You are on page 1of 11

Blastic Phase of Chronic

Myelogenous Leukemia
Merat Karbasian Esfahani, MD*
Evelyn L. Morris, MD
Janice P. Dutcher, MD
Peter H. Wiernik, MD
Address
*Our Lady of Mercy Comprehensive Cancer Center, 600 East 233rd Street,
Bronx, NY 10466, USA.
E-mail: kar1070@yahoo.com
Current Treatment Options in Oncology 2006, 7:189–199
Current Science Inc. ISSN 1527–2729
Copyright © 2006 by Current Science Inc.

Opinion statement
Chronic myelogenous leukemia (CML), also known as chronic myelocytic or chronic myeloid
leukemia, is a clonal disorder of hematopoiesis that arises in a hematopoietic stem cell or
early progenitor cell. This is characterized by the dysregulated production of mature non-
lymphoid cells with normal differentiation. Eventually, in spite of the term chronic, there is
progression to acute leukemia, usually of the myeloid variety, which is highly resistant to
current therapies. Despite recent improvements in the treatment of early-stage disease,
CML blast crisis (CMLBC) remains a therapeutic challenge. CMLBC is highly refractory to
standard induction chemotherapy, with a response rate in myeloid blast crisis of less than
30%. Conventional chemotherapy has been much less successful in this disease compared
with de novo acute leukemia, with a mean survival after diagnosis of blast crisis of only
2 to 4 months for nonresponders. Many regimens of chemotherapies have been tried in
CMLBC, with minor success. Although imatinib was evaluated in patients with CMLBC, most
CMLBC cases today arise in patients already on imatinib-based therapy and developing
blastic phase on that therapy; thus there is no standard therapy for patients with CMLBC.
Further studies of the mechanisms of transformation of chronic-phase CMLBC at a molecu-
lar level, and methods to target these molecular abnormalities, will determine the future
direction of new treatment modalities. The prognosis of CML in blast crisis remains dis-
appointing, despite great efforts. Currently, the most successful strategy for improving
survival in CML is by prolonging the chronic phase and delaying the onset of blast crisis.

Introduction
Chronic myelogenous leukemia (CML) is a clonal dis- in CML, but hematopoietic stem cell behavior is abnor-
order of hematopoiesis and a myeloproliferative stem mal in tissue culture studies [1•,2]. CML progresses
cell disorder, characterized by dysregulated production from a chronic phase to an accelerated phase, followed
and expansion of myeloid, erythroid, and megakaryo- by a terminal blastic phase of lymphoid or myeloid
cytic cells. CML accounts for 15% of adult leukemias, phenotype [3]. CML blast crisis (CMLBC) is highly
with an incidence of one to two cases per 100,000 pop- refractory to standard induction chemotherapy, with a
ulation. Most patients present in their fifth and sixth response rate in myeloid blast crisis of less than 30%.
decades of life, with a male to female ratio of 1.3:1 [1•]. Allogeneic stem cell transplantation during blast crisis
The disease is characterized by a biphasic or triphasic does not confer superior benefit, and the 5-year survival
course and the presence of a characteristic cytogenetic rate is only 6%. Blast crisis is invariably fatal and despite
abnormality, the Philadelphia chromosome (Ph) in aggressive medical efforts, the median survival contin-
leukemic cells. Myeloid differentiation is not impaired ues to be approximately 2 to 3 months [4,5]. Although
190 Chronic Leukemia

