Next Article in Journal
O-GlcNAcylation: An Emerging Protein Modification Regulating the Hippo Pathway
Previous Article in Journal
The High Proportion of Discordant EGFR Mutations among Multiple Lung Tumors
 
 
cancers-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prevalence and Prognostic Role of IDH Mutations in Acute Myeloid Leukemia: Results of the GIMEMA AML1516 Protocol

by Monica Messina 1, Alfonso Piciocchi 1, Tiziana Ottone 2,3, Stefania Paolini 4, Cristina Papayannidis 4, Federica Lessi 5, Nicola Stefano Fracchiolla 6, Fabio Forghieri 7, Anna Candoni 8, Andrea Mengarelli 9, Maria Paola Martelli 10, Adriano Venditti 2, Angelo Michele Carella 11, Francesco Albano 12, Valentina Mancini 13, Bernardi Massimo 14, Valentina Arena 1, Valeria Sargentini 1, Mariarita Sciumè 6, Domenico Pastore 15, Elisabetta Todisco 16, Giovanni Roti 17, Sergio Siragusa 18, Marco Ladetto 19, Stefano Pravato 5, Eleonora De Bellis 20, Giorgia Simonetti 21, Giovanni Marconi 22, Claudio Cerchione 22, Paola Fazi 1, Marco Vignetti 1, Sergio Amadori 2, Giovanni Martinelli 21,† and Maria Teresa Voso 2,3,*,†add Show full author list remove Hide full author list
1
GIMEMA Foundation, 00182 Roma, Italy
2
Ematologia, Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, 00133 Roma, Italy
3
Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, 00179 Roma, Italy
4
IRCCS Azienda Ospedaliero-Universitaria di Bologna Istituto di Ematologia “Seràgnoli” Bologna, 40138 Bologna, Italy
5
Ematologia ed Immunologia Clinica, Università degli Studi di Padova, 1222 Padua, Italy
6
UOC Ematologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
7
UO Ematologia-AOU Policlinico di Modena, 41125 Modena, Italy
8
Clinica Ematologica, ASUFC, Università degli Studi di Udine, 33100 Udine, Italy
9
UO Ematologia-IRCCS Istituto Nazionale Tumori Tumori Regina Elena, 00128 Roma, Italy
10
Sezione di Ematologia ed Immunologia Clinica, Università degli Studi di Perugia, 06123 Perugia, Italy
11
Ematologia e Centro Trapianti CSE Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
12
Hematology and Stem Cell Transplantation Unit, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari Aldo Moro, 70121 Bari, Italy
13
Ospedale Niguarda Ca Granda-SC Ematologia Blocco SUD, 20162 Milano, Italy
14
IRCCS San Raffaele Scientific Institute, 20132 Milano, Italy
15
UOC Ematologia Brindisi, 72100 Brindisi, Italy
16
Onco-Hematology Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
17
Azienda Ospedaliera Universitaria di Parma, Ematologia, Università di Parma, 43126 Parma, Italy
18
U.O. di Ematologia con Trapianto-A.U. Policlinico Paolo Giaccone, 90127 Palermo, Italy
19
AO SS Antonio e Biagio Arrigo, 15121 Alessandria, Italy
20
Hematology Unit, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy
21
Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, 47014 Meldola, Italy
22
Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori [M1] (IRST) IRCCS, 47014 Meldola, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(12), 3012; https://doi.org/10.3390/cancers14123012
Submission received: 13 May 2022 / Revised: 8 June 2022 / Accepted: 15 June 2022 / Published: 18 June 2022

Abstract

:

Simple Summary

IDH1/2 mutations are a common event in acute myeloid leukemia (AML) and represent a therapeutic target. We designed the GIMEMA AML1516 observational protocol to examine the prevalence of IDH1/2 mutations and the associations between IDH mutations and clinico-biological parameters in a cohort of Italian patients affected by AML. By analyzing 284 consecutive adult AML patients, we confirmed that IDH1 and IDH2 mutations are frequently detected–14% and 18%, respectively–at diagnosis. IDH1/2 mutations were significantly associated with an inferior performance status and non-complex karyotype when compared to IDH1/2-WT. With regards to the outcome, in the subset of IDH1/2-mutated patients the rate of complete remission achievement was 60.5% and overall survival at 2 years was 44.5%: these percentages did not significantly differ from IDH1/2-WT patients. However, given the availability of IDH1/2 inhibitors, it is important to recognize IDH1/2-mutated cases up-front to offer patients the most appropriate therapeutic strategy.

Abstract

IDH1/2 mutations are common in acute myeloid leukemia (AML) and represent a therapeutic target. The GIMEMA AML1516 observational protocol was designed to study the prevalence of IDH1/2 mutations and associations with clinico-biological parameters in a cohort of Italian AML patients. We analyzed a cohort of 284 AML consecutive patients at diagnosis, 139 females and 145 males, of a median age of 65 years (range: 19–86). Of these, 38 (14%) harbored IDH1 and 51 (18%) IDH2 mutations. IDH1/2 mutations were significantly associated with WHO PS >2 (p < 0.001) and non-complex karyotype (p = 0.021) when compared to IDH1/2-WT. Furthermore, patients with IDH1 mutations were more frequently NPM1-mutated (p = 0.007) and had a higher platelet count (p = 0.036). At relapse, IDH1/2 mutations were detected in 6 (25%) patients. As per the outcome, 60.5% of IDH1/2-mutated patients achieved complete remission; overall survival and event-free survival at 2 years were 44.5% and 36.1%, respectively: these rates were similar to IDH1/2-WT. In IDH1/2-mutated patients, high WBC proved to be an independent prognostic factor for survival. In conclusion, the GIMEMA AML1516 confirms that IDH1/2 mutations are frequently detected at diagnosis and underlines the importance of recognizing IDH1/2-mutated cases up-front to offer the most appropriate therapeutic strategy, given the availability of IDH1/2 inhibitors.

