Faculty Opinions recommendation of Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531.

Author(s):  
Stefan Bielack
2014 ◽  
Vol 32 (27) ◽  
pp. 3021-3032 ◽  
Author(s):  
Alan S. Gamis ◽  
Todd A. Alonzo ◽  
Soheil Meshinchi ◽  
Lillian Sung ◽  
Robert B. Gerbing ◽  
...  

Purpose To improve survival rates in children with acute myeloid leukemia (AML), we evaluated gemtuzumab-ozogamicin (GO), a humanized immunoconjugate targeted against CD33, as an alternative to further chemotherapy dose escalation. Our primary objective was to determine whether adding GO to standard chemotherapy improved event-free survival (EFS) and overall survival (OS) in children with newly diagnosed AML. Our secondary objectives examined outcomes by risk group and method of intensification. Patients and Methods Children, adolescents, and young adults ages 0 to 29 years with newly diagnosed AML were enrolled onto Children's Oncology Group trial AAML0531 and then were randomly assigned to either standard five-course chemotherapy alone or to the same chemotherapy with two doses of GO (3 mg/m2/dose) administered once in induction course 1 and once in intensification course 2 (two of three). Results There were 1,022 evaluable patients enrolled. GO significantly improved EFS (3 years: 53.1% v 46.9%; hazard ratio [HzR], 0.83; 95% CI, 0.70 to 0.99; P = .04) but not OS (3 years: 69.4% v 65.4%; HzR, 0.91; 95% CI, 0.74 to 1.13; P = .39). Although remission was not improved (88% v 85%; P = .15), posthoc analyses found relapse risk (RR) was significantly reduced among GO recipients overall (3 years: 32.8% v 41.3%; HzR, 0.73; 95% CI, 0.58 to 0.91; P = .006). Despite an increased postremission toxic mortality (3 years: 6.6% v 4.1%; HzR, 1.69; 95% CI, 0.93 to 3.08; P = .09), disease-free survival was better among GO recipients (3 years: 60.6% v 54.7%; HzR, 0.82; 95% CI, 0.67 to 1.02; P = .07). Conclusion GO added to chemotherapy improved EFS through a reduction in RR for children and adolescents with AML.


Blood ◽  
2008 ◽  
Vol 111 (8) ◽  
pp. 4329-4337 ◽  
Author(s):  
Sanne Lugthart ◽  
Ellen van Drunen ◽  
Yvette van Norden ◽  
Antoinette van Hoven ◽  
Claudia A. J. Erpelinck ◽  
...  

AbstractInappropriate expression of EVI1 (ecotropic virus integration-1), in particular splice form EVI1-1D, through chromosome 3q26 lesions or other mechanisms has been implicated in the development of high-risk acute myeloid leukemia (AML). To validate the clinical relevance of EVI1-1D, as well as of the other EVI1 splice forms and the related MDS1/EVI1 (ME) gene, real-time quantitative polymerase chain reaction was performed in 534 untreated adults with de novo AML. EVI1-1D was highly expressed in 6% of cases (n = 32), whereas 7.8% were EVI1+ (n = 41) when all splice variants were taken into account. High EVI1 predicted a distinctly worse event-free survival (HR = 1.9; P = .002) and disease-free survival (HR = 2.1, P = .006) following multivariate analysis. Importantly, we distinguished a subset of EVI1+ cases that lacked expression of ME (EVI1+ME−; n = 17) from cases that were ME+ (EVI1+ME+; n = 24). The atypical EVI1+ME− expression pattern exhibited cytogenetically detectable chromosomal 3q26 breakpoints in 8 cases. Fluorescence in situ hybridization revealed 7 more EVI1+ME− cases that carried cryptic 3q26 breakpoints, which were not found in the EVI1+ME+ group. EVI1+ME− expression predicts an extremely poor prognosis distinguishable from the general EVI1+ AML patients (overall survival [OS]: P < .001 and event-free survival [EFS]: P = .002). We argue that EVI1/ME quantitative expression analysis should be implemented in the molecular diagnostic procedures of AML.


2017 ◽  
Vol 35 (23) ◽  
pp. 2674-2682 ◽  
Author(s):  
Jatinder K. Lamba ◽  
Lata Chauhan ◽  
Miyoung Shin ◽  
Michael R. Loken ◽  
Jessica A. Pollard ◽  
...  

