• No results found

Incidence, Predisposing Factors, and Outcome of Engraftment Syndrome in Pediatric Allogeneic Stem Cell Transplant Recipients

N/A
N/A
Protected

Academic year: 2021

Share "Incidence, Predisposing Factors, and Outcome of Engraftment Syndrome in Pediatric Allogeneic Stem Cell Transplant Recipients"

Copied!
7
0
0

Loading.... (view fulltext now)

Full text

(1)

Incidence, Predisposing Factors, and Outcome of

Engraftment Syndrome in Pediatric Allogeneic

Stem Cell Transplant Recipients

Irene Schmid,1Daniel Stachel,2Philipp Pagel,3Michael H. Albert1

1Pediatric Hematology and Oncology, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-Universita¨t, Munich,

Germany;2Pediatric Hematology and Oncology, Universita¨t Erlangen-Nu¨rnberg, Erlangen, Germany; and3Lehrstuhl fu¨r Genomorientierte Bioinformatik, Technische Universita¨t Mu¨nchen, Munich, Germany

Correspondence and reprint requests: Irene Schmid, MD, Pediatric Hematology and Oncology, Dr. von Haunersches Kinderspital, Ludwig-Maximilians-University Munich, Lindwurmstr.4, D-80337 Munich, Germany (e-mail:Irene. Schmid@med.uni-muenchen.de).

Received November 4, 2007; accepted February 4, 2008

ABSTRACT

Engraftment syndrome (ES) has been recognized as an inflammatory condition during neutrophil recovery after hematopoietic stem cell transplantation (HSCT) characterized by noninfectious fever and skin rash. It has been reported to occur frequently after autologous HSCT in children and adults, and has been shown to be an inde-pendent risk factor for increased transplant-related mortality (TRM). However, virtually no data exist on its oc-currence after allogeneic HSCT in children. To determine incidence, predisposing factors for, and complications of ES in a pediatric transplant cohort, we analyzed 61 consecutive recipients of a myeloablative allogeneic HSCT for the occurrence of ES. Diagnosis of ES was established when children presented with $2 of the following symptoms within 7 days before engraftment: (1) fever .38.0C, (2) skin rash, (3) weight gain

and albumin drop, or (4) dyspnea, hypoxia, and pulmonary infiltrates. Incidence of ES in this cohort was 48% (29 of 61). In a univariate analysis, posttransplant granulocyte-colony stimulating factor (G-CSF) administration (P 5 .02), and high mononuclear cell count (MNC) (P 5 .002) were identified as significant risk factors predis-posing for the development of ES. In a multiple logistic regression analysis, amphotericin B therapy (P 5 .009) and high MNC (P 5 .004) were significant explanatory variables for ES risk. There was a slight trend toward a higher rate of chronic GVHD (cGVHD) in patients with ES (P 5 .11). However, after a median follow-up of 9.5 years overall survival (OS) (P 5 .53) and TRM (P 5 .65) did not differ between the 2 groups. ES presenting with fever, rash, weight gain, and pulmonary symptoms should be recognized as a frequent complication of al-logeneic HSCT after myeloablative conditioning in children. Treatment with G-CSF, amphotericin B, and a high nucleated cell count of the graft predisposed for the development of ES in this study. OS and TRM in this cohort were not affected by the occurrence of ES.

Ó 2008 American Society for Blood and Marrow Transplantation KEY WORDS

Pediatric  Allogeneic stem cell transplantation  Engraftment syndrome  Skin rash  Capil-lary leakage syndrome  Pulmonary infiltrates

INTRODUCTION

Engraftment syndrome (ES) or autoaggression graft-versus-host (GVHD)-like syndrome describes a characteristic constellation of symptoms that occur during neutrophil recovery after both allogeneic and autologous hematopoietic stem cell transplantation (HSCT) [1]. The syndrome consistently includes a maculopapular skin rash and noninfectious fever, and in some definitions also weight gain and

pulmo-nary edema with hypoxemia not attributable to pulmonary hemorrhage, fluid overload, infection, thromboembolism, or cardiac disease. Because there is no generally accepted standard definition, the re-ported incidence for ES has varied widely ranging from 5% to 72%[2-14].

