The management of hematologic malignancies, including chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), and acute lymphocytic leukemia (ALL), has led the way in establishing the use of regular laboratory testing to assess for residual disease on an ongoing basis. The goals of minimal residual disease (MRD) monitoring for leukemias include (1) demonstration of the effectiveness of initial therapy; (2) monitoring for treatment resistance or relapse; and (3) dissecting the mechanisms of treatment failure to help in selection of alternative therapies. The underlying biology and range of treatment options for each leukemia type influence the methodologies used for MRD monitoring and the timing and duration of such monitoring. For example, flow cytometric monitoring plays a large role in MRD monitoring for ALL and CLL, whereas it is not used currently in CML. Here, we present an overview of the various approaches to monitoring CML, and a practical summary of the current uses of molecular methodologies and their routine applications in clinical practice.

The treatment of patients with Philadelphia chromosome (Ph)–positive CML with imatinib mesylate has resulted in complete cytogenetic response rates of 65% to 85%, major molecular response rates (ie, 3-log reduction in transcript levels from a standardized baseline) of 40% to 70%, and complete molecular response rates (ie, undetectable BCR-ABL transcripts) of 10% to 40%.1,3  Routine cytogenetic analysis is still considered the gold standard for evaluating response in CML.4,,7  As most patients achieve complete cytogenetic responses with modern therapy, there is a need to develop more sensitive and accurate monitoring tools to measure residual disease.1,3  It has been assumed that such increased accuracy might help better predict outcome.

Monitoring methods now include fluorescent in situ hybridization (FISH)8,,,,13  and qualitative and quantitative polymerase chain reaction (QPCR).14,,,,,,,,,,25  Molecular studies (ie, PCR) quantify the amount of disease with an increased sensitivity of up to 10−8. Better molecular responses may correlate with improved outcome. The rapidity of molecular response may also have additional prognostic significance.25,,28  Achievement of a major molecular response after 12 months of imatinib therapy was associated with the best progression-free survival.1,24  Thus, many specialists use today molecular studies in lieu of cytogenetic analysis to monitor response.29,31 

Resistance to imatinib and to other BCR-ABL tyrosine kinase inhibitors has been associated with the development of mutations in the BCR-ABL kinase domain in 40% to 50% of instances. Mutations prevent the activity of imatinib in different ways. Some may prevent the binding of the tyrosine kinase inhibitor to BCR-ABL; others may favor the active conformation of the kinase, a conformation to which imatinib cannot bind.32,34  Different mutations may confer different levels of resistance, from an absolute resistance, to minimal or no resistance, depending on their location and their effect on the kinase. Molecular and mutational analyses have figured prominently in recent scientific studies, but have also been used in clinical practice to monitor the course of CML. It is unclear whether these more sensitive assays are of value in the management of individual patients; misinterpretation of these tests could lead to potentially harmful clinical decisions. Frequently noted examples are (1) referral of patients in complete cytogenetic response for allogeneic stem-cell transplantation because of “increasing” BCR-ABL transcript levels; (2) changing therapy to new tyrosine kinase inhibitors for similar reasons; and (3) declaring resistance to therapy in a patient who had been reported to be molecularly “negative” and who subsequently shows positive transcript levels.

Because of the confusion about the appropriate use and interpretation of these studies, we elected to review the pertinent information in the context of current clinical practice, and to recommend reasonable monitoring steps. This review emphasizes the practical rather than the technical aspects of methods, and should be considered along with recently published recommendations.30,31 

There are several tools to identify and quantify the abnormally rearranged gene. Advantages and disadvantages of these monitoring methods are shown in Table 1.

Conventional cytogenetic studies using chromosome Giemsa banding

These are still the gold standard of treatment monitoring. The current definitions of cytogenetic responses were derived from cytogenetic analysis in at least 20 evaluable metaphases. These include the following: complete cytogenetic response—0% Ph-positive metaphases; partial cytogenetic response—1% to 35% Ph-positive metaphases; and minor-minimal (or simply minor) cytogenetic response—36% to 90% or 95% Ph-positive metaphases. A major cytogenetic response refers to Ph-positive metaphases 0% to 35%.5,6,35  These definitions were established based on the strong association of major and complete cytogenetic response with improved long-term survival with interferon-alpha therapy.5,7  These prognostic associations have also been confirmed with imatinib therapy,1,3  suggesting that the relationship between response and survival may be independent of the treatment that produced the response.2  Cytogenetic studies analyze only cells in metaphase, require a marrow sample, are time consuming, and have a wide confidence interval. In a patient who demonstrates 5 of 20 Ph-positive metaphases (25% Ph-positive = partial cytogenetic response), the 95% confidence interval of such positivity is 9% to 49%. Studies of the association of cytogenetic response with long-term prognosis have so far relied on classic cytogenetic analysis, not on FISH-based results.