Purpose Gemtuzumab ozogamicin (GO), a CD33-targeted immunoconjugate, is a re-emerging therapy for acute myeloid leukemia (AML). CD33 single nucleotide polymorphism rs12459419 C>T in the splice enhancer region regulates the expression of an alternatively spliced CD33 isoform lacking exon2 (D2-CD33), thus eliminating the CD33 IgV domain, which is the antibody-binding site for GO, as well as diagnostic immunophenotypic panels. We aimed to determine the impact of the genotype of this splicing polymorphism in patients with AML treated with GO-containing chemotherapy. Patients and Methods CD33 splicing single nucleotide polymorphism was evaluated in newly diagnosed patients with AML randomly assigned to receive standard five-course chemotherapy alone (No-GO arm, n = 408) or chemotherapy with the addition of two doses of GO once during induction and once during intensification (GO arm, n = 408) as per the Children’s Oncology Group AAML0531 trial. Results The rs12459419 genotype was CC in 415 patients (51%), CT in 316 patients (39%), and TT in 85 patients (10%), with a minor allele frequency of 30%. The T allele was significantly associated with higher levels of D2-CD33 transcript ( P < 1.0E−6) and with lower diagnostic leukemic cell surface CD33 intensity ( P < 1.0E−6). Patients with the CC genotype had significantly lower relapse risk in the GO arm than in the No-GO arm (26% v 49%; P < .001). However, in patients with the CT or TT genotype, exposure to GO did not influence relapse risk (39% v 40%; P = .85). Disease-free survival was higher in patients with the CC genotype in the GO arm than in the No-GO arm (65% v 46%, respectively; P = .004), but this benefit of GO addition was not seen in patients with the CT or TT genotype. Conclusion Our results suggest that patients with the CC genotype for rs12459419 have a substantial response to GO, making this a potential biomarker for the selection of patients with a likelihood of significant response to GO.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5096-5096 ◽  
Author(s):  
Haiping Dai ◽  
Chao Ding ◽  
Suning Chen ◽  
Roderick A. F. MacLeod ◽  
Stefan Ehrentraut ◽  
...  

Abstract Emerging evidence has revealed that miRNAs can function as oncogenes or tumor suppressor genes in leukemia. By profiling miRNA expression across a cohort (n=156) with de novo acute myeloid leukemia (AML), we identified miR-130a as significantly overexpressed in AML with t(8;21). Expression of miR-130a decreased significantly once patients with t(8;21) achieved complete remission, but increased sharply at the time of relapse. In patients with t(8;21) AML, KIT mutational status was associated with miR-130a expression - with higher expression associated with KIT activating mutations. Increased miR-130a expression in t(8;21) AML was associated with worse event-free survival, however no impact on overall survival was observed. Knockdown of RUNX1/RUNX1T1 (AML1/ETO) protein in the SKNO-1 cell line resulted in decrease of expression of miR-130a. Direct binding of RUNX1/RUNX1T1 fusion protein with the promoter sequence of miR-130a was detected with luciferase reporter gene assay. Following miR-130a knockdown, SKNO-1 demonstrated increased sensitivity to etoposide. Taken together, our data suggest that miR-130a is directly activated by RUNX1/RUNX1T1, and can be used to predict leukemia burden, event-free survival and chemotherapy sensitivity in AML with t(8;21). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 899-899 ◽  
Author(s):  
Richard Aplenc ◽  
Soheil Meshinchi ◽  
Lillian Sung ◽  
Todd A. Alonzo ◽  
Jessica Pollard ◽  
...  