The pathogenesis of ES remains unknown. It is thought that cytokines released during neutrophil re-covery and products of neutrophil degranulation and

438

(2)

oxidative metabolism lead to local and systemic tissue injury[1,5,15,16]. The ensuing detrimental effects re-sult in pulmonary injury where the neutrophils may be initially trapped, noninfectious fever, and a diffuse in-crease in capillary leakage. In autologous transplanta-tion the skin rash mimics autoinflammatory acute GVHD (aGVHD)[17]. Whether this rash that coin-cides with the appearance of autoreactive T cells di-rected at histocompatibility complex class II antigens is because of an auto-GVHD or part of the inflamma-tory ES remains unknown. Past observations, however, do suggest that there is an important clinical and path-ophysiologic overlap of ES and autologous GVHD

[17,18].

In adults, a wide variety of risk factors for the de-velopment of ES have been identified in different stud-ies including breast cancer as underlying disease[7,17], a higher number of previous treatment regimens[17], a higher number of hematopoetic cells infused

[3,4,19], peripheral blood as source of stem cells

[2,3], the use of G-CSF [2,3,19], and GM-CSF [7],

early and steep neutrophil recovery[3,4], nonmyeloa-blative conditioning regimens [8], patient age [8], treatment with amphotericin B [8], a female gender

[3,8], and autologous transplantation [8]. In 1 study

of reduced intensity conditioning allogeneic trans-plants the occurrence of ES was associated with a sig-nificantly higher transplant-related mortality (TRM) (49% versus 16%) and shorter median survival (168 versus 418 days)[8].

In children, there are very few studies analyzing ES. In autologous peripheral blood stem cell transplanta-tion, ES—as defined by fever and skin rash—was re-ported to be a frequent event occurring in 30 of 156 patients (19.2%), and was found to be the only signif-icant variable that influenced transplant related mortality[10,20]. In the only prospective analysis in au-tologous peripheral blood stem cell recipients a high number of CD341 cells/kg infused, patients trans-planted in early phase, the type of malignancy (solid tu-mor), and conditioning regimens other than busulfan were risk factors for ES, which was diagnosed in 38 of 112 of patients (34%)[13]. The study of Nu¨rnberger et al. [12] investigated exclusively the occurrence of capillary leakage defined as weight gain unresponsive to furosemide. This study also contained 54 allogeneic transplantations, where an increased incidence in unre-lated compared to reunre-lated donor transplantation was found but influence on outcome was not analyzed.

Thus, although risk factors for and outcome of ES are well characterized in adult allogeneic and autolo-gous HSCT, only few data exist for autoloautolo-gous periph-eral blood HSCT in the pediatric setting. Data from allogeneic pediatric transplants are virtually missing. We therefore retrospectively analyzed 61 consecutive allogeneic HSCT after myeloablative conditioning performed at our center to determine incidence,

pre-disposing factors, nonrelapse mortality, and survival in a pediatric transplant cohort.

PATIENTS AND METHODS Study Population

Sixty-one consecutive HSCT in 60 children at a single institution from a 13-year time period from 1988 to 2001 that fulfilled the following inclusion cri-teria were analyzed retrospectively by careful chart re-view: (1) allogeneic transplantation, (2) myeloablative conditioning, and (3) achievement of neutrophil en-graftment. Disease and transplant characteristics of the patients are shown in Table 1. T cell depletion was carried out by CD34-positive selection with im-munomagnetic beads (Miltenyi Biotec, Bergisch Glad-bach, Germany) if indicated. In all other transplants no graft manipulation was done. No patient had to be ex-cluded because of engraftment failure. All patients were conditioned with myeloablative chemotherapy with or without total body irradiation or total lymph node irradiation. All patients or their respective care-givers had expressed written consent to data storage

Table 1.Patient, Disease, and Transplant Characteristics

No. of patients 61

Age, median (range), years 7.8 (0.5-20)

Female/male 28/33 Donor MFD 34 MUD 19 MMFD 8 Graft source BM 50 PB 11 T cell depletion No 50 Yes 11 Conditioning regimen Busulfan based 38 TBI based 23 Underlying disease Malignant 44 (72%) ALL 29 AML 3 CML 8 MDS 3 ALCL 1 Nonmalignant 17 (28%) SCID 3 FHLH 2 WAS 5 Thal. 3 Other 4

ALCL indicates anaplastic large cell lymphoma; ALL, acute lym-phoblastic leukemia; AML, acute myeloid leukemia; BM, bone marrow; CML, chronic myeloid leukemia; FHLH, familial he-mophagocytic lymphohistiocytosis; MDS, myelodysplastic syn-drome; MFD, matched family donor; MMFD, mismatched family donor; MUD, matched unrelated donor; SCID, severe combined immunodeficiency; Thal., thalassemia; WAS, Wiskott Aldrich syndrome.

