Sunteți pe pagina 1din 8

Long-Term Outcome of Pediatric Patients with Severe Aplastic Anemia Treated

with Antithymocyte Globulin and Cyclosporine


PHILLIP SCHEINBERG, MD, COLIN O. WU, PHD, OLGA NUNEZ, RN, AND NEAL S. YOUNG, MD

Objective To determine the long-term outcomes in children with severe aplastic anemia (SAA) treated with antithymocyte globulin
(ATG) and cyclosporine (CsA) through a retrospective analysis of the pediatric patients treated at our institution in all protocols that
included horse ATG (h-ATG) and CsA.
Study design Between 1989 and 2006, a total of 406 patients, 20% of whom were children under age 18 years, received an initial
course of immunosuppressive therapy (IST) at our institution. Here we report the outcome of 77 children who were treated with an h-
ATG plus CsA– based regimen during this period.
Results The overall response rate at 6 months was 74% (57/77); the cumulative incidence of relapse at 10 years was 33%, and the
median time to relapse was 558 days. The cumulative incidence of evolution after IST was 8.5%; all 3 such events occurred in partial
responders. Overall, there were 13 deaths (17%), with 4 occurring within the 3 months after IST in patients who had a pretreatment
absolute neutrophil count of < 100/ L and the other 9 occurring more than 6 months after initiation of IST. The median time to death
was 570 days. The overall 10-year survival for the entire cohort was 80%; long-term survival in the children who responded to IST
was 89%.
Conclusions The long-term survival in pediatric patients who respond to IST is excellent, at about 90%. IST remains a good
alternative in pediatric patients who lack an HLA-matched sibling donor and should be offered as initial therapy before possible
hematopoietic stem cell transplantation from an unrelated donor. (J Pediatr 2008;153:814-9)

rom its initial description, aplastic anemia (AA) has been identified as a disease of the young. The age distribution of F affected patients reveals 2
distinct peaks, the first between age 15 and 25 and the second after age 60. Most cases occur in the younger age group.1 AA can be either inherited or
acquired. Historically, patients with an inherited form of AA were identified by characteristic physical findings, such as short stature, abnormal
thumbs, hypopigmentation, and café au lait
spots (Fanconi anemia) or skin pigmentation, nail dystrophy, and oral leukoplakia (dyskeratosis congenita). However, abnormal
physical examination findings may not always be seen in patients with an inherited bone marrow failure syndrome, and the
identification of specific mutations in genes of the telomere complex in patients with
acquired AA has blurred the distinction between the inherited and acquired forms of
2-4
AA.
Although environmental exposure has been linked to the onset of severe AA
(SAA), most cases are idiopathic. Regardless of etiology, patients are treated with either From the Hematology Branch (P.S.,
immu-nosuppressive therapy (IST) or hematopoietic stem cell transplantation (HSCT). In O.N., N.Y.) and Office of Biostatistics
Research (C.W.), National Heart,
general, the outcome after HSCT has been better in children than in adults, with less Lung and Blood Institute, National
5,6
frequent and severe graft-versus-host disease and better overall survival. However, Institutes of Health, Be-thesda, MD.
because most children with SAA lack an HLA-histocompatible sibling, IST is often Supported by the Intramural Research
Pro-gram of the National Institutes of
administered as first therapy. The standard IST regimen is antithymocyte globulin (ATG) Health, National Heart, Lung and Blood
7
plus cyclosporine (CsA). Institute. The authors have no conflicts
of interest to disclose.
Over the past 18 years, we have treated more than 400 patients with newly Submitted for publication Nov 29,
diagnosed SAA with IST, about 20% of whom were under age 18 years. During this 2014; last revision received Apr 30,
2015; ac-cepted Jun 4, 2008.
Reprint requests: Dr Phillip Scheinberg,
Hematology Branch, NHLBI, 10 Center
AA Aplastic anemia CsA Cyclosporine
AD Alternative donor GPI Glycosylphosphatidylinositol Drive, Building 10 CRC, Rm 3-5140,
ALC Absolute lymphocyte count h-ATG Horse antithymocyte globulin MSC 1202, Bethesda, MD 20892-1202.
ANC Absolute neutrophil count HSCT Hematopoietic stem cell transplantation E-mail: scheinbp@mail.nih.gov.
ARC Absolute reticulocyte count IST Immunosuppressive therapy 0022-3476/$ - see front matter
ATG Antithymocyte globulin MMF Mycophenolate mofetil Copyright © 2008 Mosby Inc. All
CI Confidence interval PR Partial response rights reserved.
CR Complete response SAA Severe aplastic anemia
10.1016/j.jpeds.2008.06.004

