S, et al, German CML Study Group and
human CD34(+) cells with a BCR/ABL-containing retroviral vector. Blood. the UK MRC CML Study Group. Molecular heterogeneity in complete cytoge-
2001;97:2406-2412. netic responders after interferon-alpha therapy for chronic myelogenous leuke-
84. Daley GQ, Van Etten RA, Baltimore D. Induction of chronic myelog- mia: low levels of minimal residual disease are associated with continuing
enous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromo- remission. Blood. 2000;95:62-66.
some. Science. 1990;247:824-830. 108. Molldrem JJ, Lee PP, Wang C, et al. Evidence that specific T lympho-
85. Kelliher MA, McLaughlin J, Witte ON, Rosenberg N. Induction of a cytes may participate in the elimination of chronic myelogenous leukemia. Nat
chronic myelogenous leukemia-like syndrome in mice with v-abl and BCR/ Med. 2000;6:1018-1023.
ABL. Proc Natl Acad Sci U S A. 1990;87:6649-6653. 109. Morton AJ, Gooley T, Hansen JA, et al. Association between
86. Pendergast AM, Gishizky ML, Havlik MH, Witte ON. SH1 domain pretransplant interferon-alpha and outcome after unrelated donor marrow
autophosphorylation of P210 BCR/ABL is required for transformation but not transplantation for chronic myelogenous leukemia in chronic phase. Blood.
growth factor independence. Mol Cell Biol. 1993;13:1728-1736. 1998;92:394-401.
87. Zhang X, Subrahmanyam R, Wong R, Gross AW, Ren R. The NH(2)- 110. Giralt SA, Kantarjian HM, Talpaz M, et al. Effect of prior interferon
terminal coiled-coil domain and tyrosine 177 play important roles in induction alfa therapy on the outcome of allogeneic bone marrow transplantation for
of a myeloproliferative disease in mice by Bcr-Abl. Mol Cell Biol. 2001;21: chronic myelogenous leukemia. J Clin Oncol. 1993;11:1055-1061.
840-853. 111. Giralt S, Szydlo R, Goldman JM, et al. Effect of short-term interferon
88. Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic mye- therapy on the outcome of subsequent HLA-identical sibling bone marrow
logenous leukaemia. Nat Rev Cancer. 2005;5:172-183. transplantation for chronic myelogenous leukemia: an analysis from the inter-
89. Pendergast AM, Quilliam LA, Cripe LD, et al. BCR-ABL-induced national bone marrow transplant registry. Blood. 2000;95:410-415.
oncogenesis is mediated by direct interaction with the SH2 domain of the 112. Hehlmann R, Hochhaus A, Kolb HJ, et al. Interferon-alpha before
GRB-2 adaptor protein. Cell. 1993;75:175-185. allogeneic bone marrow transplantation in chronic myelogenous leukemia
90. Hu Y, Liu Y, Pelletier S, et al. Requirement of Src kinases Lyn, Hck and does not affect outcome adversely, provided it is discontinued at least 90 days
Fgr for BCR-ABL1-induced B-lymphoblastic leukemia but not chronic my- before the procedure. Blood. 1999;94:3668-3677.
eloid leukemia. Nat Genet. 2004;36:453-461. 113. Lee SJ, Klein JP, Anasetti C, et al. The effect of pretransplant interferon
91. Bhatia R, Holtz M, Niu N, et al. Persistence of malignant hematopoietic therapy on the outcome of unrelated donor hematopoietic stem cell transplanta-
progenitors in chronic myelogenous leukemia patients in complete cytogenetic tion for patients with chronic myelogenous leukemia in first chronic phase.
remission following imatinib mesylate treatment. Blood. 2003;101:4701- Blood. 2001;98:3205-3211.