(3)

and analysis concerning their transplants. The study was announced to the local ethics committee and ded-icated as not requiring formal approval because of the retrospective nature of the study.

Supportive Care

All patients were cared for in laminar air flow units and received oral gut decontamination. Aciclovir was given from the day of admission until discharge. Be-cause of concerns about potential myelotoxicity cotri-moxazole was administered until day 2 and after stable neutrophil counts had been achieved. Empirical broad spectrum antibiotic therapy with a third-generation cephalosporine and aminoglycoside was initiated in neutropenic patients at the time of the first recorded fever of $38.5C. A glycopeptide antibiotic was added if fever persisted for $72 hours and amphotericin B if the fever lasted for $96 hours. Blood, stool, and urine cultures were taken and tested for bacterial, fungal, and viral pathogens at the onset of fever. Blood cultures were repeated when anti-infectious therapy was aug-mented or changed because of persistent fever. Further investigations for infectious agents were initiated if the patients presented specific symptoms. G-CSF was ad-ministered at the treating physician’s discretion after it was licensed in 1991.

GVHD Prophylaxis

GVHD prophylaxis consisted of cyclosporine A starting on day 21 and methotrexate on days 1, 3, 6, and 11 in all recipients of non-T cell-depleted grafts. Trough levels of cyclosporine A were targeted to be in the range of 100-150 ng/mL. Steroids (prednisolone 2 mg/kg/day) were initiated if clinical signs of aGVHD grade 2 or greater occurred.

Definitions

Neutrophil engraftment was defined as the first day on which the absolute neutrophil count exceeded 500/mL for 3 consecutive days. ES was diagnosed if pa-tients presented with 2 or more of the following symp-toms within 7 days before neutrophil engraftment: (1) fever of unknown origin .38.0C with no infectious agent or other reason detectable, (2) erythematous skin rash not because of drug reactions or viral infec-tion, (3) weight gain of $5% and albumin drop to #90% of pretransplant levels, and (4) pulmonary symptoms with dyspnea, hypoxia, and pulmonary infil-trates on a chest X-ray.

Statistical Analysis

Statistical analysis comparing 2 groups was per-formed using 2  2 contingency tables and Fisher’s ex-act test for count data (one sided). For survival estimates a Kaplan-Meier life table analysis and a Mantel-Haenszel log rank test were used to compare the time

variables between both groups. Differences were con-sidered significant at P \ .05. The following software was used for statistical analyses: ‘‘R’’ version 2.6.1 (R development core team, Vienna, Austria) and Prism, version 3.0 (GraphPad Software, San Diego, CA).

RESULTS

Incidence of and Symptoms Associated with ES To assess the incidence of ES in pediatric alloge-neic HSCT recipients we classified the patients for the presence of ES according to the above-mentioned criteria.

Of the 61 consecutive allogeneic HSCT analyzed, 29 (48%) met the diagnostic criteria for ES with at least 2 symptoms. Fever of otherwise unexplained etiology was present in 90% of transplantations diagnosed with ES (26 of 29 patients), an erythematous skin rash in 72% (21 of 29), weight gain/albumin drop in 66% (19 of 29) and pulmonary symptoms in 24% (7 of 29) (Figure 1A). Incidence of these symptoms in all transplantations was: fever 64% (39 of 61), skin rash 38% (23 of 61), weight gain/albumin drop 38% (23 of 61), and pulmonary symptoms 11% (7 of 61). Other symptoms included in the criteria suggested by Spitzer [1] were less frequent: hepatic dysfunction 8% (5 of 61), renal dysfunction 3% (2 of 61), and en-cephalopathy in 0% (Figure 1A). One patient devel-oped acute respiratory distress syndrome requiring mechanical ventilation. Previous reports had indicated that G-CSF administration would predispose to an in-creased rate of pulmonary complications after HSCT, which was not significant in the population analyzed here (Figure 1B; P 5 .09). Median onset of ES was ob-served after 16 days (range: 7-24). The median time to neutrophil recovery was 18 days (8-30) for the entire cohort and 18 (8-28) and 19 (10-30) for patients with or without ES respectively (Figure 2).

