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.
2023-05-16T12:32:32.000Z

Clinical characteristics and outcomes of patients with AML with inv(3)(q21q26.2)/t(3;3)(q21;q26.2)

May 16, 2023
Share:
Learning objective: After reading this article, learners will be able to discuss the characteristics and outcomes of patients with AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2).

Bookmark this article

Inv(3)(q21q26.2) and t(3;3)(q21;q26.2) are types of chromosomal abnormalities present in ~1% of patients with newly diagnosed (ND) acute myeloid leukemia (AML) and are associated with dismal outcomes.1 The 2022 European LeukemiaNet (ELN) guidelines classify the presence of inv(3)(q21q26.2)/t(3;3)(q21;q26.2) as an adverse risk. Patients with AML with inv(3)(q21q26.2) and t(3;3)(q21;q26.2) frequently have additional chromosomal abnormalities, the most common being monosomy 7. There is a need to understand how the clinicopathological characteristics of AML with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) affect prognosis and treatment and the potential benefit of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in this patient group.1

Here, we summarize a retrospective study, published by Richard-Carpentier et al.1 in Haematologica, on the clinicopathological characteristics of patients with AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2) and their impact on clinical outcomes.

Study design and patient characteristics

This retrospective cohort study identified the presence of inv(3)(q21q26.2) or t(3;3)(q21;q26.2) in patients diagnosed with AML at the MD Anderson Cancer Center from January 2000 to September 2020. A total of 108 patients with AML with inv(3)(q21q26.2)/t(3;3)(q21;q26.2) were included; 53 patients and 55 patients were in the ND AML and relapsed/refractory (R/R) cohorts, respectively (Table 1).

Table 1. Baseline characteristics*

Characteristics, % (unless otherwise specified)

Total
(N = 108)

ND AML cohort
(n = 53)

R/R AML cohort
(n = 55)

p value

Median age (range), years

55 (16–84)

63 (16–84)

48 (18–83)

<0.01

Age ≥60 years

41

53

30

0.01

Male

63

58

67

0.46

Median WBC (range), × 109/L

3.9 (0.3–143.2)

4.1 (0.6–143.2)

3.4 (0.3–101.0)

0.06

Hb (range), g/dL

8.8 (5.7–17.1)

8.6 (5.7–17.1)

8.9 (7.0–13.8)

0.81

Platelets (range), × 109/L

67 (7–787)

80 (15–787)

50 (7–372)

0.02

Median PB blasts (range)

20 (0–96)

18 (0–96)

21 (0–93)

0.34

Median BM blasts (range)

48 (16–94)

35 (16–94)

54 (19–92)

0.02

Secondary AML

27

21

33

0.24

t-AML

18

32

5

<0.01

inv(3)(q21q26.2)

81

85

76

0.38

ACA

77

72

82

0.31

               −7/del(7q)

56

53

58

0.71

               del(5q)

16

17

15

0.93

               CK

30

25

35

0.35

               MK

49

47

51

0.84

ACA, additional chromosomal abnormalities; AML, acute myeloid leukemia; BM, bone marrow; CK, complex karyotype; del, deletion; Hb, hemoglobin; inv, inversion; MK, monosomal karyotype; ND, newly diagnosed; PB, peripheral blood; R/R, relapsed/refractory; t, translocation; t-AML, therapy-related AML; WBC, white blood cell.
*Adapted from Richard-Carpentier, et al.1
Presence of ≥3 unrelated clonal chromosomal abnormalities (≥2 ACA accompanying inv(3)(q21q26.2)/t(3;3)(q21;q26.2)).
Presence of ≥2 monosomies or ≥1 monosomy in the presence of a structural chromosomal abnormality (≥1 monosomy accompanying inv(3)(q21q26.2)/t(3;3)(q21;q26.2)).

Key findings

Genetic characteristics

AML with inv(3)(q21q26.2) was identified in 81% of patients. However, only patients with AML with inv(3)(q26.2;q21) showed a white blood cell (WBC) count ≥20 × 109/L (25% vs 0%; p = 0.02).

Mutated genes

  • The most frequently mutated gene in this population was SF3B1 (Figure 1).
  • NRAS mutations were associated with more frequent occurrence of WBC count ≥20 × 109/L (39% vs 14%; p = 0.02).
    • NRAS mutations were also more common in the R/R AML versus ND AML cohort (14% ND vs 33% R/R; p = 0.04).
  • KRAS mutations were associated with a higher median peripheral blood blast (PBB) percentage (52% vs 16%; p < 0.01).
  • TP53 mutations occurred more frequently in the ND AML compared with the R/R AML cohort (16% vs 0%; p = 0.04).
  • ASXL1 and NRAS mutations were reported more often in patients with secondary AML (ASXL, 33% vs 6%; p = 0.046 and NRAS, 46% vs 16%; p < 0.01).

Figure 1. Frequency of gene mutations in patients with AML with inv(3)(q21q26.2)/t(3;3)(q21;q26.2)* 

AML, acute myeloid leukemia; inv, inversion; t, translocation.
*Adapted from Richard-Carpentier, et al.1

Remission rates

In total, 96 patients who received treatment were evaluable for response. Composite complete remission (CRc) was achieved by 46% vs 14% (p < 0.01) of patients in the ND AML and R/R AML cohorts, respectively (Table 2). The rate of measurable residual disease negativity was 38% vs 0% in the ND AML and R/R AML cohorts, respectively. The median duration of CRc was 5.4 months in patients who received venetoclax-based regimens.

