Single-Arm, Open-Label, Multicenter Study Of Deep Molecular Response (MR4.5) With Nilotinib In Adult Patients (pts) With Newly Diagnosed Philadelphia Chromosome–Positive Chronic Myeloid Leukemia In Chronic Phase (CML-CP): ENESTnext Study Update

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4015-4015 ◽  
Author(s):  
Michael Mauro ◽  
Jerald Radich ◽  
Jorge E. Cortes ◽  
Shaker Dakhil ◽  
Christopher Keir ◽  
...  

Abstract Background Pivotal treatment-free remission (TFR) trials require prolonged deep molecular response (MR), often MR4.5, for study entry (Mahon et al. 2010), which has led to increasing focus on MR4.5 as a treatment goal for pts with CML-CP. New tyrosine kinase inhibitors (TKIs), such as nilotinib, more potently inhibit BCR-ABL and elicit significantly deeper and faster MRs than imatinib (Larson et al. 2012), requiring more sensitive techniques to monitor residual disease. In ENESTnext (registered as NCT01227577), MR was evaluated using standard reverse transcriptase quantitative polymerase chain reaction (RQ-PCR) and a microfluidic “digital” PCR assay, which is >1-log more sensitive than conventional methods. Methods Pts diagnosed with CML-CP within 6 mo of enrollment were treated with nilotinib 300 mg twice daily (BID) for up to 2 years. RQ-PCR was performed on peripheral blood samples by a central laboratory according to the International Scale (IS). Samples were taken monthly in mo 1-3 and every 3 mo thereafter. The primary endpoint was the rate of confirmed MR4.5 at 2 years, defined as ≥ 2 samples taken 3 mo apart with ≥ 4.5-log reduction of BCR-ABL transcript levels (≤ 0.0032%IS) with a minimum of 25,614 ABL control copies. Pts with suboptimal response or failing treatment (per European LeukemiaNet 2009 recommendations) could dose escalate to nilotinib 400 mg BID per physician discretion. Complete cytogenetic response (CCyR) and major MR (MMR, 3-log reduction of BCR-ABL transcript levels [≤ 0.1%IS]) were also assessed. In an exploratory analysis, samples identified as MR4.5 using conventional RQ-PCR were also evaluated using the Fluidigm digital PCR platform (Oehler et al. 2009). BCR-ABL copy number was estimated by Poisson distribution; samples were positive if copy number was > 0. The data cutoff date for this analysis was April 30, 2013. Results Of 128 pts, 64 (50%) were male and 103 (80%) were white. The mean age was 55.6 y (range, 21.0-89.0). Pts were treated for a median of 8.5 mo (range 0.1-24.1); median daily nilotinib dose was 600 mg. As of the data cut, 87, 36, 23 and 18 pts have been treated for ≥ 6, ≥ 12, ≥ 18 and ≥ 22 mo, respectively. Cumulative incidence of MR4.5, MMR and CCyR was 22 (17%), 76 (59%) and 72 (56%) pts, respectively. In the 22 pts who achieved MR4.5, the mean time to first MR4.5 was 6.4 mo (range, 1.0-22.7). Digital PCR was performed on 57 samples from these 22 pts; 15/22 pts had ≥ 2 samples from different time points. Of these, 8 were initially positive for BCR-ABL and became negative, 5 were initially negative and remained negative and 2 were initially positive and remained positive. Achieving BCR-ABL negativity using digital PCR generally occurred rapidly (within 3 mo of MR4.5). The most common (≥ 2 pts) grade 3 adverse events (AEs) were nausea (n = 4), headache (n = 3), elevated lipase level (n = 12), thrombocytopenia (n = 6), neutropenia (n = 6), hypophosphatemia (n = 5), anemia (n = 4) and increased amylase level (n = 3). Grade 4 AEs were myocardial infarction (n = 2), device-related infection (n = 1), elevated lipase level (n = 4), thrombocytopenia (n = 3), neutropenia (n = 2) and hyponatremia (n = 1). To date, 22 (17%) pts have discontinued treatment. Reasons included AEs (n = 11), withdrawn consent (n = 3), protocol deviation (n = 2), unsatisfactory therapeutic effect (n = 2), abnormal laboratory values (n = 2), abnormal test procedure result (n = 1) and loss to follow-up (n = 1). Conclusions Treatment with nilotinib 300 mg BID in pts with newly diagnosed CML-CP resulted in rapid and substantial rates of MR4.5 using conventional RQ-PCR, with a safety profile similar to that reported in previous studies. The digital PCR assay detected residual BCR-ABL in approximately 50% of samples with at least MR4.5 by RQ-PCR, and showed declining BCR-ABL levels with continued therapy. Thus, use of digital PCR may help better identify appropriate candidates for TFR studies. Based on results from the ENESTnd trial (Larson et al. 2012), MR4.5 rates are expected to increase over time. Other studies have demonstrated that pts who achieve early, deep MR have improved long-term outcomes (Marin et al. 2012). Evaluation of longer-term outcomes for patients with negative digital PCR results is ongoing. Disclosures: Mauro: Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Ariad: Consultancy, Honoraria, Research Funding, Speakers Bureau; Pfizer: Consultancy, Honoraria, Speakers Bureau. Off Label Use: This abstract describes a clinical trial evaluating the investigational agent midostaurin for use in patients with advanced systemic mastocytosis. Radich:novartis: Consultancy, Honoraria, Research Funding; Bristol Myers Squibb: Consultancy, Honoraria; Ariad: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Incyte: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Cortes:Pfizer: Consultancy, Research Funding; Novartis: Research Funding; Bristol Myers Squibb: Research Funding; Ariad: Consultancy, Research Funding; Teva: Consultancy, Research Funding. Keir:Novartis: Employment, Equity Ownership. Yi:Novartis: Employment. Goldberg:Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Bristol Myers Squibb: Consultancy, Honoraria, Research Funding, Speakers Bureau; Ariad: Consultancy, Honoraria, Research Funding, Speakers Bureau.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1796-1796
Author(s):  
Michael J. Mauro ◽  
Shaker Dakhil ◽  
Jorge E. Cortes ◽  
David A. Rizzieri ◽  
Christopher H Keir ◽  
...  

