Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 May 1.
Published in final edited form as: Biol Blood Marrow Transplant. 2019 Dec 28;26(5):884–892. doi: 10.1016/j.bbmt.2019.12.763

Prior Gemtuzumab Ozogamicin Exposure in Adults With Acute Myeloid Leukemia Does Not Increase Hepatic Veno-occlusive Disease Risk After Allogeneic Hematopoietic Cell Transplantation: a CIBMTR Analysis

Vincent T Ho 1, Andrew St Martin 2, Waleska S Pérez 2, Patricia Steinert 2, Mei-Jie Zhang 2,3, Deborah Chirnomas 4, Caroline J Hoang 4, Fausto R Loberiza Jr 4, Wael Saber 2
PMCID: PMC7439134  NIHMSID: NIHMS1556887  PMID: 31891815

Abstract

Gemtuzumab ozogamicin (GO) therapy prior to allogeneic hematopoietic cell transplantation (alloHCT) has been historically associated with an increased risk of hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) in patients with acute myeloid leukemia (AML). The current analysis examined VOD/SOS risk and outcomes in a cohort of patients who in recent years reported to the Center for International Blood & Marrow Transplant Research. Adults with AML who had GO exposure prior to myeloablative alloHCT were matched 1:4 by age and disease status at transplant to recipients without GO exposure (controls). A total of 137 patients with GO exposure and 548 matched controls who underwent alloHCT between 2008 and 2011 were included in this analysis. With a median ~8-year follow-up of survivors, the 5-year overall survival probability was similar in the 2 cohorts: 38% and 38% in the GO-exposed versus control (P = .97) group. Incidence of VOD/SOS and severe VOD/SOS, respectively, at 100 days was 4% (95% confidence interval [CI]: 1–7) and 3% (95% CI: 1–6) in GO-exposed patients and 3% (95% CI: 2–5) and 1% (95% CI: 0–2) in controls. Correspondingly, among patients who developed VOD/SOS, 1-year survival probability after VOD/SOS diagnosis was 33% (95% CI: 5–72) and 27% (95% CI: 11–47) (P = .78). In multivariate analyses, GO exposure prior to alloHCT was not associated with an increased risk of VOD/SOS (odds ratio 1.10; P = .85) or death (hazard ratio 1.08; P = .57). Three (3%) deaths in the GO group and 3 (<1%) deaths in the control group were attributed to VOD/SOS. Our results suggest that GO treatment prior to myeloablative alloHCT in the recent era is not associated with an increased risk of post-transplant VOD/SOS or death.

INTRODUCTION

Hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) is a potentially life-threatening complication that occurs primarily after hematopoietic cell transplantation (HCT) [1]. VOD/SOS is characterized by fluid retention, painful hepatomegaly, weight gain, and hyperbilirubinemia and results from injury to the sinusoidal endothelial cells due, at least in part, to the conditioning regimen [1,2]. The rate of VOD/SOS following HCT in historical cohorts ranges from 0 to 62.3%, with a mean of 13.7%, and is higher with allogeneic HCT (alloHCT) compared with autologous HCT [3]. However, the incidence of VOD/SOS following alloHCT is decreasing as a result of advances in supportive care, improved understanding of risk factors, and increased use of less toxic and reduced-intensity conditioning regimens [46].

VOD/SOS occurs primarily in the context of HCT, but is also associated with exposure to certain chemotherapeutic agents, such as gemtuzumab ozogamicin (GO) [3,79]. GO is an antibody drug conjugate composed of the CD33-directed monoclonal antibody covalently linked to the cytotoxic agent N-acetyl gamma calicheamicin. The CD33 antigen is expressed on leukemic blasts in the vast majority of patients with acute myeloid leukemia (AML) and is an important target for antibody-based AML therapies [10]. VOD/SOS has been reported in patients exposed to GO, with and without alloHCT [7]. Treatment with GO prior to alloHCT may place patients at an increased risk of developing VOD/SOS post transplant [11]. In a pooled analysis of 9 clinical trials, the rate of post-transplant VOD/SOS was 37%, with a range of 20 to 89%, among patients treated with GO prior to alloHCT [8].

Fractionated dosing of GO has been examined as a strategy for reducing hepatotoxicity while maintaining the efficacy of GO for AML treatment. The pivotal phase 3 ALFA-0701 trial examined efficacy and safety of adding fractionated, low-dose GO (3 mg/m2 on days 1, 4, and 7 of induction and day 1 of consolidation) to standard chemotherapy versus standard chemotherapy alone. Results of this trial demonstrated improved outcomes with the addition of GO; a low rate of VOD/SOS was also observed, including in patients who proceeded to HCT [1214].. Based on ALFA-0701 and other key clinical trials, GO was re-approved in the United States (US) in 2017 for the treatment of adults with newly diagnosed CD33-positive (CD33+) AML and adults and children (aged ≥2 years) with relapsed/refractory CD33+ AML [7].

