ArticlePDF Available

Impact of Additional Cytogenetic Abnormalities in Adults with Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia Undergoing Allogeneic Hematopoietic Cell Transplantation

Authors:

Abstract and Figures

The occurrence of additional cytogenetic abnormalities (ACA) is common in Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) but is of unknown significance in the tyrosine-kinase inhibitor (TKI) era. We retrospectively analyzed data from a consecutive case series of adults with Ph+ ALL who had undergone allogeneic hematopoietic cell transplantation (alloHCT) at City of Hope between 2003 and 2014. Among 130 adults with Ph+ ALL who had TKI therapy prior to alloHCT, 78 cases had available data on conventional cytogenetics at diagnosis and were eligible for outcomes analysis. ACA was observed in 41 cases (53%). There were no statistically significant differences in median age, median initial white blood count, post-HCT TKI maintenance, or disease status at the time of transplant between the Ph-chromosome only and ACA cohorts; however, the Ph-chromosome only cohort had a higher rate of minimal residual disease (MRD) positivity at the time of HCT. Three-year leukemia-free survival (LFS) [79.8% vs. 39.5%, p=0.01] and 3-year overall survival (OS) [83% vs. 45.6%, p = 0.02] were superior in the Ph chromosome only, compared to ACA cohorts, respectively. Monosomy 7 was the most common additional aberration observed in our ACA cohort (N=12). Thus, when TKI therapy and alloHCT are utilized as part of adult Ph+ ALL therapy, the presence of ACA appears to have a significant deleterious effect on outcomes post HCT. Copyright © 2015 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
The impact of additional cytogenetic abnormalities in adults with
Philadelphia chromosome-positive acute lymphoblastic
leukemia undergoing allogeneic hematopoietic cell
transplantation
Ibrahim Aldoss1, Tracey Stiller2, Thai M Cao1, Joycelynne M. Palmer2, Sandra H. Thomas1,
Stephen J. Forman1, and Vinod Pullarkat1
1Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
2Department of Information Science, City of Hope, Duarte, CA
Abstract
The occurrence of additional cytogenetic abnormalities (ACA) is common in Philadelphia
chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) but is of unknown significance in
the tyrosine-kinase inhibitor (TKI) era. We retrospectively analyzed data from a consecutive case
series of adults with Ph+ ALL who had undergone allogeneic hematopoietic cell transplantation
(alloHCT) at City of Hope between 2003 and 2014. Among 130 adults with Ph+ ALL who had
TKI therapy prior to alloHCT, 78 cases had available data on conventional cytogenetics at
diagnosis and were eligible for outcomes analysis. ACA was observed in 41 cases (53%). There
were no statistically significant differences in median age, median initial white blood count, post-
HCT TKI maintenance, or disease status at the time of transplant between the Ph-chromosome
only and ACA cohorts; however, the Ph-chromosome only cohort had a higher rate of minimal
residual disease (MRD) positivity at the time of HCT. Three-year leukemia-free survival (LFS)
[79.8% vs. 39.5%, p=0.01] and 3-year overall survival (OS) [83% vs. 45.6%, p = 0.02] were
superior in the Ph chromosome only, compared to ACA cohorts, respectively. Monosomy 7 was
the most common additional aberration observed in our ACA cohort (N=12). Thus, when TKI
therapy and alloHCT are utilized as part of adult Ph+ ALL therapy, the presence of ACA appears
to have a significant deleterious effect on outcomes post HCT.
Keywords
Philadelphia-chromosome positive; Ph+; additional cytogenetic abnormalities; monosomy 7;
allogeneic; stem cell transplant
Corresponding author: Ibrahim Aldoss, MD, Department of Hematology and Hematopoietic Cell Transplantation, 1500 E. Duarte
Road, Duarte, CA 91010-3000, Phone: 626-256-4673, Ext. 62405, Fax: 626-301-8116, ialdoss@coh.org.
Conflict of interest statement: None of the authors has conflicts of interest to declare.
HHS Public Access
Author manuscript
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Published in final edited form as:
Biol Blood Marrow Transplant. 2015 July ; 21(7): 1326–1329. doi:10.1016/j.bbmt.2015.03.021.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Introduction
Philadelphia chromosome (Ph) is the most common cytogenetic abnormality in adults with
acute lymphoblastic leukemia (ALL) [1, 2]. The presence of Ph chromosome has long been
considered an adverse prognostic feature and was associated with dismal outcomes in the
pre-tyrosine kinase inhibitor (TKI) era. For patients with Ph+ ALL treated with
chemotherapy alone, complete remission (CR) rates were low and leukemia relapse was the
norm in the absence of allogeneic hematopoietic stem cell transplantation (alloHCT) [3–6].
Therefore, alloHCT was routinely recommended for eligible patients with available donors.
However, the introduction of TKIs has drastically improved the outcome of Ph+ ALL, and
currently, remission is achieved in the majority of cases [7, 8]. Nonetheless, alloHCT
continues to be the preferred consolidation therapy in adults with Ph+ ALL due to lack of
long-term data on patients treated with combination chemotherapy and TKIs without
alloHCT.
Although additional cytogenetic abnormalities (ACA) at diagnosis are detected in
approximately two thirds of cases with Ph+ ALL [5, 9–11], its prognostic significance in
patients who are treated with TKI and alloHCT remains unclear. In this study, we
investigated the prognostic implications of ACA in adults with Ph+ ALL who had received
TKI therapy prior to consolidation with alloHCT.
Methods
We reviewed all cases of adult Ph+ ALL who had undergone alloHCT at City of Hope
Medical Center between 01/2003 and 04/2014. Only patients with available conventional
cytogenetic documentation of the Ph chromosome with an adequate number of analyzed
metaphases (≥ 20) were included in the outcomes analysis. We excluded patients younger
than 18 years of age and patients who did not receive pre-transplant TKI-based therapy.
Statistical analysis
Demographic, disease and treatment characteristics were summarized using descriptive
statistics. The Wilcoxon Rank Sum test and Chi-sqaure test were used to determine
differences in demographic and disease characteristics of interest. Survival estimates were
calculated using the Kaplan-Meier product-limit method [12] and confidence intervals were
calculated using the logit transformation and the Greenwood variance estimate [13].