Abstract Background: The BCR-ABL tyrosine kinase inhibitor nilotinib elicits faster and deeper molecular responses (MRs) vs imatinib in patients with CML-CP. Achievement of sustained deep MR is associated with improved long-term outcomes and is a key criterion for entry into treatment-free remission (TFR) studies. Given the importance of accurately measuring deep MR in patients with CML, increasingly sensitive techniques are needed for monitoring minimal residual disease. In ENESTnext, MR to nilotinib was assessed using conventional methodology (real-time quantitative reverse transcriptase polymerase chain reaction [RQ-PCR]) and a novel microfluidic digital PCR assay that is > 1 log more sensitive than standard RQ-PCR. Methods: In this single-arm, open-label, multicenter study (NCT01227577), adults with CML-CP diagnosed within 6 months of enrollment were treated with nilotinib 300 mg twice daily (BID) for up to 2 years. Dose escalation to nilotinib 400 mg BID for patients with suboptimal response or treatment failure (per modified European LeukemiaNet 2009 recommendations) was permitted per physician discretion. RQ-PCR evaluation of peripheral blood samples was performed by a central laboratory (monthly for the first 3 months and every 3 months thereafter) according to the International Scale (IS). The primary endpoint is the rate of confirmed (≥ 2 samples taken 3 months apart) MR4.5 (≥ 4.5-log reduction of BCR-ABL transcript levels; BCR-ABLIS ≤ 0.0032%) with 2 years of nilotinib therapy; complete cytogenetic response (CCyR) and major MR (MMR; 3-log reduction of BCR-ABL transcript levels; BCR-ABLIS ≤ 0.1%) were evaluated as secondary endpoints. Per protocol, assessment of cytogenetic response was not required at specified time points for all patients on study. In an exploratory analysis, samples from patients with confirmed MR4.5by conventional RQ-PCR were also evaluated using the more sensitive Fluidigm digital PCR platform. The data cutoff date for this analysis was April 30, 2014. Results: A total of 128 patients were enrolled (median age, 56.5 years [range, 21.0-89.0 years]); 64 patients (50.0%) were male and 103 (80.5%) were Caucasian. As of the data cutoff, 45 patients (35.2%) had completed the study, 49 (38.3%) remained on treatment, and 34 (26.6%) had discontinued early. With a median treatment duration of 12.7 months, 88 (68.8%), 94 (73.4%), and 32 (25.0%) patients achieved CCyR, MMR, and MR4.5, respectively, at any time (Table). Of 32 patients who achieved MR4.5, 14 achieved MR4.5 by 6 months. A total of 169 samples from 32 patients with confirmed MR4.5 by conventional RQ-PCR were analyzed by digital PCR. Using the digital PCR platform, 6 of these patients initially had detectable BCR-ABL transcripts that subsequently became undetectable with continued nilotinib therapy. Of the remaining 26 patients, 12 had BCR-ABL transcripts that were initially undetectable and remained undetectable by digital PCR, 12 had detectable BCR-ABL transcripts that remained detectable, and 2 had undetectable BCR-ABL transcripts that became detectable. The most common (≥ 4 patients) grade 3/4 adverse events (AEs) regardless of relationship to study drug were increased lipase (n = 14), thrombocytopenia (n = 11), neutropenia (n = 8), hypophosphatemia (n = 5), anemia (n = 4), and nausea (n = 4). Reasons for study discontinuation were AEs (n = 15), unsatisfactory therapeutic effect (n = 5), withdrawn consent (n = 4), death (n = 3; causes of death were other malignancy, pneumonia, and not specified/no AE [n = 1 each]), protocol deviation (n = 3), abnormal laboratory values (n = 2), loss to follow-up (n = 1), and administrative problems (n = 1). Conclusions: Frontline treatment with nilotinib 300 mg BID in patients with newly diagnosed CML-CP led to rapid achievement of MR4.5 as assessed with conventional RQ-PCR. As > 40% of samples with at least MR4.5according to standard RQ-PCR were positive using the digital PCR assay, this tool may have potential in evaluating MR to determine eligibility for TFR studies. Table Response CCyRa MMR MR4.5 Patients with response, n (%) 88 (68.8) 94 (73.4) 32 (25.0) Time to response, n (%) < 3 mo 26 (20.3) 21 (16.4) 2 (1.6) 3 to < 6 mo 42 (32.8) 41 (32.0) 12 (9.4) 6 to < 12 mo 16 (12.5) 22 (17.2) 11 (8.6) 12 to < 18 mo 4 (3.1) 9 (7.0) 7 (5.5) ≥ 18 mo 0 1 (0.8) 0 a Cytogenetic response was not assessed in all patients at all time points. Disclosures Mauro: Novartis Oncology: Consultancy; Bristol Myers Squibb: Consultancy; Ariad: Consultancy; Pfizer: Consultancy. Cortes:Ariad: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Teva: Consultancy, Research Funding. Rizzieri:Sanofi: Consultancy; Celgene: Consultancy, Speakers Bureau. Keir:Novartis: Employment, Equity Ownership. Yi:Novartis Pharmaceuticals: Employment. Heinrich:Novartis: Consultancy, Patents & Royalties, Research Funding; MolecularMD: Consultancy, Equity Ownership. Goldberg:Novartis: Honoraria, Research Funding, Speakers Bureau; Bristol Myers Squibb: Honoraria, Research Funding, Speakers Bureau; Ariad: Research Funding, Speakers Bureau; Pfizer: Research Funding. Kuriakose:Teva: Speakers Bureau; Alexion: Speakers Bureau. Radich:Novartis: Consultancy, Research Funding; Ariad: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3096-3096
Author(s):  
Bernardi Simona ◽  
Andrea Di Palma ◽  
Federica Cattina ◽  
Simone Perucca ◽  
Michele Malagola ◽  
...  