In this registry analysis, we examined VOD/SOS risk and outcomes in a recent cohort of adults with AML who received myeloablative alloHCT with and without prior GO exposure and reported to the Center for International Blood & Marrow Transplant Research (CIBMTR) database. As this cohort received alloHCT prior to the 2017 re-approval of GO by the US Food and Drug Administration (FDA), patients were administered GO based on the original FDA approval, which had a higher recommended dose and no fractionated dosing schedule [15]. However, our primary objective was to examine VOD/SOS risk after myeloablative alloHCT in GO-exposed patients in recent years, regardless of GO dosage and administration schedule.

METHODS

Patients and Study Design

The CIBMTR is a research collaboration between the Medical College of Wisconsin in Milwaukee, WI, and the National Marrow Donor Program® / Be The Match® (Minneapolis, MN; www.CIBMTR.org). CIBMTR operates a large outcomes database in HCT and, recently, non-HCT cellular therapies for diverse indications. The CIBMTR relies on the collaboration of more than 420 centers worldwide that share data on treated patients. The CIBMTR has data for more than 495,000 transplant recipients and receives data for more than 24,000 new transplants each year.

This retrospective, matched-cohort analysis included adult patients with AML who underwent first myeloablative alloHCT with versus without prior GO exposure between 2008 and 2011 and reported to the CIBMTR at the Comprehensive Report Form (CRF) level. Patients were randomly selected through a weighted randomization scheme for the CRF level of data collection, which captures detailed patient-, disease-, and transplant-related data further to the standard data collection. All data were collected pre-transplant, 100 days and 6 months post-transplant, annually until 6 years post-transplant, and biannually thereafter until death.

Adults with GO exposure before first myeloablative alloHCT were matched 1:4 to patients without GO exposure (controls) by age and disease status (complete remission [CR]1, CR≥2, and relapse/primary induction failure) at alloHCT. Matching was an iterative process whereby each case was matched to a pool of possible controls on disease status and age within 5 years, and the control with the smallest age difference was selected. This process was performed for each case, after which the process was repeated to add additional controls. The matching process was complete after each GO-exposed patient was matched to 4 controls.

Due to the small number of VOD/SOS events in the reduced-conditioning cohort and the expected differences in outcomes between myeloablative and non-myeloablative/reduced-intensity conditioning, the study population was limited to those who received myeloablative conditioning (Supplementary Table S1). The cutoff year 2011 was chosen because few GO-exposed patients in the database received alloHCT after 2011, as GO was withdrawn from the market the prior year. Patients who did not consent to research, patients whose data were embargoed from research studies, and syngeneic twins were also excluded from the analysis.

Outcomes

The primary outcome was the incidence of VOD/SOS at 100 days following alloHCT. Secondary outcomes were overall survival (OS) rates at 100 days, 6 months, and 1, 3, and 5 years, disease-free survival (DFS), relapse, and nonrelapse mortality (NRM) rates at 1, 3, and 5 years, and graft-versus-host disease (GVHD).

Diagnosis of VOD/SOS

VOD/SOS was diagnosed using the Modified Seattle criteria, the Baltimore criteria, autopsy/biopsy, or other criteria as reported by the individual centers. The Modified Seattle criteria requires ≥2 of the following within 20 days of alloHCT: serum bilirubin >34 umol/L (>2 mg/dL), hepatomegaly with right upper quadrant pain, and 2% weight gain from baseline due to fluid retention [16]. The Baltimore criteria requires serum bilirubin >34umol/L (>2 mg/dL) within 21 days of alloHCT and ≥2 of the following: hepatomegaly, 5% weight gain from baseline, and ascites [2]. Other diagnostic criteria included various combinations of ascites, serum bilirubin >2.0 mg, elevated liver enzymes, abnormal ultrasonography, hepatomegaly, and weight gain. Severe VOD/SOS was defined as VOD/SOS associated with renal impairment requiring dialysis or any noninfectious pulmonary abnormality.

Statistical Analyses

The cumulative incidences of VOD/SOS and severe VOD/SOS at 100 days were calculated using the cumulative incidence estimator to accommodate competing risks. Death without development of VOD/SOS was considered a competing risk. Probabilities of OS and DFS were calculated using the Kaplan–Meier method, and probabilities of relapse and NRM were calculated using the cumulative incidence estimator.

Multivariate analysis was performed for VOD/SOS at 100 days, OS, DFS, relapse, and NRM. To accommodate matched pairs, marginal logistic regression models were used to compare the incidence of VOD/SOS at day 100 between GO-exposed patients and controls, and marginal Cox regression models were used to examine the effect of GO exposure on OS, DFS, relapse, and NRM. Variables considered for adjustment in the models were recipient age, recipient sex, Karnofsky performance score, HCT-comorbidity index (HCT-CI), cytogenetics, disease status at alloHCT, conditioning regimen, pharmacokinetics for busulfan, donor type, cytomegalovirus match, graft type, GVHD prophylaxis, sirolimus usage, antithymocyte globulin/alemtuzumab usage, and hepatitis B and C status (Supplementary Table S2). All factors that attained a P ≤ .05 were held in the final model with the exception of GO exposure, which was held in all steps of the model building regardless of level of significance.