Differences between Kaplan-Meier curves were determined using the log-rank test. Overall
survival (OS) was defined as time from date of transplant to death from any cause.
Leukemia-free survival (LFS) was measured as time from date of transplant to relapse or
death from any cause, whichever occurred first. Patients who were alive at the time of
analysis were censored at the last contact date.
Prognostic variables analyzed included patient age at transplant, initial white blood cell
count (WBC), disease status at transplant (CR1 versus beyond CR1), and cytogenetic risk
(isolated versus ACA, and isolated versus ACA with −7, versus ACA without −7). All
analyses were performed using SAS v09.3 (SAS Institute, Cary, NC). The data were locked
for analysis August 5, 2014.
Aldoss et al. Page 2
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Results
We identified 134 consecutive adults with Ph+ ALL who had undergone alloHCT during the
selected time period. Five cases were excluded due to the lack of pretransplant TKI therapy.
Fifty-one cases with Ph positivity demonstrated only by PCR and/or FISH for BCR-ABL
were excluded due to unavailability of conventional cytogenetics [n=28], no growth/
insufficient growth [n=12], or karyotyping that did not show t(9;22)(q34.1;q11.2) [n=11]).
Patient and disease characteristics are shown in Table 1.
Among the 78 cases who met the eligibility criteria, 41 (53%) had ACA and 37 had isolated
t(9;22). There were 4 cases of ≥3-way complex BCR-ABL1 translocations (variant
translocations with one or more chromosomes involved in addition to 9 and 22) involving
chromosomes 6, 7, 10, 14 and 19, of which 3 cases were in the isolated Ph+ subgroup and 1
case was in the ACA subgroup. The median age was 41.4 years (19.8–68.4). Forty-nine of
the patients (63%) were male. Donors were matched siblings in 35 patients (45%). The
source of stem cells was peripheral blood in 65 patients (83%). The conditioning regimen
was myeloablative in 59 patients (76%). Tacrolimus/sirolimus-based GVHD prophylaxis
was the most commonly used conditioning regimen, in 59 patients (76%). Patient disease
and transplant characteristics are shown in Table 1.
Monosomy 7 (−7) was the most commonly observed ACA (29%, N =12) and was the only
ACA in 8 cases. The second most common abnormality was derivative chromosome 22 (N =
9). There were 6 cases of hyperdiploidy (> 50 chromosomes) in the ACA cohort. The most
common trisomies were 6, 8 and 21, seen in 5, 8 and 5 cases, respectively. Most trisomies
were part of other additional cytogenetic abnormalities. Excluding cases with −7, there were
8 cases that met the definition of monosomal karyotype (2 autosomal monosomies or 1
monosomy combined with structural aberration). There was one case of concurrent
occurrence of mixed myeloid leukemia (MLL) and Ph+ rearrangements. Cytogenetic
abnormalities are summarized in Table 1. There was no difference in median age (p=0.08),
median initial WBC (p=0.99), disease status at the time of transplant (p=0.58), or post-HCT
TKI maintenance therapy (p=0.93), between the isolated Ph-chromosome and ACA cohorts.
However, more patients with Philadelphia-chromosome alone had minimal residual disease
(MRD) positivity (based on BCR/ABL by PCR) prior to HCT compared to patients with
ACA (p=0.0088), as seen in Table 1. The median follow-up for surviving patients from the
time of transplant was 43 months (range: 4.1–133 months) and 47 months (range: 3.2–123
months) for isolated t(9;22) and ACA cohorts, respectively. Five (14%) patients relapsed in
the isolated t(9;22) group and eight (20%) relapsed in the ACA group. One-year non-relapse
mortality (NRM) for the whole cohort was 20%. The 3-year OS of 45.6% (95% CI: 28.2–
61.5) in the ACA group was significantly worse than the 83% (95% CI: 65.9–92.0) seen in
the isolated Ph chromosome group (p =0.02). Likewise, the 3-year LFS of 39.5% (95% CI:
23.3–55.3) for ACA was significantly worse than the 79.8% (95% CI: 62.2–89.9) in the
isolated Ph+ group (p=0.01) (Figure 1). No multivariate analysis was performed given the
small study size and the fact that no significant differences in OS or LFS were seen in
univariate analysis other than the presence of ACA. The 3-year OS and LFS for the 51 Ph+
patients excluded from the analysis were 53.9% and 38.6%, respectively.
Aldoss et al. Page 3
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Since monosomy 7 was the most common ACA, we examined its impact on HCT outcomes
by splitting the Ph+ cohort into three subgroups: isolated Ph chromosome, Ph+ with − 7, and
Ph+ with other ACA. Both 3-year OS and LFS were inferior in patients with Ph+ and other
ACA (non-7) compared to isolated Ph+ and Ph+ with −7 cohorts (Figure 2). Three-year OS
for Ph+ (other ACA) was 39.7% (95%CI: 20.3–58.6) vs. 83.0% (95%CI: 65.9–92.0) for Ph+
(alone) vs. 59.7% (95%CI: 24.1–82.9) for Ph+ (with −7), (p=0.01). Three-year LFS was
30.2% (95%CI: 13.3–49.0) for Ph+ (other ACA) vs. 79.8% (95%CI: 62.2–89.9) for Ph+
(alone) vs. 60.6% (95%CI: 25.8–83.1) Ph+ (with −7), (p=0.001)
Discussion
Our study is unique in that it examines the impact of ACA in Ph+ patients treated with TKIs
and consolidated with alloHCT, and therefore, it is relevant to contemporary management of
Ph+ALL compared to previous reports examining this issue [5, 9].
Additional cytogenetic abnormalities (ACA) occur frequently in Ph+ ALL, and have been
reported in over 60% of cases [5, 9–11]. Additional aberrations involving chromosomes 8, 7,
9, 21 and 22 were most commonly reported [9, 10]. In the pre-TKI era, two studies
investigated the prognostic significance of ACA in Ph+ ALL. The first study included 249
children with Ph+ ALL from 10 large pediatric groups diagnosed between 1986 and 1996.