Fluorescent in situ hybridization studies

Several FISH methodologies have been developed and are reviewed elsewhere.8,,,,,14  Traditional FISH uses a 5′ BCR and a 3′ ABL fluorescent probe of different colors and has a false-positive threshold of 1% to 10%.9  Newer FISH techniques use 3 or 4 probes, the latter known as double-FISH (D-FISH). D-FISH reduces the rates of false positives.11  Interphase FISH does not require cells to enter into division, and is thus applicable to a larger population of cells. It is more prone for false positivity. Hypermetaphase-FISH is applicable only to marrow dividing cells and therefore has limited utility.13  Most laboratories now report interphase FISH analyses using multiple standardized probes. FISH studies use peripheral blood samples and analyze approximately 200 interphase cells. The results have a narrower confidence interval than cytogenetic analysis (eg, 50/200 positive equals 25%; 95% confidence interval 19% to 32%). The sensitivity limit is not always reported, but most laboratories have an upper limit of false positivity of 1% to 5%. This limits the interpretation of complete cytogenetic response by FISH and its interchangeable use with routine cytogenetic studies. In a study by Testoni et al,36  there was generally a good correlation between cytogenetic and FISH results (Table 2). However, 18% of patients in complete cytogenetic response by standard cytogenetics had 1% to more than 5% FISH-positive cells.

While many oncologists use FISH interchangeably with (or in place of) cytogenetic studies, few studies have investigated the association of long-term outcome with cytogenetic response assessed by FISH. However, it is reasonable to expect that responses assessed by FISH may have similar long-term implications.36,,,40  In fact, FISH is beginning to be incorporated into current studies of tyrosine kinase inhibitors in CML,41  although most current trials exploring the efficacy of the new tyrosine kinase inhibitors and phase 3 trials comparing imatinib to the new tyrosine kinase inhibitors still rely on conventional cytogenetic analysis for cytogenetic response. Whether patients in complete cytogenetic response (ie, 0% Ph-positive by cytogenetic analysis) but FISH positive would have a similar or worse prognosis to FISH-negative patients is unknown. Clinicians should also be cautious in establishing failure to treatment based on low levels of positivity by FISH.

In clinical practice, it may be reasonable to use peripheral blood FISH as an alternative to routine cytogenetics until the FISH levels are less than 5% to 10%. At that point, a cytogenetic analysis should be used to confirm a complete cytogenetic response. Patients can subsequently be monitored with FISH studies, as long as they are reported to be negative or within the range of background positive results reported by the laboratory performing the test. Persistent low levels of FISH positivity require additional testing (discussed in “Molecular studies”).

Molecular studies, polymerase chain reaction

In earlier studies of patients after stem-cell transplantation, qualitative reverse-transcriptase polymerase chain reaction (RT-PCR) was used. More recently, quantitative techniques have been developed, now most frequently performed by real-time RT-PCR.14  Results usually report the ratio of BCR-ABL to a reference gene (recommended genes include ABL, BCR, and GUSB).17,29,31  Several technical aspects have not been standardized, including the sample source (blood or marrow), the effects of shipping and storage, the amount of sample required, the techniques for extraction of RNA, and the preferred reference gene. A detailed description of the techniques is beyond the scope of this review, but has been discussed previously, together with the variability and reproducibility/reliability of such molecular studies.29,30 

As more patients achieve complete cytogenetic responses with imatinib and other tyrosine kinase inhibitors, there is interest in obtaining better molecular responses. A major molecular response has been defined as a reduction in transcript levels (BCR-ABL/reference gene) of at least 3-logs compared with a standardized baseline obtained from patients with untreated newly diagnosed CML. The standardized baseline in the IRIS trial defined as the average ratio from 30 patients was 36%; thus, a major molecular response was defined as achieving levels of 0.036% or less. The considerable range in the values among these patients lends some imprecision to the calculation of the log reduction in an individual patient. This standardized baseline value has to be defined by individual laboratories; unfortunately, this has not been done by many commercial laboratories. The baseline value can also vary considerably from laboratory to laboratory and is not always included in the laboratory report.17,24,25  There is currently an effort to establish a harmonization of results from different laboratories using predefined control samples to adjust the results from each laboratory, such that a conversion factor can be established. Then, all laboratories can adjust their values uniformly to define a major molecular response as a value of 0.1% or less on the adjusted scale.30 

A complete molecular response is achieved when transcripts are not detectable (better referred to as undetectable transcript levels), in an assay that has at least 4- to 5-log range of detection. Unfortunately, when negative results for BCR-ABL are reported, the sensitivity is not always reported.

Molecular studies have several advantages including (1) good correlation between marrow and blood levels and (2) detection of very low levels of residual disease. Limitations include (1) a substantial incidence of false-negative tests (RNA degradation, low sensitivity of a given assay); (2) a coefficient of variability, which may be up to 0.5 log; and (3) poor reproducibility of results. Wide variations of transcript levels may be due simply to technical reasons rather than real changes in CML burden.

Mutation analysis

Analysis of mutations of the BCR-ABL kinase domain use different techniques: (1) direct sequencing of DNA following nested RT-PCR (detect mutations in 10% to 25% of tumor cells)30,42 ; (2) denaturing-high performance liquid chromatography analysis (DHPLC; sensitivity of 5% to 10%),43  and (3) fluorescent-based allele-specific oligonucleotide PCR (ASO-PCR) assays (detect mutations in 0.1% to 1% of tumor cells).44  Sequencing of DNA clones increases the sensitivity of the assays in some studies. Direct sequencing is the technique most frequently available to clinicians. Mutation studies have been performed in peripheral blood or bone marrow; a correlation between samples has not been reported.