Abstract Introduction: Despite the very high intensity of current chemotherapy regimens for children with acute myeloid leukemia (AML), approximately 50% of patients will experience disease relapse. New therapeutic strategies to improve clinical outcomes have centered on improving the efficacy of standard chemotherapy with novel agents such as gemtuzumab and other agents designed to augment standard chemotherapy. Bortezomib, a proteasome inhibitor, is one such agent. The Children's Oncology Group (COG) Phase III clinical trial AAML1031 tested the hypothesis that the addition of bortezomib to standard chemotherapy would improve treatment outcomes in pediatric patients with newly diagnosed AML. Methods: The COG AAML1031 trial randomized patients younger than 30 years of age with de novo AML to either standard chemotherapy (Arm A) or standard chemotherapy with bortezomib (Arm B). Patients with high allelic ratio FLT3 ITD were offered enrollment on a standard chemotherapy plus sorafenib (Arm C, n = 102) and are excluded from this efficacy analysis. All patients received induction chemotherapy with cytarabine, daunorubicin, and etoposide (ADE). Risk stratification occurred at the end of ADE induction and was based on the presence of high risk cytogenetic/molecular markers and/or minimal residual disease (MRD) >0.1% determined by multidimensional flow cytometry. Low risk patients received three additional courses of chemotherapy consisting of a second course of ADE, a third course of cytarabine/etoposide and a fourth course of cytarabine/mitoxantrone. High risk patients received a second course of cytarabine/mitoxantrone, a third course of cytarabine/etoposide, and then allogeneic stem cell transplant (SCT) from the best available donor. Bortezomib 1.3 mg/m2 was given on days 1, 4 and 8 of each cycle with one dose de-escalation to 1 mg/m2 allowed for dose limiting toxicity. Results: A total of 1097 patients were randomized to either standard therapy (Arm A, n = 542) or standard chemotherapy with bortezomib (Arm B, n = 555). No statistically significant differences in sex, age, race, ethnicity, WHO classification, initial blast count, or initial CNS status was observed between arms. Remission induction rate did not differ between treatment arms 89% vs 91%, p = 0.457. MRD was negative in 75% of patients on both treatment arms at the end of Induction I and mean MRD measures did not differ significantly: 2.8% vs 1.9%, p = 0.247. For all patients, event free survival (EFS) and overall survival (OS) at 3 years were 44.4% ± 3.8% and 60.6% ± 4.4%. EFS was not significantly different between Arms A and B (44.0% ± 5.2% vs 44.6% ± 5.6%, p = 0.285) (Figure 1). Likewise, OS was similar between arms (59.0 ± 6.7 vs 62.2 ± 6.0, p = 0.732) (Figure 1). One year cumulative treatment related mortality (TRM) was 14.6 ± 9.3 and 10.8 ± 7.5, p = 0.49 for Arms A and B, respectively. No significant differences were seen in OS, disease-free survival, and TRM from the end of Induction I in low and high risk groups. Cox proportional hazards analysis demonstrated that initial WBC count at diagnosis was the only consistently identified risk factor for OS, DFS, and TRM. Targeted toxicity monitoring identified increased toxicity risks in Arm B for peripheral neuropathy (Induction I/II), dose reductions (all chemotherapy courses), and PICU admissions (Induction I/II) and Intensification I). Serial monitoring of cardiac ejection fraction/shortening fraction in all patients did not demonstrate a clinically meaningful difference in drop in ejection fraction/shortening fraction by treatment arm. No other consistent differences in targeted toxicity rates were identified. Conclusions: The addition of bortezomib to standard chemotherapy increased toxicity but did not improve EFS or OS in children with newly diagnosedAML Consequently, bortezomib should not be used in children with de novo AML in combination with standard chemotherapy. Future work will evaluate the role of intensifying Induction II therapy for patients with high risk AML, further refine risk stratification, and define a more optimal role for allogeneic donor SCT in pediatric AML. Figure 1 Figure 1. Disclosures Loken: Hematologics: Employment, Equity Ownership.


Haematologica ◽  
2018 ◽  
Vol 104 (1) ◽  
pp. 113-119 ◽  
Author(s):  
Juliette Lambert ◽  
Cécile Pautas ◽  
Christine Terré ◽  
Emmanuel Raffoux ◽  
Pascal Turlure ◽  
...  

2008 ◽  
Vol 26 (14) ◽  
pp. 2390-2395 ◽  
Author(s):  
Richard Aplenc ◽  
Todd A. Alonzo ◽  
Robert B. Gerbing ◽  
Beverly J. Lange ◽  
Craig A. Hurwitz ◽  
...  

Purpose While gemtuzumab ozogamicin (GTMZ) is commonly used in the treatment of acute myeloid leukemia (AML) in combination with standard chemotherapy agents, the pediatric maximum-tolerated dose (MTD) of GMTZ in combination with chemotherapy has not been determined. Patients and Methods The Children's Oncology Group AAML00P2 trial sought to define the MTD of GMTZ in combination with cytarabine and mitoxantrone and cytarabine and l-asparaginase chemotherapy regimens. Results The MTD for GMTZ in combination with cytarabine and mitoxantrone was 3 mg/m2 while the MTD in combination with cytarabine and l-asparaginase was 2 mg/m2. Toxicities observed in both treatment regimens were typical of those seen in the relapsed AML setting and consisted primarily of infectious complications. The overall remission response rate (mean ± SE) was 45% ± 15% and the 1 year event-free survival and overall survival estimates were 38% ± 14% and 53% ± 15%, respectively. Conclusion This trial determined the pediatric MTD for GMTZ with two commonly used AML chemotherapy combinations. Based on these results, an ongoing phase III trial conducted within the Children's Oncology Group is evaluating the effect of GMTZ when added to standard AML therapy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1414-1414
Author(s):  
Donna L Johnston ◽  
Todd A. Alonzo ◽  
Robert B. Gerbing ◽  
Betsy A. Hirsch ◽  
Nyla A. Heerema ◽  
...  