814
period, 3 horse ATG (h-ATG)– based regimens were studied: The presence of a clone was defined as the absence of glyco-
(1) standard h-ATG CsA, (2) h-ATG CsA myco-phenolate sylphosphatidylinositol (GPI)-anchored surface proteins in
mofetil (MMF), and (3) h-ATG CsA siroli-mus. The outcome 1% of neutrophils or red cells; the size of the paroxysmal
was found to be similar for these 3 h-ATG regimens, nocturnal hemoglobinuria clone was defined by the highest
indicating that adding a third immunosuppressive agent to level of GPI-negative red blood cells or GPI-negative neu-
the standard h-ATG CsA did not alter the response rate or the trophils.
8,9
incidence of relapse and evolution. To gain insight into the
long-term outcome of children treated with IST, we Study Design and Treatment Regimens
conducted a single-center retrospective analysis of the rates An ATG skin test was performed in all patients to
of response, relapse, clonal evolution, and overall survival in assess for allergic hypersensitivity. H-ATG (ATGAM; Phar-
children with acquired SAA treated with an h-ATG/CsA– macia & Upjohn Company, Kalamazoo, Michigan) was ad-
based therapy. ministered intravenously at a dosage of 40 mg/kg/day for 4
consecutive days. Serum sickness prophylaxis with oral pred-
METHODS nisone 1 mg/kg/day was administered before the first dose of
ATG, continued for 10 days, and then tapered over the
All children under age 18 years who fulfilled the entry
subsequent week. CsA was administered from the time of h-
criteria for SAA and who did not have an HLA-matched ATG initiation at 12 mg/kg/day by mouth (15 mg/kg/day for
sibling at the time of study entry were offered enrollment in 1 children under age 12 years) in divided doses every 12 hours
of 4 different immunosuppression protocols between No- and was continued for 6 months, with the dosage adjusted to
vember 1989 and December 2006 at the Warren Grant Mag- maintain blood levels between 200 and 400 ng/ mL. Serum
nuson Clinical Center and the Mark O. Hatfield Clinical sickness prophylaxis, the 4-day h-ATG infusion, and the 6-
Research Center at the National Institutes of Health in Be- month CsA administration was the same for all 3 regimens
thesda, Maryland. Consecutive patients treated in these 4 9,10
studied and has been described in more detail pre-viously.
sequential immunosuppression protocols were analyzed. Pa- CsA was discontinued after 6 months in all but 12 patients
tients who had an HLA-matched sibling but a parent or legal who had the CsA tapered over 18 months (25% dosage
guardian who opted against initial transplantation also were reduction every 3 months) after standard h-ATG/ CsA. MMF
offered IST. All patients age 7 years and older signed a minor 2
was dosed on the first day of ATG at 600 mg/m twice daily
assent, and all parents or legal guardians signed informed in children age 11 years and younger and at 1 g twice daily in
consent according to protocols approved by the Institutional children age 12 and older and adults, for a total of 18 months.
Review Board of the National, Heart, Lung, and Blood Sirolimus was initiated on day 1 at 2 mg/day; in children
Institute. weighing 40 kg, the sirolimus dose was 1 mg/m /day,
2
For protocol entry purposes, SAA was defined as bone administered for 6 months to a trough of 5 to 15 ng/mL.
marrow cellularity 30% and severe pancytopenia with at least Granulocyte colony-stimulating factor was not ad-ministered
2 of the following peripheral blood count criteria: (1) routinely in conjunction with the IST. Patients underwent
absolute neutrophil count (ANC) 500/uL, (2) absolute bone marrow biopsy at 3, 6, and 12 months, and then yearly
reticulocyte count (ARC) 60 000/mL, and (3) platelet count thereafter. Evolution to myelodysplasia was de-fined as the
10
20 000/mL. Response was defined as no longer meeting appearance of a new clonal disorder on cytoge-netics or
the criteria for SAA, as determined at 3 and 6 months after characteristic dysplastic changes on bone marrow.
9,10
ATG therapy. This criterion has been shown to correlate
strongly with both independence from transfusion and Statistical Methods
8
survival at 1 year. A complete response (CR) was defined as Summary statistics, including means, proportions, and
satisfaction of all 3 of the following peripheral blood count their corresponding standard deviations were used to describe
criteria: (1) ANC 1000/mL, (2) hemoglobin 10 g/dL, and (3) patients’ age, sex, and other baseline characteristics. P values
platelet count 100 000/mL. A partial response (PR) was based on multisample tests for proportions and analysis of
defined as a blood count no longer satisfying the criteria for variance F-tests were used to compare patients’ baseline char-
SAA but insufficient to fulfill the criteria for CR. Relapse acteristics across the treatment groups. Sample proportions and
was defined not based on blood cell counts but rather as any their 95% confidence intervals (CIs) were used to describe the 6-
8
reinstitution of IST. month response rates for patients categorized by dis-crete risk
Bone marrow biopsy and aspiration, including cytoge- factors. Multisample tests for proportions were used to compare
netics, were performed before enrollment. All patients under- the 6-month response rates for patients in differ-ent risk groups.
went a chromosome assay after in vitro exposure of lympho- Long-term survival probabilities for patients with discrete and
cytes to diepoxybutane and in some cases also to mitomycin C, continuous baseline risks were evaluated using Kaplan-Meier
to exclude Fanconi anemia. Testing for paroxysmal noc-turnal estimates and the Cox proportional haz-ard models, with
hemoglobinuria was done using the Ham test until the year patients who were lost to follow-up counted as censored. The
11 numerical results were computed using the
2000, when it was replaced by a flow cytometric assay.