Cytogenetic analysis

Routine karyotype is the only routinely available technique that assesses all chromosomes. This is important because additional chromosomal abnormalities besides Ph (ie, clonal evolution) are a criterion for accelerated phase. In addition, some patients treated with tyrosine kinase inhibitors develop chromosomal abnormalities in Ph-negative cells. Most abnormalities are transient or clinically innocuous. Rare instances of transition to myelodysplastic syndromes or acute leukemias have been reported. Thus, performing a marrow aspiration with cytogenetic analysis is recommended at the time of diagnosis, to confirm complete cytogenetic response, and every 12 months thereafter. Conventional cytogenetic studies at 3, 6, and 12 months are performed more often in the setting of research programs. In routine practice, the 3- and 6-month evaluations often use FISH studies that give reasonable estimates of cytogenetic response at these time points, without resorting to a painful bone marrow aspiration procedure.

Fluorescent in situ hybridization studies

Pretreatment FISH studies provide some potential benefits: (1) In an occasional patient with a morphologic picture of CML but no detectable Ph-positive metaphases by routine cytogenetics, a FISH or a molecular analysis may detect BCR-ABL–mediated disease.45  (2) Peripheral blood FISH can confirm a diagnosis of CML (although avoiding marrow analysis is not recommended, as the percentage of blasts and basophils in the bone marrow is needed to establish the disease staging). Conversely, FISH or PCR studies can exclude the diagnosis of CML in patients with leukocytosis, obviating the need for a marrow biopsy. (3) They allow for the detection of deletions around the breakpoint of the derivative chromosome 9.46,47  These had been associated with worse prognosis in the preimatinib era,46,47  but this is now controversial48  and may not be evident with imatinib therapy.49 

FISH studies pretreatment are useful to establish a pretreatment baseline to monitor response by FISH.50  FISH studies are a useful alternative to cytogenetic studies during early Ph suppression, and are informative in a state of complete cytogenetic response if the FISH results are negative.

Molecular studies

Molecular studies before treatment may identify Ph-negative BCR-ABL–positive CML, and establish a baseline level for subsequent monitoring. Failure to achieve a cytogenetic response confers a poor prognosis. Whether molecular results can define patients in complete cytogenetic response who are destined to relapse is unclear. The results from the IRIS trial, the most widely cited in this regard, showed that patients who achieved a major molecular response at 12 months had a small but statistically significantly better probability of survival free from transformation to accelerated or blastic phases. The 5-year transformation-free survival was 100% for patients who achieved a major molecular response (≥ 3-log reduction in transcript levels) versus 95% if they had achieved a complete cytogenetic remission but not a major molecular response (P = .01).1,24  Although a similar analysis for survival was not reported, it is likely that the difference would be even smaller. Thus, not achieving a major molecular response in a patient in complete cytogenetic remission at 12 months should not trigger a major change of therapy, particularly given the variability of the assays.

Other studies have not shown a significant difference in progression-free survival or in survival in patients achieving a complete cytogenetic response at 12 months by the depth of molecular response (major vs less than major).2  Similar results were obtained using landmark analyses at 6, 18, and 24 months (H.K., unpublished data, October 2007). The proposed definitions of failure to imatinib therapy do not include failure to achieve molecular response.31  Whether any change in treatment strategy (eg, dose increase, addition of other agents, change to different inhibitors) improves the long-term outcome of patients with suboptimal responses remains to be seen.

If fluctuations in QPCR levels in complete cytogenetic response do not necessarily dictate a change of therapy, why should transcript levels be monitored in a patient in complete cytogenetic response? The first reason derives from studies that showed that a 2- to 5-fold rise in transcript levels was associated with a higher rate of detection of mutations (presumably an ominous sign).51,52  Studies from M. D. Anderson suggested a higher incidence of cytogenetic relapse only among patients who never achieved a major molecular remission and who had a 10-fold (1 log) increase of transcript levels.25  The difference among these results may be due to different methodologies or fluctuations in PCR measurements. However, they emphasize the risk of overreacting clinically, and the need to critically interpret results in individual patients even when reported by specialized academic laboratories.

A second reason to advocate molecular monitoring is that it can be used in lieu of other tests to serially monitor response or to evaluate discordant FISH results. Because of the variability of PCR results (up to 0.5-log even in high-quality laboratories), they should be performed from the same consistent compartment (usually blood rather than marrow), by the same reliable high-quality laboratory, and using high-quality collection and shipping procedures.31 BCR-ABL transcript increases of 2- to 5-fold could trigger a mutational analysis, but the yield of detection in such instances is low in many laboratories, compared with searching when there is clinical and/or cytogenetic evidence of relapse.

