Next Article in Journal
Remdesivir Decreases Mortality in COVID-19 Patients with Active Malignancy
Next Article in Special Issue
Hsa_circ_0015278 Regulates FLT3-ITD AML Progression via Ferroptosis-Related Genes
Previous Article in Journal
Mutation Patterns in Portuguese Families with Hereditary Breast and Ovarian Cancer Syndrome
Previous Article in Special Issue
Target Therapy for Extramedullary Relapse of FLT3-ITD Acute Myeloid Leukemia: Emerging Data from the Field
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Relevance of NPM1 and FLT3 Mutations in Acute Myeloid Leukaemia, Longterm Follow-Up—A Single Center Experience

1
Department of Haematology, ASST Spedali Civili, 25100 Brescia, Italy
2
Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, 25100 Brescia, Italy
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(19), 4716; https://doi.org/10.3390/cancers14194716
Submission received: 24 August 2022 / Revised: 18 September 2022 / Accepted: 23 September 2022 / Published: 28 September 2022
(This article belongs to the Special Issue Targeting FLT3 Mutations in AML (Acute Myeloid Leukemia))

Abstract

:

Simple Summary

In acute myeloid leukemia, molecular genetics abnormalities, particularly NPM1 and FLT3 mutations, have a recognized prognostic role in the ELN risk classification and guide treatment decisions, especially since the availability of FLT3-targeted drugs. The NILG-AML 01/00 protocol uses a modified induction approach (ICE) and delivers the most active cytostatic agents at maximal doses in consolidation (a high dose of ARAC plus idarubicin) with autologous stem cell support. It calls for allogeneic transplant only in ELN high-risk patients including NPM1-wt FLT3-ITD-mutated patients. The results obtained from 171 patients showed that the median survival was not reached, and 5y-OS was 58% +/− 4. The prognostic influence of co-mutated FLT3 was overcome, and the efficacy of this treatment reduced the need for early consolidation with an allogeneic transplant in double-mutated patients, in their first complete remission. These data could represent the benchmark against which results of therapeutic programs using second-generation FLT3-targeted drugs should be compared.

Abstract

The prognosis of acute myeloid leukemia depends on genetic aberrations, particularly NPM1 and FLT3-ITD mutations. The targeted drugs’ availability has renewed interest in FLT3 mutations, but the impact of these genetic alterations using these treatments is yet to be confirmed. Our objective was to evaluate the results obtained with the intensified NILG-AML 01/00 protocol (ClinicalTrials.gov Identifier: NCT 00400673) in 171 unselected patients (median age, 54.5 years, range 15–74) carrying the FLT3 (ITD or TKD) and/or NPM1 mutations. The CR rate and 5-y survival were 88.3% and 58% +/− 4, respectively, significantly higher in the NPM1-mutated (CR 93.9%, p: 0.0001; survival 71% +/− 6, p: 0.0017, respectively). In isolated ITD patients, the CR was lower (66.7%, p: 0.0009), and the 3 years-relapse-free survival worse (24%, p: <0.0002). The presence of ITD, irrespective of the allelic ratio, or TKD mutation, did not significantly affect the survival or relapse-free survival among the NPM1-co-mutated patients. Our data indicate that a high dose of ARAC plus idarubicin consolidation exerts a strong anti-leukemic effect in NPM1-mutated patients both with the FLT3 wild-type and mutated AML, while in the NPM1 wild-type and FLT3-mutated, the therapeutic effect remains unsatisfactory. New strategies incorporating target therapy with second-generation inhibitors will improve these results and their addition to this aggressive chemotherapeutic program merits testing.

1. Introduction

Acute Myeloid Leukemia (AML) is a heterogeneous disorder characterized by a wide range of cytogenetic and molecular aberrations [1], which influence prognosis and can guide the choice of treatment [2].
Mutations of Nucleophosmin-1 (NPM1) and activating mutations in FMS-like tyrosine kinase 3 (FLT3), including internal tandem duplications (ITDs) and tyrosine kinase domain (TKD) mutations represent the most frequent genetic aberrations in AML [1,3,4,5]. While NPM1 mutation is associated with a good prognosis [6,7,8], FLT3-ITD confers a poor prognosis, even after intensive chemotherapy and/or stem cell transplant [9,10,11,12].
The concomitant presence of NPM1 mutation reduces the negative prognostic impact of FLT3-ITD, which is also modulated by the FLT3-ITD/wild-type (wt) allelic ratio [13,14,15]. Mutations within TKD are the second most common type of FLT3 mutation in AML, occurring in up to 14% of adult patients with AML [16,17]. They did not, apparently, play a negative role in the prognosis of the overall AML population; however, their impact is still debated and may depend on additional mutations, as well as on the cytogenetic background [16].
With the advent of FLT3 inhibitors, the management and prognosis of this subgroup of AML patients have changed. Multiple small molecule TKIs that target FLT3 have demonstrated clinical activity as single agents or in combination with chemotherapy, as reported in the randomized AML Trial RATIFY trial, which documented the efficacy of midostaurin in combination with intensive treatment in adult AML patients aged <60 years [18,19,20].
In this study, we analysed the very long-term impact of the presence of NPM1 and/or FLT3 mutations in a large cohort of AML patients, homogeneously treated in the pre-midostaurin era with a program different from classical 3+7, whose overall intensity was higher. It consisted of the use of idarubicin and etoposide in association with cytarabine (ICE) as the induction and of idarubicin in association with High-Dose (HD) ARAC as consolidation therapy and used allogeneic transplant (allo-SCT) only in FLT3-ITD-mutated (m) patients without the NPM1 mutation. Considering the outstanding long-term results obtained without FLT3 inhibitors, evaluation of the addition of FLT3 inhibitors also to chemotherapy programs different from classical 3+7 is needed to further improve the outcome of FLT3-mutated AML.

2. Methods

2.1. Study Design and Endpoints

This retrospective study analysed a total of 366 “de novo” AML patients, diagnosed between 2005 and 2021 and considered fit for intensive treatment managed at our institution with the NILG AML-01/00 risk-oriented protocol (ClinicalTrials.gov Identifier: NCT 00400673). Patients with acute promyelocytic leukemia were excluded.
The present analysis was focused particularly on evaluating the prognostic role of FLT3 and NPM1 mutations within an unselected AML population, homogeneously treated, in terms of response to treatment, relapse-free survival (RFS), and overall survival (OS).