In summary, these data demonstrate that ES repre-sents a frequent complication of HSCT in pediatric patients.

Influence of Transplant and Disease Characteristics on the Incidence of ES

Previous studies in adult patients had identified pe-ripheral blood as stem cell source, higher number of cells infused, certain underlying diseases (breast can-cer), therapy with amphotericin B, application of G-CSF, and early recovery of neutrophil counts as risk factors associated with the development of ES [17]. We therefore analyzed the influence of these factors on the incidence of ES in our pediatric transplant co-hort. As the types of underlying diseases were very var-iable (Table 1), we categorized them as either malignant or nonmalignant.

In a univariate analysis the incidence of ES was not different depending on whether patients had received

(4)

bone marrow or peripheral blood (24 of 50 versus 5 of 11; P 5 .68), T cell depleted or not (5 of 11 versus 24 of 50; P 5 .57), from an unrelated or related donor (8 of 19 versus 21 of 42; P 5 .38), or whether they had a ma-lignant or nonmama-lignant underlying disease (22 of 44 versus 7 of 17; P 5 .37) (Table 2a). There was a nonsig-nificant trend toward a higher incidence associated with a neutrophil recovery on or before day 14 after transplantation (11 of 18 versus 18 of 43; P 5 .14) and whether patients were on amphotericin B therapy (25 of 46 versus 4 of 15; P 5 .06). The kinetics of neu-trophil recovery in patients with or without ES are dis-played inFigure 2. There was, however, a significant influence of G-CSF application with 12 of 17 patients developing ES versus 17 of 44 without G-CSF (P 5 .02). Likewise, a graft containing $7  108 mononu-clear cells (MNC) was associated with a significantly increased incidence of ES in 16 of 22 (73%) patients compared to 8 of 28 (29%) with an MNC count of \7  108(P 5 .002). Using a multiple logistic

regres-sion analysis, amphotericin B (P 5 .009) and MNC $7 108(P 5 .004) were significant risk factors, whereas G-CSF treatment was not (P 5 .35). These data are summarized inTable 2A. A 7-way multiple logistic re-gression model was built and a minimal model ob-tained by stepwise regression. The reduced model showed a trend toward a higher incidence of ES with an underlying malignant disease (P 5 .12), and again, a significant influence of amphotericin B therapy (P 5 .005) and MNC $7  108(P 5 .002) (Table 2B). T cell depletion was excluded from the multiway anal-ysis because of its perfect correlation with very low MNC values and with G-CSF therapy. This observa-tion is easily explained by the fact that all T cell deple-tions were carried out by CD34 positive selection, resulting in extremely low MNC counts, and that all these patients had received G-CSF a priori.

Taken together, a higher MNC count of the graft and G-CSF application were significant risk factors for developing ES in a univariate analysis in this pediatric transplant cohort, while amphotericin B therapy and higher MNC were significant risk factors in a multiple logistic regression analysis.

ES and GVHD

ES as an early complication of HSCT has been hy-pothesized to be the clinical correlate of a cytokine storm during engraftment. The release of high amounts of pro-inflammatory cytokines is also widely accepted to be 1 of the determining factors during the early phase of GVHD after HSCT[15]. We there-fore analyzed whether the occurrence of ES was asso-ciated with an increased incidence of GVHD in pediatric HSCT.

However, there was no difference in the incidence of aGVHD of any grade in patients with or without ES (17 of 29 versus 15 of 32; P 5 .25) and a nonsignificant trend toward a higher rate of chronic GVHD 75 symptom frequency (%) 0 25 50 100 fever rash weight gain pulmonary symptoms hepatic renal encephalopathy symptoms in ES A frequency of pulmonary symptoms (%) 0 25 G-CSF no G-CSF p=0.09 pulmonary symptoms and G-CSF B

Figure 1.Symptoms associated with ES. (A) Frequency of diagnostic symptoms in patients diagnosed with ES. The gray bars represent

symp-toms included in the diagnostic criteria used in this study. (B) Association of pulmonary sympsymp-toms with G-CSF application.

