Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Molecular Monitoring and Treatment of Chronic Myeloid Leukemia (CML)
ISSN: 2161-0681
Journal of Clinical & Experimental Pathology

Like us on:

Make the best use of Scientific Research and information from our 700+ peer reviewed, Open Access Journals that operates with the help of 50,000+ Editorial Board Members and esteemed reviewers and 1000+ Scientific associations in Medical, Clinical, Pharmaceutical, Engineering, Technology and Management Fields.
  • Review Article   
  • J Clinic Experiment Pathol 2012, Vol 2(4): 113
  • DOI: 10.4172/2161-0681.1000113

Molecular Monitoring and Treatment of Chronic Myeloid Leukemia (CML)

Zeba Singh1*, Stephen Medlin2 and Saad Z Usmani2
1Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
2Myeloma Institute for Research & Therapy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
*Corresponding Author: Zeba Singh, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 502 Little Rock, AR 72205, USA, Email: zsingh@uams.edu

Received: 09-May-2012 / Accepted Date: 14-Jun-2012 / Published Date: 17-Jun-2012 DOI: 10.4172/2161-0681.1000113

Keywords: CML; Targeted therapy; Tyrosine kinase inhibitors; Drug resistance; Molecular monitoring

Introduction

In 1960, Peter Nowell and David Hungerford made the seminal observation of an abnormally small chromosome in patients with Chronic Myeloid Leukemia (CML), which they named the Philadelphia or Ph1 chromosome [1] for the city where the discovery was made. In the early 1970’s, Janet Rowley identified the underlying non-random balanced chromosomal translocation between chromosomes 9q34 and 22q21 [2]. Molecularly, this translocation fuses the 5’ end of the BCR gene on chromosome 22 with 3’ end of the ABL1 gene on chromosome 9 and gives rise to the chimeric BCR-ABL gene [3]. Ph1 was the first disease-specific chromosomal abnormality described in cancer and is the disease defining abnormality for Chronic Myeloid Leukemia (CML). The BCR-ABL1 chimeric gene on derivative chromosome 22 has since been established to drive the oncogenesis.

In the fifty years since the first identification of the Ph1 chromosome, the sequence of discoveries and inventions made in understanding the biology, detection strategies, and treatment of CML exemplify the concept of molecular medicine. The increase in understanding of the disease has resulted in a paradigm shift in treatment from the traditional combination of Hydroxyurea, interferon-alpha, and Ara-C to a targeted approach by imatinib mesylate, (Glivec/Gleevec; formerly STI571; Novartis Pharmaceuticals, East Hanover, NJ), a tyrosine kinase inhibitor (TKI) that targets the breakpoint cluster region-v-abl, the Abelson murine leukemia viral oncogene homolog 1, also known as ABL1, and by competitive inhibition, disrupts the downstream oncogenic signals. In the IRIS (International Randomized Study of Interferon and STI571) trial, imatinib proved superior to a combination of interferon-alpha and cytarabine in having statistically significant higher rates of hematological and complete cytogeneic responses, lower risk of progression to advanced disease states, and increased progression free survival [4]. Imatinib has thus revolutionized the treatment and outcome of CML, making it the paradigm of molecularly targeted cancer therapies. The success of imatinib in the IRIS trial necessitated the development of sensitive laboratory methods for molecular monitoring of the disease. This review summarizes the recent advances in the treatment of CML and the current appropriate methods for establishing diagnosis and molecular monitoring of the disease.

Natural History of the Disease

CML is characterized by uncontrolled proliferation of primarily the granulocytic and megakaryocytic lineages resulting in accumulation of immature granulocytic precursors in the one marrow, blood and other organs. If left untreated, the natural history of CML follows a course progressing through a chronic phase to an accelerated phase (in most cases) and terminating in blast crisis over 3-5 years. Disease progression is heralded by increasing genomic instability and acquisition of additional chromosomal abnormalities, the most common of which are +8, +19, del(9)(q34), isochromosome 17, and additional Ph1 chromosome. Some recent studies have shown emergence of the RUNX1 mutations during progression of CP-CML to blast crisis [5]. The blast transformation is myeloblastic in twothirds, and lymphoblastic (precursor B-) in one-third of the patients. Blasts with mixed phenotypes representing different lineages are not uncommon, consistent with the stem cell origin of the disease [6-8].

Molecular Pathogenesis

Studies in mice have shown that translocation between BCR and v-ABL is sufficient to drive oncogenesis [9]. The chimeric BCR-ABL gene encodes for an abnormal, constitutively active tyrosine kinase, which phosphorylates and activates downstream signaling pathways including PI3K, AKT, SRC-kinase, and STAT that in turn modulate transcription factors related to cell proliferation, survival, protection from apoptosis, and differentiation. In addition, the BCR-ABL protein causes oxidative damage to the DNA by induction of reactive oxygen species and also interferes with the DNA-repair pathway [10], thus making the cells genetically unstable and prone to accumulate deleterious mutations. Nearly 80% of patients with CML have additional chromosomal abnormalities, many of which are associated with disease progression.

Laboratory Diagnosis of CML

The WHO classification has laid down definite criteria for the chronic phase, accelerated phase, and blast crisis of CML. Whereas demonstration of the defining BCR-ABL1 translocation is a must for establishing the diagnosis of CML, the diagnosis can easily be made from the peripheral smear in most cases. Leukocytosis with circulating granulocytic precursors, absolute basophilia, and thrombocytosis form the backbone of morphological diagnosis of chronic phase CML (CPCML). In CP-CML, the peripheral blood and bone marrow have less than 10% blasts. Accelerated disease is associated with increasing leukocyte count, increase in circulating or bone marrow blasts, basophilia of >20% in the peripheral blood, thrombocytosis or thrombocytopenia, and bone marrow fibrosis with proliferation and clustering of abnormal megakaryocytes. The last mentioned morphological feature, while not a definitive criterion, is often observed with the other defined WHO criteria for accelerated disease. For details on the clinical features and diagnostic criteria of the different stages of disease evolution, the reader is referred to the WHO Blue book on the Classification of tumors of the hematopoietic and lymphoid system [6].

Demonstration of the translocation t (9;22) (q34;q22) or the BCR-ABL1 chimeric gene product is quintessential to establish the diagnosis. The Ph1 chromosome can be demonstrated by G-banded metaphase cytogenetics or fluorescent in-situ hybridization (FISH), and the abnormal BCR-ABL transcript by RT-PCR (reverse transcriptase- PCR). Each of these methods has its pros and cons as a diagnostic tool or for subsequent monitoring of the disease.