Abstract Abstract 1414 Introduction: Studies of acute myeloid leukemia (AML) in adults have documented that abnormalities of chromosome 5q (-5/5q-), primarily deletions, confer a poor prognosis. However, there are no large studies that specifically focus on -5/5q- in pediatric AML. Methods: To elucidate the disease correlates of this group, we retrospectively analyzed cytogenetic data from 5 studies of childhood AML: Children's Oncology Group (COG-AAML03P1), Children's Cancer Group (CCG-2961 and 2891) and Pediatric Oncology Group (POG-8821 and 9421). Data from all patients whose cytogenetic clones included -5/5q-, with the exception of those with acute promyelocytic leukemia (M3) or Down syndrome, were included. A total of 2035 patients from these 5 studies had cytogenetic data available for review. Results: Twenty-two (1.1%) of the 2035 patients had −5 or 5q-. The majority of these patients were male (63.6%). The median age was 12.9 years (range 0.3–20.7 years) with a significant number of patients in the 11–21 year age range (63.6%, p=0.032). The median white blood cell count was 17.4 ×103/μL (range 1.4–98 ×103/μL) and the median bone marrow blast percentage was 77% (range 15–99%). Patients with -5/5q- had a significantly higher median platelet count at diagnosis than those without this abnormality (88×103/μL versus 53×103/μL, p=0.015). Of the 22 patients with -5/5q-, their FAB classification showed that a significant number of patients had M0 morphology (28.6%, p<0.001) versus patients without -5/5q- (2.6%). The remaining patients had M1 morphology (23.8%), M2 (19%), M4 (9.5%), M5 (9.5%), M6 (4.8%) and M7 (4.8%) morphology. Eighteen of the 22 patients (81.8%) had a complete response (CR) to induction chemotherapy. The 5-year event free survival (EFS) from the time of diagnosis for these 22 patients was 27% (±19%) and the 5-year overall survival (OS) was 32% (±20%). The 5-year EFS and OS for the patients on these studies without -5/5q- were 41% (±2%) and 51% (±2%) respectively. The 5-year EFS and OS rates were not significantly different between the two groups (p=0.182 and 0.120 respectively). For the 18 patients who achieved a CR with induction chemotherapy, from time of CR the 5-year disease free survival (DFS) was 33% (±22%), the 5-year OS was 39% (±25%), the 5-year relapse risk (RR) was 61% (±23%) and the 5-year treatment related mortality (TRM) was 6% (±11%). For the 1674 patients without -5/5q- who obtained a CR after induction, from the time of CR the 5-year DFS was 47% (±2%), OS was 57% (±2%), RR was 44% (±3%) and TRM was 9% (±5%). None of these values were significantly different between the groups (p>0.1). Conclusions: In this, the largest retrospective study of pediatric patients with AML and -5/5q- to date, this cytogenetic subgroup was found to have a poor outcome. The median 5-year overall survival across studies was 32%, and the median 5-year event free survival was 27%. These findings support the use of more aggressive therapy for the treatment of children with -5/5q- AML, as has been previously supported based on data from adults with -5/5q- AML. This subset of patients may also benefit from treatment with innovative agents. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
pp. JCO.20.03048
Author(s):  
Jessica A. Pollard ◽  
Erin Guest ◽  
Todd A. Alonzo ◽  
Robert B. Gerbing ◽  
Mike R. Loken ◽  
...  

PURPOSE We investigated the impact of the CD33-targeted agent gemtuzumab ozogamicin (GO) on survival in pediatric patients with KMT2A-rearranged ( KMT2A-r) acute myeloid leukemia (AML) enrolled in the Children's Oncology Group trial AAML0531 ( NCT01407757 ). METHODS Patients with KMT2A-r AML were identified and clinical characteristics described. Five-year overall survival (OS), event-free survival (EFS), disease-free survival (DFS), and relapse risk (RR) were determined overall and for higher-risk versus not high-risk translocation partners. GO's impact on response was determined and outcomes based on consolidation approach (hematopoietic stem cell transplant [HSCT] v chemotherapy) described. RESULTS Two hundred fifteen (21%) of 1,022 patients enrolled had KMT2A-r AML. Five-year EFS and OS from study entry were 38% and 58%, respectively. EFS was superior with GO treatment (EFS 48% with GO v 29% without, P = .003), although OS was comparable (63% v 53%, P = .054). For patients with KMT2A-r AML who achieved complete remission, GO was associated with lower RR (40% GO v 66% patients who did not receive GO [No-GO], P = .001) and improved 5-year DFS (GO 57% v No-GO 33%, P = .002). GO benefit was observed in both higher-risk and not high-risk KMT2A-r subsets. For patients who underwent HSCT, prior GO exposure was associated with decreased relapse (5-year RR: 28% GO and HSCT v 73% No-GO and HSCT, P = .006). In multivariable analysis, GO was independently associated with improved EFS, improved DFS, and reduced RR. CONCLUSION GO added to conventional chemotherapy improved outcomes for KMT2A-r AML; consolidation with HSCT may further enhance outcomes. Future clinical trials should study CD33-targeted agents in combination with HSCT for pediatric KMT2A-r AML.


Sign in / Sign up

Export Citation Format

Share Document