First Results of a New PCR-Based Approach for a Sensitive and Quantitative Monitoring of BCR-ABL Kinase Domain Mutations in Patients with Chronic Myeloid Leukemia.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2191-2191
Author(s):  
Oliver Pelz-Ackermann ◽  
Michael W. Deininger ◽  
Michael Cross ◽  
Ines Kovacs ◽  
Christine Guenther ◽  
...  

Abstract Mutations of the ABL kinase domain (KD) are the most frequent cause of acquired resistance to imatinib in patients with chronic myeloid leukaemia (CML), most likely due to selection of mutated clones on imatinib treatment. As new Bcr-Abl inhibitors become available, precise quantification of low level mutation will be required to monitor response. Here we report our results evaluating patients with advanced phase or imatinib resistant CML for KD mutations using a newly developed, sensitive and quantitative Ligation PCR (L-PCR) assay in comparison to direct sequencing. Twenty eight CML patients on imatinib (17 male, 11 female) with a median age of 62 (range 20 to 75) years in blast crisis (n=4), accelerated phase (n=12) or imatinib failure (n= 12) were analysed using both approaches. Sequencing of the ABL KD was performed using forward and reverse primers to ABL exons 4 and 7, while the L-PCR analysis focussed initially on the E255K and T315I mutations. Briefly, pairs of probes specific for either wild type (WT) or mutant BCR-ABL were added to the RT-PCR amplified ABL KD, then ligated under conditions optimized for specificity. Ligated probe pairs were than amplified in a quantitative PCR using universal primers. Quantification was performed using internal cell-in-cell dilutions of BaF3 cell lines expressing wt and mutant BCR/ABL and values were expressed as % BCR-ABLmut/ BCR-ABLWT. In our hands, this assay can detect 0.05 – 0.1% T315I and 0.01–0.05% E255K in a BCR-ABL WT background. The inter-assay variation at the lowest detection level was only 6.7 and 4,7% for the mutations T315I and E255K respectively. Results were scored positive only if two independent runs showed amplification exceeding the lowest controls. All patients were treated with a median imatinib dose of 600 (range 500–800) mg for a median of 10.5 (range 1 to 74) months. Dose reductions due to toxicity were necessary in 8 (29%) patients. Direct sequencing revealed E255K or T315I mutations in three patients, each with more than 20% mutated allele. L-PCR revealed these three patients plus three more with lower levels of mutation (T315I, 0.46% and E255K, 0.16, 0.17%). The patient with 0.46% T315I also showed G250E by sequencing. This patient was subsequently treated with Dasatinib but failed to respond. In twenty two patients negative by L-PCR, sequencing of exons 4 and 7 showed 15 (53%) to be WT, 6 (21%) to have KD mutations F317L, F359V (n=2), H396R, M315T with F359V, Y253F with the K247R polymorphism and one to have an 80 base pair deletion 3′ of the p-loop. In conclusion the L-PCR assay was able to detect T315I and E255K mutations in twice as many patients as did direct sequencing. These low-level mutations would most probably also have been missed by the D-HPLC-based screening. The fact that only L-PCR detected this mutation in a patient who failed to respond to dasatinib implies that the assays generate clinically useful information. mutations. The study is currently being expanded to include further mutations and longitudinal monitoring of a larger cohort of patients.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2153-2153 ◽  
Author(s):  
Jamshid S. Khorashad ◽  
Jeffrey H. Lipton ◽  
David Marin ◽  
Dragana Milojkovic ◽  
Nicholas C.P. Cross ◽  
...  

