Skip to main content
Transfusion Medicine and Hemotherapy logoLink to Transfusion Medicine and Hemotherapy
. 2022 Nov 4;50(3):234–244. doi: 10.1159/000526174

The Prognostic Value of TP53 Mutations in Adult Acute Myeloid Leukemia: A Meta-Analysis

Guoxiang Qin a,*, Xueling Han b
PMCID: PMC10331159  PMID: 37435002

Abstract

Objective

Mutations of the tumor protein p53 (TP53) gene were considered to be associated with an unfavorable prognosis in acute myeloid leukemia (AML). This meta-analysis aimed to systematically elucidate the prognostic value of TP53 mutation in adult patients with AML.

Method

A comprehensive literature search was conducted for eligible studies published before August 2021. The primary endpoint was overall survival (OS). Pooled hazard ratios (HRs) and their 95% confidence intervals (CIs) were calculated for prognostic parameters. Subgroup analyses based on intensive treatment were performed.

Results

Thirty-two studies with 7,062 patients were included. As compared to wild-type carriers, AML patients with TP53 mutations had significantly shorter OS (HR: 2.40, 95% CI: 2.16–2.67, I<sup>2</sup>: 46.6%). Similar results were found in DFS (HR: 2.87, 95% CI: 1.88–4.38), EFS (HR: 2.56, 95% CI: 1.97–3.31), and RFS (HR: 2.40, 95% CI: 1.79–3.22). Mutant TP53 predicted inferior OS (HR: 2.77, 95% CI: 2.41–3.18) in the intensively treated AML subgroup, compared with the non-intensively treated group (HR: 1.89, 95% CI: 1.58–2.26). Among intensively-treated AML patients, the age of 65 did not affect the prognostic value of TP53 mutations. Besides, TP53 mutation was also strongly associated with an elevated risk of adverse cytogenetics, which conferred a dismal OS in AML patients (HR: 2.03, 95% CI: 1.74–2.37).

Conclusion

TP53 mutation exhibits a promising potential for discriminating AML patients with a worse prognosis, thus being capable of serving as a novel tool for prognostication and therapeutic decision-making in the management of AML.

Key Words: Acute myeloid leukemia, TP53 mutations, Prognosis, Meta-analysis

Introduction

Acute myeloid leukemia (AML) is a genetically heterogeneous disorder characterized by the somatic acquisition of genetic and epigenetic alterations in hematopoietic progenitor cells that perturb normal self-renewal, proliferation, and differentiation mechanisms [1]. Of acute leukemia in adults, AML is the most common, with an incidence exceeding 20,000 cases per year in the USA alone [2]. The advent of sequencing techniques such as next-generation sequencing has allowed new insights into the molecular basis of AML, and the molecular analysis of gene alterations has been incorporated into the risk stratification and prognostication for patients with AML in clinical practice [3].

The tumor suppressor p53 (TP53) gene, located on chromosome 17p13.1, is an established cellular gatekeeper of proliferation/differentiation in response to aberrant oncogene expression. TP53 inactivation induced by gene-mutation or deletion favors the effect of the oncogene, thus contributing to the uncontrolled proliferation of cancer cells [4]. Somatic mutations of TP53 gene have been detected in more than 50% of all human tumors and represent an important prognostic and predictive marker in cancer [5]. Of note, these mutations are less frequent in hematological malignancies (10% overall); however, similar to other cancer types, they are found to show a negative impact on survival [6]. In patients with myelodysplastic syndromes, TP53 mutations (5–10% of cases) are associated with high-risk disease, secondary AML progression, and dismal outcomes [7]. Similarly, TP53 mutations are observed in proximately 5–10% of adult de novo AML cases [8], with high frequency (∼30%) in therapy-related AML. TP53 mutations are also highly associated with increased ages and complex karyotypes (CK) (52–70% of cases) [9, 10, 11].

Quite a few reports have supported the adverse prognostic impact of TP53 mutations in the setting of AML, where patients with mutant TP53 could exhibit a significantly shorter overall survival (OS) than those with wild-type TP53. The 2017 European Leukemia Net guidelines were updated to recommend upfront screening for mutations in TP53 at diagnosis for risk stratification given its dismal prognostic implications [3]. Given the relatively low frequency of TP53 mutation in this population, additional information is encouraged for establishing a prognostic profile of TP53 mutations based on previously published data in a systematic manner. Hence, this present meta-analysis was conducted to systematically validate the prognostic value of TP53 mutations in adult patients with AML through comparing the prognostic parameters between patients with mutant and wild-type TP53 to strengthen the impact of this gene mutation in clinics, and the relations between TP53 mutation status and cytogenetics would also be assessed.

Materials and Methods

Data Sources and Search Strategy

The present study was conducted following the guidelines of Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statements [12]. A database search of PubMed, Cochrane library, ScienceDirect, Embase, and Web of Science was conducted to find eligible studies published before August 2021. The following search terms with their synonyms in various combinations were used in the searching process: “Myeloblastic,” “Leukemia,” “TP53,” and “prognosis.” Reference lists of potentially related papers were cross-checked to identify additional records. After removing duplicate items with the help of Endnote X9 software, two reviewers independently screened the initial search results based on title and abstract and then, selected candidate studies for further full-text evaluation. Any difference was resolved by consensus.