Abstract Monitoring of BCR-ABL1 levels by quantitative PCR (qPCR) is essential for the management of CML patients treated with TKIs. Because of intrinsic limits, qPCR does not appear to be an optimal assay to select the best candidates to TKIs discontinuation. Up to 40% of CML patients treated with TKIs can achieve a Deep Molecular Response (DMR), but only 50% maintain a stable Treatment Free Remission (TFR) after therapy discontinuation. Digital PCR (dPCR), giving an absolute quantification of BCR-ABL1, is expected to be more sensitive and accurate than qPCR in the assessment of molecular MRD. dPCR performed on a QuantStudio 3D Digital PCR System (Life Technologies) by using a TaqMan-MGB probes targeting the BCR-ABL1 transcript was used to comparatively analyse 102 CML patients with Major (MR3.0 = 31 cases) or Deep (MR4.0= 33 cases; MR4.5 = 24 cases and MR5.0 = 14 cases) molecular response. Preliminary results showed that: a) ≥77% of deep responders (MR4.0, MR4.5 and MR5.0) fell under the value of 0.468 BCR-ABL1 copies/▢l indicated by the ROC analysis as the value below which the patients with lower levels of MRD might be dissected (spec.=71%, sens.=77%; AUC=0,79); b) BCR-ABL1 transcript levels were detectable by dPCR also in cases resulted undetectable by qPCR. In this study, we analysed the BCR-ABL1 transcript levels by dPCR in 207 samples related to 102 CML patients. Fourteen (14%) out of 102 CML patients discontinued the TKIs therapy. Among them, 3 patients (21%) lost DMR and all of them showed dPCR values > 0.468 BCR-ABL1 copies/▢l (previously described as cut-off for a deep MRD), while 11 (79%) maintained a stable DMR and 9 of them (82%) fell under the value of 0.468 BCR-ABL1 copies/▢l. These latter patients stratified in different DMR classes by qPCR and all had undetectable level of BCR-ABL1 by qPCR. In 149 out of 207, qPCR revealed DMR. They were: MR4.0= 59 samples; MR4.5= 61 cases; MR5.0 = 29 cases. One hundred twenty-five (84%) fell under the value of 0.468 BCR-ABL1 copies/▢l. A linear regression analysis in these samples did not show any correlation between BCR-ABL1 copies/▢l detected by qPCR with the ones detected by dPCR (R2 < 0.01). On the basis of our preliminary results, TFR seems to be correlated to the maintenance of dPCR values < 0.468 BCR-ABL1 copies/▢l. The concordance between qPCR and dPCR quantification in patients with DMR and with BCR-ABL1 copies/▢l value < 0.468 was poor. Patients with dPCR values < 0.468 BCR-ABL1 copies/▢l may have 75% of probability to maintain TFR status. These results suggest that dPCR may be more sensitive to detect the MRD and could be more useful for DMR monitoring and for dissecting the best candidate to discontinuation of therapy with TKIs. Disclosures Tiribelli: Ariad Pharmaceuticals: Consultancy, Speakers Bureau; Bristol-Myers Squibb: Consultancy, Speakers Bureau; Novartis: Consultancy, Speakers Bureau. Castagnetti:Bristol-Myers Squibb: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; ARIAD Pharmaceuticals: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria. Soverini:Novartis: Consultancy; Ariad: Consultancy; Bristol-Myers Squibb: Consultancy. Rosti:Pfizer: Honoraria, Research Funding, Speakers Bureau; Roche: Honoraria, Research Funding, Speakers Bureau; BMS: Honoraria, Research Funding, Speakers Bureau; Incyte: Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau. Martinelli:Roche: Consultancy; ARIAD: Consultancy; Pfizer: Consultancy, Speakers Bureau; Genentech: Consultancy; Amgen: Consultancy; MSD: Consultancy; Roche: Consultancy; ARIAD: Consultancy; Pfizer: Consultancy, Speakers Bureau; BMS: Speakers Bureau; Genentech: Consultancy; Amgen: Consultancy; Novartis: Speakers Bureau; MSD: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2763-2763
Author(s):  
Hyun-Gyung Goh ◽  
Soo-Young Choi ◽  
Ju-Hee Bang ◽  
Soo-Hyun Kim ◽  
Eun-Jung Jang ◽  
...  

Abstract Abstract 2763 Approximately 50% of CP CML patients achieve complete molecular response (CMR) at 6–7 years of first-line imatinib therapy. Although imatinib therapy is effective in CML patients and a substantial portion of patients achieve CMR with prolonged imatinib therapy, up to 10^7 leukemic cells can still be present in the absence of detectable BCR-ABL in RQ-PCR assay due to the sensitivity limit of current RQ-PCR technology. The recent data from STIM (Stop Imatinib) trial showed that the probability of persistent CMR at 12 month follow-up after imatinib discontinuation was 41%, and the conclusion was that imatinib can be safely discontinued, at least in some patients with persistent CMR. However, it is still not clearly defined whether discontinuation of imatinib therapy can be safely employed in patients with sustained CMR. In our prospective study, we examined if imatinib therapy can be safely discontinued in CML patients with sustained CMR4.5 according to strict PCR sensitivity criteria, and CMR4.5 was defined as undetectable BCR-ABL using RQ-PCR assay with at least 4.5-log sensitivity. CML patients who were treated with imatinib for more than 3 years and whose BCR-ABL was undetectable in RQ-PCR for at least 2 years were enrolled in this study. Our primary objectives were to evaluate the probability of persistent CMR4.5 at 12 month follow-up after discontinuation, and to measure the duration of persistent CMR4.5 after discontinuation. The secondary objective was to evaluate the probability of major molecular response (MMR) loss and the time taken to lose MMR at 12 month follow-up after discontinuation. In patients with loss of MMR, the probability of re-achieving MMR/CMR4.5 and the time taken to re-achieve MMR/CMR4.5 after imatinib resumption were also evaluated. After discontinuation, molecular response was monitored using RQ-PCR assay every month up to 6 month follow-up, every 2 months up to 12 month follow-up, and every 3 months thereafter. Digital PCR methodology with higher sensitivity compared to RQ-PCR assay was also applied before discontinuation and every year after discontinuation for more accurate estimation of BCR-ABL transcript levels. In case of relapse, defined as loss of MMR on 2 consecutive assessments, imatinib therapy was re-introduced and molecular response after resumption was observed using both RQ-PCR and digital PCR assays. As of data cut-off date of 15 Jul 2011, 20 patients (13 females, 7 males) who were diagnosed in Seoul St. Mary's Hospital between 20 Mar 1996 and 25 Apr 2005 were enrolled in this study with a median follow-up of 7 months (range, 2–9), and informed consents were obtained from all patients prior to participation. With a median age of 44 years (range, 25–67), the percentages of patients with low, intermediate and high Sokal risk scores were 30%, 30% and 15%, respectively with unknown Sokal risk scores in 25%. Ten patients (50%) received SCT and/or interferon therapy prior to imatinib therapy, while 10 patients (50%) received first-line imatinib therapy. The median time on imatinib therapy and the median duration of sustained CMR4.5 were 91 months (range, 40–112) and 60 months (range, 23–104), respectively, prior to discontinuation. Since discontinuation of imatinib therapy, all of 20 patients remained off therapy at the last follow-up with persistent CMR4.5 in 18 patients (90%) and loss of CMR in 2 patients (10%). Although loss of CMR was observed in 2 patients, both patients have not resumed imatinib therapy as MMR was maintained at the last follow-up. Our preliminary data show lower relapse rate after discontinuation compared to previous discontinuation studies. Strict PCR sensitivity criteria should be employed to assess the accurate measurement of BCR-ABL transcript levels prior to discontinuation, and then it might be possible to safely stop imatinib therapy in CML patients with stable CMR4.5. Through further clinical investigation on a large patient population and longer period of observation, more concrete conclusion can be made regarding the outcome of imatinib discontinuation. Updated data with longer follow-up duration will be presented in the meeting. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2771-2771 ◽  
Author(s):  
Sikander Ailawadhi ◽  
Carole B. Miller ◽  
Anand P. Jillella ◽  
Nebu Koshy ◽  
Brian Tudor ◽  
...  

