scholarly journals Reduction of imatinib dose and persistence of complete molecular response after p210 multipeptide vaccine in chronic myeloid leukaemia treated with dose escalation for acquired resistance

Author(s):  
Massimo Breccia ◽  
Monica Bocchia ◽  
Laura Cannella ◽  
Marzia Defina ◽  
Micaela Ippoliti ◽  
...  
Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5201-5201
Author(s):  
Heledd Thomas ◽  
Jane F Apperley ◽  
Richard M Szydlo ◽  
Gareth Gerrard ◽  
David Marin ◽  
...  

Abstract Background There is currently no universal definition of complete molecular response (CMR) in chronic myeloid leukaemia (CML). This has implications for studies of TKI withdrawal as CMR is the main criterion for entry into these trials. We aimed to assess the proportion of patients in our practice who achieved CMR by a variety of definitions. We hoped to identify a definition that was predictive of sustained CMR and could therefore be recommended to predict those patients who might be able to stop treatment in the longer term. Methods We conducted a retrospective analysis of a comprehensive CML database of serial RT-qPCR results of 215 patients achieving deep molecular responses on TKI therapy. The least stringent definition of CMR was defined as BCR-ABL1 transcripts <11 and ABL1 control value (CV)>10,000. The depth of CMR was categorised according to transcript number and CV. Probability of molecular relapse according to depth of CMR at 2 and 5 years from initial CMR (ICMR) was estimated using the Kaplan Meier method. Results Patients with 6-10 transcripts (any CV) were most likely to lose CMR by 2 years (86.4%) while patients with 0 transcripts (any CV) were least likely to do so (7.6%). There was no difference in the probability of molecular relapse between patients with 1-5 transcripts (CV>32,000) and those with 0 transcripts (any CV) (RR at 2 years=1 p=0.945, RR at 5 years=3.7 p = 0.114). Conclusion Depth of CMR predicts molecular relapse at 2 and 5 years from ICMR. CMR4.5 (<6 transcripts) is the least stringent response that is required to sustain CMR. Patients with <6 transcripts (CV>32,000) are equally as likely to sustain CMR as patients with 0 transcripts up to 5 years from ICMR. These patients may be suitable for a stopping trial. Disclosures: Apperley: Ariad: Honoraria; Bristol Myers Squibb: Honoraria; Novartis: Honoraria, Research Funding; Pfizer: Honoraria. Milojkovic:BMS: Honoraria; Pfizer: Honoraria; Ariad: Honoraria; Novartis: Honoraria.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1102-1102 ◽  
Author(s):  
Davi D M Ross ◽  
Andrew Grigg ◽  
Anthony Schwarer ◽  
Christopher Arthur ◽  
Kerryn Loftus ◽  
...  