Eligibility Criteria

Studies were considered eligible if satisfied the following requirements: (1) prospective or retrospective clinical studies assessing TP53 mutation status in adult patients with AML; (2) offered enough prognostic information of TP53 mutation versus TP53 wild-type subgroups, with calculation of hazard ratio (HR) and its corresponding 95% confidence intervals (CI) or with adequate data for their estimation; and (3) original articles written in English. Those articles were excluded if they had a mixed cohort with <50% of the cohort having AML without a separate analysis of AML cases. In event of overlapped samples, the study with the most sufficient data was included. Nonrelated papers, animal studies, reviews, case reports, letters, editorials, conference abstracts, duplicate publications, or studies with insufficient data were rejected from the meta-analysis.

Data Extraction

For each eligible study, we captured the following information: the first author, year of publication, country, study design, sample size, patient age, type of AML, median follow-up, therapeutic strategies, TP53 mutation status (measuring techniques and incidence of mutations), and outcomes of interest including HR for prognostic information with its 95% CI under Cox proportional hazards regression models, such as OS (the time interval from the date of diagnosis to the date of death), RFS (the time interval from the date of complete response to the date of relapse), event-free survival (EFS, defined as the time from diagnosis of AML to treatment failure relapse, death, or last follow-up), and disease-free survival (DFS, the duration from complete response until relapse or death or censorship at the last follow-up). All data were checked to ensure the study's accuracy and any discrepancies were finally resolved by consensus.

Quality Assessment

Two reviewers independently assessed the quality of each eligible study with NOS (Newcastle-Ottawa Quality Assessment Scale) [13] and reached a consensus by discussion in cases of differences. NOS covers three domains: selection (four items), comparability (two items), and outcome (three items). The overall NOS score ranges from zero to nine points. Studies with more than six points were considered to be of high quality.

Statistical Analysis

Pooled HRs and their 95% CIs for OS (the primary objective) comparing TP53 mutation and TP53 wild type were used to assess the prognostic value of TP53 mutations in patients with AML. Other prognostic parameters, such as RFS, EFS, and DFS, were secondary outcomes. The HR >1 with 95% CI exceeding 1 represents a significantly higher risk of death in favor of AML patients with TP53 mutations, compared to those with wild-type TP53. Additionally, odds ratio (OR) and 95% CI were used to reveal relations between TP53 and patients with adverse cytogenetics, with an OR and 95% CI >1, suggesting that patients with mutant TP53 were at a higher risk of adverse cytogenetics with statistical significance. Heterogeneity across studies was measured by the inconsistency index (I2), and an I2 >50% was indicative of significant heterogeneity [14]. We used a random-effects model for data pooling when I2 >50% was present; otherwise, the fixed-effect model was used [14]. Sensitivity analysis was conducted to interpret significant heterogeneity by sequentially omitting 1 study each time. Besides, subgroup analyses based on the treatment strategies (intensively treated vs. nonintensively treated) were performed for the primary outcome (OS) to investigate the impact of intensive treatment on the overall result. Studies in which AML patients treated with intensive treatment accounted for more than half (>50%) of the total cohort were grouped into the “intensively treated” subgroup. Further analysis based on median age (≥65 years old or not) within each subgroup was done to investigate the impact of age. A test of interaction was used to explore if there was a statistical difference (p < 0.05) between subgroups [15]. The potential publication bias of the primary outcome was evaluated through Egger's test, with a p > 0.05, indicating the absence of potential bias [14]. A trim-and-filled analysis was performed if there was a p < 0.05. All the data analyses were conducted by STATA version 12.0 (College Station, TX, USA).

Results

Literature Search

In total, 7,314 relevant studies were identified from electronic databases and cross-checked references. After excluding duplicated records, we screened the results based on title and abstract and selected 179 studies for further full-text evaluation. Finally, 32 studies [16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47] were included in our meta-analysis. Figure 1 shows the flowchart of study selection.

Fig. 1.

Fig. 1

Flow diagram of study selection.

Study Characteristics and Quality Assessment

Published between 2002 and 2021, 31 studies including 7,062 subjects from eight countries were included in our study. Fourteen articles (43.8%) [16, 20, 21, 22, 23, 24, 25, 29, 32, 35, 36, 37, 38, 42] were prospective studies and the remaining were in a retrospective design. The median age of enrolled AML cases in 14 studies was ≥65 years old. More detailed information is presented in Table 1. The NOS was used for quality assessment and results are summarized in Table S1 (see www.karger.com/doi/10.1159/000526174 for all online suppl. material). The average NOS score across these studies was 6.1 (Table 1), suggesting that the methodological quality was moderately high and overall acceptable.

Table 1.