Abstract Neoplastic cells bearing fusion genes that express an activated tyrosine kinase may set the scene for accumulation of genetic lesions by dysregulating DNA damage repair mechanisms and causing genetic instability. The observation that the BCR-ABL fusion gene alters pre-mRNA splicing in a variety of other genes including Ikaros and PYK2 supports this hypothesis. However, the only current evidence for acquired genetic change in the BCR-ABL gene itself is limited to finding mutations in the BCR-ABL kinase domain in patients treated with imatinib mesylate (IM). Here we report the observation that some patients with CML have abnormally small BCR-ABL transcripts both before and during treatment. Patients with sub-optimal response to IM are screened for mutations specifically within the BCR-ABL kinase domain by performing nested PCR, thereby excluding amplification of the non-translocated ABL allele. In the first round PCR amplification is performed across the fusion and the amplicons generated are subjected to a second round to yield an expected 863 bp (containing ABL exons 4 through 9 and thus the entire BCR-ABL kinase domain) PCR fragment. Smaller amplicons were observed in 49 (9.9%) of the 494 CML patients investigated. There was marked variation in the mRNA species when the abnormally small amplicons were subjected to direct sequencing; we found exon skipping, intra-exon splicing and insertion of intronic sequences. Similarly, in some cases the open reading frame was maintained whilst in others there were frame shifts leading to premature stop codons. The commonest finding, (22 of the 49 patients) was skipping of ABL exon 7 from codons 362 to 424, which includes the activation loop of the kinase domain. The smaller amplicons persisted even after the first round products had been diluted to 1:160. We also noted that the normal 863 bp fragment was present in some cases but was not detectable in others; its absence could reflect preferential amplification of the smaller transcripts. In a number of cases the same abnormally short amplicons were identified in the same patient studied serially on three or more separate occasions. We subsequently performed a second round of nested PCR with primers designed to amplify across the BCR-ABL junction such that the product included sequences from BCR 13 to ABL exon 9. With these new primers the detection frequency of abnormally small transcripts was increased. Furthermore, we observed the smaller transcripts in all of the 12 patients tested prior to beginning IM therapy. We then sought to determine if the normal ABL allele was involved; in order to avoid amplifying the BCR-ABL kinase domain, we performed a single round of PCR and restricted the analysis to patients in complete cytogenetic remission (CCyR). Only the expected 863 bp amplicon was observed in cDNA samples from 19 CML patients in CCyR whose BCR-ABL/ABL ratios ranged from 0.01 to 0.98. Furthermore, the smaller amplicons were not observed in cDNA samples from 20 normal individuals. We conclude that these abnormalities may result either from aberrant alternative splicing or from spontaneous deletions, or from a combination of both mechanisms. They may be a manifestation of the genetic instability believed to be an integral feature of CML.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2918-2918
Author(s):  
Oliver Pelz-Ackermann ◽  
Michael W.N. Deininger ◽  
Michael Cross ◽  
Kathrin Wildenberger ◽  
Rainer Krahl ◽  
...  

Abstract Objectives: Acquired resistance to imatinib (IM) in patients with chronic myeloid leukaemia (CML) is frequently due to mutations of the BCR-ABL kinase domain (KD). Current thinking holds that this results from the selection of pre-existing mutant clones on IM. As new BCR-ABL inhibitors with differential activity against KD mutant BCR-ABL have become available, precise quantification of key mutations even at a low level is required to adequately monitor responses. Here we report that the occurrence of G250E, Q252H, Y253F/H, E255K/V, T315I, F317L, F359V mutations at the time of IM resistance is independently associated with a high maximum IM dose. Patients and Methods: We have developed highly specific allele-specific ligation-PCR assays (L-PCR) to accurately quantify a panel of frequent KD mutations, including G250E, Q252H, Y253F/H, E255K/V, T315I, F317L, F359V. In limiting dilution experiments the L-PCR assays routinely detect between 0.05 and 0.1% of mutant allele in total BCR-ABL, and their average dynamic range is in the range of 4.5 log. Forty-three patients with imatinib failure were analyzed. The median age was 60 (range 20 – 75) years and the median disease duration 64 (range 3–213) months. Eleven patients were in blast crisis, 20 in accelerated phase and 12 in chronic phase. Results: Patients were treated with chemotherapy (48%) and/or interferon alpha (76%) prior to IM. The median maximal IM dose was 600 (range 500–800) mg and the median duration of total therapy until resistance was 15.5 (range 1 to 75) months. Eighteen patients (42%) had dose reductions due to toxicity. At the time of IM resistance, clonal evolution was present in 21/41 (47%) of the patients. L-PCR identified 50 mutations in 29/43 patients (67%) (table1). One, 2, 3 or 4 different mutations were identified in 14, 11, 2 and 2 patients, respectively. The T315I and E255K/V mutations accounted for 32/50 (64%) of all mutations, while Q252H and Y253H were not detected. Twelve mutations (24%) were confirmed by direct sequencing (DS) and an additional M315T mutation and a K247R polymorphism were detected. Thirty eight (76%) mutations were negative by DS for the corresponding mutation from the L-PCR panel but additional mutations (L248V, M351T, H396Rx2, L298Vx2) were identified. All 38 L-PCR positive mutations were confirmed in a second independent experiment. The mutated clone was significantly smaller in mutations with no confirmation by DS (median 0.2, range 0.05–12.63%) compared to positive by DS (median 40.76, range 13.58–100%, p=0.003, Wilcoxon test). The detection of one or more mutations was significantly more frequent in patients with a maximum IM dose of 800mg (n=15/17, 88%) compared to less than 800mg (14/26, 53%, p= 0.02 Fisher’s exact test). Multivariate analysis (Wald forward) confirmed that a maximum IM peak dose of 800mg is an independent prognostic parameter to detect a mutation from the L-PCR panel at the time of IM resistance. Conclusions:High sensitive testing with L-PCR detects mutations with 4-fold increased frequency compared to direct sequencing.Mutations, including p-loop and T315I, may be selected by exposure to higher drug levels.The predominance of T315I and E255K/V mutations at a low level is consistent with the findings in newly diagnosed patients with Ph+ALL (Pfeifer et al. Blood 2007).