Abstract Abstract 2771 Background: NIL is a potent, highly selective Bcr-Abl kinase inhibitor approved for newly diagnosed adult pts with Philadelphia-chromosome positive (Ph+) CML-CP and in Ph+ CML-CP and accelerated-phase pts who are resistant or intolerant to IM. Achieving complete cytogenetic response (CCyR) and major molecular response (MMR, 3-log reduction of Bcr-Abl transcript level from the baseline mean) are favorable prognostic factors for CML. This multicenter, open-label study (ENABL) was designed to explore nilotinib Bcr-Abl effects in pts with CCyR but who have suboptimal molecular response to IM. Methods: This study evaluates change in Bcr-Abl trends in 2 groups of CML-CP pts (total n = 18) who achieved CCyR but have suboptimal molecular response to IM defined as: (Group 1) treated ≥ 1 year with IM, but Bcr-Abl transcript levels did not reach ≤ 0.1% on the international scale (IS) (MMR); or (Group 2) > 1-log increase in Bcr-Abl transcript levels from best response regardless of IM treatment duration. Pts are treated with NIL 300 mg twice daily for ≥ 1 year. RQ-PCR analysis is performed by a central lab at screening, then every 3 months (mos) for Group 1. Group 2 pts are monitored by RQ-PCR monthly for the first 3 mos, then every 3 mos. The 1° end point is change in Bcr-Abl transcript levels from a standardized baseline value by RQ-PCR at 12 mos. The data cutoff date for this analysis was June 30, 2011. Results: Eighteen pts (Group 1, n = 17; Group 2, n = 1) have been treated with NIL for a median of 17 mos on study (range 3–34 mos). Thirteen pts have been treated for ≥ 6 mos and 10 for ≥ 12 mos. One pt was deemed ineligible due to lack of evidence of CCyR at baseline but is included in the analysis because there was at least 1 post-baseline evaluation performed. The remaining 17 pts had CCyR at baseline. Before enrollment, pts were treated with at least 400 mg once-daily IM; the mean dose of prior IM treatment was 487 mg/day (range 342–786 mg/day). Median duration of prior IM treatment was 3.4 yrs (range 1.3–10.2 yrs). Three pts had prior interferon treatment. All 18 pts were treated for ≥ 3 mos and had ≥ 1 post-baseline RQ-PCR result. Overall, 15 of 18 evaluable pts (83%) achieved MMR during treatment; 10 pts by 3 mos, 1 pt by 4.5 mos (measured at end of study), 1 pt by 6 mos, 2 pts by 9 mos, and 1 pt by 30 mos (Figure 1). The 3 pts who did not reach MMR at any point were only followed for up to 3 mos before discontinuing from the trial but showed a decreasing Bcr-Abl trend. Overall, pts achieved a median log reduction of PCR transcript levels of 3.1 (0.08% IS) at 3 mos; median 3.3-log reduction (0.05% IS) at 6 mos, and median 3.5-log reduction (0.035% IS) at 9 mos. Four pts had > 4-log (≤ 0.01% IS) reduction in Bcr-Abl; of these, 2 pts reached > 4.5-log (≤ 0.0032% IS) reduction in Bcr-Abl at least once during the study. Median Bcr-Abl transcript log reduction at 12 mos was 3.6 (0.025% IS, 1° end point) for 10 evaluable pts. All these pts reached MMR during NIL treatment; 9 pts by 12 mos, 1 pt after 30 mos. NIL was well tolerated and brief dose interruptions were sufficient to manage most adverse events (AEs). Seven of 18 pts were dose reduced for NIL-related AEs and re-escalated if the patient recovered from the AEs. Patients were permitted to dose escalate to 400 mg b.i.d. per physician's discretion if MMR was not achieved after 6 mos (n = 1). The Grade 3 AEs reported include 2 cases of rash and 1 case each of pneumonia, squamous cell carcinoma, bladder prolapse, uterine prolapse, bradycardia, hypertension, hyperbilirubinemia and hypophosphatemia. The rashes and bradycardia were suspected to be related to NIL. No Grade 4 AEs were reported. The median dose intensity was 600 mg/day (range 300–683 mg/day). Five pts were discontinued from the study (3 due to abnormal laboratory values, 1 due to an AE, and 1 due to protocol violation). No pts who experienced QTcF changes had differences > 33 msec from baseline. No QTcF prolongation > 500 msec was observed. Conclusions: NIL treatment results in high molecular response rates in CML-CP pts with suboptimal molecular responses to IM. Overall 83% of pts who switched to NIL achieved MMR, and the median Bcr-Abl log reduction for pts who reached 12 mos on study was 3.6 (0.025% IS). The IRIS study has shown that MMR rates increase with time in pts treated with IM (Hughes Blood 2010); however, this study appears to demonstrate that MMR is achieved relatively quickly in suboptimal molecular IM-treated pts when switched to NIL. Disclosures: Ailawadhi: Novartis Pharmaceuticals: Consultancy, Speakers Bureau. Miller:Incyte: Research Funding; Novartis: Honoraria, Research Funding, Speakers Bureau. Akard:Eisai: Speakers Bureau; Bristol Myers-Squibb: Speakers Bureau; Novartis: Speakers Bureau; Millenium: Speakers Bureau; Chemgenex: Consultancy. Ericson:Novartis Pharmaceuticals Corporation: Employment, Equity Ownership. Lin:Novartis: Employment, Equity Ownership. Radich:Novartis: Consultancy, Research Funding, Speakers Bureau; Pfizer: Consultancy, Speakers Bureau; Bristol-Myers Squibb: Consultancy, Speakers Bureau. DeAngelo:Novartis: Consultancy; Bristol-Myers Squibb: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1224-1224
Author(s):  
Koji Sasaki ◽  
Hagop M. Kantarjian ◽  
Susan O'Brien ◽  
Farhad Ravandi ◽  
Marina Konopleva ◽  
...  