Mutational studies

Studies of BCR-ABL KD mutations in patients not previously exposed to BCR-ABL tyrosine kinase inhibitors show low-level detection of mutations in patients in accelerated or blast phase, and in late chronic phase. Mutations prior to exposure to imatinib have not been reported in newly diagnosed chronic-phase CML. Even when detected in advanced disease, these mutations may not have prognostic significance.44  In a study by Willis et al,44  66 patients with CML (20 chronic, 27 accelerated, 19 blastic), were studied for the presence of 8 common mutations by a sensitive method of cDNA sequencing and ASO-PCR assays: 14 patients (23%) showed mutations, 8 (12%) involving T315I. Mutations were not detected in any of the 20 patients in chronic phase. Mutations did not correlate with response to imatinib, event-free survival, or survival. Among patients responding to imatinib therapy, mutations are also rarely encountered, and when present they represent 1% to less than 20% of the total CML population.53,54  Mutations in this setting did not necessarily persist or expand, and were not consistently associated with progression. In a study by Sherbenou et al using direct sequencing and denaturing HPLC, 9 (11%) of 42 patients with CML in complete cytogenetic response on imatinib had mutations. With a median follow up of 12 months, 5 patients had increased BCR-ABL transcript levels and 4 remained with low BCR-ABL transcript levels; only 1 of 9 patients had a cytogenetic relapse.54 

In patients with cytogenetic or hematologic resistance or relapse, mutational studies have yielded clinically relevant information. In patients with chronic-phase CML who develop resistance to imatinib, 30% to 50% of patients exhibit imatinib-resistant BCR-ABL KD mutations. New, more potent tyrosine inhibitors, some of which also inhibit SRC family of kinases, overcome resistance in most such cases as well as in instances where no mutations are detected.

In the setting of clinical resistance to imatinib, mutational analyses may be helpful in several ways. Identifying T315I mutations, sometimes referred to as a “gatekeeper” mutation, predicts for resistance to the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib. Such patients may be referred earlier for allogeneic stem cell transplantation (SCT) or offered investigational therapies with drugs that can inhibit T315I-bearing BCR-ABL. Since resistance in chronic phase occurs in 4% of patients, and T315I mutations account for 10% to 20% of the 40% of resistant patients who develop mutations, the development of the T315I mutations is predicted to occur in approximately 0.16% to 0.32% of newly diagnosed patients in chronic phase on therapy (prevalence of CML approximately 70 000 cases in the United States; annual number of cases with T315I mutations approximately 150 to 300).

Mutations of the BCR-ABL KD have been categorized by their location into mutations occurring in the ATP-binding domain (P-loop), the critical sites of direct contact with imatinib and other kinase inhibitors, the activation loop (A-loop), and the catalytic domain. The definition of P-loop includes amino acids 248 to 255,32  although some have suggested it should be restricted to amino acids 250 to 255. Earlier studies have associated T315I and P-loop mutations with worse outcomes.

A study by Branford et al42  reported a very poor survival in patients with P-loop mutations. The 1-year survival rate of 13 patients with P-loop mutations was 15% versus 80% with non–P-loop mutations (P = .002). This was corroborated by a multicentric study reported by Nicolini et al58  An Italian study by Soverini et al analyzed mutational status by D-HPLC and sequencing among patients who received imatinib and had less than a major cytogenetic response (Ph-positive disease > 35%) after 12 months of therapy. Among 40 such patients, 19 (48%) had detectable mutations. The presence of any mutation, or of P-loop mutations, correlated with worse time to progression and with worse survival and progression-free survival rates.43  Other studies could not confirm these findings.59,60  Jabbour et al59  analyzed 171 patients with CML after imatinib failure: 62 patients (36%) had mutations (P-loop in 24). They found a similar survival in patients with chronic-phase CML and imatinib resistance whether or not they had mutations, and whether the mutation was located in T315I, the P-loop, or other areas.59  Possible reasons behind the differences in the results were reviewed.61  Prognosis in CML after imatinib failure may be dependent on several factors including the therapeutic interventions used after imatinib failure. The “geographic” grouping of mutations is probably not appropriate for purposes of defining outcome and therapy. Different mutations may alter the kinase activity and proliferation potency of BCR-ABL differently, which may be more relevant than the location of the mutation. Thus, mutations are better described in terms of their relative sensitivity to specific therapies such as imatinib, nilotinib, and dasatinib (ie, the IC50 for each drug to inhibit BCR-ABL with the specific mutation in vitro). With further clinical experience with the newer tyrosine kinase inhibitors, mutational studies may allow selection of particular treatments based on relative sensitivities and clinical responses to different inhibitors, although the approach of selecting therapy based on the specific mutation has not been tested prospectively (Table 3).55,57  Furthermore, whether finding a mutation in patients with rising transcript levels who are still in complete cytogenetic response should prompt a change in therapy remains to be investigated. The risks and benefits of the alternative therapy should also be carefully evaluated.

When the authors discussed this section, there was only partial consensus on the optimal monitoring approach with some disagreements on the details of the sequence of tests to be used for patients in chronic phase. We all agreed that adequate monitoring is important to optimize outcome of patients with CML. We therefore elected to present several monitoring approaches as proposed and practiced by CML experts.

