Purpose: To determine the clinical significance of molecular response and relapse among patients with chronic myelogenous leukemia (CML) treated with imatinib.

Experimental Design: We analyzed the results of quantitative PCR in 280 patients with CML in chronic phase who achieved complete cytogenetic remission with imatinib (117 after IFN-α failure and 163 previously untreated). Median follow-up was 31 months (range, 3-52 months).

Results: Median BCR-ABL/ABL ratio before the start of therapy was 39.44 (range, 0.252-170.53). A major molecular response (BCR-ABL/ABL ratio <0.05%) was achieved in 174 (62%), and transcripts became undetectable (complete molecular response) in 95 (34%). By multivariate analysis, only treatment with high-dose imatinib (P = 0.02) was associated with achievement of a major molecular response. Nine of 166 (5%) patients who achieved a major molecular response lost their cytogenetic remission, compared with 25 of 68 (37%) among those who did not achieve this response (P < 0.0001). Patients achieving a major molecular response 12 months after the start of therapy had significantly better complete cytogenetic remission duration than others. A >1-log reduction in transcript levels after 3 months of therapy predicted for an improved probability of achieving a major molecular response at 24 months. Increasing levels of BCR-ABL transcripts predicted for a loss of cytogenetic remission only among patients who did not achieve a major molecular response.

Conclusions: Achieving a major molecular response, particularly within the first year of therapy, is predictive of a durable cytogenetic remission and may be the future goal of therapy in CML.

Imatinib mesylate (Gleevec, Novartis, Basel, Switzerland) is a potent and selective tyrosine kinase inhibitor (1, 2). It was initially developed as an inhibitor of Bcr-Abl, the protein product of the Philadelphia chromosome (Ph) characteristic of chronic myelogenous leukemia (CML). The activity of imatinib in CML has made it the current standard therapy in this disease. Among patients who failed prior therapy with IFN-α, 45% to 60% of patients achieve a complete cytogenetic remission, and 80% of patients remain alive and free of progression after 4 years (35). A randomized trial in patients with previously untreated CML in early chronic phase showed the higher efficacy and better toxicity profile of imatinib compared with IFN-α (6). Over 75% of these patients achieved a complete cytogenetic remission with imatinib, >95% remained alive and free of progression at 18 months (68). Higher doses of imatinib improved the complete cytogenetic remission rates to 90% both in patients who failed prior IFN-α therapy (9) and in those previously untreated (10, 11).

As the rates of complete cytogenetic remission improve, monitoring of minimal residual disease by PCR has become more important. Patients are currently routinely followed with quantitative PCR techniques (e.g., real-time PCR) to determine the levels of BCR-ABL transcripts during the course of therapy (12). The prognostic significance of levels of minimal residual disease determined by PCR have been established in CML after bone marrow transplantation and IFN-α therapy (1315). The clinical significance of molecular response among patients treated with imatinib is being investigated. The International Randomized Study of Interferon and STI571 (IRIS) trial showed that a significantly higher proportion of patients who were in complete cytogenetic remission at 6 months had at least a 3-log reduction of BCR-ABL/BCR levels if treated with imatinib compared with those treated with IFN-α (42% versus 13%, P = 0.03; ref. 16). Patients with at least a 3-log reduction of BCR-ABL/BCR levels after 12 months of therapy had a significantly better probability of disease-free survival compared with those in complete cytogenetic remission but with a less than 3-log reduction of BCR-ABL/BCR (16). This data has not been confirmed in independent series, and additional questions remain, including the factors predictive of a molecular response, the durability of response, and the significance of increasing transcript levels.

We analyzed the results of molecular monitoring in patients with CML in chronic phase treated with imatinib at our institution to determine the frequency of molecular response to imatinib and the long-term prognostic implications of molecular responses.

From December 1999 to August 2003, 377 patients with chronic phase CML were treated with imatinib. These included 190 patients receiving imatinib after failing therapy with IFN-α, and 187 who received imatinib as their first line of therapy for CML. All patients receiving at least one dose of imatinib were included in this analysis. The definitions of chronic phase and IFN-α failure were the same as those used in published studies using imatinib (3, 4, 7). Briefly, patients were considered to be in chronic phase if they had blasts <15%, blasts plus promyelocytes <30%, basophils <20%, platelets ≥100 × 109/L and no evidence of extramedullary disease. Patients with cytogenetic clonal evolution but without other features of accelerated phase were included in this analysis. IFN-α failure was defined as follows: hematologic failure included hematologic resistance (failure to achieve complete hematologic response after ≥6 months of IFN-α) or relapse (disease recurrence after achieving complete hematologic response), cytogenetic failure included resistance (Ph ≥65% after at least 12 months of IFN-α), or relapse (Ph increase >30% documented on two occasions, or a single increase to ≥65%); and intolerance defined as grades 3 to 4 non–hematologic toxicity (according to the National Cancer Institute Common Toxicity Criteria) not responding to adequate management.

