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.
2019-08-14T12:36:07.000Z

KIT mutations in pediatric, core binding factor (CBF), acute myeloid leukemia (AML) 

Aug 14, 2019
Share:

Bookmark this article

Core binding factor (CBF)* acute myeloid leukemia (AML) is present in 20–30% of pediatric cases and is associated with favorable outcomes. The co-occurrence of KIT mutations, found in 20–40% of patients, has been reported to be a negative prognostic factor in some studies, however, other contradictory studies reported no association between KIT mutation and clinical outcome.1 Whilst the European Leukemia Network (ELN) 2017 guidelines recognize that CBF AML with KIT mutations appear to be associated with a poorer prognosis, they do not recommend patients are assigned to a different genetic risk category.2

To better determine whether KIT mutations are of prognostic significance in CBF AML, Katherine Tarlock, Fred Hutchinson Cancer Research Center and Seattle Children’s Hospital, Seattle, US, and colleagues conducted a study, recently published in Clinical Cancer Research.1 They aimed to:

  • Identify the functional impact of distinct KIT mutation subsets
  • Analyze the response of different KIT mutants to tyrosine kinase inhibitors (TKIs) in vitro
  • Define the prognostic impact of KIT + CBF AML using data from a phase III trial, COG AAML0531 (NCT01407757)

* CBF AML is associated with the cytogenetic abnormalities translocation (t)(8;21)(q22;q22) and inversion(16)(p13;1q22)/t(16;16)(p13;q22)

Background1

The KIT proto-oncogene encodes a transmembrane glycoprotein type III receptor tyrosine kinase (RTK). KIT mutations cause significant oncogenic events as they affect RTK activity. In some malignancies with KIT mutations, such as gastrointestinal stromal tumors, melanomas, and mastocytosis. TKIs have proven effective and have become standard-of-care for first-line therapeutic approaches.

Study design and patient characteristics1

  • Evaluated two KIT mutations:
    • Immunoglobulin G (IgG) domain - exon 8 (E8)
    • Tyrosine kinase domain (TKD) - exon 17 (E17)
  • Used data from the phase III study COG AAML0531 (NCT01407757) to analyze patient outcomes by mutation status (KIT+ vs KIT-) and mutation location (E8 vs E17)
    • Study compared the use of the CD33-targeted antibody-drug conjugate, gemtuzumab ozogamicin (GO) in combination with conventional chemotherapy, to conventional chemotherapy alone
    • Of the total patient population (N= 1,022), 247 eligible patients were identified with CBF AML, of whom 205 had available bone marrow (BM) or peripheral blood specimens for KIT analysis
    • KIT mutations were identified in 63 patients (31%). These were located in:
      • E8 only: 22 (35%)
      • E17 only: 32 (51%)
      • Both E8 and E17: 6 (10%)
      • Alternative exons: 3 (5%)
    • Outcomes of patients with KIT+ (n= 63) were compared to those who were KIT- (n= 142)
      • No significant differences in baseline characteristics between two groups and no difference in treatment assignment
      • KIT+ patients had a higher percentage of BM blast cells and peripheral blasts cells

Results1

Growth characteristics of transduced cells in vitro

  • HEK293 cells transduced with E8 mutations in the IgG domain (D419G, D419del) showed:
    • No KIT phosphorylation
    • No interleukin-3 (IL-3) independent proliferation when transduced into Ba/F3 cells
  • HEK293 cells transduced with E17 mutations in the TKD (D816V, N822K) showed:
    • Autonomous receptor phosphorylation
    • IL-3 independent proliferation in Ba/F3 cells

Response of transduced cells to TKI in vitro

  • Ba/F3 cells (E17 variants): uniquely susceptible to the TKIs, dasatinib and crenolanib
    • Both dasatinib and crenolanib provided an apoptotic effect
    • Supports E17 mutations as therapeutic targets in AML
  • Ba/F3 cells (parental or KIT-): no response to TKIs with IC50s > 10,000 nmol/L
    • Cells transduced with E8 KIT mutations were insensitive to the cytotoxic effects of TKIs
  • These results support the investigation of using TKIs in patients with E17, but not E8, CBF AML

Clinical analysis

The prognostic significance of KIT mutations was analyzed by comparing outcomes between patients with KIT+ and KIT- CBF AML (Table 1).