The “conservative” approach

Some investigators contend that the best current information relating response and prognosis derives from routine cytogenetic studies. Based on this, they suggest that cytogenetic analysis should still be a central element of monitoring. Thus, a conservative approach would be to monitor patients by bone marrow cytogenetic analysis every 6 months until a complete cytogenetic response is achieved, and then every 6 to 12 months.

The “FISHer” approach

As valuable and historically important as they may be, cytogenetic studies usually require a marrow aspiration, which is painful. Some physicians rely only on peripheral blood FISH to follow response to avoid doing marrows. The extreme proponents of this approach may use only FISH as a surrogate for “molecular” responses, either erroneously interpreting the FISH as sensitive enough to evaluate such responses, or not being convinced that molecular responses may add much information in patients in complete cytogenetic remission (ie, those with repeatedly “negative” results) as measured by FISH.

The “molecular enthusiast” approach

Some CML experts have endorsed the molecular analysis as a substitute to cytogenetics for monitoring. In this approach, PCR studies are performed before treatment, then every 3 months in the first year, and every 3 to 6 months subsequently. This approach relies purely on peripheral blood monitoring and aims to achieve and maintain QPCR levels at a major or better molecular response (eg, BCR-ABL/ABL ratio of 0.1% or less). This response would be associated with a very low relapse rate and excellent survival. Cytogenetic responses may not even be reported, instead referring to a 2-log reduction in transcript levels (or BCR-ABL/ABL ratio of 1% in the international scale) as the equivalent of such a response. Disadvantages to this approach include (1) wide variability of the test (up to 0.5-log or 2- to 5-fold range variation) in commercial and even some “reference” laboratories; (2) disregard of any information from marrow procedures (eg, percentage of basophils and blasts, presence of myelofibrosis, cytogenetic abnormalities in Ph-positive or Ph-negative cells); (3) false-negative results in laboratories using assays of low sensitivity; and (4) unfamiliarity with the possible variability in results that may not reflect a clinically significant change in CML burden. Physicians are seldom provided with the standardized baseline for the laboratory reporting the results, making interpretation difficult. The proposed international standardization is unfortunately not yet universal. Under these circumstances, molecular monitoring may lead to improper therapeutic decisions.

The “I-don't-care-about-any-such-studies” approach

It is interesting that some oncologists and hematologists do not use any of these studies, particularly in elderly patients (eg, 70 years or older). The proponents of this approach perceive the benefit-cost ratio to be low, since they may not routinely change therapy unless there is clear-cut evidence of hematologic relapse, which can be detected by inexpensive peripheral blood count studies. The authors believe this approach not to be in the patient's best interest, as it could delay the implementation of beneficial therapeutic interventions until settings where they may be less effective (eg, advanced-stage disease). At a minimum, some assessment of the degree of CML response is mandatory, given the different options available to patients not responding to imatinib.

A “hybrid” approach

This approach mixes the relative strengths of different monitoring procedures and variations of this approach are favored by the authors. The weight given to each method, and some of the details (eg, frequency, methodology) may vary by investigator.

(a) Since a bone marrow analysis before treatment is useful to assess prognostic factors such as the percentage of marrow blasts and basophils and clonal evolution, a pretreatment cytogenetic analysis is advisable. FISH or molecular studies on the peripheral blood may not be necessary unless the physician wishes to rely on them solely for monitoring. However, they can help establish that the transcripts can indeed be detected in the individual patient by such tests. Subsequently, a marrow analysis and cytogenetic studies are ordered at 6 and 12 months, then once every 1 to 2 years after a patient has achieved a complete cytogenetic response. If, with further research, the chromosomal abnormalities in Ph-negative metaphases prove to be clinically insignificant, then marrow analyses can be performed less frequently (eg, every 2+ years).

(b) FISH studies would be performed every 3 months in the first 12 months until repeated values are negative. FISH studies would be omitted when marrow analyses are performed (cytogenetic studies done instead). After cytogenetic complete response is confirmed, regular monitoring by QPCR should be done.

(c) Patients can be monitored using real-time PCR every 3 months in the first 12 months, then every 6 months once a stable complete cytogenetic response is achieved. As long as BCR-ABL/control gene ratios are within the range of major molecular response (0.1% or less in the international scale), this indicates a solid response. If there is an increase in the ratio (greater than the coefficient of variability), this should trigger a repeat analysis, more frequent monitoring (eg, every 3 months), and repeating a bone marrow analysis and cytogenetics, particularly if QPCR levels rise to higher than 1%. Variations in BCR-ABL/ABL levels in a patient in complete cytogenetic response would not trigger a major change in therapy (eg, allogeneic stem-cell transplantation). In a patient with repeatedly rising molecular levels (eg, to higher than 0.1% and a 5-fold increase) who remains in complete cytogenetic response, it may be reasonable to increase the dose of imatinib with a switch to a new TK inhibitor should the dose increase not be effective, or to offer a clinical trial. It is also important to determine that the patient was compliant with taking his/her imatinib. Some investigators also advocate that a rise of QPCR levels by 2- to 5-fold should trigger a mutational analysis.

So, when do we do the mutational studies?