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3267-3267
Author(s):  
Jacqueline Maier ◽  
Michael Cross ◽  
Haifa Kathrin Al-Ali ◽  
Rainer Krahl ◽  
Dietger Niederwieser ◽  
...  

Abstract Abstract 3267 Poster Board III-1 Objectives: BCR-ABL kinase domain (KD) mutations can be detected at a low level prior to the start of imatinib (IM) in patients with advanced phase chronic myeloid leukemia (CML) and the presence of such mutations in CD34+ cells of CML patients in complete cytogenetic response is thought to underlie disease persistence on IM. Since new tyrosine kinase inhibitor (TKI) specific mutations have been shown to arise on nilotinib or dasatinib treatment, we have asked in this analysis whether patients with resistance to TKI may harbour additional, low-level BCR-ABL KD compound mutations in the early progenitor cells. Patients and Methods: Using a MACS® indirect CD34 Micro Bit Kit we selected a minimum of 1×104 CD34+ and CD34- cells from 16 patients at the time of TKI resistance. The median purity of the CD34+ and CD34- cell fractions was 93% (range 87 to 98%) and 95% (range 58 to 100%). Ten of these 16 patients (56%) had BCR-ABL KD mutations detectable by direct sequencing (DS M244Vx2, G250E, V299Lx2, F317I/Lx4, F359Vx2) defined as high level mutations. Complementary DNA prepared from total white cells, CD34+ and CD34- cells was used for the amplification of BCR-ABL and the sensitive detection of 8 specific BCR-ABL KD mutations (F317L, F359V, T315I, E255K, E255V, Y253H, G250E, Q252H) by quantitative Ligation-PCR, which yields a reproducible sensitivity of 0.5% BCR-ABLmutant/BCR-ABLtotal, enabling the quantitative detection of low level mutations which are not detectable by direct sequencing. Results: Of the 10 patients carrying high level mutations, 9 (90%) were represented within our specific ligation PCR-panel. In these cases, the total mutated BCR-ABL level was comparable between the total white cells (median 99%, range 14 to 100%), CD34+ (median 100, range 18.02 to 100%) and CD34- cells (median 100, range 11.73 to 100% BCR-ABLmutant/BCR-ABLtotal). However, ligation-PCR detected further low level mutations within the 16 patient panel, which where present at a higher frequency in the CD34+ cells (n=8; Y252H, E255K/V, T315I, F317Lx2, F359Vx2) than in the CD34- (n=2; Y252H, T315I) or the total white cells (n=3; Y252H, T315I, F359V). Furthermore, the proportion of mutated BCR-ABL within the patients with low level mutations was higher in the CD34+ cells (median 4.57%, range 0.64 to 7.82%) then in the CD34- (median 1.18%, range 0.75 to 1.61%) or total white cells (median 1.24%, range 0.7–6.85%). Within the 10 patients with high level mutations, the low level mutations were exclusively detected in CD34+ cells (p=0.014). Conclusions: Whereas high level mutations are present at the same level in total white cells, CD34+ and CD34- cells, we confirm our hypothesis that low level mutations are predominantly detectable in the early progenitor fraction. This is consistent with a spontaneous background of potentially resistant mutations in the stem/progenitor population which have the potential to develop a resistant leukemic phenotype on ineffective TKI treatment. Disclosures: Al-Ali: BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees. Niederwieser:BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Research Funding. Lange:BMS: Honoraria; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (5) ◽  
pp. 1208-1215 ◽  
Author(s):  
Simona Soverini ◽  
Andreas Hochhaus ◽  
Franck E. Nicolini ◽  
Franz Gruber ◽  
Thoralf Lange ◽  
...  