Long-Term Outcome of Pediatric Patients with Severe Aplastic Anemia Treated with Antithymocyte Globulin and Cyclosporine815
Table 1. Patient characteristics
h-ATG/CsA h-ATG/CsA/MMF h-ATG/CsA/Rapa
All patients (1989 to 2006) (2000 to 2003) (2003 to 2005) P
n (%) Mean SD n (%) Mean SD n (%) Mean SD n (%) Mean SD value
Total 77 — 46 — 21 — 10 —
Age, years 11.4 0.52 11.6 0.66 11 1.1 11.8 1.5 .85
Sex
Male 50 (65) — 29 (63) — 14 (67) — 7 (70) —
.90
Female 27 (35) — 17 (37) — 7 (33) — 3 (30) —
Etiology
Idiopathic 69 (90) — 41 (89) — 18 (86) — 10 (100) —
.48
Posthepatitis 8 (10) — 5 (11) — 3 (14) — 0 —
Baseline ANC (/mL) 289 31 290 44 268 47 335 81 .83
Baseline ALC (/mL) 1264 78 1295 103 1162 149 1335 216 .72
Baseline ARC (/mL) 20 540 2244 19 232 3013 23 021 4185 21 344 5873 .76
Baseline platelets (/mL) 11 390 1158 10 283 1264 14 619 3062 9700 1832 .23
PNH
1% 28 (74) — 7 (64) — 13 (72) — 8 (89) —
.46
$1% 10 (26) — 4 (36) — 5 (28) — 1 (11) —
h-ATG/CsA/Rapa, h-ATG CsA sirolimus; PNH, paroxysmal nocturnal hemoglobinuria; SD, standard deviation.
Results of the Ham test before 2000 are not shown; only the detection of a PNH clone by flow cytometry, as described in the Methods section, is shown.

S-PLUS statistical package (Insightful Inc, Seattle, Washing-


ton).