Conventional metaphase cytogenetics and fluorescent in-situ hybridization

In 95% of patients, the Ph1 chromosome can be detected by metaphase cytogenetics or G-banding [11]. Analysis of 20-25 metaphases for presence of Ph1 chromosome is standard practice. A bone marrow sample is required for obtaining metaphases from dividing cells, entailing the risk associated with an invasive procedure. In about 5% cases the result may be negative due to the overgrowth of normal bone marrow cells or inadequate metaphases for analysis. The specific translocation may be masked in the presence of a complex translocation involving three or more chromosomes, or when there is no alteration in the size of chromosome 22 due to cryptic interstitial insertions of the ABL1 within the BCR gene [12]. The main advantage of G-banding is that no a priori information about the chromosomal abnormalities is required and it can detect abnormalities that may be present in addition to the Ph1 chromosome. The commonest secondary abnormalities in CML are an additional Ph1 chromosome, +8, and isochromosome 17q. Several previous studies had reported rapid disease progression and worse outcome in the 10-15% of patients who develop deletions of the derivative chromosome 9 [13-15]; in recent studies however, authors show that the adverse effect of this chromosomal abnormality on the EFS in chronic phase patients can be mitigated by second generation TK inhibitors [4-16]. Fluorescent in-situ hybridization (FISH) has a higher sensitivity for detection of Ph1 than metaphase cytogenetics. The dual-color, dual-fusion FISH probe set is very sensitive and has high specificity of 1.0 to 0.1%. The major advantage of FISH is that it can be performed on interphase, non-dividing cells from peripheral blood, precluding requirement for the invasive, costly, and labor intensive bone marrow procedure. The results obtained from the peripheral blood sample are comparable in sensitivity to those obtained from the bone marrow cells [17]. FISH should however not be considered an alternative for metaphase cytogenetics. Current guidelines recommend that cytogenetic analysis be performed at baseline, and then at 3 to 6 month intervals after initiation of TK inhibitor therapy [18,19]. Attaining the defined levels of cytogenetic responses at specific time points correlates with response to therapy and likelihood of subsequent relapse or disease progression. The definitions for hematological, cytogenetic and molecular remission are depicted in 9 (Table 1). (Table 2) shows the NCCN and ELN recommendations for time points for metaphase cytogenetics, FISH, and quantitative PCR. Patients with a major cytogenetic response (MCyR) including those with a partial response (1-34% Ph1 positive metaphases), or a complete cytogenetic response (CCyR: no Ph1 positive metaphases) are less likely to progress to advanced phase CML. Achieving CCyR at 12 months is a major landmark in therapeutic response to imatinib [20-21].

HEMATOLOGICAL

Complete (CHR)

Partial (PHR)


WBC< 1x109/L; basophils <5%; no circulating granulocyte precursors; platelet count <450x109/L; no splenomegaly

Levels less than the above
CYTOGENETIC

Partial (PCyR)

Major (MCyR)

Complete (CCyR)


1%-35% Ph1 positive metaphases

Combination of PCyR and CCyR (≤35% Ph1 positive metaphases)

No Ph1 positive metaphases detected in 2 successive bone marrow samples after counting 30 metaphases
MOLECULAR

Major (MMR)


Complete (CMR)


3-log reduction in transcript level based on 2-consequtive molecular analyses

2 consecutive samples with no detectable transcripts provided that control gene copy numbers are adequate

Table 1: Definitions for hematological, Cytogenetic and Molecular remission.

  3 months 6 months 12 months 18 months
Response CHR PCyR CCyR MMR
Suboptimal response <CHR <PCyR (≥35% Ph1 positive metaphases) <CCyR <MMR
Failure No hematological response, stable disease, or disease progression <CHR or no CyR (≥95% Ph1 positive metaphases) <PCyR (≥35% Ph1 positive metaphases) <CCyR
Failure At any time: loss of CHR, or loss of CCyR, or Abl kinase Mutation

Table 2: NCCN and European LeukemiaNet criteria for remission, suboptimal response and failure for previously untreated chronic-phase CML patients on standard dose of imatinib.

Qualitative and quantitative PCR

Qualitative testing by PCR analysis of the BCR-ABL transcript is required to establish diagnosis only in the rare situations when interphase FISH is inconclusive due to failure of probe hybridization or sample inadequacy. At the molecular level, the translocation t(9;22)(q34;q22) consistently involves exon 2 of the ABL gene, but occurs in different exons of the BCR gene. The fusion involving the major breakpoint region (MBR) between exons 12 and 13 (or e13a2; formerly called b2a2) or 13 and 14 (or e14a2; formerly called b3a2) leads to expression of a transcript that codes for a 210kD fusion protein (p210BCR-ABL1). The breakpoint in the minor breakpoint region (m- BR) between alternate exon1 and exon 2 (e1a2) results in a smaller transcript coding for a 190 kD protein (p190BCR-ABL1) [22,23]. Most adult CML have the b2a2 or b3a2 transcript (p210BCR-ABL1), whereas the e1a2 (p190BCR-ABL1) is usually present in acute lymphoblastic leukemia and the lymphoid blast crisis of CML. Chronic phase CML with p190 BCR-ABL1 have a myelomonocytic morphology. Less commonly, the break point involves the μ-BCR (micro-BCR) or exon 19(e19a2) resulting in a p230BCR-ABL1 kD product [23]. Co-expression of the p210BCR-ABL1 and p190BCR-ABL1 encoding transcripts can occur as a result of alternative splicing in the M-BR of BCR [24-27]. Quantitative monitoring is the basis of molecular monitoring of CML and is discussed in a later section.

Treatment Strategies

Prior to the advent current therapeutic modalities, the treatment for CML was restricted to lowering the high cell counts by chemotherapy, or managing the symptoms arising from an enlarging spleen with local radiotherapy [28]. Interferon-alpha (IFNa) based regimens were the first to offer the possibility of eliminating the malignant clone, with 20-30% of patients achieving CCyR [29] and were distinctly superior to the primarily leukostatic hydroxyurea [30,31]. However, the benefit with IFNa was mostly in patients with a low Sokal risk score, and that too at the expense of profound toxicity.

Allogeneic hematopoietic stem cell transplant is curative, especially when performed early during the disease course and was very popular in the 1990s. The advantage from HSCT was however restricted to the younger patients with a suitable donor. The outcome in HSCT depends on several factors including the age of the patient, the stage of the disease, time since diagnosis, type of donor, source of stem cells, post-transplant management of infections and graft versus host disease [32]. Younger patients in chronic phase disease with matched related donors have the best outcome [33,34]. Better supportive care has largely mitigated the negative impact of age in the matched related transplant group, but not for those with matched unrelated transplants [35]. With advances in HLA typing and GVHD management, the results for fully matched unrelated transplants are now nearly similar to that for matched related donor transplants [36]. Currently, the survival following HSCT is nearly 80% in chronic phase patients, about 40%-50% in accelerated phase, and 20% in patients who receive it after blast crisis sets in [37]. The therapeutic landscape for CML has undergone a paradigm change with the advent of TK inhibitors. HSCT is no longer the choice for first-line treatment even in younger patients. At this time, HSCT is recommended for patients who develop resistant mutations that fail to respond to second line TK inhibitors as well, or for patients in the advanced stage of disease [37,38].

Imatinib as first-Line therapy

Imatinib was initially approved over a decade ago for treatment of CML patients who had failed prior therapy with IFNa. The results of this Phase I study demonstrated a remarkable response with 53/54 patients achieving complete hematological remission (CHR), and 31% achieving MCyR (major molecular remission) (13% with CCyR) [39]. In the subsequent Phase II study, the efficacy of imatinib was established in patients in all stages of the disease. The response in CP-CML was more impressive (CHR 95%, MCyR 60%, CCyR 41%) than in AP-CML and BP-CML in whom CHR was obtained in 34% and 8% patients respectively [40]. Shortly thereafter, the International Randomized Study of Interferon and STI571 (IRIS) study (Phase III, randomized, double blind crossover study) demonstrated the superiority of imatinib over the IFNa/Ara-C combination in newly diagnosed chronic phase patients [20]. Results of the IRIS study showed 87% MCyR rate in patients on imatinib versus 35% on IFNa/ Ara-C (P < 0.001), and a CCyR of 76% in the imatinib group compared with 15% in the IFNa/Ara-C group (P < 0.001). The progression free survival (PFS) at 18 months was significantly higher in the imatinib group versus IFNa/Ara-C (97% vs. 91.5%; P < 0.001) [20]. Based on the impressive results of the IRIS trial, imatinib received a fast approval by FDA for use as first line therapy of CML patients in May 2001. FDA also approved imatinib for treatment of AP and BP-CML, relapsed CML following HSCT, and Ph1 positive acute lymphoblastic leukemia. Long term follow up of the IRIS trial confirmed that the responses obtained with imatinib were durable over time and clearly improved the overall survival (OS) and PFS in patients with CML [41].