In our opinion, mutational studies should be performed only with evidence of hematologic or cytogenetic resistance relapse. Whether they should be performed in patients responding to imatinib therapy if PCR transcript levels increase by 2- to 5-fold remains an open question. But if they are performed, the results should be interpreted with caution, particularly as it relates to any proposed change in therapy. Risks and benefits based on available data should be carefully weighed. Mutational studies may also be helpful any time a change of therapy is considered: identification of specific mutations may help in the choice of one treatment approach over another (eg, allogeneic SCT if T315I mutation identified) or of a specific BCR-ABL inhibitor depending on the relative sensitivity of the particular mutation to these inhibitors (Table 3).55,57 

The outcome of patients with CML has improved significantly, and the availability of an increasing number of effective treatment options makes adequate monitoring of patients increasingly important. While some research trials are proposing to evaluate the incidences of major or complete molecular response in patients in complete cytogenetic response as the comparative efficacy end point, the prognostic significance of such molecular (major, complete) end points for long-term survival is unknown. Just as the treatment of CML is rapidly changing, concepts related to monitoring, including available techniques, the interpretation of the results, and the best use of this information are constantly and rapidly evolving. Table 4 proposes a reasonable monitoring treatment approach for CML.

Contribution: H.K., C.S., and J.C. designed research, performed research, analyzed data, and wrote the paper; D.J. performed research, analyzed data, and wrote the paper.

Conflict-of-interest disclosure: H.K., C.S., and J.C. received research grants from BMS and Novartis. D.J. declares no competing financial interests.

Correspondence: Hagop Kantarjian, Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030; e-mail: hkantarj@mdanderson.org.

