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 the MDS Alliance.

The MDS 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 MDS Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The MDS 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 MDS Hub cannot guarantee the accuracy of translated content. The MDS 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.
As of January 1st, 2024, the MDS Hub will no longer be updated. Please continue to browse our archive for valuable content. For the latest updates in MDS, visit our sister site aml-hub.com.
2021-05-17T11:58:56.000Z

Efficacy and safety of CC-486 (oral azacitidine) in patients with lower risk myelodysplastic syndromes

May 17, 2021
Share:

Bookmark this article

The most common feature of myelodysplastic syndromes (MDS) is anemia, with thrombocytopenia having a particularly poor prognostic implication for patients with low-risk MDS (LR-MDS), subsequently decreasing overall survival (OS). LR-MDS patients have conventionally been managed with transfusions, erythropoiesis stimulating agents, and hematopoietic growth factors. However, a subgroup of LR-MDS patients with red blood cell transfusion-dependency (RBC-TD) anemia, and thrombocytopenia require therapeutic agents that address both these deficiencies. There is anecdotal evidence that injectable hypomethylating agents (HMAs) may address anemia and thrombocytopenia in MDS patients.

In a randomized controlled trial, published in the Journal of Clinical Oncology, Garcia-Manero et al. evaluated CC-486 (oral azacitidine) in LR-MDS patients, and the key findings are summarized below.1

Study design

Phase III, randomized, placebo-controlled multicenter trial (NCT01566695). The patients were aged ≥18 years with LR-MDS, had an Eastern Cooperative Oncology Group (ECOG) performance status score ≤2, and having RBC-TD anemia and thrombocytopenia.

Patients (n = 216) were randomly assigned either to 300 mg of CC-486 (n = 107) or placebo (n = 109), administered once daily for 21 days per 28-day treatment cycle.

  • Primary endpoint was rate of RBC transfusion independence (TI) lasting ≥56 consecutive days at 12 months.
  • Secondary endpoint included OS, time to and duration of RBC-TI, rate and duration of ≥84 days of RBC-TI and platelet TI, hematologic improvement in the erythroid (HI-E) and platelet (HI-P) lineages, morphologic response among patients with >5% blasts at baseline, and AML progression.

Results

Baseline characteristics

The median age was 74 years, median platelet count was 25 × 109/L, and absolute platelet count was 1.3 × 109/L. Baseline characteristics were well balanced between the two groups except that a higher proportion of patients in the placebo arm had >5% blasts (28.4% versus 15.9%) (Table 1).

Table 1. Baseline characteristics.*

Characteristic

CC-486
(n = 107)

Placebo
(n = 109)

Median age, years, (range)
              Age ≥ 65 years, %

74.0 (30−89)
85.0

73.0 (44−88)
87.2

Sex, %
              Male


73.8


72.5

WHO 2008 MDS classification, %
RCMD
RAEB-1
RA
RARS
MDS-U
RT


74.8
15.9
3.7
2.8
1.9
0.9


67.0
26.6
2.8
1.8
1.8
0.0

IPSS-R risk, %
Very low (≤ 1.5)
Low (> 1.5−3)
Intermediate (> 3−4.5)
High (> 4.5−6)
Very high (> 6)
Missing


0.0
22.4
47.7
25.2
0.9
3.7


0.0
19.3
44.0
30.3
0.0
6.4

ECOG-PS score, %
0−1
2


85.0
15.0


86.2
13.8

IPSS cytogenic risk, %
Good
Intermediate
Missing


81.3
15.9
2.8


82.6
12.8
4.6

Gene mutations, %
ASXL1
TET2
RUNX1
U2AF1
SRSF2
DNMT3A


28.4
20.6
9.8
17.6
13.7
9.8


38.2
22.5
22.5
14.7
15.7
14.7

Platelet transfusion-dependent, %

28.0

32.1

Months since MDS diagnosis, median (range)

18.9 (0.9−153)

16.1 (0.4−381)

RBC transfusion requirement per 28 day, units, median (range)
> 4 units, %


3.3 (1.3−10.0)
33.6


3.3 (1.3−9.5)
31.2

Hemoglobin, g/dL, median (range)

8.3 (5.4−10.9)

8.1 (5.7−10.1)

Platelets, 109/L, median, (range)
Platelet count < 20 × 109/L, %

24 (566)
41.1

25 (573)
44.0

ANC, 109/L, median (range)

1.36 (0.07−25.2)

1.28 (0.06−20.5)

Absolute lymphocyte count, %
< 0.5 × 109/L
≥ 0.5 × 109/L


18.7
81.3


10.1
89.0

Bone marrow blasts, median (range)
< 5% blasts, %
≥ 5% blasts, %

3.0 (0.0−9.0)
83.2
15.9

3.5 (0.0−9.0)
71.6
28.4

ANC, absolute neutrophil count; ECOG-PS, Eastern Cooperative Oncology Group performance status; IPSS, International Prognostic Scoring System; IPSS-R, Revised IPSS; MDS, myelodysplastic syndromes; MDS-U, MDS-unclassified; RA, refractory anemia; RAEB, RA with excess blasts; RARS, RA with ringed sideroblasts; RBC, red blood cell; RCMD, refractory cytopenia with multilineage dysplasia; RT, refractory thrombocytopenia.

