Abnormally Small BCR-ABL Transcripts in CML Patients before and during Imatinib Treatment.

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 ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 575-575
Author(s):  
Simona Soverini ◽  
Alessandra Gnani ◽  
Caterina De Benedittis ◽  
Ilaria Iacobucci ◽  
Annalisa Lonetti ◽  
...  

Abstract Abstract 575 Incorporation of the tyrosine kinase inhibitor (TKI) imatinib in the frontline treatment of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) patients (pts) has significantly improved the anti-leukemic efficacy of induction therapy. In contrast to chronic myeloid leukemia (CML), however, responses are short-lived and relapse is frequently associated with the selection of Bcr-Abl kinase domain (KD) mutations, fostered by the high genetic instability of Ph+ ALL cells. The advent of the 2nd-generation TKIs dasatinib and nilotinib has brought additional treatment options both for newly diagnosed and for imatinib-resistant pts. To analyze the changes they have brought in mutation frequency and type, we have reviewed the database recording the results of BCR-ABL KD mutation analyses done in our laboratory from January 2004 to June 2011. Overall, 781 tests on 258 Ph+ ALL pts (number of tests per pt, range: 1–15) were performed by denaturing high-performance liquid chromatography (D-HPLC) followed by direct sequencing of D-HPLC-positive cases. One hundred and fourty-three pts were analyzed because of imatinib resistance. One hundred and one out of 143 (71%) pts scored positive for one or more KD mutations. Similarly to what is know to occur in CML, hematologic and cytogenetic resistance were by far more frequently associated with mutations than molecular resistance (Bcr-Abl transcript increase as assessed by RT-Q-PCR). Overall, mutations at thirteen residues were detected. In contrast to what can be observed in CML, three mutations were by far the most frequent, accounting for almost 75% of the mutated cases: T315I (n=38 pts, 37%), E255K/V (n=19 pts, 18%) and Y253H (n=19 pts, 18%). The other mutations were, in order of frequency: F359V/I, M244V, M351T, F317L, G250E, Q252H, L387M, D276G, L248R, E279K. Nine out of 103 (9%) pts had two mutations, in the same (2 pts) or in different (7 pts) subclones. In 84 pts who were analyzed because they were reported to have developed resistance to dasatinib (n=72) or nilotinib (n=12) as 2nd- or 3rd-line TKIs, 65 (77%) had newly acquired mutations (57/72 dasatinib-resistant pts and 8/12 nilotinib-resistant pts). The most frequent newly acquired mutation in this setting was the T315I, detected in 35/57 (61%) cases acquiring mutations on dasatinib and in 2/8 cases acquiring mutations on nilotinib. Other recurrent newly acquired mutations were F317L, V299L, T315A in dasatinib-resistant pts and Y253H and E255K in nilotinib-resistant pts. Thirty out of 65 pts (46%) were positive for multiple mutations (2 to 4 mutations, in the same or in different subclones or both) that emerged under the same TKI in 11 cases (37%) and accumulated as a consequence of multiple lines of TKI therapy in the remaining 19 (63%) cases. Mutation analysis was also performed in 15 resistant pts enrolled in a clinical trial of dasatinib as first-line treatment for Ph+ ALL. Twelve pts were positive for mutations; 11/12 had a T315I. Sixty-one pts were analyzed at the time of diagnosis in order to assess whether TKI-resistant mutations could already be detectable. Only two pts (3%) were positive for mutations: one patient had an F311L that disappeared after one month of nilotinib treatment; an additional patient was positive only by D-HPLC, but not by the less sensitive direct sequencing – most likely for the T315I mutation that shortly after the start of dasatinib treatment outgrew and led to resistance. Taking advantage of the recent availability of a next-generation sequencing platform (Roche 454), allowing high sensitivity (0.01%) mutation scanning of the KD, samples collected at the time of diagnosis and during follow-up from selected Ph+ ALL cases who developed mutations and resistance to TKI therapy were retrospectively analyzed – but the mutations were not always already detectable at diagnosis. In conclusion: a) although 2nd generation TKIs may ensure a more rapid debulking of the neoplastic clone and have much fewer insensitive mutations, long-term disease control remains a problem and the T315I becomes an even tougher enemy; b) the clinical usefulness of mutation screening of Ph+ ALL pts at diagnosis before TKI start, even with highly sensitive approaches is low – not all mutations pre-exist since genetic instability remains high and fosters mutational events anytime during treatment. Supported by PRIN, FIRB, AIL and AIRC. Disclosures: Soverini: ARIAD: Consultancy; Novartis: Consultancy; Bristol-Myers Squibb: Consultancy. Luppi:CELGENE CORPORATION: Research Funding. Foà:Bristol-Myers Squibb: Consultancy, Membership on an entity's Board of Directors or advisory committees. Baccarani:Bristol-Meyers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Martinelli:Novartis: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Pfizer: Consultancy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2002-2002
Author(s):  
Sara Ghorashian ◽  
Anna Babb ◽  
Jamshid Khorasan ◽  
Jaspal Kaeda ◽  
David Marin ◽  
...  

