High Rate of BCR-ABL Transcript Undetectability Achieved by Treating with Imatinib Mesylate Very Late CML Patients in Stable CCR after IFN.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2938-2938
Author(s):  
Alimena Giuliana ◽  
Diverio Daniela ◽  
Breccia Massimo ◽  
Mancini Marco ◽  
Giuliani Nicoletta ◽  
...  

Abstract Background. Interferon alfa (IFN a) induces complete cytogenetic response (CCR) in small proportion of CML patients, with almost all of these patients still presenting BCR-ABL transcript at the molecular level, even after prolonged period of CCR. Imatinib mesylate induces CCR in a 60–80% of patients but a high percentage of these still show residual disease as detectable by RT-PCR, and can be at risk of disease relapse. Thus combining synergistic drugs might be required to eliminate the disease effectively. Methods. We treated with imatinib 16 CML patients (8 M and 8 F) who were in very late CP and in stable CCR achieved with IFNa, but still were persistently positive at molecular level by RT-PCR. According to Sokal’s score, 11 patients were low risk (LR) and 5 were intermediate risk (IR). Median time from diagnosis was 125 mos.(r. 52–202), median duration of stable CCR was 79 mo.s (r.9–148). Imatinib was administered at the standard dose of 400 mg/die, after stopping IFN for 1 week. The level of residual disease was then monitored on BM cells at planned intervals ( baseline, 3, 6, 12 mo.s), by assaying BCR-ABL transcript at nested PCR and real-time quantitative RT-PCR; ABL was used as an internal control and results expressed as a ratio of BCR-ABL/ABL transcripts on a log scale. Results. The median transcript levels, as measured at various time points, appeared to progressively and consistently decrease in all patients with respect to the baseline values. In particular, BCR-ABL transcript undectectability was observed in 6/15 patients with evaluable analyses at 3 mo.s, in 9/16 patients analysed at 6 mo.s, and in 5/8 with available results also at 12 mo.s; of these latter patients, 4 were already negative in previous analyses and one become negative at 12 mo.s. Thus, altogether, 10/16 (62.5%) patients with at least two examinations within 12 mo.s had at least one negative molecular result. Correlations between degree of transcript reduction and clinical/biological factors detected at diagnosis and during follow-up, evidenced not significant results for age, type of transcript (either b2a3 or b3a3), baseline transcript level pre-imatinib, duration of disease and of stable CCR prior imatinib, while a trend was apparent for a higher rate of LR vs IR score among the 10 patients reaching transcript undetectability (8/10 LR vs 2/10 IR) with respect to the 6 persistently positive subjects(3 LR vs 3 IR). In present cases, imatinib was well tolerated and no side effects required drug dose reduction or discontinuation. At a median follow-up of 13 months (8–16) from start imatinib, all 16 patients are alive in CCR with progressively improving molecular response, 10 of whom with persistent transcript undetectability. Conclusion. Albeit obtained in a series of very selected patients, present results represent a further evidence stressing on the efficacy of combining imatinib and IFN a; through different, possibly complementary, mode of action, these two drugs might mutually potentiate their effect while reducing the emergence of drug resistance. The additive toxicity caused by their concurrent use might be overcome by a sequential combination; this could also be applied to patients reaching stable CCR on imatinib but still maintaining detectable residual disease.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 276-276
Author(s):  
Thoralf Lange ◽  
Thomas Bumm ◽  
Marc Mueller ◽  
Sandra Otto ◽  
Haifa K. Al-Ali ◽  
...  