in the era of imatinib initial results in blast crisis were With progression of the disease, 50% to 75% of
promising [6•], ABL mutations in late chronic-phase patients acquire chromosomal changes in addition to
CML patients with upfront cytogenetic resistance to the Ph chromosome, and this may precede or accom-
imatinib are associated with a greater likelihood of pro- pany hematologic and clinical manifestations of blastic
gression to blast crisis and shorter survival [7]. Because phase. The most frequently observed changes of karyo-
almost all chronic-phase patients are now treated with typic evolution are an additional Ph chromosome
imatinib, they are expected to be resistant when they (double Ph), isochromosome 17 (i17q), trisomy 8, and
progress to blast crisis [7,8]. trisomy 19 [1•]. Studies using fluorescent in situ
hybridization techniques are more sensitive in deter-
MOLECULAR GENETICS AND PHILADELPHIA CHROMOSOME mining additional chromosomal abnormalities. Studies
The Philadelphia chromosome (Ph), initially described have demonstrated new abnormalities in separate
in 1960 [9], is present in more than 90% of individuals subclones of blasts and in cases in which all lineages
with CML. The newly created BCR-ABL gene on the Ph demonstrate the abnormality.
chromosome encodes a 210-kd chimeric protein, which Deletions and mutations of p53 occur in 20% to
causes leukemogenesis and interferes with normal cell 30% of patients with CMLBC, associated with suppres-
cycle events, such as signal transduction and the regula- sion of apoptosis [1•]. Amplification of c-myc is seen in
tion of apoptosis and cell proliferation (p160 BCR and 20% of patients. Other rare events observed include
p145 ABL) [10,11]. This aberrant protein has enhanced mutations in ras and rearrangement and deletions of Rb
tyrosine phosphokinase enzymatic activity and is and p16. There is evidence to suggest that the biologic
involved in the molecular pathogenesis of CML. The abnormalities of CMLBC, namely the enhanced prolif-
normal ABL protein is found in the nucleus, where it eration and survival of cells and the arrest of differentia-
exerts its proapoptotic role, and in the cytoplasm. tion, are the consequence of abnormalities in the p53 or
In contrast, the BCR-ABL protein is exclusively cyto- Rb (or both) gene activity, or by dysregulation of differ-
plasmic and is anti-apoptotic. BCR-ABL affects the entiation-regulatory genes (ie, C/EBP alpha) [16].
DNA repair process, and altered DNA repair leads to The enhanced leukemic potential of CML granulocyte
genomic instability, which leads to clonal evolution macrophage progenitor cells has also been attributed to
and progression to blast crisis [12]. Primitive Ph+ the activation of ␤-catenin, resulting in these progenitor
hematopoietic progenitors from patients with CML cells acquiring the capacity of self-renewal [18]. The ␤-
show decreased adherence to bone marrow stroma [13] catenin levels in myeloid precursors in patients in blast
and fibronectin [14] because of a defect in ␤-1 integrin crisis are high and found to normalize once the patients
function, caused by the BCR-ABL protein. Although are treated with imatinib [17].
BCR and C-ABL proteins have no intrinsic oncogenic Some patients with CML demonstrate genomic
properties, the BCR-ABL protein has the ability to deletions resulting in loss of chromosome 9 and 22
transform cell lines and primary cells in vitro, behaving sequences flanking the translocation breakpoint on the
as a classical oncogene [15]. derivative [9], on additional partner chromosomes, or
on both. Such deletions are associated with a worse
BLAST TRANSFORMATION prognosis, with a median survival of 36 months (com-
Molecular biology Although the Ph chromosome trans- pared with > 90 months in those without deletions),
location may be the initiating event in CML, progres- and may influence the progression of CML [19].
sion to blast crisis appears to require the acquisition of
other chromosomal changes and/or dysregulation of Clinical manifestations At diagnosis of blast crisis, the
differentiation-regulatory genes [1•,16]. Although in most common clinical symptoms are fatigue and mal-
most cases there is a transition from an initial chronic aise. Other associated symptoms are fever, night sweats,
phase to an accelerated phase, and finally culmination bone pain, weight loss, and bleeding and infection as
in an inevitable CMLBC, in 20% to 25% of patients, mature cells are depleted [1•]. Most of the patients have
the blastic phase develops without the intermediate progressive splenomegaly often leading to left upper
accelerated phase. abdominal discomfort. Anemia is the most common
Several mechanisms have been implicated in the initial laboratory feature associated with progression to
malignant transformation produced by BCR-ABL, includ- accelerated or blastic phase. Thrombocytopenia, when
ing altered adhesion of hematopoietic cells to stroma present at the onset of blast crisis, indicates a lower
cells and extracellular matrix, activated signaling path- degree of marrow reserve and has a negative influence
ways that induce mitosis, inhibition of apoptosis, and on patient survival. Increasing eosinophilia and/or
degradation of important cellular proteins [12]. It has basophilia are signs of disease progression. Some
been postulated that the alleged anti-apoptotic effect patients develop lymphadenopathy. Complications
of BCR-ABL favors inaccurate DNA repair, whereas in associated with blast crisis include hyperleukocytosis,
normal cells, DNA damage would induce apoptosis [17]. leukostasis, bleeding, and infection. Radiation therapy
Blastic Phase of Chronic Myelogenous Leukemia Karbasian Esfahani et al. 191

is used to manage leukostasis in the cerebral or pulmo- rate of only 20%, range 11% to 60% (Table 1). The
nary vasculature, when the number of circulating blasts median survival in CMLBC is 2 to 3 months. Blast trans-
is high. It is sometimes administered to control symp- formation is most commonly myeloid [1•], but a lym-
toms of splenomegaly and is the treatment of choice phoblastic immunophenotype is seen in 20% to 30%
for cranial nerve palsies from meningeal leukemia. of cases. The blast transformation can even be mega-
When the peripheral blast count is more than 100,000 karyocytic, and this is defined immunophenotypically.
cells/␮L, leukapheresis is indicated [20]. Complete hematologic and complete marrow responses
Chronic myelogenous leukemia blast crisis resem- were seen in 29% of 56 evaluable patients with relapsed
bles acute leukemia with an absence of mature cells and or refractory Ph+ acute lymphoblastic leukemia (ALL)
is defined by the presence of extramedullary blastic or CML in lymphoid blast crisis [21]. Because of respon-
infiltrates, or of 30% or more leukemic blasts in the siveness to vincristine and prednisone, lymphoid
peripheral blood and bone marrow [1•]. CMLBC is blast crisis has a slightly better survival than does
highly resistant to therapy, with a median response myeloid blast crisis.

Treatment
Diet and lifestyle
• There is no evidence to support any role for diet and lifestyle changes in the
treatment of CMLBC.