RESULTS

Patients and Treatment

A total of 137 patients with GO exposure and 548 patients without GO exposure (controls) prior to myeloablative alloHCT were included in the analysis (Table 1). Median (range) age at transplant was 42 (18–73) years for GO-exposed patients and 38 (18–74) years for controls. In both arms, 33% of patients were in CR1, 30% in CR≥2, and 37% in relapse or primary induction failure prior to alloHCT; a majority of patients had a Karnofsky performance score ≥90. All patients received myeloablative conditioning: 56 (41%) patients with GO exposure and 140 (45%) controls received a regimen containing total body irradiation and 81 (59%) and 168 (55%) received a regimen with chemotherapy only. Cyclophosphamide was included in the conditioning regimen of 82 (60%) patients with GO exposure and 378 (69%) controls. In all, 10 (7%) patients with GO exposure and 23 (4%) controls received prophylactic ursodeoxycholic acid; no patient in either arm received prophylactic defibrotide.

Table 1.

Patient-, disease-, and transplant-related characteristics

n (%)* GO
n =137
Control
n = 548
P Value
Patient-related
Age at transplant, median (range), years 42 (18–73) 38 (18–74) .26
Male 62 (45) 291 (53) .10
Karnofsky performance score .06
 90–100 79 (58) 346 (63)
 <90 52 (38) 194 (35)
 Not reported 6 (4) 8 (1)
HCT-CI .97
 0 54 (39) 224 (41)
 1–2 (25) 137 (25)
 ≥3 46 (34) 175 (32)
 Not reported 3 (2) 12 (2)
Hepatitis B and C serostatus pre-HCT .66
 B and C positive 4 (3) 20 (4)
 B positive 1 (<1) 6 (1)
 B and C negative 129 (94) 501 (91)
 Not tested/inconclusive 3 (2) 21 (4)
Disease-related
Cytogenetics .03
 Favorable 9 (7) 65 (12)
 Intermediate 80 (58) 322 (59)
 Poor 48 (35) 149 (27)
 Not reported 0 12 (2)
Disease status .99
 First complete remission 45 (33) 180 (33)
 Second or greater complete remission 41 (30) 164 (30)
 Relapse/primary induction failure 51 (37) 204 (37)
Transplant-related
Conditioning regimen .64
 Myeloablative-total body irradiation 56 (41) 236 (43)
 Myeloablative-chemotherapy 81 (59) 312 (57)
Pharmacokinetics for busulfan 40 (52) 143 (45)
Time from receiving GO to transplant, median (range), mo
 First complete remission 4(2–10)
 Second or greater complete remission 7 (1–52)
 Relapse/primary induction failure 3 (<1–76)
Type of donor. .74
 Related 37 (27) 170 (31)
 Unrelated 73 (53) 275 (50)
 Single cord blood 7 (5) 27 (5)
 Double cord blood 20 (15) 79 (14)
Donor-recipient CMV match .55
 Negative, negative 31 (23) 116 (21)
 Positive, negative 15 (11) 56 (10)
 Negative, positive 49 (36) 169 (31)
 Positive, positive 38 (28) 181 (33)
 Not reported 4 (3) 26 (5)
Donor age, median (range), years§ 32 (19–57) 31 (19–60) .40
Graft type .58
 Bone marrow 21 (15) 68 (12)
 Peripheral blood 89 (65) 374 (68)
 Umbilical cord blood 27 (20) 106 (19)
GVHD prophylaxis .43
 Tacrolimus-based 104 (76) 406 (74)
 Cyclosporine-based 32 (23) 123 (22)
 Other 1 (<1) 19 (3)
Sirolimus usage 4 (3) 45 (8) .03
ATG/alemtuzumab .67
 ATG alone 35 (26) 142 (26)
 Alemtuzumab alone 2 (1) 3 (<1)
 No ATG oi alemtuzumab 100 (73) 402 (73)
 Not reported 0 1 (<1)
Year of transplant <.001
 2008 64 (47) 174 (32)
 2009 47 (34) 169 (31)
 2010 23 (17) 138 (25)
 2011 3 (2) 67 (12)
Follow-up of survivors, median (range), months 96 (11–123) 92 (3–124)
*

Unless otherwise noted.

Cytogenetic classification was defined as follows:

Favorable: inv(16), t(16;16), del(16q), t(15;17), [t(8;21) without del(9q) or complex].

Intermediate: normal karyotype, +8, +6, -y, del(12p), t(9;11), 11q23, MLL rearranged, and any other abnormalities not belonging to favorable or poor.