  • Five-year overall survival (OS) was comparable between both groups
  • There was an overall trend for decreased event-free survival (EFS) and disease-free survival (DFS), and increased relapse rate (RR) in KIT+ patients
  • In patients who received traditional chemotherapy, those with KIT mutations had inferior EFS, DFS and a higher RR
  • When comparing outcomes by location of KIT mutation and treatment type (Tables 2–4)
    • E8 KIT+ patients: no improvement in outcome when treated with GO
    • E17 KIT+ patients: DFS and RR were comparable to KIT- patients indicating a trend towards improved outcomes in this subset of patients. Indicates GO abrogated the negative impact of E17 KIT mutation

In the Tables below, statistically significant results are indicated in a bold font.

Table 1. Comparison of outcomes to evaluate the impact of KIT mutations

 

KIT+ (%)

KIT- (%)

p value

Five-year OS

78 ± 12

81% ± 7

0.905

Patients in CR

DFS

55 ± 14

73 ± 8

0.040

RR

43 ± 14

21 ± 8

0.005

Treatment with conventional chemotherapy only

EFS

44 ± 20

70 ± 11

0.021

DFS

43 ± 20

69 ± 12

0.034

RR

57 ± 21

28 ± 12

0.013

Table 2. Comparison of outcomes in patients with E17 KIT mutations, overall and by treatment

 

KIT+ (%)

KIT- (%)

p value

E17 KIT+ versus KIT-

 

 

 

OS

72 ± 17

81 ± 7

0.478

DFS

51 ± 18

73 ± 8

0.027

RR

46 ± 19

21 ± 8

0.007

E17 KIT+ versus KIT- treated with GO

DFS

67 ± 24

78 ± 11

0.396

RR

27 ± 24

13 ± 9

0.207

E17 KIT+ versus KIT- treated with conventional chemotherapy

DFS

35 ± 25

69 ± 12

0.021

RR

65 ± 27

28 ± 12

0.010

Table 3. Comparison of outcome in E17 KIT+ by treatment type

 

GO (%)

Conventional chemotherapy

p value

DFS

67 ± 24

35 ± 25

0.161

RR

27 ± 24

65 ± 27

0.062

Table 4. Comparison of outcomes in patients with E8 KIT mutations, overall and by treatment

 

KIT+ (%)

KIT- (%)

p value

E8 KIT+ versus KIT-

 

 

 

OS

91 ± 12

81 ± 7

0.302

DFS

60 ± 22

73 ± 8

0.305

RR

40 ± 23

21 ± 8

0.072

E8 KIT+ versus KIT- treated with GO

OS

100 ± 0

77 ± 10

0.092

DFS

64 ± 29

78 ± 11

0.456

RR

36± 31

13 ± 9

0.082

Advantages and limitations of analysis1

Advantages

  • Patients were homogenously treated with the same chemotherapy backbone
    • Provides an insight into distinct functional properties of KIT mutations in context of GO treatment
  • Large scale analysis: allowed investigation by location of KIT mutation
  • Investigated CD33-targeted therapy in KIT+ CBF AML
    • Provides an insight into the potential therapeutic efficacy of anti-CD33 agents in this population

Limitations

  • Prognostic impact of KIT mutations may be influenced by therapy (inclusion of GO and no GO patients)
  • A larger cohort size would be required to definitively confirm these results

Conclusions1

In vitro:

  • KIT mutations in E17 were associated with aberrant KIT phosphorylation, and a higher sensitivity to KIT-directed TKIs in vitro
  • KIT mutations in E8 had no functional impact

In vivo

  • Patients with E17-mutated CBF AML form a high-risk subgroup
  • Despite a lower CD33 expression than other AML subgroups, this study supports the use of CD33-targeted, intensive treatment for the E17 KIT+ subgroup
  • Adding a TKI to pediatric CBF AML treatment may be an important strategy
    • TKIs have been shown to be safe when combined with intensive chemotherapy
  1. Tarlock K. et al., Functional Properties of KIT Mutations Are Associated with Differential Clinical Outcomes and Response to Targeted Therapeutics in CBF Acute Myeloid Leukemia. Clin Can Res. 2019 Jun 10. DOI: 10.1158/1078-0432.CCR-18-1897
  2. Döhner H. et al., Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2016 Nov 28. DOI: 10.1182/blood-2016-08-733196

Your opinion matters

Do you intend to implement next-generation sequencing for measurable residual disease monitoring in MDS patients?
1 vote - 3 days left ...

Newsletter

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