scholarly journals Evidence for Clinical Differentiation and Differentiation Syndrome in Patients With Acute Myeloid Leukemia and IDH1 Mutations Treated With the Targeted Mutant IDH1 Inhibitor, AG-120

2016 ◽  
Vol 16 (8) ◽  
pp. 460-465 ◽  
Author(s):  
K.C. Birendra ◽  
Courtney D. DiNardo
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2796-2796
Author(s):  
Jorge Monge ◽  
Christine Vaupel ◽  
Ronan T Swords ◽  
Terrence Bradley ◽  
Ryan Chapman ◽  
...  

Abstract Introduction: There is a paucity of data on the distribution of acute leukemia (AL) across ethnic groups. Historically, B-cell acute lymphoblastic leukemia (B-ALL) and acutepromyelocytic leukemia (APL) are over-represented in Hispanic patients, while acute myeloid leukemia (AML) is less frequent. We analyzed the ethnic distribution of AL in Florida based on incidence rates and nativity differences using Florida Cancer Data System (FCDS). In our study, B-ALL and APL were more common in Hispanics vs. non-Hispanics (IRR of 1.627 and 1.302, respectively); however, contrary to prior reports, AML was also more common (IRR 1.533) (Swords et al, BCJ 2016, in press). No nativity differences were observed. Given this novel finding of increased AML incidence in Hispanics in South Florida, we examined AML patients at Sylvester Comprehensive Cancer Center (SCCC) to look for ethnic differences in mutational frequency. Methods: We obtained molecular genetic data on AML patients treated from 2012-2015 across multiple centers.Genomic DNA from bone marrow or peripheral blood was sequenced for ASXL1, DNMT3A, FLT3 TKD, IDH1, IDH2, KIT, NPM1, PHF6, TET2 on the IlluminaMiSeq platform using a lower limit of detection of 5% and minimum coverage of 500X. Alignment and variant calling were performed usingNextGENe® software as previously described byGenoptix, Inc. MLL PTD, FLT3 ITD and CEBPA mutations were detected by PCR amplification followed by fragment analysis.For patients treated at SCCC, we obtained IRB approval to conduct retrospective chart reviews for clinical and pathologic data. Categorical data was analyzed by Fisher exact test or Chi-square test as appropriate. Continuous data was analyzed by independent t-test and Wilcoxon-Mann Whitney test for parametric and non-parametric data, respectively. Statistical analysis was performed using Stata (version 13.0).Circos plots were created usingCircos. Results: We examined the 11-gene mutational profile of 927 unique AML patients. The frequency of these mutations was consistent with previous reports (Park et al, NEJM 2012). DNMT3A, NPM1, and FLT3 were the most common abnormalities (Figure 1A). 75 of these 927 patients were treated at our center. Of these, median age was 60.2 years; 53.3% were female; 80% were white, 13.3% black,6.7% other. 68% (n=51) self-identified as non-Hispanic and 32% (n=24) as Hispanic (Table 1). Median WBC was 3.95 K/µL (1.4-100.7); 37% had antecedent MDS or MDS-related changes; 8% had favorable-, 69% intermediate-, and 23% poor-risk cytogenetics. There were no differences in WBC, MDS, or cytogenetic risk by ethnicity. In the 75 patient SCCC cohort, TET2 (n=14, 18.7%), CEBPA (n=14, 21.9%), and DNMT3A (n=11, 14.7%) were the most common mutations. 16 patients had an IDH mutation (10 IDH1 and 6 IDH2). We found no statistically significant difference in mutation frequency in Hispanic vs. non-Hispanic patients (Figure 1B); however, there was a non-significant increase in MLL-PTD mutations in Hispanic patients (15.8% vs. 9.3%, p=0.665) and IDH1 mutations in non-Hispanics (11.8% vs. 0%, p=0.168). Conclusions: We present one of the largest examinations to date (n=927 patients) of mutational frequency in AML. Our findings corroborate prior literature; however, we did note the rare co-occurrence of TET2 and IDH mutations-showing the 2 are not always mutually exclusive. We examined demographic data on a subset of these patients treated at SCCC, where we have a large Hispanic population, and assessed for ethnic differences in mutational frequency. We found a suggestion of increased MLL PTD mutations, a poor-risk abnormality, in Hispanic patients, and increased IDH1 mutations in non-Hispanics, but found no significant differences. This may be due to our relatively small sample size. We are now examining the full 927 patient cohort for demographic data and updated results will be presented. If certain AML mutations cluster by ethnicity, this might explain differing incidence rates and outcomes in Hispanics, and examination of predisposing heritable or environmental factors should be pursued. Table 1. Distribution of mutations in an 11-gene AML molecular profile by ethnicity in 75 patients with AML. Figure 1.Circosplots characterizing the distribution of concomitant mutations in 927 patients with AML (A); and the incidence of mutations by ethnicity in 75 patients with AML (B). Figure 1 Figure 1. Figure 2 Figure 2. Disclosures Vaupel: Genoptix, a Novartis Company: Employment. Hall:Genoptix, a Novartis Company: Employment.