Patients were treated with different regimens and only 67 patients (27%) underwent
alloHCT as part of their ALL treatment. Event free survival (EFS) and overall survival (OS)
were lower in patients with ACA and loss of chromosome 7, 7p, and/or 9p, but the inferior
outcome was not significant when adjusted to other prognostic factors [5]. The second
analysis included 111 adults with Ph+ ALL who were treated in one of 7 different CALGB
studies between 1985 and 2000. Only 24 patients (22%) underwent alloHCT. Monosomy 7
was associated with a lower CR rate, and complex cytogenetics (≥ 3 abnormalities) was
associated with higher CR rate. The presence of +der(22)t(9;22) was associated with higher
risk of relapse [9]. However, these previous two studies included patients who did not
receive TKIs as part of ALL therapy, and alloHCT was only utilized in approximately a
quarter of the cases.
Our analysis shows inferior LFS and OS in patients with Ph+ ALL with ACA compared to
those with isolated t(9;22) despite a higher rate of pre-HCT MRD positivity in the latter
group. Monosomy 7 was the most common additional abnormality in our cohort and usually
occurred as an isolated extra finding. The inferior outcome of ACA cohort was more
pronounced in patients without monosomy 7 compared to those with monosomy 7. The
basis for the worse LFS in the ACA group without monosomy 7 remains unclear and may be
related to the specific nature of the more prevalent abnormalities in this group. The small
number of cases makes it difficult to draw firm conclusions regarding this observation.
In conclusion, the presence of ACA, especially in patients without monosomy 7, appears to
adversely impact the post-transplant outcomes in Ph+ALL when TKI and alloHCT are
utilized as part of the treatment program. Due to their poor outcomes, these patients are
probably best served if treated in the context of a clinical trial. We believe this data supports
further investigation in an unbiased prospective dataset, which might identify a higher risk
Aldoss et al. Page 4
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
group of Ph+ ALL who could benefit from additional treatment such as post-transplant
maintenance TKI therapy to reduce relapse risk.
Acknowledgments
This work was partially supported by the City of Hope Comprehensive Cancer Center support grant NCI CA33572.
References
1. Moorman AV, Harrison CJ, Buck GA, et al. Karyotype is an independent prognostic factor in adult
acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the
Medical Research Council (MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993
trial. Blood. 2007; 109:3189–3197. [PubMed: 17170120]
2. Cytogenetic abnormalities in adult acute lymphoblastic leukemia: correlations with hematologic
findings outcome. A Collaborative Study of the Group Francais de Cytogenetique Hematologique.
Blood. 1996; 87:3135–3142. [PubMed: 8605327]
3. Wetzler M, Dodge RK, Mrózek K, et al. Prospective Karyotype Analysis in Adult Acute
Lymphoblastic Leukemia: The Cancer and Leukemia Group B Experience. Blood. 1999; 93:3983–
3993. [PubMed: 10339508]
4. Secker-Walker LM, Prentice HG, Durrant J, Richards S, Hall E, Harrison G. Cytogenetics adds
independent prognostic information in adults with acute lymphoblastic leukaemia on MRC trial
UKALL XA. MRC Adult Leukaemia Working Party. Br J Haematol. 1997; 96:601–610. [PubMed:
9054669]
5. Dombret H, Gabert J, Boiron JM, et al. Outcome of treatment in adults with Philadelphia
chromosome-positive acute lymphoblastic leukemia--results of the prospective multicenter
LALA-94 trial. Blood. 2002; 100:2357–2366. [PubMed: 12239143]
6. Gleissner B, Gokbuget N, Bartram CR, et al. Leading prognostic relevance of the BCR-ABL
translocation in adult acute B-lineage lymphoblastic leukemia: a prospective study of the German
Multicenter Trial Group and confirmed polymerase chain reaction analysis. Blood. 2002; 99:1536–
1543. [PubMed: 11861265]
7. Yanada M, Takeuchi J, Sugiura I, et al. High complete remission rate and promising outcome by
combination of imatinib and chemotherapy for newly diagnosed BCR-ABL-positive acute
lymphoblastic leukemia: a phase II study by the Japan Adult Leukemia Study Group. J Clin Oncol.
2006; 24:460–466. [PubMed: 16344315]
8. Bassan R, Rossi G, Pogliani EM, et al. Chemotherapy-phased imatinib pulses improve long-term
outcome of adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia:
Northern Italy Leukemia Group protocol 09/00. J Clin Oncol. 2010; 28:3644–3652. [PubMed:
20606084]
9. Wetzler M, Dodge RK, Mrozek K, et al. Additional cytogenetic abnormalities in adults with
Philadelphia chromosome-positive acute lymphoblastic leukaemia: a study of the Cancer and
Leukaemia Group B. Br J Haematol. 2004; 124:275–288. [PubMed: 14717774]
10. Heerema NA, Harbott J, Galimberti S, et al. Secondary cytogenetic aberrations in childhood
Philadelphia chromosome positive acute lymphoblastic leukemia are nonrandom and may be
associated with outcome. Leukemia. 2004; 18:693–702. [PubMed: 15044926]
11. Ko BS, Tang JL, Lee FY, et al. Additional chromosomal abnormalities and variability of BCR
breakpoints in Philadelphia chromosome/BCR-ABL-positive acute lymphoblastic leukemia in
Taiwan. Am J Hematol. 2002; 71:291–299. [PubMed: 12447959]
12. Kaplan G, Meier P. Non-parametric estimations from incomplete observations. J Am Stat Assoc.
1958; 53:457–481.
13. Breslow NE, Day NE. Statistical methods in cancer research: volume II, the design and analysis of
cohort studies. IARC Sci Publ. 1987; 82:1–406.
Aldoss et al. Page 5
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Highlights
Additional cytogenetic abnormalities (ACA) are common in Ph+ ALL
Monosomy 7 was the most common ACA in our cohort
When TKI therapy and allogeneic HCT were utilized as part of adult Ph+ ALL
therapy, the presence of ACA appeared to have a significant deleterious effect
on outcomes
Aldoss et al. Page 6
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 1. Survival outcomes stratified by presence/absence of ACA
Panel A plots overall survival (OS) and Panel B plots leukemia-free survival (LFS) by
Kaplan-Meier. Solid lines represent patients with isolated Ph+ (n=37), and dashed lines
represent patients with Ph+ and additional cytogenetic abnormalities (ACA) (n=41).
