Next-Generation Sequencing Of The BCR-ABL1 Kinase Domain May Be Beneficial In Decision Making Among Chronic Myeloid Leukemia Patients With Tyrosine Kinase Inhibitor Resistance

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 384-384 ◽  
Author(s):  
Yucel Erbilgin ◽  
Ahmet Emre Eskazan ◽  
Ozden Hatirnaz Ng ◽  
Ayse Salihoglu ◽  
Tugrul Elverdi ◽  
...  

Abstract Background and Aim BCR-ABL1 mutation testing is recommended for chronic myeloid leukemia (CML) patients who have suboptimal response and/or treatment failure with tyrosine kinase inhibitor (TKI) therapy. BCR-ABL1 mutations in the kinase domain (KD) of ABL1 account for at least 40-50% of all TKI resistant cases. Thus, detection of low-level mutations after development of resistance may offer critical information to guide subsequent therapy selection. The current gold standard for BCR-ABL1 mutation detection is Sanger sequencing (SS), which has an analytical sensitivity of approximately 10-20%. In this study, our aim was to detect low level BCR-ABL1 variants in follow up samples of CML patients with TKI resistance using next-generation sequencing (NGS) approach. Methods Eight patients with CML who were resistant to imatinib had been routinely sequenced with SS for BCR-ABL1 KD mutations between December 2009 and December 2012. We then retrospectively analyzed these samples with NGS. RT and long range PCR was performed to amplify BCR-ABL1 fusion transcripts and the PCR products sequenced bidirectional after library preparation. We performed a fusion transcript based BCR-ABL1 mutation assay using Roche 454 amplicon deep-sequencing technology that is suited for detecting low level variants in pooled amplicon samples. Sequencing data was analyzed with GS Amplicon Variant Analyzer (AVA) software, and the variant frequency cut-off was adjusted to 1%. Results Clinical features, sequencing results, and the outcomes of the patients were summarized in Table 1. Four patients were male, and the median age was 37 years (range, 20-60 years). The patients were all in chronic phase at the time of the diagnosis. After imatinib resistance, 4 patients had received dasatinib (DAS), and 2 were given nilotinib (NIL) as second line TKI treatment. The remaining two patients had both received DAS and NIL (Table 1). In a set of 20 clinical samples, at different time points, NGS not only identified all the mutations detected by SS, but additionally identified low level variants present between 1 – 28.12 %. T315I and E255K/V were the most common mutations, which were detected in four patients, both by SS and NGS at the same time points (Table 1). Two patients (patient #1 and #4) had T315I, and they both progressed to blastic phase and died. E255K was detected in patients #2 and #3, and patient #2 had achieved and maintained complete cytogenetic and major molecular responses with 100 mg daily DAS, whereas patient #3 had received both NIL and DAS, but she was deceased due to myeloid blastic crisis. Among 4 patients (patients #5, #6, #7, and #8), mutation analysis was performed at eleven different time points, and these patients were wild-type with SS. We also did not detect any clinically significant mutations in these patients by NGS. Most probably mechanisms other than KD mutations were responsible for the TKI resistance among these four patients. Conclusions Polyclonal mutations in BCR-ABL1 KD are commonly identified in TKI resistant patients. Thus, detection of low-level mutations after development of resistance offers critical information to guide subsequent therapy selection. An inappropriate kinase inhibitor selection could highly increase the risk of treatment failure with clonal expansion of the resistant mutant. In our imatinib resistant cohort, we detected low level variants accompany to known mutations which may constitute background genetic variations. Although we had expected to detect mutations earlier by NGS (i.e. before these mutations can be detected by SS), we did not observe such finding in our patients. The patients' samples may not show a stable mutation spectrum between time points. Hence, it is not always possible to spot a mutation before patients show resistance to therapy. Regular NGS analysis might detect these mutations in earlier phases, which might help clinicians to choose the most suitable individual treatment modality for the patients. Acknowledgment The authors would like to thank the Interlaboratory Robustness of Next-generation sequencing (IRON) Phase II study group members, especially to Simona Soverini and Alexander Kohlmann who designed BCR-ABL primers and plates. We also would like to thank the Research Fund of the Istanbul University (Project no. 24244) and Turkish Society of Hematology for supporting the study. Disclosures: Sayitoglu: Roche Diagnostics: Research Support Other.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1295-1295 ◽  
Author(s):  
Jean-Michel Cayuela ◽  
Francois Lay ◽  
Yves Chalandon ◽  
Philippe Rousselot ◽  
Xavier Thomas ◽  
...  