Abstract Objectives: Patients with CML who achieve molecular remission (MR, defined as a RT-PCR negativity for BCR-ABL transcripts) after myeloablative stem cell transplantation (SCT) have a low risk of relapse, and the majority may be cured. The frequency of MR on imatinib varies greatly and the durability of these responses has not been reported. To investigate if MR after SCT and on imatinib are equally stable, we directly compared two cohorts of patients treated with imatinib or SCT, respectively, from the time of their first negative RT-PCR result. Patients and Methods: One hundred and forty-four CML patients in chronic (n=104) or accelerated phase (n=40) treated with standard dose imatinib were routinely monitored by conventional cytogenetics, quantitative RT-PCR (qPCR) and conventional nested PCR in case of negative qPCR results. Nineteen patients (13.2%) had at least 1 negative nested PCR. To assess the level of residual disease in patients with a single negative RT-PCR result, 10 replicate reactions were performed, each corresponding to > 106 white bone marrow cells. Thirty-six samples (median 3, range 1–4) from patients in MR on imatinib and 45 samples (median 2, range 1–3) from patients in MR after SCT were available. Twenty samples from healthy individuals were tested as controls. Results: The first negative result was noted after a median of 16.8 months (range 11.5–36.1) of imatinib therapy and 6.6 months (range 4.7–9.5) after SCT, respectively. The projected risk of molecular relapse at 12 months after the first negative RT-PCR result was 83% in patients on imatinib but only 20% in patients after SCT (P = 0.0001). Only two patients on imatinib remained in molecular remission at 13.8 and 16.6 months. While none of the patients with molecular relapse after allograft lost CCyR, one patient on imatinib progressed to cytogenetic relapse. The replicate assay was positive in 18/36 samples (50%) from patients on imatinib, 8/46 (17.4%) after allografting and 4/20 (20%) from healthy individuals. These differences were significant between patients on imatinib and after allografting (P = 0.003) and between patients on imatinib and healthy individuals (P = 0.005), but not between patients after allografting and healthy individuals (P = 0.9). Negativity by replicate testing was more stable in patients after allografting, although, even in these patients, positive replicate reactions continued to occur with longer follow-up. Conclusion: Imatinib-induced MR is usually not durable, in contrast to MR after transplant. Consistent with this, the level of residual disease in samples negative by single nested PCR is higher in patients on imatinib compared to patients after SCT. These results suggest that disease eradication with imatinib monotherapy may be rare. Patients on imatinib followed by PCR should be made aware of the fact that a single negative test does not have the same significance as in patients after SCT.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2158-2158
Author(s):  
Giuliana Alimena ◽  
Massimo Breccia ◽  
Luigia Luciano ◽  
Fabrizio Quarantelli ◽  
Daniela Diverio ◽  
...  

Abstract Imatinib mesylate was given to 26 Philadelphia positive (Ph+) chronic myeloid leukemia (CML) patients who were in late chronic phase (CP) and in stable complete cytogenetic response (CCR) after interferon-alfa (IFN-α), but showed persistent positive residual disease at PCR analysis under this treatment. At diagnosis median age was 40 years (range 21–64) and according to Sokal’s score, 18 patients were low risk and 8 were intermediate risk. Median IFN treatment was 88 mo.s (range 15–202) and median CCR duration was 73 mo.s (range 10–148). Imatinib was administered at the standard dose of 400 mg/die, after stopping IFN for 1 week. Residual disease was measured on bone marrow (BM) cells at baseline, before starting Imatinib, at 3, 6, 12, 18 mo.s and at the last follow-up (median 32 mo.s, range 21–49), by assaying BCR-ABL transcripts using quantitative PCR (RQ-PCR). The copy number (CN) of BCR/ABL and ABL transcript were derived by the interpolation of CT values to the appropriate standard curve, and the result, for each sample, was expressed as ratio of BCR/ABL mRNA copies to ABL mRNA x 100 (normalized copy number - NCN). Imatinib treatment resulted in a progressive and consistent decline of residual disease in all but one patient, from a median of 0.89 at baseline to 0.01 at the end of follow-up. Major molecular response (BCR/ABL levels <0.1) was reached in 20 patients (77%) and BCR/ABL transcripts were undetectable in 13 (50%). Achievement of molecular response was significantly correlated with post-IFN baseline transcript level (mean 1.194 for patients achieving complete molecular response vs 18,97 for those who did not; p<0.001), but not with other clinical/biological patient characteristics. In all patients, imatinib was well tolerated with no side effects requiring drug dose reduction or dose discontinuation. Albeit obtained from an unusual subset of selected patients with favourable prognosis, and likely particularly sensitive to imatinib, present results confirm the efficacy of combining Imatinib and IFN-α and further support investigating treatment approaches employing these two drugs.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2154-2154 ◽  
Author(s):  
Francois-Xavier Mahon ◽  
Francoise Huguet ◽  
Gabriel Etienne ◽  
Delphine Réa ◽  
Jean-Michel Cayuela ◽  
...  