Management of imatinib resistance

Whereas the majority of patients achieve sustained remission with imatinib, an 8-year follow up study showed that it had to be discontinued in 30%-40% of patients because of intolerance or refractoriness to the drug [42]. Resistance to imatinib is more commonly acquired and mostly due to additional genetic abnormalities, or resistant mutations in the ABL kinase domain. Primary resistance is less common and may be due to polymorphisms in drug metabolizing enzymes [43]. Secondary resistance or refractoriness to imatinib is most often due to ABL kinase mutations, from alterations in the intracellular drug availability due to overexpression of the multidrug resistance (MDR1) gene product P-glycoprotein, or decreased expression of the octamer binding protein (OCT1) gene [44].

Increasing the dose of imatinib is a rational first step in countering resistance. The effect of many of the earlier mentioned factors for resistance, including some of the ABL kinase mutations (e.g., M351T, V299L, L384M, G398R, M244V, M244I, Q252H, E355G, L387M), can be overcome by escalation of the dose from the standard 400 mg daily to 600 mg or even 800 mg per day. However, in these dose-escalation trials only a small number of patients achieved CCyR, which is predictive of a sustained favorable outcome [45-47]. The T315I is a highly resistant mutation that fails to respond to imatinib dose escalation as well as to second generation TKIs, as discussed further.

Second-generation TK inhibitors

Second line TK inhibitors- dasatinib and nilotinib are effective in patients who fail to respond to dose escalation or develop adverse effects to it. Dasatinib and nilotinib are structurally different from imatinib, hence are able to overcome the resistance induced as a result of conformational change in the molecule by ABL1 kinase mutations. In addition, dasatinib and nilotinib do not require the OCT1 drug transport protein and are not affected by a decrease in its level [48,49]. Dasatinib is several hundred times more potent than imatinib and is also effective against the Src Family Kinases. Nilotinib is 30 times more effective than imatinib against BCR-ABL1 kinase. Both dasatinib and nilotinb can counter the negative effect of many of the ABL kinase mutations including several that do not respond to escalation of imatinib dose (e.g., G250E, E255K, E255V, F486S, Y253H, Y253F, F317L, F311L) [50]. Clinical trials using dasatinib and nilotinib have demonstrated durable overall response rates, improved PFS rates, and tolerability of these second line TK inhibitors in patients with imatinib resistant or intolerant CML in CP, AP, and BP-CML resulting in FDA-approval for use of dasatinib and nilotinib in treatment of patients resistant or intolerant CML in all stages of the disease [51-54]. The choice between dasatinib and nilotinib is generally based on the adverse effect profile of these drugs in the context of a specific patient, and the nature of the specific mutations [55]. Secondary resistance from additional mutations may develop under drug pressure from second line TK inhibitors [56,57]. Mutations such as Q252H, E255K/V, V299L, F317L, and T315A are more likely to develop resistance to dasatinib, whereas others including Y253H, F359C/V, and E255K/V to nilotinib. The little overlap between mutations insensitive to both drugs allows, replacing one by the other to restore effectiveness against the mutation. The highly resistant T315I mutation however fails to respond to both dasatinib and nilotinib, requiring recourse to allogenic BMT or other alternative therapies for subsequent management [58].

Third -generation TKI

A future option may be a third-generation TK inhibitor ponatinib. Pontinib is an oral pan-BCR-ABL inhibitor developed to specifically counter the steric hindrance induced by the T315I mutation. Ponatinib overlaps the gatekeeper region of the ATP-binding site, and has an extensive network of high-potency hydrogen and triple carbon bonds that form multiple high-affinity contact points preventing destabilization of the ABL-kinase domain by the mutation [59]. It is effective against several common BCR-ABL1 mutations as well. In a cell-based mutagenesis screen, Ponatinib in therapeutic doses was able to inhibit growth of resistant BCR-ABL1 mutations, offering the possibility that it may prevent emergence of resistant mutations [60]. Initial results of a 3-month evaluation in the pivotal phase II trial of ponatinib (AP24534) in patients with refractory CML show MCyR in 57% of the 23 patients with the T315I mutation, of whom 11 achieved CCyR [59].

Second-generation TK Inhibitors as frontline therapy for CML

Results of recent clinical trials comparing the efficacy of dasatinib and nilotinib with standard dose imatinib in the first-line setting in CP-CML clearly demonstrate significantly higher rates and depth for achieving CCyR and MMR with both nilotinib and dasatinib compared with standard dose imatinib, with lesser rates of progression to accelerated phase or blast crisis, and with similar safety profiles [61,62]. Compared with imatinib, the time to attain CCyR and MMR was significantly shorter with dasatinib (p < 0.0001) [62]. FDA has approved both dasatinib and nilotinib for front line treatment of CPCML.

Overall, for most patients, the treatment options for CML are rendering it more of a chronic disease with the median survival estimated at greater than 20 years [63]. With improved overall survival for most patients, quality of life issues require more attention. In addition to common class-related adverse effects, each TK kinase inhibitor is characterized by a unique side effect profile [64]. The importance of being cognizant of these adverse effects and taking them along with the patient-specific comorbidities into account while tailoring the TK inhibitor therapy cannot be overemphasized. Early management of adverse effects is important in preventing non-compliance and to ensure maintenance of remission status in patients [65].