ND AML cohort

  • CRc rates were similar for patients treated with high- and low-intensity treatments (p = 1.00).
  • The 30-day mortality rates were comparable between high- and low-intensity treatment groups (p = 0.16).
  • In the univariate analyses, higher PBB percentage was associated with lower CRc in the ND AML cohort (odds ratio, 0.98; p = 0.02).
  • Lower CRc rates were observed in patients with secondary AML versus patients with ND AML (10% vs 38%; p = 0.02).

R/R AML cohort

  • CRc rates were not significantly different between patients who received high- and low-intensity treatments (p = 0.39).
  • The 30-day mortality rates were similar between the high- and low-intensity treatment groups (p = 1.00).

Table 2. Response rates*

Clinical outcome, % (unless otherwise specified)

Total
(n = 96)

ND AML cohort

R/R AML cohort

Total
(n = 52)

HI
(n = 35)

LI
(n = 17)

Total
(n = 44)

HI
(n = 20)

LI
(n = 24)

CRc

31

46

46

47

14

20

8

CR

17

29

26

35

2

5

0

CRi

15

17

20

12

11

15

8

MLFS

2

4

0

12

0

0

0

ORR

33

50

46

59

14

20

8

No. of cycles to remission (range)

1 (1–4)

1 (1–4)

1 (1–2)

1.5 (1–4)

1.5 (1–3)

1.5 (1–3)

1.5 (1–2)

Median days to remission (range)

39.5

(19–126)

37

(19–126)

34.5

(20–85)

53

(19–126)

53.5

(27–102)

73

(31–102)

42

(27–57)

30-day mortality

8

10

14

0

7

5

8

CRc according to treatment type

 

 

 

 

 

 

 

               With Ven

33

20

0

25

43

66

24

               Without Ven

31

49

47

54

8

12

5

               With HMA

36

50

NA

50

20

NA

20

               Without HMA

10

40

NA

40

0

NA

0

AML, acute myeloid leukemia; CR, complete remission; CRc, composite CR; CRi, CR with incomplete hematological recovery; HI, high intensity; HMA, hypomethylating agents; LI, low intensity; MLFS, morphologic leukemia-free state; ND, newly diagnosed; ORR, overall response rate; R/R, relapsed/refractory; Ven, venetoclax.
*Adapted from Richard-Carpentier, et al.1

An additional patient treated with a low-intensive venetoclax-based regimen achieved MLFS and is not included in the CRc rates.

Survival outcomes

  • At a median follow-up of 83 months, the median overall survival (OS) was 7.9 months and 5.9 months in the ND AML and R/R AML cohorts, respectively.
  • The 3-year OS rate was 8.8% vs 7.1% in the ND AML and R/R AML cohorts, respectively.
  • The median relapse-free survival (RFS) was 4.1 months, with a 3-year RFS rate of 8.6% and a 3-year cumulative incidence of relapse rate of 81.7%.
  • In total, 6% of patients were alive at the last follow up or had RFS ≥5 years.
  • Univariate analyses revealed that age (hazard ratio [HR], 1.01; p = 0.04), WBC count ≥20 × 109/L at diagnosis (HR, 1.02; p < 0.01), higher PBB percentage (HR, 1.01; p < 0.01), and secondary AML (HR, 1.81; p = 0.01) were associated with worse OS.
    • A WBC count ≥20 × 109/L was associated with significantly worse OS compared with a WBC count ≤20 x 109/L (HR, 2.19; p < 0.01).
  • Multivariable analyses revealed that higher WBC count ≥20 × 109/L (HR, 5.67; p < 0.01), secondary AML (HR, 4.14, p < 0.01), and ASXL1 mutations (HR, 2.83; p = 0.049) were independently associated with worse OS.
  • Mutation analysis revealed the following:
    • NRAS mutations were associated with inferior OS (HR, 2.39; p = 0.06) among patients in the ND AML cohort.
    • KRAS mutation was associated with poor OS (HR, 2.37; p = 0.01), particularly in the R/R AML cohort.
    • ASXL1 (HR, 2.62; p = 0.04) and DNMT3A (HR, 3.09; p = 0.02) mutations were also associated with poor OS.

Allo-HSCT

  • Among patients achieving CRc or morphologic leukemia-free state, 31% received allo-HSCT in first complete remission (CR1).
    • The median time from response to allo-HSCT was 56 days and 39 days in the ND AML and R/R AML cohorts, respectively.
  • At 4 months, allo-HSCT in CR1 was associated with improved OS (HR, 0.33; p = 0.03).
  • The 5-year OS rates were higher in patients who received allo-HSCT compared with those who did not receive allo-HSCT in CR1 (44% vs 6%).
  • The 2-year cumulative incidence of relapse was lower among patients who received allo-HSCT compared with those who did not receive allo-HSCT in CR1 (57% vs 86%).
  • Time-dependent variable analysis showed allo-HSCT in CR1 improved RFS (HR, 0.39; p = 0.046) and generally showed a trend towards improving OS (HR, 0.41; p = 0.08).

Conclusion

This retrospective study highlights the very poor prognosis associated with AML with inv(3)(q21q26.2)/t(3;3)(q21;q26.2). Current treatment options for this patient population remain limited, with similar response rates observed in both high- and low-intensity chemotherapy treatment groups in the ND AML cohort. The addition of venetoclax did not improve response rates, suggesting that hypomethylating agent-based therapies may be the preferred treatment option in this patient group. While allo-HSCT in CR1 was associated with improved survival outcomes, few patients in this study proceeded to transplantation due to high rates of relapse. This study demonstrates the need for novel therapeutics to treat patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2).

  1. Richard-Carpentier G, Rausch C R, Sasaki K, et al. Characteristics and clinical outcomes of patients with acute myeloid leukemia with inv(3)(q21q26.2) or t(3;3)(q21;q26.2). Haematologica. 2023. Online ahead of print. DOI: 3324/haematol.2022.282030

Newsletter

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