Chemotherapy
• The treatment of CMLBC is a challenge because conventional chemotherapy
has been much less successful in this disease, compared with de novo acute
leukemia. The mean survival after diagnosis of blast crisis is only 2 to 4
months for nonresponders. In responders, standard chemotherapy yields
a response rate of approximately 30%. One-year survival for this group of
patients is 25%, but all of them eventually relapse with dismal prognosis.
Agents used in chronic phase, such as interferon and hydroxyurea, are ineffec-
tive in blast crisis. Table 1 summarizes various chemotherapy regimens that
have been used in treatment of CMLBC. The most commonly used induction
regimens have been a combination of an anthracycline (daunorubicin) or
anthracenedione (mitoxantrone) and cytarabine [22,23]. Mitoxantrone has
been used as a single agent or in combination, with some benefit [5,24].
Other agents in combination or as a single agent include 5-azacytidine, etopo-
side [4], carboplatin [24], cladribine [25], plicamycin, and hydroxyurea [26].
All-trans retinoic acid and arsenic trioxide, which has been used extensively
in acute promyelocytic leukemia, have been tried in CMLBC [27]. In 2002,
Bolaman et al. [5] reported a response rate of 54% in patients with CMLBC
using mitoxantrone, cytarabine, and high-dose methylprednisolone, but the
duration of response was short.
• Table 1 summarizes different chemotherapy treatments that have been evalu-
ated from 1982 to 2005. The response rates range from 11% to 77%, with
the median survival of responders ranging from 3.5 months to 19 months
[5,22–26,28–32], likely reflecting patient population or small numbers
treated. A few patients who responded to induction chemotherapy under-
went stem cell transplantation, with anecdotal reports of survivors.
• Approximately 30% of patients who develop CMLBC have a lymphoid
phenotype. In an evaluation of 97 consecutive patients with CMLBC,
patients with lymphoid CMLBC seldom had an accelerated phase before
blast crisis, had less frequent splenomegaly and hepatomegaly, and showed
a favorable response to chemotherapy regimens including vincristine and
prednisone [33]. However, remissions, if achieved, are generally very short.
In a series of 15 patients with extramedullary CMLBC, Specchia et al. [34]
Table 1. Available chemotherapy in blast crisis of CML (1982–present)
192

Median survival 1-year survival


Study Regimen Response rate Responders Nonresponders Responders Nonresponders

Schiffer et al. [28], n = 27 5-azacytidine/etoposide 58%; one CR, 7.7 months 2.5 months 20% 10%
15 PR
Koller and Miller [26], n = 9 Plicamycin/hydroxyurea 77%; seven CR 18 months NA NA NA
Kantarjian et al. [22], n = 27 Mitoxantrone/cytosine 30%; seven CR, 6 months 3 months 25% 8%
arabinoside one PR
Chronic Leukemia

Kantarjian et al. [23], n = 48 (CMLBC = 24) Daunorubicin/high-dose 33%; eight CHR 3.5 months 2 months 20% NA
cytosine arabinoside, GMCSF
Dutcher et al. [29], n = 40 Mitoxantrone/5-azacytidine 23%; five CR, 6+ months 3 months 37% 10%
two PR, two MR
Dutcher et al. [30]* (in accelerated phase), α-Interferon/plicamycin 30%; one CR, 5+ years 24 months 100%* 100%*
n = 13 (in accelerated phase) three PR
Dutcher et al. [31], n = 36 Carboplatin 22%; five CR, 12.8 months 3.5 months 50% NA
three PR
Robak and Gora-Tybor [25], n = 12 Mitoxantrone/cladribine 17%; zero CR, NA NA NA NA
two PR
Bolaman et al. [5], n = 14 Mitoxantrone/cytosine 64%; five CR, 19 months, CR; 2 months NA NA
arabinoside/high-dose four PR 14 months, PR
methyl prednisolone
Dutcher et al. [32], n = 18 All-trans retinoic acid 11%; one CR, 10 months 6 months 40% NA
one PR, three HI
Morris et al. [24], n = 20 Mitoxantrone/carboplatin 40%; five CR, 6.5 months 2 months 38% 17%
three PR
*This study was done in CML patients in accelerated phase.
CHR—complete hematologic response; CML—chronic myelogenous leukemia; CMLBC—chronic myelogenous leukemia blast crisis; CR—complete response; GMCSF—granulocyte-macrophage
colony-stimulating factor; HI—hematologic improvement; MR—major response; NA—not available; PR—partial response.
(Adapted from Morris and Dutcher [60••]; with permission.)
Blastic Phase of Chronic Myelogenous Leukemia Karbasian Esfahani et al. 193

had a complete remission in all of the patients with lymphoid extramedul-


lary CMLBC using vincristine, daunorubicin, and prednisone. No single
regimen has emerged as a preferred treatment of CMLBC.

Allogeneic bone marrow transplantation


• Allogeneic bone marrow transplantation (BMT) is often considered the
only established “curative” therapy in chronic phase of CML [35••]. How-
ever, allogeneic BMT with a related or unrelated donor, a consideration
in patients with CMLBC, is usually unsuccessful in this later stage of the
disease, although the results report 10% to 20% of patients achieving long-
term disease-free survival. Transplantation in blastic phase is characterized
by an increased rate of leukemia relapses (80%) and treatment-related
mortality. Goldman et al. [36] in 1986 reported a series of patients with
CML who underwent allogeneic BMT. Two-year survival was 72%, with a
risk of relapse of 7% in patients treated in chronic phase, whereas patients
with more advanced disease had a 2-year survival of only 18%, with a
relapse rate of 42%. Fefer et al. [37], in a retrospective study, showed that
transplantation in CMLBC is associated with much lower long-term
survival (14%) in a follow-up time of 1 to 8 years, compared with patients
transplanted in chronic phase (58%).