Molecular monitoring of CML

With the current molecularly targeted therapy for CML the majority of patients in chronic phase achieve CCyR at the end of one year [20,66]. At the time of CCyR the tumor burden in the bone marrow may be as high as 109 leukemic cells. In the IRIS study, quantitative real time PCR testing for the BCR-ABL transcript was performed in all patients who achieved CCyR [66]. Results of the study showed that the likelihood of progression free state at 24 months was 100% in patients who additionally achieved a 3-log reduction in the BCR-ABL transcript at 12 months, compared with 95% in those who did not achieve 3-log reduction (P < 0.001) [66]. These results made it incumbent on laboratories to develop more sensitive, reproducible, and standardized methods for subsequent monitoring of the disease. Quantitative reverse transcriptase PCR (RQ-PCR) performed on real time platforms can detect presence of abnormal transcripts with a sensitivity of 1 in 104 or 1 in 105 cells, and is currently the most sensitive laboratory technique for detection of the BCR-ABL1 transcript [67-70]. RQ-PCR is particularly useful for subsequent monitoring of patients who have achieved CCyR. The assay is equally sensitive on peripheral blood and bone marrow samples and either specimen can be used for the assay, as long as the follow up is done using the same sample type for comparable results [71]. The levels of BCR-ABL transcript in the peripheral blood by RQ-PCR show excellent congruity with those of metaphase cytogenetics [71]. Quantitative reverse-transcriptase PCR (RQ-PCR) involves extraction of total RNA from the peripheral blood or bone marrow specimen, reverse-transcription of the mRNA so obtained into cDNA, and quantitative (real time) co-amplification of the target BCR-ABL cDNA and cDNA of an internal control gene (to control for RNA integrity, sample preparation, extraction and loading). Standard curves are constructed by serial dilutions of known amount of cloned plasmid containing the fusion DNA, or from serial dilutions of K562 cells in normal DNA. The value of BCR-ABL transcript extrapolated from the standard curve is expressed as a normalized ratio of the BCRABL transcript to the control gene transcript [72,73]. Despite the fact that RQ-PCR is currently performed by most laboratories nation-wide, there is a lack of uniformity in the way the results are obtained and the data expressed. This inherent variability is due to several factors that affect the performance of RQ-PCR including methods of specimen transport, storage, RNA extraction procedures, reverse transcription and PCR efficiency, and the type of real-time platform used. The use of different control genes by the laboratories can also significantly alter the BCR-ABL/control gene ratios. To develop international treatment guidelines it is essential that the molecular end-points for therapy protocols be comparable. For harmonizing the molecular monitoring of CML by RQ-PCR, recommendations were made at the CML meeting at the National Institutes of Health in Bethesda in 2005 to establish guidelines for specimen collection and transport, appropriate RNA quality, methodology for reverse transcription, PCR amplification efficiency, use of suitable control genes, test sensitivity for reporting negative results, quality assurance of the assay, generation of international reference, and a standardized method for expressing the result on an international scale [71]. To achieve this objective, a proposal was made to develop laboratory-specific conversion factors to convert the results obtained locally to a standardized international scale (expressed as % or IS units) [66]. The international scale was derived from the IRIS study in which the baseline median transcript level in 30 pre-treatment patient samples by RQ-PCR performed independently in the three principal laboratories was defined as 100% BCR-ABL level. Using this international scale, 1% BCR-ABL level correlated with the state of complete cytogenetic remission, and 1 log below this level (0.1% BCR-ABL) was defined as major molecular remission (MMR). In the IRIS study, no patient in MMR had positive Ph1 on conventional karyotyping [71]. According to the current guidelines laid by the NCCN and the European Leukemia Net [18,19], optimum response to imatinib therapy is indicated by attainment of MMR by 18 months of start of therapy. The NCCN and ELN criteria for remission, suboptimal response and failure for previously untreated patients on standard dose of imatinib are shown in (Table 3). Data from several recent studies have shown that time to response is important and the probability of a favorable long-term outcome is best for patients who have the best responses at 3 months after the start of therapy [74-76]. Using receiveroperated characteristic curves, [76] authors have confirmed that the BCR-ABL1 transcript ratio of less than 9.84% at 3 months correlates with best outcome, and compared with current NCCN and ELN cytogenetic milestones (Table 2), molecular determination of transcript levels at 6 and 12 months levels was the only independent of overall survival (p < 0.001 for both time points). These data have not yet been translated into guidelines for testing. It is clear that the success of these predictive time points depends on standardization of methodology for determining the BCR-ABL1 transcript and subsequent reporting of the results. The first World Health Organization International Genetic Reference Panel for quantitation of BCR-ABL mRNA was approved by the Expert Committee on Biological Standardization of the World Health Organization in November 2009 and is now available commercially [77,78]. It consists of 4 dilutions of lyophillized preparations of K562 cells with assigned, fixed BCR-ABL/control gene values according to the IS. This development is a major milestone towards standardized molecular monitoring of therapeutic response to tyrosine kinase inhibitors in CML.

Test Time Point
A. Metaphase cytogenetics B. FISH (peripheral blood sample is acceptable if bone marrow specimen not available) using dual color, dual fusion probes and standard technique 1. At diagnosis and 6, 12, and 18 months until CCyR achieved. If CCyR achieved at an earlier time point, then there is no need to perform metaphase cytogenetics in a stable patients.
2. Rising level of BCR-ABL1 transcript (1 log increase) without achieving MMR
QRT-PCR for BCR-ABL transcript 1. At diagnosis to establish baseline transcript level.
2. Every 3 months thereafter until patient is responding to TK inhibitors, and every 3-6 months after achieving CCyR
3. If level of BCR-ABL1 transcript is rising (1 log increase) after achieving MMR, then QRT-PCR should be performed every 1-3 months
Kinase domain mutation testing 1. At time of suboptimal response or failure
2. Before switching to another TK inhibitor

Table 3: Recommendations of NCCN and European LeukemiaNet for monitoring of CML patients on Tyrosine kinase inhibitors.

Mechanism of resistance to imatinib and molecular testing for ABL kinase inhibitors

The majority of CML patients in chronic phase achieve an optimum response with the standard recommended dose of imatinib. A minority (20%), patients develop primary resistance to imatinib, defined by failure to achieve the different levels of response i.e. hematological, cytogenetic, or molecular, at the expected time points [19]. The resistance is ‘acquired’ in the majority of patients, with loss of a previously obtained hematological or cytogenetic remission, or loss of MMR fifty- ninety percent of the acquired resistance in CML is due to BCR-ABL-kinase domain mutations. According to a recent publication from 2011, nearly 100 different ABL kinase mutations have been identified [79]. The mutations involve the catalytic domain, the highly conserved p-loop, or the activation loop, with two different mechanisms of causing resistance [58,80,81]: direct interference in binding of the drug (eg, T315I, F317L, and F359C/V), or by a causing a conformational change in the ABL kinase ATP-binding site that prevents access to the drug (eg, G250E, Q252H, Y253H, E255K/V) [80]. The mutations have different level of sensitivity or resistance to the different TK inhibitors based on the in vitro IC50 determinations. Mutations resistant to imatinib may be sensitive to second-generation TK inhibitors, underscoring the importance of mutation testing. The T315I mutation present in 4-15% of imatinib-resistant patients however, is resistant to imatinib as well as the second-generation TK inhibitors dasatinib and nilotinib [58]. Other mechanisms of acquired resistance include increased dosage of the BCR-ABL1 transcript by amplification or additional copies of the Ph1 chromosome, less commonly by involvement of different pathways such as the SRK kinase pathway that are not inhibited by imatinib, or drug efflux mechanisms that decrease the effective concentration of the drug within the cells [82,83].

The Pertinent Questions Regarding Mutation Testing for BCR-ABL1 Are Discussed Below

When should the testing be done and who should be tested?

Treatment naive patients: There are a few reports of ABL1 kinase mutations present in newly diagnosed treatment naïve patients. Most of them are in patients who present in accelerated phase or blast crisis at diagnosis [84-87]; they are rare in chronic phase treatment naïve patients [88,89]. Given the rare occurrence, and lack of any clinical evidence of an adverse outcome from mutations in chronic phase patients, routine testing of all chronic-phase patients for mutations is not recommended at this time. The present recommendation to perform BCR-ABL (kinase domain) KD mutations in treatment naïve patients is restricted to those who present with advanced disease in accelerated phase or blast crisis [87].

In patients on imatinib ss first line therapy: Per the NCCN practice guidelines [18] and ELN [19], testing for BCR-ABL KD mutations in patients on imatinib as first line therapy is to be performed in the following circumstances: 1) Failure to respond or suboptimal response at the defined time points i.e. complete hematological remission at 3 months, minor cytogenetic remission at 6 months, and CCyR at 12 months of initiation of imatinib therapy 1 [8,19]. Twenty-nine percent of chronic phase non-responders and 16% of patients with suboptimal response harbor BCR-ABL KD mutations [90-92]. 2) Confirmed loss of previously attained optimum response demonstrated by loss of MMR or disease progression [86]. Fluctuations in levels of BCRABL transcript may occur when the levels are very low and are not considered clinically significant. A trend of increasing BCR-ABL transcript level by analysis of sequential samples, or a two-fold (or one log) increase in BCR-ABL transcript level is more consistent with definitive loss of MMR and predictive of relapse [93].