1. Introduction

Progresses in the knowledge of the genetic landscape of AML—accelerated by high throughput sequencing technologies—led to a better understanding of AML pathogenesis and enhanced the development of targeted approaches.
Mutations targeting epigenetic regulators emerged as one of the most common events—accounting for >50% of AML patients—and contribute to the differentiation block typical of AML [1,2]. Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) belong to the class of epigenetic modulators and mutations of these genes occur in up to 20% of adult AML cases [3,4,5,6] and 30% of pediatric AML [7]. IDH1 and IDH2 mutations target the conserved arginine residues, namely R132 of IDH1, and R140 and R172 of IDH2 [8]. They determine an aberrant production of 2-hydroxyglutarate (2HG) that acts as an antagonist of α-KG; thus, inhibiting the activity of multiple α-KG-dependent dioxygenases, including both histones and DNA demethylases involved in epigenetic control of gene expression. As a consequence, IDH1 and IDH2 mutations determine an aberrant hypermethylated phenotype and, ultimately, influence cell differentiation [4,9].
IDH1/2 mutations are associated with intermediate-risk cytogenetics and NPM1 mutations [4,10], in particular, in the absence of DNA-damage-related and cohesin gene mutations [11]. Moreover, Chou et al. reported an association between IDH1/2 mutations and higher platelet counts, normal karyotype, and isolated trisomy 8 [12,13,14].
The impact of IDH1/2 mutations on AML prognosis is controversial, and depends on the specific AML subsets, i.e., normal karyotype or FLT3-WT/NPM1-WT, or treatment groups, i.e., standard intensive chemotherapy [5,6,15,16]. In other cohorts, IDH1/2 mutations do not impact on prognosis [17,18].
Despite this, IDH1 and IDH2 immediately qualified as promising therapeutic targets, due to the activating nature of their recurrent mutations: small inhibitory molecules have been developed and were tested in clinical trials, as monotherapy or in combination with chemotherapy or azacitidine [19,20,21,22]. Two orally bioavailable IDH inhibitors, enasidenib (IDH2 inhibitor) and ivosidenib (IDH1 inhibitor) are now FDA-approved [19,20]: the former for the treatment of IDH2-mutated relapsed-refractory AML, and the latter for both IDH1-mutated relapsed-refractory and newly diagnosed AML unfit for intensive chemotherapy.
This progress has a substantial impact on the therapeutic algorithm of AML patients harboring IDH1/2 mutations. Therefore, the assessment of IDH1/2 mutations is pivotal to identify the population of patients that might benefit from the use of IDH inhibitors, at diagnosis or at relapse.
We present here the results of the GIMEMA AML1516 protocol, designed to (i) study the prevalence of IDH1 and IDH2 mutations in patients with AML at the time of initial diagnosis and at relapse, (ii) evaluate the association between IDH mutations and patient or disease characteristics, and (iii) assess the impact on response to treatment and survival.

2. Materials and Methods

2.1. GIMEMA AML1516 Study Design

The GIMEMA AML1516 protocol (ClinicalTrials.gov Identifier: NCT02986620) is an observational study aimed at collecting data on IDH1 and IDH2 mutational status in adult AML patients in Italy treated as per clinical practice, not including IDH1/2 inhibitors. The primary objective of the trial was to estimate the prevalence and type of IDH mutations in AML at initial diagnosis and relapse. The secondary objectives were to evaluate the associations between IDH mutations and clinico-biological parameters (i.e., age, white blood cell (WBC), lactate dehydrogenase (LDH), cytogenetics, NPM1, FLT3-ITD, CEBPA alterations), AML type, treatment response, and survival.
The study was active starting from May 2017 to January 2020 and included a retrospective and a prospective cohort.
The analysis of IDH1 and IDH2 mutations was performed either by Sanger sequencing or NGS technologies at local laboratories [8,23]. The sensitivity of Sanger sequencing analysis is approximately 15–20% and the presence of chromatograms with a double peak into wild-type gene sequence identified an IDHs gene mutation. For the assessment of IDHs status by NGS assay, the detection limit of the variant allele frequency (VAF) was 5%.
Study data were collected and managed using REDCap electronic data capture tools hosted at GIMEMA Foundation [24,25].