Abstract Introduction Initial treatment with tyrosine kinase inhibitors (TKI) induces excellent response in the majority of patients with CML-CP. Current guidelines recommend periodic monitoring of BCR-ABL1 levels to monitor response. During the course of treatment, recognizing early predictors of deeper response and longer-term outcomes can help guide treatment. This is relevant not only at the specified fixed time point typically reported (i.e., 3, 6, 12 months) but at any other time point when an assessment is made. Achievement of sustained deep molecular response is a goal of increasing relevance as it opens the possibility of treatment discontinuation. The objective of this study is suggest optimal BCR-ABL transcript levels at any given time, and to suggest a prediction model for sustained molecular response 4.5 (MR4.5) (BCR-ABL ≤0.0032%) for at least 2 years according to BCR-ABL levels achieved within the first 12 months of TKI therapy. Methods Response data for 630 patients with newly diagnosed CML-CP in consecutive prospective clinical trials of frontline imatinib (n=73; NCT00048672), high-dose imatinib (n=208; NCT00038469 and NCT00050531), nilotinib (n=148; NCT00129740), dasatinib (n=150; NCT00254423), and ponatinib (n=51; NCT01570868) were analyzed. Real-time PCR analysis was performed at approximately 3 month intervals during the first year and 6 month intervals thereafter. The "best fit average" molecular response was defined by robust linear regression models, with which the estimated molecular level in patients with complete cytogenetic response (CCyR) within 1 year, major molecular response (MMR) within 1 year, and sustained MR4.5 at any point were defined. The acceptable molecular response was defined by quantile regression for the 95th percentile, with which the worst 5% BCR-ABL levels in patients with CCyR within 1 year, MMR within 1 year, and sustained MR4.5 at any point were identified. Results In 630 patients, 2512 data points of BCR-ABL levels within 1 year of TKI were identified. The median follow-up for the entire cohort was 106 months (range, 0.3-177.8). The regression equations for best fit average PCR for CCyR within 1 year was Log10(PCR) = -0.2159 x (Months) + 0.1957; for MMR within 1 year, Log10(PCR) = -0.2304 x (Months) + 0.1046; for sustained MR4.5 at any point, Log10(PCR) = -0.2154 x Months -0.1161. The regression equations for acceptable PCR for CCyR within 1 year was Log10(PCR) = -0.15796 x (Months) + 1.54839; for MMR within 1 year, Log10(PCR) = -0.20999 x (Months) + 1.54839; for sustained MR4.5, Log10(PCR) = -0.22476 x (Months) + 1.50516 (Figure 1). The best fit average PCR (i.e., estimated levels achieved by the average responder in each category) for CCyR within 1 year was 0.353%, 0.079%, 0.017%, and 0.004% at 3, 6, 9, and 12 months, respectively; for MMR within 1 year was 0.259%, 0.053%, 0.011%, and 0.002% at 3, 6, 9, and 12 months, respectively; for sustained MR4.5 at any point was 0.295%, 0.067%, 0.015%, and 0.003% at 3, 6, 9, and 12 months, respectively (Table 1). To achieve CCyR within 1 year, the acceptable PCR (i.e., levels achieved by 95% of all those who eventually reach the said endpoint) response was 11.872%, 3.987%, 1.339%, and 0.450% at 3, 6, 9, and 12 months, respectively; to achieve MMR within 1 year, 8.287%, 1.943%, 0.455%, and 0.107% at 3, 6, 9, and 12 months, respectively; to achieve sustained MR4.5 at any time, 6.774%, 1.434%, 0.304%, and 0.064% at 3, 6, 9, and 12 months, respectively. Of 289 patients who eventually achieved sustained MR4.5, 288 (99%) achieved CCyR within 1 year; 268 (93%), MMR within 1 year; 201 (70%), MR4 within 1 year; 162 (56%), MR4.5 within 1 year; 72 (25%), CMR within 1 year. Of 359 patients who achieved MMR within 1 year with a minimum follow-up of 48 months, 256 (71%) achieved sustained MR4.5; of 180 patients who achieved MR4.5 within 1 year, 151 (84%); of 72 patients who achieved CMR within 1 year, 65 (90%). Conclusion Proper interpretation of early transcript levels at any time during the course of therapy may help predict later response and outcome. Such models can be built to guide therapy for patients in a continuous basis. To achieve sustained MR4.5 for at least 2 years, deeper responses are required at each time point. Our model proposes optimal values that predict the highest probability of reaching such goal. At a minimum, CCyR within 1 year is required to achieve sustained MR4.5. Disclosures Kantarjian: Bristol-Myers Squibb: Research Funding; ARIAD: Research Funding; Amgen: Research Funding; Pfizer Inc: Research Funding; Delta-Fly Pharma: Research Funding; Novartis: Research Funding. Ravandi:Seattle Genetics: Consultancy, Honoraria, Research Funding; BMS: Research Funding. Konopleva:AbbVie: Research Funding; Genentech: Research Funding. Wierda:Novartis: Research Funding; Abbvie: Research Funding; Acerta: Research Funding; Gilead: Research Funding; Genentech: Research Funding. Daver:Ariad: Research Funding; Karyopharm: Honoraria, Research Funding; Sunesis: Consultancy, Research Funding; BMS: Research Funding; Kiromic: Research Funding; Otsuka: Consultancy, Honoraria; Pfizer: Consultancy, Research Funding. Jabbour:ARIAD: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Novartis: Research Funding; BMS: Consultancy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2301-2301
Author(s):  
Carole Miller ◽  
Sikander Ailawadhi ◽  
Anand P. Jillella ◽  
Jerald P. Radich ◽  
Daniel J DeAngelo ◽  
...  