Aldoss et al. Page 7
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 2. Survival outcomes stratified by presence/absence of monosomy 7 and other ACA
Panel A plots overall survival (OS) and Panel B plots leukemia-free survival (LFS) by
Kaplan-Meier. Solid black lines represent patients with isolated Ph+ (n=37), solid grey lines
represent patients with Ph+ and monosomy 7 (n=12), and dashed lines represent patients
with Ph+ and non-monosomy 7 additional cytogenetic abnormalities (ACA) (n=29).
Aldoss et al. Page 8
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Aldoss et al. Page 9
Table 1
Patient, disease, and treatment characteristics
Variables
No t(9;22)
Cytogenetics*
(N=51)
Isolated Ph+
Cytogenetics
(N=37)
Ph+ plus
ACA
(N=41)
Age (yrs) 44.3 (18.9–68.5) 49 (19.8–68.4) 38.6 (21.4–61.0)
Gender
Female 22 (43) 17 (46) 13 (32)
Male 29 (57) 20 (54) 28 (68)
WBC 18.2 (1.2–425) 37.1 (1.1–440.0) 22.6 (1.2–426.0)
Disease Status
CR1 37 (73) 31 (84) 34 (83)
CR2 or beyond 10 (20) 3 (8) 4 (10)
Active disease at HCT 4 (8) 3 (8) 3 (7)
Cytogenetic Remission** 40/41 (98) 26/29 (90) 29/33 (88)
MRD Status Pre-HCT
MRD positive 14 (27) 17 (46) 9 (22)
MRD negative 28 (55) 13 (35) 27 (66)
Unknown 9 (18) 7 (19) 5 (12)
Time from Diagnosis to Transplant (days) 210 (75–1748) 135 (71–434) 177 (68–1,162)
Pre-HCT TKI
Imatinib 35 (67) 23 (62) 29 (71)
Dasatinib 11 (23) 11 (30) 7 (17)
Imatinib/Dasatinib 5 (10) 3 (8) 5 (12)
Post-HCT TKI Maintenance
Yes 18 (35) 19 (51) 21 (51)
No 30 (59) 16 (43) 17 (41)
Not Applicable#3 (6) 2 (5) 3 (7)
Donor Type
Sibling 29 (57) 16 (43) 19 (46)
Unrelated 22 (43) 21 (57) 22 (54)
Stem Cell Source
Bone Marrow 3 (6) 2 (5) 6 (15)
Cord Blood/Double Cord 4 (8) 0 (0) 6 (15)
Peripheral Blood 44 (86) 35 (95) 29 (71)
Conditioning Regimen Intensity
RIC/NMA 14 (27) 11 (30) 9 (22)
MAC 37 (73) 26 (70) 32 (78)
Conditioning Regimens
FTBI/Etoposoide 29 (57) 25 (68) 27 (66)
Fludarabine/Melphalan 15 (29) 10 (27) 6 (15)
Cytoxan/Fludarabine/TBI 7 (14) 0 (0) 6 (15)
Others 0 (0) 2 (5) 2 (5)
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Aldoss et al. Page 10
Variables
No t(9;22)
Cytogenetics*
(N=51)
Isolated Ph+
Cytogenetics
(N=37)
Ph+ plus
ACA
(N=41)
Cytogenetic abnormalities
Ph+ by FISH or PCR only 51 (100) 0
Ph+ by conventional cytogenetics 0 37 (10) 41 (10)
−7 NA 0 12
−8, i(8) or +8 NA 0 11
9p del, −9, der(9), add(9p) or +9 NA 0 11
der(22) NA 0 9
Monosomy, excluding −7 NA 0 8
Hyperdiploidy NA 0 6
*By conventional cytogenetics
**Patients in CR1 or CR2 with available cytogenetics (denominator) who were also in cytogenetic remission (numerator)
#Death within 60 days of HCT
Ph+ – Philadelphia Chromosome positive, ACA – additional cytogenetic abnormalities, RIC – reduced intensity conditioning, NMA – non-
myeloablative, MAC – myeloablative conditioning, WBC – white blood cell count, CR1
Biol Blood Marrow Transplant. Author manuscript; available in PMC 2016 July 01.
... The presence of additional cytogenetic aberrations (ACAs) has been reported in approximately 40-80% of patients with Ph + ALL [9][10][11][12]. Several previous studies have investigated the prognostic role of ACA in Ph + ALL, but there was no uniform conclusion [10,[12][13]. The study aims to investigate the prognosis signi cance of common ACAs for Ph + ALL patients who had received HSCT in the TKI era. ...
... The chromosomal abnormality − 7 is the common ACA in the current cohort, as previously reported [10,12,13,24]. The analysis shows higher CIR and inferior PFS and OS in patients with Ph + ALL with − 7 compared to the sole t(9;22) group and the ACA group without monosomy 7. The prognostic value of monosomy 7 in haematological malignancy has been widely discussed. ...
... In addition, some studies have demonstrated that − 7 or 7q-is inextricably linked to an inferior survival and prognosis in Ph + ALL, consistent with the main ndings of the current study [29][30]. However, several other studies did not show an association between − 7 and poor outcomes [13,23]. ...