0 10 20 30 100 75 50 25 0 ES no ES days post tx cumulative incidence p=0.15 neutrophil recovery

Figure 2.Neutrophil recovery kinetics in ES. Cumulative incidence

of time to neutrophil recovery in transplants with (ES) and without ES (no ES) (n 5 61; P 5 .15).

(5)

(cGVHD) (7 of 21 versus 4 of 28; P 5 .11). These data are summarized inTable 3.

In summary, we could not demonstrate a correla-tion between ES and GVHD in this study.

Outcome of ES

Because earlier studies had revealed a negative im-pact of ES on survival after allogeneic HSCT in adults as well as autologous transplantation in children be-cause of an increased rate of TRM [8,20], we tested whether there was any discernible effect of ES on over-all survival (OS) and TRM in our study cohort.

However, after a median follow-up of 9.5 years (range: 5.9-17.4) OS did not differ after transplanta-tions complicated by ES compared to such without

ES (Figure 3A; P 5 .53). Likewise, TRM was identical

in both groups (Figure 3B; P 5 .65).

Taken together, we could not identify any impact of the occurrence of ES on OS or TRM.

DISCUSSION

This study represents, to our knowledge, the first published analysis to investigate the incidence, predis-posing factors, nonrelapse mortality, and survival of ES in pediatric allogeneic HSCT with a median follow-up of 9.5 years (range: 5.9-17.4). Of the 61 consecutive transplantations 48% met our diagnostic criteria for ES ($2 of the following symptoms: fever, skin rash,

weight gain/albumin drop, pulmonary symptoms). In 2001, Spitzer[1]proposed specific diagnostic criteria for ES. As major criteria, he suggested (1) temperature $38.3C with no infectious etiology, (2) erythematous rash involving .25% of body surface area and not at-tributable to a medication, and (3) noncardiogenic pul-monary edema manifested by diffuse pulpul-monary infiltrates consistent with this diagnosis, accompanied by hypoxia. As minor criteria, he suggested (1) hepatic dysfunction with either total bilirubin $2 mg/dL or transaminase levels $2 times normal, (2) renal insuffi-ciency with serum creatinine $2 times baseline, (3) weight gain $2.5% of baseline body weight, and (4) transient encephalopathy unexplainable by other causes[1]. A diagnosis of ES would be established by the presence of all 3 major criteria or 2 major criteria and 1 or more minor criteria. Although these criteria are somewhat more stringent than the ones we used in this study, they encompass most of the diagnostic items that we used. However, we do deem our criteria to better reflect the clinical situation in pediatric pa-tients as the incidence of renal or hepatic dysfunction as well as encephalopathy is rather low in children

(Figure 1A). According to the Spitzer[1]criteria, the

incidence of ES in our cohort would have been 20% (12 of 61).

Before ES was recognized as an independent entity of clinical symptoms it was often described as an early manifestation of a graft-versus-host reaction in alloge-neic transplantation[21]. However, this does not ex-plain the same high incidence of ES with a nearly identical clinical presentation in autologous transplan-tation in a study by Cahill et al.[5]. One could argue

Table 2B.Minimal Multiple Logistic Regression Analysis

Predisposing Factor:

Underlying disease malignant P 5 .12

Amphotericin B therapy yes P 5 .005

MNC $7 108 P 5 .002

Multiple logistic regression analysis was performed including 7 fac-tors (graft source, donor, underlying disease, neutrophil recov-ery, amphotericin B therapy, G-CSF therapy, and MNC). A minimal model was obtained by stepwise regression.

Table 3.Incidence of GVHD in Patients with ES

ES no ES

GVHD

acute 17/29 (59%) 15/32 (47%) P 5 .25

chronic 7/21 (33%) 4/28 (14%) P 5 .11

Table 2a.Influence of transplant and disease characteristics on the incidence of ES

Overall incidence of ES

Incidence with respect to: 29/61 (48%) Fisher’s exact test Multiple logistic regression