Imatinib
• With the understanding that the consequence of the Philadelphia chromo-
some, translocation 9;22, was the formation of a unique gene product,
efforts were aimed at developing compounds that could selectively inhibit
the aberrant tyrosine kinase resulting from this molecular rearrangement.
Imatinib, which is a signal transduction inhibitor (STI), is a potent selective
competitive inhibitor of the BCR/ABL protein tyrosine kinase and is the
cornerstone of therapy in CML [38–41].
• Imatinib was developed by Lydon and Druker in collaboration with Ciba
Geigy (now Novartis Pharmaceuticals) in the early 1990s [42] and was first
administered to patients with CML in June 1998 [43]. It is orally adminis-
tered, with a bioavailability of almost 100%. It occupies the adenosine tri-
phosphate (ATP)-binding site of the ABL tyrosine kinase component of the
BCR-ABL oncoprotein and thereby prevents phosphorylation of any sub-
strate. It was designed to act by inhibiting the tyrosine kinase activity of
the BCR-ABL, but it was also found to inhibit the tyrosine kinases coded by
platelet-derived growth factor receptor (PDGF-r) and c-KIT (stem cell factor
receptor) [35••]. Although it is highly effective in the chronic phase of CML,
its role is less clear in CMLBC. Initially, the use of imatinib in these patients
generated hopes of better treatment options (Table 2). Patients with Ph1-
positive CML in blast crisis were treated with imatinib at doses ranging from
300 to 1000 mg/day. Druker et al. [6•] reported responses to imatinib among
58 treated patients with CMLBC. Although there was a higher response rate
among those with lymphoid blast crisis (70%), none remained in remission.
The response rate in myeloid blast crisis was 55%, with 50% having complete
hematologic responses (CHRs), and 36% of these responders remained in
remission for 3 to 12 months [6•]. None of these patients had received
imatinib in chronic phase.
• In a study by Kantarjian et al. [44], imatinib was used in patients with CMLBC
(65 patients with nonlymphoid blast crisis and 10 patients with lymphoid
blast crisis; they received 300–1000 mg daily of imatinib). Response rates
were similar between nonlymphoid and lymphoid groups (52%), with 16
Table 2. Imatinib in CMLBC
194

Cytogenetic Response duration, Median survival,


Study Daily dose, mg Response rate response rate months months 1-year survival

Druker et al. [6•], n = 588, 300–1000 Myeloid blast crisis: 55%, four CR; NA 3.3–11.6 NA NA
lymphoid = 20, myeloid = 39 lymphoid blast crisis/ALL: 70%,
four CR
Kantarjian et al. [44], n = 75, 300–1000 52%; 16 CR, three PR, no 16% NA 6.5 22%
lymphoid = 10, nonlymphoid = 65 difference lymphoid/nonlymphoid
Chronic Leukemia

Sawyers et al. [45], n = 260 400–600 52%, with 31% > 4 weeks; CHR 8% 7% 10 6.9 (7.3 for 600 mg) 32%
Sureda et al. [40], n = 30 600 60% > 4 weeks 8% 5 NA 36% ± 13%
Zhang et al. [46], n = 19 400/600 + 57%; six CR, two PR NA NA NA 38%
cytarabine
ALL—acute lymphoblastic leukemia; CHR—complete hematologic response; CMLBC—chronic myelogenous leukemia blast crisis; CR—complete response; NA—not available;
PR—partial response.
(From Morris and Dutcher [60••]; with permission.)
Blastic Phase of Chronic Myelogenous Leukemia Karbasian Esfahani et al. 195

