scholarly journals Plasma concentration guided dosing of drugs used for the treatment of childhood leukaemias: protocol for a systematic review

BMJ Open ◽  
2022 ◽  
Vol 12 (1) ◽  
pp. e053308
Author(s):  
Madelé van Dyk ◽  
Chelsea Boylan ◽  
Robin Michelet ◽  
Anna M Mc Laughlin ◽  
Ganessan Kichenadasse ◽  
...  

IntroductionChildhood leukaemia is the most common type of cancer in children and represents among 25% of the diagnoses in children <15 years old. Childhood survival rates have significantly improved within the last 40 years due to a rapid advancement in therapeutic interventions. However, in high-risk groups, survival rates remain poor. Pharmacokinetic (PK) data of cancer medications in children are limited and thus current dosing regimens are based on studies with small sample sizes. In adults, large variability in PK is observed and dose individualisation (plasma concentration guided dosing) has been associated with improved clinical outcomes; whether this is true for children is still unknown. This provides an opportunity to explore this strategy in children to potentially reduce toxicities and ensure optimal dosing. This paper will provide a protocol to systematically review studies that have used dose individualisation of drugs used in the treatment of childhood leukaemias.Methods and analysisSystematic review methodology will be applied to identify, select and extract data from published plasma guided dosing studies conducted in a paediatric leukaemia cohort. Databases (eg, Ovid Embase, Ovid MEDLINE, Ovid Cochrane) and clinical trial registries (CENTRAL, ClinicalTrials.gov and ISRCTN) will be used to perform the systematic literature search (up until February 2021). Only full empirical studies will be included, with primary clinical outcomes (progression-free survival, toxicities, minimal residual disease status, complete cytogenetic response, partial cytogenetic response and major molecular response) being used to decide whether the study will be included. The quality of included studies will be undertaken, with a subgroup analysis where appropriate.Ethics and disseminationThis systematic review will not require ethics approval as there will not be collection of primary data. Findings of this review will be made available through publications in peer-reviewed journals and conference presentations. Gaps will be identified in current literature to inform future-related research.PROSPERO registration numberCRD42021225045.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3788-3788 ◽  
Author(s):  
Vikram Gota ◽  
Anupam Sharma ◽  
Amey Paradkar ◽  
Anand Patil ◽  
Navin Khattry ◽  
...  

