ISSN 1866-8836
Клеточная терапия и трансплантация

Ph-positive leukemias in the era of modern cytogenetics, molecular biology, tyrosine kinase inhibitors and hematopoietic stem cell transplantation

Nikolay N. Mamaev

St. Petersburg Pavlov State Medical University, St. Petersburg, Russia


Correspondence:
Nikolay N. Mamaev, Clinical-laboratory Diagnostics Chair, Cytogenetic Lab, St. Petersburg Pavlov State Medical University, 6/8, Tolstoy str., St. Petersburg, 198022, Russia
Phone: +7 (812) 233-12-43 (office) or +7 (812) 726-53-25 (home)
E-mail: nikmamaev@spam is badrambler.ru

doi 10.3205/ctt-2009-en-000047.01
Submitted 05 June 2009
Accepted 25 August 2009
Published 31 August 2009

Summary

The review is devoted to the complex problems of modern cytogenetic and molecular diagnostics of Ph-positive leukemias, their treatment in era tyrosine kinase inhibitors and hematopoietic stem cell transplantation at the condition of cytogenetic and molecular monitoring.

Keywords

Hematopoietic stem cell transplantation, cytogenetics, ph-positive leukemias, molecular biology, tyrosine-kinase inhibitors


Introduction

The Philadelphia (Ph1)-chromosome was discovered in 1960 year [1] and for a long time it was considered to be a specific marker of chronic myeloid leukemia (CML). Later it was found that Ph1 is the result of reciprocal translocation between chromosomes, i.e. t(9;22)(q34;q11) [2]. In fact, this translocation results in the formation of two hybrid genes, BCR-ABL on the Ph1 chromosome and ABL-BCR on 9q+. The chimeric BCR-ABL fusion product is a tyrosine kinase. Its activity accounts for more than 95% of CML cases [3-5], and 10–20% of adults and about 2–5% of children with B-cell acute lymphoblastic leukemia (ALL) [6, 7]. Occasional bona fide Ph1-positive (Ph+) cases among acute myeloid leukemias (AML) [8, 9], lymphomas [10], multiple myeloma [11], myelodysplastic syndromes [12, 13], ‘essential thrombocythemia’ [14-16], and chronic neutrophilic leukemia (CNL) [17] have also been published.

The treatment of CML has been recently revolutionized by the development of imatinib mesylate (IMT, Gleevec, STI571) and other tyrosine kinase inhibitors (TKIs), which report to be targeted inhibitors of BCR-ABL [18-20].

I. Laboratory Tests

1.1. t(9;22)(q34;q11) translocation detection
Translocation (9;22) in newly diagnosed leukemia patients is detected using conventional cytogenetics (CC) supplemented by chromosome banding analysis (Fig. 1), and supported by reverse transcriptase polymerase chain reaction (RT-PCR) [21]. The chromosome banding analysis remains the best first-line genetic test for assessing any new acute leukemia because it screens for t(9;22) as well as for alternate and nonrandom additional genetic defects, including +der(22)t(9;22), 9p abnormalities, i(17q), -7, +8, +19, +21, and so on [21].

2009-4-en-Mamaev-Figure-1.JPG

Figure 1. Karyotype of bone marrow cell from a female patient with Ph-positive acute lymphoblastic leukemia, relapsed after allogeneic stem cell transplantation, made in Germany. It illustrates two Ph-chromosomes with typical translocation t(9;22)(q34;q11) and multiple additional numeral and structural chromosome changes. 2n=52, XX, +X, t(9;22)(q34;q11), +der(22)t(9;22), +del(4)(q27), -8, +i(8)(q10), +10, +14, -20, +der(20q+), +21.

1.2. Fluorescence in situ hybridization
One of the sensitive approaches for diagnosis of Ph+ leukemias is fluorescence in situ hybridization (FISH). The FISH technique can be applied directly to non-dividing cells, it detects both cryptic and complex BCR-ABL rearrangements, including three-way translocations as well as some breaks outside of the usual major and minor cluster regions (Fig. 2). Sensitivity of the method is about 1 cell in 200 tested, or 0.5%. A problem with around 1% of CML patients who have a Ph1-negative karyotype because of a cryptic BCR/ABL1 fusion is that it can only be located by FISH at chromosome 22q11, 9q34 or a third chromosome [22-24].

2009-4-en-Mamaev-Figure-2.JPG

Figure 2. Karyotype (A) and fluorescent in situ hybridization (FISH) analysis findings on chromosomes (B, D) and interphase nuclei (C) from a patient with chronic myeloid leukemia, illustrating atypical translocation of t(6;22)(q21;q11), which was associated with formation of fusion ABL/BCR gene on chromosome der(22)t(6;22). 2n=46, XY, t(6;22)(q21;q11). The fusion gene (yellow color) (B, C) was evidenced by means of fluorescent probes to ABL (red) and BCR (green) genes. On the other hand, the atypical translocation t(6;22) was evidenced  (D) by means of fluorescent probe WCW22 directed to chromosome number 22. After FISH each chromosome plate contains three brightly stained signals, including those on der(22)t(6;22) (small signal), unchanged chromosome 22 (right big signal) and der(6)t(6;22) (left big signal). Magnification x900.

1.3. Southern blot analysis
Southern blot analysis reliably identifies BCR gene rearrangement using probes targeting either the M-bcr or m-bcr breakpoint. Although this technique is quite helpful in confirming the BCR defect associated with CML or ALL, the Southern blot method is time-consuming and costly [22].

1.4. Amplification technologies
RT-PCR is the most sensitive of today's methods for detecting BCR-ABL. One of the advantages of RT-PCR is an ability to differentiate p210 from p190 forms of BCR-ABL [22]. It should be mentioned here that the recently developed real-time quantitative PCR (RQ-PCR) technique allows the assessment of trends in the BCR-ABL load over time and, hence, the chance to control minimal residual disease (MRD) [25]. 

1.5. Analysis of BCR-ABL kinase domain (KD) mutations
There are several molecular techniques that can detect BCR-ABL kinase domain (KD) mutations in Ph+ leukemias that are resistant to tyrosine kinase inhibitors (TKI). A more effective technique among them is a recently modified liquid chromatography (D-HPLC) [26-33].  

1.6. Gene expression profiles and micro array analysis
A major advantage of array technology is the ability to evaluate an expression of thousands of genes simultaneously. For this purpose RNA is extracted from the patient’s sample and converted to labeled cRNA or cDNA before being applied to an array of complementary probes that allows the successful differentiation of BCR-ABL cases from other forms of ALL [34].

II. Laboratory and clinical findings in Ph-positive disorders

2.1. Structure of the BCR-ABL fusion genes and their transcripts
The breakpoint in the ABL gene occurs within a >300-kb segment at the 5’end of the gene, either upstream of the first alternative exon Ib, between exons Ib and Ia, or downstream of axon Ia. In the vast majority of CML patients and in about one third of ALLs, the breakpoint in the BCR gene is revealed within a 5.8-kb region known as the major breakpoint cluster region (M-bcr), spanning 5 exons historically named b1 to b5, now known to be exons e12 to e16 of the BCR gene. Regardless of the position of the ABL breakpoint, processing of the primary BCR-ABL transcript usually results in hybrid BCR-ABL mRNA molecules with e14a2 and or e13a2 junction encoding a p210BCR-ABL fusion protein [7]. In two thirds of ALLs and in very rare cases of CML and AML patients, the breakpoint in BCR falls further upstream, in the long (54.4 kb) intron between the two alternative exons e2’ and e2, known as the minor bcr (m-bcr). In these circumstances, exons e1’ and e2’ are removed by splicing, and the hybrid BCR-ABL transcript contains an e1a2 junction, and is translated into a smaller 190-kD BCR-ABL fusion protein named p190BCR-ABL. Thus, e14a2 (b3a2) and/or e13a2 (b2a2) fusion transcripts and p210BCR-ABL protein[s] are more characteristic for CML. In contrast, the p190BCR-ABL protein arising from the minor BCR rearrangement producing the e1a2 and/or e1a3 fusion transcript is seen in the majority of cases of Ph+ ALL. Meantime, the expression of e13a2 and/or e14/a2 fusion transcripts is noted in ALL, especially in adult patients [35, 36]. Furthermore, in Ph+ ALL patients some atypical BCR-ABL transcripts, including e1a3, e13a3, and e6a2, have recently been detected [36]. Additionally, occasionally cases of CML with e1a2, e19a1 and e19a2 transcripts [33, 37, 38] have been reported; the clinical significance of which remains unclear.

2.2. Prevalence of BCR-ABL


2.2.1. CML
The vast majority of CML patients have a p210 breakpoint of ABL-BCR gene, and they retain this genotype when their disease progresses to blast crisis (BC). There is also a possibility for coexistent p190 and p210 transcripts in 8% patients with accelerated-phase or BC CML [39].  

It is important to clarify whether the position of the breakpoint within the M-bcr region (5’ M-bcr v 3’ M-bcr), or the type of fusion transcript (for instance, e13a2 v e14a2) somehow influences the disease phenotype or not. The debate has been going on for several years, with some evidence in favor [33, 40, 41] and some against [42, 43] a possible link between M-bcr breakpoint location and disease features, that remains to be cleared up.

Currently many atypical BCR-ABL transcripts are documented in patients with CML, including e1a3, e6a2, e8a2, e13a3, e14a3, e19a2 and others [7, 36, 44]. The most frequent transcripts among them are e6a2 and e1a3 [36]. The clinical course of Ph+ CML with BCR breakpoints outside the above three main cluster regions (e.g. with e1a3) was more benign [45-48], although myeloid BC CML developed in most of them [36, 38, 47, 49]. It has been actively discussed that the poor prognosis of leukemias in the patients with atypical ABL/BCR transcripts might be associated with their lacking the (GET)/Abl-like domain of BCR [50]. This conclusion was based on the findings of a case in Ph-negative CML, where BCR-ABL fusion was recognized by FISH staining of metaphase chromosomes only [51], and wherein e6a2 atypical ABL-BCR fusion, encoding a hybrid p195BCR-ABL protein [50] was detected.

2.2.2. ALL

The BCR-ABL fusion gene is found in around 25% of adult ALL cases [36, 52-54] and in 2% of children [55-56]. Minor breakpoint transcripts (e1a2) encoding for p190 protein, are found in 59% to 70% of positive cases, whereas major breakpoint transcripts (e13a2 or e14a2) encoding for p210 protein are detected in 23% to 30% of cases [57]. Thus, the typical BCR-ABL mRNA transcripts are e1a2, e13a2, and e14a2. At the same time 3% to 19% of ALL patients can express both p210 and p190 fusion transcripts [35, 52-54, 57]. Preliminary data show a high prevalence of p210 copies with respect to copies of the p190 transcript in p190/p210+ cases [35]. Finally, atypical BCR-ABL transcript products are revealed in 1–2% of all Ph+ ALL cases, and include such atypical transcripts as e1a3 (most often), e6a2, e13a3, and e19a2 [7, 36, 58, 59].

The presence of BCR-ABL in ALL is interesting for two reasons. First, the patients with Ph+ ALL have a poor prognosis under conventional therapy [52] and are, therefore, considered high-risk patients and primary candidates for both intensified therapy regimens and alloSCT [36]. Secondly, BCR-ABL positive patients can have a significantly better prognosis under treatment regimens with ABL TKIs such as imatinib [60], dasatinib [61, 62], nilotinib [63] etc., and should take them whenever possible.