Poor: complex (≥3 abnormalities), −5/5q, −7/7q, abn 3q 9q 11q 21q 17p, t(6;9), t(9;22).

For busulfan-containing regimens only.

§

For unrelated donors only.

ATG indicates antithymocyte globulin; CMV, cytomegalovirus; GO, gemtuzumab ozogamicin; GVHD, graft-versus-host disease; HCT, hematopoietic cell transplantation; HCT-CI, hematopoietic cell transplantation-comorbidity index; MLL, mixed lineage leukemia

Among patients with GO exposure, median (range) time from last GO exposure to transplant was 4 (2–10) months for patients in CR1 prior to alloHCT, 7 (1–52) months for patients in CR≥2, and 3 (<1–76) months for patients in relapse/primary induction failure.

GO dosing data were obtained for 58 (42%) of the 137 patients with GO exposure. All patient-, disease-, and transplant-related characteristics were balanced between patients with versus without dosing data (Supplementary Table S3). Therefore, the subset of patients with supplemental dose data can be considered representative of all GO-exposed patients in this analysis.

Among patients with available dosing data, median (range) total GO dose was 9.5 (3.0–33.0) mg. All 58 patients received GO in combination with chemotherapy. Characteristics were generally balanced between patients who received <10 versus ≥10 mg total GO, although patients receiving <10 mg were more likely to have poor cytogenetics (43% vs. 21%) and to be in CR1 at transplant (40% vs. 29%; Table 2). Fractionated-dose GO was administered in 8 (27%) patients receiving <10 mg total dose and in 11 (39%) patients who received ≥10 mg total dose. The corresponding median (range) number of cycles of GO prior to alloHCT was 1 (1–3) versus 1 (1–3) and median (range) time from last GO dose to transplant 4 (1–12) versus 3 (1–15) months.

Table 2.

Patient-, disease-, and transplant-related characteristics by GO total dose

n (%)* GO <10 mg
Total Dose
n = 30
GO ≥10 mg
Total Dose
n = 28
Patient-related
Age at transplant, median (range), years 41 (19–63) 47 (21–70)
Males 15 (50) 13 (46)
Karnofsky performance score
 90–100 16 (53) 15 (54)
 <90 13 (43) 13 (46)
 Not reported 1 (3) 0
HCT-CI
 0 13 (43) 9 (32)
 1–2 9 (30) 9 (32)
 ≥3 8 (27) 10 (36)
Hepatitis B and C serostatus pre-HCT
 B and C positive 0 1 (4)
 B positive 1 (3) 0
 B and C negative 28 (93) 26 (93)
 Not tested/inconclusive 1 (3) 1 (4)
Disease-related
Cytogenetics
 Favorable 1 (3) 2 (7)
 Intermediate 16 (53) 20 (71)
 Poor 13 (43) 6 (21)
Disease status
 First complete remission 12 (40) 8 (29)
 Second or greater complete remission 8 (27) 9 (32)
 Relapse/primary induction failure 10 (33) 11 (39)
Transplant-related
GO administration
 Single-agent GO 0 0
 GO plus chemotherapy 30 (100) 28 (100)
 Fractionated dosing 8 (27) 11 (39)
 Cycles of GO before HCT, median (range) 1 (1–3) 1 (1–3)
 Time from receiving GO transplant, median (range), months 4 (1–12) 3 (1–15)
Conditioning regimen
 Myeloablative–total body irradiation 9 (30) 9 (32)
 Myeloablative–chemotherapy 21 (70) 19 (68)
Pharmacokinetics for busulfan 7 (41) 10 (53)
Time from receiving GO to transplant, median (range), months
 First complete remission 4 (2–7) 3 (3–7)
 Second or greater complete remission 4 (2–9) 6 (2–15)
 Relapse/primary induction failure 4 (1–12) 2 (1–51)
Type of donor
 Related 11 (37) 12 (43)
 Unrelated 11 (37) 15 (54)
 Single cord blood 2 (7) 1 (4)
 Double cord blood 6 (20) 0
Donor-recipient CMV match
 Negative, negative 4 (13) 10 (36)
 Positive, negative 5 (17) 2 (7)
 Negative, positive 12 (40) 6 (21)
 Positive, positive 9 (30) 8 (29)
 Not reported 0 2 (7)
Donor age, median (range), years§ 33 (25–45) 28 (19–44)
Graft type
 Bone marrow 4 (13) 7 (25)
 Peripheral blood 18 (60) 20 (71)
 Umbilical cord blood 8 (27) 1 (4)
GVHD prophylaxis
 Tacrolimus-based 21 (70) 27 (96)
 Cyclosporine-based 8 (27) 1 (4)
 Other 1 (3) 0
Sirolimus usage 1 (3) 0
ATG/alemtuzumab
 ATG alone 7 (23) 5 (18)
 Alemtuzumab alone 1 (3) 0
 No ATG or alemtuzumab 22 (73) 23 (82)
Year of transplant
 2008 12 (40) .4 (50)
 2009 13 (43) 9 (32)
 2010 5 (17) 4 (14)
 2011 0 1 (4)
Follow-up of survivors, median (range), months 93 (49–98) 96 (56–121)
*

Unless otherwise noted.