Abstract Abstract 3788 Introduction: Imatinib mesylate has demonstrated excellent efficacy as first-line therapy for treatment of chronic phase chronic myelogenous leukemia (CML-CP). The drug has a high inter-patient variability in pharmacokinetics (PK), and several factors are presumed to influence its PK. Imatinib is a substrate of CYP3A4 and CYP3A5 enzymes. However, the clinical significance of its metabolism on pharmacokinetics and therapeutic response is still unclear. Significantly higher in vivo CYP3A activity was reported recently in patients achieving complete molecular response than those who did not in a small cohort study. Several investigators in the past have tried to optimize imatinib dosing based on trough levels after the fourth treatment week. Picard S et al (Blood, 2007) showed that threshold plasma concentration of 1002 ng/ml on day 29 was associated with higher incidence of major molecular response. The primary objective of this study was to evaluate the association between CYP3A4 polymorphisms, trough plasma concentration of imatinib on day 8 and day 29 of treatment, and cytogenetic response. The secondary objective was to evaluate the potential of early (day 8) trough level monitoring to discriminate between responders and non-responders. Methods: All patients received standard 400 mg OD dose of imatinib. A single blood sample was collected on day 8 and day 29 prior to imatinib dosing for the determination of trough plasma concentration. Imatinib level was determined using High Performance Liquid Chromatography (HPLC). Cytogenetic response was assessed at 3 monthly intervals by Fluorescence In Situ Hybridization (FISH). Complete cytogenetic response (CCR) was defined as 0% of Philadelphia chromosome–positive cells in the bone marrow aspirate. Pharmacogenetic sample was collected from each patient at the beginning of the study to look for CYP3A4 polymorphism. Genotyping was carried out for *4 (rs55951658), *5 (rs55901263) and *18 (rs28371759) variants by PCR-RFLP technique. About 10% samples were chosen randomly for direct sequencing to confirm the PCR-RFLP findings. Median imatinib concentration in the CCR and non CCR groups was compared using non-parametric Mann-Whitney test. Receiver Operating Characteristic (ROC) curve analysis was performed to assess the discrimination potential of trough imatinib plasma levels for complete cytogenetic response (CCR). Results: Ninety eight patients were enrolled on this study, 70 males and 28 females. The median age was 33 years (Range 6 – 71 years). High interpatient variability in the trough concentration of imatinib was observed (Coefficient of variation = 64% and 56% for day 8 and day 29 respectively). However, none of the patients were found to have *4, *5 or *18 variant alleles. Genotype–PK or genotype-response association could not be evaluated as a result. Data on cytogenetic response at 12 months was available for 75 patients (60 CCR, 15 without CCR), 46 of whom also had trough imatinib levels measured on day 8 and day 29 (36 CCR, 10 without CCR). Median day 8 and day 29 trough concentrations were not significantly different in the group with CCR (1.71 and 1.91 μg/mL respectively) and the group without (1.31 and 1.64 μg/mL respectively). Regarding the discrimination potential of trough levels for CCR, day 8 concentrations failed to discriminate between responders and non-responders, whereas for the Day 29 levels, the area under the ROC curve (AUC) was 0.554, with best sensitivity (75%) and specificity (61%) at plasma threshold of 1.69 μg/mL. Conclusion: In this cohort of patients, common CYP3A4 variants did not contribute to the high inter-patient variability of imatinib levels. Early monitoring on Day 8 failed to discriminate responders from non-responders. Day 29 levels are a better predictor of complete cytogenetic response as shown in previous studies. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 25 (8) ◽  
pp. 742-745
Author(s):  
Akiko Inoue ◽  
Chiyo K. Imamura ◽  
Hiroyuki Shimada ◽  
Daisuke Katayama ◽  
Keisuke Urabe ◽  
...  

Bosutinib is a second-generation tyrosine kinase inhibitor indicated for treatment of chronic myeloid leukemia (CML) in adult patients. The safety and efficacy of bosutinib in patients younger than 18 years of age have not been established. We here report the case of a 4-year-old male with CML who was treated with bosutinib during coordination of human leukocyte antigen–matched unrelated bone-marrow transplantation because of insufficient responses to imatinib and dasatinib. The patient achieved a complete cytogenetic response immediately after starting bosutinib at 180 mg/day (290 mg/m2/day). Because toxicity was tolerable, the dose was increased to 200 mg/day (330 mg/m2/day). A complete cytogenetic response was maintained, but a major molecular response was not achieved 6 months after initiation of treatment with bosutinib. At steady state, maximum plasma concentration, minimum plasma concentration, and area under the plasma concentration-time curve were 89.2 ng/mL, 16.7 ng/mL, and 1017.4 ng·hr/mL, respectively, at 290 mg/m2/day; and 141.1 ng/mL, 18.9 ng/mL, and 1278.5 ng·hr/mL, respectively, at 330 mg/m2/day. To the best of our knowledge, this is the first case report to show the pharmacokinetics of bosutinib with efficacy and safety in a pediatric patient with CML. This rare case in a very young child with CML can also be valuable reference for clinical practice.


2021 ◽  
Vol 28 ◽  
Author(s):  
Adeleh Sahebnasagh ◽  
Farhad Najmeddin ◽  
Atabak Najafi ◽  
Fatemeh Saghafi ◽  
Amin Salehi-Abargouei ◽  
...  