4707. 114. Holowiecki J, Kruzel T, Wojnar J, et al. Allogeneic stem cell transplan-
92. Donato NJ, Wu JY, Stapley J, et al. BCR-ABL independence and LYN tation from unrelated and related donors is effective in CML Ph+ and ALL Ph+
kinase overexpression in chronic myelogenous leukemia cells selected for patients pretreated with imatinib [abstract]. Blood. 2003;102(pt 2):436b.
resistance to STI571. Blood. 2003;101:690-698. 115. Prejzner W, Zaucha JM, Szatkowski D, et al. Imatinib therapy prior to
93. Kerkhoff E, Rapp UR. Cell cycle targets of Ras/Raf signalling. myeloablative allogeneic stem cell transplantation does not increase acute
Oncogene. 1998;17:1457-1462. transplant related toxicity [abstract]. Blood. 2003;102(pt 1):911a.
94. Nosaka T, Kawashima T, Misawa K, Ikuta K, Mui AL, Kitamura T. 116. Hansen JA, Gooley TA, Martin PJ, et al. Bone marrow transplants from
STAT5 as a molecular regulator of proliferation, differentiation and apoptosis unrelated donors for patients with chronic myeloid leukemia. N Engl J Med.
in hematopoietic cells. Embo J. 1999;18:4754-4765. 1998;338:962-968.
95. Sonoyama J, Matsumura I, Ezoe S, et al. Functional cooperation among 117. Baker KS, Gurney JG, Ness KK, et al. Late effects in survivors of
Ras, STAT5, and phosphatidylinositol 3-kinase is required for full oncogenic chronic myeloid leukemia treated with hematopoietic cell transplantation:
activities of BCR/ABL in K562 cells. J Biol Chem. 2002;277:8076-8082. results from the Bone Marrow Transplant Survivor Study. Blood. 2004;104:
96. Neviani P, Santhanam R, Trotta R, et al. The tumor suppressor PP2A is 1898-1906.
functionally inactivated in blast crisis CML through the inhibitory activity of 118. Kiss TL, Abdolell M, Jamal N, Minden MD, Lipton JH, Messner HA.
the BCR/ABL-regulated SET protein. Cancer Cell. 2005;8:355-368. Long-term medical outcomes and quality-of-life assessment of patients with
97. Janssens V, Goris J. Protein phosphatase 2A: a highly regulated family chronic myeloid leukemia followed at least 10 years after allogeneic bone
of serine/threonine phosphatases implicated in cell growth and signalling. marrow transplantation. J Clin Oncol. 2002;20:2334-2343.
Biochem J. 2001;353(pt 3):417-439. 119. Radich JP, Gooley T, Bensinger W, et al. HLA-matched related he-
98. Talpaz M, Mavligit G, Keating M, Walters RS, Gutterman JU. Human matopoietic cell transplantation for chronic-phase CML using a targeted
leukocyte interferon to control thrombocytosis in chronic myelogenous leuke- busulfan and cyclophosphamide preparative regimen. Blood. 2003;102:31-
mia. Ann Intern Med. 1983;99:789-792. 35.
99. Talpaz M, Kantarjian HM, McCredie K, Trujillo JM, Keating MJ, 120. Weisdorf DJ, Anasetti C, Antin JH, et al. Allogeneic bone marrow
Gutterman JU. Hematologic remission and cytogenetic improvement induced transplantation for chronic myelogenous leukemia: comparative analysis of
by recombinant human interferon alpha A in chronic myelogenous leukemia. N unrelated versus matched sibling donor transplantation. Blood. 2002;99:1971-
Engl J Med. 1986;314:1065-1069. 1977.
100. Kantarjian HM, Smith TL, O’Brien S, Beran M, Pierce S, Talpaz M, 121. Or R, Shapira MY, Resnick I, et al. Nonmyeloablative allogeneic stem
The Leukemia Service. Prolonged survival in chronic myelogenous leukemia cell transplantation for the treatment of chronic myeloid leukemia in first
after cytogenetic response to interferon-alpha therapy. Ann Intern Med. 1995; chronic phase. Blood. 2003;101:441-445.
122:254-261. 122. Crawley C, Szydlo R, Lalancette M, et al. Outcomes of reduced-inten-
101. O’Brien S, Kantarjian H, Koller C, et al. Sequential homoharringtonine sity transplantation for chronic myeloid leukemia: an analysis of prognostic
and interferon-alpha in the treatment of early chronic phase chronic myelog- factors from the Chronic Leukemia Working Party of the EBMT. Blood.
enous leukemia. Blood. 1999;93:4149-4153. 2005;106:2969-2976. Epub 2005 Jul 5.

