scholarly journals Phase 1 study of epigenetic priming with decitabine prior to standard induction chemotherapy for patients with AML

Blood ◽  
2011 ◽  
Vol 118 (6) ◽  
pp. 1472-1480 ◽  
Author(s):  
Joseph M. Scandura ◽  
Gail J. Roboz ◽  
Michelle Moh ◽  
Ewelina Morawa ◽  
Fabienne Brenet ◽  
...  

Abstract We conducted an open-label phase 1 study exploring the feasibility, safety, and biologic activity of epigenetic priming with decitabine before standard induction chemotherapy in patients with less-than-favorable risk of acute myelogenous leukemia (AML). We directly compared the clinical and DNA-hypomethylating activity of decitabine delivered at 20 mg/m2 by either a 1-hour infusion (Arm A) or a continuous infusion (Arm B) for 3, 5, or 7 days before a single, standard induction with infusional cytarabine (100 mg/m2 for 7 days) and daunorubicin (60 mg/m2 × 3 doses). Toxicity was similar to that of standard induction chemotherapy alone. Although we did not identify a maximum tolerated dose, there was more gastro-intestinal toxicity with 7 days of decitabine priming. Decitabine induced DNA hypomethylation at all dose levels and there was a trend toward greater hypomethylation in CD34+ bone marrow cells when decitabine was delivered by a short pulse (Arm A). Twenty-seven subjects (90%) responded to therapy: 17 with complete remission (57%) and 10 with partial remission (33%). Of the patients with partial remission to protocol treatment, 8 achieved remission to their next therapy, bringing the overall complete remission rate to 83%. We conclude that epigenetic priming of intensive chemotherapy can be safely delivered in an attempt to improve response rates. This trial was registered at www.clinicaltrials.gov as NCT00538876.

Blood ◽  
1983 ◽  
Vol 62 (2) ◽  
pp. 315-319 ◽  
Author(s):  
HJ Weinstein ◽  
RJ Mayer ◽  
DS Rosenthal ◽  
FS Coral ◽  
BM Camitta ◽  
...  

Abstract We designed a protocol (VAPA) that featured 14 mo of intensive postremission induction chemotherapy in an effort to improve remission durations for patients with acute myelogenous leukemia (AML). One hundred and seven patients under 50 yr of age were entered into this study. The rate of complete remission is 70%. A Kaplan-Meier analysis of patients entering remission predicts that 56% +/- 7% (+/-SE) of patients less than 18 yr and 45% +/- 9% of patients aged 18–50 yr will remain in remission at 3 yr (median follow-up is 43 mo). Patients with the monocytic subtype had a statistically significant shorter duration of remission (2-sided p less than 0.05). There was a high incidence of primary CNS relapse in children. Thirty-one of 41 patients who completed the regimen remain in remission without maintenance therapy. We conclude that the VAPA protocol continues to offer a promising approach to treatment of AML.


2020 ◽  
Vol 95 (11) ◽  
pp. 1296-1303
Author(s):  
Gautam Borthakur ◽  
Zhihong Zeng ◽  
Jorge E. Cortes ◽  
Hsiang‐Chun Chen ◽  
Xuelin Huang ◽  
...  

2020 ◽  
Vol 4 (4) ◽  
pp. 599-606 ◽  
Author(s):  
Kirk E. Cahill ◽  
Yasmin H. Karimi ◽  
Theodore G. Karrison ◽  
Nitin Jain ◽  
Margaret Green ◽  
...  

Abstract In this phase 1 study, azacitidine (AZA) was given before high-dose cytarabine (HiDAC) and mitoxantrone (mito) based on the hypothesis that epigenetic priming with a hypomethylating agent before cytotoxic chemotherapy would improve response rates in patients with high-risk acute myeloid leukemia (AML), including relapsed/refractory disease. The primary objective was to establish the recommended phase 2 dose of AZA given before standard HiDAC/mito. In a dose escalation scheme, 46 patients (median age, 66 years) received AZA at 37.5, 50, or 75 mg/m2 subcutaneously or IV once daily on days 1 to 5 followed by HiDAC (3000 mg/m2) and mitoxantrone (30 mg/m2) once each on days 6 and 10 (the HiDAC/mito dose was reduced 33% in elderly subjects). Two dose-limiting toxicities occurred (both in the same patient): acute liver failure and kidney injury at the 50 mg/m2 dose. The 30-day induction death rate was 2.2% (1 of 46). The overall response rate, including complete remission and complete remission with incomplete count recovery, was 61% (28 of 46). Previously untreated patients aged ≥60 years with therapy-related AML and de novo AML were more likely to respond than untreated patients with AML progressing from an antecedent hematologic disorder (myelodysplastic syndrome and chronic myelomonocytic leukemia). Patients with favorable European Leukemia Network risk (P = .008), NPM1 mutations (P = .007), or IDH2 mutations (P = .03) were more likely to respond, and those with TP53 mutations (P = .03) were less likely to respond. The recommended phase 2 dose of AZA is 75 mg/m2 per day on days 1 to 5 followed by HiDAC (3000 mg/m2) and mitoxantrone (30 mg/m2) once each on days 6 and 10. This trial was registered at www.clinicaltrials.gov as #NCT01839240.