Abstract We identified all patients who developed blast crisis before the advent of imatinib therapy and in whom stored marrow samples were available from a database of CML patients in a major teaching hospital. mRNA was extracted and cDNA synthesized successfully in 20 of 22 cases. The cDNA was subjected to real time PCR for quantification of BCR-ABL transcripts. The ABL allele not involved in the t(9;22) translocation was excluded from mutational analysis by subjecting the cDNA to nested PCR. This was achieved by amplifying from exons 13 and 10 of the BCR and ABL moieties of the BCR-ABL fusion gene using primers B2A and JAMR, respectively. The resulting amplicon was then subjected to nested PCR using primers, NTPB+ and NTPE-, sited within exons 4 and 10 of the ABL gene. The nested PCR yielded an 863 bp fragment in length containing the entire BCR-ABL kinase domain. To test for successful amplification an aliquot of the PCR products were electrophoresed through 2.0% agarose gel. In all cases, a single amplicon was observed and the PCR reaction subjected to magnetic purification. The purified amplicon were then sequenced by Sanger’s dideoxy chain termination reaction using Big-Dye ABI 310 sequencer (Applied Biosystems, Foster City, USA). In each case, the sequence obtained was compared with the published ABL type 1a sequence, Genbank M14752, using Blast 2 software. No mutations were found in any patient. This suggests the absence of a mutated kinase in a proportion of greater than approximately 30% of the total BCR-ABL kinase, due to the limit of detection of the technique. Our results suggest that the predominant form of BCR-ABL in the setting of blast crisis (in the absence of imatinib) is wild type. This goes against theories that kinase mutations confer an intrinsic ‘gain of function’ or a proliferative advantage to a CML clone. Instead, it is likely that mutant forms of BCR-ABL which are relatively resistant to imatinib binding gain prominence through positive selection with the presence of imatinib within the cell milieu.


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 ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1938-1938 ◽  
Author(s):  
Alfonso Quintás-Cardama ◽  
Don L. Gibbons ◽  
Hagop Kantarjian ◽  
Moshe Talpaz ◽  
Nick Donato ◽  
...  

Abstract ABL kinase domain (AKD) mutations are the main mechanism of resistance in patients (pts) with CML who fail tyrosine kinase inhibitors (TKIs) therapy, being found in 20%–40% of cases by direct sequencing (DS). Therefore, many pts fail TKI therapy for unknown reasons. We evaluated the development of AKD mutations among 61 CML pts after imatinib-intolerance (n=10) or -resistance (n=51) enrolled in a phase I study of dasatinib by 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. Prior to imatinib (400 mg daily in 47, 600 in 13, and 800 in 1), 54 pts were in chronic (CP), 2 in accelerated (AP), and 5 in blastic (BP) phase. At the end of imatinib therapy, 26 pts were in CP, 14 in AP, and 21 in BP. AKD mutations (in ≥1 out of 10 sequenced clones) were detected in 58/61 (95%) pts by DESC (4 pts with wild type [WT] BCR-ABL1) but only in 23/55 (42%) by DS. Overall, 118 AKD mutations at 112 amino acid positions were detected by DESC, of which 77 had never been previously reported. Mutations conferring resistance to >1μM imatinib (M244V, G250E, Q252H, Y253H, E255K/V, F359V, H396R, and T315I) were detected in 20 (34%) pts by DESC, but only in 5 (8%) by DS. Combinations of mutations within the same clone (polymutants) were detected in 33/58 (57%) pts by DESC, with clones expressing 2 (n=41), 3 (n=11), 4 (n=1), or even 5 (n=2) distinct mutations. By contrast, only 1 pt was found to carry 2 different mutations (M244V and M351T) by DS. Dasatinib was subsequently given to 56/61 (92%) pts (53 evaluable for response) for a median of 17 months (range, 1 to 48). DESC available in 15 pts during dasatinib therapy revealed 16 additional mutations (15 amino acid positions), including 5 previously not reported (all in polymutants). Dasatinib-resistant mutations (L248V/R, Q252H, E255K, V299L, T315I/A, and F317L/C/I/S/V) were detected in 7/15 (47%) cases (2 with T315I) by DESC but only in 2/15 (13%) by DS. Of these 15 pts, only 3 (1 CP, 1 AP, and 1 BP) are alive. The proportion of clones harboring WT BCR-ABL1 prior to and during dasatinib therapy decreased significantly (p=0.003), particularly in pts harboring highly dasatinib-resistant mutants. Notably, pts without cytogenetic (CG) response on dasatinib had a lower proportion of WT clones compared with those who achieved at least a partial cytogenetic response (p=0.02). AFTER IMATINIB ON DASATINIB Evaluable Patients No. Clones No. WT Clones (%) Evaluable Patients No. Clones No. WT Clones (%) DASATINIB RESPONSE 53 598 268 (48) 15 130 26 (20) No CG Response 25 242 128 (49) 7 58 5 (9) CG Response Minor 10 79 33 (42) 4 35 9 (26) Partial 6 57 35 (61) 2 18 8 (44) Complete 12 123 46 (37) 2 19 4 (21) In summary, DESC greatly increases the sensitivity of detection of AKD mutants compared to DS and reveals heightened BCR-ABL1 genetic instability among pts failing TKIs, which could explain TKI resistance in pts not carrying resistant mutations by DS. This might be mediated by generation of resistant polymutant clones that perpetuate a “mutator phenotype” leading to WT BCR-ABL1 exhaustion.


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 ◽  
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