Evaluation before and during treatment. Before the start of treatment, patients were evaluated with history an physical exam, complete blood cell count (CBC) with differential, and blood chemistry including total bilirubin, creatinine, and alanino aminotransferase. All patients had a pretreatment bone marrow evaluation for morphology and cytogenetic analysis, fluorescent in situ hybridization, and real-time PCR. After treatment was started, patients were evaluated with complete blood count and blood chemistry weekly during the first 1 to 3 months, then every 2 to 6 weeks. Bone marrow aspirations for morphology, cytogenetics (fluorescent in situ hybridization when cytogenetic analysis inevaluable) and PCR were repeated every 3 to 4 months for the first year, and every 6 months thereafter. Patients were followed for survival at least every 3 months. Drug toxicity was evaluated at each visit and graded according to the National Cancer Institute Common Toxicity Criteria (version 2.0).

Response criteria. Response criteria were previously described (3). Briefly, a complete hematologic remission was defined as a WBC count of <10 × 109/L, a platelet count of <450 × 109/L, no immature cells (blasts, promyelocytes, myelocytes) in the peripheral blood, and disappearance of all signs and symptoms related to leukemia (including palpable splenomegaly) lasting for at least 4 weeks. A complete hematologic remission was further categorized by the best cytogenetic remission as complete (0% Ph-positive), partial (1-34% Ph-positive), and minor (35-90% Ph-positive). A major cytogenetic remission included complete plus partial cytogenetic remissions (i.e., Ph-positive <35%). Cytogenetic remission was judged by standard cytogenetic analysis; fluorescent in situ hybridization was used only when routine cytogenetic analysis was unanalyzable (i.e., insufficient metaphases). For the purpose of this analysis, a major molecular response was defined as a BCR-ABL/ABL ratio of <0.05%, a level that has been found to correlate with long-term remission after IFN-α-based therapy (14). A complete molecular response was defined as undetectable levels of BCR-ABL confirmed by nested PCR.

Cytogenetic analysis, fluorescent in situ hybridization, and PCR. Cytogenetic analysis was done in bone marrow cells by the G-banding technique. For chromosome analysis, at least 20 metaphases were analyzed and bone marrow specimens were examined on direct or short-term (24 hours) cultures. Bone marrow cells were analyzed by fluorescent in situ hybridization using the LSI BCR/ABL dual color extra-signal probe according to the manufacturer's instructions (Vysis, Inc., Downers Grove, IL).

BCR-ABL transcript levels was determined by real-time quantitative reverse transcription PCR (qRT-PCR), with negative results (i.e., undetectable transcript) confirmed by nested PCR. Total RNA from bone marrow aspirate (1-2 mL) and peripheral blood (10 mL) was isolated using Trizol reagent (Invitrogen Life Technologies, Gaithersburg, MD). The integrity of RNA was documented by gel electrophoresis prior to reverse transcription with samples rejected if rRNA bands were not visible. Reverse transcription was done using up to 14 μg of total RNA at a concentration of 0.5 μg/μL using Superscript II RT (Invitrogen). cDNA (5.0 μL representing ∼1 μg of total RNA) was used as a template in a 25 μL qRT-PCR reaction using primers specific for BCR-ABL and ABL. Assays were done using the ABI PRISM 7700 Sequence Detection System (Applied Biosystems, Foster City, CA).

A multiplex qRT-PCR assay was done in a single tube to simultaneously detect b2a2, b3a2, and e1a2 transcripts as well as total abl as a normalizing transcript, with each sample amplified in duplicate. Primers used are listed in Table 1, as are the fusion and normalizing abl detection probes which were labeled with 6-carboxyfluorescein (FAM) and VIC (ABI) fluorochromes, respectively. By mixing experiments with bcr-abl + cell lines (KBM-7, b2a2; K562, b3a2; and B15, e1a2), we had optimized the primer concentrations to allow for accurate quantitation of each of the fusion transcripts over the range of the assay. The bcr-abl levels for each sample were expressed as a percentage of BCR-ABL to ABL. For each run, the t(9;22)-positive cell lines KBM7, K562, and B15, that carry b2a2, b3a2, and e1a2 fusion genes, respectively, served as positive controls. The HL60 cell line was used as a negative control.

Table 1.

Sequence of primers and probes used in the TaqMan qRT-PCR

PrimersFinal concentration (nmol/L)
bcr-e1 400 GCAAGACCGGGCAGATCT 
bcr-b2 400 ACAGCATTCCGCTGACCAT 
abl-a2 400 NEDR-CGAGCGGCTTCACTCAGA 
abl 10-F 50 AGAGCTGCAGAGCACAGAGACA 
abl 11-R 50 GCTCTTTTCGAGGGAGCAATG 
   
TaqMan probes   
    bcrabl-2a 200 6FAMR-CCTGAGGCTCAAAGTCAGATGCTACTGG-TAMRA 
    abl 1011-P 100 VIC-ATGGTCCAGAGGATCGCTCTCTCCCT-TAMRA 
PrimersFinal concentration (nmol/L)
bcr-e1 400 GCAAGACCGGGCAGATCT 
bcr-b2 400 ACAGCATTCCGCTGACCAT 
abl-a2 400 NEDR-CGAGCGGCTTCACTCAGA 
abl 10-F 50 AGAGCTGCAGAGCACAGAGACA 
abl 11-R 50 GCTCTTTTCGAGGGAGCAATG 
   