Abstract After 5 years of imatinib treatment 40–50% of chronic myeloid leukaemia (CML) patients will have stable undetectable BCR-ABL by real-time quantitative RT-PCR (RQ-PCR) using strict sensitivity criteria (‘complete molecular response’, CMR). Many patients who stop imatinib in CMR will relapse, but small numbers have been reported with sustained CMR after imatinib withdrawal. We designed a non-randomised prospective Phase 2 study of imatinib withdrawal in adult chronic phase CML patients in CMR for ≥2 years (ACTRN012606000118505). Patients were treated in multiple centres around Australia, and RQ-PCR for BCR-ABL was performed centrally: monthly for the first year after imatinib withdrawal, and 2-monthly in the second year. Molecular relapse was defined as a single PCR result above the level of major molecular response (MMR) or any two consecutive positive results. Molecular relapse was treated with imatinib and patients were monitored monthly for 12 months to assess response to retreatment. Patients were enrolled in two cohorts: imatinib de novo (IM only, n=5) and imatinib after prior interferon therapy (IFN-IM, n=13). The median duration of prior IFN was 39 months. Both cohorts continue to accrue. For all 18 patients the median age at study entry was 58 years; 44% were male. The median duration of imatinib treatment was 60 months (R40-89). The Kaplan-Meier estimate of the rate of sustained CMR after 12 months off treatment was 67% (95% confidence interval 40–85%, see Figure). Ten of 13 IFN-IM patients (77%) remain in CMR, and 7 of these have been in CMR for at least 12 months without treatment (maximum 23 months). The median follow-up in the IM only patients is currently only 7 months (R1-15), and 3/5 remain in CMR. All molecular relapses in both groups have occurred within 5 months of stopping imatinib. The median duration of prior imatinib treatment was not different in the 5 patients with loss of CMR (76 months) versus those in stable CMR (60 months; p=0.59). Among the 5 patients with loss of CMR the median time to molecular relapse was 3 months (range 2–5 months). Two relapsing patients lost MMR, and 3 had detectable BCR-ABL mRNA below this level. No patient has experienced haematological relapse or developed a kinase domain mutation. At last follow-up all 5 relapsing patients had regained CMR after a median of 5 months of re-treatment with imatinib. Patient-specific DNA Q-PCR assays were developed to test whether minimal residual disease (MRD) was detectable in genomic DNA in patients in CMR defined by RQ-PCR for BCR-ABL mRNA. Results are available for 6 patients, 3 of whom have relapsed. One relapsing patient had BCR-ABL DNA detected prior to imatinib withdrawal. In the remaining 2 relapsing patients BCR-ABL DNA was detected after imatinib withdrawal, but 2–3 months prior to the detection of BCR-ABL mRNA by RQ-PCR. BCR-ABL DNA increased by at least 1-log between the time of the first positive result and the detection of molecular relapse by RQ-PCR. The 3 patients in stable CMR had no detectable BCR-ABL DNA. In conclusion, with close molecular monitoring imatinib withdrawal in stable CMR appears to be safe: currently all patients are either in stable CMR off treatment or back in CMR after re-treatment. Withdrawal of effective treatment outside the setting of a clinical trial is not recommended. Monitoring of MRD by genomic DNA Q-PCR was able to detect molecular relapse prior to mRNA RQ-PCR, and shows promise for the prospective identification of patients at high risk of relapse. There is an apparent dichotomy of response between early molecular relapse and durable CMR, at least in patients treated with imatinib after IFN. It is too early to identify clinical or laboratory factors (such as prior IFN treatment) that may influence the probability of sustained CMR without treatment. Figure Figure


2010 ◽  
pp. NA-NA
Author(s):  
Massimo Breccia ◽  
Fabio Stagno ◽  
Paolo Vigneri ◽  
Roberto Latagliata ◽  
Laura Cannella ◽  
...  

Author(s):  
Hilbeen Hisham Saifullah ◽  
Claire Marie Lucas

Following the development of tyrosine kinase inhibitors (TKI), the survival of patients with chronic myeloid leukaemia (CML) drastically improved. With the introduction of these agents, CML is now considered a chronic disease, for some patients. Taking into consideration the side effects, toxicity, and high cost, discontinuing TKIs became a goal for patients with chronic phase CML. Patients who achieved deep molecular response (DMR) and discontinued TKI, remained in treatment-free remission (TFR). Currently, the data from the published literature demonstrate that 40-60% of patients achieve TFR, with relapses occurring within the first six months. In addition, almost all patients who relapsed regained a molecular response upon re-treatment, indicating TKI discontinuation is safe. However, there is still a gap in the understanding the mechanisms behind TFR, and whether there are prognostic factors that can predict the best candidates who qualify for TKI discontinuation with a view to keeping them in TFR. Furthermore, the information about a second TFR attempt and the role of gradual de-escalation of TKI before complete cessation is limited. This review highlights the factors predicting success or failure of TFR. In addition, it ex-amines the feasibility of a second TFR attempt after the failure of the first one, and the current guidelines concerning TFR in clinical practice.