Abstract The BCR-ABL tyrosine kinase inhibitor imatinib mesylate (Gleevec) induces complete cytogenetic responses (CCR) in more than 85% of patients with chronic myelogenous leukemia (CML). However, patients in CCR relapse after imatinib interruption in case of detectable residual disease. In fact, less than 10% of patients achieve a molecular remission, defined by an undetectable residual disease using real time quantitative polymerase chain reaction (RTQ-PCR). We previously reported the outcome of CML patients in CCR after cessation of interferon-alpha during the pre-imatinib era. Seven (all with a negative PCR) out of 15 patients did not relapse (J Clin. Oncol.,20,2002:214–220). In the present study, we address the issue of the discontinuation of imatinib in CML with undetectable residual disease for more than 2 years in 15 patients. The median duration of RTQ-PCR negativity and imatinib therapy were respectively 32 months (24–46) and 45 months (32–56) before imatinib interruption. Eight patients displayed a molecular relapse with a detectable BCR-ABL transcript appearance between the first 6 months. Imatinib was then re-introduced and led to a novel molecular response in most patients. Seven other patients have still an undetectable level of BCR-ABL transcript after a median follow up of 20 months (9–24). With the assumption that the doubling time of a proliferative CML cell is 8 days, it will take a maximum of 6 months if only one leukemic cell persists and proliferates to reach 107 cells i.e corresponding to a residual disease detectable by RTQ-PCR. Relapses observed within 6 months may reflect the kinetic of undetectable dividing CML cells. Those cells may be eradicated or controlled in long term non relapsing patients described in our study.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1052-1052 ◽  
Author(s):  
Delphine Rea ◽  
Gabriel Etienne ◽  
Selim Corm ◽  
Pascale Cony-Makhoul ◽  
Martine Gardembas ◽  
...  

Abstract In chronic phase-chronic myeloid leukemia (CP-CML), a complete cytogenetic response (CCR) along with a major molecular response (MMR) on imatinib mesylate (IM) at 400mg/d represents a strong factor predicting survival. Suboptimal cytogenetic responders (minimal or minor CR (mCR) by 6 months or partial CR (pCR) by 12 months, ELN) have a probability of further achievement of CCR of only 50%. Suboptimal molecular responders (CCR without MMR by 18 months, ELN) have a decreased probability of remaining event-free survivors when compared to optimal responders. Since non-randomized trials suggest that high-dose IM at CML diagnosis produces high rates of optimal responses, dose escalation can be recommended for suboptimal responders to standard dose of IM, but this strategy has not been yet evaluated. Here, we present the results from a series of 24 CP-CML patients who experienced IM-dose escalation for cytogenetic or molecular suboptimal response to standard doses of IM. Suboptimal cytogenetic responders (n=10) included 9 males, median age was 51.3 years-old (27.7–64.2), all were in early CP. Sokal scores were low (n=5), int (n=2), high (n=2) and unknown (n=1). All patients were treated with IM frontline at 400mg/d (n=9) or 600mg/d (n=1) and 2 received PEGIFN associated with IM at 400mg/d (withdrawn after 3 months for intolerance in 1). Prior to dose escalation, 7 patients were in pCR at 12 months and 3 in mCR at 6 months. The search for BCR-ABL mutations was negative in 6 patients tested. IM was increased to 600mg/d (n=7) or 800mg/d (n=3) after a median time of 13.7 months (5.6–15.2) on initial IM treatment. Median follow-up from IM at standard and escalated doses were respectively 28 (16.1–79.1) and 14.9 months (2.2–73.5). Of 9 patients with cytogenetic evaluation, 100% obtained CCR after a median duration of high-dose IM of 6.2 months (2.4–12.6). Five patients (50%) achieved a MMR after a median duration of high-dose IM of 9.7 months (2.9–45). Only one patient treated with PEGIFN and IM increased to 600mg/d obtained a complete molecular response (CMR) 19.9 months after high-dose IM. Suboptimal molecular responders (n=14) included 11 males, median age was 38.2 years-old (20.9–63.2), 9 were in early CP and 5 in late CP. Six had previously received IFN for a median of 4 months (4–53). Sokal scores were low (n=5), int (n=5), high (n=3) and unknown (n=1). All patients had received IM at 400mg/d, for a median duration of 27.3 months (16.7–73.3). BCR-ABL mutations were detected in 2/8 patients tested (M244V and Q252R). IM was increased to 600mg/d (n=13) or to 800mg/d (n=1). Median BCR-ABL prior to dose increase was 0.79% (0.15–3.06). Median follow-up from standard and escalated doses of IM were respectively 45.2 (26.9–85.9) and 12.5 months (3.3–38.1). Six patients (43%) obtained a MMR after a median of 6.7 months (2–25.4) of high-dose IM, including 1 with the M244V mutation. None achieved a CMR. To conclude, IM-dose escalation is beneficial to suboptimal cytogenetic responders with a rate of achievement of CCR and MMR of respectively 100 and 50%. Regarding molecular suboptimal responders, the rate of MMR after dose increase in only 43% and other strategies should be considered.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1499-1499
Author(s):  
Soo-Young Choi ◽  
Sung-Eun Lee ◽  
Yun Jeong Oh ◽  
Soo-Hyun Kim ◽  
Richard C. Woodman ◽  
...  