complete and three partial hematologic responses; 12 had hematologic


improvement, and seven returned to chronic phase. The overall median sur-
vival was 6.5 months, and the 1-year survival was 22%.
• Concurrently, during a phase II trial of imatinib in CMLBC, Sawyers et al.
[45] evaluated 229 patients. The hematologic response was 52%, with a
sustained response of 31% (beyond 4 weeks with median of 10 months).
CHRs were reported in 8% of patients, with major cytogenetic responses
(CyRs) in 16% and complete CyRs in 7%. The median survival of all
treated patients was 6.9 months. Using imatinib with a higher dosage of
600 mg daily appeared to produce a longer median survival (7.3 months),
compared with 4.5 months with 400 mg daily.
• Other smaller series have been reported with response rates of 57% to 60%
using different dosages of imatinib (Table 2). Sureda et al. [40] reported 18
of 30 (60%) patients achieving sustained hematologic response (> 4 weeks),
with a median duration of 5 months. They observed a better response to
higher doses of imatinib. Also, the presence of a complex karyotype and/or a
long time interval between the diagnosis of the blast crisis and the institution
of imatinib therapy significantly worsened the overall survival and event-free
survival of patients [40].
• In a study by Zhang et al. [46], 19 patients were treated with imatinib
400 mg/day plus standard cytarabine-based chemotherapy. After 4 weeks,
if no remission was obtained, imatinib was increased to 600 mg/day for
another 8 weeks. Induction chemotherapy could not achieve a remission
in most patients (16/19), but prolonged high-dose imatinib produced a
complete remission in six of 16 patients. Also, two patients had partial
remissions and one returned to the chronic phase, with 1-year survival of
38% in responding patients.
• Side effects of imatinib include myelosuppression and toxicities observed
with its use in chronic phase. Reports have shown that significant progres-
sion of marrow reticulin fibrosis during imatinib therapy can be an indica-
tor for progression of CML [27]. Also, imatinib may promote cytogenetic
clonal evolution, resulting in a poor response to treatment [27]. Resistance
also has been described [47]. Pfeifer et al. [48] reported three patients with
CML who rapidly progressed from chronic phase to blast crisis while taking
imatinib for 7 to 10 months. Increased frequency of extramedullary blast
crisis, including central nervous system relapse, has been reported [48,49].
Imatinib has a limited ability to penetrate the intact blood-brain barrier [50]
and thus may not be sufficient therapy for patients in blast crisis with central
nervous system involvement [49]. Drug resistance has been associated with
molecular or cytogenetic causes such as reactivation of Bcr/Abl signal trans-
duction, amino acid substitutions that changed the conformation of the
ATP-binding site, and additional chromosomal aberrations [51].
• Relapses of imatinib-treated chronic phase patients have demonstrated new
clinical patterns, including patients with complete CyR who have abruptly
developed blast crisis, suggesting that mechanisms additional to BCR-ABL
are important in the transition to blast crisis [49,52].
• At present, the role of imatinib in patients in blast crisis is not clear,
particularly because many have now received imatinib in chronic phase.
Most patients with CMLBC who respond will relapse relatively quickly
[10]. Nevertheless, imatinib provides a less toxic induction therapy and
is associated with fewer deaths during induction [44]. In vitro culture
studies have indicated that CD34+ CML progenitor cells can remain
viable in a quiescent state in the presence of imatinib and growth factors.
These cells could be responsible for resistance or relapse during treatment
with imatinib or after cessation of imatinib therapy [53]. To overcome
196 Chronic Leukemia

Table 3. Dasatinib in CMLBC and accelerated phase


Study Dosage Response rate Cytogenetic response

Talpaz and Rousselot [55•], n = 74 70 mg orally twice daily (dose 55% MHR, 24% CHR 45%, with 21% CR and 17% PR
escalation: 100 mg orally twice
daily; dose reduction: 50 and
40 mg orally twice daily)
Guilhot et al. [56•], n = 35 70 mg orally twice daily (dose 66% MHR, 20% CHR 54%, with four of 24 complete
escalation: 100 mg orally twice CyR (0% Ph+) and two of 24
daily; dose reduction: 50 and partial CyR (1% to 35% Ph+)
40 mg orally twice daily)
CHR—complete hematologic response; CMLBC—chronic myelogenous leukemia blast crisis; CR—complete response; CyR—cytogenetic
response; MHR—major hematologic response; Ph—Philadelphia chromosome; PR—partial response.

resistance to imatinib, several approaches have been studied, including


escalation of the dose of imatinib and the combination of imatinib with
chemotherapeutic drugs [54].

Dasatinib
• Dasatinib (BMS-354825) is an oral multitargeted kinase inhibitor, which
targets BCR-ABL and SRC kinases. Talpaz and Rousselot [55•] conducted
a phase II trial with dasatinib in myeloid CMLBC. Seventy-four imatinib-
resistant (IM-R) or intolerant (IM-I) patients participated. The preliminary
data were reported for 29 IM-R and five IM-I patients. Median age of the
patients was 54 years (71% male and 29% female), and median duration of
CML from first diagnosis was 49.3 months. In 44% of patients, the highest
dose of imatinib was 600 mg/day, and 41% of patients received imatinib
for more than 3 years, with CHR in 82% of patients. Other prior treatments
included BMT and interferon. Dasatinib was administered at the dosage of
70 mg orally twice daily, with dose escalation to 100 mg orally twice daily
(in 32% of patients) or dose reduction to 50 mg and 40 mg orally twice
daily in some patients based on the hematologic response or persistent
toxicity. The results were as follows: major hematologic response was 16/29
(55%), with seven CHRs; nine patients had no evidence of leukemia; 13
patients (45%) showed CyR, including six (21%) complete (0% Ph+) and
five (17%) partial responses (1% to 35% Ph+) [55•]. A full report on the
74 patients is awaited with interest (Table 3).
• Dasatinib also has been used in patients with accelerated-phase CML who
are resistant or intolerant to imatinib (Table 3). In a phase II study, prelim-
inary results were reported on 35 patients, using the earlier-mentioned
dosages in the study by Talpaz and Rousselot [55•]. Major hematologic
response was reported in 23 patients (66%), with seven CHRs and 16
patients with no evidence of leukemia. In addition, CyR was seen in 13/24
patients (54%), with four complete CyRs and two partial CyRs [56•]. Side
effects of dasatinib include profound myelosuppression. Nonhematologic
toxicities were uncommon.
• These data suggest that dasatinib has potent activity with high hematologic
and CyR rates in myeloid CMLBC and CML accelerated phase in patients
who are resistant or intolerant to imatinib (Table 3). Further clinical evalu-
ation and understanding of molecular pathways for overcoming resistance
is currently underway.
Blastic Phase of Chronic Myelogenous Leukemia Karbasian Esfahani et al. 197

Emerging therapies
• Parthenolide, a naturally occurring small molecule, induces robust apoptosis
in primary human acute myelocytic leukemia cells and CMLBC cells, while
sparing normal hematopoietic cells. The activity of parthenolide triggers
leukemia stem cell (LSC)-specific apoptosis and as such may represent a
potentially important new class of drugs for LSC-targeted therapy [57].
• Recent studies by Yu et al. [58] and Dai et al. [59] examined the interaction
between bortezomib, a proteosome inhibitor, and histone deacetylase
inhibitors in BCR-ABL (+) human leukemia cells and have demonstrated
synergistic anti-leukemic activity. There was an increase in mitochondrial
injury, caspase activation, and apoptosis. This was evident even in apoptosis-
resistant cell lines. These data also can be viewed as an efficient way to treat
CMLBC. Clinical evaluation is ongoing.