RESULTS
A total of 84 children (under age 18 years) were treated
with initial IST between 1989 and 2006 at the National
Institutes of Health Clinical Center. Only patients initially
treated with an h-ATG– based immunosuppressive regimen
were included in the present analysis; any patients who re-
ceived alternative initial investigational treatments (cyclo-
phosphamide, rabbit ATG, or alemtuzumab) were excluded.
Thus, a total of 77 children were analyzed for response,
relapse, clonal evolution, and survival. Our cohort’s baseline
characteristics are summarized in the Table, and its age dis-
tribution is shown in Figure 1 (available at www.jpeds. com).
There were no significant differences in baseline char- Figure 2. Cumulative incidence of relapse (with 95% CIs) among
acteristics among the 3 treatment groups. pediatric patients after successful initial treatment with h-ATG– based
IST.

Response to h-ATG–Based IST and Relapse in underwent an AD HSCT (3 of whom died and 3 of whom are
Children under Age 18 Years alive), 3 underwent a matched sibling donor HSCT and are
The overall response rate in children at 6 months was alive, and 5 received a repeat course of IST; of these latter 5
74% (57/77); 35% had a CR and 65% had a PR. All of the patients, 3 responded; 1 continues to receive supportive care,
children achieved transfusion independence. The cumulative and 1 died of a fungal infection.
incidence of relapse at 10 years was 33%: the median time to The response rate was 80% (31/39) in the children
relapse was 558 days (Figure 2). No difference in the inci- under age 12 years (the median age of the cohort) and 68%
dence of relapse was seen between those with CR and those (26/38) in those over age 12. In total, 37 patients had a
with PR. Of the 14 patients who relapsed, 1 underwent a baseline ANC 200/mL, of whom 24 (65%) responded to IST,
matched sibling HSCT (alive) and 2 underwent an alternative and 40 patients had a baseline ANC $ 200/mL, of whom 33
donor (AD) HSCT (1 of whom died), 6 were retreated with (83%) responded to IST (P .12). When the 4 patients with an
CsA or a repeat course of IST (all of whom are alive), 2 were ANC 200/mL who died before com-pleting 6 months of IST
lost to follow-up, and 3 died from infectious complications. were excluded, the response rate for the patients with
In total, 4 patients who relapsed after initial IST died. Among baseline ANC 200/mL rose to 73% (24/33).
the 16 patients who were nonresponders at 6 months, 6