Graft source BM 24/50 (48%) PB 5/11 (45%) p50.68 p50.99

T-cell depletion yes 5/11 (45%) no 24/50 (48%) p50.57 §§

Donor unrelated 8/19 (42%) related 21/42 (50%) p50.38 p50.92

Underlying disease non-malignant 7/17 (41%) malignant 22/44 (50%) p50.37 p50.11

Neutrophil recovery after d14 18/43 (42%) until d14 11/18 (61%) p50.14 p50.22

Amphotericin B therapy no 4/15 (27%) yes 25/46 (54%) p50.06 p50.009

G-CSF therapy no 17/44 (39%) yes 12/17 (71%) p50.02 p50.35

MNC \7*108 8/28 (29%) 7*108 16/22 (73%) p50.002§ p50.004

BM: bone marrow, PB: peripheral blood, G-CSF: granulocyte colony stimulating factor, MNC: mononuclear cell count of the graft. §transplants with T-cell depletion were excluded from MNC analysis, due to their extremely low MNC count caused by graft manipulation. §§transplants with T-cell depletion were excluded from the multiple logistic regression analysis due to perfect correlation with MNC \7*108 and G-CSF use.

(6)

that in the allogeneic setting early GVHD may be mis-diagnosed as ES, but in this case one would expect a higher incidence of later GVHD manifestations in patients diagnosed with ES. In our study we could not identify an association between ES and the subse-quent development of aGVHD or cGVHD, adding further evidence that this syndrome occurs indepen-dently of graft-versus-host pathology. With cytokine release being a hallmark both of early GVHD develop-ment[15]as well as ES[5], however, one can still spec-ulate that there may be a certain overlap between both syndromes. Clinical situations might occur where a dif-ferentiation between the 2 might prove difficult, such as when rash develops before engraftment without fur-ther indication of GVHD. In these cases eifur-ther addi-tional symptoms fulfilling the above-mentioned criteria for ES or skin biopsies might help to draw a dis-tinction. Routine skin biopsies for rash were not per-formed in this study.

Sinusoidal obstruction syndrome (SOS) is another condition after HSCT that might sometimes be con-fused with ES, especially because some criteria for the diagnosis of both overlap, such as increase in serum bilirubin and weight gain. However, there are also cri-teria such as rash or fever for ES or right upper quad-rant pain for SOS that can help to differentiate between the 2. This is further illustrated by the fact that in this cohort only 2 of the ES patients were also diagnosed with SOS using the Seattle criteria [22], whereas this was the case in 1 patient in the group without ES.

As noted above, various predisposing factors for ES had been identified in previous studies. Most of them have only been found in some studies, likely reflecting the variable criteria used for diagnosing ES. One of the significant factors identified by univar-iate analysis in this study was granulocyte-colony stim-ulating factor (G-CSF) application, which was also found in the study by Lee et al.[2]. G-CSF application also lead to a nonsignificant increase in the rate of pul-monary complications. It can be speculated that the neutrophils further triggered by G-CSF during recov-ery are trapped in the small pulmonary vessels as a

con-sequence of cytokine release, endothelial lung damage by cytotoxic drugs, and previous radiotherapy and de-fective phagocytosis. The other significant risk factor in univariate analysis was a high MNC count, which had also been identified in 2 other adult studies[3,4]. When we analyzed our data in a multiple logistic gression model followed by an automated stepwise re-gression procedure, amphotericin B therapy evolved as a significant risk factor. MNC count was still signifi-cant, but G-CSF treatment was not anymore. This is very likely because of the fact that all patients receiving CD34 positive selected grafts had received G-CSF a priori, and these data sets had to be removed from this statistical analysis, thereby likely reducing the pos-sible influence of G-CSF treatment in this analysis. The incidence of ES was not affected by the source of stem cells, T cell depletion, type of donor, underly-ing disease, and time of neutrophil recovery, indicatunderly-ing that in the pediatric allogeneic transplant setting other factors may be important in the development of ES than in adult or autologous transplantation. In the only prospective study in pediatric autologous HSCT, high numbers of CD341cells/kg infused, chil-dren transplanted in an early phase, the type of malig-nancy (solid tumor), and a conditioning regimen other than busulfan based were significantly associated with ES in a multivariate analysis[13].