2.2.3. Clinical  significance of BCR/ABL isoforms
The majority of BCR-ABL positive adult [52] and childhood ALLs [55, 56, 64] have common or pre-B cell immunological variants with frequent co-expression of CD10, CD13, and CD33 antigens. A special analysis of gene expression patterns showed PILRB, STS-1 and SPRY genes to be overexpressed, whereas TSPAN16 and ADAMTSL4 genes are under-expressed in p190BCR-ABL-positive cases relative to p210BCR-ABL-positive ALLs [54, 57]. No difference was seen in the overall survival of patients with p190BCR-ABL-positive vs p210BCR-ABL-positive ALL. On the basis of this data a gene expression- and interaction-based outcome predictor consisting of 27 genes (including GRB2, GAB1, GLI1, IRS1, RUNX2, and SPP1) has been constructed which, in turn, correlated with overall survival (p=0.0001) in BCR-ABL-positive adult ALL, independent of age (p=0.25) and WBC count at presentation (p=0.003) [54].

The overall response to induction therapy (without TKIs) as well as the type of transplants did not differ significantly between patients with p190+ and p210+ transcripts, although the probabilities of DFS and OS are dependant on BCR/ABL isoforms [35]. Multivariate analysis performed in BCR/ABL+ patients showed that among the variables analyzed (i.e., age, white cell count, expression of CD34, CD10, CD13 and CD33, and type of BCR/ABL transcript), the presence of the p190 fusion transcript was the only powerful independent prognostic factor that favorably influenced disease-free survival (p=0.031) [35]. Besides this, the p190 fusion appears to be the only independent prognostic factor with regard to DFS and OS [35, 65].

2.3. Treatment of BCR-ABL+ leukemias with TKIs

2.3.1. CML
The deregulated tyrosine kinase activity of the BCR-ABL and represents an extremely attractive target for therapeutic intervention. The first original Abl tyrosine kinase inhibitor (TKI) imatinib mesylate (imatinib, IMT) has been developed for clinical use as a result of collaboration between Brian Druker [66-69] and investigators at Ciba-Geigy [70]. Its efficacy was evidenced both on cell lines and the great material collected from patients with CML [69, 71-83]. With a median follow-up of 19 months [77], the estimated rates of complete hematologic remission (CHR) for patients whose initial treatment was imatinib were 96%; major cytogenetic response (MCyR) was 87%; and from CCyR-treated patients, 76%. In patients with accelerated phase CML, imatinib results in a CHR rate of 82% and a MCyR rate of 24% [84]. However, in BC patients CML responses to IMT were of short duration [68, 71], although around 7% patients remain alive after a median observation time of 6 years [85]. In this case imatinib results in a CHR rate of 8–11%, having a MCyR response rate of 16% only, and the median time to relapse in responding patients with BC was about 3 months [86].

The striking efficacy of IMT in CML [25, 72, 74, 75, 78, 79, 87-89] has established this therapy as a new standard for care for the disease. However, resistance is an emerging problem. which has prompted the design of several second-generation TKIs. To overcome the resistance to imatinib [90-93], high-dose IMT therapy was used [94] as well as alternative second-generation TKIs, including dasatinib, nilotinib, INNO-406, MK-0457, and bosutinib (BST).

Dasatinib (BMS-354825; Bristol-Myers Squibb) [68, 92] is active against many of the kinase domain mutants responsible for imatinib resistance [95, 96]. One remarkable exception appears to be the T315I mutation. As shown in a special study of 21 Ph+ patients who failed to respond or relapsed during dasatinib therapy, all patients but one had mutations at residue 315 and/or at residue 317 [30].

Clinical experience in treatment of IMT-resistant or refractory CML patients with dasatinib and nilotinib has been recently reviewed by Goldman [75]. As a result, MHR rates with dasatinib among intolerant or resistant to imatinib patients were 63% for accelerated phase CML (follow-up > or =9 months), 34% for patients with myeloid BC CML, and 35% for those with lymphoid BC CML (follow-up > or =12 months; START-B and START-L trials) [92]. In another study, CHR responses with dasatinib were achieved in 81% IMT-resistant patients with CP CML, while MCyR was demonstrated in 57% of patients [97]. As for high resistance to dasatinib, it was related to the T315I and F317L Abl KD mutations [30, 98, 99].

The next of the second-line BCR-ABL TKI is nilotinib (formerly AMN107), which is 20 to 50 times more active than imatinib against CML cells [87, 93]. The first experience with nilotinib in 119 IMT-resistant patients with CML showed the following. Of 30 patients at BP of the disease, 13 (39%) experienced HR and 9 patients (27%) had a CyR, of whom 6 had a MCyR (Ph+ cells in metaphase, ≤35%). Of 46 patients with accelerated phase CML 33 had a HR and 22 had a CyR. Lastly, 11 of 12 patients with the CP CML had a CHR [90]. As for the resistance to nilotinib, it was again associated with T315I and F317L ABL KD mutation [30] that allows a choice of such TKIs as INNO-406 or MK-0457 for treatment patients with the above unfavorable mutations [30, 75, 98, 99].

2.3.2. ALL

The treatment of Ph+ ALL has also changed dramatically since imatinib was introduced [100-105]. Early phase I and II studies of imatinib in Ph+ ALL revealed its significant, albeit unsustain, activity against relapsed or refractory leukemias [73, 81, 106]. On the other hand, combined with chemotherapy, or even as a single agent, it can produce CR rates of 90% or higher in newly diagnosed patients [6, 85, 100-102, 104, 105, 107-109]. However, these responses were not durable (median, 9 months) [60], whereas further intensification of chemotherapy has had no substantial impact on the unfavorable course of de novo Ph+ adult ALL or in patients who fail first-line therapy [110, 111]. A special study of alternating vs concurrent schedules of imatinib and chemotherapy as front-line for Ph+ ALL demonstrated that co-administration of imatinib and induction cycle 2 results in a CHR rate of 95% and RT-PCR negativity for BCR-ABL in 52% of patients, compared with 19% in patients in the alternating treatment cohort (p=0.01). Further, in each cohort, 77% of patients underwent alloHSCT in CR1. As a result, both schedules of imatinib had acceptable levels of toxicity to patients, but concurrent administration of IMT and chemotherapy had resulted greater anti-leukemic efficacy [109].

It should be noted also, that the Ph+ ALL is very characteristic for elderly patients, wherein it has a poor prognosis, with a low CHR rate, high induction mortality, and short remission duration [112]. On the other hand, imatinib as a single agent for elderly patients is toxic enough whereas a potentially curative alloHSCT is not generally applicable [110]. Therefore, it is noteworthy, that the overall CHR rate in the group of elderly patients where imatinib was used for remission induction, was significantly higher (93.5%) than that in patients wherein for induction remission was used multi-agent chemotherapy (50%, p=0.0001) [112].

Despite the similar cytogenetic and molecular characteristics, the resistance to imatinib in patients with Ph+ ALL develops earlier and more frequently than that in CML, and therefore second-line TKIs are necessary. In part, CHR and CyR may be achieved with dasatinib [68, 90], and nilotinib [113], which appear to be ineffective in cases with T315I and F317L KD mutations [99].

Analogous with CML, the best drugs for treatment of patients who seem to be resistant to or intolerant to imatinib, were dasatinib, nilotinib, or bosutinib [6, 63, 91]. According to published data [90, 92], dasatinib induced CHRs in 42% of patients with IMT-resistant Ph+ ALL (follow-up > or =12 months), which is even higher than those in patients with myeloid and lymphoid variants of BC CML.

2.4. BCR-ABL kinase domain (KD) mutations

2.4.1. CML
More than 90 different point mutations encoding for distinct single amino acid substitutions in the BCR-ABL KD have been so far identified, but 10 of them occur in >80% of the cases [20]. The presence of KD mutations has been studied mainly in the advanced phase, and in CP patients when they become resistant to imatinib [113]. Meantime, a special screening for the mutation status of 319 newly-diagnosed patients with CP CML showed the identification of a mutation without other evidence of imatinib resistance to be highly predictive for loss of CCyR (RR, 3.8; p=0.005) and for progression to advanced phase (RR, 2.3; p=0.01). On the basis of this data it was concluded that mutations in the P-loop (excluding residue 244) were associated with a higher risk of progression than mutations elsewhere [114]. Although T315I was the most frequent mutant to emerge, other mutations like T315A, V299L, and F317I were detected that demonstrated retention of sensitivity to imatinib and potentially to nilotinib [30, 115]. Among patients with CP CML who develop (secondary) resistance to imatinib, 30% to 50% have one or more BCR-ABL KD mutations [116-120]. Some DK mutations (V299L, T315I, and F317L/I) were greatly specific for dasatinib [122-125], and others (G250E, Y253H, E255K, T351I, or F311I) for NLT [122] or MK-0457 [126]. Serial investigations showed that the longest period between detection of a mutation and subsequent relapse is observed in patients harboring M351T mutations [125]. In contrast, patients harboring T315I or P-loop mutations demonstrated a 5–25-fold need for immediate withdrawal of imatinib [28, 29].

According to the three largest retrospective studies [28, 117, 127], the incidence of the T315I mutation in IMT-resistant patients was 4%, 11%, and 19%, respectively whereas the frequency of P-loop mutations in the same series of patients was much higher, being 28%, 46%, and 39%, respectively. In general, survival of patients with this mutation remains dependent on the stage of the disease, it having an indolent course in many CP patients [98]. Meantime it is expected that patients with the T315I mutation will also reveal resistance to the treatment with either dasatinib or nilotinib [29, 30, 68, 87, 128]. That is why INNO and ON012380, which are binding regions of BCR-ABL other than the ATP binding pocket, are recommended first of all for these patients [30]. It should be noticed that except for the lack of response to second TKIs (p=0.002), other patient characteristics, including outcomes between patients with T315I and those with other or no mutations were similar.

So the prognostic significance of such KD mutations as T315I, and P-loop region of ABL is undoubted. It is also important that T315I and P-loop mutations were preferentially presented in accelerated phase and of BC CML, compared to other types of mutations in CP CML (p=0.003). In addition, overall survival (OS) after imatiunib initiation was significantly worse for P-loop (28.3 months) and for T315I (12.6 months), than that for other mutations (p=0.0004). Meantime, multivariate analysis of data from CP patients demonstrated a worse OS for P-loop mutations only (p=0.014), but a worse progression-free survival (PFS) for T315I mutations (p=0.014).