Abstract Background In chronic myeloid leukemia (CML), achievement of optimal responses by time point has improved long-term outcomes. In IRIS study, patients who achieved major molecular response (MMR) at 18 months had event-free survival (EFS) benefit, compared to those who achieved complete cytogenetic response (CCyR) without MMR. However, the best treatment for these patients is still not confirmed. By the previous studies, sustaining standard-dose of imatinib (IM) is expected to yield less than 20 percent of additive MMR. In this study, we investigated the efficacy of switching to nilotinib (NIL) versus high-dose IM versus standard-dose IM for patients with suboptimal molecular response to IM as first-line therapy. Methods Early chronic phase (CP) CML patients who have achieved CCyR but no MMR after at least 18 months and up to 24 months ( 18 to 24 months) on first-line IM therapy at a daily dose of 400 mg were enrolled in RE-NICE study, and informed consents were obtained from all patients. In NIL arm, patients received 400 mg BID (800 mg/day) and in high-dose IM arm, patients received 800 mg/day administrated as 400 mg BID. Primary endpoint is to evaluate the cumulative MMR rates by 12 months, and secondary endpoints are to evaluate the cumulative MMR, MR4.0 and undetectable molecular residual disease (UMRD) rates during further 24 month follow-up. The efficacy of switching to NIL or high-dose IM were compared with that of the patients who maintained standard-dose of IM. Patients with standard-dose IM were selected with the same inclusion criteria and maintained standard-dose IM after enrollment period. To compare the efficacy among three groups, MMR rate, MR4.0 and undetectable molecular residual disease (UMRD) rates by 12 months were analyzed. Safety profiles also be assessed. Patients showing lack of response (lack of complete hematologic response (CHR) at 6 months, increasing WBC, no major cytogenetic response (MCyR) at 24 months), loss of response (loss of CHR or MCyR) or severe intolerance to treatment were allowed to crossover to the alternative treatment. Results With a data cut-off date of 15 Jul 2013, a total of 52 patients were randomized into NIL arm (n = 26) or high-dose IM arm (n = 26) and 16 patients were included in standard-dose IM group. With a median follow-up of 21 months (range, 1-36) in NIL arm and high-dose IM arm, all patients have maintained CCyR without progression to advanced disease, and progressive decrease in BCR-ABL1 transcript levels was observed in all patients. With a median follow-up of 12 months (range, 1-60), all patients in standard-dose IM group have maintained CCyR without progression to advanced disease. Cumulative incidence (CI) of MMR by 12 months showed no significant difference between NIL arm and high-dose IM arm (41.1% vs 28.8, P = 0.334). Only in NIL arm, 2 in 26 (8%) achieved confirmed MR4.0 and UMRD. By 12 months, 10 in 26 (39%), 7 in 26 (27%) and 3 in 16 (19%) patients achieved MMR, in NIL arm, high-dose IM arm and standard-dose IM group respectively. Overall, the patients treated with high-dose IM showed toxicities more frequently, such as leucopenia, anemia, Thrombocytopenia, edema, fatigue, dyspnea and decreased phosphate. In addition, 14 patients in high-dose IM arm have cross-over to NIL treatment due to lack of response (n=11) and intolerance (n=3), and the median duration of NIL treatment was 23 months (range, 2-36 months). Among them, 6 (43%) patients have achieved MMR with a median NIL treatment duration of 12 months (range, 0-18). Conclusions These results demonstrate that early switching to NIL or dose escalation of IM could be recommended, considering the results of standard dose of IM in suboptimal molecular responders. When the tolerability of treatment was considered for switching to NIL or high-dose IM, NIL may be preferred. Through further clinical investigation on a large patient population and longer period observation, the efficacy and safety of early intervention of suboptimal molecular response using NIL or dose escalation of IM will be needed. Updated data with longer follow-up duration will be presented in the meeting. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2720-2720
Author(s):  
Giovanni Martinelli ◽  
Angela Poerio ◽  
Marilina Amabile ◽  
Ilaria Iacobucci ◽  
Simona Soverini ◽  
...  