Study characteristic of the included studies

First author Year N Median age M, % Type of AML Median OS/FU NOS
Bories 2020 279 76 55.6 De novo AML, sAML, tAML FU 66.1 months 7
Chen 2021 204 54.4 50.5 De novo AML / 5
Daher-Reyes 2021 435 59 52.0 De novo AML, sAML, tAML FU 475 days 8
Falk 2015 189 64 50.3 De novo AML / 4
Fenwarth 2019 187 (18–59) 52.0 De novo AML / 7
Fernandez 2017 143 60 50.3 AML-MRC, AML-NOS, AML-RGA, tAML / 5
Fujiwara 2019 98 71 59.2 De novo AML / 6
Grossmann 2021 1,000 66.8 / No mentioned FU 23.7 months 7
Hong 2020 125 66 53.6 AML-RGA, AML-MRC, AML-NOS, tAML 16 months/12 months 7
Hou 2015 500 51 57.0 De novo AML FU 55 months 8
Inokuchi 2002 170 57 62.9 De novo AML FU 20.6 months 6
Kihara 2014 197 <60 (89%) / De novo AML FU 32.5 months 6
Middeke 2016 97 51 / De novo AML, sAML FU 67 months 8
Milosevic 2012 86 68 57.0 sAML / 6
Montalban-Bravo 2020 415 70 64.0 AML-MRC 10.5 months/28.3 months 5
Morsia 2020 44 73.5 61.4 De novo AML, sAML, tAML 11 months/7 months 7
Mrózek 2019 136 53, 59 66.9 De novo AML FU 81.6 months 7
Najima 2021 120 51 59.2 De novo AML, sAML, tAML FU 60 months 6
Ni 2020 92 67 56.5 De novo AML (Elderly) OS 9.7 months 5
116 43 56.0 De novo AML (Younger <60 years) /
Ohgami 2015 93 55 48.4 AML-MRC, AML-RGA OS 221 days 5
Ok 2015 108 68 57.4 tMDS/AML 8 months/12.9 months 7
Österroos 2020 182 74 50.5 De novo AML, tAML 8.2 months/57 months 7
Rucker 2012 155 59 51.6 De novo AML, sAML, tAML FU 36.6 months 8
Stahl 2018 655 65 58.2 De novo AML, sAML OS 6.7 months 6
Stahl 2021 86 67 63.0 De novo AML, sAML, tAML 6.1m/12 months 6
Terada 2018 412 55.1 57.5 De novo AML / 4
Thol 2018 96 50.1, 51.7 41.6 De novo AML, sAML FU 74.4 months 7
Venton 2018 73 70 63.0 Post-MDN sAML OS 4.7 months 5
Wang 2021 347 57 56.5 De novo AML / 4
Wu 2021 81 54 54.0 No mentioned OS 4 months 5
Yonada 2016 77 69, 70 62.3 AML-MRC, AML-RGA, tAML, AML-NOS FU 48 months 6
Yu 2019 64 27.5 60.9 De novo AML FU 23.5 months 6

AML, acute myeloid leukemia; AML-MRC, AML with myelodysplasia-related changes; AML-NOS, AML not otherwise specified; AML-RGA, AML with recurrent genetic abnormalities; FU, follow-up; M, male; MDS/AML, acute myeloid leukemia evolving from MDS; N, number; NOS, Newcastle-Ottawa Scale; OS, overall survival; Post-MDN sAML, acute myeloid leukemias secondary to myeloproliferative neoplasms (MPN); sAML, secondary acute myeloid leukemia; tAML, therapy-related acute myeloid leukemia; t-MDS, therapy-related myelodysplastic syndrome.

Prognostic Value of TP53 Mutations

Table 2 summarizes the TP53 mutational status across included studies. The frequency of TP53 mutation ranged from 2.6% (Ni et al. [34], younger AML cohorts) to 66.5% (Rucker et al. [38], AML cases with CK). Based on 11 studies with available data [16, 19, 23, 24, 25, 29, 30, 31, 32, 36, 38, 43], the median OS of AML patients with mutant TP53 ranged from 1.8 months to 10 months, while that of patients with wild-type TP53 ranged from 5.6 months to 35.6 months. The meta-analysis of data from 30 included studies (969 AML cases with TP53 mutation and 4,828 with TP53 wild-type) showed that TP53 mutation was associated with a statistically significant negative effect on the OS of AML patients with an HR of 2.40 (95% CI: 2.16–2.67, p < 0.05, Fig. 2) under a fixed-effect model due to nonsignificant heterogeneity (I2: 46.6%). As shown in Figure 3, patients with mutant TP53 also exhibited a worse prognosis than those with TP53 wild type in terms of RFS (HR: 2.40, 95% CI: 1.79–3.22), EFS (HR: 2.56, 95% CI: 1.97–3.31), and DFS (HR: 2.87, 95% CI: 1.88–4.38). A fixed-effect model was used owing to low heterogeneity between studies (I2 <50%, Fig. 3).

Table 2.

Summary of TP53 status among included studies

Study ID Applying NGS? TP53 mutation
TP53 wild type
Frequency of TP53 mutation (%)
n median OS n median OS
Bories et al. [16] 2020 No 55 7.9 months 169 12.6 months 24.6
Chen et al. [17] 2021 Yes 15 / 189 / 7.4
Daher-Reyes et al. [18] 2021 Yes 25 / 153 / 14.0
Falk et al. [19] 2015 No 19 2 months 170 16 months 10.1
Fenwarth et al. [20] 2019 No 17 / 170 / 9.1
Fernandez-Pol et al. [21] 2017 No 12 / 55 / 17.9
Fujiwara et al. [22] 2019 No 12 / 86 / 12.2
Grossmann et al. [23] 2012 Yes 115 4.6 months 885 35.6 months 11.5
Hong et al. [24] 2020 Yes 15 10 months 110 18 months 12.0
Hou et al. [25] 2015 No 35 5 months 465 35 months 7.0
Inokuchi et al. [26] 2002 No 20 / 150 / 11.8
Kihara et al. [27] 2014 No 7 / 190 / 3.6
Middeke et al. [28] 2016 Yes 40 10% (3 years OS) 57 33.1% (3 years OS) 41.2
Milosevic et al. [29] 2012 No 14 1.8 months 59 5.6 months 19.2
Montalban-Bravo et al. [30] 2020 Yes 152 6.6 months 263 11.1 months 36.6
Morsia et al. [31] 2020 Yes 9 8 months 35 14 months 20.5
Mrózek et al. [32] 2019 Yes 69 4.8 months 67 7.2 months 50.7
Najima et al. [33] 2021 Yes 21 9.5% (2 years OS) 99 30.1% (2 years OS) 17.5
Ni et al. [34] 2020 Yes 13 / 79 / 14.1
Yes 3 / 113 / 2.6
Ohgami et al. [35] 2015 Yes 17 / 76 / 18.3
Ok et al. [36] 2015 No 40 6.1 months 68 14.1 months 37.0
Österroos et al. [37] 2020 No 25 / 157 / 13.7
Rucker et al. [38] 2012 No 103 4.14 months 52 10.97 months 66.5
Stahl et al. [39] 2018 No 7 / 86 / 7.5
Stahl et al. [40] 2021 No 11 / 70 / 13.6
Terada et al. [41] 2018 No 31 14.5% (5 years OS) 381 33.7% (5 years OS) 7.5
Thol et al. [42] 2018 Yes 12 / 84 / 12.5
Venton et al. [43] 2018 Yes 20 4.4 months 36 6.5 months 35.7
Wang et al. [44] 2021 No 26 / 321 / 7.5
Wu et al. [45] 2021 Yes 10 / 71 / 12.3
Yonada et al. [46] 2016 No 12 / 65 / 15.6
Yu et al. [47] 2019 Yes 5 / 59 / 7.8