2.2. Statistical Analysis

Characteristics of patients were summarized by means of cross-tabulations or quantiles.
IDH1-IDH2 mutation detection was evaluated in terms of percentage of patients at the time of initial diagnosis and relapse.
Non-parametric tests were applied, in univariate analysis, for comparisons between groups, chi-squared and Fisher exact test for difference in terms of categorical variables or mutation rate, Mann–Whitney and Kruskal–Wallis tests for difference in terms of continuous variables. All clinical parameters, genetic subtypes, and treatment received were considered in the univariate analyses. The multivariate models considered all relevant clinical/biologic variables or covariates with a p-value less than 0.15 in the univariate analysis.
Logistic regression models were used in univariate and multivariate analyses to assess if the clinical and biological parameters are associated to response outcomes (CR and ORR rate). Odds ratios (OR) and 95% confidence intervals were reported as parameter results of the logistic regression models.
Survival distributions (e.g., overall survival (OS), event-free survival (EFS)) were estimated using the Kaplan–Meier product limit estimator. Subgroup comparisons with clinical and biological parameters were performed for descriptive purposes.
Differences in terms of time to response OS and EFS were evaluated by means of log-rank test or Cox regression model in univariate and multivariate analyses, after assessment of proportionality of hazards.
Hazard ratios (HR) and 95% confidence interval were reported as parameter results of the Cox regression models.

3. Results

3.1. Study Population

Between 5/2017 and 1/2020, 393 consecutive patients were diagnosed with AML at 17 Italian Hematology Centers and members of the GIMEMA working group, enrolled in the AML1516 study. Of them, 388 were deemed eligible. IDH1/2 mutational status was available for 361 patients (337 at diagnosis and 24 at relapse).
The present analysis is based on 284 patients studied at diagnosis with available IDH1/2 mutation status, treatment and follow-up data. At diagnosis, 145 (51%) patients were males and 139 (49%) were females. Median age was 65 (range 19–86) years. In total, 229 (81%) patients had a de novo, 37 (13%) a secondary, and 16 (5.7%) a therapy-related AML. Cytogenetics was available for 259 patients, 132 (50.9%) had a normal and 29 (11.2%) a complex karyotype. As per the main chromosomal aberrations, anomalies of chromosome 5 (del5q, monosomy 5) occurred in 20 patients (7.7%), and aberrations of chromosome 7 (del7q, monosomy 7) were detected in a total of 20 patients (7.7%); in 23 patients a trisomy 8 was documented. Recurrent rearrangements, including RUNX1T1-RUNX1 and CBF-MYH11 were detected in 5 (1.9%) and 10 (3.9%) patients. FLT3 mutations were detected in 60/271 (22.4%), NPM1 in 71/266 (26.7%).
With regards to the treatment received, 201 (71%) patients were treated with conventional chemotherapy, 76 (27%) with hypomethylating agents, and the remaining 7 patients with other treatment schemes.
Demographic characteristics are summarized in Table 1.

3.2. Incidence, Type of IDH1/2 Mutations, and Patients’ Clinico-Biological Features

Of 284 patients studied at diagnosis, 38 (14%) carried IDH1 mutations and 51 (18%) IDH2 mutations (Figure 1A). With regards to the type of IDH1 mutations, the majority (32, 84.2%) targeted R132, with R132C and R132H being the most common substitutions. Similarly, R140 was the most commonly involved residue of IDH2 (30, 58.8%)—with R140Q accounting for the vast majority of substitutions—followed by R172K detected in 19 cases (37.2%), as depicted in Figure 1B,C.
IDH1/2 mutations were significantly associated with WHO PS >2 (p < 0.001) and non-complex karyotype (p = 0.021) when compared to IDH1/2-WT. As per MDS-related anomalies, a WT status of IDH1/IDH2 was associated with del5q (p = 0.035). Furthermore, patients with IDH1 mutations had higher platelet counts (p = 0.036) and were more frequently NPM1-mutated (p = 0.007, Table 1).
At relapse, 5 (21%) patients carried IDH1 mutations, all targeting R132 with R132H being the most common substitution (3 out of 5); 1 patient had a concomitant FLT3-ITD mutation and another patient a concurrent TP53 mutation. Only 1 patient (4.2%) harbored a IDH2 mutation, that targeted the R172 residue.