TaqMan probes   
    bcrabl-2a 200 6FAMR-CCTGAGGCTCAAAGTCAGATGCTACTGG-TAMRA 
    abl 1011-P 100 VIC-ATGGTCCAGAGGATCGCTCTCTCCCT-TAMRA 

A plasmid containing cloned sequences spanning abl exons 10 and 11 was used for generating the ABL standard curve. A plasmid containing clone BCR-ABL fusion sequences derived from K562 was used to generate the BCR-ABL standard curve. A 5-log dilution of each of these plasmid standards were included in each run as a test for assay sensitivity across the dynamic range. Dilutions of K562 RNA (1:10,000 and 1:100,000) into HL-60 RNA were also included in each run as a control for reverse transcription and PCR conditions. The sensitivity of each run was confirmed to be in the range of 1:100,000. PCR results were rejected if the sensitivity of the run was <10,000 or if the total level of normalizing ABL transcript in any given sample was <10,000 copies. This value sets a lower limit of sensitivity of 1:10,000 for detection of BCR-ABL transcripts in posttreatment samples. Interassay variability, assayed by repeated BCR-ABL/ABL determination starting from RNA from control samples, was estimated to be between 15% and 40% over the range of the assay, with highest variability noted at the high and low BCR-ABL copy numbers.

To determine the type(s) of BCR-ABL fusion transcripts that were amplified by qRT-PCR, capillary electrophoresis was subsequently done on all samples. The products were detected due to the abl exon 2 primer in the qRT-PCR reaction being labeled at the 5′ end with NED fluorescent dye (ABI). Specimens that were inadequate or negative for BCR-ABL fusion transcript detection by qRT-PCR/capillary electrophoresis were subsequently tested by nested PCR and then quantitated by competitive PCR, as previously reported (17, 18). The sensitivity of the nested PCR assay, established by dilution of control RNA samples is approximately 1:100,000 to 1:1,000,000. Given the differing methodologies, these values were not regarded as directly comparable to the qRT-PCR values and were thus not used for the statistical analyses below. The qRT-PCR methodology described above has been used routinely since October 2001. Before this date, a different methodology was used and the results cannot be extrapolated to current values. Thus, for consistency, only those results obtained with this methodology are considered for this analysis.

Statistical analysis. Complete cytogenetic remission duration was considered from the time a complete cytogenetic remission was first achieved to the time when any Ph-positive metaphases were first detected again. Survival was calculated from the time the treatment began until death from any cause, or last follow-up. The probability of remission duration was estimated using the Kaplan-Mier product-limit method (19) and compared by means of the log-rank test (20). Associations between categorical variables were assessed via cross-tabulation and Fisher's exact test or the χ2 test (21). Multivariate logistic regression analysis was used to assess the relationship between various predictors of interest and molecular response.

Patients. A complete hematologic remission was achieved in 293 of 305 (96%) patients treated with imatinib for CML in active chronic phase (i.e., abnormal peripheral blood counts and/or splenomegaly). These included 177 of 181 (98%) in early chronic phase (i.e., within 12 months from diagnosis), and 116 of 124 (94%) in chronic phase after IFN-α failure. A major cytogenetic remission was achieved in 310 of 369 (84%) patients who had Ph-positive metaphases by standard cytogenetic analysis at the start of therapy, including 279 (76%) that achieved a complete cytogenetic remission. An additional seven patients had Ph-negative BCR-ABL-positive disease at the start of therapy. Among 286 patients in complete cytogenetic remission, 280 had at least one PCR test done for follow-up. These patients constitute the focus of this analysis. The median age of the study group was 50 years (range, 15-84 years). Among them, 117 (42%) patients received imatinib after IFN-α failure and 163 (58%) as first line of therapy. The starting dose of imatinib was 400 mg daily for 144 (51%) patients and 800 mg for 136 (49%) patients. The median BCR-ABL/ABL ratio before treatment with imatinib was started was 39.44% (range, 0.252-170.53%). The median follow up from the time treatment was started is 31 months (range, 3-52 months). Five patients have died while on therapy with imatinib, with an estimated overall survival of 95% at 48 months. Transformation while on therapy occurred in two patients.

Molecular response. A major molecular response was achieved in 174 patients (62%; 46% of all patients treated) and 95 patients (34%; 25% of all patients treated) had at least one analysis showing undetectable BCR-ABL (i.e., complete molecular response). Among these, 81 have had subsequent PCR analyses and 44 (54%) have had at least two consecutive undetectable assays. The median time from start of treatment to achievement of a major molecular response was 10 months (range, 2.8-46 months), and 16.7 months (range, 3-48 months) for achievement of a complete molecular response. However, as shown in Fig. 1, these responses may occur late and there is no clear evidence of a plateau for time to molecular response.

Fig. 1.

Cumulative probability of achieving a molecular response on imatinib for 280 patients with CML in chronic phase who achieved a complete cytogenetic remission on imatinib. All times are calculated from the start of therapy with imatinib.