NGS, next-generation sequencing; OS, overall survival.

Fig. 2.

Fig. 2

Forest plot of pooled HRs for OS comparing AML patients with TP53 mutation versus TP53 wild type. The HR with 95% CI exceeding 1 suggested a significantly higher risk of death in AML patients with TP53 mutation. HR, hazard ratio; CI, confidence interval; OS, overall survival.

Fig. 3.

Fig. 3

Forest plot of pooled HRs for DFS, EFS, and RFS comparing AML patients with TP53 mutation versus TP53 wild type. HR, hazard ratio; CI, confidence interval; DFS, disease-free survival; EFS, event-free survival; RFS, relapse-free survival.

Egger's test revealed that potential bias might be potentially present in the analysis of OS (p = 0.02) and a trim-and-filled was used. Ten missing studies were filled by the trim-and-fill method, but the pooled estimate did not significantly change under the fixed-effect model (before: LogHR 0.874, 95% CI: 0.768–0.981; after filled analysis: LogHR 0.748, 95% CI: 0.648–0.847), indicating that the result remained robust albeit with the presence of potential bias.

Additional Subgroup Analysis

As displayed in Figure 4a, subgroup analyses stratified by intensive treatment suggested that TP53 mutation was associated with a more negative OS in intensively treated patients (HR: 2.73, 95% CI: 2.39–3.13), compared with that in patients treated by nonintensive therapy (HR: 1.89, 95% CI: 1.58–2.26) (test of interaction: p = 0.001). We further analyzed each subgroup based on the median age (≥65 years old or <65 years old) of enrolled subjects. In the “intensively treated” group, no significant difference was found between median age <65 and ≥65 years groups (test of interaction: p = 0.83), indicating that intensively treated AML cases with mutant TP53 might demonstrate a worse OS regardless of age (Fig. 4b). Of note, the median age of the “nonintensively treated” group was more than 65; therefore, among AML subjects with median age ≥65, the nonintensive treatment showed a better OS than the intensive treatment (HR: 1.89 vs. 2.98, test of interaction: p < 0.05; Fig. 4).

Fig. 4.

Fig. 4

Subgroup analysis of pooled HRs for OS according to intensive treatment in total AML patients (a) and further analysis of pooled HRs for OS according to the median age of 65 years old in intensively treated AML patients (b).

Relations between TP53 Mutations and Cytogenetics

Cytogenetics information was frequently used for AML risk stratification, and we conducted additional analyses to investigate the relation between adverse cytogenetics and TP53 mutations. Pooled by 12 studies, results suggested that there was a significant difference in OS between patients with adverse cytogenetics and patients with favorable or intermediate cytogenetics (HR: 2.03, 95% CI: 1.74–2.37, I2: 32.4%). Furthermore, compared to patients with TP53 wild type, patients harboring TP53 mutation were at an elevated risk of adverse cytogenetics (OR: 23.22, 95% CI: 13.84–38.95, I2: 18.6%). It is indicated that TP53 mutation was significantly correlated with adverse cytogenetics, which might predict a worse OS. Forest plot is shown in Figure 5.

Fig. 5.

Fig. 5

Forest plot of pooled HRs for OS comparing AML patients with adverse cytogenetics versus nonadverse cytogenetics (a) and the relation between TP53 mutation and adverse cytogenetics (b). HR, hazard ratio; CI, confidence interval; OR, odds ratio; OS, overall survival.

Discussion

In this meta-analysis based on 32 eligible studies involving 7,062 adult AML cases, results demonstrated that patients with TP53 mutations exhibited a significantly worse survival than those with TP53 wild type, given the significant HRs greater than 1 for OS, EFS, DFS, or RFS, which validated the prognostic potential of TP53 mutation in patients with AML in a comprehensive manner.