2021 ◽  
Vol 70 (1) ◽  
pp. 83-97
Author(s):  
Remco J. Molenaar ◽  
Johanna W. Wilmink

Isocitrate dehydrogenase 1 and 2 (IDH1/2) are enzymes recurrently mutated in various types of cancer, including glioma, cholangiocarcinoma, chondrosarcoma, and acute myeloid leukemia. Mutant IDH1/2 induce a block in differentiation and thereby contribute to the stemness and oncogenesis of their cells of origin. Recently, small-molecule inhibitors of mutant IDH1/2 have been Food and Drug Administration–approved for the treatment of IDH1/2-mutated acute myeloid leukemia. These inhibitors decrease the stemness of the targeted IDH1/2-mutated cancer cells and induce their differentiation to more mature cells. In this review, we elucidate the mechanisms by which mutant IDH1/2 induce a block in differentiation and the biological and clinical effects of the release into differentiation by mutant-IDH1/2 inhibitors. (J Histochem Cytochem 70:83–97, 2022)


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 787-787
Author(s):  
Ujunwa Cynthia Okoye-Okafor ◽  
Boris Bartholdy ◽  
Jessy Cartier ◽  
Enoch Gao ◽  
Beth Pietrak ◽  
...  

Abstract Mutations in the isocitrate dehydrogenase 1 (IDH1) gene are known driver mutations in acute myeloid leukemia (AML) and other cancer types. Patient outcomes in AML have remained poor, especially for patients above 60 years of age who typically do not tolerate high dose chemotherapy and stem cell transplantation, leading to cure rates below 20%. The development of novel targeted therapies for defined AML subtypes is urgently desired. Inhibitors of mutants of the closely related IDH2 gene as well as IDH1 have recently been described and show promising pre-clinical and early phase clinical activity. However, the specific molecular and functional effects of IDH1 inhibitors in AML, including in primary patients' cells, have not been reported yet. Here, we report the development of novel allosteric inhibitors of mutant IDH1 for differentiation therapy of acute myeloid leukemia. A high-throughput biochemical screen targeting an IDH1 heterodimer composed of R132H and WT IDH1 led to the identification of a tetrahydropyrazolopyridine series of inhibitors. Structural and biochemical analyses revealed that these novel compounds bind to an allosteric site that does not contact any of the mutant residues in the enzymes active site and inhibit enzymatic turnover. The enzyme complex locked in the catalytically inactive conformation inhibits the production of the oncometabolite 2-hydroxyglutarate (2-HG). In biochemical studies, we observed potent inhibition of several different clinically relevant R132 mutants in the presence or absence of the cofactor NADPH, accompanied by significant decrease in H3K9me2 levels. Allosteric inhibitor treatment of primary AML patients' cells with different clinically relevant R132 mutants of IDH1 ex vivo uniformly led to a decrease in intracellular 2-HG, abrogation of the myeloid differentiation block, increased cell death and induction of differentiation both at the level of leukemic blasts and immature stem-like cells. Allosteric inhibition of IDH1 also led to a decrease in blasts in an in vivo xenotransplantation model. At the molecular level, enhanced reduced representation bisulfite sequencing showed that treatment with allosteric IDH1 inhibitors led to a significant reversal of the DNA cytosine hypermethylation pattern induced by mutant IDH1, accompanied by gene expression changes of key sets of genes and pathways, including "Cell Cycle", "G1/S transition", "Cellular growth and proliferation", and "Cell death and survival". Taken together, our findings provide novel insight into the cellular and molecular effects of inhibition of mutant IDH1 in primary AML patients' cells. Furthermore, our study provides proof-of-concept for the molecular and biological activity of novel allosteric inhibitors for targeting of different mutant forms of IDH1 in leukemia, and opens new avenues for future investigations with these and other allosteric inhibitors for targeting mutant IDH1 in leukemia and other cancers. Disclosures Gao: GlaxoSmithKline: Employment. Pietrak:GlaxoSmithKline: Employment. Rendina:GlaxoSmithKline: Employment. Rominger:GlaxoSmithKline: Employment. Quinn:GlaxoSmithKline: Employment. Smallwood:GlaxoSmithKline: Employment. Wiggall:GlaxoSmithKline: Employment. Reif:GlaxoSmithKline: Employment. Schmidt:GlaxoSmithKline: Employment. Qi:GlaxoSmithKline: Employment. Zhao:GlaxoSmithKline: Employment. Joberty:GlaxoSmithKline: Employment. Faelth-Savitski:GlaxoSmithKline: Employment. Bantscheff:GlaxoSmithKline: Employment. Drewes:GlaxoSmithKline: Employment. Duraiswami:GlaxoSmithKline: Employment. Brady:GlaxoSmithKline: Employment. Concha:GlaxoSmithKline: Employment. Adams:GlaxoSmithKline: Employment. Schwartz:GlaxoSmithKline: Employment. McCabe:GlaxoSmithKline: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1699-1699
Author(s):  
Anna Andersson ◽  
David Miller ◽  
John Lynch ◽  
Andrew Lemoff ◽  
Zhongling Cai ◽  
...  