Fig. 1.

Cumulative probability of achieving a molecular response on imatinib for 280 patients with CML in chronic phase who achieved a complete cytogenetic remission on imatinib. All times are calculated from the start of therapy with imatinib.

Close modal

We then analyzed the pretreatment clinical characteristics associated with achieving a molecular response. Table 2 shows the clinical characteristics at the start of imatinib therapy for all 280 patients. Characteristics associated with a higher probability of achieving a major molecular response included a lower percentage of peripheral blood blasts, Sokal low-risk group, treatment with high-dose imatinib, and treatment in early chronic phase. Achievement of undetectable levels of BCR-ABL was associated with Ph-positive metaphases ≤90% at the start of therapy. In a multivariate analysis, treatment with high-dose imatinib was the only variable independently associated with an increased probability of achieving a major molecular remission (P = 0.02). Multivariate analysis for complete molecular remission identified a lower percentage of Ph-positive metaphases (<90%) as the only variable associated with this response (P = 0.01); treatment with high-dose imatinib was marginally significant (P = 0.08).

Table 2.

Associations of patient and disease characteristics with molecular response (n = 280)

VariablesCategorynMMRPCMRP
Age (y) <60 199 122 (61) 0.75 65 (33) 0.57 
 ≥60 81 52 (64)  30 (37)  
Splenomegaly No 232 148 (64) 0.25 77 (33) 0.62 
 Yes 48 26 (54)  18 (38)  
Hemoglobin (g/dL) <12 110 63 (57) 0.23 36 (33) 0.80 
 ≥12 169 110 (65)  59 (35)  
WBC (× 109/L) ≤20 159 97 (61) 0.39 61 (38) 0.20 
 20.1-50 64 44 (69)  17 (26)  
 >50 56 32 (57)  17 (30)  
Platelets (× 109/L) ≤450 205 120 (58) 0.06 63 (31) 0.08 
 >450 75 54 (72)  32 (43)  
Peripheral basophils (%) <7 238 151 (63) 0.30 79 (33) 0.60 
 ≥7 42 23 (55)  16 (38)  
Peripheral blasts (%) 0-1 254 163 (64) 0.04 89 (35) 0.35 
 18 9 (50)  5 (28)  
 ≥3 2 (25)  1 (12)  
Marrow basophils (%) <4 220 140 (64) 0.40 79 (36) 0.22 
 ≥4 60 34 (57)  16 (27)  
Marrow blasts (%) <5 261 166 (64) 0.08 91 (35) 0.32 
 ≥5 19 8 (42)  4 (21)  
Cytogenetic clonal evolution No 258 159 (62) 0.82 84 (32) 0.23 
 Yes 21 14 (67)  10 (48)  
Sokal risk* Low 98 76 (78) 0.008 34 (35) 0.53 
 Intermediate 69 41 (59)  26 (38)  
 High 34 18 (53)  9 (26)  
% Ph at pretreatment ≤90 56 39 (70) 0.28 27 (48) 0.02 
 >90 221 134 (61)  67 (30)  
Duration of chronic phase (months) <12 175 112 (64) 0.49 54 (31) 0.30 
 12-35 65 36 (55)  27 (42)  
 ≥36 40 26 (65)  14 (35)  
Study group ECP 163 110 (67) 0.04 52 (32) 0.47 
 IFN failure 117 64 (55)  43 (37)  
Starting dose of imatinib (mg) 400 144 77 (53) 0.003 42 (29) 0.11 
 800 136 97 (71)  53 (39)  
VariablesCategorynMMRPCMRP
Age (y) <60 199 122 (61) 0.75 65 (33) 0.57 
 ≥60 81 52 (64)  30 (37)  
Splenomegaly No 232 148 (64) 0.25 77 (33) 0.62 
 Yes 48 26 (54)  18 (38)  
Hemoglobin (g/dL) <12 110 63 (57) 0.23 36 (33) 0.80 
 ≥12 169 110 (65)  59 (35)  
WBC (× 109/L) ≤20 159 97 (61) 0.39 61 (38) 0.20 
 20.1-50 64 44 (69)  17 (26)  
 >50 56 32 (57)  17 (30)  
Platelets (× 109/L) ≤450 205 120 (58) 0.06 63 (31) 0.08 
 >450 75 54 (72)  32 (43)  
Peripheral basophils (%) <7 238 151 (63) 0.30 79 (33) 0.60 
 ≥7 42 23 (55)  16 (38)  
Peripheral blasts (%) 0-1 254 163 (64) 0.04 89 (35) 0.35 
 18 9 (50)  5 (28)  
 ≥3 2 (25)  1 (12)  
Marrow basophils (%) <4 220 140 (64) 0.40 79 (36) 0.22 
 ≥4 60 34 (57)  16 (27)  
Marrow blasts (%) <5 261 166 (64) 0.08 91 (35) 0.32 
 ≥5 19 8 (42)  4 (21)  
Cytogenetic clonal evolution No 258 159 (62) 0.82 84 (32) 0.23 
 Yes 21 14 (67)  10 (48)  
Sokal risk* Low 98 76 (78) 0.008 34 (35) 0.53 
 Intermediate 69 41 (59)  26 (38)  
 High 34 18 (53)  9 (26)  
% Ph at pretreatment ≤90 56 39 (70) 0.28 27 (48) 0.02 
 >90 221 134 (61)  67 (30)  
Duration of chronic phase (months) <12 175 112 (64) 0.49 54 (31) 0.30 
 12-35 65 36 (55)  27 (42)  
 ≥36 40 26 (65)  14 (35)  
Study group ECP 163 110 (67) 0.04 52 (32) 0.47 
 IFN failure 117 64 (55)  43 (37)  
Starting dose of imatinib (mg) 400 144 77 (53) 0.003 42 (29) 0.11 
 800 136 97 (71)  53 (39)  