Mutation analysis in patients on second-line TKIs: KD mutations arise under selective drug pressure. Thus, it is not unexpected to see emergence of additional resistant mutations in patients who are put on second-generation TK inhibitors following resistance to imatinib [94]. Mutations resistant to dasatinib (V299L, T315A, F317L/V/I/C) or to nilotinib (E255K/V, Y253H, F359V/C/I) have been described in patients developing secondary response failure with second-generation TK inhibitors [95,96]. The data on resistant mutations to the second generation TKIs is limited; currently KD mutation testing is done in event of failure to respond, or suboptimal response, similar to that for imatinib. Preliminary data show a better projected one-year survival in patients receiving dasatinib as second or subsequent-line therapy if they attain MCyR at 12 months, and suggest alternative therapies if there is no cytogenetic response in 3 to 6 months [97,98].

Methods for Mutation Analysis

Many different approaches can be applied for mutation detection with sensitivities varying from 20% (one mutant allele in a background of 5 normal alleles) by Sanger sequencing to 0.01%-0.1% by allelespecific PCR(AS-PCR) [71-79,99-101]. Despite the low sensitivity of Sanger sequencing, it is the most commonly used method because of its availability in most molecular laboratories, and its ability to detect a large number of mutations in the activation-loop, the P-loop, as well as the catalytic loop [71-79,99,100]. Bidirectional sequencing is recommended to avoid false positive calls [102]. A two-step process of initial screening by DHPLC (Denaturing High Performance Lliquid Chromatography) increases the sensitivity of subsequent sequencing [99-101]. The threshold of detection at 20% is acceptable since the clinical significance of detecting very low-level mutations is unclear [101]. According to a recent study, sensitive detection methods such as mass-spectrometry can predict a sub group of poor-responders to dasatinib/nilotinib at the time of switchover, by detecting multiple low-level mutations that are not detected by routine direct sequencing method [103]. The clinical validity of these results is yet to be seen in large multicenter studies. The sensitivity of detection will be greatly increased when the high-throughput deep sequencing technology is established in the clinical laboratory.

Reporting of Results and Interpretation

The purpose of mutation detection is to guide further management using one of the second or third-generation TK inhibitors. The report should indicate the specific mutation detected, the location in the specific region of the ABL kinase genome, the nucleotide change, and the resultant amino acid change. In addition, the report should include the specific BCR-ABL isoform-p190, p210 or rarely p230, and the RQPCR results leading to the testing. Several laboratories have developed large databases that include a list of mutations reported previously and the sensitivity to the different TK inhibitors (IC50). Soverini et al. [96] provide a meta-analysis of published IC50 values of imatinib, dasatinib, and nilotinib for the most commonly occurring mutations as compared with the IC50 for the wild type BCR-ABL. A reference for these databases is useful for making clinical decisions for second-line TKIs, and is a useful addition to the report.

Current Consideration

The durable remissions now possible with TK inhibitors enable us to ask the pertinent question whether therapy can be stopped in patients in stable remission. This question has been addressed by at least two studies with similar conclusions that at least 40% of patients who have been in complete molecular remission (CMR: level of BCR-ABL1 transcript 1-2 logs below the detection limit of the current sensitive qRT-PCR method of detection) for at least 2 years, will remain in CMR for 2 years after discontinuing imatinib [103,104]. Longer-term data are not yet available. Early molecular relapse in 60% of these patients with stable CMR, relapses occurring after nearly a decade in patients who underwent allogenic stem cell transplant [105] clearly indicate persistence of leukemic cells at a very low level. Indeed, the unique genomic BCR-ABL1 fusion of the original leukemic clone could be detected by patient-specific, highly sensitive, DNA quantitative PCR in patients who were transcript negative and in ‘functional remission’ [106,107]. The factors determining sustained remission, or relapse in a patient after stopping TK inhibitor therapy are presently unclear. It is believed that low level persistence of leukemic stem cells that are independent of BCR-ABL1 for their survival, and hence immune to the TK inhibitors, are responsible for subsequent relapses [108], implying that ‘cure’ in CML is dependent on eradication of this resistant leukemic stem cell clone. Three hypothetical models of operational cure have been proposed that include depletion of the stem cell clone by prolonged imatinib therapy in combination with therapies that target pathways other than those mediated by BCR-ABL1, exhaustion of the stem cell from asymmetric division by depleting the mutable mature leukemic clone that replenishes the self renewing cells, and third by boosting the host T- cell dependent cancer surveillance by immunomodulation to maintain the initially established remission [103].

In summary, the current management of CML patients is able to provide sustained CCyR and MMR in a significant number of patients. The goals are to maintain the remission status in these patients, recognize causes of relapse in the remaining subset of patients, and eventually gear towards a ‘cure’ for this disease. An approach to achieve PCyR or reducing the BCR-ABL transcript to 10% within 3 months by newer second-generation TK inhibitors, assiduous monitoring for early detection of resistance by standardized sensitive methodology, judicious use of alternate TK inhibitors and HSCT to counter resistant mutations, and ensuring compliance by management of adverse effects is required to meet these goals.