2018 ◽  
Vol 10 (1) ◽  
pp. 2018027 ◽  
Author(s):  
Ekaterina Chelysheva ◽  
Sergey Aleshin ◽  
Evgenia Polushkina ◽  
Roman Shmakov ◽  
Igor Shokhin ◽  
...  

Breastfeeding in patients with chronic myeloid leukaemia (CML) who take tyrosine kinase inhibitors (TKIs) is not recommended but interruption of TKI treatment may cause the loss of remission. We observed the kinetics of the leukaemic clone in 3 women with CML in accordance with treatment interruptions for pregnancy and breastfeeding. The concentrations of nilotinib and imatinib in maternal milk were measured when the breastfeeding period was over. Nilotinib transfer into human breast milk was demonstrated for the first time and had a maximum concentration (Cmax) 129 ng/ml after 4 hours of the drug intake at a dose of 400 mg. The Cmax of imatinib in maternal milk ranged from 420 to 1411 ng/ml after 4-8 hours of the drug intake at a dose of 400-600 mg. Breastfeeding without TKI treatment may be safe with molecular monitoring, but preferably in those patients with CML who have durable deep molecular response.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3302-3302
Author(s):  
Massimo Breccia ◽  
Fabio Stagno ◽  
Roberto Latagliata ◽  
Paolo Vigneri ◽  
Laura Cannella ◽  
...  

Abstract Abstract 3302 Poster Board III-190 Introduction Imatinib mesylate (IM) given at a daily dose of 400 mg currently represents the gold standard of care for patients with chronic myeloid leukemia (CML) in chronic phase (CP). European LeukemiaNet (ELN) guidelines propose IM dose escalation to rescue those CML patients with either suboptimal response or drug resistance. We report on the long-term efficacy of IM dose escalation in 74 patients with CP-CML after suboptimal response or failure to IM conventional dose. Patients and methods Median age was 50 years (range 19-85), there were 52 males and 22 females. Thirteen patients were classified as hematologic failure (10 primary and 3 secondary), 57 patients as cytogenetic resistance (24 primary and 33 acquired). Three patients escalated the dose for cytogenetic suboptimal response and one patient for molecular suboptimal response at 18 months. Fifty-four received IM dose escalation from 400 to 600 mg and 20 patients from 400 to 800 mg. Results Overall, after a median follow-up of 36 months, 68/74 (91.8%) patients maintained or achieved a complete haematologic response (CHR); this was maintained in all patients who escalated the dose for cytogenetic failure or suboptimal response. A major cytogenetic response (MCyR) was achieved in 41 patients (72%) who escalated the dose for cytogenetic failure and in 6/13 (46%) patients who escalated imatinib for hematologic failure (p=0.002). Overall, complete cytogenetic responses (CCR) were achieved in 27 (37%) out of 74 CML patients: of the 13 hematologic failure patients, only 5 achieved CCyR: all patients had prior acquired resistance to imatinib. Of the 57 cytogenetic failure, 22 reached CCR: this response was obtained in 27% of the primary cytogenetic resistant, and in 50% of the acquired cytogenetic resistant patients (p=0.02). Three patients who escalated the dose for cytogenetic suboptimal response obtained CCR and complete molecular response (CMR), whereas one patient who escalated the dose for molecular suboptimal response at 18 months did not obtain CMR. Median time to cytogenetic response was 3.5 months. Cytogenetic responses occurred in 37/50 patients who escalated the dose to 600 mg and in 10/20 patients who escalated to 800 mg daily (p=0.234). CMR was obtained in 10 patients: in 7 patients who escalated the dose for cytogenetic failure and in 3 patients who escalated imatinib for suboptimal cytogenetic response. Estimated 2 year-progression free survival (PFS) and overall survival (OS) is 87% and 85% respectively. Sixteen patients (21.6%) experienced toxicities and had temporarily IM interruption. Conclusions Imatinib dose escalation can induce sustained responses in a subset of patients with cytogenetic resistance and a prior suboptimal cytogenetic response to standard-dose imatinib, whereas it appears less effective in haematologic failure patients or in molecular sub-optimal responders. The availability of second generation TKI should be taken into account in these letter categories of patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4036-4036
Author(s):  
Alexandra Smith ◽  
Eve Roman ◽  
Daniel Painter ◽  
Debra Howell ◽  
Graeme Smith ◽  
...  