2.2. Diagnostic Workup and Molecular and Cytogenetic Analysis

The diagnosis of AML was made on stained bone marrow aspiration/biopsies according to ELN criteria [2].
Cytogenetic results were described according to the International System for Human Cytogenetic Nomenclature [21]. Karyotype risk classification was performed according to MRC [22]. The presence of NPM1 and FLT3 mutations was evaluated by PCR as described by Gorello, et al. [23] and by Nakao et al. [24], respectively. The FLT3-ITD allelic ratio and minimal residual disease (MRD) evaluation in NPM1-mutated (m) patients were performed on stored samples in patients diagnosed before 2017, according to ELN recommendations [25]. As the period of observation is very long (17 years), we did not perform a comprehensive genetic study on all the patients at the beginning of the study.

2.3. Treatment Plan, Risk Assessment, and Criteria of Response

The treatment protocol (NILG AML-01/00 study; NCT00400673) is shown in Figure S1 and detailed in Appendix A.
The main differences compared to standard daunomycin+cytarabine 3+7 induction and HD-ARAC consolidation were the addition of etoposide in the first induction cycle, the use of idarubicin instead of daunomycin in induction, the mobilization of CD34+ peripheral blood stem cells (PBSCs) with a short HD-ARAC course, and the delivery of three consolidation cycles with maximal doses of ARAC, which included also the addition of idarubicin. To minimize haematological toxicity due to HD-ARAC (A20 courses), the patients received the reinfusion of a limited amount of CD34+ PBSC (1–2 × 106/Kg) after each course.
Allo-SCT was planned in patients with an FLT3-ITD-isolated mutation and adverse karyotype or late responder (CR after 2 courses) in patients with a persistent molecular disease detectable at the end of the entire treatment program and in patients with haematological or molecular relapse.
Response evaluation was performed between days 28 and 35 following the start of induction. Complete remission (CR) was defined as a reconstitution of normal marrow cellularity with <5% blast cells and more than 1 × 109/L neutrophils and at least 1 × 1011/L platelets in the peripheral blood. Refractory AML was defined as less than CR and persistence of >5% AML blasts in the bone marrow and/or in peripheral blood. Relapse was defined as the reappearance of leukemic blasts in the peripheral blood, recurrence of >5% blasts in the bone marrow, or appearance of extramedullary leukaemia.

2.4. Statistical Analysis

The overall survival (OS) was calculated from the date of diagnosis to the date of death of any cause. The relapse-free survival (RFS) was calculated from the date of CR until the date of the first haematological or molecular AML relapse or the patient’s death.
Survival was evaluated using the Kaplan–Maier method and compared using log-rank tests. The statistical significance threshold was set at a p value < 0.05.
Fisher’s exact test was applied to compare dichotomous variables.
Variables found to be significant in univariate analysis were tested in multivariate analysis by the Cox proportional hazard regression model. Analysis was performed with SPSS version 22 (SPSS Inc./IBM, Chicago, IL, USA).

3. Results

3.1. Prevalence of NPM1 and FLT3 Mutations

A total of 366 cases of AML were analysed. The patients’ characteristics are summarized in Table S1. The median age was 51 years (range 15–74) and 203 (55.4%) were males. NPM1 or FLT3 mutations or both were detected in 46.7% of patients. The frequency of the NPM1 mutation was 40.3%. The FLT3 mutations were present in 27.8% (ITD: 18.4%; TKD: 10.4%). Both mutations (NPM1 and FLT3) were present in 19.3% of patients.

3.2. Study Cohort: Characteristics of Patients

According to the NILG AML 00/01 protocol, 171 patients with the NPM1 mutation, FLT3 mutations, or both were treated, and these represented the study cohort (Figure 1). The characteristics of this cohort are shown in Table 1.
Based on the molecular analysis for the NPM1 and FLT3-ITD mutations, 27 (15.8%) patients had isolated FLT3-ITD (ITDm/NPM1wt), 38 (22.2%) concomitant FLT3-ITD, and NPM1 mutations (ITDm/NPM1m), and 73 (42.7%) had an isolated NPM1 mutation (FLT3wt/NPM1m). The FLT3-TKD mutations were present in 33 (19.3%) patients, 21 of whom were also NPM1-mutated (TKDm/NPM1m).
The median age of the population studied was 54.5 years, without differences between the molecular subgroups. There were 32 patients aged over 64 years (19%). Most of them (28/32; 87.5%) showed an NPM1 mutation (14 isolated and 14 associated with FLT3 mutations).
In eleven patients, an extramedullary disease was documented, nine of whom were NPM1-mutated. The sites were the skin (ten), central nervous system (two), and nasopharynx (one), (two patients had both CNS and skin), without differences between the molecular groups.
According to the protocol criteria, 26 patients received allo-SCT in their first CR (CR1), after a median of 5 months, of which 18 (69.2%) were FLT3-ITD-mutated (m) (Table 2).