The vast number of mutation reports published later have demonstrated number of IMT-resistant patients with different amino acid substitution to be greatly high [89]. Many of these BCR-ABL DK mutations are associated with a relatively modest increase in resistance to imatinib, which may be overcome by dose increase only [30, 51]. On the contrary, mutations such as T315I, and those falling within the P-loop region, i.e., G250E, Y253F/H, and Ee55K/V, confer a highly resistant phenotype [28-30]. An analysis of BCR-ABL mutations by means of denaturating high-performance liquid chromatography found them in 127 of 297 (43%) of the available Ph+ patients. These mutations were revealed in 27% of CP CML patients (14% treated with IMT frontline; 31% treated with IMT post INF-α failure), 52% of accelerated-phase patients, 75% of myeloid BC patients, and 83% of lymphoid BC/Ph+ ALL patients. It should be mentioned that mutations were associated in 30% of patients with primary resistance to imatinib (44% hematologic and 28% cytogenetic), and in 57% of patients with acquired resistance (23% patients who lost CyR; 55% patients who lost HR; and 87% patients who progressed to accelerated phase/BC). On the basis of these findings the following conclusions were drawn. Firstly, amino acid substitution of seven residues (M244V, G250E, V253F/H, E255K/V, T315I, M351T, and F359V) accounts for 85% of all resistance-associated mutations. Secondly, the search for mutations is important both in case of IMT failure and in case of a loss of response at the hematologic or cytogenetic level.

It is interesting to note that in the majority of the resistance patients with CML, mutations were detectable at various intervals prior to hematologic relapse (range 0.9–44.2 months), which was registered at a median of 12.9 months after the start of imatinib therapy. On the other hand, BCR-ABL mutations first became detectable at a median of 5.8 months (range 0–30.5) after commencing imatinib. The difference between time to hematologic relapse and earliest detection of a mutation was highly significant (p<0.0001).

Since BCR-ABL KD mutations in some of CML and Ph+ ALL patients can be found without clinical evidence of resistant disease [26, 129-131], the questions arises as to when tests for DK mutations should be done and by what methods. According to international recommendations, BCR-ABL KD mutation screening in chronic phase CML is only recommended for patients with inadequate initial response to TKIs or those with evidence of loss of response. Mutation screening is also recommended at the time of progression to accelerated or BC CML [26, 132, 133]. Criteria for inadequate initial response (i.e., primary resistance) include lack of CHR, mCyR (66-95% Ph+ metaphases in BM) or lack of MCyR at 3, 6, and 12 months respectively which are similar to the criteria adopted by the European Leukemia Net [89]. Criteria for loss of response to TKI (i.e., secondary resistance) are also based on cytogenetic and/or hematological relapse, with variable use of molecular relapse criteria. One proposed molecular trigger for mutation testing is a tenfold or greater increase in BCR-ABL transcript levels, although smaller rises in BCR-ABL transcript levels may also be predictive of mutation development.

2.4.2. ALL
In comparison with CML, BCR-ABL KD mutations in relapsed Ph+ ALL occur much more frequently (80% to 90% of cases), especially in patients who have been treated with TKIs as initial or ongoing therapy [26, 32, 33, 134].  However, more sensitive detection methods reveal low levels of a point mutation clone in some Ph+ ALL cases before exposure to TKIs. The latter convinces that resistant clones in Ph+ ALL may precede TKI selection [134].

A study of the ABL KD mutation status in newly diagnosed Ph+ ALL patients showed that IMT-resistant TK domain mutations were detected in 38% patients even before exposure to IMT. Although the frequency of the mutant allele was low in such patients at the time of relapse, the same mutation was present as the dominant clone in 90% of the relapsing cases [134].

2.5. Hematopoietic stem cell transplantation (HSCT)

2.5.1. CML
Until the advent of imatinib, alloHSCT was considered to be the only known curative therapy for patients younger than 50 years having suitable HLA-identical siblings, HLA-matched family members, or HLA-matched volunteer unrelated donors [75]. Although the morbidity and indeed mortality attributable to the procedure were both appreciable, the probability of survival could be predicted with reasonable accuracy via the scoring system developed for the European Group for Blood and Marrow Transplantation (EBMT) [135]. Graft-versus-leukemia (GvL) effects contribute substantially to the curative potential of this approach [25, 136]. However, relapse occurs in approximately 5% to 20% of patients transplanted in chronic phase. While some patients remain in a stable MRD status, others rapidly progress to overt clinical relapse [137]. In order to re-induce MolR through augmenting or re-establishing the GvL effect, cessation of immunosuppression [138] and, particularly, donor lymphocyte infusion (DLI) are standard approaches for patients with relapse after alloHSCT [139, 140]. Unfortunately, this approach is frequently associated with substantial risk, mainly induction of graft-versus-host disease (GvHD) or BM suppression [139-144], which in large surveys have been observed in approximately 48% and 18% of patients [25, 145]. As a result, over 80% of patients with low risk of GvHD (i.e., 0–2) had a probability of being alive at 5 years [146] and the probability of subsequent relapse for patients free of detectable CML was extremely low [147].

Although the treatment with imatinib is less toxic, it may not cure the disease and, being used for a long time makes it very expensive. Additionally, resistance to imatinib can also occur in the course of the therapy. Therefore, alloHSCT for the treatment of some CML patients remains  necessary [25]. Firstly, transplanted HSC have anti-leukemic activity and contribute to maintaining remissions. Secondly, the effect of imatinib may be increased by potential side effects of DLI and vice versa [148]. Since suppression of MRD with imatinib can theoretically allow re-establishment of complete chimerism as well as restore full GvL effects, this approach has been recently tested successfully in clinical practice [37, 77, 149]. Additionally, a special molecular investigation showed that 70% of patients with CML who relapsed after alloHSCT can achieve CMolR after imatinib treatment and DLI [139, 140, 142, 148].

2.5.2.  ALL
The poor outcome with conventional chemotherapy made alloHSCT an attractive option for patients with Ph+ ALL [100, 101, 108, 109, 150-157]. The first study of allo BMT in 10 patients with Ph+ showed that 4 patients died of transplant-related complications, while 6 patients survived the transplant and remained in CR (median follow-up; 19 months) [158]. A retrospective analysis in 67 Ph+ ALL patients aged 5–49 years who underwent alloHSCT, showed that 21 of them (31%) were in continuous CR≥2 years after transplant [159]. There were no significant differences in TRM, DFS, and relapse rate between patients who had been transplanted in first or later CR. However, 2-year DFS was much lower in patients who had never achieved CR before transplant. Better results were reported later [151, 154, 160, 161]. For instance, Snyder et al [151] reported a 65% DFS rate (15/23) at 3 year post-transplant, whereas the estimated relapse rate during that period was only 12%. According to data from the French Bone Marrow Transplantation Society [162], in a study where 76 adult patients were in CR1 at the time of transplant, 2-year OS was demonstrated in 50% of patients, whereas the 2-year relapse incidence rate was 37%. In the largest prospective alloHSCT study (conducted during the pre IMT era), 5-year OS was superior in those who underwent MRD alloHSCT compared with chemotherapy or autologous HSCT [163, 164]. Unfortunately, despite its efficacy, currently only 20% to 60% of patients actually undergo alloHSCT. Moreover, even after alloHSCT in CR1 the probability of relapse for Ph+ ALL patients is approximately 30%, which together with a high transplant-related mortality of 20% to 40% highlights the limitations of current therapy [109, 165, 166].

The recent appearance of TKIs in clinical practice has changed the up-front treatment paradigm and appears to affect the outcome after HCST [167]. In contrast with CML, wherein TKIs have changed our attitude to HSCT as the treatment of first choice [168], TKIs in Ph+ ALL appear to be seen only as a “bridge to transplant”, helping more patients to achieve and maintain sufficiently durable remission before [156] or after [63] HSCT. The latter is desirable to undergo in CR1. In such a case the long-term survival rates may be greater than 35%. Furthermore, the results can be essentially improved after first-line imatinib interim treatment [107, 157]. The data suggests that the 3-year estimated probabilities of relapse, DFS, and OS were 3.5% vs 47.3% in controls (p=0.02), and 78.1% vs 38% in controls (p=0.001), respectively, without much difference in the transplant-related toxicities. In this context, other studies have also demonstrated the feasibility of giving imatinib following alloHSCT for BCR-ABL+ ALL either preemptively or to treat any MRD detected after transplant prior to relapse instead of using a donor lymphocyte infusion [169, 170]. The management of patients with Ph+ ALL relapsing after alloHSCT represents a major challenge with limited chances of success. The efficacy of DLI in Ph+ ALL is much lower that that in CML [171], which is explained by the diverse immune escape mechanisms of ALL blasts from GVL effects of DLI [172]. On the other hand, the addition of cytoreductive chemotherapy to DLI does not seem to improve the outcome of relapsed ALL patients [173], whereas  a combination of TKIs and DLI may be greatly effective [63].

2.6. Monitoring treatment and/or minimal residual disease

2.6.1. CML

2.6.1.1. Blood counts and bone marrow karyotyping

Complete blood counts should be performed at least weekly until they have stabilized, with greater intervals thereafter. Once CHR has been documented, monitoring continues with karyotyping of bone marrow metaphases, which is recommended at 6, 12, and 18 months, or until CCyR has been achieved [38, 89, 174, 175]. If there are fewer than 20 metaphases, the cytogenetic response can be validated by determining the level of BCR-ABL transcripts and by molecular cytogenetics, or FISH. Of note is that FISH can be performed on metaphases or more frequently and more conveniently on interphase cells (IP-FISH) [38, 176]. In fact, all FISH data correlates very significantly with chromosome banding data [177], as well as with BCR-ABL transcript levels [176, 177]. Moreover, FISH can detect deletions of the long arm of chromosome 9 and variant translocations.

2.6.1.2.  Monitoring BCR-ABL transcript levels
Molecular monitoring of BCR-ABL transcript levels with RQ-PCR has become an integral part of management of patients with CML [132, 178-180]. This is particularly relevant in the era of imatinib therapy, when residual levels of leukemia usually fall below the level of detection via bone marrow cytogenetic analysis. Indeed, even small increases in the BCR-ABL level can identify patients with TK domain mutations that lead to IMT resistance.

Currently, the best molecular approach for monitoring BCR-ABL transcript levels is considered to be RQ-PCR [133, 178, 179]. The best gene for internal reference appears to be β-glucuronidase and not the widely-used ABL gene [180]. In fact, a failure to achieve a major MolR by 18 months after starting imatinib therapy is considered a suboptimal response requiring careful re-evaluation and possible reassessment of therapy [92]. Various prerequisites for achieving optimal sensitivity and standardization have been agreed upon and published [133, 181, 182]. According to recent international reporting scale, major MolR is established at BCR-ABL 0.1%, whereas a value of 1.0% is approximately equivalent to the achievement of CCyR  [38].  From a practical point of view, it has been suggested that RQ-PCR must be performed every 3 months even in patients who achieve a MolR [179]. If, after alloHSCT, the attainment of BCR-ABL-negative status is conferred, the overall survival is significantly improved (57% vs 12%; p=0.006) [176]. In contrast, in the case where BCR-ABL levels increase, there is the possibility of identifying the patients who do not respond properly to the proposed therapy or even to start the search for BCR-ABL mutations [183, 184]. However, even considering that RQ-PCR is fundamental for monitoring patients with CCyR, there are other reasons to suggest that cytogenetics should not be completely replaced by RQ-PCR in the follow-up of CML patients. Firstly, additional chromosome abnormalities present at diagnosis or arising during the disease may have a prognostic influence [183]. Secondly, there is evidence of clonal cytogenetic abnormalities in the Ph-negative cells [185, 186] which appeared after suppression of Ph-positive clone by imatinib and, in a majority of cases, could be a characteristic of future myelodysplasias [38]

2.6.1.3. Monitoring BCR-ABL kinase domain mutations
A mutation at the TK domain of the oncogenic BCR-ABL protein is frequently detected in patients with CML who fail to respond to TKIs or lose the response [187>]. The best approach for screening for BCR-ABL mutations during therapy with TKIs is D-HPLC [28, 31]. The studies show that mutations may be detectable several months before relapse, but the occurrence of BCR-ABL mutations during imatinib therapy is predictive of relapse.
     