Background: The pathophysiology of cardiac arrest (CA) involves over-activation of systemic inflammatory responses, relative adrenal insufficiency, and glycocalyx damage. Corticosteroids have beneficial effects in preventing the perturbation of the endothelial glycocalyx. Objectives: The aim of this systematic review was to determine the efficacy of glucocorticoids in patients with cardiac arrest. Methods: We searched PubMed, Scopus, ISI Web of Science, Google Scholar and Cochrane central register for relevant clinical trials and cohort studies until September 2019. Results: We retrieved 7 peer-reviewed published studies for the systematic review. Two studies were clinical trials evaluating 147 patients, while five illustrated cohort design, evaluating 196,192 patients. In total, 196,339 patients were assessed. There was limited evidence and conflicting results to establish a correlation between glucocorticoids and the survival of patients suffering from cardiac arrest. However, the link between these medications and survival-to-admission, survival-to discharge, and 1-year survival rates was strong and consistent in observational studies. Conclusion: The clinical evidence regarding the efficacy and safety of glucocorticoids in CA is limited to observational studies with inconsistent methodology and few clinical trials with small sample size. Nevertheless, it seems that glucocorticoid supplementation during and after cardiopulmonary resuscitation (CPR) may have significant effects in terms of survival-to-admission, survival to discharge, 1-year survival rates and an improved return of spontaneous circulation (ROSC) rate, especially in patients with hemodynamic instability and cardiovascular diseases (i.e., refractory hemodynamic shock). Future studies with high-quality, large-scale, long-term intervention and precise baseline characteristics are needed to evaluate the exact effective dose, duration, and efficacy of glucocorticoids in CA.


Materials ◽  
2021 ◽  
Vol 14 (12) ◽  
pp. 3251
Author(s):  
Francesca Delucchi ◽  
Emanuele De Giovanni ◽  
Paolo Pesce ◽  
Francesco Bagnasco ◽  
Francesco Pera ◽  
...  

The purpose of this systematic review was to investigate the clinical outcomes of frameworks made of different materials in patients with implant-supported full-arch prostheses. A literature search was conducted on MEDLINE, Scopus and Cochrane Library, until the 1st of March 2021, with the following search terms: framework or substructure combined with “dental implants”. The outcomes evaluated were: implant and prosthesis survival, bone resorption, biological and technical complications. The Cochrane Handbook for Systematic Reviews of Interventions was employed to assess the risk of bias in randomized clinical trials. The Newcastle–Ottawa quality assessment scale was used for non-randomized studies. In total, 924 records were evaluated for title and abstract, and 11 studies were included in the review: 4 clinical randomized trials and 7 cohort studies. The framework materials investigated were: gold alloy, titanium, silver-palladium alloy, zirconia and polymers including acrylic resin and carbon-fiber-reinforced composites. High implant and prosthetic cumulative survival rates were recorded by all included studies. Various materials and different fabrication techniques are now available as alternatives to traditional cast metal frameworks, for full-arch implant-supported rehabilitations. Further long-term studies are needed to validate the use of these materials and clarify their specific clinical indications and manufacturing protocols to optimize their clinical outcomes.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1025-1025
Author(s):  
Jennifer L Beaumont ◽  
Cindy Nowinski ◽  
John Coombs ◽  
Tomasz Szczudlo ◽  
Rick E. Blakesley ◽  
...  