3.3. Response and Toxicities

After the first induction cycle, CR was achieved in 151/171 patients (88.3%) and 11 further patients obtained CR after two courses (162/171, 94.7%). The sixty-day treatment-related mortality was 2/171 (1.1%), due to uncontrolled infections.
Table 2 shows the outcome of the NPM1 or FLT3 patients according to the molecular subgroups.
Overall, the CR rate after the first induction was significantly higher in NPM1m patients compared to NPM1wt [124/132 (93.9%) vs. 27/39 (69.2%): Fisher’s exact test, p: 0.0001)], whereas it was significantly lower in ITDm/NPM1wt compared to other subgroups [18/27 (66.7%) vs. 133/144 (92.4%): Fisher’s exact text p: 0.0009]. In TKDm, the CR rate was 90.9%, significantly higher in patients also with the NPM1 mutation (21/21 in NPM1m vs. 9/12 in NPM1wt: Fisher’s exact test p: 0.04).
The FLT3-ITD allelic ratio was available in 28 patients; it did not influence the CR rate (95% and 87.5% for patients with a low or high allelic, respectively; Fisher’s exact test p: 0.49).
An NPM1 MRD assessment was available in 62/151 of the CR patients (41%). After the first consolidation, 35/62 (56.4%) of the patients had achieved NPM1-MRD negativity in their peripheral blood, with no difference between NPM1m patients with or without a concomitant ITD mutation (26/46, 56.5%, and 9/16, 56.2%, respectively).
Overall, most patients were able to fully receive the pre-planned dosage of the consolidation course, and only seven patients have not completed the program. After the consolidation course with HD-ARAC, neutrophil recovery occurred a median of 10 days (6–17) after CD34+ cells reinfusion. The extra-hematological toxicities were gastrointestinal (nausea, stomatitis, and diarrhea) of grades 2 or 3 WHO, in 42% of the courses and neurological in two patients (one reversible cerebellar dysfunction with a grade 3 WHO and one reversible motor neuropathy with a grade 2 WHO). No cardiac side effects were observed. This intensive chemotherapy program was, overall, well-tolerated; the reinfusion of small amounts of autologous PBSC after each course of high-dose therapy may have potentially contributed to reducing toxicity.
In summary, the CR rate after the ICE course was high in the whole population (88.3%), significantly higher in NPM1m patients, and lower in the ITD-isolated, without differences according to the allelic ratio. The treatment program was well-tolerated.

3.4. Relapse: Relapse Rate (RR) and Relapse-Free Survival (RFS)

After a median follow-up of 63.3 months (CI 95%: 51.6–100 months), 77 patients relapsed (52.1%).
The relapse rate was lower in patients with the NPM1 mutation [54/127 (44.6%) vs. 23/35 (65.7%); p: 0.021], while it was higher among isolated ITDm patients if compared to patients with either co-mutated or isolated NPM1m (70.8%, 47.4%, 36.7%, respectively, p: 0.015).
The timing of relapse differed between FLT3-ITD patients and NPM1-mutated patients, as 41% (7/17) of the ITDm/NPM1wt patients relapsed within 6 months and 88% within 12 (15/17) months compared to only 11% (3/26) of the NPM1m/FLT3wt within 6 months and 35% (9/26) within 12 months.
NPM1 MRD positivity in the peripheral blood after consolidation impacted their relapse adversely [MRD-negative 10/35 (28.6%) and MRD-positive 16/27 (59.2%); p: 0.02], particularly in the subgroup with an NPM1m/ITDm co-mutation (relapse risk: 85.7% in the MRD-positive vs. 11% in the MRD-negative, p: 0.0087).
Among patients who achieved CR, the median RFS was 42 months. At 3-years (y), the RFS was 52% +/− 8 (SE) (Figure 2a), without significant differences according to age [3-y RFS 45% +/− 10 (SE) in the older vs. 61% +/− 4.5 (SE) in the younger; p: 0.13].
RFS was not significantly different among the NPM1-mutated, with or without concomitant FLT3-ITD [3-y survival 67% +/− 5 (SE) and 50% +/− 8 (SE), respectively, p: 0.09] (Figure 2b). The allelic burden did not impact their RFS (p: 0.5), whereas patients with isolated FLT3-ITD had a significantly lower RFS (3-y 24%, p: 0.0002) (Figure 2b).
In TKDm patients, RFS was intermediate between the NPM1m and ITDm patients, and it was not influenced by the presence of the NPM1 mutation [3-y RFS: 41% +/− 14 (SE) and 47% +/− 11 (SE) in the NPM1wt and in NPM1m patients, respectively; p: 0.9].
In summary, the relapse rate was lower in NPM1m patients, as opposed to ITDm-isolated patients, where it was higher and earlier. In NPM1m patients, the peripheral blood MRD positivity adversely impacted their relapse. The RFS was better in the NPM1m than in the FLT3-ITDm patients (p < 0.0001); a trend towards statistical significance was also observed in patients showing both mutations (FLT3-ITDm/NPM1m) (p: 0.09).

3.5. Overall Survival

In the whole cohort, sixty-nine out of one hundred and seventy-one (40%) patients died, fifty-four of disease progression, two of other causes (lung neoplasia and Amyotrophic Lateral Sclerosis), and ten from treatment-related causes (three of infection and seven from transplant-related mortality). Three patients developed a fatal secondary myelodysplastic disease, at 6, 11, and 24 months after their AML diagnosis, respectively.
The median survival was undefined and the OS at 3 and 5 years was 66% +/− 3 (SE) and 58% +/− 4 (SE), respectively. At 10 years survival, it was 51.5% +/− 4.6 (SE) (Figure 3a).
According to the molecular groups, in NPM1m patients, the median survival was undefined [OS at 3y and 5y: 72.4% +/− 4 (SE) and 63.6% +/− 5 (SE), respectively], significantly better than in the NPM1wt patients [median survival: 32 months, OS at 3y and 5y: 49.7% +/− 7(SE) and 43.6% +/− 7 (SE), respectively; p: 0.0017]; while in the FLT3m patients, the median survival was 57 months (OS at 3 and 5y: 55% +/− 5 and 49% +/− 5, respectively) significantly worse compared to the FLT3wt patients (p: 0.001).
Concerning the FLT3-ITD and NPM1 reciprocal mutation status (Figure 3b), the median survival of the patients with FLT3-ITDm/NPM1wt was 18.5 months [OS at 3y and 5y: 32% +/− 8.8 (SE) and 23% +/− 8 (SE), respectively], significantly worse compared to the other subgroups (p: <0.0001 compared to the FLT3-ITDwt/NPM1m and p: 0.0076 compared to the FLT3-ITDm/NPM1m). Particularly, the median survival in patients with both mutations (FLT3-ITDm/NPM1m) was 98 months [OS at 3y and 5y 60% +/− 8 (SE) and 56% +/− 8 (SE), respectively], worse than in the FLT3-ITDwt/NPM1m, with a borderline significance (p: 0.06) [in the ITDwt/NPM1m: the median survival was undefined, at 3y and 5y OS 80% +/− 4.8 (SE) and 71% +/− 6 (SE), respectively]. The allelic ratio of the FLT3-ITD had no impact on the survival rates (p: 0.16).
In the TKDm patients, the median survival was 93.2 months, intermediate between the NPM1m-isolated and ITDm and without differences between the other group (p: 0.075 if compared to the ITD and p: 0.3 if compared to the NPM1m).
In the univariable and multivariable analysis, patients aged < 60 years and with the presence of NPM1 mutations were correlated with significantly better survival (p 0.019 and p < 0.0001, respectively), while the presence of FLT3-ITD correlated with significantly worse survival (p < 0.0001).
Analysing the NPM1m and FLT3-ITD patients according to age, an FLT3-ITD mutation indicated poor survival in the younger patients (<65 years; p: 0.004), but it had no effect in the older patients (>66 years; p: 0.08), whereas NPM1m indicated better survival in both the older patients and in the younger patients (with any age cut off considered, <55 years, <60 years, and <65 years; p 0.000). In FLT3-ITDm/NPM1m patients, the survival was not impacted by age.
Allo-SCT was performed in 61 patients, in 26 in their CR1, according to protocol, and in 35 after haematological or molecular relapse. The median survival was 78 months, without differences between CR1 and CR2 (second CR) patients (5y-OS 55.7% +/− 10 (SE) in CR1 vs. 56.7% +/− 10 (SE) in CR2; p: 0.2). It was similar to the OS of non-transplanted patients [59.7% +/− 4 (SE) at 5y]. Overall, in our series, 44% (12/27) of all FLT3-ITD-mutated AML and only 10% (6/59) of co-mutated (NPM1m/FLT3m), underwent allo-SCT in their CR1.
In summary, the NPM1 mutation correlated with significantly better survival, while the ITD mutation with significantly worse survival, as confirmed in the multivariable analysis. In co-mutated patients (FLT3-ITDm/NPM1m), the outcome was intermediate: worse than in FLT3-ITDwt/NPM1m, with a borderline significance (p: 0.06), but significantly better than FLT3-ITDm/NPM1wt (p: 0.0076).