3.3. Treatment Response and Survival According to IDH1/2 Mutations

Out of 284 patients with therapy information, 201 (71%) were treated with a conventional chemotherapy approach (CHT), 76 (27%) with a hypomethylating agent (HMA), and 7 (2.5%) with other regimens.
Overall, 228 patients were evaluable for response and 128 (56%) achieved complete remission (CR). When considering CHT vs. HMA, 113 of 181 (62.4%) treated with conventional CHT achieved a CR while only 13 of 41 (31.7%) treated with HMA obtained a CR (p < 0.0001). There were no differences in CR rate when stratifying patients according to IDH1/2 mutational status (60.5% CR in IDH1/2-mutated vs. 64% in IDH1/2-WT patients).
Indeed, the parameters with an impact on CR, resulting from the logistic regression model, were younger age (OR 0.96 95% CI 0.94–0.98, p < 0.001), WHO performance status (OR 0.2 95% CI 0.07, 0.51, p = 0.001), de novo AML (OR 0.17, 95% CI 0.06–0.39, p < 0.001), NPM1 mutations (OR 2.26, 95% CI 1.29–4.42, p = 0.013), and conventional CHT treatment (OR 0.28, 95% CI 0.13–0.57, p < 0.001, Table S1). WHO PS and AML type retained statistical significance also in the multivariate model.
Overall response was obtained by 167 (73%) patients, at a similar rate in IDH1/2-mutated (71%) and IDH1/2-WT (68%) patients.
With a median follow-up of 22.5 months (13.5–35.7), overall survival (OS) at 24 months was 43.7% (95% CI 37.5–50.9) and event-free survival (EFS) was 30% (95% CI 24.5–36.6).
Overall, there were no differences in OS or EFS in patients with IDH1/2-mutated vs. WT AML (44.5% vs. 43.3% Figure 2A, and 36.1% vs. 26.6%, Figure 2B, respectively).
Additionally, we did not document any difference in OS and EFS grouping patients according to the IDH mutation type (IDH1-R132 vs. IDH2-R140 vs. IDH2-R172 vs. IDH-WT, Figure 2C,D).
Analyzing the clinico-biological parameters that impact on survival outcomes, the univariate analyses showed that patients treated with CHT when compared to HMT, had a significantly longer OS (49.5% vs. 21%, p < 0.001) and EFS (34.4% vs. 15.3%, p = 0.0013). Furthermore, in the univariate model, age (HR 1.04, 95% CI 1.02–1.05, p < 0.001), high WBC (HR 1.0, 95% CI 1.0–1.0, p = 0.004), WHO PS (1 vs. 0: HR 1.9, 95% CI 1.3–2.9, p < 0.0001; 2 vs. 0: HR 2.7, 95% CI 1.6–4.5, p < 0.0001), complex karyotype (HR 2.83, 95% CI 1.79–4.45, p < 0.001), and HMA treatment (HR 2.2, 95% CI 1.56–3.10, p < 0.001) negatively impacted on OS and EFS. Age, WHO PS, and complex karyotype proved independent prognostic factors for OS and EFS in the multivariable model, as detailed in Table 2.
When restricting the analysis to IDH1/2-mutated patients, the univariate analyses for OS and EFS confirmed also in this AML subset that age (HR 1.03, 95% CI 1.00–1.06, p = 0.019), WBC (HR 1.0, 95% CI 1.00–1.00, p = 0.007), and HMA (HR 2.03, 95% CI 1.07–3.83, p = 0.030) were associated with inferior survival. In the multivariate analysis, only WBC was confirmed as an independent prognostic factor (Table 2).

4. Discussion

The GIMEMA AML1516 study illustrates the prevalence of IDH1/2 mutations in AML in Italy and adds further evidence on the value of screening these aberrations for therapeutic purposes.
By analyzing a cohort of 284 adult AML mainly de novo, we found that 32% of patients carried either a IDH1 or IDH2 mutation at diagnosis. Compared to the literature, this incidence is higher than reported in other patient cohorts (30% vs. 20%) and this may be due to the observational nature of the study, and to a selection bias [3,4,5,6]. As already reported, the most common IDH1 changes were R132C and R132H, while R140Q and R172K were the most frequent substitutions in IDH2 [8]. As per the association with clinico-biological parameters, we documented that IDH1/2-mutated AML had more frequently a WHO PS >2. We did not document any association with a specific cytogenetic subset, but we found that IDH1/2-mutated AML more frequently display a non-complex karyotype. Additionally, IDH1 mutation was associated with higher platelet counts and with NPM1 mutations, in agreement with Chou et al. [12].
Next, we analyzed the outcome of patients with IDH1/2-mutated AML in comparison with IDH1/2-WT. We did not document any difference neither in terms of CR achievement, nor of survival. In our cohort, the CR rate was 60% in IDH1/2-mutated cases, while OS and EFS at 24 months were 44% and 36%, respectively. Additionally, there was no impact on outcome when grouping patients according to common IDH1/2 mutations (i.e., IDH1-R132, IDH2-R140, and IDH2-R172).
Therefore, our data reinforce the notion that IDH1/2 mutational status does not impact on prognosis, in line with the reports by Di Nardo and Chotirat [17,18]. Accordingly, Middeke et al. recently reported on the prognostic role of IDH1/2 mutations in the largest AML cohort treated with intensive chemotherapy [26]. They did not find any difference in response rates, nor in survival for patients carrying IDH1/2 mutations when compared to IDH1/2-WT patients. However, when the most common IDH1/2 substitutions were analyzed, they found that IDH1-R132C is associated with a lower rate of complete remission and a trend towards shorter OS compared to other IDH1 mutations and IDH1/2-WT. On the contrary, patients with IDH2-R172K-mutated AML had a better OS within the ELN2017 intermediate/adverse risk groups, compared to IDH1/2-WT. Most likely, we failed to detect this difference because of the low number of cases included in these subgroups.
Other parameters, such as age and WBC count, had an impact on survival, independent of IDH mutations.
Lastly, we evaluated the incidence of IDH1/2 mutations at relapse and we found that 25% of cases were IDH1/2 mutated. However, this data relies on the analysis of only 24 patients; thus, representing a limitation of the present study. To this regard, Chou and colleagues reported that IDH mutations are stable during disease course and even detected at CR [12,27].
As discussed above, the prognostic relevance of IDH1/2 mutations is not straightforward. Notwithstanding, IDH1 and IDH2 display a prominent therapeutic role since they can be pharmacologically targeted. In the past few years, clinical trials showed that IDH1/2 inhibitors are well-tolerated and efficacious as monotherapy. In particular, the first IDH inhibitors approved by FDA were enasidenib, a selective allosteric inhibitor of IDH2-mutated, and ivosidenib that competes with magnesium for binding to mutated IDH1 enzyme.
Both were approved for the treatment of relapsed/refractory AML and induced a CR/CRh rate of approximately 30% [19,20], and a median OS of roughly 9 months. Subsequently, ivosidenib was approved also for newly diagnosed patients ineligible for chemotherapy. In this subset, the CR/CRh rate was 42% and OS 12% [28]. In newly diagnosed AML, further improvements were obtained with the use of combination approaches. Indeed, in combination with azacytidine and with the standard 7 + 3 intensive chemotherapy approach, IDH1/2 inhibitors induced CR/CRh rates exceeding 60% [21,22,29,30,31]. Another option is the combination with venetoclax that is highly effective in NPM1/IDH-mutated AML [32].
Second generation and “pan” IDH inhibitors are currently under development.
This progress broadens the therapeutic armamentarium of IDH1/2-mutated AML; thus, contributing to the shift to targeted regimens alone or in combination or in sequence. Therefore, the possible use of these strategies makes the screening of IDH1/2 mutations of utmost importance.