A high proportion of Ph-positive Acute Lymphoblastic Leukemia (Ph+ ALL) patients still undergo relapses despite the use of Tyrosine Kinase Inhibitors (TKIs) in addition to chemotherapy as frontline therapy. In this leukemia subtype, the role of BCR-ABL1 kinase domain (KD) mutations as a driver of resistance to TKIs has already been documented by previous studies and such mutations have been reported in up to 80% of the patients at relapse. Next-generation sequencing (NGS) has been proposed to characterize these mutations with a higher sensitivity than Sanger. We report here a prospective study aiming at detecting potentially resistant cell populations by NGS in Ph+ ALL patients enrolled in the GRAAPH 2014-trial. Between March 2016 and February 2019, 156 patients aged 18 to 59 years with newly diagnosed Ph+ and/or BCR-ABL1 positive ALL have been included in the GRAAPH 2014 trial (NCT02619630). BCR-ABL1 isoforms were E1A2 69%, B2A2/B3A2 29%, atypical 2%. After a prephase of steroid, treatment consisted of 4 blocks of chemotherapy + nilotinib. 118 patients (76%) underwent allogeneic or autologous stem cell transplantation (SCT). 22 medullary relapses were recorded within a median time of 9 months (range, 2 - 35). Blood and marrow samples harvested at diagnosis, after each treatment block, before and 3 months after SCT, and at relapse, were sequenced if BCR-ABL1/ABL1 ratio were above 0.001. Mutated BCR-ABL1 transcripts were detected by sequencing the KD of BCR-ABL1 transcripts by NGS with a limit of detection (LOD) of 0.03. T315I allele specific oligonucleotide (ASO) droplet digital RT-PCR (ddRT-PCR) with a LOD of 0.0005 was also performed at diagnosis on a subset of 63 patients, including 5 who have subsequently developed a T315I clone. NGS. At diagnosis, no KD mutation was found by NGS in pretreatment samples of 137 patients. During follow-up (FU), only 12 mutations were found by NGS in 7 out of the 88 patients tested (81, 45, 30, 20, 19, 9 after block 1, 2, 3, 4, before and 3 months after SCT, respectively). Mutations were T315I (N=6), Y253H (N=1), E255K (N=2), E255V (N=1), Q252H (N=1), Y253F (N=1). At relapse, 16 mutations were identified by NGS in 12 patients out of the 17 tested (71%). Mutations were T315I (N=7), Y253H (N=n=3), F359V (N=2), E255K (N=1), E255V (N=1), Q252H (N=1), Y253F (N=1). More than 1 mutated clone were present in 2 patients (E255V+T315I+F359V and Y253H+F359V), and a compound mutation was found in 1 patient (Q252H/Y253F). Out of the 7 patients found mutated during FU, 5 have relapsed with a rapid expansion (1 to 3 months) of the mutated clone. One patient harboring a sub-clonal (10%) E255K at MRD1 has relapsed 9 months later without any detectable mutation. One patient identified with 3 mutated clones (E255K 10%, E255V 10%, T315I 80%) underwent SCT and has not relapsed so far. We failed to anticipate expansion of any mutated clone in the 7 remaining patients found mutated at relapse. T315I ASO ddRT-PCR on diagnostic samples. Low-level T315I mutated BCR-ABL1 transcripts (0.00051 to 0.0013) were detected in 14 out of 63 patients (24%) tested. Only one has expanded a T315I clone later on. In the context of the GRAAPH 2014 trial, 71% of the 17 relapses tested so far were associated with BCR-ABL1 KD mutations. Expansion of the mutated clone could have been characterized before the onset of hematological relapses in only 5 out of 12 patients (42%). Unfortunately in these cases, lags between first detection and relapse were very short (1 to 3 months). On the contrary, occurrences of relapses associated with expansion of KD-mutated clones could not have been anticipated in 58%. All mutations identified, including T315I, F359V, E255K/V and Y253F/H, Q252H/Y253F are known for conferring resistance to nilotinib. NGS is a valuable method for KD mutation detection in Ph+ ALL. It allows a quantitative characterization of KD mutations at relapse. However in our hands and in the context of an intensive therapy combining chemotherapy, nilotinib and SCT, its enhanced sensitivity as compared to Sanger (3% vs 20%) does not translate into the capacity of anticipating expansion of KD-mutated clones. Moreover, in this study, NGS did not detect any mutation in pre-therapeutic samples while T315I mutated BCR-ABL1 transcripts were found at low-level in 24% of these samples by ddRT-PCR. However it should be emphasized that when detected, low-level T315I mutated sub-clones present at diagnosis failed to expand in most instances. Disclosures Cayuela: Incyte: Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau. Chalandon:Pfizer: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Incyte Biosciences: Consultancy, Honoraria; Novartis: Consultancy, Honoraria. Rousselot:Pfizer: Research Funding. Thomas:PFIZER: Honoraria; ABBVIE: Honoraria; INCYTE: Honoraria; DAICHI: Honoraria. Huguet:Amgen: Honoraria; Servier: Honoraria; Jazz Pharmaceuticals: Honoraria; BMS: Honoraria; Pfizer: Honoraria; Incyte Biosciences: Honoraria; Novartis: Honoraria. Chevallier:Incyte: Consultancy, Honoraria; Jazz Pharmaceuticals: Honoraria; Daiichi Sankyo: Honoraria. Boissel:NOVARTIS: Consultancy. Vey:Novartis: Consultancy, Honoraria; Janssen: Honoraria. Berthon:PFIZER: Other: DISCLOSURE BOARD; JAZZPHARMACEUTICAL: Other: DISCLOSURE BOARD; CELGEN: Other: DISCLOSURE BOARD.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1654-1654
Author(s):  
Jayachandran PK ◽  
Trivadi S Ganesan ◽  
Nikita Mehra ◽  
Krishnarathinam Kannan ◽  
Manikandan Dhanushkodi ◽  
...  