Cancer ◽  
2013 ◽  
Vol 119 (10) ◽  
pp. 1823-1831 ◽  
Author(s):  
Matthew G. Fury ◽  
Eric Sherman ◽  
Alan L. Ho ◽  
Han Xiao ◽  
Frank Tsai ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4008-4008
Author(s):  
Wendy Stock ◽  
Karen WL Yee ◽  
Eunice S. Wang ◽  
Sari Enschede ◽  
Joyce Steinberg ◽  
...  

Abstract ABT-869 is an orally bioavailable, potent and specific inhibitor of multiple receptor tyrosine kinases (RTKs) including vascular endothelial and platelet growth factor, FLT3, STAT5, and ERK receptors. Since multiple RTKs, particularly FLT3, are commonly expressed and activated in AML/MDS, ABT-869 may prove to be an attractive therapeutic option. The current multi-center, two arm, dose-escalation study was designed to assess the safety, pharmacodynamics, pharmacokinetics (PK) and preliminary anti-tumor activity of ABT-869 as monotherapy in arm A, and to determine the PK, safety profile and potential of a PK or PD mediated drug interaction in patients with AML or MDS treated with ABT-869 plus Cytosine arabinoside (Ara-C) in arm B. No dose was administered on D7 (arm A) and D12 (arm B). In single-agent Arm A enrollment is complete (N=29) with 9, 4, 12 and 4 patients enrolled in each of the 10, 12.5, 15, and 20 mg cohorts, respectively. Dosing began with the 10 mg/day cohort and escalated to the 20 mg/day cohort to define a recommended phase 2 dose (RP2D) of 15 mg. During dose escalation, in the 10 mg cohort, 2 patients experienced DLTs of fatigue while in the 20 mg cohort, 3 patients experienced DLTs of fatigue and 1 patient experienced a DLT of proteinuria. In currently enrolling arm B (N=17), ABT-869 was administered in combination with Ara-C. ABT-869 was initiated on day 6 following administration of Ara-C at 1.5 mg/m2 on days 1–3 (patients ≥64 years old) or days 1–4 (patients <64 years old); dosing began at 10 mg and escalated to 12.5 mg. In arm B, 2 patients on the 12.5 mg cohort experienced DLTs, one of gr 3 fatigue and one of gr 3 proteinuria, leading to selection of an RP2D of 10 mg. In arm A, the average effective half-life for ABT-869 ranged from 11.7–13.5 h, and the average CL ranged from 4.9–6.6 L/h. The dose-normalized (DN) Cmax were similar between arm A (N=27) and arm B (N=9) at 0.016 ± 0.007 (mean ± SD) and 0.014 ± 0.004 (μg/mL/mg), respectively. The DN Cmax geometric mean ratio of arm B vs. A was 0.938 (90% confidence interval 0.71 – 1.24). DN AUC24 was different between arm A and arm B (0.21 ± 0.10 vs. 0.10 ± 0.04 μg*hr/mL/mg, respectively). The DN AUC24 geometric mean ratio of arm B vs. A was 0.494 (90% confidence interval 0.36–0.68). The DLTs observed in this study were consistent with VEGF receptor tyrosine kinase inhibition including fatigue and proteinuria as seen in both arms A and B. The most common adverse events across all grades (majority being grade 1/2) as reported from the available arm A data were diarrhea, fatigue, dry mouth, nausea, vomiting, asthenia, anorexia, and decreased appetite. In addition, grade 3 events observed were; asthenia (12%), fatigue (6%), febrile neutropenia (6%) and pancytopenia (6%). The latter two events are reflective of the pancytopenia present at the time of study enrollment. ABT-869 has an acceptable safety profile. Preliminary results from this ongoing phase 1 study demonstrate that in arm A the PK of ABT-869 are dose proportional over the 10–20 mg dose range. Differences in ABT- 869 AUC between arm A and arm B are being further investigated and will be correlated with pharmacodynamic endpoints.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2487-2487
Author(s):  
Brian Parkin ◽  
Peter Ouillette ◽  
Yifeng Li ◽  
Cheng Li ◽  
Kerby Shedden ◽  
...  