5. Conclusions

The GIMEMA AML1516 confirms that IDH1/2 mutations are frequently detected at diagnosis in an Italian AML cohort of patients and underlines the importance of recognizing IDH1/2-mutated cases up-front to offer patients the most appropriate therapeutic strategy.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14123012/s1, Table S1: Univariate Logistic regression model for CR.

Author Contributions

Conceptualization, S.P. (Stefania Paolini), C.P., G.S., G.M. (Giovanni Marconi), S.A., G.M. (Giovanni Martinelli) and M.T.V.; data curation, M.M., A.P., T.O., F.L., N.S.F., F.F., A.C., A.M., M.P.M., A.V., A.M.C., F.A., V.M., B.M., V.A., V.S., M.S., D.P., E.T., G.R., S.S., M.L., S.P. (Stefano Pravato), E.D.B., G.S. and C.C.; formal analysis, A.P. and V.A.; funding acquisition, G.M. (Giovanni Martinelli); investigation, M.M., A.P., G.M. (Giovanni Martinelli) and M.T.V.; project administration, S.P. (Stefania Paolini), P.F. and M.V.; supervision, V.S., P.F., M.V. and M.T.V.; writing—original draft, M.M.; writing—review and editing, N.S.F., M.P.M., A.V., G.S., G.M. (Giovanni Marconi), P.F., M.V., S.A., G.M. (Giovanni Martinelli) and M.T.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Bristol-Myers Squibb.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by Comitato Etico Indipendente dell’AOU di Bologna, Policlinico S. Orsola—Malpigni Approval Code (EC Study reference number): n° 42/2017/U/Oss Approval Date: 16 May 2017.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