2.6.2. ALL

2.6.2.1. Cytogenetic monitoring

In contrast to CML, cytogenetic changes in Ph+ ALL in the course of chemotherapy do not last long. Therefore, the main tasks of cytogenetics, including FISH, in these patients are: a) to verify a CCyR; or, in turn, b) to detect a resistance to chemotherapy, TKI or HSCT.

2.6.2.2. BCR-ABL transcript level monitoring
BCR-ABL RNA transcripts are suitable molecular markers for MRD analysis and for guiding therapy [25, 188, 189]. However, MRD analysis with RT-PCR is affected by intra- and inter-assay variability, which appear to reflect different proportions of leukemic blast cells. A special investigation of samples from 56 patients showed levels of BCR-ABL to be higher in bone marrow than in peripheral blood. In fact, they did not differ when normalized to 100% blasts. Moreover, the numbers of BCR-ABL transcripts per blast in 25 sequential BM and 8 sequential PB did not change significantly during evolution of Ph+ ALL. As for the mean number of p210 copies per blast concern they were 1.1 log higher than that for p190 (p=0.0006).

2.6.2.3. BCR-ABL kinase domain mutation monitoring
In contrast to CML, pre-existence of mutations including the T315I mutation did not adversely effect either the CR rate following imatinib or chemotherapy induction or the achievement of BCR-ABL negativity in response to combination therapy, when compared with patients exhibiting only non-mutated BCR-ABL at diagnosis [190]. In our opinion, this might be explained by the earlier use of high-doses chemotherapy followed by alloHSCT.

2.7. Prognosis

2.7.1. CML
In the TKI era the prognosis for CML patients has greatly improved. The majority of patients can now expect to survive 10 to 20 years [74, 88, 191]. It is even possible that some patients treated for a number of years can stop the imatinib and be regarded as cured of their leukemia [192]. Hence, any possibility of recognizing the minority of patients who respond poorly to imatinib at the earliest opportunity is very important [193]. Therefore, a series of empirical recommendations developed on behalf of the European LeukemiaNet (Table 1) [89] are used by clinicians to identify those CP CML patients responding poorly to imatinib at standard dosage. As for the other prognostically poor and verified factors in CML patients, they include the following: a) age; b) advanced stages CML; and c) BCR-ABL domain mutations, mainly, T315I.

Table 1. Modified European LeukemiaNet definition of failure and suboptimal response for previously untreated, early CP CML patients treated with 400 mg IMT daily
Failure implies that the patient should be moved to other treatments whenever available; NA indicates not applicable; HR, hematological response; CHR, complete HR; CyR, cytogenetic response; CCyR, complete CyR; MMolR, major molecular response; ACA, additional chromosome aberrations, and TK tyrosine kinase

Time Failure Suboptimal response Warnings
Diagnosis NA NA High risk
      del 9q+; addit chrom abnormalities (ACA) in Ph+ cells
3 months No HR Less than CHR  
6 months Less than CHR Less than partial CyR (Ph+ >35%)  
12 months Less than partial CyR (Ph+ >35%) Less than CCyR Less than MMolR
18 months Less than CCyR Less than MMolR  
At any time Loss of CHR; loss of CCyR; TK domain mutation ACA in Ph+ cells; Loss of MMolR; TK domain mutation Any rise in transcript level; ACA in Ph- cells

2.7.2. ALL
The prognosis of Ph+ ALL – whatever the fusion protein – is poor in both adults and children [70, 194]. Detailed analysis of prognostic-related factors showed that deletion of the IKZF1 gene [195-199], as well as the secondary chromosome aberrations at diagnosis [200-204], in addition to t(9;22), had a significant association with shorter RFS. The latter has been reported in both before the imatinib [177, 178] ] and during the imatinib era [21]. Partly, there is data from 80 Ph+ ALL patients treated with imatinib, which was followed by alloHSCT in 60 of them. In fact it was found that the 2-year RFS for those who had undergone alloHSCT during CR1 was 62.6±7.5% compared to 62.1±12.3% for those who had not undergone alloHSCT. When alloHSCT was considered as a time-dependent covariant, it was shown to have no significant effect on RFS. On the other hand, a statistically significant negative impact on RFS for +der(22)t(9;22) and 9p abnormalities (p<0.001 and p<0.005) [21] was revealed, although it had not been demonstrated earlier [204].

Conclusions and future directions

The study of patients with Ph+ leukemias allowed a clarification of many sides of the nature and pathogenesis of leukemias [127, 128], and helped to develop new therapeutic approaches [208, 209]. The treatment of these disorders was partly revolutionized with TKIs. As a result, nowadays the vast majority of the patients with CML can be treated without alloHSCT. As far as HSCT is concerned, this must be reserved for those CML patients who become resistant to TKIs. The main reason for the resistance formation appears is considered to be mutations of BCR-ABL KD, primarily T315I. For achieving effect in relapsed after HSCT patients may be effectively used DLI which is the ideal approach for treating these CML patients [164].

In contrast to CML, alloHSCT is the only known curative modality for patients with Ph+ ALL under 55 years old. Naturally, it must be coupled by high dose chemotherapy and TKIs before transplantation [157, 170]. If such therapy is unsuccessful, testing for BCR-ABL domain mutations is necessary. Besides other prognostic factors, the following are considered to be important: a) additional chromosome changes, including the 2nd Ph-chromosome and 9p abnormalities; b) deletion of genes (i.e., IKAROS) needed for natural differentiation of B-cells; c) some cryptic chromosome changes; and d) micro-deletions of many important genes, which respond for natural differentiation of lymphoid and myeloid hematopoietic cells. The latest  investigations appear to be directed onto: a) the development of new techniques of mutation status testing;  b) the active introduction into clinical practice of TKIs of second- and third-lines; c) the further elucidation of mutated genes’ participation in both pathogenesis and resistance formation to treatment of Ph+ leukemias. As a result, one can expect that discoveries in these regions will allow the development of a concepts for target therapy of Ph+ leukemias, enabling more effective treatment than ever before.

References

1. Nowell PC, Hungerford D. A minute chromosome in human chronic granulocytic leukemia. Science. 1960;132:1497-9.

2. Rowley JD. Letter: A new consistent chromosome abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973;243:290-3.

3. Deininger MWN, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000;96:3343-56.

4. Advani AS, Penderqast AM. Bcr-Abl variants: Biological and clinical aspects. Leuk Res. 2002;26:713-20. doi: 10.1016/S0145-2126(01)00197-7.

5. Pane F, Intrieri M, Quintarelli C, et al. BCR/ABL genes and leukemic phenotype: from molecular mechanisms to clinical correlations. Oncogene. 2002;21:8652-67.

6. Ottmann O, Dombret H,  Martinelli G, et al. Dasatinib induces rapid hematologic and cytogenetic responses in adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia with resistance or intolerance to imatinib: interim results of a phase 2 study. Blood. 2007;110:2309-15. doi: 10.1182/blood-2007-02-073528.

7. Melo JV. The diversity of BCR-ABL fusion proteins and their relationship to leukemia phenotype.  Blood. 1996;88:2375-84.

8. Secker-Walker LM, Morgan GJ, Min T, et al. Inversion of chromosome 16 with the Philadelphia chromosome in acute myelomonocytic leukemia with eosinophils. Report of two cases. Cancer Genet Cytogenet. 1992;58:29-34. doi:10.1016/j.leukres.2005.06.003.

9. Alimena G, Cedrone M, Nanni M, et al. Acute leukemia presenting a variant Ph chromosome with p190 expression, dup 3q and -7, developed after malignant lymphoma treated with alkilating agents and topoisomerase II inhibitors. Leukemia. 1995;9:1483-6. pmid: 7658716.

10. Mitani K, Sato Y, Tojo A, et al. Philadelphia chromosome positive B-cell type malignant lymphoma expressing an aberrant 190 kDa bcr-abl protein. Br J Haematol. 1990;76:221-5. pmid: 2094324.

11. Martiat P, Meccucci C, Nizet Y, et al. P190 BCR/ABL transcript in a case of Philadelphia-positive multiple myeloma. Leukemia. 1990;4:751-4. pmid: 2232886.

12. Verhoef G, Meens P, Stul M, et al. Cytogenetic and molecular studies of the Philadelphia translocation in myelodysplastic syndrome. Report of two cases and review of the literature. Cancer Genet Cytogenet. 1992;59:161-6.

13. Keung YK, Beaty M, Powell BL, et al. Philadelphia chromosome positive myelodysplastic syndrome and acute myeloid leukemia – retrospective study and review of literature. Leuk Res. 2004;28:579-86. doi: 10.1016/j.leukres.2003.10.027.

14. Stoll DB, Peterson P, Exten R, et al. Clinical presentation and natural history of patients with essential thrombocythemia and the Philadelphia chromosome. Am J Hematol. 1988;27:77-83. pmid: 3422539.

15. Martiat P, Ifrah N, Rassool F, et al. Molecular analysis of Philadelphia positive essential thrombocythemia. Leukemia. 1989;3:563-5. pmid: 2747291.

16. Cervantes F, Urbano Ispizua A, Villamor N, et al. Ph-positive chronic myeloid leukemia mimicking essential thrombocythemia and terminating into megakaryoblastic blast crisis: Report of two cases with molecular studies. Leukemia. 1993;7:327-30. pmid: 8426485.

17. Pane F, Frigeri F, Sindona M, et al. Neutrophilic-chronic myelogenous leukemia: A distinct disease with a specific molecular marker (BCR-ABL with c3a2 junction). Blood. 1996;88:2410-4.

18. Dorshkind K, Witte ON. Linking the hematopoietic microenvironment to imatinib-resistant Ph+ B-ALL. Genes Dev. 2007;21:2249-52. doi: 10.1101/gad.1600307.

19. Piccaluga PP, Paolini S, Martinelli G. Tyrosine kinase inhibitors for the treatment of Philadelphia chromosome-positive adult acute lymphoblastic leukemia. Cancer. 2007;110:1178-86. doi: 10.1002/cncr.22881.

20. Melo JV, Chuah C. Novel agents in CML therapy: tyrosine kinase inhibitors and beyond. Hematology Am Soc Hematol Educ Program. 2008;427-35.

21. Yanada M, Takeuchi J, Sugiura I, et al. Karyotype at diagnosis is the major prognostic factor predicting relapse-free survival for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia treated with imatinib-combined chemotherapy. Haematologica. 2008;93:287-90. doi:10.3324/haematol.11891.

22. Nashed AL, Rao KW, Gulley ML. Clinical applications of BCR-ABL molecular testing in acute leukemia. J Mol Diagnostics. 2003;5:63-72.

23. Qiu H, Miao K, Wang R, et al.  The application of fluorescence in situ hybridization in detecting chronic myeloid leukemia. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2009;26:207-10. Chinese. pmid: 19350518.