Abstract Abstract 2720 Treatment strategies based on second generation tyrosine kinase inhibitors such as Nilotinib, have improved overall chronic myeloid leukemia (CML) treatment results, with a rapid and major molecular remission (MCR), at 12 months rate ranging 75%. Therefore, the detection of residual leukemic cells by measuring BCR-ABL1 transcript level is absolutely required to monitor minimal residual disease. To investigate the molecular therapeutic efficacy of nilotinib 400 mg BID in previously untreated, ECP, Philadelphia-positive CML patients, the Italian GIMEMA CML Working Party is conducting an multicenter phase II study (ClinicalTrials.gov NCT00481052). The primary endpoint is the CCgR rate at 1 year. The kinetic of molecular response is being studied by Q-PCR at baseline and after 1, 2, 3, 6, 9, 12, 18 and 24 months from treatment start by a TaqMan absolute quantitative PCR (Applied Biosystems 7900HT Fast Real-Time PCR System) and by the 12.765 Digital array (Fluidigm) which is is a nanofluidic biochip consisting in twelve panels, each containing 765 individual reaction chambers of 6 nL volume. Fluidigm analysis was done in parallel on same samples (at diagnosis,1, 2, 3, 6, 9, 12, and were possible at 18 and 24 months). Patient with longest follow up was 727 days. A Major Molecular Response, defined as a BCR-ABL:ABL ratio <0.1% according to the International Scale, and obtained by conventional RT-PCR was already achieved in 52% at 3 months, 66% at 6 months 73% at 9 months and 85% at 12 months. The median BCR-ABL transcripts level at 3, 6, 9 and 12 months was 0.063, 0.018, 0.018 and 0.006, respectively. On 73 patients with the longest follow up, a complete molecular response (CMR) defined as a BCR-ABL:ABL ratio <0.0032%IS and a negative nested PCR, was achieved in 7 patients at 12 months. The log-reduction in BCR-ABL/ABL level at 3 months was predictive of the probability of MMR at 12 months. Probability to obtain a MMR at 12 months was of 42%, 62% and 95%, respectively. The molecular results achieved in our study strongly support the notion that in early chronic phase CML patients molecular responses to nilotinib are substantially faster and “deeper” than those to IM. More rapid reduction of residual disease might help to reduce failures and to improve the late outcome of therapy. Supported by: European LeukemiaNet, AIL, AIRC, Fondazione Del Monte di Bologna e Ravenna, FIRB 2006, PRIN 2008, Ateneo RFO grants, Project of integrated program (PIO), Programma di Ricerca Regione – Università 2007 – 2009. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1918-1918 ◽  
Author(s):  
Thomas Ernst ◽  
Vivien Schäfer ◽  
Helen E White ◽  
Susanne Saussele ◽  
Thoralf Lange ◽  
...  