Abstract Abstract 2301 Background: Nilotinib is a potent, highly selective Bcr-Abl kinase inhibitor approved for adult patients with Ph+ CML in chronic and accelerated phase who are resistant or intolerant to IM and for frontline CML treatment. The achievement of a complete cytogenetic response (CCyR) and a major molecular response (MMR), defined as ≥ 3 log reduction of Bcr-Abl transcript levels from a standardized baseline (equivalent to ≤ 0.1% international scale [IS]) are favorable prognostic factors. Achieving CCyR and MMR are associated with significantly lower rates of disease progression (Saglio, G. et al. N Engl J Med 2010; 362:2251). This multi-center, open-label US study was designed to assess the impact of nilotinib on Bcr-Abl molecular response dynamics in pts with CCyR but have demonstrated a suboptimal molecular response to IM. Methods: This study evaluates the change in Bcr-Abl kinetics in 2 groups of CML-CP pts (target enrollment n=50) who achieved CCyR but have a suboptimal molecular response to IM. Suboptimal molecular response was defined either as: (Group 1) pts treated ≥ 1 year with IM, but have not reached MMR; or (Group 2) pts with > 1 log increase in Bcr-Abl transcript levels from best response regardless of the IM treatment duration. Pts are treated with nilotinib 300 mg BID on study; if dose reductions are required, pts are treated with nilotinib 400 mg QD. Quantitative reverse transcriptase polymerase chain reaction (RQ-PCR) analysis is performed by a central lab at baseline and then every 3 months (mos) for Group 1. Group 2 pts are monitored by RQ-PCR at baseline, monthly for the first 3 mos and then every 3 mos on study. The primary endpoint is to measure the change on a logarithmic scale of Bcr-Abl transcript levels from a standardized baseline value by RQ-PCR after 12 mos on treatment with nilotinib. This analysis was performed on the 14 pts enrolled as of the data cut-off date of June 30, 2010. Results: Fourteen pts (Group 1:13; Group 2:1) have been treated with a median of 9.8 mos (range: 3.4–22.3 mos) on nilotinib. Thirteen pts entered the trial with a baseline CCyR. One pt (Group 1) was discontinued due to lack of evidence of CCyR at baseline (protocol deviation); however was included in the analysis since the pt had at least one post-baseline evaluation performed. Prior to enrollment, pts were treated with ≥ 400mg QD IM; the mean dose of prior IM treatment was 505 mg/day (range 377 – 786 mg/day). The median duration of prior IM treatment was 40.5 mos (range 15.3 – 115.8 mos). The median Bcr-Abl log reduction at baseline was 2.5 (0.32%IS). Overall 12/14 pts achieved MMR on study; 9 pts after 3 mos, 1 pt after 4.5 mos (measured at end of study due to a protocol deviation), and 2 pts after 9 mos. Overall, pts achieved a median 3.11 log reduction (0.078%IS) at Month (Mo) 3; median 3.33 log reduction (0.047%IS) at Mo 6, and a median 3.72 log reduction (0.019% IS) at Mo 9. Of the pts who were treated at least 12 mos, 6/7 (85.7%) reached MMR after switching to nilotinib and the median Bcr-Abl transcript log reduction at 12 mos was 3.66 (0.022% IS,1° endpoint). Nilotinib was well tolerated and brief dose interruptions were sufficient to manage most adverse events (AEs). Five of 14 pts were dose reduced for nilotinib-related AEs. The median dose intensity was 536 mg (range 300 – 600 mg/day). One of each of the following Grade 3 AEs were reported: rash, pneumonia, squamous cell carcinoma, bladder prolapse, and uterine prolapse. Only the rash was suspected to be due to nilotinib. No Grade 4 AEs were reported. One pt experienced serious AEs; pancreatitis was suspected to be related to nilotinib and pneumonia was not suspected to be related to nilotinib. Nilotinib was interrupted and the pt recovered from both events. Four pts were discontinued from the study, 3 due to abnormal labs (Grade 2–3 ALT, Grade 2 bilirubin) and 1 due to a protocol deviation. The median Bcr-Abl log reduction of these 4 pts at end of study was 3.03 logs (0.096% IS). A protocol amendment has since instated a more liberal dose reduction guideline. No pts who experienced QTcF changes had differences > 33 msec from baseline. No QTcF prolongation >500 msec was observed. Conclusions: Nilotinib treatment resulted in an improvement of molecular response in pts switched from IM and was well tolerated. Overall 12/14 (85.7%) of the pts who switched to nilotinib achieved MMR at the time of analysis, and the median Bcr-Abl log reduction for pts who reached 12 mos on study was 3.66 from the standardized baseline (0.022% IS). Disclosures: Miller: Novartis: Consultancy, Honoraria, Research Funding. Off Label Use: Nilotinib is being studied patients with suboptimal response in the context of a clinical trial. Ailawadhi: Novartis: Consultancy, Honoraria. Radich: Novartis: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria. DeAngelo: Novartis: Consultancy; BMS: Consultancy. Goldberg: Novartis: Consultancy, Honoraria, Research Funding. Williams: Novartis: Employment, Equity Ownership. Lin: Novartis: Employment. Akard: Novartis: Consultancy, Honoraria.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4028-4028
Author(s):  
Simona Bernardi ◽  
Andrea Di Palma ◽  
Mario Tiribelli ◽  
Erika Codarin ◽  
Giuseppina Ruggeri ◽  
...  

Abstract Monitoring BCR-ABL1 transcript by quantitative PCR (qPCR) is essential for the management of CML patients treated with TKIs. Currently, up to 40-50% of CML patients treated with TKIs can achieve a deep molecular response (DMR = BCR-ABL1 minor or equal to0,01% IS), but only 50% of them are reported to maintain a stable Treatment Free Remission (TFR). Since qPCR has some intrinsic limitations in detection and quantification of BCR-ABL1 transcript, this method does not appear optimal in the selection of patients eligible for TKIs cessation. A precise monitoring of BCR-ABL1 transcript levels can help even better the clinicians in managing CML patients treated with TKIs and in selecting the best candidates for discontinuation of TKIs without relapse. Digital PCR (dPCR) can be more advantageous than qPCR. It gives the absolute quantification of target nucleic acidsby partitioning the PCR reaction mix over a large number of wells, each containing a single copy or no copies of the target region. Based on the assumption of Poisson's distribution, the number of template copies originally presenting in the sample can be calculated from the number of partitions in which amplification has successfully occurred. In that way, standard curves cannot be necessary and data are more accurate. In this study we set a dPCR assay to quantify the BCR-ABL1 transcript in a preliminary cohort of CML patients with major (MMR or MR3.0) or deep molecular response (MR4.0, MR4.5 and MR5.0). The analysis by dPCR were based on a TaqMan-MGB probes targeting the BCR-ABL1 transcript. Custom assay was designed and produced with a FAM-label, basing on the sequence of routinely-used probes. The experiments were performed on a QuantStudio 3D Digital PCR System (Life Technologies). A commercial BCR-ABL1 Mbcr standard dilutions (Qiagen) and 10 replicates of blank samples (DNA-free, RNA-free water) were used to set dPCR assay and to determine the Limit of Detection and the Limit of Quantification of our method. The values of absolute quantities of BCR-ABL1 transcript assessed by dPCR were expressed as number of copies/ul. Samples for dPCR testing were obtained from peripheral blood (10 ml in EDTA tubes) of CML patients treated with TKIs, namely imatinib, nilotinib, or dasatinib, on time-checks planned for monitoring MMR or DMR through the conventional qPCR. To the purpose of the study, we evaluated 10 cases with stable MR3.0, 7 cases with stable MR4.0, 6 cases with stable MR4.5, 7 cases with stable MR5.0. It has been considered as stable any MR3.0, MR4.0, MR4.5, or MR5.0 molecular response measured by conventional qPCR and detected in the last three consecutive checks performed during the last 12 months. Blank samples served as negative controls, while 10 peripheral blood samples of healthy donors served as normal controls. Digital PCR (dPCR) revealed different levels of BCR-ABL1 copies/µl among the CML patients achieving the major (MR3.0) or deep (MR4.0, MR4.5 AND MR5.0) molecular response with TKIs. Moving from MR3.0 to MR5.0 molecular response the median of BCR-ABL1 copies/µl assessed by dPCR were progressively decreasing (Figure 1), with blanks and healthy controls approximately to zero. Medians with ranges were 0.957 (0.472-1.692) for MR3.0; 0.319 (0.072-0.906) for MR4.0; 0.231 (0.063-0.651) for MR4.5; 0.219 (0.074-0.399) for MR5.0; 0.076 (0.000-0.118) for healthy controls, and 0.000 (0.000-0.129) for blanks. Moreover, dPCR revealed different BCR-ABL1 copies/µl among the patients of each class of molecular response. Importantly, in patients with MR4.5, MR5.0 and with undetectable levels of BCR-ABL1 % IS as measured with qPCR, discrete variable levels of BCR-ABL1 copies/µl have been detected by dPCR. These data, revealing different levels of BCR-ABL1 copies/µl beyond the limit of detection and quantification of conventional qPCR, may explain why no correlation was observed between BCR-ABL1 % IS levels measured by qPCR and numbers of BCR-ABL1 copies/µl measured by dPCR. We are screening CML patients with MMR and DMR, but these preliminary results show that dPCR appears to be more accurate than qPCR for detection and quantification of BCR-ABL1 transcript and it should be seen as a useful step forward in order to better manage the TKI therapy and to better select the candidates for TFR. Acknowledgments: Department of Clinical and Experimental Sciences, University of Brescia; PRIN2009; European Leukaemia Net; BCC "Pompiano e Franciacorta". Figure 1. Figure 1. Disclosures Tiribelli: Bristol Myers Squibb: Consultancy, Speakers Bureau; Ariad Pharmaceuticals: Consultancy, Speakers Bureau; Novartis Farma: Consultancy, Speakers Bureau. Rosti:Bristol Myers Squibb: Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau. Martinelli:Roche: Consultancy; BMS: Speakers Bureau; MSD: Consultancy; ARIAD: Consultancy; Novartis: Speakers Bureau; Pfizer: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4250-4250 ◽  
Author(s):  
Nicholas L Jackson Chornenki ◽  
Christopher M. Hillis ◽  
Isabelle Bence-Bruckler ◽  
Lambert Busque ◽  
Caroline Hamm ◽  
...  

