scholarly journals Venetoclax, Actinomycin D and Low Dose Cytarabine for Relapsed or Refractory Acute Myeloid Leukemia in Clinical Practice Setting

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3422-3422
Author(s):  
Andrius Zucenka ◽  
Vilmantė Vaitekėnaitė ◽  
Kazimieras Maneikis ◽  
Regina Pileckytė ◽  
Igoris Trociukas ◽  
...  

Abstract Background Venetoclax based therapies have produced varying results in the relapsed or refractory acute myeloid leukemia (R/R AML) setting. Highest response rates were demonstrated after Venetoclax in combination with high dose chemotherapy. However, this approach is feasible mainly in fit, younger patients. Herein, we present a lower intensity combination therapy consisting of Venetoclax, low dose Cytarabine and Actinomycin D (ACTIVE) in patients with R/R AML administered in real-life clinical practice setting. Methods We performed an observational, retrospective, single centre study. The patients were at least 18 years of age and had R/R AML. All patients provided informed consent for treatment as well as data collection. Venetoclax ramp-up was administered over either 3 or 5 days until the daily dose of 600mg/d was reached. The ACTIVE regimen consisted of Venetoclax 600mg/d p/o on days 1-28, Cytarabine 20mg/m 2 s/c on days 1-10, Actinomycin D 12.5 µg/kg i/v on days 1, 2 and 3 (on days 1 and 2 for patients ≥65 years). Strong/moderate CYP3A inhibitors/inducers or Venetoclax dose reductions were not allowed. Indications for stopping Venetoclax before Day 28 were life threatening infectious complications or faster hematological recovery with the addition of G-CSF in responders. A second ACTIVE cycle was administered in non-responders without evidence of progressive disease after Cycle 1 or in responders with positive minimal residual disease (MRD). We evaluated baseline characteristics, composite CR (CRc = CR + CRi + CRp), overall response (ORR = CRc + MLFS), MRD negativity rates, overall survival (OS), relapse-free survival (RFS), event-free survival (EFS), Grade 3-5 non-hematological toxicities and Day 30 and Day 60 mortality rates. Results Fifty R/R AML patients were treated with ACTIVE and 56% (28/50) were male. The median age was 65 years (20-84). The median Eastern Cooperative Oncology Group (ECOG) status was 1 (0-3) and 40% (20/50) had ECOG status of 2 or 3. Secondary AML was confirmed in 48% (24/50) of cases. Adverse cytogenetics were identified in 28% (14/50) of patients whereas 60% (30/50) were stratified to adverse risk group in accordance with ELN 2017 guidelines. The most common gene mutations were IDH1/2 30% (15/50), FLT3 28% (14/50), ASXL1 22% (11/50), NPM1 14% (7/50), N/KRAS 12% (6/50) and RUNX1 12% (6/50). The median number of prior therapies was 2 (1-5). Intensive chemotherapy was administered in 80% (40/50) of whom 38% (15/40) had primary refractory disease and 48% (19/40) had previously received Fludarabine + Cytarabine + Idarubicin (FLAG-Ida) or Cytarabine + Mitoxantrone (HAM). Eight percent (4/50) had had prior Venetoclax exposure. Thirty-six percent (18/50) had relapsed after allogeneic stem cell transplantation (alloSCT) with a median time of 7.4 months (1.6-37.3) from transplant to relapse. One cycle of ACTIVE therapy was administered in 76% (38/50) of cases, whereas 24% (12/50) received 2 cycles. The median number of Venetoclax 600mg/d days per cycle was 18 (8-28). Additional FLT3/RAS/BCR-ABL1 inhibitors Gilteritinib, Trametinib or Dasatinib were administered in 12% (6/50). Forty-nine patients were evaluable for response and one patient died of sepsis before response evaluation (Table 1). The ORR was 73% (36/49) and the CRc rate was 67% (31/46). MRD negativity was confirmed in 61% (19/31) of CRc patients. Sixteen patients had undergone additional early bone marrow evaluations. Blast count reduction to <5% was observed in 50% (8/16) after Venetoclax ramp-up and in 88% (14/16) on Day 4. Half of the ACTIVE responders (18/36) continued maintenance therapy with Venetoclax + low dose Cytarabine and optional DLI, whereas 36% (13/36) proceeded to either first or second alloSCT. After 16.4 months of median follow-up, the median OS, RFS and EFS were 13.1, 7.2 and 4.5 months, respectively (Figure 1A). Median OS was not reached in MRD negative patients (Figure 1B). In multivariable Cox regression analysis, adverse cytogenetics (HR 3.48, 95% CI 1.39 -8.55) and primary refractory disease (HR 2.54, 95% CI 1.05-6.12) were associated with worse OS. The most common grade 3-5 non-hematological adverse events were febrile neutropenia (54%, 27/50) and bacteremia/sepsis (34%, 17/50). Day 30 and Day 60 mortality rates were 8% (4/50) and 16% (8/50), respectively. Conclusions ACTIVE was effective and well tolerated in this unselected prognostically unfavourable older R/R AML patient population. Figure 1 Figure 1. Disclosures Zucenka: Jannsen: Honoraria, Other: Travel-expenses; Takeda: Other: Travel Expenses; Novartis: Honoraria, Other: Travel Expenses; Pfizer: Honoraria, Other: Travel Expenses; Astellas: Honoraria; Abbvie: Honoraria, Other: Travel Expenses. Maneikis: Abbvie: Honoraria. Pileckytė: Abbvie: Honoraria, Other: Travel Expenses. Griškevičius: Abbvie: Other: Travel Expenses. OffLabel Disclosure: Venetoclax has been used off-label for the treatment of R/R AML