1
Druker
B
Guilhot
F
O'Brien
S
et al
Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia.
N Engl J Med
2006
355
2408
2417
2
Kantarjian
H
Talpaz
M
O'Brien
S
et al
Survival benefit with imatinib mesylate versus interferon alpha-based regimens in newly diagnosed chronic phase chronic myelogenous leukemia.
Blood
2006
108
1835
1840
3
O'Brien
SG
Guilhot
F
Larson
R
et al
Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia.
N Engl J Med
2003
348
994
1004
4
Guilhot
F
Chastang
C
Michallet
M
et al
Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia.
N Engl J Med
1997
337
223
229
5
Kantarjian
HM
Smith
TL
O'Brien
S
et al
Prolonged survival in chronic myelogenous leukemia after cytogenetic response to interferon.
Ann Intern Med
1995
122
254
261
6
Kantarjian
H
O'Brien
S
Cortes
J
et al
Complete cytogenetic and molecular responses to interferon-α-based therapy for chronic myelogenous leukemia are associated with excellent long-term prognosis.
Cancer
2003
97
1033
1041
7
Bonifazi
F
de Vivo
A
Rosti
G
et al
Chronic myeloid leukemia and interferon-α: a study of complete cytogenetic responders.
Blood
2001
98
3074
3081
8
Landstrom
A
Tefferi
A
Fluorescent in situ hybridization in the diagnosis, prognosis, and treatment monitoring of chronic myeloid leukemia.
Leukemia Lymphoma
2006
47
397
402
9
Dewald
GW
Schad
CR
Christensen
ER
Tiede
AL
Zinsmeister
AR
Spurbeck
JL
The application of fluorescent in situ hybridization to detect Mbcr/abl fusion in variant Ph chromosomes in CML and ALL.
Cancer Genet Cytogenet
1993
71
7
14
10
Sinclair
PB
Green
AR
Grace
C
Nacheva
EP
Improved sensitivity of BCR-ABL detection: a triple-probe three-color fluorescence in situ hybridization system.
Blood
1997
90
1395
1402
11
Dewald
GW
Wyatt
WA
Juneau
AL
Carlson
RO
Zinsmeister
AR
Jalal
SM
Highly sensitive fluorescence in situ hybridization method to detect double BCR/ABL fusion and monitor response to therapy in chronic myeloid leukemia.
Blood
1998
91
3357
3365
12
Dewald
G
Stallard
R
Alsaadi
A
et al
A multicenter investigation with D-FISH BCR/ABL1 probes.
Cancer Genet Cytogenet
2000
116
97
104
13
Seong
DC
Kantarjian
H
Ro
JY
et al
Hypermetaphase fluorescence in situ hybridization for quantitative monitoring of Philadelphia chromosome-positive cells in patients with chronic myelogenous leukemia during treatment.
Blood
1995
86
2343
2349
14
Wang
YL
Bagg
A
Pear
W
Nowell
PC
Hess
J
Chronic myelogenous leukemia: laboratory diagnosis and monitoring.
Genes Chromosomes Cancer
2001
32
97
111
15
Cross
NC
Feng
L
Chase
A
Bungey
J
Hughes
TP
Goldman
JM
Competitive polymerase chain reaction to estimate the number of BCR-ABL transcripts in chronic myeloid leukemia patients after bone marrow transplantation.
Blood
1993
82
1929
1936
16
Branford
S
Hughes
TP
Rudzki
Z
Monitoring chronic myeloid leukaemia therapy by real-time quantitative PCR in blood is a reliable alternative to bone marrow cytogenetics.
Br J Haematol
1999
107
587
599
17
Hochhaus
A
Lin
F
Reiter
A
et al
Quantification of residual disease in chronic myelogenous leukemia patients on interferon-alpha therapy by competitive polymerase chain reaction.
Blood
1996
87
1549
1555
18
Kantarjian
H
Talpaz
M
Cortes
J
et al
Quantitative polymerase chain reaction monitoring of BCR-ABL during therapy with imatinib mesylate (STI571; Gleevec) in chronic phase chronic myelogenous leukemia.
Clin Cancer Res
2003
9
160
166
19
Luthra
R
Sanchez-Vega
B
Medeiros
LJ
Quantitative TaqMan t (9;22) (q34;q11) PCR coupled with capillary electrophoresis-based identification of bcr-abl fusion sequences.
Mod Pathol
2004
17
96
103
20
Men sink
E
van de Locht
A
Quantitation of minimal residual disease in Philadelphia chromosome positive chronic myeloid leukemia patients using real-time quantitative RT-PCR.
Br J Haematol
1998
102
768
774
21
Emig
M
Saussele
S
Wittor
H
et al
Accurate and rapid analysis of residual disease in patients with CML using specific fluorescent hybridization probes for real time quantitative RT-PCR.
Leukemia
1999
13
1825
1832
22
Preudhomme
C
Revillion
F
Merlat
A
et al
Detection of BCR-ABL transcripts in chronic myeloid leukemia (CML) using a “real time quantitative RT-PCR assay.
Leukemia
1999
13
957
964
23
Wang
L
Pearson
K
Pilletteri
C
et al
Serial monitoring of BCR-ABL by peripheral blood real-time polymerase chain reaction predicts the bone marrow cytogenetic response to imatinib mesylate in chronic myeloid leukaemia.
Br J Haematol
2002
118
771
777
24
Hughes
TP
Kaeda
J
Branford
S
et al
Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia.
N Engl J Med
2003
349
1423
1432
25
Cortes
J
Talpaz
M
O'Brien
S
et al
Molecular responses in patients with chronic myelogenous leukemia in chronic phase treated with imatinib mesylate.
Clin Cancer Res
2005
11
3425
3432
26
Merx
K
Muller
MC
Kreil
S
et al
Early reduction of BCR-ABL mRNA transcript levels predicts cytogenetic response in chronic phase CML patients treated with imatinib after failure of interferon α.
Leukemia
2002
16
1579
1583
27
Muller
MC
Gattermann
N
Lahaye
T
et al
Dynamics of BCR-ABL mRNA expression in first-line therapy of chronic myelogenous leukemia patients with imatinib or interferon α/ara-C.
Leukemia
2003
17
2392
2400
28
Wang
L
Pearson
K
Ferguson
JE
Clark
RE
The early molecular response to imatinib predicts cytogenetic and clinical outcome in chronic myeloid leukaemia.
Br J Haematol
2003
120
990
999
29
Branford
S
Rudzki
Z
Harper
A
et al
Imatinib produces significantly superior molecular responses compared to interferon plus cytarabine in patients with newly diagnosed chronic myeloid leukemia in chronic phase.
Leukemia
2003
17
2401
2409
30
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
31
Baccarani
M
Saglio
G
Goldman
J
et al
Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet.
Blood
2006
108
1809
1820
32
Shah
NP
Nicoll
JM
Nagar
B
et al
Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia.