Preprint
Full-text available
Additional chromosomal abnormalities(ACAs) at diagnosis are associated with inferior prognosis in chronic myelogenous leukemia. However, the prognostic significance of ACAs in adult patients with Philadelphia Chromosome Positive acute lymphoblastic leukemia (Ph + ALL) receiving TKI-targeted drugs and allogeneic hematopoietic stem cell transplantation(HSCT) is unknown. One hundred eighty-eight adult patients with Ph + ALL were retrospectively analysed, evaluating the effect of ACAs on outcomes of transplantation. One hundred thirty-six patients were included in the study. ACAs are observed in 60 cases (44%). The major-route ACAs are detected in more than 5% are as follows: +der(22), +der(9), + 8, -7 and complex karyotype. The median follow-up was 26.4 months. In the subgroup analyses of major route ACAs, three-year cumulative incidence of relapse (CIR) and progression-free survival(PFS) are statistically significant in + 8[66.7% vs.23.7%, P = 0.024; 77.8% vs. 23.7%, P = 0.0087], -7[53.8% vs. 23.7%, P = 0.035%; 61.5% vs. 32.9%, P = 0.033], and complex karyotypes[42.9% vs. 23.7%, P = 0.027; 47.6% vs. 23.7%] compared with isolated t(9;22). Additionally, the 3-year CIR for Ph + ALL with + der(22) is 44% vs. 23.7% for standard Ph + ALL(P = 0.045). The 3-year overall survival (OS) in the − 7 group is 46.5%, which is statistically significant compared with the other groups(P = 0.001). In multivariate analyses, three years CIR and PFS are statistically significant in + der(22), + 8, -7 and complex karyotype compared with standard Ph + ALL(P < 0.05). More importantly, Ph + ALL with − 7 was negatively associated with the rate of 3-year OS(P = 0.012). Partial ACAs at diagnosis appear to have a significant prognostic impact on transplantation outcomes in patients with Ph + ALL.
... A complex karyotype was defined by the presence of at least 3 abnormalities (ie, t[9;22] and 2 or more additional aberrations) as previously described. [7][8][9] We analyzed data from 206 de novo adult Ph+ALL patients (Suppl. Table S1). ...
... The OS of high-risk patients was worse than that of low-risk patients, even when censored at the time of allo-SCT ( Although it has been reported that +der(22)t(9;22) is an ACA with a poor prognosis 1 , we clarified that prognoses differ depending on the coexistence of a complex karyotype. It is known that an ACA is often observed in Ph+ALL patients, but reports of whether ACAs affect prognosis are inconsistent: some studies have reported that ACAs were a poor prognostic factor 7,9 whereas others have reported that ACAs were not a poor prognostic factor. [10][11][12] Considering the types and combinations of ACAs, we were able to clarify a subgroup of ACAs with poor prognosis in Ph+ALL. ...
... Additional chromosomal abnormalities (ACAs) can be detected in 64-78% of patients with Ph+ ALL [4,10]. The prognostic impact of some ACAs is established, and the presence of −7, + 8, −9/9p, + der(22)t(9;22), hypodiploidy (<44 chromosomes), and complex karyotype (≥5 chromosome abnormalities) constitute high-risk karyotypes [10,11]. Poor-risk ACAs were prognostic in patients who received imatinib and dasatinib but not in those who received ponatinib [10]. ...
... Confirmation of Ph+ ALL relies on karyotyping and/or molecular genetics (cross reference). Additional chromosomal aberrations have been linked to inferior outcome [106][107][108] . PCR analysis of BCR::ABL1 should not be the sole diagnostic test as atypical BCR::ABL1 transcripts may be missed but determining the BCR::ABL1 isoform is important for subsequent MRD analysis. ...
Article
Full-text available
Experts from the European Leukemia Net (ELN) working group for adult acute lymphoblastic leukemia have identified an unmet need for guidance regarding management of adult ALL from diagnosis to aftercare. The group has previously summarized their recommendations regarding diagnostic approaches, prognostic factors and assessment of ALL (cross-reference). The current recommendation summarizes clinical management. It covers treatment approaches including the use of new immunotherapies, application of MRD for treatment decisions, management of specific subgroups and challenging treatment situations as well as late effects and supportive care. The recommendation provides guidance for physicians caring for adult ALL patients which has to be complemented by regional expertise preferably provided by national academic study groups. -
... For instance, Aldoss et al. reported that among 78 adult patients with Ph+ ALL who underwent alloSCT and had available cytogenetic data, 53% had any ACA, with monosomy 7 being the most common (29%). In this cohort, primarily treated with imatinib-based regimens, a significant difference in EFS was observed (79.8% versus 39.5%, p = 0.01) [27]. In contrast, a study by Akahoshi et al., focused on allografted patients with Ph+ ALL, examined 1375 patients from the Japanese transplant registry. ...
Article
Full-text available
Simple Summary Remarkable strides have been performed in the treatment of adults diagnosed with Philadelphia-positive lymphoblastic leukemia (Ph+ ALL) through the integration of newer-generation tyrosine-kinase inhibitors and monoclonal antibodies. However, it is crucial to acknowledge that most medical centers worldwide lack access to these therapies. As a result, primary strategies employed for curing this disease continue to rely on a combination of chemotherapy and allogeneic stem-cell transplantation. Additionally, the scarcity of comprehensive literature makes it particularly challenging to provide straightforward treatment recommendations. In this narrative review, our aim is to offer a real-world perspective on the monitoring and management of Ph+ ALL patients, with an emphasis on less-resourced scenarios. Abstract Recent studies have indicated that more than half of adult patients newly diagnosed with Ph+ ALL can now achieve a cure. However, determining the most suitable protocol for less-resourced settings can be challenging. In these situations, we must consider the potential for treatment toxicity and limited access to newer agents and alloSCT facilities. Currently, it is advisable to use less intensive induction regimens for Ph+ ALL. These regimens can achieve high rates of complete remission while causing fewer induction deaths. For consolidation therapy, chemotherapy should remain relatively intensive, with careful monitoring of the BCR-ABL1 molecular transcript and minimal residual disease. AlloSCT may be considered, especially for patients who do not achieve complete molecular remission or have high-risk genetic abnormalities, such as IKZF1-plus. If there is a loss of molecular response, it is essential to screen patients for ABL mutations and, ideally, change the TKI therapy. The T315I mutation is the most common mechanism for disease resistance, being targetable to ponatinib. Blinatumomab, a bispecific antibody, has shown significant synergy with TKIs in treating this disease. It serves as an excellent salvage therapy, aside from achieving outstanding results when incorporated into the frontline.