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 388-388 ◽  
Author(s):  
Gautam Borthakur ◽  
Hagop M. Kantarjian ◽  
Susan O'Brien ◽  
Guillermo Garcia-Manero ◽  
Elias Jabbour ◽  
...  

Abstract Background: FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) mutation in acute myeloid leukemia (AML) is associated with early relapse and poor survival. Quizartinib potently and selectively inhibits FLT3 kinase activity. In a phase I and II studies the composite response rate (CRR) was approximately 50% among patients with FLT3-ITD. There is in-vitro synergy between quizartinib and 5-AZA or LDAC. We hypothesize that adding quizartinib to a hypomethylating agent such as 5-azacitidine (AZA) or cytarabine may improve the response rate expected from the use of either agent alone. Objectives: The primary objective of phase I part is to determine the dose limiting toxicity (DLT) and maximally tolerated dose (MTD) of the combination of quizartinib (AC220) with either AZA or low-dose cytarabine (LDAC); for phase II is to determine the clinical activity of both combinations. This planned interim analysis reports on the recommended phase II dose (RP2D) and first futility analysis. Methods: For phase I, patients with relapsed/refractory high-risk myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia (CMML) or AML were eligible irrespective of FLT3 mutation and salvage status. For phase II, presence of FLT3-ITD is a requisite. Phase II enrollment is limited to patients >60 years with untreated MDS/CMML/AML, or any age receiving first salvage treatment. Additional eligibilities include performance status ECOG ≤2, adequate organ function, normal electrolytes (potassium, calcium and magnesium). Important exclusions include QTcF> 450 mSec, concomitant drugs that prolong QT/QTc interval or strong CYP3A4 inhibitors or inducers with the exception of antibiotics, antifungals, and antivirals that are used as standard of care. Treatment cycle is defined as 28 days. Treatment comprises of AZA 75 mg/m2 subcutaneously (SQ) or intravenously (IV) for 7 days of every cycle (Days 1-7), or cytarabine 20 mg SQ twice daily for 10 days of every cycle (Days 1-10) along with quizartinib at two planned dose levels: 60 mg (dose level 1) or 90 mg orally daily (dose level 2) uninterrupted. Patients are assigned to AZA or LDAC arm by physician choice or slot availability. Planned accrual for each arm in phase 2 is 26 pts each and an ORR of ≥50% will be considered favorable. Accrual of 26 pts will give a 95% credible interval for overall response rate of (0.32, 0.68). The study will be stopped for toxicity (>30%) and/or futility (ORR <50%) at interim analysis for each arm. Results: Twenty-six (Phase I=12, phase II=14) pts have been enrolled: 18 to AZA arm and 8 to LDAC arm. Median age is 62 years (range, 25-79 years), 7 (27%) are female. Cytogenetics are diploid=14, +8=2, -7=2, miscellaneous=6, 11q and t(8;21)= 1 each. Median number of prior therapies is 2 (range, 0-7), 7 patients received prior FLT3 inhibitor. For both schedules quizartinib 60 mg daily was identified as the recommended phase II dose (RP2D) based on emerging results from separate dose-finding study. Eighteen [5 in LDAC arm (63%) and 13 (72%) in AZA arm; all with FLT3-ITD mutation without D835 mutation] of 26 total pts (69%) have responded (CR=1/ CRp=3/ CRi=2/ MLFS=10/PR=1/HI=1). Among patients with FLT3-ITD (N=22), ORR is 82%. Four of 7 (57%) patients with prior FLT3 inhibitor exposure responded. Median number of days to respond is 57 days (range, 25-102 days). Among responders two patients died (MLFS=1, PR=1): one with gastro-intestinal bleeding and other with progressive pneumonia. Three additional responders have discontinued therapy for stem cell transplant (1), withdrawal of consent (1), and loss of response with emergence of D835 mutation (1). Nine responders (CR=1, CRi=1, CRp=1, PR=1, MLS=5) had >50% reduction of FLT3-ITD allelic burden and 2 additional pts (CR=1, CRi=1) had no detectable FLT3-ITD at response. Number of pts with treatment emergent grade 3/4 toxicities irrespective of attribution include hypokalemia (15), hypophosphatemia (5), hyponatremia (4), hypocalcemia (4), hyperbilirubinemia (3), increase in ALT (1), hypernatremia (1hyperglycemia (1), hypotension (1), QTcF prolongation (1, grade 3). Conclusion: Combination of quizartinib and AZA or LDAC is highly active among patients with AML/MDS/CMML with FLT3-ITD . Response rates appear higher than expected with either agent alone. Clinically significant QTcF prolongation is infrequent. Accrual to both arms of the current trial continues. Disclosures Cortes: Ambit Biosciences: Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5108-5108
Author(s):  
Li Yang ◽  
Ligen Liu ◽  
Limin Zhao ◽  
Yingting Lu ◽  
Jing Zhang ◽  
...  