Cytogenetic classification was defined as follows:

Favorable: inv(16), t(16;16), del(16q), t(15;17), [t(8;21) without del(9q) or complex].

Intermediate: normal karyotype, +8, +6, –y, del(12p), t(9;11), 11q23, MLL rearranged, and any other abnormalities not belonging to favorable or poor.

Poor: complex (≥3 abnormalities), –5/5q, –7/7q, abn 3q 9q 11q 21q 17p, t(6;9), t(9;22).

For busulfan-containing regimens only.

§

For unrelated donors only.

ATG indicates antithymocyte globulin; CMV, cytomegalovirus; GO, gemtuzumab ozogamicin; GVHD, graft-versus-host disease; HCT, hematopoietic cell transplantation; HCT-CI, hematopoietic cell transplantation-comorbidity index; MLL, mixed lineage leukemia

Incidence of VOD/SOS

The cumulative incidence (95% confidence interval [CI]) of VOD/SOS and severe VOD/SOS, respectively, at 100 days was 4% (1–7%) and 3% (2–5%) in patients with GO exposure and 3% (1–6%) and 1% (0–2%) in controls (Table 3). In patients who received cyclophosphamide as part of the conditioning regimen, the cumulative incidence (95% CI) of VOD/SOS at 100 days was 2% (0–7%) and 2% (1–4%) in the GO and control arms, respectively.

Table 3.

Outcomes in all patients

Outcomes, % (95% CI)* GO
n = 137
Controls
n = 548
P Value
VOD/SOS
 Patients who developed 6 22 -
 VOD/SOS, n
 VOD/SOS at 100 days 4 (1–7) 3 (2–5) .76
 Severe VOD/SOS at 100 days 3 (1–6) 1 (0–2) .18
Overall survival
 100 days 80 (73–86) 81 (78–84) .81
 6 months 66 (58–74) 69 (65–73) .52
 1 year 53 (44–61) 57 (53–61) .33
 3 years 41 (33–49) 42 (38–46) .83
 5 years 38 (30–46) 38 (34–42) .97
Disease-free survival
 1 year 45 (36–53) 49 (45–54) .31
 3 years 34 (26–42) 38 (34–42) .45
 5 years 31 (24–39) 34 (30–38) .46
Relapse
 1 year 40 (32–48) 32 (28–36) .09
 3 years 45 (37–54) 38 (34–42) .13
 5 years 48 (39–56) 40 (36–44) .10
Nonrelapse mortality
 1 year 15 (10–22) 18 (15–22) .37
 3 years 21 (14–28) 24 (21–28) .34
 5 years 21 (15–29) 26 (22–30) .26
*

From date of transplant.

CI indicates confidence interval; GO, gemtuzumab ozogamicin; VOD/SOS, veno-occlusive disease/sinusoidal obstruction syndrome

Six patients with GO exposure developed VOD/SOS; diagnosis was ascertained by autopsy/biopsy (n = 1), Baltimore criteria (n = 1), and other clinical diagnostic criteria (n = 4). Median (range) time from last GO dose to alloHCT among these 6 patients was 3 (2–5) months. A total of 22 controls developed VOD/SOS; diagnosis was ascertained by autopsy/biopsy (n = 8), Baltimore criteria (n = 2), modified Seattle criteria (n = 2), and other clinical diagnostic criteria (n = 10). No patient in either arm received therapeutic defibrotide for VOD/SOS.

In a multivariate analysis, GO exposure was not associated with an increased risk of VOD/SOS (odds ratio [OR] 1.10, 95% CI: 0.43–2.81; P = .85), nor were any of the other factors considered in the analysis (Table 4 and Supplementary Table S2).

Table 4.

Multivariate analysis of outcomes

Covariates* VOD/SOS at 100 Days OS DFS Relapse NRM
OR HR HR HR HR
(95% CI) P (95% CI) P (95% CI) P (95% CI) P (95% CI) P
GO exposure
 GO vs. no GO 1.10 (0.43–2.81) .85 1.08 (0.86–1.37) .57 1.23 (0.98–1.53) .07 1.46 (1.11–1.93) .007 0.91 (0.64–1.29) .58
Age, years
 ≥50 vs. <50 1.39 (1.14–1.69) .001 1.33 (1.10–1.61) .003 1.88 (1.35–2.62) <.001
HCT-CI
 ≥3 vs. 0–2 1.43 (1.20–1.72) <.001 1.41 (1.18–1.70) <.001 1.43 (1.12–1.83) .004
Cytogenetics
 Poor vs. favorable/ intermediate 1.27 (1.04–1.54) .021 1.33 (1.10–1.62) .004 1.51 (1.18–1.92) .004
Disease status
 Relapse/PIF vs. CR1 2.30 (1.82–2.92) <.001 2.51 (1.99–3..8) <.001 3.56 (2.64–4.79) <.001
Type of donor
 Cord blood vs. HLA-identical sibling 1.63 (1.22–2.17) .001 1.51 (1.16–1.96) .002 3.22 (2.02–5.13) <.001
 Other vs. HLA-identical sibling 2.36 (1.46–3.79) <.001

OR/HR >1 indicates a worse outcome for the comparison (first) group.