Background: Imatinib resistance mutation analysis (IRMA) or abl kinase domain mutation analysis is performed in patients with Chronic myeloid leukemia (CML) whenever the response to treatment is inadequate. We have analyzed the reports of IRMA at our centre. Methods: The clinical details of 71 patients with CML on Imatinib, who underwent IRMA testing during the period of January 2017 to March 2019 were collected from the patient records and analyzed. IRMA was performed for failure or warning or progression, anytime during the course of treatment. IRMA was done by either Sanger sequencing (n=45) or next generation sequencing (n=26, Illumina, NGS). The associations between variables were tested using Chi - Square test. Results: Median age at diagnosis of 71 patients was 44 years (Range 18 - 71 years). Males constituted 70% (n=50). At diagnosis, 92% (n=65) of patients were in chronic phase and the remainder were in accelerated phase (n=4) or blast crisis (n=2). Mutations in the abl kinase domain were detected in 26 patients (37%). Next Generation Sequencing (NGS) could identify more mutations (13/26 - 50%) compared to conventional Sanger Sequencing (13/45 - 29%), but the difference was not significant (p=0.07). NGS could identify three or more mutations in 5 patients in contrast to Sanger. All the mutations detected were those previously described except for an insertion of 35bp near the C-Terminal which was identified in 3 patients. E459K translocation was identified in 6 patients. E355G translocation was identified in 4 patients. F359V, M351T, Y253H, G250E, H396R, T315I translocations were identified in 3 patients each. Patients who were not compliant to therapy had increased frequency of mutations (14/26 - 54%) compared to those who were compliant (12/45 - 27%), which was significantly different (p=0.02). Patients who had loss of complete hematological response (CHR) had significantly higher frequency of mutations (14/21- 67%) compared to other reasons for performing the test (p=0.001). Patients who had failure to achieve targets at various time points had a significantly lower frequency of mutations (4/23 - 17%, p=0.02) compared to other reasons for performing the test. Conclusion: Patients who were not compliant for treatment were more likely to have mutations. Loss of CHR showed an increased frequency compared to other reasons. NGS could identify mutations in more number of patients. NGS identified numerically higher mutations in patients. Larger prospective data are needed to confirm these observations. Table Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2771-2771
Author(s):  
Marcin M Machnicki ◽  
Iwona Solarska ◽  
Rafal Ploski ◽  
Ilona Seferynska ◽  
Tomasz Stoklosa

