All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know AML.

The AML Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your AML Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The AML Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the AML Hub cannot guarantee the accuracy of translated content. The AML Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2022-03-04T13:40:50.000Z

Molecular analysis of TP53-mutant AML and MDS-EB

Mar 4, 2022
Share:

Bookmark this article

The presence of TP53 mutations in acute myeloid leukemia (AML) and myelodysplastic syndromes with excess blasts (MDS-EB) has been identified as a prognostic factor for poor survival outcomes and complex karyotypes. Despite this, a comparison of the molecular characteristics and treatment responses in patients with TP53-mutated AML and MDS-EB has not yet been performed. If mutant TP53 AML and MDS-EB were found to be similar with respect to molecular characteristics and survival, such findings could suggest a new classification of these disease subtypes as a single molecular disease entity.

An in-depth clinical and molecular analysis of TP53-mutated AML/MDS-EB was recently published by Grob et al.1 in Blood. We summarize key results below.

Methods

A total of 2,200 patients with newly diagnosed AML/MDS-EB who received standard induction chemotherapy were analyzed via next-generation sequencing (NGS).

The study design is summarized in Figure 1.

Figure 1. Study design*

AML, acute myeloid leukemia; MDS-EB, myelodysplastic syndromes with excess blasts; MRD, minimal residual disease.
*Adapted from Grob et al.1

Results

Patient characteristics for the AML and MDS-EB cohorts are summarized in Table 1. No significant differences in patient characteristics were observed apart from a greater presence of deletion 5q in patients with MDS-EB (p = 0.025).

Table 1. Patient characteristics for AML and MDS-EB cohorts*

Characteristic

AML
(n = 186)

MDS-EB
(n = 44)

Median age, years (range)

62 (18–80)

63 (35–73)

Female, %

40

43

White blood cell count ≤100, %

99

100

Allogeneic HSCT, %

74

75

Cytogenetics, %

              Monosomy 5

28

27

              Deletion 5q

44

63

              Monosomy 7

32

34

              Monosomy 17

40

24

              Abnormality 17p

18

15

              Complex karyotype

83

90

              Monosomal karyotype

78

85

AML, acute myeloid leukemia; HSCT, hematopoietic stem cell transplantation; MDS-EB, myelodysplastic syndromes with excess blasts.
*Adapted from Grob et al.1

Molecular analysis

A molecular analysis of mutant allele status, number of TP53 mutations, TP53 variant allele frequency (VAF), and concurrent mutations are summarized in Table 2. No significant difference was reported in any of the molecular characteristics between TP53-mutated AML and MDS-EB. The most prevalent concurrent mutations reported in the entire AML/MDS-EB cohort included DNMT3A (13%), TET2 (9%), ASXL1 (5%), RUNX1 (5%), and SRSF2 (6%).

Most patients with AML/MDS-EB had complex karyotype (84%)

  • The presence of complex karyotype was observed in 97% of patients with bi-allelic TP53-mutated AML/MDS-EB, 94% of patients with multiple TP53 mutations, and 94% of patients with larger TP53 clones (VAF >40%)
  • Concurrent mutations were more prevalent in patients with a non-complex karyotype

Table 2. Molecular characterization of TP53-mutated AML/MDS-EB*

Characteristic

AML
(n = 186)

MDS
(n = 44)

AML/MDS-EB
(n = 230)

TP53-mutant bi-allelic status, %

77

68

76

Multiple TP53 mutations, %

19

30

21

TP53 VAF, median (range)

48 (1–97)

41 (3–91)

47 (1–97)

Concurrent mutation, %

51

41

49

AML, acute myeloid leukemia; MDS, myelodysplastic syndromes; MDS-EB, myelodysplastic syndromes with excess blasts; VAF, variant allele frequency.
*Adapted from Grob et al.1

Treatment outcomes

  • Overall, the presence of TP53 mutations was associated with reduced overall survival (OS) when compared with wildtype patients (2-year OS, 12.8% vs 42.5%, respectively; p < 0.001).
  • Notably, OS was comparable between TP53-mutated AML and MDS-EB (p = 0.549), and so further analysis grouped these diseases together.
  • Evaluating the relationship of molecular characteristics with OS, mono- or bi-allelic TP53 demonstrated comparable poor survival (p = 0.327).
    • Similarly, neither the number of TP53 mutations, rising VAF, or aberrations involving chromosome 17 had a significant impact on OS
    • In contrast, the presence of concurrent mutations produced a small survival benefit (p = 0.042)
  • Complex karyotype was associated with a reduced 2-year OS compared with non-complex karyotype (9% vs 34%; p = 0.002), which was not impacted by the type of consolidation therapy.
    • OS in patients with non-complex karyotype TP53-mutated AML/MDS-EB was still poor
  • The presence of wildtype TP53 appeared to improve survival outcomes in patients with complex karyotype when analyzing the whole cohort of 2,200 patients (p < 0.001).

Molecular minimal residual disease (MRD)

  • As molecular MRD can predict survival outcomes, the authors sequenced bone marrow samples from 62 patients in morphologic complete remission.
  • A total of 45 out of 62 patients had detectable TP53 mutations, but this was not associated with reduced OS (p = 0.653).

Conclusion

Overall, detailed molecular analysis of TP53-mutated AML/MDS-EB revealed homogeneity of molecular characteristics and clinical outcomes between these disease types. As such, the authors indicated a new classification of TP53-mutated AML/MDS-EB as a single disease entity.

  1. Grob T, Al Hinai ASA, Sanders MA, et al. Molecular characterization of mutant TP53 acute myeloid leukemia and high-risk myelodysplastic syndrome. 2022; blood.2021014472. DOI: 10.1182/blood.2021014472

Your opinion matters

Do you intend to implement next generation sequencing for measurable residual disease monitoring in AML patients?
0 votes - 6 days left ...

Newsletter

Subscribe to get the best content related to AML delivered to your inbox