Background. Patients with relapsed or refractory acute myeloid leukemia (R/R AML)who are intolerant of strong regimen chemotherapy have poor responsibility to chemotherapy and clinic remission is lower. The DNMT is depleted to reactivate the silencing tumor suppressor gene to exert anti-tumor effects. Vitro experiments confirmed that ATRA has an anti-leukemia effect on non-M3-AML cell lines. We perform the 2 phase, single-arm, multicenter study of Decitabine in Combination with ATRA, G-CSF and low dose cytarabine(DLAAG Regimen)in R/R AML not suitable for strong chemotherapy (NCT03356080). Methods. R/R AML patients aged more than 18 years not suitable for strong chemotherapy were eligible to enroll on this study. The objective was to assess the responses and safety of this therapy. All patients received subcutaneous decitabine injection 0.1-0.2mg/kg on days 1-3 ,8-10,15-17 .ATRA was taken orally at 45mg/m2 on day 4-6 and 15mg/m2on days 7-20. Subcutaneous injection cytarabine 15mg/m2 every 12 hours on days 1-10 and G-CSF 300ug/d on day 0 until disease remission or absolute neutrophils count ≥ 0.5×109/L were administered. The FLT3 inhibitor, arsenious acid, and the JAK-2 inhibitor, rotectinib, were allowed to be combined.All patients received one or two courses of induction treatments. Results. During the period from December 1, 2017 to July 2019, a total of 33 patients were recruited, including 17 women and 16 men. The median age is 64 years (range 46-82). In 33 patients, the overall response rate (ORR = CR + PR) was 36.3%, and the complete response rate (CRR = CR + CRi) was 33.3%. The early treatment-related mortality rate was 6.0%. One died of infection and one died of organ bleeding. Of the 26 patients with karyotype data, 11 received CR. Seven patients (63.6%) in CR showed normal karyotype, 2 patients (18.1%) in CR, and 1 patient (9.0%) in PR showed other moderate karyotypes. One case (9.0%) of the CR group had a poor karyotype. Statistical analysis showed that there was a statistically significant difference between the different karyotype groups (P < 0.05). Only 2 of the 10 FLT3 mutant patients achieved CR (20%). Two patients with abnormal MLL gene did not receive CR. Conclusions DLAAG regimen is effective and tolerated in patients with R/R AML who are not suitable for intensive therapy and could be as a bridging therapy regimen followed by HSCT. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 100 (5) ◽  
pp. 1195-1202
Author(s):  
Andrius Zucenka ◽  
Kazimieras Maneikis ◽  
Birute Pugaciute ◽  
Ugne Ringeleviciute ◽  
Austeja Dapkeviciute ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1336-1336 ◽  
Author(s):  
Agnieszka Wierzbowska ◽  
Ewa Wawrzyniak ◽  
Agnieszka Pluta ◽  
Tadeusz Robak ◽  
Grzegorz J. Mazur ◽  
...  