Future directions
• Further studies of the mechanisms of transformation of chronic phase
CMLBC at a molecular level, and methods to target these molecular abnor-
malities, will determine the future direction of new treatment modalities.
Different chemotherapy agents and STIs have been tried. The prognosis of
CML in blast crisis remains disappointing, despite these efforts. Currently,
the most successful strategy for improving survival in CML is by prolonging
the chronic phase and delaying the onset of blast crisis. Nevertheless, there
is considerable enthusiasm that further understanding of the molecular
events may lead to additional targeted approaches to then prevent the
transformation to CMLBC. Imatinib and subsequently dasatinib trial
results have been promising in treating the CMLBC. In the interim, clinical
trials in CMLBC using agents that may induce a return to chronic phase
continue to be important.

References and Recommended Reading


Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
1.• Faderl S, Talpaz M, Estrov Z, et al.: Chronic myelogenous 5. Bolaman Z, Koseoglu M, Ayyildiz O, et al.: Treatment of
leukemia: biology and therapy. Ann Intern Med 1999, blastic phase chronic myeloid leukemia with mitox-
131:207–219; and The biology of chronic myeloid antrone, cytosine arabinoside and high dose methyl-
leukemia. N Engl J Med 1999, 341:164. prednisolone. Hematologica 2002, 32:49–57.
This study provides a useful overview of switching treatment 6.• Druker BJ, Sawyers CL, Kantarjian HM, et al.: Activity of
of CML from chemotherapy to targeted therapy based on the a specific inhibitor of the BCR-ABL tyrosine kinase
biology of the disease. in the blast crisis of chronic myeloid leukemia and
2. Deininger MW, Goldman JM, Melo JV: The molecular acute lymphoblastic leukemia with the Philadelphia
biology of chronic myeloid leukemia. Blood 2000, chromosome. N Engl J Med 2001, 344:1038–1042.
96:3343–3356. This pilot study is one of the pioneer studies that evaluated the
3. Giles FJ, Cortes JE, Kantarjian HM, et al.: Accelerated effect of tyrosine kinase inhibition in patients with CMLBC
and blastic phases of chronic myelogenous leukemia. and Ph-positive ALL.
Hematol Oncol Clin North Am 2004, 18:753–774. 7. Soverini S, Martinelli G, Rosti G, et al.: ABL mutations
4. Axdorph U, Stenke L, Grimfors G, et al.: Intensive in late chronic phase chronic myeloid leukemia
chemotherapy in patients with chronic myelogenous patients with up-front cytogenetic resistance to ima-
leukemia (CML) in accelerated or blastic phase--a tinib are associated with a greater likelihood of pro-
report from the Swedish CML group. Br J Haematol gression to blast crisis and shorter survival: a study
2002, 118:1048–1054. by the GIMEMA Working Party on Chronic Myeloid
Leukemia. J Clin Oncol 2005, 23:4100–4109.
198 Chronic Leukemia