References

  1. NOWELL PC, HUNGERFORD DA (1960) Chromosome studies on normal and leukemic human leukocytes. J Natl Cancer Inst 25: 85-109.
  2. Rowley JD (1973) Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 243: 290-293.
  3. de Klein A, van Kessel AG, Grosveld G, Bartram CR, Hagemeijer A, et al. (1982) A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukaemia. Nature 300: 765-767.
  4. O'Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, et al. (2003) Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 348: 994-1004.
  5. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA,et al. (2008). Pathology and genetics of tumours of haematopoietic and lymphoid tissues. World health organization classification of tumours. 4th ed. Lyon, France: IARC Press.
  6. Anastasi J, Feng J, Dickstein JI, Le Beau MM, Rubin CM, et al. (1996) Lineage involvement by BCR/ABL in Ph+ lymphoblastic leukemias: chronic myelogenous leukemia presenting in lymphoid blast vs Ph+ acute lymphoblastic leukemia. Leukemia 10: 795-802.
  7. Khalidi HS, Brynes RK, Medeiros LJ, Chang KL, Slovak ML, et al. (1998) The immunophenotype of blast transformation of chronic myelogenous leukemia: a high frequency of mixed lineage phenotype in "lymphoid" blasts and A comparison of morphologic, immunophenotypic, and molecular findings. Mod Pathol 11: 1211-1221.
  8. Daley GQ, Van Etten RA, Baltimore D (1990) Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science 247: 824-830.
  9. Antonio Valeri A, Alonso-Ferrero ME, Río P, Roser Pujol M, Casado JA,et al. (2010) Jacome A, Agirre X, José Calasanz M, Hanenberg H, et al. Bcr/Abl interferes with the fanconi Anemia/BRCA pathway: Implications in the chromosomal instability of chronic myeloid leukemia cells. PLoS One 5: e15525.
  10. Wang YL, Bagg A, Pear W, Nowell PC, Hess JL (2001) Chronic myelogenous leukemia: laboratory diagnosis and monitoring. Genes Chromosomes Cancer 32: 97-111.
  11. Albano F, Anelli L, Zagaria A, Coccaro N, Casieri P, et al. (2010) Non random distribution of genomic features in breakpoint regions involved in chronic myeloid leukemia cases with variant t(9;22) or additional chromosomal rearrangements. Mol Cancer 9: 120.
  12. De Melo VA, Milojkovic D, Marin D, Apperley JF, Nacheva EP, et al. (2008) Deletions adjacent to BCR and ABL1 breakpoints occur in a substantial minority of chronic myeloid leukemia patients with masked Philadelphia rearrangements. Cancer Genet Cytogenet 182: 111-115.
  13. Huntly BJ, Bench A, Green AR (2003) Double jeopardy from a single translocation: deletions of the derivative chromosome 9 in chronic myeloid leukemia. Blood 102: 1160-1168.
  14. Lee YK, Kim YR, Min HC, Oh BR, Kim TY, et al. (2006) Deletion of any part of the BCR or ABL gene on the derivative chromosome 9 is a poor prognostic marker in chronic myelogenous leukemia. Cancer Genet Cytogenet 166: 65-73.
  15. Quintás-Cardama A, Kantarjian H, Shan J, Jabbour E, Abruzzo LV, et al. (2011) Prognostic impact of deletions of derivative chromosome 9 in patients with chronic myelogenous leukemia treated with nilotinib or dasatinib. Cancer 117: 5085-5093.
  16. Kantarjian H, Schiffer C, Jones D, Cortes J (2008) Monitoring the response and course of chronic myeloid leukemia in the modern era of BCR-ABL tyrosine kinase inhibitors: Practical advice on the use and interpretation of monitoring methods. Blood 111: 1774-17780.
  17. NCCN GuidelinesTM Version 2.2012. Chronic Myelogenous Leukemia [Internet]: NCCN; c2012. Available from: http://guidelines.nccn.org/epc-guideline/guideline/id/EBA4F5EF-5A9B-E0B4-C6A4-BFDD8FA8BF78?jumpTo=false#.
  18. Baccarani M, Castagnetti F, Gugliotta G, Palandri F, Soverini S (2009) Response definitions and European Leukemianet Management recommendations. Best Pract Res Clin Haematol 22: 331-341.
  19. O'Brien SG, Deininger MW (2003) Imatinib in patients with newly diagnosed chronic-phase chronic myeloid leukemia. Semin Hematol 40: 26-30.
  20. Vigil CE, Griffiths EA, Wang ES, Wetzler M (2011) Interpretation of cytogenetic and molecular results in patients treated for CML. Blood Rev 25: 139-146.
  21. Verma D, Kantarjian HM, Jones D, Luthra R, Borthakur G, et al. (2009) Chronic myeloid leukemia (CML) with P190 BCR-ABL: analysis of characteristics, outcomes, and prognostic significance. Blood 114: 2232-2235.
  22. Westbrook CA, Hooberman AL, Spino C, Dodge RK, Larson RA, et al. (1992) Clinical significance of the BCR-ABL fusion gene in adult acute lymphoblastic leukemia: a Cancer and Leukemia Group B Study (8762). Blood 80: 2983-2990.
  23. Melo JV (1997) BCR-ABL gene variants. Baillieres Clin Haematol 10: 203-222.
  24. Kunieda Y, Okabe M, Kurosawa M, Itaya T, Kakinuma M, et al. (1994) Chronic myeloid leukemia presenting ALL-type BCR/ABL transcript. Ann Hematol 69: 189-193.
  25. Lichty BD, Keating A, Callum J, Yee K, Croxford R, et al. (1998) Expression of p210 and p190 BCR-ABL due to alternative splicing in chronic myelogenous leukaemia. Br J Haematol 103: 711-715.
  26. van Rhee F, Hochhaus A, Lin F, Melo JV, Goldman JM, et al. (1996) p190 BCR-ABL mRNA is expressed at low levels in p210-positive chronic myeloid and acute lymphoblastic leukemias. Blood 87: 5213-5217
  27. Goldman JM (2010) Chronic myeloid leukemia: a historical perspective. Semin Hematol 47: 302-311.
  28. Kantarjian HM, O'Brien S, Cortes JE, Shan J, Giles FJ, Rios,et al. (2003) Complete cytogenetic and molecular responses to interferon-alpha-based therapy for chronic myelogenous leukemia are associated with excellent long-term prognosis. Cancer 97: 1033-1041.
  29. [No authors listed] (1997) Interferon alfa versus chemotherapy for chronic myeloid leukemia: a meta-analysis of seven randomized trials: Chronic Myeloid Leukemia Trialists' Collaborative Group. J Natl Cancer Inst 89: 1616-1620.
  30. Hehlmann R, Heimpel H (1996) Current aspects of drug therapy in Philadelphia-positive CML: correlation of tumor burden with survival. Leuk Lymphoma 1: 161-167.
  31. Gratwohl A, Hermans J, Goldman JM, Arcese W, Carreras E, et al. (1998) Risk assessment for patients with chronic myeloid leukaemia before allogeneic blood or marrow transplantation. Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Lancet 352: 1087-1092.
  32. Gratwohl A, Stern M, Brand R, Apperley J, Baldomero H, et al. (2009) Risk score for outcome after allogeneic hematopoietic stem cell transplantation: a retrospective analysis. Cancer 115: 4715-4726.
  33. Clift RA, Buckner CD, Thomas ED, Bryant E, Anasetti C, et al. (1994) Marrow transplantation for patients in accelerated phase of chronic myeloid leukemia. Blood 84: 4368-4373.
  34. Hansen JA, Gooley TA, Martin PJ, Appelbaum F, Chauncey TR, et al. (1998) Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia. N Engl J Med 338: 962-968.
  35. Davies SM, DeFor TE, McGlave PB, Miller JS, Verfaillie CM, et al. (2001) Equivalent outcomes in patients with chronic myelogenous leukemia after early transplantation of phenotypically matched bone marrow from related or unrelated donors. Am J Med 110: 339-346.
  36. Jain N, van Besien K (2011) Chronic myelogenous leukemia: role of stem cell transplant in the imatinib era. Hematol Oncol Clin North Am 25: 1025-1048, vi.
  37. Heim D, Gratwohl A (2008) Role of allogeneic transplantation in chronic myeloid leukemia. Expert Rev Hematol 1: 41-50.
  38. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, et al. (2001) Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med 344: 1031-1037.
  39. Talpaz M, Silver RT, Druker BJ, Goldman JM, Gambacorti-Passerini C, et al. (2002) Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study. Blood 99: 1928-1937.