Abstract Background: Plasma levels of imatinib have been shown to be predictive of disease response in chronic phase chronic myeloid leukemia (CML). However, the ultimate site of action of imatinib is intracellular. While intracellular imatinib has been reported to be correlated with plasma imatinib, the use of intracellular imatinib for the prediction of clinical outcomes is unclear. The concentration of intracellular imatinib depends on many factors including the level of alpha-1-acid glycoprotein, which binds to imatinib and prevents intracellular uptake, and the OCT-1 transporter, which mediates the influx of imatinib across the plasma membrane of leukemic cells. We conducted the present study of newly diagnosed CML patients with the primary objective of determining if intracellular levels of imatinib two weeks after treatment initiation predicted major molecular response. The secondary objectives were to elucidate the relationships between the levels of OCT-1 and plasma imatinib with intracellular imatinib. Methods: We prospectively studied newly diagnosed chronic phase CML patients in Canada who were treated with standard dose imatinib (400 mg). We measured both intracellular and extracellular (plasma) levels of imatinib by tandem mass spectrometry at two weeks, four weeks, and twelve months after enrollment. Additionally, we measured transcript levels of OCT-1 and BCR-Abl by q-rt-PCR before treatment, at six months, and at twelve months to determine therapeutic response. Results: Eighty-one patients were screened. A total of 76 patients entered the study, and 55 completed the study per protocol. Patient information is shown in Table 1. There was a significant correlation between intracellular imatinib levels at two weeks and a 2-log reduction of BCR-Abl transcript at six months (r=0.390; 95% CI = 0.136 - 0.595; p = 0.004) (Figure 1), but not at twelve months (r=0.183; 95% CI = -0.094 - 0.434; p = 0.194). Notably, intracellular imatinib levels and plasma levels of imatinib were highly correlated at two weeks (r=0.698; 95% CI = 0.559-0.799; p <0.001) and four weeks (0.699; 95% CI = 0.555 - 0.802; p < 0.001) but not between four weeks and twelve months (r= 0.230; 95% CI = -0.040 - 0.469; p = 0.094). There was no correlation between intracellular imatinib levels at two weeks and the pre-treatment OCT-1 transcript level (r=0.072; 95% CI = -0.166 to 0.301; p =0.556). Conclusions: Intracellular imatinib levels at two weeks was moderately predictive of a disease response at six months as indicated by a 2-log reduction in BCR-Abl transcript. OCT-1 transcript levels did not have utility for predicting intracellular imatinib levels. Measurement of intracellular Imatinib levels may prove to have utility in identifying patients who would benefit from adjustments to therapy. Disclosures Hillis: Bristol-Myers Squibb: Honoraria; Novartis: Honoraria. Busque:Novartis: Consultancy; Pfizer: Consultancy; Paladin: Consultancy; BMS: Consultancy. Lipton:Bristol-Myers Squibb: Consultancy, Research Funding; ARIAD: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding. Savoie:Pfizer: Consultancy; Novartis: Consultancy, Honoraria; BMS: Consultancy, Honoraria. Leber:Novartis Canada: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis Canada: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2713-2713
Author(s):  
Sarah Huet ◽  
Denis Souche ◽  
Jean-Pierre Magaud ◽  
Pascale Cony-Makhoul ◽  
Yann Guillermin ◽  
...  

Abstract Introduction Recent studies demonstrated that early molecular response was predictive for outcome in chronic phase chronic myeloid leukemia (CML) patients on imatinib (IM) (Hanfstein et al., Leukemia 2012, Marin et al., JCO 2012). It is thus essential to identify patients likely to achieve better long-term outcome if they are switched early to second-line therapy. In reference laboratories, BCR-ABL1 transcript levels are expressed as BCR-ABL/ABL ratios (%) according to the international scale (IS, Hughes et al., Blood 2006). Nevertheless, for high levels of BCR-ABL1, the copy number of total-ABL1 comprises a part of BCR-ABL1, which results in underestimation of the ratio at diagnosis. But the BCR-ABL1 levels for which this underestimation begins to be significant is currently unknown. Besides, since the cut-off value of 10% seems critical to predict outcome for patients treated with IM for 3 months, it stresses the necessity of a precise quantification at this level. In this study, we evaluated the ability of GUS, identified as another suitable control gene (CG) (Beillard et al., Leukemia 2003), to accurately define the relevant thresholds for known therapeutic decision levels (10% and major molecular response MMR: 0.1%) in a series of front line therapy patients on IM. Methods We collected 141 blood samples of CML patients between August 2000 and December 2012, including a first group of diagnosis samples (n= 83) and follow-up samples chosen according to their transcript value: the second group with a BCR-ABL1/ABL1 ratio between 6 and 14% (n=21) and the third group at the MMR threshold of 0.1% (n=37). The expression of BCR-ABL1, total-ABL1 and GUS were quantified as previously described (Gabert et al., Leukemia 2003). Results were expressed as percent ratios relative either to ABL1, with original values converted to IS, or to GUS, without transformation of the original values. Results The mean BCR-ABL1/ABL1 ratio was 55.91% (range: 34.83-98.01%) at diagnosis, 9.66% (range: 6.72-13.53%) in the second group (6-14% values) and 0.093% (range: 0.05-0.18%) in the third group (MMR patients). To determine the impact of GUS as CG on the MMR threshold, we compared in the third group the copy number ratios of BCR-ABL1 relative either to ABL1 (=0.093%) or GUS. The BCR-ABL1/GUS mean value was 0.036%. Thus, the translation of the MMR threshold from BCR-ABL1/ABL1 to BCR-ABL1/GUS would need the introduction of an additional conversion factor (Figure 1). This factor is defined as the ratio of (BCR-ABL1/ABL1)/(BCR-ABL1/GUS) =GUS/ABL1 for each measurement and its mean was valued at 2.774 (2.507-3.040, 95% confidence interval). To assess whether this factor was constant within the different disease time, we next calculated it for the two other groups (figure 2). In diagnosis samples, the mean value of the factor was 3.772 (3.554-3.991, 95%CI) whereas it valued at 2.668 (2.263-3.072, 95%CI) in samples with a 6-14% BCR-ABL1/ABL1 ratio. Comparison of means showed a significant difference between the 3 groups (p<0.001, ANOVA test). This difference over time in the course of the disease was due to GUS overexpression in CML cells at diagnosis, GUS overexpression being superior to the overestimation of ABL1 at diagnosis due to the quantification test bias (Figure 3). This overexpression of GUS, previously reported (Beillard et al., Leukemia 2003), impacts the BCR-ABL1/GUS ratio at diagnosis and warrants the use of a unique translation factor for all disease levels. The number of copies of GUS did not differ significantly between the 6-14% samples and MMR samples (p=0.123, Student's T-Test), nor did the ABL1 copy number (p=0.06). This means that the quantification bias observed for diagnosis samples does not impact the values under 14%. Conclusion The use of GUS as CG for the MRD would require defining the factor allowing translation between BCR-ABL1/GUS and BCR-ABL1/ABL1 ratios, which implies a new standardization of the international scale. Nevertheless, this factor seems to depend on the transcripts levels, since the value of BCR-ABL1/GUS obtained at diagnosis is impacted by the higher GUS copy number in CML cells. As the copy number of ABL1 and GUS did not differ significantly between the groups of patients with 0.1% and 6-14% BCR-ABL1/ABL1 ratio, we concluded that there is no quantification bias at these critical therapeutic decision levels. As a consequence, ABL1 as GUS may be used efficiently as CG to assess early response to TKI. Disclosures: Cony-Makhoul: BMS: Honoraria. Michallet:MSD: Consultancy, Honoraria, Research Funding; Astellas: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Genzyme: Consultancy, Honoraria, Research Funding. Nicolini:Novartis, Ariad, Teva, BMS and Pfizer: Honoraria from Novartis, Ariad, Teva, BMS and Pfizer. Grants from Novartis. Other.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. LBA-6-LBA-6 ◽  
Author(s):  
Jerald P. Radich ◽  
Kenneth J. Kopecky ◽  
Suzanne Kamel-Reid ◽  
Wendy Stock ◽  
Elisabeth Paietta ◽  
...  