8. Ilaria RL Jr: Pathobiology of lymphoid and myeloid 24. Morris EL, Dutcher JP, Paietta E, et al.: A pilot study of
blast crisis and management issues. Hematology Am Soc carboplatin and mitoxantrone in blast crisis of chronic
Hematol Educ Program 2005, 188–194. myeloid leukemia. Leuk Lymphoma 2006, Submitted.
9. Goldman JM, Melo JV: Chronic myeloid leukemia-- 25. Robak T, Gora-Tybor J: Cladribine combined with
advances in biology and new approaches to treat- mitoxantrone in the treatment of blastic phase of
ment. N Engl J Med 2003, 349:1451–1464. chronic myeloid leukemia. Neoplasm 2001, 48:203–207.
10. Kurzrock R, Kantarjian HM, Druker BJ, Talpaz M: Phila- 26. Koller CA, Miller DM: Preliminary observations on the
delphia chromosome-positive leukemias: from basic therapy of the myeloid blast phase of chronic granu-
mechanisms to molecular therapeutics. Ann Intern Med locytic leukemia with plicamycin and hydroxyurea.
2003, 138:819–830. N Engl J Med 1986, 315:1433–1438.
11. Warmuth M, Danhauser-Riedl S, Hallek M: Molecular 27. Kantarjian HM, Keating MJ, Talpaz M, et al.: Chronic
pathogenesis of chronic myeloid leukemia: implica- myelogenous leukemia in blast crisis. Analysis of 242
tions for new therapeutic strategies. Ann Hematol 1999, patients. Am J Med 1987, 83:445–454.
78:49–64. 28. Schiffer CA, De Bellis R, Kasdorf H, et al.: Treatment of
12. Melo JV, Deininger MW: Biology of chronic myeloge- the blast crisis of chronic myelogenous leukemia with
nous leukemia--signaling pathways of initiation and 5-azacitidine and VP-16-213. Cancer Treat Rep 1982,
transformation. Hematol Oncol Clin North Am 2004, 66:267–271.
18:545–568. 29. Dutcher JP, Eudey L, Wiernik PH, et al.: Phase II study
13. Gordon MY, Dowding CR, Riley GP, et al.: Altered adhe- of mitoxantrone and 5-azacytidine for accelerated and
sive interactions with marrow stroma of hematopoietic blast crisis of chronic myelogenous leukemia: a study
progenitor cells in chronic myeloid leukemia. Nature of the Eastern Cooperative Oncology Group. Leukemia
1987, 328:342–344. 1992, 6:770–775.
14. Verfaillie CM, McCarthy JB, McGlave PB: Mechanisms 30. Dutcher JP, Coletti D, Paietta E, et al.: A pilot study of
underlying abnormal trafficking of malignant progen- alpha-interferon and plicamycin for accelerated
itors in chronic myelogenous leukemia. Decreased phase of chronic myeloid leukemia. Leuk Res 1997,
adhesion to stroma and fibronectin but increased 21:375–380.
adhesion to the basement membrane components 31. Dutcher JP, Lee S, Paietta E, et al.: Phase II study of car-
laminin and collagen type IV. J Clin Invest 1992, boplatin in blast crisis of chronic myeloid leukemia:
90:1232–1241. Eastern Cooperative Oncology Group study (E 1992).
15. Van Etten RA: Cycling, stressed-out and nervous: cellular Leukemia 1998, 12:1037–1040.
functions of c-Abl. Trends Cell Biol 1999, 9:179–186. 32. Dutcher JP, Lee S, Wiernik PH, et al.: Phase II study of
16. Calabretta B, Prerotti D: The biology of CML blast crisis. All-Trans retinoic acid (ATRA) in accelerated phase
Blood 2004, 103:4010. or early blastic phase of chronic myeloid leukemia.
17. Deininger MW, Goldman JM, Melo JV: The molecular A study of the Eastern Cooperative Oncology Group
biology of chronic myeloid leukemia. Blood 2000, (E1993). Leuk Lymphoma 2005, 46:377–385.
96:3343–3356. 33. Cervantes F, Villamor N, Esteve J, et al.: “Lymphoid” blast
18. Jamieson CH, Ailles LE, Dylla SJ, et al.: Granulocyte- crisis of chronic myeloid leukaemia is associated with
macrophage progenitors as candidate leukemic distinct clinicohaematological features. Br J Haematol
stem cells in blast-crisis CML. N Engl J Med 2004, 1998, 100:123–128.
351:657–667. 34. Specchia G, Palumbo G, Pastore D, et al.: Extra-medullary
19. Sinclair PB, Nacheva EP, Leversha M, et al.: Large dele- blast crisis in chronic myeloid leukemia. Leuk Res 1996,
tions at the t(9;22) breakpoint are common and may 20:905–908.
identify a poor-prognosis subgroup of patients with 35.••Neylon AJ, O’Brien SG: Diagnosis and treatment of
chronic myeloid leukemia. Blood 2000, 95:738–743. CML. In Neoplastic Diseases of the Blood, edn 4. Edited by
20. Dutcher JP, Wiernik PH: Accelerated and blastic phase Wiernik PH. New York: Cambridge University Press;
of chronic myeloid leukemia. Curr Treat Options Oncol 2003:53–69.
2000, 1:51–52. This chapter provides a comprehensive review of CML and
21. Ottmann OG, Druker BJ, Sawyers CL, et al.: A phase 2 CMLBC disease, including pathophysiology, diagnosis,
study of imatinib in patients with relapsed or refractory and treatment.
Philadelphia chromosome-positive acute lymphoid 36. Goldman JM, Apperley JF, Jones L, et al.: Bone marrow
leukemias. Blood 2002, 100:1965. transplantation for patients with chronic myeloid
22. Kantarjian HM, Walters RS, Keating MJ, et al.: Treatment leukemia. N Engl J Med 1986, 314:202–207.
of blastic phase of chronic myelogenous leukemia 37. Fefer A, Clift RA, Thomas ED: Allogeneic marrow trans-
with mitoxantrone and high-dose cytosine arabino- plantation for chronic granulocytic leukemia. J Natl
side. Cancer 1988, 62:672–676. Cancer Inst 1986, 76:1295–1299.
23. Kantarjian HM, Talpaz M, Gutterman J, et al.: Treatment 38. Kantarjian HM, Cortes JE, O’Brien S, et al.: Long-term
of chronic myelogenous leukemia in accelerated and survival benefit and improved complete cytogenetic
blastic phases with daunorubicin, high dose cytara- and molecular response rates with imatinib mesylate
bine, and granulocyte-macrophage colony-stimulating in Philadelphia chromosome-positive chronic-phase
factor. J Clin Oncol 1992, 10:398–405. chronic myeloid leukemia after failure of interferon-
alpha. Blood 2004, 104:1979–1988.
Blastic Phase of Chronic Myelogenous Leukemia Karbasian Esfahani et al. 199