Abstract Background: Cytogenetics is one of the most important prognostic factors in acute myeloid leukemia (AML). Monosomal karyotype (MK), defined as two or more distinct autosomal monosomies or one single autosomal monosomy in the presence of at least one structural abnormality, has been identified as a new cytogenetic category associated with particularly poor prognosis (Breems, JCO, 2008). AML patients (pts) with MK+ have shorter overall survival (OS) compared to other unfavorable karyotypes, irrespective of treatment intensity. However, preliminary results from a phase II trial of decitabine (DEC) as first-line treatment for elderly AML pts demonstrated that an objective response rate (CR+PR) in MK+ pts was higher than in the MK- cases with abnormal cytogenetics. Moreover, the OS of MK+ pts was comparable to that of MK- pts (Lübbert, Haematologica, 2012). This observation might suggest a positive effect of DEC in patients with MK+ AML. Objective: To determine the effects of treatment with DEC vs low-dose cytarabine or best supportive care (BSC) as a treatment of choice (TC) on OS, progression free survival (PFS) and response rate (CR/CRp) in the subset of pts with AML MK+, who were treated in the randomized phase III DACO-16 study. A second objective is to compare the OS, PFS and CR/CRp of MK+ vs other unfavorable karyotype cases. Methods: Eligible pts were ≥65 years old with newly diagnosed, de novo or secondary AML with ≥20% bone marrow blasts, ECOG performance status 0-2, WBC count ≤40x109/L. According to study protocol pts were randomly assigned 1:1 to receive DEC 20 mg/m2 per day as a 1-hour i.v. infusion for five days every 4 weeks or TC (BSC or cytarabine 20 mg/m2 s.c. for 10 days every 4 weeks). This analysis included pts identified from the clinical database whose locally obtained cytogenetics data indicated they had MK+. Pts with poor-risk cytogenetics (Southwest Oncology Group classification) but MK- (poor-risk MK-) were used as the comparison group. Median OS (Kaplan-Meier method) and PFS were compared between decitabine vs TC treatment arms for pts MK+ and poor-risk MK-. Remission was assessed using modified IWG AML criteria (Cheson, Blood, 2003). Results: Of 480 pts in DACO-16 study, 64 (13.3%) fulfilled the criteria for MK+ and additional 99 (20.6%) had poor-risk MK- cytogenetics. In MK+ group 33 pts received DEC and 31 pts were assigned to TC. In poor-risk MK- group DEC and TC was administered to 49 and 50 pts respectively. Baseline characteristics of treatment subgroups were comparable. Outcome of MK+ and poor-risk MK- pts according to treatment arm The CR/CRp rate in MK+ pts was higher in the DEC arm compared to TC (21% vs 3%; OR=8.1; 95% CI, 0.93 to 70.04; P = 0.054). The median PFS was also improved in the DEC arm vs TC (2.6 vs 1.3 mos, HR=0.53; 95% CI, 0.31 to 0.9; P = 0.018) Fig.1. There was a trend towards longer OS by ~4 mos in the DEC arm vs TC (2.6 vs 6.3 mos; HR=0.67; 95% CI, 0.39 to 1.15; p=0.14) Fig.2. In contrast, no difference in CR/CRp rate between DEC vs TC was observed in poor-risk MK- pts (respectively 8% vs 10%; OR=0.8; 95% CI, 0.2 to 3.17; P = 1.0). Similarly, no improvement in PFS as well as OS was observed in poor-risk MK- cases treated with DEC compared to TC. Overall survival in decitabine and TC treatment arms according to cytogenetics In AML patients receiving low-dose cytarabine or BSC, patients with MK+ appeared to have a poorer outcome than pts with poor-risk MK-. The median OS was 2.6 vs 4.7 mos (HR=1.52; 95% CI, 0.91 to 2.54; p=0.11) in MK+ and poor-risk MK- pts, respectively. In contrast, in the DEC arm MK+ pts did not do worse than MK- cases with poor-risk cytogenetics. The median OS was similar: 6.3 vs 5.0 mos for MK+ and poor-risk MK- (HR=0.98; 95% CI, 0.6 to 1.58; p=0.92), respectively. Conclusions: In older patients with AML-MK+, decitabine improved CR/CRp rate and PFS compared with standard therapies. A survival analysis suggesting a benefit of about 4 months in median OS for decitabine in MK+ AML needs confirmation in further studies with larger sample size. These data might suggest decitabine overcomes the extremely poor prognosis associated with MK and may be a beneficial initial treatment as an alternative to low-dose cytarabine or best supportive care. Figure 1. PFS of patients with AML MK+ according to treatment Figure 1. PFS of patients with AML MK+ according to treatment Figure 2. OS of patients with AML MK+ according to treatment Figure 2. OS of patients with AML MK+ according to treatment Disclosures Wierzbowska: Janssen, Celgene: Consultancy. Off Label Use: Decitabine in elderly AML patients. Robak:Eisai Inc: Research Funding. Oriol:Celgene, Janssen, Amgen: Consultancy, Speakers Bureau.


Sign in / Sign up

Export Citation Format

Share Document