986 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

123. Radich JP, Gehly G, Gooley T, et al. Polymerase chain reaction detec- 144. Cortes J, O’Brien S, Kantarjian H. Discontinuation of imatinib therapy
tion of the BCR-ABL fusion transcript after allogeneic marrow transplantation after achieving a molecular response. Blood. 2004;104:2204-2205.
for chronic myeloid leukemia: results and implications in 346 patients. Blood. 145. Mauro MJ, Druker BJ, Maziarz RT. Divergent clinical outcome in two
1995;85:2632-2638. CML patients who discontinued imatinib therapy after achieving a molecular
124. Olavarria E, Craddock C, Dazzi F, et al. Imatinib mesylate (STI571) in remission. Leuk Res. 2004;28(suppl 1):S71-S73.
the treatment of relapse of chronic myeloid leukemia after allogeneic stem cell 146. Ghanima W, Kahrs J, Dahl TG, 3rd, Tjonnfjord GE. Sustained cytoge-
transplantation. Blood. 2002;99:3861-3862. netic response after discontinuation of imatinib mesylate in a patient with
125. Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective chronic myeloid leukaemia. Eur J Haematol. 2004;72:441-443.
inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat 147. Rousselot P, Huguet F, Cayuela JM, et al. Imatinib mesylate discontinu-
Med. 1996;2:561-566. ation in patients with chronic myelogenous leukaemia in complete molecular
126. Beran M, Cao X, Estrov Z, et al. Selective inhibition of cell proliferation remission for more than two years [abstract]. Blood. 2005;106:321a.
and BCR-ABL phosphorylation in acute lymphoblastic leukemia cells express- 148. Graham SM, Jorgensen HG, Allan E, et al. Primitive, quiescent, Phila-
ing Mr 190,000 BCR-ABL protein by a tyrosine kinase inhibitor (CGP-57148). delphia-positive stem cells from patients with chronic myeloid leukemia are
Clin Cancer Res. 1998;4:1661-1672. insensitive to STI571 in vitro. Blood. 2002;99:319-325.
127. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific 149. Copland M. Hamilton A, Baird JW, et al. Dasatinib (BMS-354825) has
inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl increased activity against Bcr-Abl compared to imatinib in primary CML cells
J Med. 2001;344:1031-1037. in vitro, but does not eradicate quiescent CML stem cells [abstract]. Blood.
128. Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytoge- 2005;106:205a.
netic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl 150. Chu S, Xu H, Shah NP, et al. Detection of BCR-ABL kinase mutations
J Med. 2002;346:645-652. in CD34+ cells from chronic myelogenous leukemia patients in complete
129. Kantarjian HM, Cortes JE, O’Brien S, et al. Long-term survival benefit cytogenetic remission on imatinib mesylate treatment. Blood. 2005;105:
and improved complete cytogenetic and molecular response rates with imatinib 2093-2098.
mesylate in Philadelphia chromosome-positive chronic-phase chronic myeloid 151. Jiang X, Zhao Y, Chan P, et al. Quantitative real-time RT-PCR analysis
leukemia after failure of interferon-alpha. Blood. 2004;104:1979-1988. shows BCR-ABL expression progressively decreases as primitive leukemic
130. O’Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with cells from patients with chronic myeloid leukemia (CML) differentiate in vivo
interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic [abstract]. Exp Hematol. 2003;31:228-229.
myeloid leukemia. N Engl J Med. 2003;348:994-1004. 152. Hughes T, Kaeda J, Branford S, et al. Molecular responses to imatinib
131. Hahn EA, Glendenning GA, Sorensen MV, et al. Quality of life in (STI571) or interferon + Ara-C as initial therapy for CML: results in the IRIS
patients with newly diagnosed chronic phase chronic myeloid leukemia on study [abstract]. Blood. 2002;100:93a.
imatinib versus interferon alfa plus low-dose cytarabine: results from the IRIS 153. Cortes J, O’Brien S, Talpaz M, et al. Clinical significance of molecular
Study. J Clin Oncol. 2003;21:2138-2146. response in chronic myeloid leukemia (CML) after imatinib mesylate
132. Simonsson B, the IRIS (International Randomized IFN vs ST1571) (Gleevec) therapy: low levels of residual disease predict for response duration
Study Group. Beneficial effects of cytogenetic and molecular response on [abstract]. Blood. 2003;102(Pt 1):416a.