M.V.: Amgen; Novartis, Mattioli srl; IQVIA; Dephaforum S.r.l.; M.L.: AbbVie, Acerta, Amgen, ADC Therapeutics, Astra Zeneca, BeiGene Celgene, GSKI, Gentili, Gilead/Kite, Novartis, Incyte J and J, Jazz, Regeneron, Roche, Sandoz, Takeda. Non-financial interests: PI or strategic investigator in studies supported by: Celgene, J&J, BeiGene, ADC Therapeutics; GMartinelli: Abbvie; Incyte; Pfizer; Celgene/BMS; Amgen, Roche; GlaxoSmithKline; Astellas; Daiichi Sankyo; Takeda; Janssen; Servier. Research support from: Pfizer, AbbVie, AstraZeneca, Daiichi Sankyo, Takeda, and Ariad/Incyte. GMarconi: Menarini/stemline, Pfizer, Servier, and Astellas and research support from Pfizer, AbbVie, and AstraZeneca. The other authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Ley, T.J.; Miller, C.; Ding, L.; Raphael, B.J.; Mungall, A.J.; Robertson, A.; Hoadley, K.; Triche, T.J., Jr.; Laird, P.W. Cancer Genome Atlas Research Network, Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tyner, J.W.; Tognon, C.E.; Bottomly, D.; Wilmot, B.; Kurtz, S.E.; Savage, S.L.; Long, N.; Schultz, A.R.; Traer, E.; Abel, M.; et al. Functional genomic landscape of acute myeloid leukaemia. Nature 2018, 562, 526–531. [Google Scholar] [CrossRef]
  3. Mardis, E.R.; Ding, L.; Dooling, D.J.; Larson, D.E.; McLellan, M.D.; Chen, K.; Koboldt, D.C.; Fulton, R.S.; Delehaunty, K.D.; McGrath, S.D.; et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 2009, 361, 1058–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Figueroa, M.E.; Abdel-Wahab, O.; Lu, C.; Ward, P.S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H.F.; et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18, 553–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Paschka, P.; Schlenk, R.F.; Gaidzik, V.I.; Habdank, M.; Krönke, J.; Bullinger, L.; Späth, D.; Kayser, S.; Zucknick, M.; Götze, K.; et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J. Clin. Oncol. 2010, 28, 3636–3643. [Google Scholar] [CrossRef] [PubMed]
  6. Boissel, N.; Nibourel, O.; Renneville, A.; Gardin, C.; Reman, O.; Contentin, N.; Bordessoule, D.; Pautas, C.; de Revel, T.; Quesnel, B.; et al. Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: A study by the Acute Leukemia French Association group. J. Clin. Oncol. 2010, 28, 3717–3723. [Google Scholar] [CrossRef]
  7. Damm, F.; Thol, F.; Hollink, I.; Zimmermann, M.; Reinhardt, K.; van den Heuvel-Eibrink, M.M.; Zwaan, C.M.; de Haas, V.; Creutzig, U.; Klusmann, J.H.; et al. Prevalence and prognostic value of IDH1 and IDH2 mutations in childhood AML: A study of the AML-BFM and DCOG study groups. Leukemias 2011, 25, 1704–1710. [Google Scholar] [CrossRef]
  8. Marcucci, G.; Maharry, K.; Wu, Y.Z.; Radmacher, M.D.; Mrózek, K.; Margeson, D.; Holland, K.B.; Whitman, S.P.; Becker, H.; Schwind, S.; et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: A Cancer and Leukemia Group B study. J. Clin. Oncol. 2010, 28, 2348–2355. [Google Scholar] [CrossRef] [Green Version]
  9. Ye, D.; Xiong, Y.; Guan, K.L. The mechanisms of IDH mutations in tumorigenesis. Cell Res. 2012, 22, 1102–1104. [Google Scholar] [CrossRef] [Green Version]
  10. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
  11. Simonetti, G.; Mengucci, C.; Padella, A.; Fonzi, E.; Picone, G.; Delpino, C.; Nanni, J.; De Tommaso, R.; Franchini, E.; Papayannidis, C.; et al. Integrated genomic-metabolic classification of acute myeloid leukemia defines a subgroup with NPM1 and cohesin/DNA damage mutations. Leukemias 2021, 35, 2813–2826. [Google Scholar] [CrossRef] [PubMed]
  12. Chou, W.C.; Lei, W.C.; Ko, B.S.; Hou, H.A.; Chen, C.Y.; Tang, J.L.; Yao, M.; Tsay, W.; Wu, S.J.; Huang, S.Y.; et al. The prognostic impact and stability of Isocitrate dehydrogenase 2 mutation in adult patients with acute myeloid leukemia. Leukemias 2011, 25, 246–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Falini, B.; Spinelli, O.; Meggendorfer, M.; Martelli, M.P.; Bigerna, B.; Ascani SStein, H.; Rambaldi, A.; Haferlach, T. IDH1-R132 changes vary according to NPM1 and other mutations status in AML. Leukemias 2019, 33, 1043–1047. [Google Scholar] [CrossRef]
  14. Meggendorfer, M.; Cappelli, L.V.; Walter, W.; Haferlach, C.; Kern, W.; Falini, B.; Haferlach, T. IDH1R132, IDH2R140 and IDH2R172 in AML: Different genetic landscapes correlate with outcome and may influence targeted treatment strategies. Leukemias 2018, 32, 1249–1253. [Google Scholar] [CrossRef]