24. Virgili A, Brazma D, Reid A, et al. FISH mapping of Philadelphia negative BCR/ABL I positive CML. Mol Cytogenetics. 2008;1:755-8. doi: 10.1186/1755-8166-1-14.

25. Hardling M, Wei Y, Palmqvist L, et al.  Serial monitoring of BCR-ABL transcripts in chronic myelogenous leukemia (CML) treated with imatinib mesylate. Med Oncol. 2004;21:349-58. doi: 10.1385/MO:21:4:349.

26. Doi Y, Sasaki D, Terada C, et al. High-resolution melting analysis for a reliable and two-step scanning of mutations in the tyrosine kinase domain of the chimerical bcr-abl gene. Int J Hematol. 2009 May 23. [Epub ahead of print]. doi: 10.1007/s12185-009-0337-y.

27. Jones D, Kamel-Reid S, Bahler D, et al. Laboratory practice guidelines for detecting and reporting BCR-ABL drug resistance mutations in chronic myelogenous leukemia and acute lymphoblastic leukemia. A report of the association for molecular pathology. J Mol Diagnostics. 2009;11:4-11. pmid: 19095773.

28. Soverini S, Colarossi S, Gnani A, et al. Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res. 2006;12:7374-9. doi: 10.1158/1078-0432.CCR-06-1516.

29. Soverini S, Colarossi S, Gnani A, et al. Resistance to dasatinib in Philadelphia-positive leukemia patients is mainly mediated by the presence or the selection of mutations at residues 315 and 317 in the Bcr-Abl kinase domain. Haematologica. 2007;109:401-4. doi: 10.3324/haematol.10822.

30. Soverini S, Iacobucci I, Baccarani M, and Martinelli G. Targeted therapy and the T315I mutation in Philadelphia-positive leukemias. Haematologica. 2007;92:437-9. doi: 10.3324/haematol.11248.

31. Ernst T, Erben P, Miller M, et al. Dynamics of BCR-ABL mutated clones prior to hematologic or cytogenetic resistance to imatinib. Haematologica. 2008;93:186-92. doi: 10.3324/haematol.11993.

32. Jones D, Thomas D, Yin CC, et al. Kinase domain point mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia emerge after therapy with BCR-ABL kinase inhibitors. Cancer. 2008;113:985-94.

33. Jones D, Luthra R, Cortes J, et al.  BCR-ABL fusion transcript types and levels and their interaction with secondary genetic changes in determining the phenotype of Philadelphia chromosome–positive leukemias. Blood. 2008;112:5190-2. pmid: 18809762.

34. Yeoh EJ, Ross ME, Shurtleff SA, et al. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell. 2002;1:133-43. doi: 10.1016/S1535-6108(02)00032-6.

35. Cimino G, Pane F, Elia M, et al. The role of BCR/ABL isoforms in the presentation and outcome of patients with Philadelphia-positive acute lymphoblastic leukemia: a seven-year update of the GIMEMA 0496 trial. Haematologica. 2006;91:377-80.

36. Burmeister T, Schwartz S, Taubald A, et al. Atypical BCR-ABL mRNA transcripts in adult acute lymphoblastic leukemia. Haematologica. 2007;92:1699-702. doi: 10.3324/haematol.11737.

37. Ohsaka A, Shina S, Kobayashi M, et al. Philadelphia chromosome-positive chronic myeloid leukemia expressing p190(BCR-ABL). Intern Med. 2002;41:1183-7.

38. Jiang DZ, Chen ZM, Lou JY, et al. Cytogenetic and molecular analysis of 1193 cases with chronic myeloid leukemia Zhonghua Xue Ye Xue Za Zhi. 2007;28:1-5. Сhinese. pmid: 17649716.

39. Dhingra K, Talpaz M, Kantarjian H, et al. Appearance of acute leukemia-associated P190 BCR-ABL in chronic myelogenous leukemia may correlate with disease progression. Leukemia. 1991;5:191-5. pmid: 2013978.

40. Melo JV, Myint H, Galton DA., and Goldman JM.  P190BCR-ABL chronic myeloid leukemia: The missing link with chronic myelomonocytic leukemia? Leukemia. 1994;8:208-11. pmid: 8289491.

41. Elliott SL, Taylor KM, Taylor DL, et al. Cytogenetic response to alpha-interferon is predicted in early chronic phase chronic myeloid leukemia by M-bcr breakpoint location. Leukemia. 1995;9:946-50. pmid: 7596182.

42. Shepherd P, Suffolk R, Halsey J, et al. Analysis of molecular breakpoint and m-RNA transcripts in a prospective randomized trial of interferon in chronic myeloid leukemia: No correlation with clinical features, cytogenetic response, duration of chronic phase or survival. Br J Haematol. 1995;89:546-54. pmid: 7734353.

43. Rozman C, Urbano Ispizua A, Cervantes F, et al. Analysis of the clinical relevance of the breakpoint location within M-BCR and the type of chimeric mRNA in chronic myelogenous leukemia. Leukemia. 1995;9:1104-7. pmid: 7596178.

44. Kutsev S, Velchenko M. The role of BCR-ABL gene mutation analysis in the optimization of target therapy of chronic myeloid leukemia. Clin Oncohematol. 2008;1:190-9. Russian.

45. Al-Ali HK, Leiblein S, Kovacs I, et al. CML with an e1a3 BCR-ABL fusion: rare, benign, and a potential diagnostic pitfall. Blood. 2002;100:1092-3.

46. Snyder DS, McMahon R, Cohen SR, et al. Chronic myeloid leukemia with an e13a3 BCR-ABL fusion: benign course responsive to imatinib with an RT-PCR advisory. Am J Hematol. 2004;75:92-5. doi: 10.1002/ajh.10456.

47. Lee JJ, Kim HJ, Lee S, et al. Imatinib induces a cytogenetic response in blast crisis of interferon failure chronic myeloid leukemia patients with e19a2 BCR-ABL transcripts. Leukemia. 2004;18:1539-40. doi: 10.1038/sj.leu.2403454.

48. Popovici C, Cailleres S, David M, et al. E6a2 BCR-ABL fusion with BCR exon 5-deleted transcript in a Philadelphia positive CML responsive to Imatinib. Leuk Lymphoma. 2005;46:1375-7. doi: 10.1080/10428190500138138.

49. Grégoire M-J, Latger-Cannard V, Staal A, et al. Identification of an acute basophilic leukemia carrying a rare e6a2 BCR-ABL transcript. Acta Haematol. 2006;116;216-8. doi: 10.1159/000094686.

50. Colla S, Sammarelli G, Voltorini S, et al. e6a2 BCR-ABL transcript in chronic myeloid leukemia: is it associated with aggressive disease? Haematologica. 2004;89:611-3.

51. Hochhaus A, Reiter A, Sklandy H, et al. A novel BCR-ABL fusion gene (e6a2) in a patient with Philadelphia chromosome negative chronic myelogenous leukemia. Blood. 1996;88:2236-40.

52. Gleiβner B, Gökbuget 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;99:1536–43.

53. Elia LI, Manchini M, Moleti L, et al. A multiplex reverse transcriptase-polymerase chain reaction strategy for the diagnostic molecular screening of chimeric genes: a clinical evaluation on 170 patients with acute lymphoblastic leukemia. Haematologica. 2003;88:275-9.

54. Juric D, Lacayo NJ, Ramsey MC, et al. Differential gene expression patterns and interaction networks in BCR-ABL-positive and –negative adult acute lymphoblastic leukemias. J Clin Oncol. 2007;25:1341-9. doi: 10.1200/JCO.2006.09.3534.

55. Schlieben S, Borkhardt A, Reinisch I, et al. Incidence and clinical outcome of children with BCR/ABL-positive acute lymphoblastic leukemia (ALL). A prospective RT-PCR study based on 673 patients enrolled in the German pediatric multicenter therapy trials ALL-BFM-90 and CoALL-05–92. Leukemia. 1996;10:957-63. pmid: 8667652.

56. Uckun FM, Nachman JB, Sather HN, et al. Clinical significance of Philadelphia chromosome-positive pediatric acute lymphoblastic leukemia in context of contemporary intensive therapies: a report from the children’s cancer group. Cancer. 1998;83:2030–9. doi: 10.1002/(SICI)1097-0142(19981101)83:9<2030::AID-CNCR21>3.0.CO;2-Q.

57. Scheuring US, Pfeifer H, Wassmann B, et al. Methodologic and biological variability of quantitative real-time polymerase chain reaction analysis of Bcr-Abl expression in Philadelphia chromosome-positive acute lymphoblastic leukemia. Haematologica. 2003;88:1074-5.

58. Rotoli B, Pane F, Salvatore F, et al. The e19a2 bcr/abl breakpoint in acute lymphoblastic leukemia. Br J Haematol. 2000;110:493-6.

59. Wilson GA, Van den Berghe EA, Politt RC, et al. Are aberrant BCR-ABL transcripts more common than previously thought? Br J Hematol. 2000;111:1109-11.

60. Wassmann B, Gökbuget N, Scheuring UJ, et al. A randomized multicenter open label phase II study to determine the safety and efficacy of induction therapy with imatinib (Glivec, formerly STI571) in comparison with standard induction chemotherapy in elderly (>55 years) patients with Philadelphia chromosome-positive (Ph+/BCR-ABL+) acute lymphoblastic leukemia (ALL) (CSTI571ADE10). Ann Hematol. 2003;82:716-20.

61. Talpaz M, Shah N, Kantarjian H, et al. Dasatinib in imatinib-resistant Philadelphia-chromosome-positive leukemias. N Engl J Med. 2006;354:2531-41.

62. Volkova MA. New possibilities in the therapy of the chronic myelocytic leukemia: dasatinib. Clin Onkohematol. 2008;1:218-26. Russian.

63. Tiribelli M, Sperotto A, Candoni A, et al. Nilotinib and donor lymphocyte infusion in the treatment of Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL) relapsing after allogeneic stem cell transplantation and resistant to imatinib. Leukemia Res. 2009;33:174-7.

64. Schultz KR, Pullen DJ, Sather HN, et al. Risk and response-based classification of childhood B-precursor acute lymphoblastic leukemia: a combined analysis of prognostic markers from the Pediatric Oncology Group (POG) and Children's Cancer Group (CCG). Blood. 2007;109: 926-35. doi: 10.1182/blood-2006-01-024729.

65. Secker-Walker LM, Craig JM. Prognostic implications of breakpoint and lineage heterogeneity in Philadelphia-positive acute lymphoblastic leukemia: a review. Leukemia. 1993;7:147–51. pmid: 8426467.

66. Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of BCR-ABL positive cells. Nat Med. 1996;2:561-6. pmid: 8616716.

67. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. New Engl J Med. 2001;344:1031-7.

68. Druker BJ, Sawyers CL, Kantarjian H, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001;344:1038-42.

69. Druker BJ, Guilhot F, O’Brien SG, et al. Five-year follow-up of imatinib therapy for newly diagnosed chronic leukemia in chronic-phase shows sustained responses and high overall survival. N Engl J Med. 2006;355:2408-17.

70. Goldman JM. How I treat chronic myeloid leukemia in the imatinib era. Blood. 2007;110:2828-37. doi: 10.1182/blood-2007-04-038943.