*

Significant covariates (P < .05) reported with the exception of GO exposure, which is reported for all outcomes regardless of significance.

CI indicates confidence interval; CR, complete remission; GO, gemtuzumab ozogamicin; HCT-CI, hematopoietic cell transplantation-comorbidity index; HLA, human leukocyte antigen; HR, hazard ratio; NRM, nonrelapse mortality; OR, odds ratio; PIF, primary induction failure; VOD/SOS, veno-occlusive disease/sinusoidal obstruction syndrome

Secondary Outcomes

With a median ~8-year follow-up of survivors, 5-year survival probability (95% CI) was 38% (30–46%) in patients with GO exposure and 38% (34–42%) in controls (Table 3). For the corresponding groups of patients who developed VOD/SOS, 1-year survival probability (95% CI) was 33% (5–72%) and 27% (11–47%) from the onset of VOD/SOS (Supplementary Table S4). Three (3%) deaths in the GO group and 3 (<1%) deaths in the control group were attributed to VOD/SOS (Table 5). Five-year (95% CI) rates of DFS were 31% (24–39%) and 34% (30–38%) in GO-exposed patients and controls, respectively; corresponding 5-year relapse (95% CI) rates were 48% (39–56%) and 40% (36–44%) and 5-year NRM (95% CI) rates were 21% (15–29%) and 26% (22–30%) (Table 3).

Table 5.

Death summary

Cause, n (%) GO
n = 137
Controls
n = 548
Total deaths 88 355
Primary disease 48 (55) 185 (52)
Graft failure 0 6 (2)
GVHD 10 (11) 29 (8)
Infection 9 (10) 46 (13)
Interstitial pneumonia/ARDS 1 (1) 16 (5)
Organ failure
 VOD/SOS 3 (3) 3 (<1)
 Liver failure 1 (1) 3 (<1)
 Cardiac failure 2 (2) 9 (2)
 Pulmonary failure 3 (3) 13 (4)
 CNS failure 0 1 (<1)
 Renal failure 0 1 (<1)
 Multiple organ failure 2 (2) 8 (2)
 Organ failure, not specified 1 (1) 4 (1)
Secondary malignancy 0 6 (2)
Hemorrhage 2 (2) 7 (2)
Other 1 (1) 8 (2)
Not reported 5 (6) 10 (3)

ARDS indicates acute respiratory distress syndrome; CNS, central nervous system; GO, gemtuzumab ozogamicin; GVHD, graft-versus-host disease; VOD/SOS, veno-occlusive disease/sinusoidal obstruction syndrome

In multivariate analyses, GO exposure was associated with an increased rate of relapse (HR 1.46, 95% CI: 1.11–1.93; P = .007) but was not significantly associated with OS (HR 1.08, 95% CI: 0.86–1.37; P = .57), DFS (HR 1.23, 95% CI: 0.98–1.53; P = .07), or NRM (HR 0.91, 95% CI: 0.64–1.29; P = .58; Table 4). Other factors associated with increased relapse were HCT-CI ≥3, poor cytogenetics, and a disease status of relapse or primary induction failure. Factors associated with decreased OS and DFS were age ≥50 years, HCT-CI ≥3, poor cytogenetics, a disease status of relapse or primary induction failure at the time of alloHCT, and a cord blood donor. Factors associated with increased NRM were age ≥50 years and a cord blood or other donor type (Table 4).

In an additional multivariate analysis, GO exposure was associated with an increased rate of grade 2–4 acute GVHD (HR 1.58, 95% CI 1.17–2.12; P = .003) but not chronic GVHD (HR 1.25, 95% CI: 0.96–1.63; P = .09).

Among patients with a total GO dose <10 mg versus ≥10 mg, respectively, who had available dosing data, 5-year survival probability (95% CI) was 25% (11–42%) and 39% (22–58%); 5-year (95% CI) rate of DFS was 19% (7–35%) versus 36% (19–54%), relapse 53% (36–71%) versus 50% (32–68%), and NRM 28% (13–46%) versus 14% (4–29%) (Supplementary Table S5). Formal statistical analyses to compare outcomes between dosing groups were not performed due to the sample size.

