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.
2020-05-07T08:45:25.000Z

High immune checkpoint expression levels in patients with AML correlated with poor outcome

May 7, 2020
Share:

Bookmark this article

Immune checkpoint inhibitors (ICIs) in acute myeloid leukemia (AML) therapy are being evaluated in a variety of clinical trials. Unfortunately, significant differences in response rate have been observed amongst clinical trials using ICIs, probably because of different immune checkpoint (IC) expression levels or expression patterns that yet need to be fully characterized.

Here we report the results of a study recently published in the Journal of Hematology & Oncology by Cunte Chen et al., evaluating the IC expression patterns and its prognostic value in patients with newly diagnosed or de novo AML.1

The use of ICIs in AML therapy is the current AML Hub editorial theme, read more information here.

Study design1

  • RNA-seq and mutation data from patients with de novo AML (n = 176), from the Cancer Genome Atlas (TCGA) database, were used for overall survival (OS) analysis
  • The results of this analysis were confirmed by analyzing bone marrow samples from 62 patients with de novo AML, from now on referred to as the validation group
  • The cutoff value for gene expression levels was determined by a bioinformatic tool (X-tile software)

Results

  • The TCGA database analysis showed a correlation between higher expression of PD-1, PD-L1, and PD-L2 with an inferior OS; these results were confirmed in the validation group. The OS rates are reported in Table 1

Table 1. 3-year OS in AML patients with high or low PD-1, PD-L1, or PD-L2 expression1

OS, overall survival

 

3-year OS, %

p value

TCGA group (n = 176)

PD-1

High

Low

PD-L1

High

Low

PD-L2

High

Low

 

 

23

38

 

19

46

 

15

40

 

0.004

 

 

0.019

 

 

0.001

Validation group (n = 62)

PD-1

High

Low

PD-L1

High

Low

PD-L2

High

Low

 

 

40

68

 

22

64

 

42

68

 

0.020

 

 

< 0.001

 

 

0.012

  • Additionally, other relevant IC patterns of expression were studied. The cytotoxic T-lymphocyte associated protein 4 (CTLA-4), and lymphocyte activation gene-3 (LAG-3) are both inhibitory molecules identified as promising immunotherapy targets for various hematological and solid tumors.2 In this study, poorer OS rates were observed in patients with AML with high expression of CTLA-4 and LAG-3 vs patients with low expression1
    • TCGA group, 3-year OS
      • CTLA-4: 9% vs 36% (p < 0.001)
      • LAG-3: 13% vs 40% (p = 0.002)
    • Validation group, 3-year OS
      • CTLA-4: 34% vs 66% (p = 0.001)
      • LAG-3: 33% vs 70% (p < 0.001)
  • In the TCGA group, a positive correlation of PD-1, PD-L1, or PD-L2 with CTLA-4 and LAG-3 was observed. These results were confirmed in the validation group (PD-L1/CTLA-4, PD-L2/CTLA-4, PD-L1/ LAG-3, and PD-L2/LAG-3 showed the stronger correlation with r value ˃ 0.9) and open the possibility to potentially improve responses by combining different ICIs according to these expression patterns1
  • In both the TCGA database and validation group, patients with concomitant high expression of PD-1 or PD-L2, and CTLA-4 showed poor OS, while patients with PD-L1high CTLA-4high showed significantly worse OS in comparison to patients with PD-L1high CTLA-4low, in the TCGA dataset but not in the validation group. The OS analysis in patients with concomitant high expression of PD-1, PD-L1 or PD-L2, and LAG-3 did not result in any significant differences except for patients with PD-1high LAG-3high showing a significantly worse OS in comparison to patients with PD-1high LAG-3high (31% vs 70%; p = 0.025) in the validation group. The OS rates for patients with concomitant expression of CTLA-4 are reported in Table 2, but these results need to be taken carefully since some subgroups have limited samples1

Table 2. 3-year OS in patients with high expression of PD-1, PD-L1 or PD-L2 concomitant with CTLA-4low or CTLA-4high

CTLA-4, cytotoxic T-lymphocyte associated protein 4; OS, overall survival

*Value indicated in bold is not statistically significant

 

3-year OS, %

p value

TCGA group

PD-1high

CTLA-4low

CTLA-4high

PD-L1high

CTLA-4low

CTLA-4high

PD-L2high

CTLA-4low

CTLA-4high

 

 

30

0

 

24

0

 

20

0

 

 

0.036

 

 

< 0.001

 

 

0.041

Validation group

PD-1high

CTLA-4low

CTLA-4high

PD-L1high

CTLA-4low

CTLA-4high

PD-L2high

CTLA-4low

CTLA-4high

 

 

57

31

 

33

20

 

57

33

 

 

0.042

 

 

0.353

 

 

0.043

  • An exploratory analysis was performed with the TCGA group to try to identify poor prognosis IC patterns of expression in patients with common AML mutations. The authors state that the OS of patients with FLT3, RUNX1, or TET2 mutations revealed that high co-expressions of PD-1/PD-L1, PD-1/PD-L1/PD-L2, and PD-1/LAG-3 respectively, were associated with poor OS in comparison with patients without these mutations (OS rates are reported in Table 3)1

Table 3. 1-year OS in patients with FLT3, RUNX1, or TET2 mutations and co-expressions of PD-1, PD-L1, PD-L2 or LAG-31

LAG-3, lymphocyte activation gene-3; OS, overall survival

 

1-year OS, %

p value

FLT3mut

PD-1high/PD-L1high

PD-1high or PD-L1high

PD-1low/PD-L1low

 

0

58

49

0.029

RUNX1mut

PD-1high/PD-L1high/PD-L2high

PD-1high or PD-L1high or PD-L2high

PD-1low/PD-L1low/PD-L2low

 

0

56

100

0.003

TET2 mut

PD-1high/LAG-3high

PD-1high or LAG-3high

PD-1low/LAG-3low

 

0

100

63

< 0.001

Conclusion1

This study showed that patients with AML and high expression of PD-1, PD-L1, or PD-L2 have significantly shorter survival. Moreover, the correlation between higher expression of the named ICs with CTLA-4 and LAG-3 gives the biological rationale to test dual-targeted combinations of ICs in these patients.

Furthermore, the co-expression of PD-1/ CTLA-4 or PD-L2/CTLA-4 was associated with poorer OS in patients with de novo AML. In addition, the concomitant high expression of PD-1/PD-L1, PD-1/PD-L1/PD-L2, and PD-1/LAG-3 was associated with poor OS in patients with FLT3, RUNX1, or TET2 mutations, respectively. This co-expression patterns might be potential biomarkers for designing novel AML targeted therapy and need to be further explored. Altogether, these results demonstrated that high expression of ICs correlated with poor outcome in patients with de novo AML, highlighting their potential prognostic value.

  1. Chen C, Liang C, Wang S et al. Expression patterns of immune checkpoints in acute myeloid leukemia. J Hematol Oncol. 2020;13(1):28-32. DOI: 1186/s13045-020-00853-x
  2. Hobo W, Hutten TJA, Schaap NPM, et al. Immune checkpoint molecules in acute myeloid leukaemia: managing the double-edged sword. Br J Haematol. 2018;181(1):38-53. DOI: 1111/bjh.15078. 

Your opinion matters

Do you intend to implement next-generation sequencing for measurable residual disease monitoring in MDS patients?
1 vote - 2 days left ...

Newsletter

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