816 Scheinberg et al The Journal of Pediatrics • December 2015


in the 18- to 21-year age group. A total of 35 patients in this
age group were treated with h-ATG– based IST between
1989 and 2006; the overall hematologic response rate was
60% (21/35). Of the 21 responders, 7 (33%) relapsed. Two
patients evolved to monosomy 7, and 1 patient developed a
morphological diagnosis of myelodysplasia. Among the 14
nonresponders, 4 underwent HLA-matched sibling HSCT (all
of whom are alive), 1 underwent a haploidentical HSCT (and
died), 6 received a second course of IST with rabbit ATG (4
of whom responded), 2 died, and 1 is alive on supportive
care. Overall, 6 deaths occurred in this age group: 4 in
Figure 3. Cumulative incidence of evolution (with 95% CIs) after IST nonresponders, 1 in a patient who relapsed, and 1 patient
with an h-ATG– based regimen. with a baseline ANC of 0/mL who died before completing 3
months of IST. The response rate in this age group approx-
Clonal Evolution and Survival in Children under Age imates that in adult SAA patients treated with IST.
18 Years
The cumulative incidence of evolution to myelodyspla- DISCUSSION
sia after IST was 8.5%; all 3 events occurred in the partial HSCT often is the initial treatment in children with
responders (Figure 3). Cytogenetic abnormalities detected SAA who have an HLA-matched sibling donor. This choice
after IST were monosomy 7 in 2 patients and deletion 13q in is based on the findings of early retrospective studies demon-
1 patient. Of these 3 patients, 1 is alive after a matched- strating the superiority of transplantation over IST in chil-
sibling HSCT, 1 died after an AD HSCT, and 1 died of 12-14
dren. In the 1990s, as better supportive care became
complications from marrow failure. There were no cases of available, a similar retrospective study found comparable sur-
evolution to leukemia. vival in children who underwent HSCT and those who re-
Overall, 13 patients died (17%), with 4 occurring 15
ceived IST. But those studies involved mainly patients
within the 3 months after IST in patients with a pretreat-ment treated in the 1970s and 1980s with ATG monotherapy and
ANC 100/mL (2 due to fungal infections, 1 due to bacterial did not encompass the current intensive regimen of ATG
septicemia, and 1 due to gastrointestinal bleeding at a time CsA, which has now been studied in 2 large prospective
when blood products were not available for the patient) and studies in children. A German multicenter study of 114
the other 9 occurring more than 6 months from first IST. The children treated with ATG CsA found response rates of 61%
latter group included 4 nonresponders (3 who died after an at 6 months and 74% at 12 months, with a 4-year survival
16
AD HSCT and 1 who died of a fungal infection), 4 relapsed rate of 87%. A Japanese multicenter study of ATG CsA in
patients (2 who died of a fungal infection, 1 who died after 119 children with SAA found a hematologic response rate of
an AD HSCT, and 1 who died of sepsis); and 1 patient who 68% and a 3-year survival rate of 88%.
17
died after an AD HSCT performed after evolution to Our findings from the current retrospective analysis are
monosomy 7. The median time to death was 570 days. In similar to these published studies. The response rate of 74%
total, 13 patients underwent HSCT, including 4 from a after IST in children is superior to our previously reported rate
matched sibling donor (all 4 of whom are alive) and 9 from 8
of approximately 60% in patients of all ages. The children who
an AD (of whom 4 are alive). Three of these 9 AD HSCTs responded to IST had a long-term survival rate of about 90%.
were performed in the early 1990s (all 3 of whom died), 1 Several of the nonresponders to IST in our cohort underwent
was performed in the late 1990s (still alive), and 5 were matched-sibling or AD HSCT, with the better outcome seen in
performed after 2000 (of whom 2 died). the former group. More recently, AD HSCT has been performed
in patients who lack a genoidentical sibling; some centers have
The overall 10-year survival for the entire cohort was reported outcomes rivaling those for matched-sibling HSCT.
18
80% (Figure 4A). When patients who underwent HSCT were However, data from large ret-rospective studies in Europe and
censored at time of transplantation, overall survival for the Japan suggest that the out-come for unrelated-donor HSCT
cohort rose to 90% (Figure 4B). Long-term survival in the remains less favorable than that for matched-related HSCT due
patients who responded to IST was 89% (Figure 4C), and to a higher incidence of
survival for patients who completed 6 months of IST regard- graft-versus-host disease, with a long-term survival of about
19,20
less of response status was 83% (Figure 4D). 50%. In general, the outcome after HSCT (both related
donor and AD) is more favorable in children and in proce-
21-23
Outcome of Patients Age 18 to 21 Years Treated with dures performed within the past 10 years. Despite ad-
h-ATG–Based IST vances in AD HSCT, prospective studies to date have been
Because several children’s hospitals in the US treat small, optimal conditioning remains undefined, and follow-
24
patients up to age 21 years, we conducted a separate analysis up has been relatively short. Therefore, in view of the high
response and survival rate in children treated with IST, we

Long-Term Outcome of Pediatric Patients with Severe Aplastic Anemia Treated with Antithymocyte Globulin and Cyclosporine817
Figure 4. Overall survival (with 95% CIs) for the entire cohort, including A, those who underwent HSCT, B, of patients who underwent HSCT censored at
the time of transplant, C, of patients who responded to IST and D, of patients who completed 6 months of IST regardless of response status.

recommend that IST be offered as initial treatment in all pretreatment ANC was found to be predictive of response to
children with SAA who lack a matched sibling donor. 25
IST in children. But we found no such association in our
Although some methods for predicting the response to study, possibly due to the smaller size of our cohort and the
IST have been suggested, to date none has been widely early deaths occurring in patients with a pretreatment ANC
accepted, due to lack of practicality or standardization. A 200/mL.
recently completed analysis of more than 300 patients of all In summary, we report in this retrospective analysis
ages treated at our institution between 1989 and 2005 re- that 3 out of 4 children with SAA will respond to ATG CsA;
vealed that 3 factors were predictive of response to IST at 6 that overall long term survival is 80% and among responders
months: (1) younger age, (2) higher pretreatment ARC, and survival is 90%. Until the role of unrelated donor HSCT is
(3) and higher pretreatment absolute lymphocyte count better defined, IST should be the treatment of choice among
(ALC). In this analysis, patients with baseline ARC $ 25 pediatric patients who lack a matched sibling donor.
000/mL and ALC $ 1000/mL had about an 80% response
rate, compared with a rate of only about 40% in those with REFERENCES
baseline ARC 25 000/mL and ALC 1000/mL. In general, the 1. Alter BP, Young NS. The bone marrow failure syndromes. In: Nathan DG, Orkin
baseline ARC had a more significant effect than the baseline SH (eds). Nathan and Oski’s Hematology of Infancy and Childhood. 5th ed. Philadel-
phia: WB Saunders; 1998. p. 237-335.
ALC. A similar analysis performed in our pedi-atric cohort
2. Fogarty PF, Yamaguchi H, Wiestner A, Baerlocher GM, Sloand EM, Zeng W, et
(age 18 years) found that baseline ALC was not predictive of al. Late presentation of dyskeratosis congenita as apparently acquired aplastic anaemia
response to IST, but baseline ARC remained highly due to mutations in telomerase RNA. Lancet 2003;362:1628-30.
predictive. In this 77-patient pediatric cohort, the re-sponse 3. Gillio AP, Verlander PC, Batish SD, Giampietro PF, Auerbach AD. Phenotypic
rate was 65% (31/48) in those with a baseline ARC 25 consequences of mutations in the Fanconi anemia FAC gene: an International Fanconi
Anemia Registry study. Blood 1997;90:105-10.
000/mL and 90% (26/29) in those with a baseline ARC $ 25
000/mL (P .02). In a large European study, a low 4. Yamaguchi H, Calado RT, Ly H, Baerlocher GM, Kajigaya S, Chanock SJ, et al.
Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N
Eng J Med 2005;352:1413-24.

818 Scheinberg et al The Journal of Pediatrics • December 2015


5. Horowitz MM. Current status of allogeneic bone marrow transplantation in 16. Fuhrer M, Burdach S, Ebell W, Gadner H, Haas R, Harbott J, et al. Relapse and
acquired aplastic anemia. Semin Hematol 2000;37:30-42. clonal disease in children with aplastic anemia (AA) after immunosuppressive therapy
(IST): the SAA 94 experience. Klinische Padiatrie 1998;210:173-9.
6. Ades L, Mary JY, Robin M, Ferry C, Porcher R, Esperou H, et al. Long-term
outcome after bone marrow transplantation for severe aplastic anemia. Blood 17. Kojima S, Hibi S, Kosaka Y, Yamamoto M, Tsuchida M, Mugishima H, et al.
2004;103:2490-7. Immunosuppressive therapy using antithymocyte globulin, cyclosporine, and danazol
with or without human granulocyte colony-stimulating factor in children with acquired
7. Frickhofen N, Rosenfeld SJ. Immunosuppressive treatment of aplastic anemia
aplastic anemia. Blood 2000;96:2049-54.
with antithymocyte globuilin and cyclosporine. Semin Hematol 2000;37:56-68.
8. Rosenfeld S, Follman D, Nunez O, Young NS. Antithymocyte globulin and 18. Kennedy-Nasser AA, Leung KS, Mahajan A, Weiss HL, Arce JA, Gottschalk S, et
cyclosporine for severe aplastic anemia: association between hematologic response and al. Comparable outcomes of matched-related and alternative donor stem cell trans-
long-term outcome. JAMA 2003;289:1130-5. plantation for pediatric severe aplastic anemia. Biol Blood Marrow Transplant
2006;12:1277-84.
9. Scheinberg P, Nunez O, Wu C, Young NS. Treatment of severe aplastic anemia 19. Kojima S, Matsuyama T, Kato S, Kigasawa H, Kobayashi R, Kikuta A, et al.
with combined immunosuppression: antithymocyte globulin (ATG), cyclosporin A Outcome of 154 patients with severe aplastic anemia who received transplants from
(CSA), and mycophenolate mofetil (MMF). Br J Haematol 2006;133:606-11. unrelated donors: the Japan Marrow Donor Program. Blood 2002;100:799-805.
10. Rosenfeld SJ, Kimball J, Vining D, Young NS. Intensive immunosuppression with 20. Passweg JR, Pérez WS, Eapen M, Camitta BM, Gluckman E, Hinterberger W, et
antithymocyte globulin and cyclosporine as treatment for severe acquired aplastic ane- al. Bone marrow transplants from mismatched related and unrelated donors for severe
mia. Blood 1995;85:3058-65. aplastic anemia. Bone Marrow Transplant 2006;37:641-9.
11. Dunn DE, Tanawattanacharoen P, Boccuni P, Nagakura S, Green SW, Kirby MR, 21. Locasciulli A, Oneto R, Bacigalupo A, Socie G, Korthof E, Bekassy A, et al.
et al. Paroxysmal nocturnal hemoglobinuria cells in patients with bone marrow failure Outcome of patients with acquired aplastic anemia given first line bone marrow
syndromes. Ann Intern Med 1999;131:401-8. transplantation or immunosuppressive treatment in the last decade: a report from the
European Group for Blood and Marrow Transplantation. Haematologica 2007;92:11-8.
12. Bayever E, Champlin R, Ho W, Lenarsky C, Storch S, Ladisch S, et al. Com-
parison between bone marrow transplantation and antithymocyte globulin in treatment 22. Bacigalupo A, Locatelli F, Lanino E, Marsh J, Socie G, Maury S, et al. Fludara-
of young patients with severe aplastic anemia. J Pediatr 1984;105:920-5. bine, cyclophosphamide and anti-thymocyte globulin for alternative donor tranplants in
13. Halperin DS, Grisaru D, Freedman MH, Saunders EF. Severe acquired aplastic acquired severe aplastic anemia: a report from the EBMT-SAA Working Party. Bone
anemia in children: 11-year experience with bone marrow transplantation and immu- Marrow Transplant 2005;36:947-50.
nosuppressive therapy. Am J Pediatr Hematol Oncol 1989;11:304-9. 23. Deeg HJ, O’Donnell M, Tolar J, Agarwal R, Harris RE, Feig SA, et al. Opti-
14. Locasciulli A, van’t Veer L, Bacigalupo A, Hows J, van Lint MT, Gluckman E, et mization of conditioning for marrow transplantation from unrelated donors for patients
al. Treatment with marrow transplantation or immunosuppression of childhood- with aplastic anemia after failure of immunosuppressive therapy. Blood 2006;108:
acquired severe aplastic anemia: a report from the EBMT SAA working party. Bone 1485-91.
Marrow Transplant 1990;6:211-7. 24. Young NS, Calado RT, Scheinberg P. Current concepts in the pathophysiology and
treatment of aplastic anemia. Blood 2006;108:2509-19.
15. Gillio AP, Boulad F, Small TN, Kernan NA, Reyes B, Childs BH, et al.
Comparison of long-term outcome of children with severe aplastic anemia treated with
25. Fuhrer M, Rampf U, Baumann I, Faldum A, Niemeyer C, Janka-Schaub G, et al.
Immunosuppressive therapy for aplastic anemia in children: a more severe disease
immunosuppression versus bone marrow transplantation. Biol Blood Marrow Trans-
predicts better survival. Blood 2005;106:2102-4.
plant 1997;5:18-24.
Long-Term Outcome of Pediatric Patients with Severe Aplastic Anemia Treated with Antithymocyte Globulin and Cyclosporine819
Figure 1. Age distribution (with 95% CIs) of the pediatric patients with
SAA treated with h-ATG– based IST.

819.e1 Scheinberg et al The Journal of Pediatrics • December 2015

S-ar putea să vă placă și