Abbreviations. MMR, major molecular response; CMR, complete molecular response; ECP, early chronic phase.

*

Two hundred and one patients evaluable.

This analysis does not take into account the time difference in follow-up between patients. Thus, to adjust for the time to achieving molecular response, we repeated the multivariate analysis considering achieving a major or complete molecular remission at 12 months as the end point. For this analysis, we focused on patients treated in early chronic phase as this is the setting where most patients are currently being treated with imatinib, and for these patients, the percentage of Ph-positive metaphases <90% rarely applies. In this analysis, treatment with high-dose imatinib was the only variable associated with an increased probability of achieving a major (P = 0.01) or complete molecular response (P = 0.02).

Durability of cytogenetic response by molecular response. We then investigated the long-term clinical significance of achieving a molecular response (Table 3). Because of the small number of deaths and transformations during therapy, the analysis of the clinical implications of molecular response focused on duration of complete cytogenetic remission. Patients achieving a major molecular response were followed for a median of 31 months (range, 7.5-52 months) from the start of imatinib therapy. Eight patients achieved a major molecular response only on their last evaluation and therefore have no follow-up at the time of this report. Among the 166 patients who had at least one additional cytogenetic analysis after achieving a major molecular response, 9 (5%) lost their complete cytogenetic remission, compared with 25 of 68 (37%) who did not achieve a major molecular response (P = 0.0001). Two and four patients eventually re-gained a complete cytogenetic remission, respectively (one after increasing the dose of imatinib, all others without additional intervention). Patients with undetectable levels of BCR-ABL were followed for a median of 14 months (range, 3-27 months) after reaching this milestone, not considering 13 who became undetectable only on their latest follow-up evaluation. Among the 82 patients with subsequent evaluations, only 3 (4%) lost their complete cytogenetic remission (one of them later regained a complete cytogenetic remission), compared with 5 of 74 (7%) who had detectable BCR-ABL levels but <0.05%. These differences are not due to a difference in follow up, as there is no difference in follow-up time between patients who did and did not achieve a major or complete molecular response.

Table 3.

Significance of achieving molecular response in sustaining a complete cytogenetic remission

Best Bcr-Abl/Abl ratioNo. evaluableNo. who lost complete cytogenetic remission to
PMedian follow-up from molecular response (range)
PR/Minor/>90%Total (%)
≥0.05% 68 12 + 4*/5+1*/3 25 (37) <0.0001 11.2 (3.0-28.9) 
<0.05% 166 6 + 2*/0/1 9 (5)  18.4 (3-33.4) 
>0 and <0.05% 74 5/0/0 5 (7) 0.48 15.0 (3.4-30.6) 
Undetectable 82 1 + 1*/0/1 3 (4)  14.3 (3-27) 
Best Bcr-Abl/Abl ratioNo. evaluableNo. who lost complete cytogenetic remission to
PMedian follow-up from molecular response (range)
PR/Minor/>90%Total (%)
≥0.05% 68 12 + 4*/5+1*/3 25 (37) <0.0001 11.2 (3.0-28.9) 
<0.05% 166 6 + 2*/0/1 9 (5)  18.4 (3-33.4) 
>0 and <0.05% 74 5/0/0 5 (7) 0.48 15.0 (3.4-30.6) 
Undetectable 82 1 + 1*/0/1 3 (4)  14.3 (3-27) 
*

Patients lost complete cytogenetic remission only transiently.

To further define the long-term prognostic implications of molecular response and account for the potential lead-time bias, we investigated the complete cytogenetic remission duration according to molecular response at different time points. For this analysis, only patients who had both a molecular analysis and were still on therapy and in complete cytogenetic remission at the specified time points were evaluable. Patients who achieved a major molecular remission by 12 months after the start of therapy have significantly longer cytogenetic remission durations than those without this magnitude of molecular response (Fig. 2C). Molecular response at earlier time points (i.e., 3 and 6 months) shows a similar trend (Fig. 2A and B). Most patients who lost their complete cytogenetic remission still have at least a partial cytogenetic remission. Thus, we repeated the analysis considering only loss to at least 35% Ph-positive metaphases. Because of the few number of events, the results are not statistically significant. In fact, the analysis could not be done for response at 12 months as no patients lost their major cytogenetic response in either group (with or without major molecular response). A trend could be observed in favor of the earlier responders at 3 (P = 0.29) and 6 months (P = 0.11; data not shown).