  15. Thol, F.; Damm, F.; Wagner, K.; Göhring, G.; Schlegelberger, B.; Hoelzer, D.; Lübbert, M.; Heit, W.; Kanz, L.; Schlimok, G.; et al. Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia. Blood 2010, 116, 614–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Abbas, S.; Lugthart, S.; Kavelaars, F.G.; Schelen, A.; Koenders, J.E.; Zeilemaker, A.; van Putten, W.J.; Rijneveld, A.W.; Löwenberg, B.; Valk, P.J. Acquired mutations in the genes 525 encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid 526 leukemia: Prevalence and prognostic value. Blood 2010, 116, 2122–2126. [Google Scholar] [CrossRef]
  17. Chotirat, S.; Thongnoppakhun, W.; Promsuwicha, O.; Boonthimat, C.; Auewarakul, C.U. Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients. J. Hematol. Oncol. 2012, 5, 5. [Google Scholar] [CrossRef] [Green Version]
  18. DiNardo, C.D.; Ravandi, F.; Agresta, S.; Konopleva, M.; Takahashi, K.; Kadia, T.; Routbort, M.; Patel, K.P.; Brandt, M.; Pierce, S.; et al. Characteristics, clinical outcome, and 538 prognostic significance of IDH mutations in AML. Am. J. Hematol. 2015, 90, 732–736. [Google Scholar] [CrossRef] [Green Version]
  19. Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
  20. DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
  21. DiNardo, C.D.; Stein, A.S.; Stein, E.M.; Fathi, A.T.; Frankfurt, O.; Schuh, A.C.; Döhner, H.; Martinelli, G.; Patel, P.A.; Raffoux, E.; et al. Mutant Isocitrate Dehydrogenase 1 Inhibitor Ivosidenib in Combination With Azacitidine for Newly Diagnosed Acute Myeloid Leukemia. J. Clin. Oncol. 2021, 39, 57–65. [Google Scholar] [CrossRef] [PubMed]
  22. DiNardo, C.D.; Schuh, A.C.; Stein, E.M.; Montesinos, P.; Wei, A.H.; de Botton, S.; Zeidan, A.M.; Fathi, A.T.; Kantarjian, H.M.; Bennett, J.M.; et al. Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): A single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 2021, 22, 1597–1608. [Google Scholar] [CrossRef]
  23. Aguilera-Diaz, A.; Vazquez, I.; Ariceta, B.; Mañú, A.; Blasco-Iturri, Z.; Palomino-Echeverría, S.; Larrayoz, M.J.; García-Sanz, R.; Prieto-Conde, M.I.; Del Carmen Chillón, M.; et al. Assessment of the clinical utility of four NGS panels in myeloid malignancies. Suggestions for NGS panel choice or design. PLoS ONE 2020, 15, e0227986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Harris, P.A.; Taylor, R.; Thielke, R.; Payne, J.; Gonzalez, N.; Conde, J.G. Research electronic data capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J. Biomed. Inform. 2009, 42, 377–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Harris, P.A.; Taylor, R.; Minor, B.L.; Elliott, V.; Fernandez, M.; O’Neal, L.; McLeod, L.; Delacqua, G.; Delacqua, F.; Kirby, J.; et al. REDCap Consortium, The REDCap consortium: Building an international community of software partners. J. Biomed. Inform. 2019, 95, 103208. [Google Scholar] [CrossRef] [PubMed]
  26. Middeke, J.M.; Metzeler, K.H.; Röllig, C.; Kramer, M.; Eckardt, J.N.; Stasik, S.; Greif, P.A.; Spiekermann, K.; Rothenberg-Thurley, M.; Krug, U.; et al. Differential impact of IDH1/2 mutational subclasses on outcome in adult AML: Results from a large multicenter study. Blood Adv. 2022, 6, 1394–1405. [Google Scholar] [CrossRef]
  27. Chou, W.C.; Peng, K.Y.; Lei, W.C.; Ko, B.S.; Tsay, W.; Kuo, C.H.; Tien, H.F. Persistence of mutant isocitrate dehydrogenase in patients with acute myeloid leukemia in remission. Leukemias 2012, 26, 527–529. [Google Scholar] [CrossRef] [Green Version]
  28. Roboz, G.J.; DiNardo, C.D.; Stein, E.M.; de Botton, S.; Mims, A.S.; Prince, G.T.; Altman, J.K.; Arellano, M.L.; Donnellan, W.; Erba, H.P.; et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020, 135, 463–471. [Google Scholar] [CrossRef] [Green Version]
  29. Stein, E.M.; DiNardo, C.D.; Fathi, A.T.; Mims, A.S.; Pratz, K.W.; Savona, M.R.; Stein, A.S.; Stone, R.M.; Winer, E.S.; Seet, C.S.; et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: A phase 1 study. Blood 2021, 137, 1792–1803. [Google Scholar] [CrossRef]
  30. Venugopal, S.; Takahashi, K.; Daver, N.; Maiti, A.; Borthakur, G.; Loghavi, S.; Short, N.J.; Ohanian, M.; Masarova, L.; Issa, G.; et al. Efficacy and safety of enasidenib and azacitidine combination in patients with IDH2 mutated acute myeloid leukemia and not eligible for intensive chemotherapy. Blood Cancer J. 2022, 12, 10. [Google Scholar] [CrossRef]
  31. DiNardo, C.D.; Tiong, I.S.; Quaglieri, A.; MacRaild, S.; Loghavi, S.; Brown, F.C.; Thijssen, R.; Pomilio, G.; Ivey, A.; Salmon, J.M.; et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020, 135, 791–803. [Google Scholar] [CrossRef] [PubMed]
  32. Lachowiez, C.A.; Borthakur, G.; Loghavi, S.; Zeng, Z.; Kadia, T.M.; Masarova, L.; Takahashi, K.; Tippett, G.D.; Naqvi, K.; Bose, P.; et al. A phase Ib/II study of ivosidenib with venetoclax +/− azacitidine in IDH1-mutated myeloid malignancies. J. Clin. Oncol. 2020, 38, 7500. [Google Scholar] [CrossRef]
Figure 1. (A) Incidence of IDH1/2 mutations at AML diagnosis; distribution of IDH1 (B) and IDH2 (C) mutation subtypes.