4. Discussion

In this monocentric, real-life study, AML patients carrying the two most frequent mutations, FLT3 and or NPM1, homogeneously treated over a period of 17 years with an aggressive idarubicin-based induction and the integration of idarubicin in the HD-ARAC consolidation, achieved an outstanding CR rate of 88.3% after one course of induction (ICE) and overall survival at 5 years of 58%. These results are similar to those obtained with the same NILG program in a multicentric setting, where in standard risk patients the CR rate after ICE was 82% and the 5y-OS 60% [26].
The choice of idarubicin in the induction instead of daunorubicin, and more importantly, its addition also to the HD-ARAC consolidation courses, may have played a role in improving the results of the treatment. It has been shown that the use of mitoxantrone and idarubicin instead of daunorubicin, during both the induction and consolidation in adult patients with AML who do not receive an allogeneic SCT, enhanced the long-term efficacy of chemotherapy, reducing their relapse risk [27]. A metanalysis of randomized trials comparing idarubicin with daunorubicin reported a survival advantage in AML patients treated with idarubicin [28]. Also, it has been shown that the combination of anthracyclines with high doses of cytarabine rather than their use as single agents or at lower doses may enhance the in vitro antileukemic T-cell immune response [29].
The optimization of antileukemic chemotherapy in our treatment program may explain the excellent outcome for NPM1-mutated patients. Indeed, the cytoplasmic delocalization of mutated NPM1 reduces the anti-apoptotic activity of the NPM1 protein, increases genomic instability, and may favour an increased sensitivity to high levels of cytotoxic agents [30]. In NPM1-mutated patients, their CR rate and survival at 3y and 5y were 93.9%, 72%, and 64%, respectively. These results compare favourably with the historical data of conventional chemotherapy regimens (CR 70% and 3y-OS 72% [13]; CR 91% and 5y-OS 49% [14]) and even with those obtained with the addition of Gemtuzumab Ozogamicin to the 3+7 regimen (CR 88.9%, 2y-OS: 68.3%, [31]) or with the use of a high-dose of cytarabine in the induction in the FLAG-Ida regimen (CR 90% and 3y-OS 63.5% [30]).
Monitoring of the NPM1-mutant measurable residual disease (MRD) allowed us to identify patients considered at an increased risk of relapse, as reported in [32,33]. In our study, NPM1 MRD positivity in the peripheral blood after consolidation impacted relapse, particularly in the subgroup of NPM1m/ITDm AML. In our series, the strategy of using MRD positivity in CR is an indication that allo-SCT may have improved the results, allowing us to better stratify patients to be addressed for transplant. Therefore, the MRD evaluation should be included in all prospective studies and guide the clinician’s decision.
The very good long-term outcome of NPM1-mutated patients was adversely impacted neither by the presence of the FLT3-ITD or TKD mutation nor by their allelic burden. In these subgroups, the RATIFY trial proved that the addition of midostaurin, a first- generation FLT3-inhibitor, to the standard 3+7 induction regimen, improved the OS compared to placebo in patients aged <60 years of age, regardless of their allelic burden (AR ≤ 0.7 or > 0.7) or the type of mutation (ITD or TKD) (the median OS of 74.7 months in patients receiving midostaurin vs. 25.6 months in patients receiving placebo plus chemotherapy [18,20]). The benefit was confirmed in the different ELN risk categories. The 5-y OS of patients treated with midostaurin were 73%, 52%, and 43% in favourable, intermediate, and adverse ELN risk, respectively [19]. In the present study, without the addition of midostaurin, the median survival and the 5y-survival of the NPM1 and FLT3 double-mutated patients were 98 months and 56% +/− 8 (SE) in the ITD-mutated and 93 months and 70.8% +/− 11 (SE) in the TKD-mutated patients. These results do not compare unfavourably, although indirectly, with those obtained in the RATIFY trial (5y-OS: in TDKm, 70.5% and in ITDm, 63% low AR and 45% high AR). Interestingly, in an Australian real-life study, which included idarubicin in the therapeutic program, the results were better when compared with those obtained in the placebo arm of the RATIFY study. These data suggest that trying to optimize the treatment backbone to which newly available targeted agents are added could be an important step to further improve treatment results in AML [34]. Recently, an international expert panel updated the ELN guidelines, classifying FLT3-mutated AML in the intermediate group, independently of NPM1 mutations’ status and allelic ratio. Our series seem to confirm an intermediate prognosis in patients with both the NPM1 and FLT3-ITD mutation, which does not imply allo-SCT as a mandatory consolidation [35].
Despite the small number of patients, the results in NPM1-wt FLT3-ITD-mutated patients were unsatisfactory, with a lower CR and higher early relapse rates. In this subset of patients, where allo-SCT in their CR1 is universally recognized as the consolidation treatment of choice, the increased intensity of our chemotherapy program before allo-SCT did not significantly improve the outcomes, as some patients (3/27: 11%) never achieved complete remission, even after two induction courses, while others (9/27: 33.3%) relapsed early and failed to undergo transplantation. Indeed, for patients harbouring the FLT3-ITD mutation, as the pivotal AML mutation, the recently approved, highly selective, second-generation FLT3 inhibitors may represent the optimal bridge to transplant, as recently demonstrated in the QuANTUM-First trial with the addition of quizartinib to a 3+7 induction treatment [36]. The new drugs would obtain a deeper remission, allowing for the gain of more time to proceed to consolidation with allo-SCT before relapse.
Although data on the prognostic significance of the FLT3 TKD mutation are still controversial, the limited efficacy of our treatment program on FLT3-mutated patients emerged also in this subgroup of patients. The CR, relapse rate, RFS, and OS were intermediate between the ITD- and NPM1-mutated. Thus, the addition of targeted agents should also be pursued in the subgroup of patients with an FLT3-TKD-isolated mutation, as recently suggested by the data shown by Voso et al. [19].
The limitations of the present study are its retrospective nature and the limited availability of data on the FLT3 allelic ratio and on NPM1-MRD, which derive from its very long observation time, which spanned periods when these techniques were not yet available.