AbstractMutations in the Bcr-Abl kinase domain may cause, or contribute to, resistance to tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia patients. Recommendations aimed to rationalize the use of BCR-ABL mutation testing in chronic myeloid leukemia have been compiled by a panel of experts appointed by the European LeukemiaNet (ELN) and European Treatment and Outcome Study and are here reported. Based on a critical review of the literature and, whenever necessary, on panelists' experience, key issues were identified and discussed concerning: (1) when to perform mutation analysis, (2) how to perform it, and (3) how to translate results into clinical practice. In chronic phase patients receiving imatinib first-line, mutation analysis is recommended only in case of failure or suboptimal response according to the ELN criteria. In imatinib-resistant patients receiving an alternative TKI, mutation analysis is recommended in case of hematologic or cytogenetic failure as provisionally defined by the ELN. The recommended methodology is direct sequencing, although it may be preceded by screening with other techniques, such as denaturing-high performance liquid chromatography. In all the cases outlined within this abstract, a positive result is an indication for therapeutic change. Some specific mutations weigh on TKI selection.


2004 ◽  
Vol 50 (7) ◽  
pp. 1205-1213 ◽  
Author(s):  
Simona Soverini ◽  
Giovanni Martinelli ◽  
Marilina Amabile ◽  
Angela Poerio ◽  
Michele Bianchini ◽  
...  

Abstract Background: Despite the efficacy of the BCR-ABL tyrosine kinase inhibitor Imatinib mesylate for the treatment of chronic myeloid leukemia (CML), resistance has been observed in a proportion of cases, especially those with advanced stages of the disease. Point mutations within the ABL kinase domain are emerging as the most frequent mechanism for reactivation of kinase activity within the leukemic clone. Methods: We developed a denaturing-HPLC (D-HPLC)-based assay for screening for ABL point mutations. For each sample, two partially overlapping fragments of 393 and 482 bp corresponding to the kinase domain were amplified by nested reverse transcription-PCR and analyzed under selected temperature and acetonitrile gradient conditions. Fifty-one bone marrow and/or peripheral blood specimens from 27 CML patients who showed cytogenetic resistance to Imatinib were screened in parallel by D-HPLC and by direct sequencing. Results: In 12 of 27 (44%) patients, D-HPLC showed an abnormal elution profile suggesting the presence of a nucleotide change. Direct sequencing confirmed the presence of a point mutation in all cases. Conversely, all samples scored as wild type by D-HPLC showed no evidence of mutations by direct sequencing. In two cases, novel amino acid substitutions at codons already known for being hot-spots of mutation were identified (F311I and E355D). Conclusions: The proposed D-HPLC-based assay is highly specific and at least as sensitive as sequencing; with respect to the latter, it provides a much faster and less expensive semiautomated system for mutational screening. It may therefore potentially be a valuable tool for regular, large-scale testing of patients undergoing Imatinib treatment.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1952-1952
Author(s):  
Alfonso Quintás-Cardama ◽  
Don L. Gibbons ◽  
Hagop Kantarjian ◽  
Moshe Talpaz ◽  
Nicholas Donato ◽  
...  