Figure 1. (A) Incidence of IDH1/2 mutations at AML diagnosis; distribution of IDH1 (B) and IDH2 (C) mutation subtypes.
Cancers 14 03012 g001
Figure 2. (A) OS by IDH1/2 mutations; (B) EFS by IDH1/2 mutations; (C) OS by the most frequent IDH1/2 mutations; (D) EFS by the most frequent IDH1/2 mutations.
Figure 2. (A) OS by IDH1/2 mutations; (B) EFS by IDH1/2 mutations; (C) OS by the most frequent IDH1/2 mutations; (D) EFS by the most frequent IDH1/2 mutations.
Cancers 14 03012 g002
Table 1. Patient characteristics by IDH1/2 mutations.
Table 1. Patient characteristics by IDH1/2 mutations.
IDH1-IDH2 Mutated vs. IDH1-IDH2 (Both)WT
CharacteristicOverall, n = 284 IDH1-IDH2 WT, n = 195IDH1-Mut, n = 38IDH2-Mut, n = 51p-Value 1
Gender, n (%) 0.15
M145 (51%)93 (48%)20 (53%)32 (63%)
F139 (49%)102 (52%)18 (47%)19 (37%)
Age starting treatment,
median (range)
65 (19, 86) 65 (19, 85) 66 (22, 86) 65 (32, 85) 0.86
WBC (109/L), median (range) 7 (0.5, 800) 8 (0.5, 347) 5 (1, 600) 4 (0.4, 800) 0.063
HB (g/dL), median (range)9.00 (2.50, 14.9) 9.00 (4.50, 14.2)8.80 (7.20, 13.20) 9.30 (2.50, 14.90) 0.28
PLTS (109/L), median (range)56 (4, 789) 53 (4, 664) 110 (6, 742) 56 (10, 789) 0.036
Blasts (% in BM), median (range)50 (3, 99) 48 (4, 99) 75 (3, 96) 70 (4, 95) 0.027
WHO PS, n (%) <0.001
0118 (44%) 79 (43%) 15 (39%) 24 (50%)
I111 (41%) 89 (48%) 12 (32%) 10 (21%)
II 34 (13%) 16 (8.6%) 7 (18%) 11 (23%)
III 8 (3.0%) 1 (0.5%) 4 (11%) 3 (6.2%)
AML type, n (%) 0.63
de novo 229 (81%) 154 (80%) 31 (82%) 44 (86%)
secondary 37 (13%) 25 (13%) 6 (16%) 6 (12%)
therapy related 16 (5.7%) 14 (7.3%) 1 (2.6%) 1 (2.0%)
AML secondary, n (%) 0.71
MDS24 (65%) 16 (64%)4 (67%) 4 (67%)
ET0 (0%)0 (0%)0 (0%)0 (0%)
PV3 (8.1%) 3 (12%) 0 (0%) 0 (0%)
MF5 (14%) 2 (8.0%) 1 (17%) 2 (33%)
FLT3, n (%) 0.177
ITD48 (18%) 29 (15%) 13 (38%) 6 (14%)
TKD10 (3.7%) 9 (4.6%) 1 (2.9%) 0 (0%)
ITD and TKD2 (0.7%)2 (1.0%) 0 (0%) 0 (0%)
Mutated NPM1, n (%)71 (27%) 44 (23%) 17 (50%) 10 (25%) 0.007
Mutated TP53, n (%)1 (6.2%) 1 (8.3%) 0 (NA%) 0 (0%) >0.99
Mutated CEBPA, n (%)3 (9.7%) 2 (15%) 1 (10%) 0 (0%) 0.77
Mutated IDH1, n (%) 38 (13%) 0 (0%) 38 (100%) 0 (0%) <0.001
Mutated IDH2, n (%)51 (18%) 0 (0%) 0 (0%) 51 (100%) <0.001
Complex karyotype, n (%)29 (11%) 26 (14%) 0 (0%) 3 (6.4%) 0.021
Treatment, n (%) 0.071
Conventional CHT201 (71%) 136 (70%) 29 (76%) 36 (71%)
Hypomethylating76 (27%) 57 (29%) 8 (21%) 11 (22%)
1 Significant p-values are indicated in bold.
Table 2. Multivariate models for OS in the whole population of study and in the subset of IDH1/2 mutated patients.
Table 2. Multivariate models for OS in the whole population of study and in the subset of IDH1/2 mutated patients.
AML
UnivariateMultivariate
CharacteristicHR 195% CI 1p-ValueHR 195% CI 1p-Value
Age1.041.02, 1.05<0.0011.031.01, 1.050.002
WBC1.001.00, 1.000.0041.001.00, 1.000.026
WHO PS
0
I1.961.34, 2.88<0.0011.651.09, 2.490.018
II2.701.63, 4.47<0.0012.451.32, 4.530.005
III0.700.17, 2.880.620.540.07, 3.990.55
Complex karyotype vs. other
other karyotype-- --
complex karyotype2.831.79, 4.45<0.0013.171.91, 5.26<0.001
Treatment
Standard CHT
Hypomethylating2.201.56, 3.10<0.0011.070.66, 1.760.78
IDH1/2-mutated AML
UnivariateMultivariate
CharacteristicHR 195% CI 1p-ValueHR 195% CI 1p-Value 2
Age1.031.00, 1.060.019
WBC1.001.00, 1.000.0071.001.00, 1.010.005
Hb0.840.70, 1.000.049
Treatment
Standard CHT
Hypomethylating2.031.07, 3.830.031.090.48, 2.480.84
1 HR = hazard ratio; CI = confidence interval; 2 Significant p-values are indicated in bold.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Messina, M.; Piciocchi, A.; Ottone, T.; Paolini, S.; Papayannidis, C.; Lessi, F.; Fracchiolla, N.S.; Forghieri, F.; Candoni, A.; Mengarelli, A.; et al. Prevalence and Prognostic Role of IDH Mutations in Acute Myeloid Leukemia: Results of the GIMEMA AML1516 Protocol. Cancers 2022, 14, 3012. https://doi.org/10.3390/cancers14123012

AMA Style

Messina M, Piciocchi A, Ottone T, Paolini S, Papayannidis C, Lessi F, Fracchiolla NS, Forghieri F, Candoni A, Mengarelli A, et al. Prevalence and Prognostic Role of IDH Mutations in Acute Myeloid Leukemia: Results of the GIMEMA AML1516 Protocol. Cancers. 2022; 14(12):3012. https://doi.org/10.3390/cancers14123012

Chicago/Turabian Style

Messina, Monica, Alfonso Piciocchi, Tiziana Ottone, Stefania Paolini, Cristina Papayannidis, Federica Lessi, Nicola Stefano Fracchiolla, Fabio Forghieri, Anna Candoni, Andrea Mengarelli, and et al. 2022. "Prevalence and Prognostic Role of IDH Mutations in Acute Myeloid Leukemia: Results of the GIMEMA AML1516 Protocol" Cancers 14, no. 12: 3012. https://doi.org/10.3390/cancers14123012

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop