In contrast to severe aplastic anemia (sAA), the appropriate management of patients with moderate pancytopenia is unclear. In this study, we examined the efficacy of a humanized monoclonal antibody recognizing interleukin-2 receptor (daclizumab), which has proven to be a successful immunosuppressive agent in solid organ and bone marrow transplantation. We treated 17 patients with moderate aplastic anemia (mAA) with 1 mg/kg every 2 weeks for 3 months. mAA was defined as depression of 2 of the 3 blood counts: absolute neutrophil count 1200/mm3 or less, platelet count 70 000/mm3 or less, hemoglobin level 8.5 g/dL or lower, and absolute reticulocyte count 60 000/mm3 or less. The primary end point of our protocol was a hematologic response in at least one affected peripheral blood value. Daclizumab had little toxicity. Six of the 16 (38%) evaluable patients responded to treatment. Two patients with previously chronic disease showed complete return of normal counts, which were sustained for more than 2 years following treatment. Four patients had single-lineage responses. Two previously transfusion-dependent patients became transfusion independent; one patient with many neutropenia-related infections had a normal neutrophil count following treatment. Daclizumab appears safe; its efficacy in this pilot protocol suggests that expanded study of this monoclonal antibody in immune-mediated bone marrow failure syndrome is warranted. (Blood. 2003; 102:3584-3586)

Immunosuppression with antithymocyte globulin (ATG) and cyclosporine (CsA) has been extraordinarily successful in producing clinical responses and prolonging survival of patients with aplastic anemia (AA).1  Intensive immunosuppressive treatment, however, is associated with significant toxicity and requires hospitalization and considerable supportive care. Although such treatment is appropriate in patients with severe aplastic anemia (sAA; defined as absolute neutrophil count [ANC] ≤ 500/mm3, platelet count ≤ 20 000/mm3, and absolute reticulocyte count ≤ 60 000/mm3), who require urgent management, it may be argued that patients with moderate or nonsevere aplastic anemia (mAA) may be observed for prolonged periods of time without definitive treatment.2-4  However, failure to treat might subject the patient's bone marrow to continued immune attack and lead to progressive loss of progenitor cells over time, ultimately resulting in the development of severe disease.5  Patients with mAA received less aggressive modalities such as androgens and growth factors,6-14  but these treatments do not clearly address the immune pathology and may only result in improvement in cytopenias without sparing progenitor cells. mAA has been treated with CsA alone12,15  and although good response rates have been achieved, patients are required to take the drug for substantial periods and frequently have a relapse following its discontinuation. In addition, irreversible renal pathology is a risk of therapy.

Daclizumab is a genetically engineered human IgG1 incorporating the antigen-binding regions of a parent murine monoclonal antibody (mAb) that specifically recognizes the 55-kDa α chain of the heterotrimeric interleukin 2 receptor (IL-2R).16  IL-2R is present only on activated T cells and is required for their clonal expansion and continued viability; blockade of IL-2R results in decreased activation and proliferation of T-cell clones. The mechanism of action of anti-IL-2R mAb is believed to be similar to that of ATG. Daclizumab has had few toxicities when used to treat patients rejecting solid organ transplants. Thus, daclizumab is an attractive, less toxic alternative to ATG.

In this study we treated a group of patients with mAA with daclizumab, assessing its toxicity, efficacy, and biologic effects.

Patients

Eligible patients with mAA were entered into the study after obtaining informed consent according to protocol approved by the Institutional Review Board of the National, Heart, Lung and Blood Institute (Bethesda, MD). We treated all consecutive patients over the age of 7 years with mAA, defined according to the Consensus Conference on Treatment of Aplastic Anemia,2  by a hypocellular bone marrow (cellularity < 30%) and depression of at least 2 of 3 blood counts below the normal values: ANC 1200/mm3 or less, platelet count ≤ 70 000/mm3 or less, anemia with hemoglobin level 8.5 g/dL or less, and absolute reticulocyte count 60 000/mm3 or less. Patients with severe pancytopenia were excluded from the protocol. The average of 3 measurements of blood counts obtained within a 2-week period prior to enrollment into the study was used to assess study eligibility. Transfusion independence was not an exclusion criterion. For the purpose of this study, transfusion dependence was defined as requirements of at least 2 U red blood cells (RBCs) or 5 U platelets per month for a period of 2 months prior to enrollment. We excluded patients with a current diagnosis or past history of myelodysplastic syndrome, abnormal cytogenetics, Fanconi anemia, or other congenital forms of AA.

Patients with a history of initial failure to respond to ATG, antilymphocyte globulin (ALG), or another immunosuppressive agent were also excluded. Patients received 1 mg/kg daclizumab infused intravenously every other week for a total of 5 doses. Response was defined at 3 months by the criteria delineated in Table 1; the primary end point of this trial was a hematologic response in at least one affected peripheral blood parameter as determined by 3 measurements in the first 12 weeks after completion of the study. Change in the transfusion requirements served as a secondary end point. Patients had a bone marrow evaluation with cytogenetics prior to and 3 months following treatment. During the course of the study weekly blood counts were obtained.

Table 1.

Criteria for response


Neutrophils 

 
If baseline ANC below 500/mm3  Increase in ANC ≥ 300/mm3 
If baseline ANC above 500/mm3  Increase in ANC ≥ 500/mm3 
Platelets  
If baseline platelet count 70 000/mm3 or below but above 50 000/mm3  Increase in platelet count ≥ 30 000/mm3 
If baseline platelet count 50 000/mm3 or below   Increase in platelet count ≥ 20 000/mm3 blood  
Transfusion dependence  
Transfusion independence   No transfusions for > 8 wk  
Decreased requirement
 
Decrease in transfusion number over time >50% for either RBC, platelets, or both
 

Neutrophils 

 
If baseline ANC below 500/mm3  Increase in ANC ≥ 300/mm3 
If baseline ANC above 500/mm3  Increase in ANC ≥ 500/mm3 
Platelets  
If baseline platelet count 70 000/mm3 or below but above 50 000/mm3  Increase in platelet count ≥ 30 000/mm3 
If baseline platelet count 50 000/mm3 or below   Increase in platelet count ≥ 20 000/mm3 blood  
Transfusion dependence  
Transfusion independence   No transfusions for > 8 wk  
Decreased requirement
 
Decrease in transfusion number over time >50% for either RBC, platelets, or both
 

Flow cytometry

Intracellular staining for interferon γ (IFN-γ) expression was performed using the PharMingen intracellular staining kit (San Diego, CA).17  Double-color surface staining was first performed with phycoerythrin (PE)-conjugated anti-CD4 and anti-CD8 mAbs and then cells were permeabilized using a saponin-based method (PharMingen) and stained with fluorescein isothiocyanate (FITC)-anti-IFN-γ or IL-4 mAbs (PharMingen; BioSource, Camarillo, CA). Specificity of the antibody was confirmed by showing elimination of staining with blocking antibody. To perform this test 10 μg of a purified unconjugated antibody was added to the fixed/permeabilized cells, which was then incubated for 20 minutes prior to addition of the conjugated antibody. Samples were analyzed using the Coulter EPICS V flow cytometer (Hialeah, FL). Lymphocytes were gated initially by forward scatter/side scatter; secondary gates were set based on staining with isotypic control mAbs such that less than 1% of cells stained positive. Staining was compared to that for 30 normal controls. Results of 30 normal controls as well as the reproducibility of the technique were previously published by our laboaratory.17 

Six of 16 evaluable patients (38%) with mAA responded to treatment within 90 days of receiving the last dose of daclizumab (Table 2; Figure 1). One patient, lost to follow-up, was excluded from analysis and was not evaluated at the 3-month evaluation period. Of the 16 evaluable patients with mAA (one patient was lost to follow-up), 3 were newly diagnosed and 13 had chronic mAA. Patients no. 1 and 2 with previously chronic disease showed complete return of normal blood counts within the first 4 months of receiving daclizumab. Single-lineage responses were seen in 3 patients with chronic disease (patients no. 3, 9, and 16). Two patients (nos. 1 and 9) previously dependent on RBC transfusions no longer require transfusions after more than 2 years of follow-up. Another patient (no.3), who had experienced many neutropenia-related infections, now has a normal white blood cell count. Only one patient progressed despite treatment and was later treated with ATG/CsA and responded. Although follow-up time was limited, only one patient had a relapse, but she responded promptly to further daclizumab treatments. Daclizumab was associated with no toxicity except for a mild first infusion reaction in one patient (itching), which did not require treatment and did not recur. As in a previous study of sAA,17  CD8 cells from 11 patients were tested for the presence of IFN-γ; 5 of the 6 testing positive by the assay responded to daclizumab, whereas all 5 patients testing negative failed to clinically respond to daclizumab (Figure 2).

Table 2.

Patient characteristics


Patient no.




ANC/mm3

Platelet count/mm3

Reticulocyte count/mm3



Follow-up, mo
Age, y
Ethnicity
Sex
Before
After
Before
After
Before
After
TI*
Response
1   57   White   F   1852   3148   45   78   55   134   T1   CR at 5 mo   31  
2   43   White   M   670   1360   45   98   49   45    CR at 4 mo   32  
3   58   White   F   667   1490   78   80   57   55    PR   25  
4   14   Asian   F   2832   ND   5   ND   4   ND    LTF   LTF  
5   53   White   F   888   786   55   61   96   66    NR   25  
6   43   Hispanic   F   1200   1398   65   32   119   153    NR   24  
7   41   White   M   625   494   34   39   72   79    NR   23  
8   30   White   M   616   584   24   33   53   51    NR   22  
9   9   White   F   1088   2400   35   30   22   134   T1   PR   21  
10   37   White   F   3012   3229   33   26   21   28    NR   19  
11   40   Hispanic   F   760   1124   18   20   58   59    NR   17  
12   30   Hispanic   M   1068   1225   115   111   22   22    NR   20  
13   25   White   M   1293   1060   33   36   71   102    NR   4  
14   26   White   M   804   928   23   22   34   36    NR   10  
15   67   Asian   M   800   100   47   57   70   115    PR   13  
16   38   White   F   1134   1800   51   51   35   105    PR   11  
17
 
24
 
Asian
 
F
 
1378
 
500
 
25
 
24
 
72
 
65
 

 
NR
 
20
 

Patient no.




ANC/mm3

Platelet count/mm3

Reticulocyte count/mm3



Follow-up, mo
Age, y
Ethnicity
Sex
Before
After
Before
After
Before
After
TI*
Response
1   57   White   F   1852   3148   45   78   55   134   T1   CR at 5 mo   31  
2   43   White   M   670   1360   45   98   49   45    CR at 4 mo   32  
3   58   White   F   667   1490   78   80   57   55    PR   25  
4   14   Asian   F   2832   ND   5   ND   4   ND    LTF   LTF  
5   53   White   F   888   786   55   61   96   66    NR   25  
6   43   Hispanic   F   1200   1398   65   32   119   153    NR   24  
7   41   White   M   625   494   34   39   72   79    NR   23  
8   30   White   M   616   584   24   33   53   51    NR   22  
9   9   White   F   1088   2400   35   30   22   134   T1   PR   21  
10   37   White   F   3012   3229   33   26   21   28    NR   19  
11   40   Hispanic   F   760   1124   18   20   58   59    NR   17  
12   30   Hispanic   M   1068   1225   115   111   22   22    NR   20  
13   25   White   M   1293   1060   33   36   71   102    NR   4  
14   26   White   M   804   928   23   22   34   36    NR   10  
15   67   Asian   M   800   100   47   57   70   115    PR   13  
16   38   White   F   1134   1800   51   51   35   105    PR   11  
17
 
24
 
Asian
 
F
 
1378
 
500
 
25
 
24
 
72
 
65
 

 
NR
 
20
 

Seventeen patients with mAA were treated with daclizumab. Of these, 16 were evaluable; patient no. 5 was lost to follow-up before the 3-month evaluation period. The ANC, absolute reticulocyte count, and platelet counts on entry to the study are shown.

Blood counts are the average of 3 consecutive measurements performed prior to study entry and 3 months after the last dose of daclizumab (first course as in the case of patient 1).

TI indicates transfusion independence; CR, complete response; PR, partial response; ND, not done; LTF, lost to follow-up; NR, no response.

*

Transfusion independence in patients who were previously transfusion dependent.

Figure 1.

Response of patients with mAA to daclizumab. Serial blood counts of patients treated with daclizumab. Each panel represents data from 1 patient. Platelets and red cells were given to Patient 1 (panel A) for bleeding.

Figure 1.

Response of patients with mAA to daclizumab. Serial blood counts of patients treated with daclizumab. Each panel represents data from 1 patient. Platelets and red cells were given to Patient 1 (panel A) for bleeding.

Close modal
Figure 2.

IFN-γ staining of peripheral blood lymphocytes of responding and nonresponding patients with mAA. Lymphocytes from 11 previously untreated patients with mAA were surface stained with CD4-tricolor (phycoerythrin-cyanine 5) and CD8-PE and intracellularly stained with IFN-γ-FITC. Patients with mAA received daclizumab therapy and were designated as responders or nonresponders based on improvement of bone marrow function by 3 months following treatment. The figure shows the ratio of MCFsample/MCFisotype of staining of peripheral blood lymphocytes. Values more than 2 SD above the mean of normal were said to be positive for IFN-γ.17  MCF indicates mean channel fluorescence.

Figure 2.

IFN-γ staining of peripheral blood lymphocytes of responding and nonresponding patients with mAA. Lymphocytes from 11 previously untreated patients with mAA were surface stained with CD4-tricolor (phycoerythrin-cyanine 5) and CD8-PE and intracellularly stained with IFN-γ-FITC. Patients with mAA received daclizumab therapy and were designated as responders or nonresponders based on improvement of bone marrow function by 3 months following treatment. The figure shows the ratio of MCFsample/MCFisotype of staining of peripheral blood lymphocytes. Values more than 2 SD above the mean of normal were said to be positive for IFN-γ.17  MCF indicates mean channel fluorescence.

Close modal

The goal of immunosuppressive regimens in AA is elimination of T cells effecting the immune-mediated destruction of hematopoietic progenitor and stem cells. Although this study included only patients who were symptomatic from their bone marrow failure, immunosuppressive therapy, if sufficiently innocuous and also convenient, could potentially be used to prevent stem cell depletion in patients with evidence of immune-mediated bone marrow destruction in an early phase of hematologic disease. The relapse rate after treatment with daclizumab is not known at this point, but the one relapse reported here might be indicative of the need for further immunosuppression in some patients. Further long-term studies are needed both to determine systematically the effect of early treatment on the course of mAA, as well as to establish the optimal dosing regimen for daclizumab and the need for further immunosuppression. Other roles for daclizumab in AA can now be envisioned including treatment of relapsed sAA, in myelodysplastic syndrome, for pure RBC aplasia and other single-lineage failure diseases, and as a replacement for CsA in treatment-dependent patients with sAA where CsA is contraindicated or has shown toxicity.

Prepublished online as Blood First Edition Paper, July 24, 2003; DOI 10.1182/blood-2003-04-1032.

J.P.M. and E.M.S. contributed equally to this study.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Rosenfeld SJ, Kimball J, Vining D, Young NS. Intensive immunosuppression with antithymocyte globulin and cyclosporine as treatment for severe acquired aplastic anemia.
Blood.
1995
;
85
:
3058
-3065.
2
Kojima S, Nakao S, Tomonaga M, et al. Consensus conference on the treatment of aplastic anemia.
Int J Hematol.
2000
;
72
:
118
-123.
3
Ohara A, Kojima S, Hamajima N, et al. Myelodysplastic syndrome and acute myelogenous leukemia as a late clonal complication in children with acquired aplastic anemia.
Blood.
1997
;
90
:
1009
-1013.
4
Shahidi NT. Aplastic anaemia: aetiological and therapeutic dilemmas.
Br J Haematol.
1979
;
43
:
163
-165.
5
Khatib Z, Wilimas J, Wang W. Outcome of moderate aplastic anemia in children.
Am J Pediatr Hematol Oncol.
1994
;
16
:
80
-85.
6
Kurzrock R, Talpaz M, Salewski E, Gutterman JU. Phase I study of recombinant human interleukin-3 in patients with bone marrow failure.
J Clin Oncol.
1991
;
9
:
1241
-1250.
7
Kurzrock R, Talpaz M, Gomez JA, et al. Differential dose-related haematological effects of GM-CSF in pancytopenia: evidence supporting the advantage of low-over high-dose administration in selected patients.
Br J Haematol.
1991
;
78
:
352
-358.
8
Kurzrock R, Talpaz M, Gutterman JU. Very low doses of GM-CSF administered alone or with erythropoietin in aplastic anemia.
Am J Med.
1992
;
93
:
41
-48.
9
Kurzrock R, Cortes J, Thomas DA, et al. Pilot study of low-dose interleukin-11 in patients with bone marrow failure.
J Clin Oncol.
2001
;
19
:
4165
-4172.
10
Bennett JM, Young MS, Liesveld JL, et al. Phase II study of combination human recombinant GM-CSF with intermediate-dose cytarabine and mitoxantrone chemotherapy in patients with high-risk myelodysplastic syndromes (RAEB, RAEBT, and CMML): an Eastern Cooperative Oncology Group Study.
Am J Hematol.
2001
;
66
:
23
-27.
11
Maschan A, Bogatcheva N, Kryjanovskii O, et al. Results at a single centre of immunosuppression with cyclosporine A in 66 children with aplastic anaemia.
Br J Haematol.
1999
;
106
:
967
-970.
12
Bessho M, Hirashima K, Asano S, et al. Treatment of the anemia of aplastic anemia patients with recombinant human erythropoietin in combination with granulocyte colony-stimulating factor: a multicenter randomized controlled study.
Eur J Haematol.
1997
;
58
:
265
-272.
13
Young N, Griffith P, Brittain E, et al. A multicenter trial of anti-thymocyte globulin in aplastic anemia and related diseases.
Blood.
1988
;
72
:
1861
-1869.
14
Kojima S, Fukuda M, Miyajima Y, Matsuyama T, Horibe K. Treatment of aplastic anemia in children with recombinant human granulocyte colony-stimulating factor.
Blood.
1991
;
77
:
937
-941.
15
Marsh J, Schrezenmeier H, Marin P, et al. Prospective randomized multicenter study comparing cyclosporin alone versus the combination of antithymocyte globulin and cyclosporin for treatment of patients with nonsevere aplastic anemia: a report from the European Blood and Marrow Transplant (EBMT) Severe Aplastic Anaemia Working Party.
Blood.
1999
;
93
:
2191
-2195.
16
Carswell CI, Plosker GL, Wagstaff AJ. Daclizumab: a review of its use in the management of organ transplantation.
BioDrugs.
2001
;
15
:
745
-773.
17
Sloand E, Kim S, Maciejewski JP. et al. Intracellular interferon-gamma in circulating and marrow T cells detected by flow cytometry and the response to immunosuppressive therapy in patients with aplastic anemia.
Blood.
2002
;
100
:
1185
-1191.
Sign in via your Institution