AML is characterized by a spectrum of gene mutations, including TP53, that stimulate the self-renewal of leukemia stem cells or leukemia-initiating cells (LICs) with blockade of differentiation [48]. TP53 mutations usually result in loss of function of the p53 protein, which demonstrates pivotal functions in normal hematopoietic stem cells (HSC) being involved in their proliferation, differentiation, and apoptosis [6]. Wild-type p53 maintains HSC quiescence and negatively regulates HSC aberrant self-renewal [49]. Zhao et al. [49] demonstrated based on a combined mosaic mouse model that mutant p53, in concert with oncogenic Kras, induced aberrant HSC self-renewal and repopulated leukemic blasts, paving a way for the formation of LICs and propagating AML. Also, another mouse model showed that mutant p53 synergizing with FLT3-ITD enhanced the self-renewal potential of LICs, which contributed to leukemia development [50]. Given the critical role of p53 in the initiation, development, and progression of AML, TP53 mutations may constitute a useful tool for prognostication of AML patients, with its prognostic potential systematically validated by this meta-analysis using a collection of previously published data.

Development of AML represents a variety of cytogenetic and molecular abnormalities. Cytogenetics at diagnosis serves as the single most important prognostic indicator for AML and patients with adverse cytogenetics were reported to be less likely to achieve a complete response and long-term survival [46]. However, almost half of AML patients lack cytogenetic abnormality, thus being allocated to the intermediate-risk group with significant clinical heterogeneity [17]. Also, there exists a time gap between the initiation of induction therapy and the obtainment of available cytogenetic information [46]. Thus, to improve the therapeutic outcomes and prognosis, it is of importance to establish a prognostic stratification that takes into consideration a combination of cytogenetic abnormality with gene aberrations, such as TP53 mutation, which have been proved to correlate with CK [38]. The present analysis showed an elevated risk of unfavorable cytogenetics in favor of patients with TP53 mutation and a close association between adverse cytogenetics and poor prognosis, supporting the role of TP53 mutation in prognostic stratification and in assisting the risk stratification scheme based on cytogenetic abnormality.

It has been reported that p53 loss of function correlates with chemotherapy resistance [38]. Results of our subgroup analyses are consistent with previous evidence. TP53 mutations were more significantly associated with a poor OS in adult AML patients treated with intensive chemotherapy treatment, as compared to those treated with nonintensive treatment. Similar results were observed when subgroups were stratified according to the median age of 65. The aforementioned results indicated the nonuse of intensive therapies for newly diagnosed AML patients harboring TP53 mutations. Notably enough, treatment options beyond standard intensive therapies for these patients represent a conundrum. Nonintensive therapies with hypomethylating agents, including azacitidine and decitabine, are frequently provided, especially to elderly AML, yet their curative effects are limited [51]. APR-246 is a novel small molecule designed to shift mutant p53 toward the wild-type conformation and induce apoptosis of TP53 mutated cancer cells. It has been reported that the combination of APR-246 and azacitidine in AML patients with mutant TP53 showed encouraging response rate with good tolerability [52]; however, the current evidence has yet reached the sufficient level and additional research aimed at therapies targeting TP53 mutations in AML patients is needed.

TP53 mutations have been evaluated as a dichotomous variable (mutant vs. wild type) in most of the studies performed so far, as shown in this meta-analysis. More attention should be paid to other novel aspects of TP53 mutations, like functional classifications and variant allele frequencies (VAF), in future studies. There are distinct types of TP53 mutations – missense, nonsense, insertions, deletions, and splice-site mutations. However, it is not yet elucidated whether these types would exert a uniformly poor outcome in AML. Different methods have been applied for the functional classification of TP53 mutations. Of them, relative fitness score, representing an algorithm based on the average of the relative enhancement or depletion of a specific TP53 mutation detected at three time points, has been suggested to hold prognostic significance and awaits further investigation [4, 53]. Besides, Prochazka et al. categorized TP53 mutations according to their VAF into groups of >40% (major AML clone), 20–40%, and <20% (subclones), suggesting that a dismal outcome also occurred in leukemic subclones with a VAF <20% [54]. Thus, more robust evidence for the prognostic role of subclonal TP53 mutations is encouraged.

Based on the close relation between TP53 mutations and an adverse prognosis, their utility in epidemiology and diagnostic workup should be noted. Somatic TP53 mutations observed at diagnosis of AML have been well characterized, yet the presence of detectable mutations years before diagnosis should also be attached importance. Desai and colleagues [55] reported that all healthy women with mutations in TP53 at baseline were eventually developed into AML during a median follow-up of 9.6 years. It is reasonable for genetic screening for mutations in TP53 to be utilized in the setting of AML surveillance.

Several limitations should be seen in our analysis. First, there existed a mixture of prospective and retrospective studies, involving different types of AML in varied centers, which could be an important source of heterogeneity. Subgroup analyses should be treated with caution as the grouping was made based on the available information provided by the included studies where heterogeneity was also inherent regarding the actual treatment regimens. Also, there are limited data in included studies for us to investigate the prognostic value of TP53 mutations based on different VAFs or distinct types. Another limitation included the presence of publication bias, which might be attributed to the fact that we only included studies published in English.

Conclusion

In conclusion, this present meta-analysis investigated the prognostic value of TP53 mutations in adult AML in a comprehensive manner, demonstrating that TP53 mutations were significantly associated with an unfavorable prognosis and could be incorporated into the prognostication and decision-making in the management of AML. Future studies are encouraged to investigate the novel aspects of TP53 mutations in terms of VAF and functional classification and to explore the therapies targeting TP53 mutations in the setting of AML.

Statement of Ethics

An ethics statement is not applicable because this study is based exclusively on the published literature.

Conflict of Interest Statement

The authors have no conflicts of interest to declare.

Funding Sources

This study did not receive any funding.