Abstract Abstract 2487 Introduction: Despite significant advances in the understanding of the biology of adult acute myelogenous leukemia (AML), overall survival remains poor due chiefly to the high rate of relapse after achieving complete remission as well as primary failure of induction chemotherapy. Efforts to further unravel the mechanisms leading to relapse and primary refractory disease are critical in order to guide the development of effective and durable treatment strategies for AML. To that end, this study seeks to elucidate the clonal relationship of AML in various disease phases. Methods: We employed SNP 6.0 array-based genomic profiling of acquired copy number aberrations (aCNA) and copy neutral LOH (cnLOH) together with sequence analysis of recurrently mutated genes to characterize paired AML genomes. We analyzed 28 AML sample pairs from patients that achieved complete remission with chemotherapy and subsequently relapsed (median remission duration 272 days [range 25 – 1249 days]) and 11 sample pairs from patients with persistent disease following induction chemotherapy. AML cell samples were isolated with a Ficoll gradient, negatively selected using Miltenyi microbead columns, and then further purified with flow cytometric cell sorting. Processed DNA isolated from highly purified AML blasts and paired buccal DNA was hybridized to Affymetrix SNP 6.0 arrays. aCNA were visually identified using the dChip program in paired data displays and corroborated by algorithmic lesion scoring, and cnLOH was detected using internally developed software. In addition, 11 genes known to be recurrently mutated in AML (CEBPA, DNMT3A, IDH1, IDH2, RUNX1, BCORL1, NPM1, NRAS, KRAS, FLT3 and TP53) were resequenced in all 39 presentation samples to identify somatically acquired mutations. Genes found mutated in individual AML cases were subsequently tested for the persistence of the mutation in paired samples. Results: For the 28 paired specimens in the relapsed cohort, comparison of aCNA and cnLOH occurrences, gene mutation patterns and karyotypes revealed 6 cases that carried no aCNA/cnLOH at either presentation or relapse, but at presentation carried at least 1 gene mutation, all of which but one were stable in relapse (1 case lost a RUNX1 mutation but carried a t(8;21) in both disease stages); 11 cases that were characterized by the presence of aCNA/cnLOH at presentation, of which 55% (6 of 11) gained additional aCNA/cnLOH at relapse; 6 cases without aCNA/cnLOH at presentation that gained aCNA/cnLOH at relapse, of which 2 concurrently lost a FLT3-ITD or CEPBA mutation; and 5 cases that carried no informative genomic events. For the 11 paired specimens in the persistent AML cohort, the same comparison revealed 2 cases without aCNA/cnLOH before or after chemotherapy and stable gene mutations; 5 cases with aCNA/cnLOH at presentation that carried the same genomic lesions and gene mutations before and after chemotherapy; 3 cases with aCNA/cnLOH present at enrollment that lost some but not all of these aCNA/cnLOH and gained none after initial induction therapy; and 1 additional case that lost a FLT3-ITD. Comparative analysis of these patterns demonstrates that relapsed AML invariably represents reemergence or evolution of an antecedent clone. Furthermore, all individual aCNA or cnLOH detected at presentation persisted at relapse indicating that this lesion type is proximally involved in AML evolution. Analysis of informative paired persistent AML disease samples uncovered at least two coexisting dominant clones of which at least one was chemotherapy sensitive and one resistant. Conclusion: This detailed genomic analysis supports the conclusion that incomplete eradication of AML founder clones rather than stochastic emergence of fully unrelated novel clones underlies AML relapse and persistence with direct implications for clinical AML research. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5335-5335
Author(s):  
Nathaniel M. Cook ◽  
James N. Butera ◽  
John L Reagan

Abstract Introduction: Many patients with Acute Myelogenous Leukemia (AML) will transiently develop blasts in the peripheral blood upon recovery of induction chemotherapy, despite going on to obtain a complete remission. These are presumably non-leukemic marrow blasts. Recovery blasts can sometimes reach over 20% in the peripheral blood in remission patients. The significance of recovery blasts cells after induction chemotherapy in patients with AML is not clear. We sought to define the incidence of recovery blastocytosis (RB) and determine if the presence of RB has prognostic relevance. Methods: We performed a retrospective analysis of 72 patients with newly diagnosed AML who underwent induction chemotherapy with 7+3 (cytarabine and daunorubicin) with or without etoposide between the years of 2006 and 2013 at our institution. Patients were included in the analysis as having RB only if their blasts resolved by day 14 of induction chemotherapy, blasts reappeared upon recovery after day 14, recovery blasts were > 5% in the peripheral blood of a duration of >5 days, and subsequently resolved by day 45 and remission was obtained. We recorded patient's absolute and percentage peripheral blast counts on presentation, upon nadir and throughout recovery of induction chemotherapy. The incidence and level of RB in patients who achieved a remission was tabulated. This group (group 1) was compared to the patients who achieved a remission whom did not develop RB (group 2). Results: 45/77 AML patients obtained a CR after induction chemotherapy. Recovery blasts were seen in 9/45 (20%) remission patients (group 1), with a median RB of 9% for a median duration of 8 days. Of these 9 patients, 4 developed RB > 10%, and 2 developed RB >20 % Group 1 did not differ in respect to age, gender, cytogenetic profile, etoposide use, or whether the patient received 1 or 2 induction therapies from the 36/45 remission AML patients who did not develop RB (group 2). Median survival for group 1 was = 1,376 days, and 1,036 days for group 2 (p= .035). Conclusion: Recovery Blasts in the peripheral blood after induction chemotherapy is seen commonly in AML patients who obtain a complete remission. RB can be seen even as high as > 20% in remission patients. AML patients who develop RB on their way to remission after induction chemotherapy have a better overall survival then patients who do not develop RB. Figure 1 Figure 1. Graph 1. Kaplan-Meier survival curves for AML patients with RB (group 1) and without RB (group 2). Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document