ES was determined to be the most important cause of TRM in the study by Foncillas et al.[20], which an-alyzed autologous transplantation in children, a situa-tion in which TRM is and should be a very rare event. Therefore, this finding cannot be compared to the fact that diagnosis of ES did not translate into a higher rate of TRM in the allogeneic setting in this study. There was also no difference in overall survival in the group diagnosed with ES. However, because of the retrospective nature of this study we cannot ex-clude an influence of ES-directed treatment on the outcome of our patients. Steroids are commonly used to treat ES, thereby utilizing its immunosuppressive and anti-inflammatory effect. However, there is no agreement on the most effective dose [2,6,11] and whether the clinical outcome is influenced by this overall survival 5 10 15 0 25 50 75 100 ES no ES p=0.53 years % survival A TRM 5 10 15 100 75 50 25 0 p=0.65 years %TRM ES no ES B

(7)

treatment[11]. However, steroids do seem to have an effect on ES, because steroid prophylaxis (0.5 mg/kg/ day) from day 14 to 114 significantly decreased the incidence of ES in adults by almost 10-fold following autologous PBSCT without increasing the risk of in-fection[23]. In our patient group, empirical treatment with prednisolone in standard doses (2 mg/kg/day) was administered in 18 of 29 (62%) of ES patients at the time of diagnosis of ES and seemed to be effective, as no patient died as a direct consequence of ES-related morbidity. Usually within 3 days after initiation of the steroid therapy the clinical condition improved dramatically, so that we could rapidly taper the steroid dose. However, because of the possible detrimental ef-fects of steroid therapy after HSCT such as increased risk of infection and abrogation of graft-versus-tumor reaction, we clearly need prospective studies to evalu-ate the role of steroids in the treatment of ES.

In summary, we found a high incidence of ES in children undergoing allogeneic HSCT with G-CSF use, amphotericin B treatment and a high MNC count of the graft as significant risk factors. ES had no effect on survival and TRM in this cohort. Further studies to elu-cidate the pathophysiology of ES and common diagnos-tic criteria to describe this clinical entity after HSCT are needed to differentiate ES from GVHD, SOS, and infections. Steroids seem to be an effective therapy for ES, but in the future it will be necessary to investigate the effects of steroid therapy without reducing antitu-mor activity and increasing infectious complications.

REFERENCES

1. Spitzer TR. Engraftment syndrome following hematopoietic stem cell transplantation. Bone Marrow Transplant. 2001;27: 893-898.

2. Lee CK, Gingrich RD, Hohl RJ, Ajram KA. Engraftment syn-drome in autologous bone marrow and peripheral stem cell transplantation. Bone Marrow Transplant. 1995;16:175-182. 3. Edenfield WJ, Moores LK, Goodwin G, Lee N. An engraftment

syndrome in autologous stem cell transplantation related to mononuclear cell dose. Bone Marrow Transplant. 2000;25: 405-409.

4. Ravoet C, Feremans W, Husson B, et al. Clinical evidence for an engraftment syndrome associated with early and steep neutro-phil recovery after autologous blood stem cell transplantation. Bone Marrow Transplant. 1996;18:943-947.

5. Cahill RA, Spitzer TR, Mazumder A. Marrow engraftment and clinical manifestations of capillary leak syndrome. Bone Marrow Transplant. 1996;18:177-184.

6. Capizzi SA, Kumar S, Huneke NE, et al. Peri-engraftment re-spiratory distress syndrome during autologous hematopoietic stem cell transplantation. Bone Marrow Transplant. 2001;27: 1299-1303.

7. Akasheh M, Eastwood D, Vesole DH. Engraftment syndrome after autologous hematopoietic stem cell transplant supported by granulocyte-colony-stimulating factor (G-CSF) versus gran-ulocyte-macrophage colony-stimulating factor (GM-CSF). Bone Marrow Transplant. 2003;31:113-116.

8. Gorak E, Geller N, Srinivasan R, et al. Engraftment syndrome after nonmyeloablative allogeneic hematopoietic stem cell trans-plantation: incidence and effects on survival. Biol Blood Marrow Transplant. 2005;11:542-550.

9. Khan SA, Gaa B, Pollock BH, et al. Engraftment syndrome in breast cancer patients after stem cell transplantation is associated with poor long-term survival. Biol Blood Marrow Transplant. 2001;7:433-438.

10. Madero L, Vicent MG, Sevilla J, Prudencio M, Rodriguez F, Diaz MA. Engraftment syndrome in children undergoing autol-ogous peripheral blood progenitor cell transplantation. Bone Marrow Transplant. 2002;30:355-358.