Author Contributions

Conception and design, administrative support, provision of study materials, collection and assembly of data, data analysis and interpretation, manuscript writing, and final approval of manuscript: all authors.

Data Availability Statement

All data generated or analyzed during this study are included in this article and its online supplementary material. Further enquiries can be directed to the corresponding author.

Supplementary Material

Supplementary data

Funding Statement

This study did not receive any funding.

References

  • 1.Dohner H, Gaidzik VI. Impact of genetic features on treatment decisions in AML. Hematology Am Soc Hematol Educ Program. 2011;2011:36–42. doi: 10.1182/asheducation-2011.1.36. [DOI] [PubMed] [Google Scholar]
  • 2.De Kouchkovsky I, Abdul-Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 2016 Jul 1;6((7)):e441. doi: 10.1038/bcj.2016.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017 Jan 26;129((4)):424–447. doi: 10.1182/blood-2016-08-733196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Molica M, Mazzone C, Niscola P, de Fabritiis P. TP53 Mutations in acute myeloid leukemia: still a daunting challenge? Front Oncol. 2020;10:610820. doi: 10.3389/fonc.2020.610820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers: origins, consequences, and clinical use. Cold Spring Harbor Perspect Biol. 2010 Jan;2((1)):a001008. doi: 10.1101/cshperspect.a001008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stengel A, Kern W, Haferlach T, Meggendorfer M, Fasan A, Haferlach C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: an analysis of 3307 cases. Leukemia. 2017 Mar;31((3)):705–711. doi: 10.1038/leu.2016.263. [DOI] [PubMed] [Google Scholar]
  • 7.Zhang L, McGraw KL, Sallman DA, List AF. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications. Leuk Lymphoma. 2017 Aug;58((8)):1777–1790. doi: 10.1080/10428194.2016.1266625. [DOI] [PubMed] [Google Scholar]
  • 8.Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, et al. Cancer Genome Atlas Research Network Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013 May 30;368((22)):2059–2074. doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.DiNardo C, Lachowiez C. Acute myeloid leukemia: from mutation profiling to treatment decisions. Curr Hematol Malig Rep. 2019 Oct;14((5)):386–394. doi: 10.1007/s11899-019-00535-7. [DOI] [PubMed] [Google Scholar]
  • 10.Hunter AM, Sallman DA. Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol. 2019 Jun;32((2)):134–144. doi: 10.1016/j.beha.2019.05.004. [DOI] [PubMed] [Google Scholar]
  • 11.McCurdy SR, Levis MJ. Emerging molecular predictive and prognostic factors in acute myeloid leukemia. Leuk Lymphoma. 2018;59((9)):2021–2039. doi: 10.1080/10428194.2017.1393669. [DOI] [PubMed] [Google Scholar]
  • 12.Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Bmj. 2021 Mar 29;372:n71. doi: 10.1136/bmj.n71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wells G, Shea B, O'Connell D, Peterson J, Welch V, Losos M, et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. 2010. Available from: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp.
  • 14.Higgins JPT. Thomas J. In: Cochrane Handbook for Systematic Reviews of Interventions (Version 6.2) Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editors. 2021. [Google Scholar]
  • 15.Altman DG, Bland JM. Interaction revisited: the difference between two estimates. Bmj. 2003;326((7382)):219. doi: 10.1136/bmj.326.7382.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bories P, Prade N, Lagarde S, Cabarrou B, Largeaud L, Plenecassagnes J, et al. Impact of TP53 mutations in acute myeloid leukemia patients treated with azacitidine. PLoS One. 2020;15((10)):e0238795. doi: 10.1371/journal.pone.0238795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen X, Zhu H, Qiao C, Zhao S, Liu L, Wang Y, et al. Next-generation sequencing reveals gene mutations landscape and clonal evolution in patients with acute myeloid leukemia. Hematology. 2021 Dec;26((1)):111–122. doi: 10.1080/16078454.2020.1858610. [DOI] [PubMed] [Google Scholar]
  • 18.Daher-Reyes G, Kim T, Novitzky-Basso I, Kim KH, Smith A, Stockley T, et al. Prognostic impact of the adverse molecular-genetic profile on long-term outcomes following allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia. Bone Marrow Transplant. 2021 Mar 25;56((8)):1908–1918. doi: 10.1038/s41409-021-01255-4. [DOI] [PubMed] [Google Scholar]
  • 19.Falk IJ, Willander K, Chaireti R, Lund J, Nahi H, Hermanson M, et al. TP53 mutations and MDM2(SNP309) identify subgroups of AML patients with impaired outcome. Eur J Haematol. 2015 Apr;94((4)):355–362. doi: 10.1111/ejh.12438. [DOI] [PubMed] [Google Scholar]