... Our patient population consisted of the AYA age subgroup, recognized as a unique population with specific characteristics and needs. Our AYA Ph+ ALL patients with high white blood count [12] and additional cytogenetics abnormalities on karyotyping had inferior survival outcomes similar to what is demonstrated in other age subgroups [13]. Patients were treated with either adult chemotherapy protocols (hyper-CVAD) or pediatric-inspired protocols like (BFM 2000 or UK-ALL regimens). ...
Article
Full-text available
Background Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) accounts for 25% of acute lymphoblastic leukemia cases in the adolescent and young adult (AYA) age subgroup. It is associated with poor outcomes and is considered a standard indication for allogeneic stem cell transplant (Allo-SCT). Improved outcomes have been reported with addition of tyrosine kinase inhibitors (TKIs) to chemotherapy in children and the role of Allo-SCT is now being debated in the first remission. Complete response (CR) at three months is associated with improved survival even without Allo-SCT in first CR. In this study, we have analyzed disease-free survival (DFS), overall survival (OS), and factors affecting survival outcomes of Ph+ ALL in the AYA subgroup, in resource-limited settings treated with chemotherapy and TKIs. Materials and methods This is a retrospective, multicenter cohort study of Ph+ ALL AYA patients, aged 18-40 years, and registered between January 2015 and December 2020. Primary objectives are to calculate disease-free survival (DFS) and overall survival (OS). Secondary objectives are to identify prognostic factors affecting response rates and outcomes. List of cases was obtained from hospital information system (HIS) and data were collected from patient case notes and electronic medical records. Data analysis was done utilizing the SPSS statistical program (Armonk, NY: IBM Corp.). Results Forty-nine patients were identified with Ph+ ALL with a median age of 23 years (range: 18-40 years) and a male-to-female ratio of 2.5:1. None of the patients had central nervous system (CNS) disease. White cell count was >30,000 per mm³ in 26% of patients, while 13% had additional cytogenetic abnormalities. Thirty-three percent patients received adult (hyper-cyclophosphamide, vincristine, Adriamycin, and dexamethasone {CVAD}) protocols while 67% received pediatric-inspired (Berlin-Frankfurt-Munster {BFM} 2000 or UK-ALL 2003/2011) protocols. TKI therapy was received by 66% of patients during treatment (early: 37%; late: 29%) and 34% did not receive TKIs due to financial constraints. CR after induction was achieved in 69% cases. Induction mortality was 16%. The median DFS for the entire cohort was 27 months (0.93-53.06) and the median OS was 29 months (8.89-49.10). The median OS in Allo-SCT group was not reached vs 8.0±8.8 months (p=0.05) with chemotherapy only. The OS was significantly better in patients with no additional cytogenetic abnormalities, pediatric-inspired chemotherapy protocols, early use of TKIs in induction phase, Allo-SCT, and post-Allo-SCT use of TKIs. Conclusion Addition of TKIs to pediatric-inspired chemotherapy protocols in Ph+ ALL AYA patients and Allo-SCT results in better overall survival. TKI availability remains a significant issue in low-income countries due to significant financial burden on the patients. Allo-SCT continues to be an attractive option, particularly in low-income countries providing an option for cure in Ph+ ALL.
Chapter
The distinct Philadelphia chromosome-positive (Ph+) subtype of acute lymphoblastic leukaemia (ALL) has historically been difficult to manage and has resulted in poor long-term survival in both paediatric and adult populations. However, drastic improvements have occurred with the addition of BCR-ABL1 tyrosine kinase inhibitors (TKIs) such as imatinib, dasatinib, and ponatinib to traditional cytotoxic chemotherapy regimens. Diagnostic advances including sensitive minimal residual disease (MRD) measurement by PCR and multi-parameter flow cytometry methods as well as BCR-ABL1 mutation analysis have enabled more accurate assessment of disease response allowing the type and intensity of treatments to be tailored accordingly. This chapter covers the diagnosis and management of Ph+ ALL in both children and adults, including discussion of TKIs, their combination with chemotherapy, and the role of haematopoeitic stem cell transplantation in this disease.
Article
Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) carried a very poor prognosis prior to the advent of tyrosine kinase inhibitors (TKIs) that block the activity of the BCR-ABL1 oncoprotein. With improvements in TKI efficacy and allogeneic hematopoietic cell transplantation (HCT), survival has improved over the past 3 decades, and the role of chemotherapy and allogeneic HCT is now changing. Better risk stratification, the application of the third-generation TKI ponatinib, and the use of immunotherapy with the CD19-CD3 bifunctional T-cell engaging antibody blinatumomab in place of chemotherapy has made therapy for Ph+ ALL more tolerable and arguably more efficacious, especially for older patients who comprise most patients with Ph+ ALL.
Article
Full-text available
Short imatinib pulses were added to chemotherapy to improve the long-term survival of adult patients with Philadelphia chromosome (Ph) -positive acute lymphoblastic leukemia (ALL), to optimize complete remission (CR) and stem-cell transplantation (SCT) rates. Of 94 total patients (age range, 19 to 66 years), 35 represented the control cohort (ie, imatinib-negative [IM-negative] group), and 59 received imatinib 600 mg/d orally for 7 consecutive days (ie, imatinib-positive [IM-positive] group), starting from day 15 of chemotherapy course 1 and from 3 days before chemotherapy during courses 2 to 8. Patients in CR were eligible for allogeneic SCT or, alternatively, for high-dose therapy with autologous SCT followed by long-term maintenance with intermittent imatinib. CR and SCT rates were greater in the IM-positive group (CR: 92% v 80.5%; P = .08; allogeneic SCT: 63% v 39%; P = .041). At a median observation time of 5 years (range, 0.6 to 9.2 years), 22 patients in the IM-positive group versus five patients in the IM-negative group were alive in first CR (P = .037). Patients in the IM-positive group had significantly greater overall and disease-free survival probabilities (overall: 0.38 v 0.23; P = .009; disease free: 0.39 v 0.25; P = .044) and a lower incidence of relapse (P = .005). SCT-related mortality was 28% (ie, 15 of 54 patients), and postgraft survival probability was 0.46 overall. This imatinib-based protocol improved long-term outcome of adult patients with Ph-positive ALL. With SCT, post-transplantation mortality and relapse remain the major hindrance to additional therapeutic improvement. Additional intensification of imatinib therapy should warrant a better molecular response and clinical outcome, both in patients selected for SCT and in those unable to undergo this procedure.