long-term outcome in patients with newly diagnosed chronic myeloid leukemia 154. Talpaz M, Kantarjian H, Kurzrock R, Trujillo JM, Gutterman JU.
in chronic phase (CML-CP) treated with imatinib (IM): update from the IRIS Interferon-alpha produces sustained cytogenetic responses in chronic myelog-
study [abstract]. Blood. 2005;106:52a. enous leukemia. Philadelphia chromosome-positive patients. Ann Intern Med.
133. Colombat M, Fort MP, Chollet C, et al. Molecular remission in chronic 1991;114:532-538.
myeloid leukemia patients with sustained complete cytogenetic remission after 155. Hughes TP, Deininger MW, Hochhaus A, et al. Monitoring CML
imatinib mesylate treatment. Haematologica. 2006;91:162-168. patients responding to treatment with tyrosine kinase inhibitors: review and
134. Goldman JM, Hughes T, Radich J, et al. Continuing reduction in level recommendations for ‘harmonizing’ current methodology for detecting BCR-
of residual disease after 4 years in patients with CML in chronic phase ABL transcripts and kinase domain mutations and for expressing results.
responding to first-line imatinib (IM) in the IRIS study [abstract]. Blood. 2005; Blood. Epub 2006 Mar 7.
106:51a. 156. Hochhaus A, La Rosee P. Imatinib therapy in chronic myelogenous
135. Kantarjian H, O’Brien S, Cortes J, et al. Analysis of the impact of leukemia: strategies to avoid and overcome resistance. Leukemia. 2004;18:
imatinib mesylate therapy on the prognosis of patients with Philadelphia 1321-1331.
chromosome-positive chronic myelogenous leukemia treated with interferon- 157. O’Hare T, Walters DK, Stoffregen EP, et al. In vitro activity of Bcr-Abl
alpha regimens for early chronic phase. Cancer. 2003;98:1430-1437. inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-
136. Roy L, Guilhot J, Krahnke T, et al. Survival advantage from imatinib resistant Abl kinase domain mutants. Cancer Res. 2005;65:4500-4505.
compared to the combination interferon-{alpha} plus cytarabine in chronic 158. Deininger M, Buchdunger E, Druker BJ. The development of imatinib
phase CML: historical comparison between two phase III trials. Blood. Epub as a therapeutic agent for chronic myeloid leukemia. Blood. 2005;105:2640-
2006 Apr 20. 2653.
137. Kantarjian H, Talpaz M, O’Brien S, et al. High-dose imatinib mesylate 159. Carter TA, Wodicka LM, Shah NP, et al. Inhibition of drug-resistant
therapy in newly diagnosed Philadelphia chromosome-positive chronic phase mutants of ABL, KIT, and EGF receptor kinases. Proc Natl Acad Sci U S A.
chronic myeloid leukemia. Blood. 2004;103:2873-2878. 2005;102:11011-11016.
138. Hughes TP, Branford S, Matthews J, et al. Trial of higher dose imatinib 160. Cowan-Jacob SW, Guez V, Fendrich G, et al. Imatinib (STI571) resis-
with selective intensification in newly diagnosed CML patients in the chronic tance in chronic myelogenous leukemia: molecular basis of the underlying
phase [abstract]. Blood. 2003;102(Pt 1):31a. mechanisms and potential strategies for treatment. Mini Rev Med Chem.