Fig. 2.

Landmark analysis of duration of complete cytogenetic remission according to the molecular response at (A) 3 months, (B) 6 months, and (C) 12 months. Patients are grouped based on their BCR-ABL/ABL ratio <0.05% versus ≥0.05% at the specified time point. An event is considered the recurrence of any Ph-positive metaphases after achieving a complete cytogenetic remission. For this analysis, the starting point in the horizontal axis is the time when patients achieved complete cytogenetic remission.

Fig. 2.

Landmark analysis of duration of complete cytogenetic remission according to the molecular response at (A) 3 months, (B) 6 months, and (C) 12 months. Patients are grouped based on their BCR-ABL/ABL ratio <0.05% versus ≥0.05% at the specified time point. An event is considered the recurrence of any Ph-positive metaphases after achieving a complete cytogenetic remission. For this analysis, the starting point in the horizontal axis is the time when patients achieved complete cytogenetic remission.

Close modal

Other studies used a 3-log reduction rather than an absolute value as a measure of molecular response for patients treated with imatinib (16). We thus repeated the analysis using a 3-log reduction as a measure of major molecular response. For this purpose, published reports have used the median calculated from measuring pretreatment values from a small number of patients as baseline levels. For our analysis we calculated a 3-log reduction in two different ways. First, we used the median of the total population (i.e., 39.44%) as the baseline value. Thus, a 3-log reduction would be considered achieving a ratio of ≤0.039%. The results are nearly identical to those considering a value <0.05% as a major molecular response. We then used each individual patients' pretreatment BCR-ABL/ABL value as their own baseline to determine log reduction. Data for this analysis is available for 79 patients. Although the difference is not statistically significant, the same trend is observed in favor of patients achieving a molecular response at each of these early time points.

Since we identified that achieving a major molecular response after 12 months of therapy is predictive of cytogenetic remission duration, we investigated the probability of achieving this response at 12 months based on the response achieved at 3 months. Patients who achieve ≤1-log reduction after 3 months of therapy have a significantly lower probability (55%) of achieving a major molecular response at 24 months, compared with those with greater than 1-log or 2-log reduction (84% and 95%, respectively; P = 0.0002).

Durability of molecular response. We next investigated the durability of molecular responses. Among the 174 patients who achieved a major molecular response, 165 (95%) have had repeated molecular testing (median, five follow-up tests, range, two to eight). The molecular response was sustained in 118 (72%) of them, for a median of 19 months (range, 3-33 months). Subsequent evaluation is available for 81 of the 95 (85%) patients who achieved undetectable levels of BCR-ABL, with a median of two follow-up tests (range, one to six). In 43 patients (53%), BCR-ABL/ABL became detectable again in subsequent testing, whereas 38 (47%) had sustained undetectable BCR-ABL levels over a median of 18 months (range, 3-27 months). Among 47 patients who lost a major molecular response, 13 (28%) did so after having therapy interrupted temporarily or permanently. The most common reasons for treatment interruptions were noncompliance in six, toxicity in five [liver (n = 3), gastrointestinal (n = 1), and weight gain (n = 1)], and hospitalization for other reasons (n = 2).

To determine the significance of an increasing BCR-ABL/ABL ratio while on therapy, we investigated the clinical significance of a <1-log (n = 23), 1-log (n = 30), or 2-log (n = 8) increase in the BCR-ABL/ABL ratio from the lowest level achieved. Only four (17%), four (13%), and two (25%) patients with each of these magnitudes of increasing transcript levels, respectively, lost their complete cytogenetic remission (P = 0.72). However, 9 of the 10 patients who lost the response had the lowest BCR-ABL/ABL ratio of >0.05, whereas only 1 of 37 patients with nadir levels of ≤0.05% had lost their response, regardless of their log increase (P = 0.04). If the absolute increase is considered instead of the log increase, an increment of BCR-ABL/ABL of more than 1.0 percentage point from the nadir is associated with an increased risk of cytogenetic relapse (5 of 11, 45%) compared with 6 of 33 (18%) of those with percentage point increments of 0.05 to 1, and 0 of 44 of those with increments of <0.05 (P = 0.0001). Again, patients with nadir levels of ≥0.05 had the highest probability of relapse.

Imatinib has become the standard therapy for patients with CML. A complete cytogenetic remission is achieved in 75% to 90% of patients treated with imatinib in early chronic phase (6, 7, 11). With standard-dose imatinib, ∼40% of patients have been reported to achieve at least a 3-log reduction in BCR-ABL/BCR levels, a reduction associated with an increased probability of progression-free survival (16). In these studies, 4% to 10% of patients have achieved undetectable levels of BCR-ABL (16). Studies using higher doses of imatinib have reported major molecular responses (i.e., 3-log reduction or <0.05%) in ∼65% of patients, and undetectable levels in 35% to 40% (9, 11). In this study, we investigated the clinical significance of these molecular responses among patients in chronic phase treated with imatinib. Among 280 patients who achieved a complete cytogenetic remission with imatinib, 174 (62%) achieved a major molecular response, and 95 (34%) had undetectable BCR-ABL transcripts at least once. It should be recognized that variations in the assay sensitivity may account for differences in the rate of patients with undetectable transcripts among different reports. The relevance of the sensitivity of the assay is further emphasized by the fact that approximately half of the patients that had undetectable transcript levels on at least one occasion later had low levels detectable. Similar results have been reported from a subset of patients from the IRIS trial. In that study, 10 of 26 (38%) patients had undetectable transcripts on at least one occasion, but only 4 (14%) had consistently undetectable levels for more than 6 months (22).