Abstract Chronic myeloid leukemia (CML) is currently a true chronic disease for majority of patients who achieve durable remission with tyrosine kinase inhibitors (TKI) and remain in chronic phase (CML-CP) for several years. However, a number of patients develop TKI resistance and may progress to blastic phase (CML-BP) which represent major therapeutic challenge. Although CML-BP resembles acute myeloid leukemia (AML) in many aspects, including selected genetic aberrations, pathogenesis of CML progression to acute phase is not fully understood. Therefore there is a strong rationale for studying underlying mechanisms of progression to CML-BP. To gain comprehensive insight into genetic background of CML progression, we performed targeted high-throughput sequencing of sequential DNA samples from patients experiencing a treatment failure and progression to CML-BP. We inquired whether gene mutations previously described in human malignancies (with special focus on genes involved in leukemogenesis) were gained during progression or if they existed already at diagnosis. Sequential samples were collected from 5 patients, who progressed to CML-BP, despite TKI therapy and for whom samples were available from both diagnosis and progression. Roche NimbleGen SeqCap EZ custom-capture was used to acquire exonic sequences of approximately 1000 cancer-related genes, comprising genes from commercially available cancer panels (such as Illumina TrueSight Cancer, NimbleGen Comprehensive Cancer Panel) and also genes altered in hematological malignancies as reported in current literature. Common variants (>1%) gathered in ESP6500 and 1000 genomes projects and our internal exome database were filtered out. Patient CML-1 was diagnosed in CML-BP and relapsed within 13 months despite treatment with 2 TKIs. Both at the diagnosis and progression, a DNMT3A p. P799T mutation was detected with similar allele frequency. Patient CML-2 was diagnosed in CML-CP, underwent alloHSCT within 6 months from diagnosis and achieved short-term remission. However, within few months she relapsed with lymphoid BP and despite short-lived responses to combined chemotherapy and TKI, she developed 2nd BP and 3rd BP with BCR-ABL1 p. Y253H and p. T315I mutations, respectively. Strikingly, p. R320* RUNX1 mutation was present at all four time points (diagnosis in CML-CP and all three BPs), though at diagnosis frequency of this mutation was approx. 1%. Patient CML-3 wasdiagnosed in CML-BP and was treated with TKIs plus chemotherapy. After 25 months patient developed resistance and progressed. We detected IDH1 p. R132S mutation, typical for AML, exclusively in the BP sample from progression, while it was absent in diagnostic sample. Patient CML-4 was diagnosed in CML-CP, developed resistance to 3 TKIs and finally progressed to CML-BP after 5 years. P. Y183C IDH1 mutation was detected in the BP sample. Patient CML-5 was diagnosed in CP and despite treatment with TKI, progressed to CML-BP within 3,5 years. P. R139L and p. R166L RUNX1 mutations were detected only in the sample from CML-BP. All detected mutations were confirmed independently by Sanger sequencing or deep amplicon sequencing for low frequency variants. Detected mutations are summarized in table 1. Our analysis of sequential samples from CML patients proves that mutations in genes commonly mutated in myeloid malignancies (DNMT3A, IDH1, RUNX1) may be preexisting or may arise during progression, independently of BCR-ABL1 mutation. With regard to preexisting mutations, this may lead to clonal evolution of the disease. Importantly, such preexisting mutations could have been missed in the previous studies, presented or published, employing next-generation sequencing strategy, since most of those studies used algorithms to detect newly gained aberrations in CML-BP as compared to CML-CP. Table 1. Mutations detected in patients experiencing TKI resistance and progression with no detectable BCR-ABL1 mutations. Patient Time to progression [months] Mutations CML-1 13 DNMT3A p. P799T preexisting CML-2 9 RUNX1 p. R320* preexisting CML-3 25 IDH1 p. R132S acquired CML-4 63 IDH1 p. Y183C acquired CML-5 41 RUNX1 p. R139L RUNX1 p. R166L acquired acquired Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4042-4042
Author(s):  
Pascal Vannuffel ◽  
Barbara Cauwelier ◽  
Céline De Rop ◽  
Friedel Nollet