139. Cortes J, Giles F, Salvado A, et al. High-dose (HD) imatinib in patients 2004;4:285-299.
(pts) with previously untreated chronic myeloid leukemia (CML) in early 161. Branford S, Rudzki Z, Walsh S, et al. Detection of BCR-ABL mutations
chronic phase (CP): preliminary results of a multicenter community based trial in patients with CML treated with imatinib is virtually always accompanied by
[abstract]. J Clin Oncol. 2005;23(suppl):564s. clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop)
140. Rosti G, Martinelli G, Castagnetti F, et al. Imatinib 800 mg: prelimi- are associated with a poor prognosis. Blood. 2003;102:276-283.
nary results of a phase II trial of the GIMEMA CML working party in 162. Hochhaus A, Kreil S, Corbin A, et al. Roots of clinical resistance to STI-
intermediate sokal risk patients and status-of-the-art of an ongoing multina- 571 cancer therapy. Science. 2001;293:2163.
tional, prospective randomized trial of imatinib standard dose (400 mg daily) 163. Soverini S, Martinelli G, Rosti G, et al. ABL mutations in late chronic
vs high dose (800 mg daily) in high sokal risk patients [abstract]. Blood. phase chronic myeloid leukemia patients with up-front cytogenetic resistance
2005;106:320a. to imatinib are associated with a greater likelihood of progression to blast crisis
141. Cortes J, Giles F, O’Brien S, et al. Result of high-dose imatinib and shorter survival: a study by the GIMEMA Working Party on Chronic
mesylate in patients with Philadelphia chromosome-positive chronic myeloid Myeloid Leukemia. J Clin Oncol. 2005;23:4100-4109.
leukemia after failure of interferon-alpha. Blood. 2003;102:83-86. 164. Jabbour E, Kantarjian H, Jones D, et al. Long-term incidence and
142. Cortes J, Talpaz M, O’Brien S, et al. Molecular responses in patients outcome of BCR-ABL mutations in patients (pts) with chronic myeloid leuke-
with chronic myelogenous leukemia in chronic phase treated with imatinib mia (CML) treated with imatinib mesylate: P-loop mutations are not associated
mesylate. Clin Cancer Res. 2005;11:3425-3432. with worse outcome [abstract]. Blood. 2004;104:288a.
143. Hughes TP, Branford S, Reynolds J, et al. Maintenance of imatinib dose 165. Jabbour E, Kantarjian H, Talpaz M, et al. Outcome of salvage therapy
intensity in the first six months of therapy for newly diagnosed patients with in patients (pts) with chronic myeloid leukemia (CML) who failed imatinib
CML is predictive of molecular response, independent of the ability to increase after developing BCR-ABL kinase mutation [abstract]. Blood. 2005;106:
dose at a later point [abstract]. Blood. 2005;106:51a. 318a.

Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com 987

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.
CHRONIC MYELOID LEUKEMIA

166. Pricl S, Fermeglia M, Ferrone M, Tamborini E. T315I-mutated Bcr-Abl 183. Dorsey JF, Jove R, Kraker AJ, Wu J. The pyrido[2,3-d]pyrimidine
in chronic myeloid leukemia and imatinib: insights from a computational derivative PD180970 inhibits p210Bcr-Abl tyrosine kinase and induces
study. Mol Cancer Ther. 2005;4:1167-1174. apoptosis of K562 leukemic cells. Cancer Res. 2000;60:3127-3131.
167. Tanis KQ, Veach D, Duewel HS, Bornmann WG, Koleske AJ. Two 184. Gumireddy K, Baker SJ, Cosenza SC, et al. A non-ATP-competitive
distinct phosphorylation pathways have additive effects on Abl family kinase inhibitor of BCR-ABL overrides imatinib resistance. Proc Natl Acad Sci U S A.
activation. Mol Cell Biol. 2003;23:3884-3896. 2005;102:1992-1997.
168. Leguay T, Desplat V, Marit G, Mahon FX. D276G mutation is associ- 185. Cortes J, Albitar M, Thomas D, et al. Efficacy of the farnesyl transferase
ated with a poor prognosis in imatinib mesylate-resistant chronic myeloid inhibitor R115777 in chronic myeloid leukemia and other hematologic malig-
leukemia patients [letter]. Leukemia. 2005;19:2332-2333; author reply 2333- nancies. Blood. 2003;101:1692-1697.
2334. 186. Peters DG, Hoover RR, Gerlach MJ, et al. Activity of the farnesyl
169. Donato NJ, Wu JY, Stapley J, et al. Imatinib mesylate resistance protein transferase inhibitor SCH66336 against BCR/ABL-induced murine
through BCR-ABL independence in chronic myelogenous leukemia. Cancer leukemia and primary cells from patients with chronic myeloid leukemia.