Abstract Abstract 1699 Pediatric de novo acute myeloid leukemia (AML) is a heterogeneous disease that can be divided into clinically distinct subtypes based on the presence of specific chromosomal abnormalities, gene mutations, or morphologic and immunophenotypic features. The best characterized subtypes include leukemias with alterations of the gene encoding the core-binding transcription factor complex, (t(8;21)[AML1- ETO] and inv(16)/t(16;16)[CBFβ-MYH11], rearrangements of the MLL gene on chromosome 11q23, normal cytogenetics, or distinct morphology including acute promyeloctic leukemia with t(15;17)[PML-RARA] and acute megakaryoblastic leukemia (FAB-M7). In AMLs with normal cytogenetics, mutations have also been identified in a number of genes, with alterations in NPM1, FLT3 and CEBPA occurring at an appreciable frequency and influencing therapeutic responses. Recent genome-wide sequencing efforts have led to the identification of a number of new candidate genes involved in the pathogenesis of this disease. Foremost among this list are isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2). IDH1 mutations were initially identified in a whole exome sequencing of glioblastoma multiforme (GBM), but were subsequently shown to be mutated in a variety of myeloid malignancies including up to 16% of adult AMLs with normal cytogenetics. The mutations in both GBM and myeloid malignancies have been heterozygous and restricted to arginine 132 in exon 4 of IDH1, or to either the homologous residue in IDH2, R172, or to a second arginine, R140, also located in its substrate binding pocket. Although the distribution of specific IDH1/IDH2 mutations varies between GBM and AML, each of the analyzed mutations result in a loss of the enzymes ability to catalyze the oxidative carboxylation of isocitrate to a-ketoglutarate (a-KG), coupled with a gain-of-function to catalyze the NADPH-dependent reduction of a–KG to 2-hydroxyglutarate (2-HG). This shift in enzymatic activity results in a dramatic increase in the levels of 2-HG within the leukemic cells; however, how the increase in this metabolite contributes to transformation remains to be determined. To investigate the frequency of IDH1 and IDH2 mutations in pediatric AML, we sequenced these genes in diagnostic samples from 227 pediatric AML patients. Our analysis identified somatic IDH1/2 mutations in 4% of cases (IDH1 N=3 and IDH2 N=5), with the frequency slightly higher in AMLs with a normal karyotype (7%). IDH1 mutations occurred in codon 132 resulting in replacement of arginine with either cysteine (N=2) or histidine (N=1). By contrast, the mutations in IDH2 did not affect the homologous residue but instead altered codon 140, resulting in replacement of an arginine with either glutamine (N=4) or tryptophan (N=1). Structural modeling studies of IDH2 suggested that the codon 140 mutations should disrupt the enzyme's ability to bind its substrate isocitrate. Consistent with this prediction, enzymology studies showed that recombinant IDH2 R140Q and R140W were unable to carry out the decarboxylation of isocitrate to α-ketoglutarate (α-KG), but instead gained the neomorphic activity to reduce α-KG to R(-)-2-hydroxyglutarate (2-HG). Analysis of primary leukemic blasts using mass spectrometry confirmed high levels of 2-HG in samples with IDH1/2 mutations. Interestingly, 3/5 leukemias also had FLT3 activating mutations, raising the possibility that these two mutations directly cooperate in leukemogenesis. Defining the biological role of the IDH1/2 mutations in leukemogenesis will benefit by a direct assessment of the biological effect of the mutations on normal murine hematopoietic cell differentiation using both in vitro and in vivo systems. Disclosures: No relevant conflicts of interest to declare.