Treatment with high-dose imatinib (i.e., 800 mg) was the most significant factor associated with in increased probability of achieving a molecular response, particularly at earlier time points (i.e., 12 months). However, the overall rate of major and complete molecular response is not different between patients treated with standard versus high-dose imatinib. Because the follow-up is significantly longer for patients treated with standard dose and patients may continue to improve their cytogenetic and molecular response with continuation of therapy, further follow-up is required to determine whether there is an absolute difference in molecular response with high-dose imatinib or if it is a means of getting earlier to the same end point.

The definition of molecular responses is still evolving. For this analysis, we considered a major molecular response as reaching an absolute value of BCR-ABL/ABL ratio <0.05%, a value that has been found predictive of duration of cytogenetic response after IFN-α-based therapy by our group and others. Interestingly, it is also the median value after 12 months of therapy for the study population in this report. Other studies have reported the log decrease of BCR-ABL transcripts and considered a 3-log reduction as a major response associated with improved outcome (16). The studies have used a standardized baseline value for patients derived from the median value obtained from 30 patients studied before the start of therapy (16). Thus, it was not necessary to know the BCR-ABL level of a patient at baseline to calculate the subsequent reduction (16). The median baseline value for our patient population was 39.44. If we were to use a 3-log reduction from this value instead of an absolute ratio of <0.05% to determine a major molecular response, only five patients would not be considered to have achieved a major molecular response, and the results of the analysis would be similar.

Our results suggest that major (i.e., BCR-ABL/ABL <0.05%) or complete molecular remissions (i.e., undetectable BCR-ABL) were associated with more durable cytogenetic remissions. Only 5% of patients who have achieved a major molecular response and 4% of those reaching undetectable levels of BCR-ABL/ABL have lost their complete cytogenetic remission, compared with 37% of patients not reaching these degrees of response (P < 0.0001). This difference is not due to a lead-time bias as patients in both groups were followed for a similar period of time. A similar trend was reported in a smaller series by Rosti et al. (23). One of 18 patients who had a 2-log reduction in BCR-ABL/B2M ratio (6%) lost a complete cytogenetic remission, compared with 3 of 9 patients who had a <2-log reduction.

The importance of achieving an early molecular response was first reported from the IRIS trial. Among patients who achieved a complete cytogenetic remission after 12 months of therapy, those with at least a 3-log reduction in BCR-ABL transcript levels had a significantly better progression-free survival compared with those with less than a 3-log reduction (16). Other smaller series' and subsets of the larger IRIS cohort have reported that early declines in transcript levels are associated with achievement of cytogenetic response (2426). In our series, patients who achieved a major molecular remission by 12 months after the start of therapy had an improved probability of a sustained complete cytogenetic remission. A similar trend was seen at earlier time points (i.e., 3 and 6 months) but it was not statistically significant, probably because of the relatively small number of patients who reach these levels at earlier time points. However, if patients are divided by quartiles of BCR-ABL transcript levels at 3 months, those with the lowest levels (i.e., <0.0847) have a significantly better probability of a sustained cytogenetic remission. Branford et al. (27) reported similar results for patients reaching a 2-log reduction at 6 months. Patients with greater than a 1-log reduction (our series) or 2-log reduction (27) at 3 months have an improved probability of later achieving a molecular response. These results emphasize the need for frequent and early molecular monitoring of patients treated with imatinib, and the importance of achieving the best response as early as possible.

There is little available data on the durability of molecular responses. In our study, major molecular responses have been sustained in 72% of patients, whereas complete responses have been sustained in nearly half of the patients. In nearly a third of these patients, the cause for loss of response was treatment interruption. Despite the loss of molecular responses, this has not translated into loss of cytogenetic remissions in most patients. An important question then is the significance of increasing levels of BCR-ABL transcripts during therapy. Our results suggest that there is a high risk of loss of cytogenetic remission with greater increases of measurable molecular disease (i.e., >2-log increase or an absolute increase >1), but the risk is particularly high for patients who did not achieve a major molecular remission. However, it is possible that patients that achieved lower transcript levels will only require longer times to show a loss of their cytogenetic response. Continued follow-up will answer this question. Different treatment strategies, such as adding other agents to imatinib, should be investigated in these cases of increasing molecular disease.