5. Conclusions

The good outcome achieved without frontline allo-SCT in the group of NPM1-mutated patients, irrespective of their FLT3 mutational status, suggests that the use of idarubicin, both in the induction and in consolidation, and of HD-ARAC plus limited autologous stem cell support, may reduce the need for early allo-SCT consolidation in the whole group of non-high-risk patients, including NPM1-mutated/high-burden FLT3-ITD patients. In addition, in NPM1 patients, MRD monitoring PCR based during treatment may help to identify patients with a suboptimal response or in the early molecular relapse, allowing pre-emptive strategies of salvage therapy and more focused use of allo-SCT consolidation. Postponing allo-SCT after haematological or molecular relapse did not affect the transplant outcome in our series since the results were similar to patients undergoing allo-SCT in their first CR.
The results of this program could provide a benchmark for the evaluation of the additional benefit of incorporating second-generation FLT3 inhibitors in effective chemotherapy programs in patients with FLT3 and NPM1 mutations.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14194716/s1, Figure S1: Treatment Plan. Table S1: Characteristics of 366 AML patients and incidence of molecular mutations (NPM1 and FLT-3).

Author Contributions

Conceptualization: E.B. and G.R.; project administration, E.B., G.R. and A.T.; methodology: E.B.; formal analysis: E.B.; investigation: E.B., D.B., C.G. and S.A.; resources: D.B., E.C., N.B., M.O., S.B., A.P., C.B., C.P., T.Z. and S.A.; data curation M.S. and C.G.; writing—original draft preparation: E.B. and G.R.; writing—review & editing: G.R.; validation: C.C., A.T., E.B. and G.R.; visualization: C.C., E.B. and A.T.; supervision: C.C., E.B. and A.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

All procedures performed in the studies involving human participants were in accordance with the ethical standards of the institutional and or national research committee and with the Helsinki Declaration and its later amendments or comparable ethical standards. The observational and retrospective study for data collection was approved by the Ethical Committee of the Spedali Civili and the University of Brescia (NP 5590). The results of NILG AML 00/01 protocol were previously reported [26,37].

Informed Consent Statement

Written informed consent for biological sample analysis and for data collection was obtained for each patient.

Data Availability Statement

Data available on request due to restrictions, e.g., privacy or ethical.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Treatment protocol
The first induction cycle consisted of ICE (idarubicin 12 mg/sqm days 1–3; Cytarabine 100 mg/sqm/12 h days 1–7, and Etoposide 100 mg/sqm days 1–5).
The patients received IC consolidation (idarubicin 10 mg/sqm days 1–2; Cytarabine 100 mg/sqm/12 h days 1–7) if CR was achieved, or SPLIT (HD-ARAC 3 g/sqm/12 h days 2–3 and 9–10 and HD-Idarubicin 17.5 mg/sqm day1 and day 8) if the patient was refractory to ICE. The treatment plan continues with risk-adapted consolidation.
Patients at high risk (FLT3-ITD-mutated isolated, adverse karyotype or late responder (CR after 2 courses) were scheduled for allo-SCT in the first remission from any donor as soon as a CR was achieved.
All the other risks received 3 consolidation cycles with HD-ARAC plus idarubicin, defined A20, (Cytarabine 2 g/sqm/12 h for 5 days and idarubicin 8 mg/sqm days 1–2), followed by reinfusion of 1–2 × 106/Kg CD34+ peripheral blood stem cell (PBSC), at 1–2 month intervals upon hematological recovery. Patients with insufficient CD34+ PBSC yield received 2 courses of A10 (ARAC 1 g/sqm/12 h for 5 days and idarubicin 10 mg/sqm for day 1).
For PBSC mobilization, one course of intermediate dose ARAC 1 g/sqm/12 h for 4 days (A8) was given to all patients achieving CR to allow the collection of 3–6 × 106/Kg CD34+ PBSC, which were stored in three aliquots. The total dose of cytarabine planned during the consolidation phase was 68 g/sqm and 28 g/sqm in patients receiving A20 and A10 courses, respectively. The total dose of idarubicin planned to be delivered was 48 mg/sqm and 20 mg/sqm in patients treated with A20 and A10 courses, respectively.
Patients aged >70 years received as consolidation 2 or 3 courses of Ara-C at a reduced dose (total dose administered 38 g/sqm or 54 g/sqm, respectively).