71. Kantarjian HM, Cortes J, O’Brien S, et al. Imatinib mesylate (STI571) therapy for Philadelphia chromosome positive chronic myelogenous leukemia in blast phase. Blood. 2002;99:3547-53.

72. Kantarjian HM, O’Brien S, Cortes JE, et al. Imatinib mesylate therapy for relapse after allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood. 2002;100:1590-5.

73. Kantarjian HM, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med. 2003;346:645-52.

74. Kantarjian HM, Talpaz M, O’Brien S, et al. Dose escalation of imatinib mesylate can overcome resistance to standard-dose therapy in patients with chronic myelogenous leukemia. Blood. 2003;101:473-5. doi: 10.1182/blood-2002-05-1451.

75. Kantarjian HM, Talpaz M, O’Brien S, et al. Survival benefit with imatinib mesylate versus-interferon alfa-based regimens in newly diagnosed chronic phase chronic myelogenous leukemia. Blood. 2006a;108:1835-40.

76. Ottmann OG, Druker BJ, Sawyers CL, et al. A phase 2 study of imatinib in patients with relapsed or refractory Philadelphia chromosome-positive acute lymphoid leukemias. Blood. 2002;100:1965-71. doi: 10.1182/blood-2001-12-0181.

77. Radojkovic M, Ristic S, Pavlovic S, Colovic M. Molecular response to imatinib in patient with Ph negative p190 BCR-ABL transcript positive chronic myeloid leukemia with cyclic leukocytosis. Leuk Res. 2009;33:10-2. doi: 10.1016/j.leukres.2008.10.028.

78. Roy L, Guilhot J, Krahnke T, et al. Survival advantage with imatinib compared to the combination interferon-α plus cytarabine in chronic phase CML: historical comparison between two phases III trials. Blood. 2006;108:1478-84. doi: 10.1182/blood-2006-02-001495.

79. de Labarthe A, Rousselot P, Huguet-Rigal F, et al. Imatinib combined with induction or consolidation chemotherapy in patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the GRAAPH-2003 study. Blood. 2007;109:1408-13. doi: 10.1182/blood-2006-03-011908.

80. Vignetti M, Fazi P, Cimino G, et al. Imatinib plus steroids induces complete remissions and prolonged survival in elderly Philadelphia chromosome-positive patients with acute lymphoblastic leukemia without additional chemotherapy: results of the Gruppo Italiano Malattie Ematologiche dell’ Adulto (GIMEMA) LAL201-B protocol. Blood. 2007;109:3676-8. doi: 10.1182/blood-2006-10-052746.

81. Zaritsky AYu, Lomaia EG, Vinogradova OYu, et al. Prognosis factors in Imatinib mesylate therapy in patients with a chronic phase of Ph-positive chronic myeloid leukemia: data from a multicenter non-randomized trial in Russia. Ther Arkh. 2007;79(8):17-22. Russian.

82. Palandri F, Iacobucci I, Martinelli G, et al. Long-term outcome of complete cytogenetic responders after imatinib 400 mg in late chronic phase, Philadelphia-positive chronic myeloid leukemia: The GIMENA Working Party on CML. J Clin Oncol. 2008;26:106-11. doi:  10.1200/JCO.2007.13.2373.

83. Druker BJ. Translation of the Philadelphia chromosome into therapy for CML. Blood. 2008;112:4808-17.

84. Talpaz M, Silver RT, Druker BJ, et al. Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study. Blood. 2002;99:1928-37.

85. Palandri F, Castagnetti F, Testoni N, et al. Chronic myeloid leukemia in blast crisis treated with imatinib 600 mg: outcome of the patients alive after a 6-year follow-up. Haematologica. 2008;93:1792-6. doi: 10.3324/haematol.13068.

86. Sawyers CL, Hochhaus A, Feldman E, et al. Imatinib induces hematologic and cytogenetic responses in patients with chronic myelogenous leukemia in myeloid blast crisis: results of a phase II study. Blood. 2002;99:3530-9.

87. Kantarjian HM, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006;354:2542-51.

88. De Lavallade H, Apperley JF, Khorashad JK, et al. Imatinib for newly diagnose patients with chronic myeloid leukemia: Incidence of sustained responses in an intention-to-treat analysis. J Clin Oncol. 2008;26:3358-63.

89. Baccarani M, Saglio G, Goldman J, et al. Evolving concepts in the managements of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2006;108:1809-20. doi: 10.1182/blood-2006-02-005686.

90. Brave M, Goodman V, dvardas Kaminskas E, et al. Sprycel for chronic myeloid leukemia and Philadelphia chromosome–positive acute lymphoblastic leukemia resistant to or intolerant of imatinib mesylate. Clin Cancer Res. 2008;14:352-9. doi: 10.1158/1078-0432.CCR-07-4175.

91. Steinberg M. Dasatinib: a tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. Clin Ther. 2007;29:2289-308. doi: 10.1016/j.clinthera.2007.11.005.

92. Keam SJ. Dasatinib: in chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. BioDrugs. 2008;22:59-69. pmid: 18215092.

93. Le Coutre P, Ottmann OG, Giles F, et al. Nilotinib (formally AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is active in patients with imatinib-resistant or -intolerant accelerated-phase chronic myelogenous leukemia. Blood. 2008;111:1834-9. doi: 10.1182/blood-2007-04-083196.

94. Castagnetti F, Palandri F, Amabile M, et al. Results of high-dose imatinib mesylate in intermediate Sokal risk chronic myeloid leukemia patients in early chronic phase: a phase 2 trial of the GIMEMA CML Working Party. Blood. 2009;113:3428-34. doi: 10.1182/blood-2007-08-103499.

95. Palandri F, Castagnetti F, Alimena G, et al. The long-term durability of cytogenetic responses in patients with accelerated phase chronic myeloid leukemia treated with imatinib 600 mg: the GIMEMA CML Working Party experience after a 7-year follow-up. Haematologica. 2009;94:205-212. doi: 10.3324/haematol.13529.

96. Tokarski JS, Newitt JA, Cheng CY, et al. The structure of dasatinib (BMS-354825) bound to activated ABL kinase domain elucidates its inhibitory activity against imatinib-resistant ABL mutants. Cancer Res. 2006;66:5790-7. 97. Guilhot F, Apperley J, Kim D-W, et al. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or- intolerant chronic myeloid leukemia in accelerated phase. Blood. 2007;109:4143-50.

97. Guilhot F, Apperley J, Kim D-W, et al. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or- intolerant chronic myeloid leukemia in accelerated phase. Blood. 2007;109:4143-50. doi: 10.1182/blood-2006-09-046839.

98. Jabbour E, Kantarjian H, Jones D, et al. Characteristics and outcomes of patients with chronic myeloid leukemia and T315I mutation following failure of imatinib mesylate therapy. Blood. 2008;112:53-5. doi: 10.1182/blood-2007-11-123950.

99. Jabbour E, Kantarjian HM, Jones D, et al.  Characteristics and outcome of chronic myeloid leukemia patients with F317L BCR-ABL kinase domain mutation after  therapy with tyrosine kinase inhibitors. Blood. 2008;112:4839-42. doi: 10.1182/blood-2008-04-149948.

100. Thomas DA, Faderl S, Cortes J, et al. Treatment of Philadelphia chromosome-positive acute lymphocytic leukemia with hyper-CVAD and imatinib mesylate. Blood 2004; 103:4396-407. doi: 10.1182/blood-2003-08-2958.

101. Thomas D.A. Philadelphia chromosome–positive acute lymphocytic leukemia: a new era of challenges. Hematology Am Soc Hematol Educ Program Book. 2007:435-43.

102. Towatari M, Yanada M, Usui N, et al. Combination of intensive chemotherapy and imatinib can rapidly induce high-quality complete remission for a majority of patients with newly diagnosed BCR-ABL-positive acute lymphoblastic leukemia. Blood. 2004;104:3507-12. doi: 10.1182/blood-2004-04-1389.

103. Ottmann OG, Wassmann B. Treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia. Hematology. Am Soc Hematol Educ Program. 2005:118-22.

104. Yanada M, Naoe T. Imatinib combined chemotherapy for Philadelphia chromosome-positive acute lymphoblastic leukemia: major challenges in current practice. Leuk Lymphoma. 2006;47:1474-53. doi: 10.1080/10428190600634085.

105. 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;24:460-6.

106. Wassmann B, Pfeifer H, Scheuring UJ, et al. Early prediction of response in patients with relapsed or refractory Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) treated with imatinib. Blood. 2004;103:1495-8. doi: 10.1182/blood-2003-01-0154.

107. Lee S, Kim Y-J, Min C-K, et al.  The effect of first-line imatinib interim therapy on the outcome of allogeneic stem cell transplantation in adults with newly diagnosed Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood. 2005;105:3449-57. doi: 10.1182/blood-2004-09-3785.

108. Wassmann B, Pfeifer H, Stadier M, et al. Early molecular response to post transplantation imatinib determines outcome in MRD+ Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL). Blood. 2005;106:458-63. doi: 10.1182/blood-2004-05-1746.

109. Wassmann B, Pfeifer H, Gökbuget N, et al. Alternating versus concurrent schedules of imatinib and chemotherapy as front-line therapy for Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL). Blood. 2006;108:1469-77. doi: 10.1182/blood-2005-11-4386.

110. Ohno R. Treatment of adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia. Curr Oncol Rep. 2008;10:379-87. pmid: 18706265.

111. Scheuring US, Pfeifer H, Wassmann B, et al. Early minimal residual disease (MRD) analysis during treatment of Philadelphia chromosome/Bcr-Abl-positive acute lymphoblastic leukemia with the Abl-tyrosine kinase inhibitor imatinib (STI571). Blood. 2003;101:85-90. doi: 10.1182/blood-2002-02-0360.

112. Ottmann OG, Wassmann B, Pfeifer H, et al. Imatinib compared with chemotherapy as front-line treatment of elderly patients with Philadelphia chromosome-positive acute lymphoblastic leukemia. Cancer. 2007;109:2068-76.

113. Apperley JF. Mechanisms of resistance to imatinib in chronic myeloid leukemia. Part I. Lancet Oncol. 2007;8:1018-29. pmid: 17976612.

114. Khorashad JS, de Lavallade H, Apperley JF, et al. Finding of kinase domain mutations in patients with chronic phase chronic myeloid leukemia responding to imatinib may identify those at high risk of disease progression. J Clin Oncol. 2008;26:4806-13. pmid: 18645191.

115. Saglio G, Ulishiani S, Bosa M, et al. New therapeutic approaches and prognostic factors in chronic myeloid leukemia. Leuk Lymphoma. 2008;49:625-8. doi: 10.1080/10428190801896210.

116. Verma D, Fava C, Kantarjian H, Cortes J. Complexity of BCR-ABL kinase domain mutations during the course of therapy with tyrosine kinase inhibitors in chronic myeloid leukemia. Am J Hematol. 2009;84:256-7. doi: 10.1002/ajh.21366.

117. Jabbour E, Kantarjian H, Jones D, et al. Frequency and clinical significance of BCR-ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate. Leukemia. 2006;20:1767-73. doi: 10.1038/sj.leu.2404318.

118. Branford S. Chronic myeloid leukemia: molecular monitoring in clinical practice. Hematology Am Soc Hematol Educ Program. 2007:376-83.