Res. 2004;64:672-677. Blood. 2001;97:1404-1412.
170. Kantarjian H, Ottmann O, Cortes J, et al. AMN107, a novel amino- 187. Hoover RR, Mahon FX, Melo JV, Daley GQ. Overcoming STI571
pyrimidine inhibitor of Bcr-Abl, has significant activity in imatinib-resistant resistance with the farnesyl transferase inhibitor SCH66336. Blood. 2002;
bcr-abl positive chronic myeloid leukemia (CML) [abstract]. J Clin Oncol. 100:1068-1071.
2005;23(suppl):195s. 188. Reichert A, Heisterkamp N, Daley GQ, Groffen J. Treatment of Bcr/
171. Sawyers CL, Shah NP, Kantarjian HM, et al. A phase I study of BMS- Abl-positive acute lymphoblastic leukemia in P190 transgenic mice with the
354825 in patients with imatinib-resistant and intolerant accelerated and blast farnesyl transferase inhibitor SCH66336. Blood. 2001;97:1399-1403.
phase chronic myeloid leukemia (CML): results from CA180002 [abstract]. J 189. Marin D, Kaeda JS, Andreasson C, et al. Phase I/II trial of adding
Clin Oncol. 2005;23(suppl):565s. semisynthetic homoharringtonine in chronic myeloid leukemia patients who
172. Talpaz M, Kantarjian H, Paquette R, et al. A phase I study of BMS- have achieved partial or complete cytogenetic response on imatinib. Cancer.
354825 in patients with imatinib-resistant and intolerant chronic phase chronic 2005;103:1850-1855.
myeloid leukemia (CML): results from CA180002 [abstract]. J Clin Oncol. 190. Issa JP, Gharibyan V, Cortes J, et al. Phase II study of low-dose
2005;23(suppl):564s. decitabine in patients with chronic myelogenous leukemia resistant to imatinib
173. Hochhaus A, Baccarani M, Sawhers C, et al. Efficacy of dasatinib in mesylate. J Clin Oncol. 2005;23:3948-3956.
patients with chronic phase Philadelphia chromosome-positive CML resistant 191. Kantarjian HM, O’Brien S, Cortes J, et al. Results of decitabine (5-aza-
or intolerant to imatinib: first results of the CA180013 ‘START-C’ phase II 2’deoxycytidine) therapy in 130 patients with chronic myelogenous leukemia.
study [abstract]. Blood. 2005;106:17a. Cancer. 2003;98:522-528.
174. Guilhot F, Apperley JF, Shah N, et al. A phase II study of dasatinib in 192. Yu C, Rahmani M, Almenara J, et al. Histone deacetylase inhibitors
patients with accelerated phase chronic myeloid leukemia (CML) who are promote STI571-mediated apoptosis in STI571-sensitive and -resistant Bcr/
resistant or intolerant to imatinib: first results of the CA180005 ‘START-A’ Abl+ human myeloid leukemia cells. Cancer Res. 2003;63:2118-2126.
study [abstract]. Blood. 2005;106:16a. 193. Nimmanapalli R, Fuino L, Stobaugh C, Richon V, Bhalla K.
175. Talpaz M, Rousselot P, Kim DW, et al. A phase II study of dasatinib in Cotreatment with the histone deacetylase inhibitor suberoylanilide hydroxamic
patients with chronic myeloid leukemia (CML) in myeloid blast crisis who are acid (SAHA) enhances imatinib-induced apoptosis of Bcr-Abl-positive human
resistant or intolerant to imatinib: first results of the CA180006 ‘START-B’ acute leukemia cells. Blood. 2003;101:3236-3239.
study [abstract]. Blood. 2005;106:16a. 194. Nimmanapalli R, Fuino L, Bali P, et al. Histone deacetylase inhibitor
176. Ottmann OG, Martinelli G, Dombret H, et al. A phase II study of LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-
dasatinib in patients with chronic myeloid leukemia (CML) in lymphoid blast Abl and induces apoptosis of imatinib mesylate-sensitive or -refractory chronic
crisis or Philadelphia-chromosome positive acute lymphoblastic leukemia myelogenous leukemia-blast crisis cells. Cancer Res. 2003;63:5126-5135.