  40. Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, et al. (2006) Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med 355: 2408-2417.
  41. Deininger M, O'Brien SG, Guilhot F, Goldman JMH, Hughes TP,et el. (2009) International randomized study of interferon vs STI571 (IRIS) 8-year follow up: Sustained survival and low risk for progression or events in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) treated with imatinib. ASH annual meeting abstracts. Blood 114: 1126.
  42. Zhang WW, Cortes JE, Yao H, Zhang L, Reddy NG, et al. (2009) Predictors of primary imatinib resistance in chronic myelogenous leukemia are distinct from those in secondary imatinib resistance. J Clin Oncol 27: 3642-3649.
  43. Thomas J, Wang L, Clark RE, Pirmohamed M (2004) Active transport of imatinib into and out of cells: implications for drug resistance. Blood 104: 3739-3745.
  44. Breccia M, Stagno F, Vigneri P, Latagliata R, Cannella L, et al. (2010) Imatinib dose escalation in 74 failure or suboptimal response chronic myeloid leukaemia patients at 3-year follow-up. Am J Hematol 85: 375-377.
  45. Jabbour E, Kantarjian HM, Jones D, Shan J, O'Brien S, et al. (2009) Imatinib mesylate dose escalation is associated with durable responses in patients with chronic myeloid leukemia after cytogenetic failure on standard-dose imatinib therapy. Blood 113: 2154-2160.
  46. Marin D, Goldman JM, Olavarria E, Apperley JF (2003) Transient benefit only from increasing the imatinib dose in CML patients who do not achieve complete cytogenetic remissions on conventional doses. Blood 102: 2702-2703.
  47. Davies A, Jordanides NE, Giannoudis A, Lucas CM, Hatziieremia S, et al. (2009) Nilotinib concentration in cell lines and primary CD34(+) chronic myeloid leukemia cells is not mediated by active uptake or efflux by major drug transporters. Leukemia 23: 1999-2006.
  48. Giannoudis A, Davies A, Lucas CM, Harris RJ, Pirmohamed M, et al. (2008) Effective dasatinib uptake may occur without human organic cation transporter 1 (hOCT1): implications for the treatment of imatinib-resistant chronic myeloid leukemia. Blood 112: 3348-3354.
  49. Laneuville P, Dilea C, Yin OQ, Woodman RC, Mestan J, et al. (2010) Comparative in vitro cellular data alone are insufficient to predict clinical responses and guide the choice of BCR-ABL inhibitor for treating imatinib-resistant chronic myeloid leukemia. J Clin Oncol 28: e169-e171.
  50. Apperley JF, Cortes JE, Kim DW, Roy L, Roboz GJ, et al. (2009) Dasatinib in the treatment of chronic myeloid leukemia in accelerated phase after imatinib failure: the START a trial. J Clin Oncol 27: 3472-3479.
  51. Hochhaus A, Kantarjian HM, Baccarani M, Lipton JH, Apperley JF, et al. (2007) Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy. Blood 109: 2303-2309.
  52. Kantarjian H, Pasquini R, Lévy V, Jootar S, Holowiecki J, et al. (2009) Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily: two-year follow-up of a randomized phase 2 study (START-R). Cancer 115: 4136-4147.
  53. Kantarjian HM, Giles F, Gattermann N, Bhalla K, Alimena G,et al. (20070 Palandri F, Ossenkoppele GJ, Nicolini FE, O'Brien SG, Litzow M, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood 110: 3540-3546.
  54. Jabbour E, Branford S, Saglio G, Jones D, Cortes JE, et al. (2011) Practical advice for determining the role of BCR-ABL mutations in guiding tyrosine kinase inhibitor therapy in patients with chronic myeloid leukemia. Cancer 117: 1800-1811.
  55. Cortes J, Jabbour E, Kantarjian H, Yin CC, Shan J, et al. (2007) Dynamics of BCR-ABL kinase domain mutations in chronic myeloid leukemia after sequential treatment with multiple tyrosine kinase inhibitors. Blood 110: 4005-4011.
  56. Shah NP, Skaggs BJ, Branford S, Hughes TP, Nicoll JM, et al. (2007) Sequential ABL kinase inhibitor therapy selects for compound drug-resistant BCR-ABL mutations with altered oncogenic potency. J Clin Invest 117: 2562-2569.
  57. O'Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, et al. (2009) AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 16: 401-412.
  58. Soverini S, Colarossi S, Gnani A, Castagnetti F, Rosti G, et al. (2007) Resistance to dasatinib in Philadelphia-positive leukemia patients and the presence or the selection of mutations at residues 315 and 317 in the BCR-ABL kinase domain. Haematologica 92: 401-404.
  59. Initial findings from the PACE trial: A pivotal phase 2 study of ponatinib in patients with CML and Ph+ALL resistant or intolerant to dasatinib or nilotinib, or with the T315I mutation.
  60. Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, et al. (2010) Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med 362: 2251-2259.
  61. Kantarjian H, Shah NP, Hochhaus A, Cortes J, Shah S,et al. (2010) Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 362: 2260-2270.
  62. Hochhaus A (2011) Educational session: managing chronic myeloid leukemia as a chronic disease. Hematology Am Soc Hematol Educ Program 2011: 128-135.
  63. Jabbour E, Deininger M, Hochhaus A (2011) Management of adverse events associated with tyrosine kinase inhibitors in the treatment of chronic myeloid leukemia. Leukemia 25: 201-210.
  64. Ibrahim AR, Eliasson L, Apperley JF, Milojkovic D, Bua M, et al. (2011) Poor adherence is the main reason for loss of CCyR and imatinib failure for chronic myeloid leukemia patients on long-term therapy. Blood 117: 3733-3736.
  65. Hughes TP, Kaeda J, Branford S, Rudzki Z, Hochhaus A, et al. (2003) Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia. N Engl J Med 349: 1423-1432.
  66. Branford S, Fletcher L, Cross NC, Muller MC, Hochhaus A,et al. (2008) Desirable performance characteristics for BCR-ABL measurement on an international reporting scale to allow consistent interpretation of individual patient response and comparison of response rates between clinical trials. Blood 112: 3330-3338.
  67. Branford S, Prime J (2011) Chronic myelogenous leukemia: monitoring response to therapy. Curr Hematol Malig Rep 6: 75-81.
  68. Breccia M, Alimena G (2011) The significance of early, major and stable molecular responses in chronic myeloid leukemia in the imatinib era. Crit Rev Oncol Hematol 79: 135-143.
  69. Cortes J, Quintás-Cardama A, Kantarjian HM (2011) Monitoring molecular response in chronic myeloid leukemia. Cancer 117: 1113-1122.
  70. Hughes T, Deininger M, Hochhaus A, Branford S, Radich J,et al. (2006) 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 108: 28-37.
  71. Fan H, Robetorye RS (2010) Real-time quantitative reverse transcriptase polymerase chain reaction. Methods Mol Biol 630: 199-213.
  72. Gerrard G, Mudge K, Stewart R, Foskett P, Stevens D, et al. (2011) Duplex quantitative PCR for molecular monitoring of BCR-ABL1-associated hematological malignancies. Am J Hematol 86: 313-315.
  73. Hughes T, Branford S (2006) Molecular monitoring of BCR-ABL as a guide to clinical management in chronic myeloid leukaemia. Blood Rev 20: 29-41.
  74. Quintás-Cardama A, Kantarjian H, Jones D, Shan J, Borthakur G, et al. (2009) Delayed achievement of cytogenetic and molecular response is associated with increased risk of progression among patients with chronic myeloid leukemia in early chronic phase receiving high-dose or standard-dose imatinib therapy. Blood 113: 6315-6321.
  75. Marin D, Ibrahim AR, Lucas C, Gerrard G, Wang L, Szydlo RM, Clark RE, Apperley JF, Milojkovic D, Bua M, et al. Assessment of BCR-ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. J Clin Oncol 2012 Jan 20;30(3):232-8.