Abstract Approximately 2‐3% of CML patients (pts) have variant BCR‐ABL fusions transcripts that cannot be monitored by RQ‐PCR using standard methodologies. Most rare variants are accounted for by seven variant fusions (e1a2/3, b2/3a3, e6a2, e8a2, e19a2). Within the European Treatment and Outcome Study (EUTOS) we sought to establish and validate robust RQ-PCR methods for each of these abnormalities. RNA was extracted from leukocytes obtained from 20 mL peripheral blood. Samples were received either locally or by mail and spent between 1 to 3 days in transit. Multiplex reverse transcription polymerase chain reaction (RT-PCR) experiments were performed to identify BCR-ABL1 transcripts (Cross et al., Leukemia 1994). In all cases, gel electrophoresis showed an unusual band. PCR products were then sequenced to characterize BCR-ABL1 rearrangements. Monitoring of residual disease was carried out by RQ-PCR with specific forward and/or reverse primers and probes using a serial dilution of individual BCR-ABL1 or GUSB plasmid DNA calibrators. GUSB transcripts were quantified as internal control and results were expressed as the BCR-ABL1/GUSB ratio with no conversion according to the international scale (IS) as it is recommended for only typical major (M) BCR-ABL1 and cannot be applied to other transcripts in CML. 43 pts from 8 prospective studies and pts outside of studies were investigated. A total of 435 samples (1-72 per patient, median 8) were analyzed. Pts expressed 6 different atypical BCR-ABL1 transcripts (e19a2, n=18; e1a2, n=10; b2a3, n=5; b3a3, n=5; b2a3&b3a3, n=2, e8a2, n=3). At the start of the observation period, ratios BCR-ABL1/GUSB ranged between 0.01% and 179.1% (median 13.75%). Follow-up results were determined in comparison to the starting level and expressed as relative log reduction. 9 pts were in deep molecular remission considering a minimum GUSB transcript level of 20,000/reaction. During the observation period, six pts relapsed with a significant increase of BCR-ABL1/GUSB ratios. We conclude that very few pts are diagnosed with unusual BCR-ABL1 transcripts, but their characterization is important for proper assessment of treatment response in affected patients, and to avoid false negative assessment of residual disease status. Based on the individual PCR strategies, results cannot be expressed with precision on the International Scale and thus the common molecular milestones and triggers for treatment discontinuation are difficult to apply. We suggest that residual disease in these cases should be assessed primarily by considering trends in BCR-ABL/reference gene ratios over time. Disclosures Saussele: NOVARTIS PHARMA: Consultancy, Honoraria; BMS: Honoraria. Cross:Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 434-434 ◽  
Author(s):  
Daniel W. Sherbenou ◽  
Matthew J. Wong ◽  
Adil Humayun ◽  
Laura S. McGreevey ◽  
Rui Yang ◽  
...  

Abstract Background: Imatinib mesylate (IM) induces complete cytogenetic remission (CCR) in 82% of newly diagnosed patients with CML in chronic phase. Most of these patients display minimal residual disease by nested RT-PCR for BCR-ABL. A previous study suggested that kinase domain (KD) mutations in BCR-ABL may be responsible for disease persistence in a significant percentage (9/13) of CCR patients (Chu et al. Blood, 2005). As the relapse rate in this small cohort was high, we decided to investigate the incidence of KD mutants in a more representative group of patients with stable CCR. Methods: We performed nested RT-PCR for BCR-ABL in 72 CCR patients, using 2 sets of primers spanning the breakpoint and entire kinase domain, with ABL as a control gene. Mutation analysis was performed with direct sequencing and denaturing-HPLC of BCR-ABL amplicons. Median time on IM in these patients was 32 months (range, 8–65). In case of wild-type sequence, PCR products were subcloned and a median of 16 individual clones (range, 11–20) sequenced. Results: Amplification was successful in 42/72 patients (58%). We detected 11 different point mutations in 9/42 (21%), of which 3 were novel (C305S, T212R, N231D). The remaining 8 mutants are known to be IM-resistant. C305S was found only by analysis of subclones, while all other mutations were detected upon initial analysis. 5/9 patients with a mutation had a corresponding rise in BCR-ABL transcripts by qRT-PCR. In the other 4, low or undetectable levels of BCR-ABL were maintained. For 2 of these, follow-up mutation analysis detected wild-type BCR-ABL, indicating a decrease of the mutant clone below the detection threshold. For patient 5, with G250E, an initial rise in BCR-ABL was followed by a continual decrease and stable CCR. Although most of the mutants detected were expected to lead to hematologic relapse, at a median follow-up of 12 months, this occurred in only one case. Conclusions: KD mutations were detected in a modest proportion of CCR patients (12% of all patients and 21% of BCR-ABL-positive patients). The relatively high rate of PCR negativity may be due to the long amplicons (1.4kb). Our data imply that, in the majority of patients, disease persistence must be mediated by mechanisms other than KD mutations. In addition, some mutants failed to persist upon follow-up analysis, suggesting that the mutations occurred in transiently amplifying cells that are not capable of prolonged survival. Characteristics of CCR patients with a KD mutation. Patient No. Age at Study (years) Time on IM (months) CD34+ Mutation Detected CD34-/MNC Mutation Detected Time to Last Follow-up (months) Current Cytogenetic Status Follow-up Sample Mutation Status No Amp: No PCR amplification. NA: No sample available. Mutations detected by nested RT-PCR and sequencing (1), or D-HPLC (2), or both in parallel (3). Subcloning results are shown in parentases. 3 84 40 NA T212R3(15,15) 11 5% Ph+ In Progress 5 27 28 No Amp G250E3(17/17) 14 CCR G250E 19 52 18 N231D2 WT3(C305S:3/20) 5 CCR NA 20 43 43 Y253F, M244V2 M244V3(20/20) NA NA NA 21 24 63 WT3 (16/16) G321E, E355G2 NA NA NA 28 80 31 NA T315I1 (15/15) 12 CCR In Progress 30 51 39 NA Y253H1 (16/16) 10 CCR WT 31 39 43 NA T315I2 17 CCR No Amp 33 69 32 NA F359V2 5 100% Ph+ NA