Abstract Abstract LBA-6 Background. The optimal tyrosine kinase inhibitor (TKI) for patients (pts) with newly diagnosed CML-CP is unknown. While dasatinib (DAS) is a more potent TKI in vitro than imatinib (IM), it is unclear if this will translate into improved long-term clinical outcomes for pts with newly diagnosed CML-CP. In this open-label phase II trial pts with newly diagnosed CML-CP were randomized to IM 400 mg po qd or DAS 100 mg po qd by four North American cooperative groups (SWOG, ECOG, CALGB, NCIC-CTG). The primary endpoint was >4 log reduction in BCR-ABL transcript at 12 months (mos). The study design, with 240 evaluable pts, provided >90% power to detect a difference in this endpoint of >20 percentage points (two-sided alpha=5%). Patients. 253 pts were randomized (12/2006 to 2/2009). Seven were ineligible, primarily due to diagnosis other than CML-CP, or nonevaluable because they received no protocol treatment or withdrew consent. Pretreatment characteristics were balanced between the arms. Treatment outcomes. Outcomes of the 246 included pts (age 18–90, median 49; 60% male; 35% / 30% with Hasford intermediate / high risk) are shown in Table 1. Eighteen DAS (15%) and 13 IM 400mg (11%) pts discontinued study drug because of a variety of toxicities. Eleven pts (3 DAS [2%], 8 IM 400mg [7%]) discontinued due to refusal, and 36 others (12 DAS [10%], 24 IM 400mg [20%]) for other reasons, most often physician or pt concerns about inadequate response, recurrence or progression. Molecular response at 12 months was deeper in the DAS arm (median 3.3 log reduction in BCR-ABL transcript level vs 2.8 with IM 400mg; Wilcoxon P=0.048), although the proportions achieving >4 log or >4.5 log reductions did not differ significantly (molecular response at 12 mos was based on 189 rather than the planned 240 pts, but this provided >80% power to detect a difference of >20 percentage points). The rates of hematologic CR (HemCR) and cytogenetic CR (CCyR) were not significantly higher with DAS, though 11% and 5% of DAS and IM 400mg pts were not adequately assessed for HemCR, and CCyR data were only available for 51% of pts. Overall survival (OS) and progression-free survival (PFS) were similar in the two arms, with very few deaths, relapses or progressions. Among pts with HemCR, 2-year relapse-free survival was 97% in the DAS arm, 95% in the IM 400mg arm. Toxicity. There were no fatal toxicities. The most common grade 3 and 4 toxicities were hematologic, including thrombocytopenia (<50×109/L) in 18% and 8% of DAS and IM 400mg pts, respectively (P=0.024). A variety of grade 4 non-hematologic toxicities were reported for 6% of DAS pts but no IM 400mg pts. An additional 30% and 17% of DAS and IM 400mg pts had a variety of grade 3 non-hematologic toxicities, while another 57% and 79% had non-hematologic grade 1–2 toxicities. Pleural effusion of any grade was reported for 11% and 2% of DAS and IM 400mg pts (P=0.0017); <2% in either arm were grade 3. Deaths. Seven pts have died, all >8 months after entering the study. Three DAS pts died: one at 8 months after progression to blast crisis, one from lung cancer diagnosed 10 months after DAS started, and one in an automobile accident. Two IM 400mg pts died of CML, and two others (ages 70 and 75 at treatment start) of cardiac arrest unrelated to CML or treatment. Conclusions. Both IM 400mg and DAS are highly effective and generally well-tolerated therapies for newly diagnosed CML-CP. DAS induced deeper molecular responses at 12 months, but not significantly higher rates of >4 log or >4.5 log reduction in BCR-ABL, compared to IM 400mg. 12-month PFS and OS were similar between the two arms, with very few events so far. DAS was associated with more grade 3–4 toxicity. Clinical follow-up is continuing to study whether the short-term deeper molecular response seen with DAS will translate into improved long-term outcomes. Disclosures: Radich: Novartis: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy; Pfizer: Consultancy. Kopecky:Bristol-Myers Squibb: Research Funding. Kamel-Reid:Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding. Larson:Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Emanuel:Novartis: Research Funding; Bristol-Myers Squibb: Research Funding; Genzyme: Consultancy. Lipton:Novartis: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding. Deininger:Bristol-Myers Squibb: Advisory Board, Consultancy, Research Funding; Novartis: Advisory Board, Consultancy. Druker:MolecularMD: Equity Ownership; Novartis: Clinical Trial Funding; Bristol-Myers Squibb: Clinical Trial Funding.


Sign in / Sign up

Export Citation Format

Share Document