PLoS ONE ◽  
2013 ◽  
Vol 8 (12) ◽  
pp. e83334 ◽  
Author(s):  
Lixun Guan ◽  
Li Gao ◽  
Lili Wang ◽  
Meng Li ◽  
Yue Yin ◽  
...  

2021 ◽  
Vol 11 (6) ◽  
Author(s):  
Ghayas C. Issa ◽  
Courtney D. DiNardo

AbstractAcute myeloid leukemia is a genetically heterogeneous hematologic malignancy; approximately 20% of AML harbors a mutation in the isocitrate dehydrogenase (IDH) genes, IDH1 or IDH2. These recurrent mutations in key metabolic enzymes lead to the production of the oncometabolite 2-hydroxyglutarate, which promotes leukemogenesis through a block in normal myeloid differentiation. Since this discovery, selective oral inhibitors of mutant IDH1 and IDH2 have subsequently been developed and are now approved as single agent therapy, based on clinical efficacy observed within the original first-in-human trials. The investigation of IDH inhibitors in combination with standard therapies such as azacytidine, with intensive chemotherapy, and with other small molecule targeted therapies in rational combinations are currently under evaluation to further improve upon clinical efficacy.


2014 ◽  
Vol 26 (1) ◽  
pp. 43-49 ◽  
Author(s):  
Ghada M. Elsayed ◽  
Hanan R. Nassar ◽  
Amr Zaher ◽  
Essam H. Elnoshokaty ◽  
Manar M. Moneer

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4469-4469
Author(s):  
Amy Song ◽  
Jasmine Mahajan ◽  
Nivetha Srinivasan ◽  
Kendra Sweet ◽  
David A. Sallman ◽  
...  