DISCUSSION

Hepatic VOD/SOS is a serious complication that occurs primarily following myeloablative alloHCT but has also been associated with the use of certain chemotherapies, including GO [3,79]. Results of this retrospective study showed the rate of VOD/SOS among adult patients who underwent myeloablative alloHCT for AML between 2008 and 2011, as reported to the CIBMTR, was similar in those with versus without prior GO exposure. Furthermore, after controlling for prognostic variables such as age, sex, disease status, cytogenetics, and conditioning regimen, GO exposure, at least in this cohort, did not significantly increase the risk of developing VOD/SOS or death among patients who received myeloablative alloHCT.

In this study, the incidence of VOD/SOS among adult patients with GO exposure prior to alloHCT was 4%; 4 (3%) cases were classified as severe and 3 of 88 (3%) deaths were due to VOD/SOS. This rate is in accordance with another recent retrospective study, which also found a low incidence of post-transplant VOD/SOS (8%) in adult patients previously treated with GO, although the study included patients receiving either myeloablative or reduced-intensity conditioning [17]. Historically, the reported incidence of VOD/SOS in GO-exposed patients has been much higher; pooled data from 9 clinical trials published in 2006 or earlier revealed a rate of 37% after alloHCT in this population [8].

The low incidence of VOD/SOS observed here and in other recent reports may reflect a number of factors. Improvements in identifying and managing patient- and treatment-related VOD/SOS risk factors and advances in preventative therapeutic strategies over the past 2 decades have contributed to a general decline in the occurrence of VOD/SOS following alloHCT [4]. In addition, a shift away from the use of conditioning regimens containing dual alkylating agents or high doses of total body irradiation and toward the use of so-called “reduced toxicity myeloablative” busulfan/fludarabine-containing regimens has also contributed to this decreasing trend [4,6,18,19]. Specific to the cohort in our study, all patients received first alloHCT, which carries a lower risk of VOD/SOS compared with second or greater alloHCT [20]. Extending the interval between GO administration and alloHCT may also reduce risk [11]. In this study, the median time to alloHCT following GO was 4 months in patients in CR1, 7 months in patients in CR≥2, and 3 months in patients in relapse or with primary induction failure. Relatively long intervals between transplant and GO may have benefited these patients, particularly those in CR1 and CR≥2. Among the 6 patients in the GO arm who developed VOD/SOS, the median time from last GO dose to alloHCT was 3 months. Due to the relatively low rates of VOD/SOS, we were not able to demonstrate a relationship between time from last GO exposure to transplant and occurrence of VOD/SOS.

After controlling for a number of prognostic factors, GO exposure was not found to be a significant risk factor for the development of VOD/SOS or death in adult patients but was associated with an increased risk of relapse following alloHCT. An explanation for this latter finding is not immediately apparent. The risk of relapse depends on several factors, such as age, disease status prior to alloHCT, conditioning regimen, donor type, and cytogenetic and molecular markers [2123]. In this study, these characteristics were generally well-balanced between treatment arms, although a slightly higher percentage of patients had poor cytogenetics in the GO (35%) versus control (27%) arm. Post-transplant outcomes in patients receiving GO prior to HCT have also been examined in the ALFA-0701 trial wherein no difference in relapse or NRM was observed between those who received GO prior to HCT versus those treated with standard chemotherapy alone [24]. However, patients in ALFA-0701 received fractionated-dose GO in combination with standard chemotherapy, whereas the patients in the current study were treated with GO using various dosages and administration schedules, thereby making a direct comparison difficult. It is worth noting that in the current analysis, GO exposure was associated with an increased risk of acute GVHD. Earlier studies have shown reduced risk of relapse in patients developing GVHD, as GVHD develops via the same mechanism as the beneficial elimination of residual malignant cells known as the graft-versus-leukemia effect [2527]. Therefore, our results suggest increased relapse in GO-exposed patients in this cohort is not the result of the blunting of the graft-versus-leukemia effect with GO treatment.

Consistent with prior reports [2832], older age, HCT-CI ≥3, poor cytogenetics, disease status of relapse or primary induction failure at transplant, and cord blood transplants were significant predictors of poorer outcomes following alloHCT. However, no factors were found to be associated with increased VOD/SOS risk. In contrast, the CIBMTR recently published a systematic evaluation of VOD/SOS risk factors in which a number of prognostic factors were identified, including, but not limited to, young age, Karnofsky score, disease status, and conditioning regimen [33]. The lack of findings in the current analysis may be attributable to insufficient power, given the smaller sample size and low event rate. Among GO-exposed patients, those who received <10 mg versus ≥10 mg total GO dose had slightly lower rates of OS and DFS at 5 years and a slightly higher rate of NRM, which could be due, at least in part, to between-group differences in disease status and/or cytogenetics. Due to the sample size, formal statistical comparisons were not performed by dose.