Article
Cytogenetic analyses performed at diagnosis on 443 adult patients with acute lymphoblastic leukemia (ALL) were reviewed by the Groupe Français de Cytogénétique Hématologique, correlated with hematologic data, and compared with findings for childhood ALL. This study showed that the same recurrent abnormalities as those reported in childhood ALL are found in adults, and it determined their frequencies and distribution according to age. Hyperdiploidy greater than 50 chromosomes with a standard pattern of chromosome gains had a lower frequency (7%) than in children, and was associated with the Philadelphia chromosome (Ph) in 11 of 30 cases. Tetraploidy (2%) and triploidy (3%) were more frequent than that in childhood ALL. Hypodiploidy 30-39 chromosomes (2%), characterized by a specific pattern of chromosome losses, might be related to the triploid group that evoked a duplication of the 30-39 hypodiploidy. Both groups shared similar hematologic features. Ph⁺ ALL (29%) peaked in the 40- to 50-year-old age range (49%) and showed a high frequency of myeloid antigens (24%). ALL with t(1;19) (3%) occurred in young adults (median age, 22 years). In T-cell ALL (T-ALL), frequencies of 14q11 breakpoints (26%) and of t(10;14)(q24;q11) (14%) were higher than those in childhood ALL. New recurrent changes were identified, ie, monosomies 7 present in Ph-ALL (17%) and also in other ALL (8%) and two new recurrent translocations, t(1;11)(p34;p11) in T-ALL and t(1;7)(q11-21;q35-36) in Ph⁺ ALL. The ploidy groups with a favorable prognostic impact were hyperdiploidy greater than 50 without Ph chromosome (median event-free survival [EFS], 46 months) and tetraploidy (median EFS, 46 months). The recurrent abnormalities associated with better response to therapy were also significantly correlated to T-cell lineage. Among them, t(10;14)(q24;q11) (median EFS, 46 months) conferred the best prognostic impact (3-year EFS, 75%). Hypodiploidy 30-39 chromosomes and the related triploidy were associated with poor outcome. All Ph-ALL had short EFS (median EFS, 5 months), and no additional change affected this prognostic impact. Most patients with t(1;19) failed therapy within 1 year. Patients with 11q23 changes not because of t(4;11) had a poor outcome, although they did not present the high-risk factors found in t(4;11).
Article
The BCR-ABL fusion, the molecular equivalent of the Philadelphia translocation, gains importance for treatment stratification in adult acute lymphoblastic leukemia (ALL). In this prospective study, samples from 478 patients with CD10+ B-cell precursor ALL (c-ALL and pre-B ALL) underwent BCR-ABL reverse transcription–polymerase chain reaction (RT-PCR) analysis with double testing of positive samples. Patients were stratified according to the PCR result and treated in 2 German Multicenter Trials of Adult ALL. The outcome was followed and the prognostic impact of BCR-ABL was compared to clinical risk features. Of the 478 samples, 432 had an evaluable BCR-ABL result. Thirty-seven percent of the c-ALL and pre-B ALL patients were BCR-ABL+ (p190, 77%; p210, 20%; simultaneous p190/p210, 3%). BCR-ABL positivity was associated with the high-risk features of older age (45 years versus 30 years median age; P = .0001) and higher white blood cell counts (23 500/μL versus 11 550/μL; P = .0001). Univariate and multivariate analyses revealed BCR-ABL as the leading factor for a poor prognosis ( P = .0001) in comparison to clinical risk criteria. Irrespective of the breakpoint, presence of any BCR-ABL transcript predicted a lower chance of initial treatment response (68.4% versus 84.6%; P = .001) and a lower probability of disease-free survival at 3 years (0.13 versus 0.47; P = .0001). This bad outcome was not influenced by postinduction high-dose treatment stratifications. The results show a high prevalence of BCR-ABL fusion transcripts with predominance of p190. BCR-ABL RT-PCR is confirmed as a sensitive, rapid method to diagnose t(9;22), and p190 and p210 are unequivocally demonstrated as the most important predictors of poor long-term survival despite intensified chemotherapy.
Article
Cytogenetic classification of 350 adults with acute lymphoblastic leukaemia on MRC UKALL XA trial showed the following statistically significant associations: t(9;22) (11%) increased with increasing age and leucocyte counts (WBC) and most had a C/pre-B immunophenotype. t(4;11) (3%) was associated with higher WBCs, increasing age and null immunophenotype. Other abnormalities of 11q (abn11q) (4%) were associated with male sex and T-cell ALL. High hyperdiploidy (7%) and abn9p (5%) decreased with increasing WBC. High hyperdiploid patients were younger and tended to have C/pre-B ALL. Triploidy/tetraploidy (3%) decreased and pseudodiploidy (11%) increased with increasing WBC. Cytogenetic classification was prognostically important (chi-square for heterogeneity of classification = 53.56; P < 0.0001) and added significance to age, sex and WBC. A poor prognosis for patients classed as t(9;22) (13% disease-free survival at 3 years), as t(4;11) 24% at 3 years) and hypodiploid (11% at 3 years), and good prognosis for abn12p (4% of subjects) and high hyperdiploidy (74% and 59% at 3 years respectively) were statistically significant, but the 54% 3-year disease-free survival for patients with t(1;19) was not. The prognosis of patients classed as t(9;22) was independent of other single variables. Abn12p, abnormalities of 11q (including t(4;11) cases) and hypodiploidy added prognostic significance to all other variables combined.