119. Al-Ali HK, Heinrich MC, Lange T, et al.  High incidence of BCR-ABL kinase domain mutations and absence of mutations of the PDGFR and KIT activation loops in CML patients with secondary resistance to imatinib. Hematol J. 2004;5:55-60.

120. Lahaye T, Riehm B, Berger U, et al.  Response and resistance in 300 patients with BCR-ABL-positive leukemias treated with imatinib in a single center: a 4.5-year follow-up. Cancer. 2005;103:1659-69. doi: 10.1002/cncr.20922.

121. Quintas-Cardama A, Kantarjian H, Jones D, et al. Dasatinib (BMS-354825) is active in Philadelphia chromosome-positive chronic myelogenous leukemia after imatinib and nilotinib (AMN107) therapy failure. Blood. 2007;109:497-9. doi: 10.1182/blood-2006-07-035493.

122. Cortes J, Rousselot P, Kim D, et al. Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or intolerant chronic myeloid leukemia in accelerated phase. Blood. 2007;109:3207-13. doi: 10.1182/blood-2006-09-046888.

123. Shah NP, Skaggs BJ, Branford S, et al. Sequential ABL kinase inhibitor therapy selects for compound drug-resistant BCR-ABL mutations with altered oncogenic potency. J Clin Invest. 2007;117:2562-9. doi: 10.1172/JCI30890.

124. Soverini S, Martinelli G, Colarossi S, et al. Second-line treatment with dasatinib in patients resistant to imatinib can select novel inhibitor-specific BCR-ABL mutants in Ph+ ALL. Lancet Oncol. 2007;8:273-4. doi: 10.1016/S1470-2045(07)70078-5.

125. Jabbour E, Kantarjian HM, Jones D, et al. Characteristics and outcome of chronic myeloid leukemia patients with F317L BCR-ABL kinase domain mutation after therapy with tyrosine kinase inhibitors. Blood. 2008;112:4839-42. doi: 10.1182/blood-2008-04-149948.

126. Giles FJ, Cortes J, Jones D, et al. VK-047, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation. Blood. 2007;109:500-2. doi: 10.1182/blood-2006-05-025049.

127. Nicolini FE, Corm S, Lĕ QH, et al. Mutation status and clinical outcome of 89 imatinib mesylate-resistant chronic myelogenous leukemia patients: a retrospective analysis from the French intergroup of CML (Fi(phi)-LMC GROUP). Leukemia. 2006;20:1061-6. doi: 10.1038/sj.leu.2404236.

128. Hochhaus A, Kantarjian H, Baccarani M, et al. Dasatinib induces notable hematologic and cytogenetic responses in chronic- phase chronic myeloid leukemia after failure of imatinib therapy. Blood. 2007;109:2303-9. doi: 10.1182/blood-2006-09-047266.

129. Zonder JA, Pemberton P, Brandt H, et al. The effect of dose increase of imatinib mesylate in patients with chronic or accelerated phase chronic myelogenous leukemia with inadequate hematologic or cytogenetic response to imatinib treatment. Clin Cancer Res. 2003;9:2092-7.

130. Chu S, Xu H, Snyder DS, et al. Detection of BCR-ABL kinase mutations in CD34+ cells from chronic myelogenous leukemia patients in complete cytogenetic remission on imatinib mesylate treatment. Blood. 2005;105:2093-8. doi: 10.1182/blood-2004-03-1114.

131. Sherbenou DW, Wong MJ, Humayun A, et al. Mutations of the BCR-ABL-kinase domain occur in a minority of patients with stable complete cytogenetic responses to imatinib. Leukemia. 2007;21:489-93.

132. Hughes T. ABL kinase inhibitor therapy for CML: baseline assessments and response monitoring. Hematology Am Soc Hematol Educ Program Book. 2006:211-8.

133. Hughes T, Deininger M, Hochhaus A, et al. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 2006;108:28-37. doi: 10.1182/blood-2006-01-0092.

134. Pfeifer H, Wassmann B, Pavlova A, et al. Kinase domain mutations of BCR-ABL frequency precede Imatinib-based therapy and give to relapse in patients with de novo Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL). Blood. 2007;110:727-34. doi: 10.1182/blood-2006-11-052373.

135. Passweg JR, Walker I, Sobocinski KA, et al. Validation and extension of the EBMT risk score for patients with chronic myeloid leukemia (CML) receiving allogeneic haemopoietic stem cell transplants. Br J Haematol. 2004;125:613-20.

136. Sullivan KM, Weiden PL, Storb R, et al.  Influence of acute and chronic graft-versus-host disease on relapse and survival after bone transplantation from HLA-identical siblings as treatment of acute and chronic leukemia.  Blood. 1989;73:1720-8.

137. Serrano J, Roman J, Sanchez J, et al.  Molecular analysis of lineage-specific chimerism and minimal residual disease by RT-PCR of p210 (BCR-ABL) and p190 (BCRABL) after allogeneic bone marrow transplantation for chronic myeloid leukemia: increasing mixed myeloid chimerism and p190 (BCR- ABL) detection precedes cytogenetic relapse. Blood. 2000;95:2659–65.

138. Collins RH, Roggers ZR, Bennett M, et al. Hematologic relapse of chronic myelogenous leukemia following allogeneic bone marrow transplantation: Apparent graft-versus-leukemia effect following abrupt discontinuation of immunosuppression. Bone Marrow Transplant. 1992;10:391-5. pmid: 1422499.

139. Kolb HJ, Schattenberg A, Goldman JM, et al. Graft-versus leukemia effect of donor lymphocyte transfusions in marrow grafted patients. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia. Blood. 1995;86:2041-50.

140. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371-83.

141. Mackinnon S, Papadopoulos EB, Carabasi MH, et al. Adoptive immunotherapy evaluating escalating doses of donor leukocytes  for relapse of chronic myeloid leukemia after bone marrow transplantation: Separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86:1261-8.

142. Collins RH, Shpilberg O, Drobyski WR, et al. Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. J Clin Oncol. 1997;15:433-44. pmid: 9053463.

143. Dazzi F, Szydio RM, Goldman JM. Donor lymphocyte infusion for relapse of chronic myeloid leukemia after allogeneic stem cell transplant: Where we now stand. Exp Hematol. 1999;27:1477-86.

144. Dazzi F, Szydio RM, Craddock C, et al. Comparison of single-dose and escalating-dose regimens of donor lymphocyte infusion for relapse after allografting for chronic myeloid leukemia. Blood. 2000;95:67-71.

145. Guglielmi C, Arcese W, Dazzi F, et al. Donor lymphocyte infusion for relapsed chronic myelogenous leukemia: Prognostic relevance of the initial cell dose. Blood. 2002;100:397-405.

146. Olavarria E, Ottman OG, Deininger M, et al. Chronic Leukemia Working Party of the European Group of Bone and Marrow Transplantation (EBMT). Response to imatinib in patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Leukemia. 2003;17:1707-12.

147. Olavarria E, Siddique Sh, Griffiths MJ, et al. Posttransplantation imatinib as a strategy to postpone the requirement for immunotherapy in patients undergoing reduced-intensity allografts for chronic myeloid leukemia. Blood. 2007;110:4614-7. doi: 10.1182/blood-2007-04-082990.

148. Tomblyn M, Lazarus HM. Donor lymphocyte infusion: the long and winding road: how should it be traveled? Bone Marrow Transplant. 2008;42:569-79. doi: 10.1038/bmt.2008.259. 

149. DeAngelo DJ, Hochberg EP, Alyes EP, et al. Extended follow-up of patients treated with imatinib mesylate (Gleevec) for chronic myelogenous leukemia relapse after allogeneic transplantation. Durable cytogenetic remission and conversion to complete donor chimerism without graft-versus-host disease. Clin Cancer Res. 2004;10:5065-71. doi: 10.1158/1078-0432.CCR-03-0580.

150. Duniop LC, Powles R, Singhal S, et al. Bone marrow transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia. Bone Marrow Transplant. 1996;17:365-9. pmid: 8704688.

151. Snyder DS, Nademanee AP, O’Donnell MR, et al. Long-term follow-up of 23 patients with Philadelphia chromosome-positive acute lymphoblastic leukemia treated with allogeneic bone marrow transplant in first complete remission. Leukemia. 1999;13:2053-8. pmid: 10602428.

152. Laport GG, Alvarnas JC, Palmer JM, et al. Long-term remission of Philadelphia chromosome-positive acute lymphoblastic leukemia after allogeneic hematopoietic cell transplantation from matched sibling donors: a 20-year experience with the fractioned total body irradiation-etoposide regimen. Blood. 2008;112:903-9. doi: 10.1182/blood-2008-03-143115.

153. Hoelzer D, Gökbuget N, Ottmann OG. Targeted therapies in the treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia. Semin Hematol. 2002;39:32-7. pmid: 12447850.

154. Avivi I, Goldstone AH. Bone marrow transplant in Ph+ ALL patients. Bone Marrow Transplant. 2003;31:623-32. doi: 10.1038/sj.bmt.1703899.

155. Fielding AK, Goldstone AH. Allogeneic hematopoietic stem cell transplant in Philadelphia-positive acute lymphoblastic leukemia. Bone Marrow Transplant. 2008;41:447-53. doi: 10.1038/sj.bmt.1705904.

156. Bachanova V, Weisdorf D. Unrelated donor allogeneic transplantation for adult acute lymphoblastic leukemia: a review. Bone Marrow Transplant. 2008;41:455-64. doi: 10.1038/sj.bmt.1705889.

157. Lee S, Kim YJ, Chung NG, et al. The extent of minimal residual disease reduction after the first 4-week imatinib therapy determines outcome of allogeneic stem cell transplantation in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia. Cancer. 2009;115:561-70. doi: 10.1002/cncr.24026.

158. Forman SJ, O’Donnell MR, Nademanee AP, et al. Bone marrow transplantation for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood. 1987;70:587-8.

159. Barrett AJ, Horowitz MM, Ash RC, et al. Bone marrow transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood. 1992;79:3067-70.

160. Arico M, Valsecchi MG, Camitta B, et al. Outcome of treatment in children with Philadelphia chromosome-positive acute lymphoblastic leukemia. N Engl J Med. 2000;342:998-1006.

161. Dombert 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;100:2357-66. doi: 10.1182/blood-2002-03-0704.

162. Esperou H, Boiron JM, Cayuela JM, et al. A potential graft-versus leukemia effect after allogeneic hematopoietic stem cell transplantation for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: results from the French Bone Marrow Transplantation Society. Bone Marrow Transplant. 2003;31:909-18. doi: 10.1038/sj.bmt.1703951.

163. Gupta V, Yi QL, Brandwei J, et al. The role of allogeneic bone marrow transplantation in adult patients below the age of 55 years with acute lymphoblastic leukemia in first complete remission: a donor vs no donor comparison. Bone Marrow Transplant. 2004;33:397-404. doi: 10.1038/sj.bmt.1704368.

164. Goldstoune AH, Chopra R, Buck G, et al. The outcome of 267 Philadelphia positive adults in the International UKALLXII/ECOG E2993 study. Final analysis and the role of allogeneic transplant in those under 50 years [abstract]. Blood. 2003;102:80a.