References

  1. 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] [PubMed]
  2. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
  3. Schlenk, R.F.; Döhner, K.; Krauter, J.; Fröhling, S.; Corbacioglu, A.; Bullinger, L.; Habdank, M.; Späth, D.; Morgan, M.; Benner, A.; et al. German-Austrian Acute Myeloid Leukemia Study Group. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 2008, 358, 1909–1918. [Google Scholar] [CrossRef] [PubMed]
  4. Kayser, S.; Schlenk, R.F.; Londono, M.C.; Breitenbuecher, F.; Wittke, K.; Du, J.; Groner, S.; Späth, D.; Krauter, J.; Ganser, A.; et al. German-Austrian AML Study Group (AMLSG). Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood 2009, 114, 2386–2392. [Google Scholar] [CrossRef]
  5. Patel, J.P.; Gonen, M.; Figueroa, M.E.; Fernandez, H.; Sun, Z.; Racevskis, J.; Van Vlierberghe, P.; Dolgalev, I.; Thomas, S.; Aminova, O.; et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 2012, 366, 1079–1089. [Google Scholar] [CrossRef]
  6. Scholl, S.; Theuer, C.; Scheble, V.; Kunert, C.; Heller, A.; Mügge, L.O.; Fricke, H.J.; Höffken, K.; Wedding, U. Clinical impact of nucleophosmin mutations and Flt3 internal tandem duplication in patents older than 60yr with acute myeloid leukemia. Eur. J. Haematol. 2008, 80, 208–215. [Google Scholar] [CrossRef]
  7. Thiede, C.; Koch, S.; Creutzig, E.; Steudel, C.; Illmer, T.; Schaich, M.; Ehninger, G. Prevalence and prognostic impact of NPM1 mutations in 1485 adult with acute myeloid leukemia (AML). Blood 2006, 107, 4011–4020. [Google Scholar] [CrossRef]
  8. Schnittger, S.; Schoch, C.; Kern, W.; Mecucci, C.; Tschulik, C.; Martelli, M.F.; Haferlach, T.; Hiddemann, W.; Falini, B. Nucleophosfomin gene mutations are predictor of favourable prognosis in acute myelogenous leukemia with normal karyotype. Blood 2005, 106, 3733–3739. [Google Scholar] [CrossRef]
  9. Oran, B.; Cortes, J.; Beitinjaneh, A.; Chen, H.C.; de Lima, M.; Patel, K.; Ravandi, F.; Wang, X.; Brandt, M.; Andersson, B.S.; et al. Allogeneic transplantation in first remission improves outcomes irrespective of FLT3-ITD allelic ratio in FLT3-ITD positive acute myelogenous leukemia. Biol. Blood Marrow. Transplant. 2016, 22, 1218–1226. [Google Scholar] [CrossRef]
  10. Gale, R.E.; Hills, R.; Kottaridis, P.D.; Srirangan, S.; Wheatley, K.; Burnett, A.K.; Linch, D.C. No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): An analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials. Blood 2005, 106, 3658–3665. [Google Scholar] [CrossRef] [Green Version]
  11. Bornhauser, M.; Illmer, T.; Schaich, M.; Soucek, S.; Ehninger, G.; Thiede, C. AML SHG 96 study group. Improved outcome after stem-cell transplantation in FLT3/ITDpositive AML. Blood 2007, 109, 2264–2265. [Google Scholar] [CrossRef] [PubMed]
  12. Brunet, S.; Labopin, M.; Esteve, J.; Cornelissen, J.; Socié, G.; Iori, A.P.; Verdonck, L.F.; Volin, L.; Gratwohl, A.-; Sierra, J.; et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: A retrospective analysis. J. Clin. Oncol. 2012, 30, 735–741. [Google Scholar] [CrossRef] [PubMed]
  13. Gale, R.E.; Green, C.; Allen, C.; Mead, A.J.; Burnett, A.K.; Hills, R.K.; Linch, D.C. Medical Research Council Adult Leukaemia Working Party. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008, 111, 2776–2784. [Google Scholar] [CrossRef] [PubMed]
  14. Schnittger, S.; Bacher, U.; Kern, W.; Alpermann, T.; Haferlach, C.; Haferlach, T. Prognostic impact of FLT3-ITD load in NPM1 mutated acute myeloid leukemia. Leukemia 2011, 25, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
  15. Pratcorona, M.; Brunet, S.; Nomdedéu, J.; Ribera, J.M.; Tormo, M.; Duarte, R.; Escoda, L.; Guàrdia, R.; de Llano, M.P.Q.; Salamero, O.; et al. Grupo Cooperativo Para el Estudio y Tratamiento de las Leucemias Agudas Mieloblásticas. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: Relevance to post-remission therapy. Blood 2013, 121, 2734–2738. [Google Scholar]
  16. Bacher, U.; Haferlach, C.; Kern, W.; Haferlach, T.; Schnittger, S. Prognostic relevance of FLT3–TKD mutations in AML: The combination matters-an analysis of 3082 patients. Blood 2008, 111, 2527–2537. [Google Scholar] [CrossRef]
  17. Thiede, C.; Steudel, C.; Mohr, B.; Schaich, M.; Schäkel, U.; Platzbecker, U.; Wermke, M.; Bornhäuser, M.; Ritter, M.; Neubauer, A.; et al. Analysis of FLT3- activating mutations in 979 patients with acute myelogenous leukemia: Association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002, 99, 4326–4335. [Google Scholar] [CrossRef]
  18. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  19. Voso, M.T.; Larson, R.A.; Jones, D.; Marcucci, G.; Prior, T.; Krauter, J.; Heuser, M.; Lavorgna, S.; Nomdede, J.; Geyer, S.M.; et al. Midostaurin in patients with acute myelod leukemia and FLT3-TKD mutations: A sub analysis from the Ratify trial. Blood Adv. 2020, 13, 4945–4954. [Google Scholar] [CrossRef]