Abstract Abstract 1025 Background: Nilotinib is a more potent and more selective inhibitor of BCR-ABL in-vitro than imatinib. A randomized Phase III study was conducted comparing these two therapies in adult patients with newly diagnosed Philadelphia Chromosome positive (Ph+) chronic myelogenous leukemia (CML) in chronic phase (CP). Aim: To evaluate whether baseline health-related quality of life (HRQL) or symptom scores were predictive of clinical outcome in this phase III trial. Methods: A total of 846 patients were randomized to receive nilotinib 300 mg BID (n=282), nilotinib 400 mg BID (n=281) or imatinib 400mg QD (n=283). For these analyses to evaluate the impact of baseline HRQL on clinical outcomes, the treatment arms were combined. HRQL was assessed using the Functional Assessment of Cancer Therapy–Leukemia (FACT-Leu). The FACT-Leu consists of four general subscales measuring physical, social/family, emotional, and functional well-being and a 17-item leukemia-specific subscale (LeuS). The FACT-G score is the sum of the four general well-being subscales. The questionnaire was administered at baseline, and at 3, 12, and 24 months. Baseline FACT-G and LeuS scores were divided into three equal-sized groups (i.e., tertiles). Patients with high, mid, and low baseline scores were compared on several clinical outcomes: best molecular response by Month 24, best cytogenetic response by Month 24, treatment discontinuation, hospitalization, dose modification of any kind (interruption, increase, or reduction), grade 3 or 4 adverse events, and missed cytogenetic tests. Chi-square tests were used to compare these dichotomous and categorical outcomes between baseline HRQL groups. The relationship between baseline HRQL scores and Sokal Risk groups was evaluated using analysis of variance. Higher scores on the FACT-G and LeuS indicate better HRQL and less symptom burden. Results: Mean baseline FACT-G scores were 97.8, 85.0, and 66.3, and mean baseline LeuS scores were 61.6, 53.8, and 42.2, for the high/mid/low tertile groups, respectively. There was no significant association between best molecular response by Month 24 or cytogenetic response by Month 24 and baseline FACT-G scores (p=0.149 and p=0.094, respectively). There was an association between best molecular response by Month 24 and baseline LeuS scores (p=0.043) but no significant association with best cytogenetic response by Month 24 (p=0.316). There were no significant associations with either dose modifications (p=0.252 for FACT-G, p=0.643 for LeuS), grade 3 or 4 adverse events (p=0.531 for FACT-G, p=0.831 for LeuS), or missed cytogenetic tests (p=0.722 for FACT-G, p=0.374 for LeuS). There was a significant association between treatment discontinuation and baseline FACT-G scores (p=0.007). Only 18% of patients with the highest baseline FACT-G scores discontinued treatment compared to 26% in the middle group and 31% in the group with the lowest baseline FACT-G scores. This relationship was not statistically significant for baseline LeuS scores (p=0.070). Fourteen percent of patients with high baseline FACT-G scores were hospitalized at some point during the study, compared to 15% of patients with mid FACT-G scores, and 22% with low baseline FACT-G (p=0.099). However, 11% of patients with high LeuS scores were hospitalized compared to 20% of patients with mid-range LeuS scores, and 21% of patients with low LeuS scores (p=0.018). The mean baseline FACT-G scores were 81.4, 83.5, and 83.4 (p=0.288), and the mean baseline LeuS scores were 51.1, 53.4, and 52.5 (p=0.042), for patients with high, intermediate, and low Sokal scores, respectively. Conclusions: In patients with newly diagnosed CML-CP, worse general HRQL at baseline was predictive for treatment discontinuation, but not predictive for best molecular response. Leukemia related symptoms at baseline were associated with a greater likelihood of subsequent hospitalization and moderately associated with molecular response. Baseline HRQL was not clearly associated with Sokal scores. These findings suggest that among patients with newly diagnosed CML-CP, examination of baseline HRQL and symptoms may allow patients and clinicians to better anticipate outcomes such as hospitalizations and continuation of therapy. Disclosures: Beaumont: Novartis: Research Funding. Nowinski:Novartis: Research Funding. Coombs:Novartis: Employment, Equity Ownership. Szczudlo:Novartis: Employment, Equity Ownership. Blakesley:Novartis: Employment, Equity Ownership. Gallagher:Novartis: Employment, Equity Ownership. Burns:Novartis: Research Funding. Cella:Novartis: Research Funding.


2017 ◽  
Vol 46 (14) ◽  
pp. 3541-3549 ◽  
Author(s):  
Filippo Familiari ◽  
Mark E. Cinque ◽  
Jorge Chahla ◽  
Jonathan A. Godin ◽  
Morten Lykke Olesen ◽  
...  