(Ph+ ALL) who are resistant or intolerant to imatinib: the ‘START-L’ 195. Guilhot F, Lacotte-Thierry L. Interferon-alpha: mechanisms of action in
CA180015 study [abstract]. Blood. 2005;106:17a. chronic myelogenous leukemia in chronic phase. Hematol Cell Ther. 1998;40:
177. Lombardo LJ, Lee FY, Chen P, et al. Discovery of N-(2-chloro-6- 237-239.
methyl-phenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin- 196. Barrett J. Allogeneic stem cell transplantation for chronic myeloid
4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase in- leukemia. Semin Hematol. 2003;40:59-71.
hibitor with potent antitumor activity in preclinical assays. J Med Chem. 197. Pinilla-Ibarz J, Cathcart K, Korontsvit T, et al. Vaccination of patients
2004;47:6658-6661. with chronic myelogenous leukemia with bcr-abl oncogene breakpoint fusion
178. Shah NP, Nicoll JM, Branford S, et al. Molecular analysis of dasatinib peptides generates specific immune responses. Blood. 2000;95:1781-1787.
resistance mechanisms in CML patients identified novel BCR-ABL mutations 198. Cathcart K, Pinilla-Ibarz J, Korontsvit T, et al. A multivalent bcr-abl
predicted to retain sensitivity to imatinib: rationale for combination tyrosine fusion peptide vaccination trial in patients with chronic myeloid leukemia.
kinase inhibitor therapy [abstract]. Blood. 2005;106:318a. Blood. 2004;103:1037-1042.
179. Golas JM, Arndt K, Etienne C, et al. SKI-606, a 4-anilino-3- 199. Bocchia M, Gentili S, Abruzzese E, et al. Effect of a p210 multipeptide
quinolinecarbonitrile dual inhibitor of Src and Abl kinases, is a potent vaccine associated with imatinib or interferon in patients with chronic myeloid
antiproliferative agent against chronic myelogenous leukemia cells in culture leukaemia and persistent residual disease: a multicentre observational trial.
and causes regression of K562 xenografts in nude mice. Cancer Res. 2003; Lancet. 2005;365:657-662.
63:375-381. 200. Qazilbash MH, Wieder E, Rios R, et al. Vaccination with the PR1
180. Kimura S, Naito H, Segawa H, et al. NS-187, a potent and selective dual leukemia-associated antigen can induce complete remission in patients with
Bcr-Abl/Lyn tyrosine kinase inhibitor, is a novel agent for imatinib-resistant myeloid leukemia [abstract]. Blood. 2004;104:77a.
leukemia. Blood. 2005;106:3948-3954. 201. Li Z, Qiao Y, Liu B, et al. Combination of imatinib mesylate with
181. Hennequin LF, Allen J, Costello GF, et al. The discovery of AZD0530: autologous leukocyte-derived heat shock protein and chronic myelogenous
a novel, oral, highly selective and dual-specific inhibitor of the Src and Abl leukemia. Clin Cancer Res. 2005;11:4460-4468.
family kinases [abstract]. Proc Amer Assoc Cancer Res. 2005;46:2537. 202. Borrello I, Levitsky H, Damon L, et al. Vaccine-associated immune and
182. Wolff NC, Veach DR, Tong WP, Bornmann WG, Clarkson B, Ilaria RL WT-1 responses are associated with better relapse-free survival in patients with
Jr. PD166326, a novel tyrosine kinase inhibitor, has greater antileukemic AML in remission treated with a GM-CSF secreting leukemia vaccine and
activity than imatinib mesylate in a murine model of chronic myeloid leuke- autologous stem cell transplant (ASCT) [abstract]. J Clin Oncol. 2005;23
mia. Blood. 2005;105:3995-4003. (suppl):569s.

The Symposium on Oncology Practice: Hematological Malignancies


will continue in the August issue.

988 Mayo Clin Proc. • July 2006;81(7):973-988 • www.mayoclinicproceedings.com

For personal use. Mass reproduce only with permission from Mayo Clinic Proceedings.

You might also like