  20. Doöhner, K.; Thiede, C.; Jahn, N.; Panina, E.; Gambietz, A.; Larson, R.A.; Prior, T.W.; Marcucci, G.-; Jones, D.; Krauter, J.; et al. Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood 2020, 135, 371–380. [Google Scholar] [CrossRef]
  21. International System for Cytogenetic Nomenclature. Guidelines for Cancer Cytogenetics. Supplement to: An International System for Human Cytogenetic Nomenclature; Felix Mitelmann: Memphis, TN, USA, 1995. [Google Scholar]
  22. Grimwade, D.; Hills, R.K.; Moorman, A.V.; Walker, H.; Chatters, S.; Goldstone, A.H.; Wheatley, K.; Harrison, C.J.; Burnett, A.K. National Cancer Research Institute Adult Leukaemia Working Group. Refinement of cytogenetic classification in acute myeloid leukemia: Determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010, 116, 354–365. [Google Scholar] [PubMed] [Green Version]
  23. Gorello, P.; Cazzaniga, G.; Alberti, F.; Dell’Oro, M.G.; Gottardi, E.; Specchia, G.; Roti, G.; Rosati, R.; Martelli, M.F.; Diverio, D.; et al. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying Nucleophosmin (NPM1) gene mutations. Leukemia 2006, 20, 1103–1108. [Google Scholar] [CrossRef] [PubMed]
  24. Nakao, M.; Yokota, S.; Iwai, T.; Kaneko, H.; Horiike, S.; Kashima, K.; Sonoda, Y.; Fujimoto, T.; Misawa, S. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 1996, 10, 1911–1918. [Google Scholar] [PubMed]
  25. Schuurhuis, J.G.; Heuser, M.; Freeman, S.; Béné, M.C.; Buccisano, F.; Cloos, J.; Grimwade, D.; Haferlach, T.; Hills, R.K.; Hourigan, C.S.; et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2018, 131, 1275–1291. [Google Scholar] [CrossRef]
  26. Bassan, R.; Intermesoli, T.; Masciulli, A.; Pavoni, C.; Boschini, C.; Gianfaldoni, G.; Marmont, F.; Cavattoni, I.; Mattei, D.; Terruzzi, E.; et al. Randomized trial comparing standard vs sequential high-dose chemotherapy for inducing early CR in adult AML. Blood Adv. 2019, 3, 1103–1117. [Google Scholar] [CrossRef]
  27. Mandelli, F.; Vignetti, M.; Suciu, S.; Stasi, R.; Petti, M.C.; Meloni, G.; Muus, P.; Marmont, F.; Marie, J.P.; Labar, B.; et al. Daunorubicin versus mitoxantrone versus idarubicin as induction and consolidation chemotherapy for adults with acute myeloid leukemia: The EORTC and GIMEMA Groups Study AML-10. J. Clin. Oncol. 2009, 27, 5397–5403. [Google Scholar] [CrossRef]
  28. The AML Collaborative Group. A systematic collaborative overview of randomized trials comparing idarubicin with daunorubicin (or other anthracyclines) as induction therapy for acute myeloid leukaemia. Br. J. Haematol. 1998, 103, 100–109. [Google Scholar] [CrossRef]
  29. Aurelius, J.; Möllgård, L.; Kiffin, R.; Sander, E.F.; Nilsson, S.; Thorén, F.B.; Hellstrand, K.; Martner, A. Anthracycline-based consolidation may determine outcome of post-consolidation immunotherapy in AML. Leuk. Lymphoma 2019, 60, 2771–2778. [Google Scholar] [CrossRef]
  30. Minetto, P.; Candoni, A.; Guolo, F.; Clavio, M.; Zannier, M.E.; Miglino, M.; Dubbini, M.V.; Carminati, E.; Sicuranza, A.; Ciofini, S.; et al. Fludarabine, High-Dose Cytarabine and Idarubicin-Based Induction May Overcome the Negative Prognostic Impact of FLT3-ITD in NPM1 Mutated AML, Irrespectively of FLT3-ITD Allelic Burden. Cancers 2020, 24, 34. [Google Scholar] [CrossRef]
  31. Castaigne, S.; Pautas, C.; Terré, C.; Raffoux, E.; Bordessoule, D.; Bastie, J.-N.; Legrand, O.; Thomas, X.; Turlure, P.; Reman, O.; et al. Acute Leukemia French Association. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): A randomised, open-label, phase 3 study. Lancet 2012, 379, 1508–1516. [Google Scholar] [CrossRef]
  32. Ivey, A.; Hills, R.K.; Simpson, M.A.; Grech, G.A.; Patel, G.Y.; Bhudia, N.; Farah, H.; Mason, J.; Wall, K.; Akiki, S.; et al. UK National Cancer Research Institute AML Working Group. Assessment of minimal residual disease in standard-risk AML. N. Engl. J. Med. 2016, 374, 422–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Balsat, M.; Renneville, A.; Thomas, X.; de Botton, S.; Caillot, D.; Marceau, A.; Lemasle, E.; Marolleau, J.P.; Nibourel, O.; Berthon, C.; et al. Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: A study by the Acute Leukemia French Association Group. J. Clin. Oncol. 2017, 35, 185–193. [Google Scholar] [CrossRef] [PubMed]
  34. Chua, C.; Grigg, A.; Singh, J.; Droogleever, M.P.; Zhang, L.; Lim, A.; Fong, C.Y.; Ting, S.B.; Schwarer, A.; Tiong, I.S.; et al. Treatment practice and outcome in FLT3-mutant acute myeloid leukemia in the re-midostaurin era: A real world experience from Australian tertiary hospitals. Leuk. Lymphoma 2020, 61, 848–854. [Google Scholar] [CrossRef] [PubMed]
  35. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and Management of AML in Adults: 2022 ELN Recommendations from an International Expert Panel. Blood 2022, online ahead of print. [CrossRef]