Abstract ABL kinase domain mutations represent the most frequent cause of resistance to tyrosine kinase inhibitors (TKIs). The BCR-ABL1T315I mutation affects a highly conserved “gatekeeper” threonine near the ABL catalytic domain, thus causing steric hindrance that precludes ABL TKIs binding. BCR-ABL1T315I retains kinase activity even in the presence of micromolar concentrations of imatinib or dasatinib. Thus, early detection of BCR-ABL1T315I has important prognostic and therapeutic implications. We evaluated the sensitivity of detection of BCR-ABL1T315I in 62 CML pts after failure (n=51) or intolerance (n=11) of imatinib enrolled in a phase I study of dasatinib by direct sequencing (DS) of nested PCR-amplified BCR-ABL1 products as well as by DNA expansion of specific clones (DESC) followed by DNA sequencing of at least 10 clones. Ten (15%) pts were found to carry BCR-ABL1T315I, 4 prior to dasatinib start and 6 during dasatinib therapy. Four pts never responded to imatinib whereas 5 had achieved a complete hematologic response (CHR) and 1 a complete cytogenetic response (CCyR). Imatinib was stopped due to rash (n=1), hematologic resistance (n=4), and progression to accelerated (AP; n=1) or blastic (BP; n=4) phase. Of the 4 pts in whom BCR-ABL1T315I was detected by DESC prior to dasatinib start, 2 are dead and 2 are alive. DS performed on the same samples detected BCR-ABL1T315I only in 2 of them. In pts in whom direct sequencing failed to detect BCR-ABL1T315I, the percentage of clones carrying BCR-ABL1T315I was 10% and 100%, respectively. In one of them, DS detected BCR-ABL1T315I 12 months later, whereas in the other case, DS failed to detect BCR-ABL1T315I in 3 separate occasions. These 2 pts are still alive (1 on hydrea, 1 on bosutinib) but never achieved any cytogenetic response. BCR-ABL1T315I was detected in 6 additional pts after a median time of 5 months (range, 1–6) on dasatinib (dosing ranging from 70 to 140 mg/d): 5 of 5 analyzed by DESC and 1 of 2 determined by DS. Of them, 4 had no response to dasatinib and 2 had transient cytogenetic responses (1 minor, 1 partial) and only one is still alive. In 1 that had no response, for whom paired samples were available, BCR-ABL1T315I was present in 10% of clones but was not detected by DS. Overall, the median number of clones harboring BCR-ABL1T315I was 90% (range, 10%–100%) and the median number of mutants co-expressed with BCR-ABL1T315I was 3 (range, 1–9). DS failed to identify 40 non-BCR-ABL1T315I mutants (including F317L in 1 patient), regardless of the percentage of clones in which they were expressed, except for E355G in 1 patient. Eight pts received dasatinib for more than 3 weeks (median, 5 months; range, 2–13) and were evaluable for response. Six failed to achieve any cytogenetic response and 2 had transient cytogenetic responses (1 minor and 1 partial). Seven (70%) pts died and 3 are alive with no cytogenetic response. In conclusion, DS has a poor sensitivity to detect ABL kinase mutations, particularly when the proportion of mutated clones is low. With the advent of novel T315I inhibitors, prompt detection of this highly-resistant mutation must be prioritized and included in therapeutic algorithms. To maximize the sensitivity of T315I detection, sequencing of subcloned PCR products might be preferable to DS.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4458-4458
Author(s):  
A. M. Carella ◽  
A. Garuti ◽  
G. Cirmena ◽  
G. Catania ◽  
G. Pica ◽  
...  

Abstract Abstract 4458 Acquired resistance to Imatinib in advanced phase of CML has been associated with mutations in the kinase domain (KD) of BCR-ABL. We have recently reviewed the status of the mutations in 52 patients with early CP-CML on the samples collected at diagnosis. Mutations were identified by direct sequency (DS) with BidDye Terminator V1.1. cycle sequencing kit and analyzed with a 3130 AB capillary electrophoresis system. Twenty-eight patients had low risk, 10 intermediate risk and 14 high risk, according to Sokal/Euro. Ten out of 14 high Sokal risk patients showed the following mutations: Y253C, S265R, E255K, F359Y, N374S, E255V, E255V, E255V, R332L, E334G. Three of these patients progressed during Imatinib and second-line TKIs and died of blastic phase CML at 23, 33 and 69 months. Curiously, S265R and N374S mutations disappeared during Imatinib treatment but were substituted during follow-up by other two mutations: E255L and H396R. The patient carrying E255L mutation died in blastic phase at 33 months and the one with mutation H396R was well controlled by Nilotinib and he is now alive in CMR 26 months after. Only one out of the 10 intermediate Sokal risk carried KD mutations at diagnosis (D363G). This patient is alive in MMR at 26 months after diagnosis under Imatinib. None of the 28 low Sokal risk patients carried KD mutations at diagnosis and no patients developed cytogenetic evolution while on treatment. In conclusion, the fact that KD mutations were more present in patients with high Sokal risk supports the hypothesis that the probability developing a mutation is related to the basic biology of the disease rather than being merely a random event. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1985-1985
Author(s):  
Giovanni Cazzaniga ◽  
Barbara Corradi ◽  
Rocco Piazza ◽  
Simona Soverini ◽  
Giovanni Martinelli ◽  
...  