In summary, molecular monitoring provides valuable information for the follow-up of patients with CML in chronic phase treated with imatinib. Achieving a major molecular response correlated with an improved probability of a durable cytogenetic remission, particularly when achieved early during the course of therapy. Most major molecular remissions have been durable, and increases in BCR-ABL transcripts have not translated in loss of cytogenetic remission for most patients with molecular response. Thus, a major goal of therapy with imatinib should be to achieve an early major molecular response.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: J. Cortes is a Clinical Research Scholar for the Leukemia and Lymphoma Society.

1
Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells.
Nat Med
1996
;
2
:
561
–6.
2
Beran M, Cao X, Estrov Z, et al. Selective inhibition of cell proliferation and BCR-ABL phosphorylation in acute lymphoblastic leukemia cells expressing Mr 190,000 BCR-ABL protein by a tyrosine kinase inhibitor (CGP-57148).
Clin Cancer Res
1998
;
4
:
1661
–72.
3
Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia.
N Engl J Med
2002
;
346
:
645
–52.
4
Kantarjian HM, Talpaz M, O'Brien S, et al. Imatinib mesylate for Philadelphia chromosome-positive, chronic-phase myeloid leukemia after failure of interferon-α: follow-up results.
Clin Cancer Res
2002
;
8
:
2177
–87.
5
Kantarjian H, O'Brien S, Cortes J, et al. Survival advantage with imatinib mesylate therapy in chronic-phase chronic myelogenous leukemia (CML-CP) after IFN-α failure and in late CML-CP, comparison, with historical controls.
Clin Cancer Res
2004
;
10
:
68
–75.
6
O'Brien SG, Guilhot F, Larson RA, 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.
7
Kantarjian HM, Cortes JE, O'Brien S, et al. Imatinib mesylate therapy in newly diagnosed patients with Philadelphia chromosome-positive chronic myelogenous leukemia: high incidence of early complete and major cytogenetic responses.
Blood
2003
;
101
:
97
–100.
8
Cervantes F, on behalf of the IRIS (International Randomized IFN vs. STI571) Study Group. Durability of responses to imatinib in newly diagnosed chronic-phase chronic myeloid leukemia (CML): 24-month update from the IRIS Study [abstract #633]. Blood 2003.
9
Cortes J, Giles F, O'Brien S, et al. Result of high-dose imatinib mesylate in patients with Philadelphia chromosome-positive chronic myeloid leukemia after failure of interferon-α.
Blood
2003
;
102
:
83
–6.
10
Hughes T, Branford S, Matthews J, et al. Trial of higher dose imatinib with selective intensification in newly diagnosed CML patients in the chronic phase [abstract #95].
Blood
2003
;
102
:
31a.
11
Kantarjian H, Talpaz M, O'Brien S, et al. High-dose imatinib mesylate therapy in newly diagnosed Philadelphia chromosome-positive chronic phase chronic myeloid leukemia.
Blood
2004
;
103
:
2873
–8.
12
Guo JQ, Lin H, Kantarjian H, et al. Comparison of competitive-nested PCR and real-time PCR in detecting BCR-ABL fusion transcripts in chronic myeloid leukemia patients.
Leukemia
2002
;
16
:
2447
–53.
13
Hochhaus A, Weisser A, La Rosee P, et al. Detection and quantification of residual disease in chronic myelogenous leukemia.
Leukemia
2000
;
14
:
998
–1005.
14
Hochhaus A, Reiter A, Saussele S, et al. Molecular heterogeneity in complete cytogenetic responders after interferon-α therapy for chronic myelogenous leukemia: low levels of minimal residual disease are associated with continuing remission. German CML Study Group and the UK Medical Research Council CML Study Group.
Blood
2000
;
95
:
62
–6.
15
Kantarjian HM, O'Brien S, Cortes JE, 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
–41.
16
Hughes TP, Kaeda J, Branford S, et al. Frequency of major molecular responses to imatinib or interferon α plus cytarabine in newly diagnosed chronic myeloid leukemia.
N Engl J Med
2003
;
349
:
1423
–32.
17
Kantarjian HM, 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
–6.
18
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.
19
Kaplan EL, Meier P. Nonparametric estimation from incomplete observations.
J Am Stat Assoc
1958
;
53
:
157
–160.
20
Peto R, Peto J. Asymptotically efficient rank invariant procedures.
J R Stat Soc [Ser A]
1972
;
135
:
185
–207.
21
Agresti A. Categorical data analysis. 7th ed. New York: John Wiley & Sons; 1972.
22
Branford S, Rudzki Z, Grigg A, et al. BCR-ABL levels continue to decrease up to 42 months after commencement of standard dose imatinib in patients with newly diagnosed chronic phase CML who achieve a major molecular response [abstract #274].
Blood
2004
;
104
:
82a
.
23
Rosti G, Martinelli G, Bassi S, et al. Molecular response to imatinib in late chronic-phase chronic myeloid leukemia.
Blood
2004
;
103
:
2284
–90.
24
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
–83.
25
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
–9.
26
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
–400.
27
Branford S, Rudzki Z, Harper A, et al. Imatinib produces significantly superior molecular responses compared to interferon α plus cytarabine in patients with newly diagnosed chronic myeloid leukemia in chronic phase.
Leukemia
2003
;
17
:
2401
–9.