Abstract The use of tyrosine kinase inhibitors (TKIs) has transformed the treatment of chronic myeloid leukaemia (CML), changing it from a disease that was fatal in non-transplant patients to a long-term condition managed by oral medication. However, survival rates in the US SEER populations are much poorer than predicted by clinical trials. The aim of this study was to investigate whether these differences were also apparent in the UK. To address this, we present contemporary (2004-13) population-based (3.6 million) data from an established patient cohort (www.hmrn.org). All of the patients were treated within the UK's National Health Service (NHS) where TKIs are freely available on the basis of clinical need. 242 patients were diagnosed with CML in the study region Sept 2004 to Aug 2011; a crude annual incidence rate of 0.97 per 100,000. Using established methods, time to event analyses was used to calculate overall survival (OS), relative survival (RS), and loss of molecular response; and the quintile distribution of the income domain of the index of deprivation (IMD) was used as marker of socio-economic status. 47 (19.5%) patients died before the 1st April 2013; minimum follow-up 1.5 yrs and maximum 8.5 yrs. The 5-year OS was 78.9% and RS, taking into account background general population mortality, was 88.6% (Fig 1). Only 8 (3.3%) of the 242 patients were not treated with TKIs within the study region: 2 died before treatment could be started, 1 refused treatment, 1 had another cancer, 2 had supportive care only, and 2 moved to another part of the country (both were alive on 1st April 2013). 219 (93.6%) of the 234 treated patients received first-line Imatinib, the remainder receiving Dasatinib as part of a clinical trial. The efficacy of TKI treatment across all ages is demonstrated in Fig 2. Whilst the age-specific crude survival curves continue to diverge (P=0.001), the relative survival curves for the two age groups remain closely aligned (5-year RS< 60 =89.9%, ≥ 60= 87.2%). Gender had little impact on outcome (5-year RS men = 90.1%, women = 89.1%). By contrast, the deprivation-specific RS curves remain as disparate from each other as the OS curves (P=0.0014); the 5-year RS estimates of the most affluent (quintiles 1 to 3) and the least affluent (quintiles 4 to 5) being 94.9% and 79.5% respectively. These deprivation differences could not be explained by variations in acquisition of TKI resistance (n=10) or acquisition of additional cytogenetic abnormalities (n=7), both of which were rare in this population: for deprivation categories1-3 and 4-5 TKI resistance was 4/140 and 6/94 respectively, and for acquisition of additional cytogenetic abnormalities it was 7/140 and 0/94 respectively. As can be seen from Table 1, however, differences in survival associated with deprivation were evident both for molecular response achievement and maintenance; defined here as one or more readings ≤ 0.1 (MMR) or ≤ 1.0 (MR). Whilst the differences by deprivation are not statistically significant in the case of the former, the findings for loss of response combined with death from CML are, as expected, consistent with the findings for survival (Fig 2). In contrast to findings reported by SEER, the outcome for patients with CML in the UK treated mainly with Imatinib is excellent, and almost identical to the results of clinical trials. Age and gender no longer have prognostic significance and incidence of adverse biological events is low and appears sporadic. However, despite free access to TKI therapy, clinical outcomes are significantly poorer in lower socio-economic groups, and this is now one of the strongest predictors of outcome in CML in the UK. Identification of the mechanism of this effect and the design of appropriate interventions could further improve survival. Disclosures: Patmore: Roche: Consultancy, Honoraria. Jack:Roche /Genentech: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document