Abstract The most frequently identified mechanism of acquired Imatinib resistance in Ph+ leukemia is BCR-ABL kinase domain point mutations. The observation that 90% of patients with mutations eventually relapsed suggests that harboring any BCR-ABL mutation has prognostic value. Thus, the detection of BCR-ABL mutants prior to and during the course of Imatinib therapy may aid in determining therapeutic strategies. However, current methods for mutation screening have either low sensitivity or are too time consuming and labor intensive to be routinely feasible. In addition, to date approximately 30 different point mutations in the BCR-ABL kinase domain have been isolated from Ph+ patients resistant to Imatinib treatment. This may require a method of mutation detection at large spectrum. We tested the feasibility of using the “SNP mining tool” on the Nanogen microarray device (San Diego, CA) for a rapid and sensitive identification of BCR/ABL mutations. The Nanogen SNP mining tool allows de novo discovery of genetic variations, including SNPs and mutations. The method combines the features of electronically controlled DNA hybridization on open-format microarrays, with mutation detection by a fluorescence-labeled mismatch-binding protein (mutS protein). If the test DNA contains a point mutation when compared to the reference DNA, the heteroduplex will contain a mismatch. This mismatch is detectable by the fluorescent-labeled mutS protein that binds to DNA containing a mismatch at any position of the DNA sequence. Since 500bp DNA fragments can be analyzed simultaneously, the entire ABL tyrosine kinase domain can be monitored by only two overlapping PCR reactions, thus substituting high throughput sequencing as a preliminary screening of point mutations. Electronic addressing of DNA strands to distinct test sites of the chip allows parallel analysis of several individuals or several SNPs/mutations in few individuals. We first performed a preliminary series of experiments on samples known to contain mutations by sequencing of PCR clones. A point mutation in ABL could be identified in a 10 copies dilution of a mutated plasmid. Different point mutations in ABL could be identified in 6 patients in which at least 1 mutated clone was found by PCR, cloning and sequencing. Four patients resistant to Imatinib were tested in 10-fold cDNA serial dilutions. A positive value was found till a 10−4 dilution in 3/4 patients. A sensitivity of 10−3 was found in 1 patient. We also monitored a single CML p210-positive patient in late chronic phase, resistant to IFN and treated with 400mg/die Imatinib. In this patient, always positive by RT-PCR of BCR-ABL, we could demonstrate the presence of the mutation prior to Imatinib and in all follow up time points during Imatinib therapy, since 1 month to 6 months. The follow up of additional Ph+ cases will be analyzed. In conclusion, the sensitivity and the short time required for the analyses of different patients samples, or multiple time points during follow-up, make the Nanogen device as an alternative to DHPLC or cloning, for the rapid identification of BCR/ABL mutations and monitoring of BCR/ABL mutations during Imatinib treatment. Early diagnosis of resistance and prompt treatment with appropriate drug dosing may be essential for increasing the success of cure in Ph+ leukemia.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 275-275 ◽  
Author(s):  
Selim Corm ◽  
Franck Nicollini ◽  
Dominique Borie ◽  
Nathalie Sorel ◽  
Thibault Leguay ◽  
...  

Abstract Imatinib mesylate (IM) is highly effective in newly diagnosed CML. Unfortunately, acquired abl-kinase domain mutations are commonly identified to be associated to resistance or relapse in patients with advanced disease. Distinct mutation exhibit varying degrees of resistance, and mutation occurring in the ATP-binding loop (P loop) or in positions in direct contact with IM (i.e. T315) may be correlated with subsequent disease progression. In this study, we investigated the bcr-abl mutation status of imatinib-resistant CML patients and correlated to the disease outcome. Patients and methods: From CML patients diagnosed in 5 French centers, 67 IM-resistants patients presenting mutations by direct sequencing of the abl kinase domain at time of resistance or relapse were included in this study. When starting IM monotherapy (J1) at 400 to 600 mg per day, 45 patients were in chronic phase of CML, 12 patients were in accelerated phase of the disease and 7 patients were in blast crisis. The median duration of IM is 29 months (range 3–54) and the median delay between diagnosis and J1 is 34 months (range 0–168). At the end of the study, 23 patients were dead. According to the mutation status, a Kaplan-Meier curve was construct for time to death to J1, and survival were compared using log rank test. Results: By direct sequencing, 17 patients presented mutations in the P-loop (residues 252 to 255), 10 patients presented the T315M mutation alone, 1 patient had the T315 and the M351 mutations, and 39 patients presented mutations in other kinase domains. According to the mutation status, survival difference between patients presenting P-loop mutations or mutations in an other abl-kinase (including T315), no significant difference was observed (p=0.07). However, survival difference between patients presenting P-loop mutations, T315 mutation and mutations in an other abl-kinase domains was highly significant (p=0.0031, figure). The median survival for the P-loop and T315 mutated patients was of 39 months and 29 months respectively, and median survival not reached for others. Conclusion: By this study we confirm that either the T315 mutation or mutations in the P-loop had a poor prognosis. Detailed correlation analysis of mutation status according to clinical features and response to increased IM-dose will be reported. Figure Figure


Sign in / Sign up

Export Citation Format

Share Document