  • 20.Fenwarth L, Duployez N, Thomas X, Boissel N, Geffroy S, Marceau-Renaut A, et al. Clofarabine improves relapse-free survival of acute myeloid leukemia in younger adults with micro-complex karyotype. Cancers. 2019 Dec 30;12((1)):E88. doi: 10.3390/cancers12010088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fernandez-Pol S, Ma L, Ohgami RS, Arber DA. Immunohistochemistry for p53 is a useful tool to identify cases of acute myeloid leukemia with myelodysplasia-related changes that are TP53 mutated, have complex karyotype, and have poor prognosis. Mod Pathol. 2017 Mar;30((3)):382–392. doi: 10.1038/modpathol.2016.206. [DOI] [PubMed] [Google Scholar]
  • 22.Fujiwara Y, Yamaguchi H, Yui S, Tokura T, Inai K, Onai D, et al. Importance of prognostic stratification via gene mutation analysis in elderly patients with acute myelogenous leukemia. Int J Lab Hematol. 2019 Aug;41((4)):461–471. doi: 10.1111/ijlh.13025. [DOI] [PubMed] [Google Scholar]
  • 23.Grossmann V, Schnittger S, Kohlmann A, Eder C, Roller A, Dicker F, et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood. 2012 Oct 11;120((15)):2963–2972. doi: 10.1182/blood-2012-03-419622. [DOI] [PubMed] [Google Scholar]
  • 24.Hong M, Zhu H, Sun Q, Zhu Y, Miao Y, Yang H, et al. Decitabine in combination with low-dose cytarabine, aclarubicin and G-CSF tends to improve prognosis in elderly patients with high-risk AML. Aging. 2020 Apr 1;12((7)):5792–5811. doi: 10.18632/aging.102973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hou HA, Chou WC, Kuo YY, Liu CY, Lin LI, Tseng MH, et al. TP53 mutations in de novo acute myeloid leukemia patients: longitudinal follow-ups show the mutation is stable during disease evolution. Blood Cancer J. 2015 Jul 31;5((7)):e331. doi: 10.1038/bcj.2015.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Inokuchi K, Yamaguchi H, Hanawa H, Tanosaki S, Nakamura K, Tarusawa M, et al. Loss of DCC gene expression is of prognostic importance in acute myelogenous leukemia. Clinical Cancer Res. 2002 Jun;8((6)):1882–1888. [PubMed] [Google Scholar]
  • 27.Kihara R, Nagata Y, Kiyoi H, Kato T, Yamamoto E, Suzuki K, et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia. 2014 Aug;28((8)):1586–1595. doi: 10.1038/leu.2014.55. [DOI] [PubMed] [Google Scholar]
  • 28.Middeke JM, Herold S, Rucker-Braun E, Berdel WE, Stelljes M, Kaufmann M, et al. TP53 mutation in patients with high-risk acute myeloid leukaemia treated with allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2016 Mar;172((6)):914–922. doi: 10.1111/bjh.13912. [DOI] [PubMed] [Google Scholar]
  • 29.Milosevic JD, Puda A, Malcovati L, Berg T, Hofbauer M, Stukalov A, et al. Clinical significance of genetic aberrations in secondary acute myeloid leukemia. Am J Hematol. 2012 Nov;87((11)):1010–1016. doi: 10.1002/ajh.23309. [DOI] [PubMed] [Google Scholar]
  • 30.Montalban-Bravo G, Kanagal-Shamanna R, Class CA, Sasaki K, Ravandi F, Cortes JE, et al. Outcomes of acute myeloid leukemia with myelodysplasia related changes depend on diagnostic criteria and therapy. Am J Hematol. 2020 Jun;95((6)):612–622. doi: 10.1002/ajh.25769. [DOI] [PubMed] [Google Scholar]
  • 31.Morsia E, McCullough K, Joshi M, Cook J, Alkhateeb HB, Al-Kali A, et al. Venetoclax and hypomethylating agents in acute myeloid leukemia: mayo clinic series on 86 patients. Am J Hematol. 2020 Dec;95((12)):1511–1521. doi: 10.1002/ajh.25978. [DOI] [PubMed] [Google Scholar]
  • 32.Mrózek K, Eisfeld AK, Kohlschmidt J, Carroll AJ, Walker CJ, Nicolet D, et al. Complex karyotype in de novo acute myeloid leukemia: typical and atypical subtypes differ molecularly and clinically. Leukemia. 2019 Jul;33((7)):1620–1634. doi: 10.1038/s41375-019-0390-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Najima Y, Sadato D, Harada Y, Oboki K, Hirama C, Toya T, et al. Prognostic impact of TP53 mutation, monosomal karyotype, and prior myeloid disorder in nonremission acute myeloid leukemia at allo-HSCT. Bone Marrow Transplant. 2021 Feb;56((2)):334–346. doi: 10.1038/s41409-020-01016-9. [DOI] [PubMed] [Google Scholar]
  • 34.Ni J, Hong J, Long Z, Li Q, Xia R, Zeng Q. Mutation profile and prognostic relevance in elderly patients with de novo acute myeloid leukemia treated with decitabine-based chemotherapy. Int J lab Hematol. 2020 Dec;42((6)):849–857. doi: 10.1111/ijlh.13299. [DOI] [PubMed] [Google Scholar]
  • 35.Ohgami RS, Ma L, Merker JD, Gotlib JR, Schrijver I, Zehnder JL, et al. Next-generation sequencing of acute myeloid leukemia identifies the significance of TP53, U2AF1, ASXL1, and TET2 mutations. Mod Pathol. 2015 May;28((5)):706–714. doi: 10.1038/modpathol.2014.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ok CY, Patel KP, Garcia-Manero G, Routbort MJ, Peng J, Tang G, et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J Hematol Oncol. 2015 May 8;8:45. doi: 10.1186/s13045-015-0139-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Österroos A, Björklund M, Eriksson A, Lindberg J, Nilsson C, Mareschal S, et al. Integrated transcriptomic and genomic analysis improves prediction of complete remission and survival in elderly patients with acute myeloid leukemia. Blood Cancer J. 2020 Jun 11;10((6)):67. doi: 10.1038/s41408-020-0332-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rucker FG, Schlenk RF, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012 Mar 1;119((9)):2114–2121. doi: 10.1182/blood-2011-08-375758. [DOI] [PubMed] [Google Scholar]