11. Maiolino A, Biasoli I, Lima J, Portugal AC, Pulcheri W, Nucci M. Engraftment syndrome following autologous hemato-poietic stem cell transplantation: definition of diagnostic crite-ria. Bone Marrow Transplant. 2003;31:393-397.

12. Nurnberger W, Willers R, Burdach S, Gobel U. Risk factors for capillary leakage syndrome after bone marrow transplantation. Ann Hematol. 1997;74:221-224.

13. Gonzalez-Vicent M, Ramirez M, Sevilla J, et al. Engraftment syndrome after autologous peripheral blood progenitor cell transplantation in pediatric patients: a prospective evaluation of risk factors and outcome. Bone Marrow Transplant. 2004;34: 1051-1055.

14. Oyama Y, Cohen B, Traynor A, Brush M, Rodriguez J, Burt RK. Engraftment syndrome: a common cause for rash and fever following autologous hematopoietic stem cell transplantation for multiple sclerosis. Bone Marrow Transplant. 2002;29:81-85. 15. Antin JH, Ferrara JL. Cytokine dysregulation and acute

graft-versus-host disease. Blood. 1992;80:2964-2968.

16. Takatsuka H, Takemoto Y, Yamada S, et al. Complications after bone marrow transplantation are manifestations of systemic in-flammatory response syndrome. Bone Marrow Transplant. 2000; 26:419-426.

17. Moreb JS, Kubilis PS, Mullins DL, Myers L, Youngblood M, Hutcheson C. Increased frequency of autoaggression syndrome associated with autologous stem cell transplantation in breast cancer patients. Bone Marrow Transplant. 1997;19:101-106. 18. Inaba H, Hale G, Leung W, et al. Diagnostic challenge in

recur-rent skin rash after autologous bone marrow transplantation. J Pediatr Hematol Oncol. 2006;28:525-528.

19. Kawano C, Muroi K, Kuribara R, et al. Engraftment syndrome after autologous peripheral blood stem cell transplantation with high numbers of peripheral blood stem cells followed by granu-locyte colony-stimulating factor administration. Bone Marrow Transplant. 2000;25:228-229.

20. Foncillas MA, Diaz MA, Sevilla J, et al. Engraftment syndrome emerges as the main cause of transplant-related mortality in pe-diatric patients receiving autologous peripheral blood progeni-tor cell transplantation. J Pediatr Hematol Oncol. 2004;26: 492-496.

21. Powles RL, Morgenstern GR, Kay HE, et al. Mismatched family donors for bone-marrow transplantation as treatment for acute leukaemia. Lancet. 1983;1:612-615.

22. McDonald GB, Hinds MS, Fisher LD, et al. Veno-occlusive dis-ease of the liver and multiorgan failure after bone marrow trans-plantation: a cohort study of 355 patients. Ann Intern Med. 1993; 118:255-267.

23. Mossad S, Kalaycio M, Sobecks R, et al. Steroids prevent en-graftment syndrome after autologous hematopoietic stem cell transplantation without increasing the risk of infection. Bone Marrow Transplant. 2005;35:375-381.

References

Related documents

88, 105 (1992) (“In assessing the impact of a state law on the federal scheme, we have refused to rely solely on the legislature’s professed purpose and have looked as well to

In this article, after briefly reviewing the literature about the rebound effect in freight transport (method and findings), what could be considered the prism of almost all

Students were asked to further rank the tools they have used in the last 6 months to contact their teachers, 37% identified their usage of face to face consultation whilst email

Educational Trends, Federal Legislation, Government Role, Information Networks, National Programs, Policy Formation, Public Policy, Social Change, Trend Analysis, User

18Mcmfi govms & ~r~on of the computer within one cycle of the computer’s internal clock; it is part of the computers oper~~g sYstem. @W@~ility of mic~e WM upheld in NEC COT.

To meet the aims of this review, a study had to meet the following criteria: (1) use a randomized controlled design; (2) utilize a nontreatment, treatment as usual,

Abstract: The paper points out certain quantitative methods largely ignored by library service providers, highlights the importance of customer participation in service

Union of European Football Associations decided to create a new legal instrument, the Financial Fair Play Regulations, in order to introduce some financial rationality