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4762-4762
Author(s):  
Sang Kyun Sohn ◽  
Yoon Young Cho ◽  
Byung Min Ahn ◽  
Yee Soo Chae ◽  
Jong Gwang Kim ◽  
...  

Abstract This study examines the treatment outcome for imatinib mesylate (IM) and allogeneic stem cell transplantation (SCT) in patients with chronic myelogenous leukemia (CML) based on quantitative PCR data and the efficacy of a dose increment of IM in patients with cytogenetic or hematologic resistance. A total of 60 Ph+ CML patients that received IM and 17 patients that received allogeneic SCT were enrolled in the current study. The cumulative incidence of a complete hematologic response (CHR) was estimated as 71% after 1 month and 93% after 3 months, while the cumulative incidence of a major molecular response (MMR) was estimated as 53% after 6 months and 73% after 1 year. Five (22.7%) out of 22 evaluable patients achieved MMR after 3 months of IM treatment. At 24 months, 10 patients (45.5%) achieved MMR, while 7 patients (31.8%) remained as non-responders. Thrirteen patients that exhibited a cytogenetic or hematologic resistance to the standard dose of IM were treated with an increased dose of IM. Four (80%) out of 5 patients who had low levels (10–55%) of Ph+ cells in their bone marrow at the time of cytogenetic resistance achieved a complete cytogenetic response (CCR) or MMR again with the increased dose of IM, while all the patients (n=8) who failed to respond cytogenetically or hmatologically to the increased dose of IM had 100% Ph+ cells in their bone marrow at the time of cytogenetic resistance. The Philadelphia positivity in the bone marrow indicated a cytogenetic or molecular response to an increased dose of IM (P=0.009). The overall survival rate at 3 years was 88% for the IM group at the median follow-up duration of 851 days (range, 13–3346 days) and 47% for the SCT group at the median follow-up duration of 456 days (range, 34–2455 days), respectively (P&lt;0.001). Plus, the progression-free survival rate at 3 years was 61% for the IM and 47% for the SCT group, respectively (P=0.037). In conclusion, IM dose escalation can be regarded as an important treatment option in terms of reserving other treatment options, especially in patients with low levels (≤55%) of Ph+ cells in their marrow who are resistant cytogenetically or hematologically to the standard dose of IM.


Blood ◽  
2006 ◽  
Vol 109 (1) ◽  
pp. 58-60 ◽  
Author(s):  
Philippe Rousselot ◽  
Francoise Huguet ◽  
Delphine Rea ◽  
Laurence Legros ◽  
Jean Michel Cayuela ◽  
...  

Abstract In the present study, we address the issue of the discontinuation of imatinib mesylate (Gleevec) in chronic myelogenous leukemia with undetectable residual disease for more than 2 years. Twelve patients were included. The median duration of real-time quantitative–polymerase chain reaction (RTQ-PCR) negativity and imatinib therapy were, respectively, 32 months (range, 24-46 months) and 45 months (range, 32-56 months) before imatinib interruption. Six patients displayed a molecular relapse with a detectable BCR-ABL transcript at 1, 1, 2, 3, 4, and 5 months. Imatinib was then reintroduced and led to a novel molecular response in most patients. Six other patients (50%) still have an undetectable level of BCR-ABL transcript after a median follow-up of 18 months (range, 9-24 months). We hypothesize that relapses observed within 6 months reflect the kinetics of undetectable dividing chronic myelogenous leukemia (CML) cells. Those cells may be eradicated or controlled in long-term nonrelapsing patients, as described in our study.


Sign in / Sign up

Export Citation Format

Share Document