Cancer Cell
2002
2
117
125
33
Branford
S
Rudzki
Z
Walsh
S
et al
High frequency of point mutations clustered within the ATP binding region of BCR/ABL in CML and Ph-positive ALL patients who develop imatinib (STI571) resistance.
Blood
2002
99
3472
3475
34
Corbin
AS
La Rosee
P
Stoffregen
EP
et al
Several Bcr-Abl kinase domain mutants associated with imatinib mesylate resistance remain sensitive to imatinib.
Blood
2003
101
4611
4614
35
Talpaz
M
Kantarjian
H
McCredie
K
Keating
M
Trujillo
J
Gutterman
J
Clinical investigation of human alpha interferon in chronic myelogenous leukemia.
Blood
1987
69
1280
1288
36
Testoni
N
Luatti
S
Marzocchi
G
et al
A prospective study in Ph+ chronic myeloid leukemia (CML) patients showing that interphase fluorescence in situ hybridization (FISH) is as effective as conventional cytogenetics for definition of cytogenetic response. Correlation with molecular response [abstract].
Blood
2006
108
suppl 1
Abstract 4779
37
Lesser
ML
Dewald
GW
Sison
CP
Silver
RT
Correlation of three methods of measuring cytogenetic response in chronic myelocytic leukemia.
Cancer Genet Cytogenet
2002
137
79
84
38
Douet-Guilbert
N
Morel
F
Charpentier
TL
Bris
MJL
Herry
A
Morice
P
Interphase FISH for follow-up of Philadelphia chromosome-positive chronic myeloid leukemia treatment.
Anticancer Res
2004
24
2535
2539
39
Schoch
C
Schnittger
S
Bursch
S
et al
Comparison of chromosome banding analysis, interphase- and hypermetaphase-FISH, qualitative and quantitative PCR for diagnosis and for follow-up in chronic myeloid leukemia: a study on 350 cases.
Leukemia
2002
16
53
59
40
Cuneo
A
Bigoni
R
Emmanuel
B
Smit
E
Rigolin
G
Roberti
M
Fluorescence in situ hybridization for the detection and monitoring of the Ph-positive clone in chronic myelogenous leukemia: comparison with metaphase banding analysis.
Leukemia
1998
12
1718
1723
41
Kantarjian
H
Giles
F
Gattermann
N
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
2007
110
3540
3546
42
Branford
S
Rudzki
Z
Walsh
S
et al
Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis.
Blood
2003
102
276
283
43
Soverini
S
Martinelli
G
Rosti
G
et al
ABL mutations in late chronic phase chronic myeloid leukemia patients with up-front cytogenetic resistance to imatinib are associated with a greater likelihood of progression to blast crisis and shorter survival: a study by the GIMEMA Working Party on Chronic Myeloid Leukemia.
J Clin Oncol
2005
23
4100
4109
44
Willis
S
Lange
T
Demehri
S
et al
High-sensitivity detection of BCR-ABL kinase domain mutations in imatinib-naive patients: correlation with clonal cytogenetic evolution but not response to therapy.
Blood
2005
106
2128
2137
45
Seong
D
Kantarjian
H
Albitar
M
et al
Analysis of Philadelphia chromosome-negative BCR-ABL-positive chronic myelogenous leukemia by hypermetaphase fluorescence in situ hybridization.
Ann Oncol
1999
10
955
959
46
Sinclair
PB
Nacheva
EP
Leversha
M
Telford
N
Chang
J
Reid
A
Large deletions at the t(9;22) breakpoint are common and may identify a poor-prognosis subgroup of patients with chronic myeloid leukemia.
Blood
2000
95
738
743
47
Huntly
BJP
Reid
AG
Bench
AJ
Campbell
LJ
Telford
N
Shepherd
P
Deletions of the derivative chromosome 9 occur at the time of the Philadelphia translocation and provide a powerful and independent prognostic indicator in chronic myeloid leukemia.
Blood
2001
98
1732
1738
48
Yoong
Y
Van DeWalker
TJ
Carlson
RO
Dewald
GW
Tefferi
A
Clinical correlates of submicroscopic deletions involving the ABL-BCR translocation region in chronic myeloid leukemia.
Eur J Haematol
2005
74
124
127
49
Quintas-Cardama
A
Kantarjian
H
Talpaz
M
O'Brien
S
Garcia-Manero
G
Verstovsek
S
Imatinib mesylate therapy may overcome the poor prognostic significance of deletions of derivative chromosome 9 in patients with chronic myelogenous leukemia.
Blood
2005
105
2281
2286
50
Bornhauser
M
Mohr
B
Ehrenlechner
U
Neubauer
A
Ehninger
G
Fluorescence in situ hybridization for the BCR/ABL rearrangement is dependent on the percentage of nonlymphocytic cells in peripheral blood stem cell harvests.
J Hematother
1998
7
425
430
51
Branford
S
Rudzk
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
2932
52
Press
RD
Love
Z
Tronnes
AA
et al
BCR-ABL mRNA levels at and after the time of a complete cytogenetic response (CCR) predict the duration of CCR in imatinib mesylate-treated patients with CML.
Blood
2006
107
4250
4256
53
Roche-Lestienne
C
Soenen-Cornu
V
Grardel-Duflos
N
et al
Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment.
Blood
2002
100
1014
1018
54
Sherbenou
D
Wong
M
Humayun
A
et al
In chronic myeloid leukemia (CML) patients with complete cytogenetic response to imatinib, BCR-ABL kinase domain mutations are relatively rare and not consistently associated with subsequent relapse [abstract].
Blood
2005
106
Abstract 434
55
O'Hare
T
Walters
D
Stoffregen
E
et al
In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants.
Cancer Res
2005
65
4500
4505
56
Weisberg
E
Manley
P
Mestan
J
Cowan-Jacob
S
Ray
A
Griffin
JD
AMN107 (nilotinib): a novel and selective inhibitor of BCR-ABL.
Br J Cancer
2006
94
1765
1769
57
Bradeen
H
Eide
C
O'Hare
T
et al
Comparison of imatinib mesylate, dasatinib (BMS-354825), and nilotinib (AMN107) in an N-ethyl-N-nitrosourea (ENU)–based mutagenesis screen: high efficacy of drug combinations.
Blood
2006
108
2332
2338
58
Nicolini
F
Corm
S
Q-H
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(ϕ)-LMC GROUP).
Leukemia
2006
20
1061
1066
59
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
1773
60
Hochhaus
A
Ernst
T
Erben
P
et al
Long term observation of CML patients after imatinib resistance associated with BCR-ABL mutations [abstract].
Blood
2005
106
Abstract 1086
61
Nicolini
FE
Corm
S
Le
QH
Roche-Lestienne
C
Preudhomme
C
The prognosis impact of BCR-ABL P-loop mutations: worse or not worse?
Leukemia
2007
21
193
194
Sign in via your Institution