39. Marin D, Marktel S, Bua M, et al.: The use of imatinib 52. Breccia M, Nanni M, Mancini F, et al.: Extramedullary
(STI571) in chronic myeloid leukemia: some practical blast crisis occurring in a Philadelphia-positive
considerations. Haematologica 2002, 87:979–988. chronic myeloid leukemia patient with major cyto-
40. Sureda A, Carrasco M, de Miguel M, et al.: Imatinib genetic response to imatinib. Haematologica 2004,
mesylate as treatment for blastic transformation of 89:ECR11.
Philadelphia chromosome positive chronic myeloge- 53. Graham SM, Jorgensen HG, Allan E, et al.: Primitive,
nous leukemia. Haematologica 2003, 88:1213–1220. quiescent Philadelphia-positive stem cells from
41. Deininger MW, Goldman JM, Melo JV: The molecular patients with CML are insensitive to ST1571 in vitro.
biology of chronic myeloid leukemia. Blood 2000, Blood 2002, 99:319.
96:3343–3356. 54. Yoshida C, Melo JV: Biology of chronic myeloid leukemia
42. Druker BJ, Lydon NB: Lessons learned from the devel- and possible therapeutic approaches to imatinib-
opment of an Abl tyrosine kinase inhibitor for resistant disease. Int J Hematol 2004, 79:420–433.
chronic myelogenous leukemia. J Clin Invest 2000, 55.• Talpaz M, Rousselot P: A phase II study of dasatinib in
105:3–7. patients with CMLBC who are resistant or intolerant
43. Marin D, Marktel S, Bua M, et al.: The use of imatinib to imatinib: first results of the CA 180006 ‘START-B’
(STI571) in chronic myeloid leukemia: some practical study [abstract 40]. http://www.abstracts2view.com/
considerations. Haematologica 2002, 87:979–988. hem_ash05atlanta/. Accessed February 6, 2006.
44. Kantarjian HM, Cortes J, O'Brien S, et al.: Imatinib This study describes the first phase II trial of dasatinib, with
mesylate (STI571) therapy for Philadelphia chromo- significant success rate in IM-R CMLBC patients.
some-positive chronic myelogenous leukemia in blast 56.• Guilhot F, Apperley JF, Shah N, et al.: A phase II study of
phase. Blood 2002, 99:3547–3553. dasatinib in patients with accelerated-phase CML who
45. Sawyers CL, Hochhaus A, Feldman E, et al.: Imatinib are resistant or intolerant to imatinib: first results of
induces hematologic and cytogenetic responses in the CA 180005 ‘START-A’ study [abstract 39]. http://
patients with chronic myelogenous leukemia in mye- www.abstracts2view.com/hem_ash05atlanta/. Accessed
loid blast crisis: results of a phase II study. Blood 2002, February 6, 2006.
99:3530–3539. This study describes the first phase II trial of dasatinib in
accelerated phase of CML. The results are important because
46. Zhang GC, Zheng D, Li QH, et al.: Efficacy of imatinib
the patients were all resistant or intolerant to imatinib.
mesylate, a specific inhibitor of BCR/ABL tyrosine
kinase, on chronic myeloid leukemia in blast phase. 57. Guzman ML, Rossi RM, Karnischky L, et al.: The sesquit-
Ai Zheng 2004, 23:1696–1699. erpene lactone parthenolide induces apoptosis of
human acute myelogenous leukemia stem and pro-
47. Weisberg E, Griffin JD: Resistance to imatinib (Glivec):
genitor cells. Blood 2005, 105:4163–4169.
update on clinical mechanisms. Drug Resist Updat
2003, 6:231–238. 58. Yu C, Rahmani M, Conrad D, et al.: The proteasome
inhibitor bortezomib interacts synergistically with
48. Pfeifer H, Wassmann B, Hofmann WK, et al.: Risk and
histone deacetylase inhibitors to induce apoptosis in
prognosis of central nervous system leukemia in Bcr/Abl+ cells sensitive and resistant to STI571. Blood
patients with Philadelphia chromosome-positive
2003, 10:3765–3774.
acute leukemias treated with imatinib mesylate.
Clin Cancer Res 2003, 9:4674–4681. 59. Dai Y, Rahmani M, Pei XY, et al.: Bortezomib and flavo-
piridol interact synergistically to induce apoptosis in
49. Bujassoum S, Rifkind J, Lipton JH: Isolated central ner-
chronic myeloid leukemia cells resistant to imatinib
vous system relapse in lymphoid blast crisis chronic mesylate through both Bcr/Abl-dependent and -inde-
myeloid leukemia and acute lymphoblastic leukemia
pendent mechanisms. Blood 2004, 104:509–518.
in patients on imatinib therapy. Leuk Lymphoma 2004,
45:401–403. 60.••Morris EL, Dutcher JP: Blastic phase of chronic myel-
ogenous leukemia. Clin Adv Hematol Oncol 2005,
50. Wolff NC, Richardson JA, Egorin M, et al.: The CNS is 3:547–552.
a sanctuary for leukemic cells in mice receiving
This is an important review of the molecular biology and
imatinib mesylate for BCR-ABL-induced leukemia. evolution of treatment of the blastic phase of CML.
Blood 2003, 101:5010.
51. Tsao AS, Kantarijian H, Talpaz M: STI-571 in chronic
myelogenous leukaemia. Br J Haematol 2002, 119:15.

You might also like