*Adapted from Garcia-Manero et al.1

Patients were considered platelet transfusion-dependent at baseline if they had received ≥2 platelet transfusions within the 56 days before random assignment and had no consecutive 28-day period during which no platelet transfusions were administered.

The average of RBC transfusion units per 28 days was derived using transfusion records for the 84 days or 56 days before random assignment.

Efficacy

  • Patients in the CC-486 arm achieved significantly higher RBC-TI ≥ 56 consecutive days compared with placebo (30.8% vs 11.1%) (Table 2). Similarly, the CC-486 arm achieved a higher RBC-TI ≥84 days compared with the placebo arm (28% vs 5.6%).
  • The median time to RBC-TI was 2.4 vs 2 months in CC-486 and placebo arm respectively. The median RBC-TI duration was much higher in the CC-486 arm (11.1 vs 5 months) compared with the placebo arm.
  • RBC transfusion reductions were sustained longer for a median of 10 months in the CC-486 arm compared with 2.3 months in the placebo arm.
  • HI-P response was higher in the CC-486 arm (24.3% vs 6.5%) compared with placebo.
  • Platelet-TI was attained in 16.7% of patients in the CC-486 arm compared with 14.3% in the placebo arm. It was also sustained longer in the CC-486 arm for a median of 12.1 months (95% CI, 8.3−not estimable [NE]) compared with 4.4 months (95% CI, 2.3−NE) in the placebo arm.

Table 2. Efficacy.*

Efficacy

CC-486 (n = 107)

Placebo (n = 109)

Odds ratio

95% CI; p value

RBC-TI ≥ 56 consecutive days, %

30.8

11.1

3.6

1.7–7.4; 0.0002

RBC-TI ≥ 84 consecutive days, %

28.0

5.6

6.6

2.6–16.7; < 0.0001

HI-E response, %

43.0

31.5

1.6

0.9–2.9; 0.12

HI-P response, %

24.3

6.5

4.6

1.9–11.2; 0.0003

≥1.5 g/dL increase in hemoglobin from baseline, %

23.4

4.6

6.3

2.3–17.1; < 0.0001

RBC

transfusion reductions of ≥ 4 units/56 days, %

42.1

30.6

1.7

0.9–2.9; 0.12

CI, confidence interval; HI-E, hematologic improvement in erythroid; HI-P, hematologic improvement in platelet; RBC, red blood cell; RBC-TI, red blood cell transfusion independence.

*Data from Garcia-Manero et al.1

 Overall survival

  • Overall, there were 140 deaths; 39 (CC-486, n = 25; placebo, n = 14) during treatment, and 101 (CC-486, n = 44; placebo, n = 57) during posttreatment follow-up.
  • There was no difference in OS between CC-486 and placebo (median, 17.3 vs 16.2 months, hazards ratio, 0.99 (95% CI, 0.70–1.40; p = 0.96).
  • Rate of AML progression was nearly half in the CC-486 arm (7.5% vs 16.7%; HR, 0.40; 95% CI, 0.16–0.97; p = 0.04) compared with placebo.

Safety

  • The most common adverse events (AEs) in both arms were lower grade gastrointestinal (GI) events (100% in CC-486, and 99.1% in placebo).
  • Grade 3−4 hematologic TEAEs and infections were more frequent in the CC-486 arm compared with placebo (Table 3).
  • 29% of patients in the CC-486 arm required ≥ 1 dose reduction due to TEAEs. Patients who permanently discontinued the study due to TEAEs were similar across both groups (29.9% in the CC-486 group and 28.4% in the placebo group).
  • Early deaths were imbalanced between groups, with 16 in the CC-486 and six in the placebo, occurring during the first 56 days.

Table 3. Grade 3−4 TEAEs.*

TEAE

CC-486 (n = 107), %

Placebo (n = 109), %

≥ 1 Grade 3−4 TEAEs

89.7

73.4

Neutropenia

46.7

11.9

Thrombocytopenia

29.0

15.6

Febrile neutropenia

28.0

10.1

Anemia

18.7

16.5

Pneumonia

12.1

9.2

TEAEs, treatment-emergent adverse events.

*Data from Garcia-Manero et al.1

 Conclusions

The findings from this study demonstrate that CC-486 can provide clinically significant reductions in the RBC transfusion burden and improve thrombocytopenia in patients with LR-MDS. Although a higher rate of early deaths occurred in patients in the CC-486 arm, most of these were related to infections in patients with significant pretreatment neutropenia. Further studies are needed to evaluate CC-486 in different subgroups of patients with MDS.

  1. Garcia-Manero G, Santini V, Almeida A, et al. Phase III, randomized, placebo-controlled trial of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes. J Clin Oncol. 2021;39(13):1426-1436. DOI: 1200/JCO.20.02619

Newsletter

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