Background: Cartilage lesions are a significant cause of morbidity and impaired knee function; however, cartilage repair procedures have failed to reproduce native cartilage to date. Thus, osteochondral allograft (OCA) transplantation represents a 1-step procedure to repair large chondral defects without the donor site morbidity of osteochondral autograft transplantation. Purpose: To perform a systematic review of clinical outcomes and failure rates after OCA transplantation in the knee at a minimum mean 2 years’ follow-up. Study Design: Systematic review; Level of evidence, 4. Methods: A systematic review of the literature regarding the existing evidence for clinical outcomes and failure rates of OCA transplantation in the knee joint was performed using the Cochrane Database of Systematic Reviews, the Cochrane Central Register of Controlled Trials, PubMed, and MEDLINE from studies published between 1980 and 2017. Inclusion criteria were as follows: clinical outcomes and failure rates of OCAs for the treatment of chondral defects in the knee joint, English language, mean follow-up of 2 years and minimum follow-up of 18 months, minimum study size of 20 patients, and human studies. The methodological quality of each study was assessed using a modified version of the Coleman methodology score. Results: The systematic search identified 19 studies with a total of 1036 patients. The mean 5-year survival rate across the studies included in this review was 86.7% (range, 64.1%-100.0%), while the mean 10-year survival rate was 78.7% (range, 39.0%-93.0%). The mean survival rate was 72.8% at 15 years (range, 55.8%-84.0%) and 67.5% at 20 years (range, 66.0%-69.0%). The weighted mean patient age was 31.5 years (range, 10-82 years), and the weighted mean follow-up was 8.7 years (range, 2-32 years). The following outcome measures showed significant improvement from preoperatively to postoperatively: d’Aubigné-Postel, International Knee Documentation Committee, Knee Society function, and Lysholm scores. The weighted mean reoperation rate was 30.2% (range, 0%-63%). The weighted mean failure rate was 18.2% (range, 0%-31%). Of note, revision cases, patellar lesions, and bipolar lesions demonstrated worse survival rates. Conclusion: Improved patient-reported outcomes can be expected after OCA transplantation, with a survival rate of 78.7% at 10 years. Revision cases, patellar lesions, and bipolar lesions were associated with worse survival rates; therefore, utilization of the most appropriate index cartilage restoration procedure and proper patient selection are key to improving results.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3603-3603
Author(s):  
Massimo Breccia ◽  
Luigia Luciano ◽  
Mario Annunziata ◽  
Imma Attolico ◽  
Alessandra Malato ◽  
...  