  • 39.Stahl M, DeVeaux M, Montesinos P, Itzykson R, Ritchie EK, Sekeres MA, et al. Hypomethylating agents in relapsed and refractory AML: outcomes and their predictors in a large international patient cohort. Blood Adv. 2018 Apr;2((8)):923–932. doi: 10.1182/bloodadvances.2018016121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stahl M, Menghrajani K, Derkach A, Chan A, Xiao W, Glass J, et al. Clinical and molecular predictors of response and survival following venetoclax therapy in relapsed/refractory AML. Blood Adv. 2021 Mar 9;5((5)):1552–1564. doi: 10.1182/bloodadvances.2020003734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Terada K, Yamaguchi H, Ueki T, Usuki K, Kobayashi Y, Tajika K, et al. Full-length mutation search of the TP53 gene in acute myeloid leukemia has increased significance as a prognostic factor. Ann Hematol. 2018 Jan;97((1)):51–61. doi: 10.1007/s00277-017-3143-2. [DOI] [PubMed] [Google Scholar]
  • 42.Thol F, Gabdoulline R, Liebich A, Klement P, Schiller J, Kandziora C, et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood. 2018 Oct 18;132((16)):1703–1713. doi: 10.1182/blood-2018-02-829911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Venton G, Courtier F, Charbonnier A, D'Incan E, Saillard C, Mohty B, et al. Impact of gene mutations on treatment response and prognosis of acute myeloid leukemia secondary to myeloproliferative neoplasms. Am J Hematol. 2018 Mar;93((3)):330–338. doi: 10.1002/ajh.24973. [DOI] [PubMed] [Google Scholar]
  • 44.Wang YH, Lin CC, Hsu CL, Hung SY, Yao CY, Lee SH, et al. Distinct clinical and biological characteristics of acute myeloid leukemia with higher expression of long noncoding RNA KIAA0125. Ann Hematol. 2021 Feb;100((2)):487–498. doi: 10.1007/s00277-020-04358-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wu M, Guo ZW, Huang GN, Ye YB. Features and impacts on the prognosis of gene mutations in patients with acute myeloid leukemia. Neoplasma. 2021 May 17;68((5)):1072–1078. doi: 10.4149/neo_2021_201230N1426. [DOI] [PubMed] [Google Scholar]
  • 46.Yanada M, Yamamoto Y, Iba S, Okamoto A, Inaguma Y, Tokuda M, et al. TP53 mutations in older adults with acute myeloid leukemia. Int J Hematol. 2016 Apr;103((4)):429–435. doi: 10.1007/s12185-016-1942-1. [DOI] [PubMed] [Google Scholar]
  • 47.Yu G, Yin C, Wu F, Jiang L, Zheng Z, Xu D, et al. Gene mutation profile and risk stratification in AML1-ETO-positive acute myeloid leukemia based on next-generation sequencing. Oncol Rep. 2019 Dec;42((6)):2333–2344. doi: 10.3892/or.2019.7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nabinger SC, Chen S, Gao R, Yao C, Kobayashi M, Vemula S, et al. Mutant p53 enhances leukemia-initiating cell self-renewal to promote leukemia development. Leukemia. 2019 Jun;33((6)):1535–1539. doi: 10.1038/s41375-019-0377-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Liu Y, Elf SE, Miyata Y, Sashida G, Liu Y, Huang G, et al. p53 regulates hematopoietic stem cell quiescence. Cell Stem Cell. 2009 Jan 9;4((1)):37–48. doi: 10.1016/j.stem.2008.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhao Z, Zuber J, Diaz-Flores E, Lintault L, Kogan SC, Shannon K, et al. p53 loss promotes acute myeloid leukemia by enabling aberrant self-renewal. Genes Dev. 2010 Jul 1;24((13)):1389–1402. doi: 10.1101/gad.1940710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dombret H, Seymour JF, Butrym A, Wierzbowska A, Selleslag D, Jang JH, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015 Jul 16;126((3)):291–299. doi: 10.1182/blood-2015-01-621664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Maslah N, Salomao N, Drevon L, Verger E, Partouche N, Ly P, et al. Synergistic effects of PRIMA-1(Met) (APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia. Haematologica. 2020 Jun;105((6)):1539–1551. doi: 10.3324/haematol.2019.218453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dutta S, Pregartner G, Rucker FG, Heitzer E, Zebisch A, Bullinger L, et al. Functional classification of TP53 mutations in acute myeloid leukemia. Cancers. 2020 Mar 10;12((3)):E637. doi: 10.3390/cancers12030637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Prochazka KT, Pregartner G, Rucker FG, Heitzer E, Pabst G, Wolfler A, et al. Clinical implications of subclonal TP53 mutations in acute myeloid leukemia. Haematologica. 2019 Mar;104((3)):516–523. doi: 10.3324/haematol.2018.205013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Desai P, Mencia-Trinchant N, Savenkov O, Simon MS, Cheang G, Lee S, et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat Med. 2018 Jul;24((7)):1015–1023. doi: 10.1038/s41591-018-0081-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary data

Data Availability Statement

All data generated or analyzed during this study are included in this article and its online supplementary material. Further enquiries can be directed to the corresponding author.


Articles from Transfusion Medicine and Hemotherapy are provided here courtesy of Karger Publishers

RESOURCES