Abstract Background. Among myeloproliferative diseases, development of chronic myeloid leukaemia (CML) is associated with the emergence of the fusion oncogene BCR-ABL1 resulting from a t(9,22) chromosomal translocation (Philadelphia chromosome). Mutations of the BCR-ABL1 kinase domain constitute a major cause of treatment failure in CML patients receiving tyrosine kinase inhibitor (TKI) treatment. Moreover, the occurrence of cells with multiple mutations is frequently associated with a higher resistance rate to the different TKI (Imatinib, Dasatinib or Nilotinib). So far, the gold standard procedure to detect BCR-ABL1 mutations remains the conventional Sanger Sequencing (SS), endowed with an analytical sensitivity of 10 to 20 %. The recent implementation of Next Generation Sequencing (NGS) allows lowering the sensitivity level and quantitative follow-up of the mutated subclone(s), which probably improves CML patient's treatments management. Aims. In this retrospective study, we evaluate the advantage of NGS approach to i) identify patients harbouring (low level) mutations that have not been not assessed by conventional methods, ii) detect the emergence of mutated clones earlier than SS and iii) monitor evolution of mutations. Methods. Total BCR-ABL1 RNA was transcribed into a long range cDNA covering the kinase, the regulatory, and the SH2/SH3 domains of either p190 or p210 BCR-ABL1 transcripts (exons 4 to 10). From primers designed with the AmpliseqTM Designer Software, a set of 10 amplicons was generated according to the AmpliSeqTM protocol. Bar-coded libraries were sequenced on the Ion Torrent PGM platform and data were analysed with Torrent Suite and NextGene softwares. Serial dilutions of samples harbouring mutations at different levels were used to determine a variant frequency cut-off. Our methodology was applied to a group of 36 patients presenting with poor response to TKI and with no mutation detected by SS and to a set of 100 samples, corresponding to 20 mutated patients, at different time points before the time of mutation identification by SS. Results. From the serial dilutions experiment, the detection limit of the assay was set up to 2 % (R² > 0.997). An overall coverage ranging from 20 000 to 50 000 reads can be achieved for the hotspot mutations when up to 12 samples were tested together on a 316 Ion chip. On the 36 patients tested by NGS versus SS, no one was found to harbour TKI-resistance mutation. NGS successfully detected all mutations identified by SS; mutations were typically detected within 4 months (18/20 patients) and were also detected up to 9 months prior to detection by SS, even in patients with a low abundance of BCR-ABL1 transcripts and in sequencing failure by SS. In 2 patients presenting with up to 3 mutations, evolution of mutations (emergence, expansion or depletion) correlates with clinical data of treatment decisions, i.e. E255K (patient-1) and L248V (patient-2) depletions when switching from Imatinib to Dasatinib, F317L (patient-1), G250E (patient-1) and T315I (patient-2) expansions under Dasatinib and a complete but transient depletion of T315I (patient-2) with the protein synthesis inhibitor homoharringtonine (Omacetaxine). Finally, assessment of the mutation status of one patient with compound mutations following an Illumina protocol on a MiSeq platform had allowed comparison of technologies performances. Conclusions. NGS did not detect mutations in 36 patients poorly responding to TKI with no detectable BCR-ABL1 TK mutation by SS. For 20 patients showing BCR-ABL1 TK mutation by SS, NGS was able to detect the mutation in samples taken up to 9 months prior to the moment when the mutation was observed by SS. Advances in sequencing technologies and further lowering sensitivity levels can contribute even more to earlier detection of mutations and guide an earlier switch of TKI. Quantitative and sensitive monitoring of mutation evolution can also inform the most appropriate and optimized treatment algorithms. A prospective evaluation of the clinical impact of NGS-based BCR-ABL1 mutation detection is ongoing. Disclosures Vannuffel: ARIAD Pharmaceuticals: Research Funding.


2019 ◽  
Vol 12 (1) ◽  
Author(s):  
Simona Soverini ◽  
Elisabetta Abruzzese ◽  
Monica Bocchia ◽  
Massimiliano Bonifacio ◽  
Sara Galimberti ◽  
...  

AbstractBCR-ABL1 kinase domain (KD) mutation status is considered to be an important element of clinical decision algorithms for chronic myeloid leukemia (CML) patients who do not achieve an optimal response to tyrosine kinase inhibitors (TKIs). Conventional Sanger sequencing is the method currently recommended to test BCR-ABL1 KD mutations. However, Sanger sequencing has limited sensitivity and cannot always discriminate between polyclonal and compound mutations. The use of next-generation sequencing (NGS) is increasingly widespread in diagnostic laboratories and represents an attractive alternative. Currently available data on the clinical impact of NGS-based mutational testing in CML patients do not allow recommendations with a high grade of evidence to be prepared. This article reports the results of a group discussion among an ad hoc expert panel with the objective of producing recommendations on the appropriateness of clinical decisions about the indication for NGS, the performance characteristics of NGS platforms, and the therapeutic changes that could be applied based on the use of NGS in CML. Overall, these recommendations might be employed to inform clinicians about the practical use of NGS in CML.


2017 ◽  
Vol 34 (7) ◽  
Author(s):  
Matthew K. Stein ◽  
Lindsay Morris ◽  
Jennifer L. Sullivan ◽  
Moon Fenton ◽  
Ari VanderWalde ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document