Abstract Background. Ponatinib is a third-generation tyrosine kinase inhibitor indicated for adults with resistant or intolerant chronic phase (CP), accelerated phase (AP), or blast phase (BP) chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia, or those carrying the T315I mutation. Ponatinib has been commercially available since January 2015, yet there is a paucity of data on its use in the real-world setting. The goal of the Observational study of Iclusig ® (ponatinib) Treatment in patients with CML in Italy (OITI) was to evaluate treatment patterns and outcomes, including safety and efficacy, in patients with CML treated in Italy since the approval of ponatinib. Methods. This noninterventional study included patients aged ≥18 years with CP, AP, or BP CML who initiated ponatinib treatment in routine clinical practice across 26 Italian centers (academic and hospital settings). The study population comprised a prospective cohort, including patients who started treatment with ponatinib after site activation during the 12-month enrollment period; a retrospective cohort, including patients who started treatment with ponatinib but died or were lost to follow-up prior to site activation; and a retrospective/prospective cohort, including patients who started treatment with ponatinib prior to site activation and were still on treatment or in follow-up at the end of the study. Demographic, efficacy, and safety data were collected from patient medical charts at study entry and routine visits. The primary endpoint was the complete cytogenetic response (CCyR) rate in CP CML patients 6 months after starting ponatinib. In the absence of cytogenetic evaluation, molecular assessment was used, considering patients in MR2 or better to be in CCyR. Here, the primary analysis of all evaluable patients for the primary endpoint (median follow-up, 23.7 months [range, 1.3-70.8]) is presented. Results. A total of 119 patients (110 CP, 6 AP, and 3 BP CML) were analyzed. Fifty-nine (49.6%) received ponatinib in second-line (2L), 41 (34.4%) in 3L, and 19 (16.0%) in ≥4L. Prior cardiovascular disease/hypertension was recorded in 56 (47.1%) patients. Median age at ponatinib start was 60 years (range, 19-93). Of 68 evaluated patients, 24 (35.3%) had a confirmed ABL1 mutation, including 7 (10.3%) with the T315I mutation. Starting doses of ponatinib were 45 mg (37.0%), 30 mg (41.2%), or 15 mg (21.8%). Median treatment duration was 22.8 months (range, 1.4-73.6). At baseline, 56 patients with CP CML had less than CCyR and 53 were in CCyR. For 1 patient, assessment was not available. At 6 months, 80/107 evaluable patients with CP CML were in CCyR; 29/56 (51.8%) achieved and 50/50 (100%) maintained CCyR compared to baseline, respectively. For 1 patient, baseline data were unavailable. Additionally, 37 (34.6%) and 19 (17.8%) patients with CP CML achieved a major molecular response (MMR; MR3) and a deep molecular response (MR4-MR5), respectively (Table 1). Progression-free survival rates estimated for patients with CP CML at 12 and 24 months were 92.6% (95% CI, 87.8-97.7%) and 84.6% (95% CI, 77.2-92.6%), respectively. Corresponding overall survival rates were 95.4% (95% CI, 91.6-99.4%), and 88.4% (95% CI, 81.7-95.7%). Seventy-one (59.7%) patients had treatment-related adverse events (AEs), most commonly rash, hypertension (Grade 1-2), and thrombocytopenia (Grade 3). Treatment-related arterial occlusive events occurred in 2 (1.7%) patients. Dose modifications occurred in 77 patients: 37.1% were due to AEs, 13.5% were reductions after at least major cytogenetic response, 10.6% were increases due to lack of efficacy, 1.7% were medical decisions, and 37.1% were for other reasons. Thirty-three patients discontinued ponatinib due to AEs (33.3%), medical decisions (24.2%), and other reasons (42.4%), such as death, progression, and hematopoietic stem cell transplantation. Conclusions. This observational study demonstrates that ponatinib has a favorable efficacy and safety profile in patients with CML treated in standard clinical practice. By 6 months, 74.8% of evaluable patients were in CCyR and 52.3% achieved at least MMR. Further, the probability of survival at 2 years was &gt;88%. No new safety signals emerged with ponatinib treatment compared to prior studies. The early use of ponatinib and careful dose selection appear key to the safety and efficacy outcomes observed in this real-world study. Figure 1 Figure 1. Disclosures Breccia: Abbvie: Honoraria; Bristol Myers Squibb/Celgene: Honoraria; Incyte: Honoraria; Novartis: Honoraria; Pfizer: Honoraria. Abruzzese: Novartis: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Bristol Myers Squibb: Consultancy, Honoraria; Incyte: Consultancy, Honoraria. Bonifacio: Pfizer: Honoraria; Amgen: Honoraria; Bristol Myers Squibb: Honoraria; Novartis: Honoraria. Castagnetti: Novartis: Consultancy, Honoraria; Incyte: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Bristol Myers Squibb: Consultancy, Honoraria. Galimberti: Inctye Bioscience Italy Srl: Current Employment. Iurlo: Incyte: Speakers Bureau; Novartis: Speakers Bureau; Pfizer: Speakers Bureau; Bristol Myers Squibb: Speakers Bureau.


2006 ◽  
Vol 24 (3) ◽  
pp. 454-459 ◽  
Author(s):  
Ilaria Iacobucci ◽  
Gianantonio Rosti ◽  
Marilina Amabile ◽  
Angela Poerio ◽  
Simona Soverini ◽  
...  