  76. White HE, Matejtschuk P, Rigsby P, Gabert J, Lin F, et al. (2010) Establishment of the first World Health Organization International Genetic Reference Panel for quantitation of BCR-ABL mRNA. Blood 116: e111-117.
  77. Genetic Reference Panel, for the quantitation of BCR-ABL translocation by RQ-PCR (1st I.S.) [Internet]. Available from: http://www.nibsc.ac.uk/products/biological_reference_materials /product_catalogue/ detail_page.aspx?CatId=09/138.
  78. Branford S, Hughes TP (2011) Mutational analysis in chronic myeloid leukemia: when and what to do? Curr Opin Hematol 18: 111-116.
  79. Shah NP (2005) Loss of response to imatinib: mechanisms and management. Hematology Am Soc Hematol Educ Program .
  80. Azam M, Latek RR, Daley GQ (2003) Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell 112: 831-843.
  81. Deininger MW (2008) Milestones and monitoring in patients with CML treated with imatinib. Hematology Am Soc Hematol Educ Program 419-426.
  82. La Rosée P, Deininger MW (2010) Resistance to imatinib: mutations and beyond. Semin Hematol 47: 335-343.
  83. Ernst T, Erben P, Müller MC, Paschka P, Schenk T, et al. (2008) Dynamics of BCR-ABL mutated clones prior to hematologic or cytogenetic resistance to imatinib. Haematologica 93: 186-192.
  84. Kuila N, Dash N, Sahoo DP, Pattnayak NC, Biswas G, et al. (2009) Presence of a new BCR-ABL kinase domain mutation, C330G in an imatinib naive patient with chronic myeloid leukemia: very low prevalence of BCR-ABL kinase domain mutations in patients with chronic myeloid leukemia from eastern India. Leuk Lymphoma 50: 663-666.
  85. Soverini S, Hochhaus A, Nicolini FE, Gruber F, Lange T, et al. (2011) BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 118: 1208-1215.
  86. Willis SG, Lange T, Demehri S, Otto S, Crossman L, et al. (2005) High-sensitivity detection of BCR-ABL kinase domain mutations in imatinib-naive patients: correlation with clonal cytogenetic evolution but not response to therapy. Blood 106: 2128-2137.
  87. Khorashad JS, de Lavallade H, Apperley JF, Milojkovic D, Reid AG, et al. (2008) 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 26: 4806-4813.
  88. Carella AM, Garuti A, Cirmena G, Catania G, Rocco I, et al. (2010) Kinase domain mutations of BCR-ABL identified at diagnosis before imatinib-based therapy are associated with progression in patients with high Sokal risk chronic phase chronic myeloid leukemia. Leuk Lymphoma 51: 275-278.
  89. Soverini S, Colarossi S, Gnani A, Rosti G, Castagnetti F, et al. (2006) 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 12: 7374-7379.
  90. Breccia M, Orlandi SM, Latagliata R, Grammatico S, Diverio D, et al. (2011) Suboptimal response to imatinib according to 2006-2009 European LeukaemiaNet criteria: a 'grey zone' at 3, 6 and 12 months identifies chronic myeloid leukaemia patients who need early intervention. Br J Haematol 152: 119-121.
  91. Marin D, Milojkovic D, Olavarria E, Khorashad JS, de Lavallade H, et al. (2008) 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 112: 4437-4444.
  92. Press RD, Willis SG, Laudadio J, Mauro MJ, Deininger MW (2009) Determining the rise in BCR-ABL RNA that optimally predicts a kinase domain mutation in patients with chronic myeloid leukemia on imatinib. Blood 114: 2598-2605.
  93. Breccia M, Frustaci AM, Cannella L, Soverini S, Stefanizzi C, et al. (2009) Sequential development of mutant clones in an imatinib resistant chronic myeloid leukaemia patient following sequential treatment with multiple tyrosine kinase inhibitors: an emerging problem? Cancer Chemother Pharmacol 64: 195-197.
  94. Soverini S, Gnani A, Colarossi S, Castagnetti F, Abruzzese E, et al. (2009) Philadelphia-positive patients who already harbor imatinib-resistant Bcr-Abl kinase domain mutations have a higher likelihood of developing additional mutations associated with resistance to second- or third-line tyrosine kinase inhibitors. Blood 114: 2168-2171.
  95. Soverini S, Rosti G, Iacobucci I, Baccarani M, Martinelli G (2011) Choosing the best second-line tyrosine kinase inhibitor in imatinib-resistant chronic mveloid leukemia patients harboring Bcr-Abl kinase domain mutations: how reliable is the IC50? 16: 868-876 .
  96. Milojkovic D, Nicholson E, Apperley JF, Holyoake TL, Shepherd P, et al. (2010) Early prediction of success or failure of treatment with second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukemia. Haematologica 95: 224-231.
  97. Tam CS, Kantarjian H, Garcia-Manero G, Borthakur G, O'Brien S, et al. (2008) Failure to achieve a major cytogenetic response by 12 months defines inadequate response in patients receiving nilotinib or dasatinib as second or subsequent line therapy for chronic myeloid leukemia. Blood 112: 516-518.
  98. Alikian M, Gerrard G, Subramanian PG, Mudge K, Foskett P, et al. (2012) BCR-ABL1 kinase domain mutations: methodology and clinical evaluation. Am J Hematol 87: 298-304.
  99. Branford S, Hughes TP (2010) Practical considerations for monitoring patients with chronic myeloid leukemia. Semin Hematol 47: 327-334.
  100. Ernst T, Gruber FX, Pelz-Ackermann O, Maier J, Pfirrmann M, et al. (2009) A co-operative evaluation of different methods of detecting BCR-ABL kinase domain mutations in patients with chronic myeloid leukemia on second-line dasatinib or nilotinib therapy after failure of imatinib. Haematologica 94: 1227-1235.
  101. Jones D, Kamel-Reid S, Bahler D, Dong H, Elenitoba-Johnson K,et al. (2009) 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 Diagn 11: 4-11.
  102. Parker WT, Ho M, Scott HS, Hughes TP, Branford S (2012) Poor response to second-line kinase inhibitors in chronic myeloid leukemia patients with multiple low-level mutations, irrespective of their resistance profile. Blood 119: 2234-2238.
  103. Melo JV, Ross DM (2011) Minimal residual disease and discontinuation of therapy in chronic myeloid leukemia: can we aim at a cure? Hematology Am Soc Hematol Educ Program 2011: 136-142.
  104. Mahon FX, Rea D, Guilhot J, Guilhot F, Huguet F,et al (2010) Nicolini F, Legros L, Charbonnier A, Guerci A, Varet B, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: The prospective, multicentre stop imatinib (STIM) trial. Lancet Oncol 11: 1029-1035.
  105. Gratwohl A, Brand R, Apperley J, Crawley C, Ruutu T,et al. (2006) Allogeneic hematopoietic stem cell transplantation for chronic myeloid leukemia in europe Transplant activity, long-term data and current results. an analysis by the chronic leukemia working party of the european group for blood and marrow transplantation (EBMT). Haematologica 91: 513-521.
  106. Sobrinho-Simões M, Wilczek V, Score J, Cross NC, Apperley JF, et al. (2010) In search of the original leukemic clone in chronic myeloid leukemia patients in complete molecular remission after stem cell transplantation or imatinib. Blood 116: 1329-1335.
  107. Ross DM, Branford S, Seymour JF, Schwarer AP, Arthur C, et al. (2010) Patients with chronic myeloid leukemia who maintain a complete molecular response after stopping imatinib treatment have evidence of persistent leukemia by DNA PCR. Leukemia 24: 1719-1724.
  108. Hamilton A, Helgason GV, Schemionek M, Zhang B, Myssina S, et al. (2012) Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood 119: 1501-1510.

Citation: Singh Z, Medlin S, Usmani SZ (2012) Molecular Monitoring and Treatment of Chronic Myeloid Leukemia (CML). J Clin Exp Pathol 2:113. Doi: 10.4172/2161-0681.1000113

Copyright: © 2012 Singh Z, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top