Abstract Introduction: Isocitrate dehydrogenase 1 (IDH1) gene is mutated in 7-14% of acute myeloid leukemia (AML) patients. IDH1 encodes for an enzyme that catalyzes the conversion of isocitrate to α- ketoglutarate. IDH1 mutation leads to accumulation of the oncometabolite 2-hydroxyglutarate. Clonal evolutionary dynamics of IDH1 mutations in AML have not been clearly characterized. The introduction of targeted small-molecule therapy, Ivosidenib, in AML treatment underscores the significance of understanding the topography of clonal dynamics in IDH1-mutated AML to optimize precision medicine. Methods: We analyzed ~6000 patients with next-generation sequencing (NGS) data and identified 107 patients with IDH1 mutated AML. Disease status was determined for each NGS test date by manual chart review. IDH1 mutation status was characterized during course of AML at diagnosis, remission, relapse, and with persistent disease. Cytogenic risk category was determined using ELN 2017 guidelines. Kaplan Meier survival analysis and log-rank test were used to determine significant differences in overall survival among patient groups. Results: Of the 107 total patients, 39 patients (36%) had AML with myelodysplastic-related changes (AML-MRC) and 39 patients (36%) had AML-NOS. The most frequently co-mutated genes were SRSF2, DNMT3A, ASXL1, RUNX1, NRAS, BCOR, STAG2, NPM1, JAK2, and FLT-3 in order of frequency. Of the total patients, 74 patients (69%) had good cytogenetics, 17 patients (16%) had intermediate cytogenetics, and 16 patients (15%) had poor/very poor cytogenetics. Among the patients with IDH1-mutated AML, 85 patients (79%) were IDH1-positive at initial diagnosis, while 22 patients (21%) were IDH1- negative at diagnosis and acquired the mutation later in disease course. Of those 22 patients, 18 patients gained the mutation in the setting of persistent disease, 3 patients at remission, and 1 patient at relapse. In those with persistent AML (n=42), 30 patients (71%) remained IDH1-positive while 12 patients (29%) lost the mutation. In those achieving remission (n=66), 12 patients (18%) who were IDH1-positive remained IDH1-positive while 51 patients (77%) cleared the mutation. In those with relapsed disease (n= 21), 17 patients (81%) with IDH1-positive AML remained IDH1-positive while 4 patients (19%) lost the mutation at relapse. There were no significant differences in median overall survival in patients who were positive or negative for IDH1 mutations at diagnosis, positive or negative with disease persistence, or among patients who remained IDH1-positive or lost the IDH1 mutation at disease relapse. Patients that were IDH1-positive at diagnosis were more likely to have poorer cytogenetics than patients who were IDH1-negative at diagnosis (p=0.0016). Conclusion: In summary, this study found that IDH1 mutations are unstable throughout the course of AML and periodic genetic testing of AML patients is necessary for optimizing precision medicine approaches. In disease remission, most patients (77%) cleared the IDH1 mutation. In the relapse setting, 81% of patients retained IDH1-positive status. Our study, the largest of its kind to our knowledge, shows that serial genomic profiling for the IDH1 mutation across disease course may be beneficial in helping to tailor targeted therapy for IDH1+ AML. Disclosures Sweet: Bristol Meyers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; AROG: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees. Sallman: Kite: Membership on an entity's Board of Directors or advisory committees; Agios: Membership on an entity's Board of Directors or advisory committees; Aprea: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Intellia: Membership on an entity's Board of Directors or advisory committees; Magenta: Consultancy; Incyte: Speakers Bureau; AbbVie: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy; Shattuck Labs: Membership on an entity's Board of Directors or advisory committees; Syndax: Membership on an entity's Board of Directors or advisory committees. Hussaini: Adaptive: Consultancy, Honoraria, Speakers Bureau; Stemline: Consultancy; Amgen: Consultancy; Seattle Genetics: Consultancy; Celegene: Consultancy; Decibio: Consultancy; Guidepoint: Consultancy; Bluprint Medicine: Consultancy.


2021 ◽  
Vol 67 (3) ◽  
pp. 92-98
Author(s):  
Amal Ezzat Abd El-Lateef ◽  
Manar M. Ismail ◽  
Mohammed Almohammadi ◽  
Amr M. Gawaly

Despite the great advance in treatment, cytogenetically normal Acute myeloid leukemia (CN-AML) is still a challenging entity. The discovery of IDH1 mutation in AML together with the frequent co-mutations; NPM1 and FLT3-ITD throughs a new insight into the pathogenesis and outcome of CN-AML. Recently, there has been an increasing number of recurring mutations in other genes for which the forecasting effect is still required. Despite the large number of risk variables established, there are relatively few prognostic indicators that can help in treatment decisions in AML patients. This study aimed at recording the frequency of IDH1 and NPM1 mutations in newly diagnosed AML and, dual clinicopathological significance. IDH1 and NPM1 mutations were analyzed using High-Resolution Melting curve analysis PCR in 78 newly diagnosed AML patients; 30 pediatric and 48 adult AML patients. IDH1 mutation was detected in 6 out of the 48 adult AML cases (12.5%) and all of them had intermediate cytogenetic prognostic stratification. 5/6 mutant IDH1 patients showed NPM1 co-mutation (P-value= 0.008). Mutant IDH1 patients showed significant resistance to induction therapy (P-value <0.001) and even those who achieved complete remission were relapsed later. Within the intermediate cytogenetic group, the IDH1 mutated patients had short overall survival (HR 12.9, 95% CI (3.1- 53.45) and event-free survival (HR 15.7, 95% CI (2.99-82.72) and P-value <0.001). IDH1 mutation is closely linked to the intermediate cytogenetic stratified group and in particular old age patients and has a great impact on their survival.


Sign in / Sign up

Export Citation Format

Share Document