Article
The Cancer and Leukemia Group B (CALGB) has been conducting a prospective cytogenetic companion study (CALGB 8461) to all CALGB treatment protocols for newly diagnosed adults with acute lymphoblastic leukemia (ALL). These protocols underwent a significant change in 1988 when a new intensive chemotherapy program was introduced (CALGB 8811). We asked whether karyotype continued to represent a significant prognostic factor in adult ALL patients after the change. A total of 256 patients had adequate pretreatment cytogenetic analyses: 67 before 1988 and 189 subsequently. The complete remission (CR) rate for the whole group was 80%. Patients with t(9;22), t(4;11), -7, or +8 had significantly lower probabilities of continuous CR and survival at 5 years (.11 and.12) than patients with a normal karyotype (.38 and.37) and patients with miscellaneous cytogenetic abnormalities (.52 and.49; P <.001 for each comparison). When analyzed by treatment period, the CR rate before CALGB 8811 was 63%; subsequently, it was 86% (P <.001). Patients with cytogenetic abnormalities other than t(9;22), t(4;11), -7, or +8 had better CR rates, disease-free survival (DFS), and survivals (P =.001, P =.04, and P =.004, respectively) after the change to the more intensive chemotherapy regimens. Patients with normal cytogenetics had improved CR rate but no improved DFS or survival, whereas no significant benefit for patients with t(9;22), t(4;11), -7, or +8 was seen. In a multivariate analysis, karyotype retained its prognostic significance for DFS but not for survival; it remained the most important factor for DFS. We conclude that cytogenetic analysis at diagnosis should be used to guide treatment decisions in adults with ALL.
Article
The BCR-ABL fusion, the molecular equivalent of the Philadelphia translocation, gains importance for treatment stratification in adult acute lymphoblastic leukemia (ALL). In this prospective study, samples from 478 patients with CD10(+) B-cell precursor ALL (c-ALL and pre-B ALL) underwent BCR-ABL reverse transcription-polymerase chain reaction (RT-PCR) analysis with double testing of positive samples. Patients were stratified according to the PCR result and treated in 2 German Multicenter Trials of Adult ALL. The outcome was followed and the prognostic impact of BCR-ABL was compared to clinical risk features. Of the 478 samples, 432 had an evaluable BCR-ABL result. Thirty-seven percent of the c-ALL and pre-B ALL patients were BCR-ABL(+) (p190, 77%; p210, 20%; simultaneous p190/p210, 3%). BCR-ABL positivity was associated with the high-risk features of older age (45 years versus 30 years median age; P =.0001) and higher white blood cell counts (23 500/microL versus 11 550/microL; P =.0001). Univariate and multivariate analyses revealed BCR-ABL as the leading factor for a poor prognosis (P =.0001) in comparison to clinical risk criteria. Irrespective of the breakpoint, presence of any BCR-ABL transcript predicted a lower chance of initial treatment response (68.4% versus 84.6%; P =.001) and a lower probability of disease-free survival at 3 years (0.13 versus 0.47; P =.0001). This bad outcome was not influenced by postinduction high-dose treatment stratifications. The results show a high prevalence of BCR-ABL fusion transcripts with predominance of p190. BCR-ABL RT-PCR is confirmed as a sensitive, rapid method to diagnose t(9;22), and p190 and p210 are unequivocally demonstrated as the most important predictors of poor long-term survival despite intensified chemotherapy.
Article
From 1994 to 2000, 154 adults with Philadelphia chromosome-positive (Ph(+)) and/or BCR-ABL(+) acute lymphoblastic leukemia (ALL) were treated according to a prospective trial (median follow-up, 4.5 years) with the aim to study the prognostic value of early response to therapy and the role of stem cell transplantation (SCT) in first complete remission (CR). All patients received a standard induction course followed by a course of mitoxantrone and intermediate-dose cytarabine (HAM). After each course, minimal residual disease was tested by specific reverse transcriptase-polymerase chain reaction (RT-PCR) (median sensitivity, 10(-5)). Allogeneic SCT (if a donor) or autologous SCT (if not) was planned at 3 months in all patients in CR after HAM. CR rates after induction, after HAM, and at 3 months were 53%, 67%, and 62%, respectively. High leukocyte count and m-bcr subtype were the 2 identified bad-prognosis factors for CR at 3 months, both superseded by a poor early response assessed at day 8 of the induction course. HAM-associated salvage rate was higher in patients with M-bcr than in those with m-bcr ALL (55% vs 30%; P =.05). In the 103 patients eligible for SCT, the existence of a donor and the negative BCR-ABL status after HAM were independently predictive of remission duration (P <.001 and.01, respectively) and survival (P =.02 and.01, respectively). Relapse was the most common cause of treatment failure in all patient groups. Allogeneic SCT in first CR is the current best treatment option in adults with the disease. New strategies must be tested during early phases of therapy to increase the rate of BCR-ABL(-) remissions.
Article
From 1986 to 1998, 26 (23%) of 114 adult acute lymphoblastic leukemia (ALL) patients and 11 (4%) of 328 pediatric patients were found to have Philadelphia (Ph) chromosome. In the 30 patients with available data at diagnosis, 18 (60%) had extra-chromosomal abnormalities. They included 1q duplication (5/18, 28%), supernumerary Ph chromosome (4/18, 22%), 9p abnormalities (3/18, 17%), 7q deletion/monosomy 7 (3/18, 17%), trisomy 19 (1/18, 6%), and trisomy 8 (1/18, 6%). Excluding those with specific cytogenetic changes, only one patient had hyperdiploid karyotype with more than 50 chromosomes. The incidence of 1q duplication was higher and that of hyperdiploidy was lower in this study than has been previously reported. There was no prognostic implication of these additional cytogenetic abnormalities. With fluorescence in situ hybridization (FISH) and reverse transcription-polymerase chain reaction (RT-PCR), 14 (27%) of 53 unselected adult ALL patients and 2 (5%) of 38 unselected pediatric patients were BCR-ABL-positive, including one adult and two children without Ph chromosome. The BCR-ABL fusion genes/transcripts were also present in all other 16 selected Ph-positive ALL patients. The BCR-ABL fusion subtypes were determined in all these 32 patients: 91% (11/12) childhood cases showed m-type fusion gene while 45% (9/20) adult ones did so (P = 0.0083). The clinical outcome was similar between the two groups of patients with m-type and M-type BCR-ABL. In conclusion, both cytogenetic and molecular studies are very helpful for identifying the subgroup of ALL patients with Ph/BCR-ABL. The additional cytogenetic abnormalities and subtypes of BCR-ABL fusion genes/transcripts had no significant implications in this group of patients.