Purpose Imatinib mesylate is a potent inhibitor of BCR-ABL, the constitutively active tyrosine kinase protein critical for the pathogenesis of chronic myeloid leukemia. Patients and Methods We reviewed 284 patients with late chronic-phase Philadelphia chromosome (Ph) –positive chronic myeloid leukemia treated with imatinib 400 mg daily after interferon-α failure. In a retrospective study, we evaluated the pattern and rapidity of the response to imatinib, comparing the cytogenetic and molecular responses, progression-free and overall survival rates in patients who obtained a complete cytogenetic response within 1 year of treatment (early responders), and in patients where a complete cytogenetic response was detected after 12 months (late responders). Results After 3 or 4 years of treatment, the molecular response of the late cytogenetic responders was similar to that of the early cytogenetic responders. At 36 months of treatment the amount of residual disease measured by standardized quantitative reverse-transcriptase polymerase chain reaction was 0.00047 in late responders versus 0.00022 in early responders, and at 48 months it was 0.00019 versus 0.00026 (median values, P value = nonsignificant). The estimated 4-year progression-free survival rate was 88% for early responders and 100% for late responders, while the estimated 4-year overall survival rates were 92% and 100% for early and late responders, respectively. Conclusion The sensitivity and the response (cytogenic and molecular) to imatinib may require 1 year or more. Long-term follow-up results continue to improve in terms of rates and durability of the complete cytogenetic response, major or complete molecular response, and progession-free and overall survival.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1375-1375 ◽  
Author(s):  
Stephanie Haiat ◽  
Xavier Decleves ◽  
Benedicte Mittaine ◽  
Ollivier Legrand ◽  
Stephanie Francart ◽  
...  

Abstract Imatinib exerts a potent and selective inhibition of tyrosine kinase Bcr-Abl. It is currently used for treatment of chronic myeloid leukaemia (CML) and acute leukaemia (AL). Pharmacokinetic data indicate large variability especially in plasma exposure with many factors which could be involved (absorption, metabolism, drug-drug interactions). Compliance must also be taken in count. The aim of this study was to determine the variability of imatinib plasma concentrations in a « natural clinical setting» (outpatients on long-term treatment for CML or AL). Secondary objective was to perform a pilot assessment of the relationship between imatinib plasma concentration and clinical outcomes. Analysis was performed by HPLC. Sample treatment (500 μL of patient plasma) consisted of a liquid-solid extraction. Imatinib and internal standard (CGP 53716) were eluted on a Lichrosorb® RP8 column with methanol-THF-sodium acetate buffer 0.1M pH 5. Detection was performed with UV spectrophotometer (262 nm). The limit of quantification was set at 200 ng/mL. Within-day and inter-day precisions were lower than 15%. Blood samples were collected at steady-state (trough and peak values, ie before and 3h after drug administration) in outpatients treated for CML or AL with one imatinib daily dose, at clinical visits. Clinical evolution was considered as successful when following responses were obtained: complete haematological response within 3 months, or major cytogenetic response within 6 months, or complete cytogenetic response within 12 months, or major molecular response within 18 months in the CML group, and complete cytogenetic response after induction in the AL group. 68 imatinib measurements were obtained from 24 patients (13 CML and 11 AL). Mean age was 49 years (range 21–74) and weight was 73 kg (49–100). In patients receiving imatinib 400mg daily dose, trough and peak concentrations were respectively 1.60 ±1.28 (0.5–5.1) μg/mL (n=19), and 3.51 ±3.04 (1.0–11.1) μg/mL (n=9). In patients treated with a daily dose 600mg, trough and peak concentrations were respectively 2.62 ±2.10 (0.8–6.5) μg/mL (n=22), and 5.85 ±4.15 (1.2–15.1) μg/mL (n=18). Variability of trough concentrations of imatinib were 80% in both groups (400 and 600mg). 18 patients were evaluable. 15 efficient responses were obtained and all these patients showed imatinib trough levels above 0.5 μg/mL (target plasma concentration required to induce death of leukaemic cells). 3 patients with failure showed nevertheless imatinib trough levels of 1.5, 1.8 and 4.5 μg/mL. 3 patients showed accumulation of imatinib (trough levels higher than 5.0 μg/mL) due to hepatic impairment but without side effects. The method of analysis is very simple, sensitive and specific. Our results confirm the large variability of imatinib plasma concentrations and are consistent with previous results. These preliminary results showed a « safe » imatinib exposure which leads to therapeutic concentrations (higher than 0.5 μg/mL). Therapeutic drug monitoring seems to be not systematicaly necessary but could be reserved to specific cases of poor compliance, major risks of interaction or cases of failure or resistance. Studies of correlation with a larger cohort of patients is necessary to clarify the role of imatinib therapeutic drug monitoring for improving its use.


Sign in / Sign up

Export Citation Format

Share Document