  36. Erba, H.; Montesinos, P.; Vrhovac, R.; Patkowska, E.; Kim, H.J.; Zak, P.; Wang, P.N.; Mitov, T.; Hanyok, J.; Liu, L.; et al. Quizartinib prolunged survival vs placebo plus intensive induction and consolidation therapy followed by single agent continuation in patients aged 18–75 years with newly diagnosed FLT3-ITD+ AML. In Proceedings of the 27th Congress of European Hematology Association (EHA 2022), Wien, Austria, 9–12 June 2022. [Google Scholar]
  37. Borlenghi, E.; Cattaneo, C.; Cerqui, E.; Archetti, S.; Bertoli, D.; Bellotti, D.; Gramegna, D.; Soverini, G.; Oberti, M.; Schieppati, F.; et al. Postremission therapy with repeated courses of high-dose cytarabine, idarubicin, and limited autologous stem cell support achieves a very good long-term outcome in European leukemia net favorable and intermediate-risk acute myeloid leukemia. Hematol. Oncol. 2020, 38, 754–762. [Google Scholar] [CrossRef]
Figure 1. Study Cohort.
Figure 1. Study Cohort.
Cancers 14 04716 g001
Figure 2. Relapse-Free Survival (RFS): (a) in the whole study cohort and according to age. (b) According to molecular data (NPM1/FLT3-ITD): NPM1m/ITDwt vs. NPM1wt/ITDm, vs. NPM1m/ITDm.
Figure 2. Relapse-Free Survival (RFS): (a) in the whole study cohort and according to age. (b) According to molecular data (NPM1/FLT3-ITD): NPM1m/ITDwt vs. NPM1wt/ITDm, vs. NPM1m/ITDm.
Cancers 14 04716 g002
Figure 3. Overall Survival (OS): (a) in the whole study cohort; (b) According to molecular data: NPM1/FLT3-ITD status.
Figure 3. Overall Survival (OS): (a) in the whole study cohort; (b) According to molecular data: NPM1/FLT3-ITD status.
Cancers 14 04716 g003
Table 1. Characteristics of 171 NPM1or FLT3-mutated patients (study cohort), divided according to molecular data.
Table 1. Characteristics of 171 NPM1or FLT3-mutated patients (study cohort), divided according to molecular data.
ITDm/NPM1wt
n 27
ITDm/NPM1m
n 38
TKDm/NPM1wt
n 12
TKDm/NPM1m
n 21
FLT3wt/NPM1m
n 73
Total
n 171
Median age (years)55555454.55554.5
Male n (%)15 (55)16 (42.1)7 (58)13 (61.9)35 (47.9)86 (50.3)
Extramedullary disease n (%)01 (2.6)2 (16)2 (9.5)6 (8.2)11 (6.4)
Favorable K n (%)1 (3.7)-2 (16.7)--3 (1.8)
Intermediate K n (%)23 (85.2)38 (100)8 (66.6)21 (100)71 (97.3)161 (94.2)
Adverse K n (%)3 (11.1)-2 (16.7)-2 (2.7)7 (4)
Abbreviations: n: number, K: karyotype; m: mutated, and wt: wild-type.
Table 2. Complete Response, duration of CR, relapse rate, RFS, and OS, according to molecular groups.
Table 2. Complete Response, duration of CR, relapse rate, RFS, and OS, according to molecular groups.
ITDm/NPM1wt
n (%) 27 (15.8)
ITDm/NPM1m
n (%) 38 (22.2)
TKDm/NPM1wt
n (%) 12 (7)
TKDm/NPM1m
n (%) 21 (12.3)
FLT3wt/NPM1m
n (%) 73 (42.7)
Total
n 171
CR rate after ICE18 (66.7)37 (97.3)9 (75)21 (100)66 (90)151 (88.3)
CR rate after 2 courses24 (88.9)38 (100)11 (92)-68 (93.2)162 (94.7)
Relapse rate17 (70.8)18 (47)6 (54.5)11 (52.4)25 (36.7)77 (52.1)
Duration CR
<6 months
6 (25)8 (21)1 (9)2 (9.5)2 (2.9)19/162 (11.7)
Median duration CR (months)113515.919.74832.5
Median RFS (months)10353233undef42
Median OS (months)18.4698undef93undefundef
5y-OS (% +/− SE)23 +/− 856 +/− 854 +/− 1570.8 +/− 1171 +/− 658 +/− 4
Allo-SCT
in CR1
during disease course
15
12 (80)
3
11
6 (67)
5
7
4 (57)
3
6
-
6
22
4(18)
18
61
26 (42.6)
35 (57)
Median RFS (months)
censored at allo-SCT
10undef11.548undefundef
Median OS (months)
censored at allo-SCT
10undef3193undefundef
5y-OS (% +/− SE)
censored at allo-SCT
16.7 +/− 1157 +/− 940 +/− 2168 +/− 1375 +/− 760 +/− 4
Abbreviations: m: mutated, wt: wild-type, Allo-SCT: allogenic stem cell transplantation, CR: complete remission, CR1: first complete remission, SE: standard Error, OS: overall survival, y: years, and RFS: relapse-free survival.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Borlenghi, E.; Cattaneo, C.; Bertoli, D.; Cerqui, E.; Archetti, S.; Passi, A.; Oberti, M.; Zollner, T.; Giupponi, C.; Pagani, C.; et al. Prognostic Relevance of NPM1 and FLT3 Mutations in Acute Myeloid Leukaemia, Longterm Follow-Up—A Single Center Experience. Cancers 2022, 14, 4716. https://doi.org/10.3390/cancers14194716

AMA Style

Borlenghi E, Cattaneo C, Bertoli D, Cerqui E, Archetti S, Passi A, Oberti M, Zollner T, Giupponi C, Pagani C, et al. Prognostic Relevance of NPM1 and FLT3 Mutations in Acute Myeloid Leukaemia, Longterm Follow-Up—A Single Center Experience. Cancers. 2022; 14(19):4716. https://doi.org/10.3390/cancers14194716

Chicago/Turabian Style

Borlenghi, Erika, Chiara Cattaneo, Diego Bertoli, Elisa Cerqui, Silvana Archetti, Angela Passi, Margherita Oberti, Tatiana Zollner, Carlotta Giupponi, Chiara Pagani, and et al. 2022. "Prognostic Relevance of NPM1 and FLT3 Mutations in Acute Myeloid Leukaemia, Longterm Follow-Up—A Single Center Experience" Cancers 14, no. 19: 4716. https://doi.org/10.3390/cancers14194716

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