165. Martin TG, Gajewski JL. Allogeneic stem cell transplantation for acute lymphocytic leukemia in adults: advances in the treatment of adult acute lymphocytic leukemia. Hematol Oncol Clin North Am. 2001;15:97-120.

166. Abou Mourad YR, Fernandez HF, Kharfan-Dabaja MA. Allogeneic hematopoietic cell transplantation for adult Philadelphia-positive acute lymphoblastic leukemia in the era of tyrosine kinase inhibitors. Biol Blood Marrow Transplant. 2008;14:949-58.

167. Stein A, Forman SJ. Allogeneic transplantation for ALL in adults. Bone Marrow Transplant. 2008;41:439-46. doi: 10.1038/bmt.2008.1.

168. Kovacsovics T, Maziarz R.T. Philadelphia chromosome-positive acute lymphoblastic leukemia: impact of imatinib treatment on remission induction and allogeneic stem cell transplantation. Curr Oncol Rep. 2006;8:343-51. pmid: 16901395.

169. Carpenter PA, Snyder DS, Flowers ME. Prophylactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia. Blood. 2007;109:2791-3. doi: 10.1182/blood-2006-04-019836.

170. Merante S, Colombo AA, Calatroni S, et al. Nilotinib restores long-term full-donor chimerism in Ph-positive acute lymphoblastic leukemia relapsed after allogeneic transplantation. Bone Marrow Transplant. 2009; Feb 9. [Epub ahead of print]. doi: 10.1038/bmt.2009.6.

171. Collins Jr RH, Goldstein S, Giralt S, et al. Donor leukocyte infusions in acute lymphocytic leukemia. Bone Marrow Transplant. 2000;26:769-74.

172. Kolb HJ, Schmid C, Barrett AJ, et al. Graft-versus-leukemia reactions in allogeneic chimeras. Blood. 2004;103:767-76. doi: 10.1182/blood-2003-02-0342.

173. Choi SJ, Lee JH, Lee JH, et al. Treatment of relapsed acute lymphoblastic leukemia after allogeneic bone marrow transplantation with chemotherapy follower by G-CSF-primed donor leukocyte infusion: a prospective study. Bone Marrow Transplant. 2005;36:163-9.

174. Deiniger MW. Milestones and monitoring in patients with CML treated with imatinib. Hematology Am Soc Hematol Educ Program. 2008;419-26.

175. Goldman J. Monitoring minimal residual disease in BCR-ABL-positive chronic myeloid leukemia in the imatinib era. Curr Opin Hematol. 2005;12(1):33-9. pmid: 15604889.

176. Raanani P, Ben-Bassat I, Gan S, et al. Assessment of the response to imatinib in chronic myeloid leukemia patients: comparison between the FISH, multiplex and RT-PCR methods. Eur J Haematol. 2004;73:243-50. doi: 10.1111/j.1600-0609.2004.00287.x.

177. Schoch C, Schnittger S, Bursch S, et al. Comparison of chromosome banding analysis, interphase- and hyper metaphase-FISH, qualitative and quantitative PCR for diagnosis and for follow-up in chronic myeloid leukemia: a study on 350 cases. Leukemia. 2002;16:53-9.

178. Branford S, Hughes T. Diagnosis and monitoring of chronic myeloid leukemia by qualitative and quantitative RT-PCR.  Methods Mol Med. 2006;125:69-92. pmid: 16502578.

179. Hughes T, Branford S. Molecular monitoring of BCR-ABL as a guide to clinical management in chronic myeloid leukemia. Blood Rev. 2006;20:29-41. doi: 10.1016/j.blre.2005.01.008.

180. Wang Y, Wu D, Sun A, et al. Allogeneic hematopoietic stem cell transplantation for patients with chronic myeloid leukemia in second chronic phase attained by imatinib after onset of blast crisis. Int J Hematol. 2008;87167-71. doi: 10.1007/s12185-008-0032-4.

181. Branford S, Cross NC, Hochhaus A, et al. Rationale for the recommendations for harmonizing current methodology for detecting BCR-ABL transcripts in patients with chronic myeloid leukemia. Leukemia. 2006;20:1925-30. doi: 10.1038/sj.leu.2404388.

182. Muller MC, Saglio G, Lin F, et al. An international study to standardize the detection and quantitation of BCR-ABL transcripts from stabilized peripheral blood preparations by quantitative RT-PCR. Haematologica. 2007;92:970-3. doi: 10.3324/haematol.11172.

183. O’Dwyer ME, Mauro MJ, Blasdel C, et al. Clonal evolution and lack of cytogenetic response are adverse prognostic factors for hematologic relapse of chronic phase CML patients treated with imatinib mesylate. Blood. 2004;103:451-5. doi: 10.1182/blood-2003-02-0371.

184. Branford S, Rudzki Z, Parkinson I, et al. Real-time quantitative PCR analysis can be used as a primary screen to identify patients with CML treated with imatinib who have BCR-ABL kinase domain mutations. Blood. 2004;104:2926-32. doi: 10.1182/blood-2004-03-1134.

185. Jabbour E, Kantarjian HM, Abruzzo LV, et al. Chromosomal abnormalities in Philadelphia chromosome negative metaphases appearing during imatinib mesylate therapy in patients with newly diagnosed chronic myeloid leukemia in chronic phase. Blood. 2007;110:2991-5. doi: 10.1182/blood-2007-01-070045.

186. Martynkevich IS, Martynenko LS, Ivanova M.P, et al. Clonal evolution in Ph-positive CML Gleevec treated patients. Bulletin Hematol. 2007;3(1):30-4. Russian.

187. Machova Polakova K, Zmekova V, Rulcova J, et al. BCR-ABL mutations in chronic myeloid leukemia – Not only detection. Leukemia Lymphoma. 2008;49:1620-2.

188. Spinelli O, Peruta B, Tosi M, et al. Clearance of minimal residual disease after allogeneic stem cell transplantation and the prediction of the clinical outcome of adult patients with high-risk acute lymphoblastic leukemia. Haematologica. 2007;92:612-8. doi: 10.3324/haematol.10965.

189. Thörn I, Botling J, Hermansson M, et al. Monitoring minimal residual disease with flow cytometry, antigen-receptor gene rearrangements and fusion transcript quantification in Philadelphia-positive childhood acute lymphoblastic leukemia. Leuk Res. 2009;33:1047-1054. doi: 10.1016/j.leukres.2008.11.031.

190. Willis SG, Lange T, Demehri S, et al. High sensitivity detection of Bcr-ABL kinase domain mutations in imatinib-naïve patients: correlation with clonal cytogenetic evolution but not response to therapy. Blood. 2005;106:2128-37. doi: 10.1182/blood-2005-03-1036.

191. Marin D, Milojkovic D, Olavarria E, et al. European LeukemiaNet criteria for failure or suboptimal response reliably identify patients with CML in early chronic phase treated with imatinib whose eventual outcome is poor. Blood. 2008;112:4437-44. doi: 10.1182/blood-2008-06-162388.

192. Rousselot P, Huguet F, Rea D, et al. Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood. 2007;109:58-60. doi: 10.1182/blood-2006-03-011239.

193. Fava C, Kantarjian HM, Jabbour E, et al. Failure to achieve a complete hematologic response at the time of achievement of a major cytogenetic response with second generation tyrosine kinase inhibitors is associated with a poor prognosis among patients with chronic myeloid leukemia in accelerated or blast phase. Blood. 2009;00:00-00. doi: 10.1182/blood-2008-10-184960.

194. Jones LK, Saha V. Philadelphia positive acute lymphoblastic leukemia of childhood. Br J Haematol. 2005;130:489-500. doi: 10.1111/j.1365-2141.2005.05611.x.

195. Iacobucci I, Lonetti A, Cilloni D, et al. Identification of different Ikaros transcripts in Philadelphia-positive adult acute lymphoblastic leukemia by a high-throughput capillary electrophoresis sizing method. Haematologica. 2008;93:1814-21. doi: 10.3324/haematol.13260.

196. Iacobucci I, Lonetti A, Messa F, et al. Expression of spliced oncogenic Ikaros isoforms in Philadelphia-positive acute lymphoblastic leukemia patients treated with tyrosine kinase inhibitors: implications for a new mechanism of resistance. Blood. 2008;112:3847-55. doi: 10.1182/blood-2007-09-112631.

197. Mullighan CG, Miller CB, Phillips LA, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453:110-4. doi: 10.1038/nature06866.

198. Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360:470-80.

199. Georgopoulos K. Acute lymphoblastic leukemia – on the wings of IKAROS. N Engl J Med. 2009;360:524-6.

200. Rieder H, Ludwig WD, Gassmann W, et al. Prognostic significance of additional chromosome abnormalities in adult patients with Philadelphia chromosome positive acute lymphoblastic leukemia. Br J Haematol. 1996;95:678-91. pmid: 8982045.

201. Thomas X, Thiebaut A, Olteanu N, et al. Philadelphia chromosome positive adult acute lymphoblastic leukemia: characteristics, prognostic factors and treatment outcome. Hematol Cell Ther. 1998;40:119-28. pmid: 9698220.

202. Wetzler M, Dodge RK, Mrozek K, et al. Additional cytogenetic abnormalities in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia: a study of the Cancer and Leukemia Group B. Br J Haematol. 2004;124:275-88.

203. Moormann 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) UKALL111/Eastern Cooperative Oncology Group (ECOG) 2993 trial. Blood. 2007;109:3189-97. doi: 10.1182/blood-2006-10-051912.

204. Pullarkat V, Slovak ML, Kopecky KJ, et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111:2563-72. doi: 10.1182/blood-2007-10-116186.

205. Li Y, Qiu L, Zou D, Zhao Y, et al. Additional chromosomal abnormalities and their prognostic significance in adult Philadelphia-positive acute lymphoblastic leukemia: with or without imatinib in chemotherapy. Ann Hematol. 2009; Mar 10. [Epub ahead of print]. doi: 10.1007/s00277-009-0720-z.

206. Yanada M, Takeuchi J, Sugiura I, et al. Karyotype at diagnosis is the major prognostic factor predicting relapse-free survival for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia treated with imatinib-combined chemotherapy. Haematologica. 2008;93:287-290. doi: 10.3324/haematol.11891.

207. Paulsson K, Cazier J-B, MacDougall F, et al. Microdeletions are a general feature of adult and adolescent acute lymphoblastic leukemia: Unexpected similarities with pediatric disease. Proc National Acad Sci. 2008;105:6708-13.

208. Eiring AM, Neviani P, Santhanam R, et al. Identification of novel posttranscriptional targets of the BCR/ABL oncoprotein by ribonomics: requirement of E2F3 for BCR/ABL leukemogenesis. Blood. 2008;111:816-28. doi: 10.1182/blood-2007-05-090472.

209. Laport GG, Alvarnas JC, Palmer JM, et al. Long-term remission of Philadelphia chromosome-positive acute lymphoblastic leukemia after allogeneic hematopoietic cell transplantation from matched sibling donors: a 20-year experience with the fractionated total body irradiation-etoposide regimen. Blood. 2008;112:903-9. doi: 10.1182/blood-2008-03-143115.

Volume 1, Number 4
03/01/2010

Download PDF version

doi 10.3205/ctt-2009-en-000047.01
Submitted 05 June 2009
Accepted 25 August 2009
Published 31 August 2009

Back to the list