The findings of this study are in contrast to a similar analysis recently reported in a pediatric population, which found GO to be a significant risk factor for the development of VOD/SOS and a higher incidence of VOD/SOS (16%) in GO-exposed pediatric patients compared with that observed here [34]. In a recent CIBMTR analysis, younger age was identified and then subsequently validated as being a strong adverse risk factor for the development of VOD/SOS [33]. The results of these two studies suggest that in the context of young children, who are at higher risk of VOD/SOS at baseline, the use of GO pre-HCT may be associated with an increased risk of VOD/SOS, whereas in adult patients without other risk factors for VOD/SOS, the incremental risk of VOD/SOS after the modern day myeloablative conditioning transplant may not be evident without studying a very large cohort.

A few limitations of this analysis should be noted. First, VOD/SOS, especially milder cases, may have been under-reported due to the retrospective nature of the study. However, VOD/SOS is a serious illness that is usually recorded in medical records and likely would have been systematically reported to the CIBMTR. Variability in methods for ascertaining VOD/SOS could also have contributed to under-reporting of VOD/SOS in this population [4]. Lastly, it is possible that patients who received a lower GO dose or had more remote exposure (i.e., a longer interval between last GO dose and alloHCT) were more likely to be offered myeloablative alloHCT, thereby introducing an element of selection bias to this analysis.

In conclusion, we found that adult patients with AML who received GO prior to myeloablative alloHCT in recent years had a low incidence of post-transplant VOD/SOS, and exposure to GO was not associated with an increased risk of VOD/SOS or death. Our results suggest that prior use of GO should not, in itself, preclude adult patients without other obvious VOD/SOS risk factors from receiving myeloablative alloHCT in the current era. Nonetheless, physicians should continue to be vigilant in their monitoring of VOD/SOS in patients who undergo alloHCT, especially in those who have received prior GO therapy. Prospective studies are planned to evaluate the relationship of GO with VOD/SOS in patients receiving alloHCT in the era of fractionated GO dosing.

Supplementary Material

1

Highlights.

  • VOD/SOS incidence at 100 days post HCT was 4% vs 3% in GO-exposed adults vs controls.

  • Five-year overall survival probability was 38% in both groups.

  • GO exposure before HCT was not associated with an increased risk of VOD/SOS or death.

Acknowledgments

This study was sponsored by Pfizer. Medical writing support was provided by Emily Balevich, PhD, of Engage Scientific Solutions and was funded by Pfizer.

CIBMTR SUPPORT LIST

The CIBMTR is supported primarily by Public Health Service grant/cooperative agreement U24CA076518 with the National Cancer Institute (NCI), the National Heart, Lung and Blood Institute (NHLBI) and the National Institute of Allergy and Infectious Diseases (NIAID); grant/cooperative agreement U24HL138660 with NHLBI and NCI; grant U24CA233032 from the NCI; grants OT3HL147741, R21HL140314 and U01HL128568 from the NHLBI; contract HHSH250201700006C with Health Resources and Services Administration (HRSA); grants N00014-18-1-2888 and N00014-17-1-2850 from the Office of Naval Research; subaward from prime contract award SC1MC31881-01-00 with HRSA; subawards from prime grant awards R01HL131731 and R01HL126589 from NHLBI; subawards from prime grant awards 5P01CA111412, 5R01HL129472, R01CA152108, 1R01HL131731, 1U01AI126612 and 1R01CA231141 from the NIH; and commercial funds from Actinium Pharmaceuticals, Inc.; Adaptive Biotechnologies; Allovir, Inc.; Amgen, Inc.; Anonymous donation to the Medical College of Wisconsin; Anthem, Inc.; Astellas Pharma US; Atara Biotherapeutics, Inc.; BARDA; Be the Match Foundation; bluebird bio, Inc.; Boston Children’s Hospital; Bristol Myers Squibb Co.; Celgene Corp.; Children’s Hospital of Los Angeles; Chimerix, Inc.; City of Hope Medical Center; CSL Behring; CytoSen Therapeutics, Inc.; Daiichi Sankyo Co., Ltd.; Dana Farber Cancer Institute; Enterprise Science and Computing, Inc.; Fred Hutchinson Cancer Research Center; Gamida-Cell, Ltd.; Genzyme; Gilead Sciences, Inc.; GlaxoSmithKline (GSK); HistoGenetics, Inc.; Immucor; Incyte Corporation; Janssen Biotech, Inc.; Janssen Pharmaceuticals, Inc.; Janssen Research & Development, LLC; Janssen Scientific Affairs, LLC; Japan Hematopoietic Cell Transplantation Data Center; Jazz Pharmaceuticals, Inc.; Karius, Inc.; Karyopharm Therapeutics, Inc.; Kite, a Gilead Company; Kyowa Kirin; Magenta Therapeutics; Mayo Clinic and Foundation Rochester; Medac GmbH; Mediware; Memorial Sloan Kettering Cancer Center; Merck & Company, Inc.; Mesoblast; MesoScale Diagnostics, Inc.; Millennium, the Takeda Oncology Co.; Miltenyi Biotec, Inc.; Mundipharma EDO; National Marrow Donor Program; Novartis Oncology; Novartis Pharmaceuticals Corporation; Omeros Corporation; Oncoimmune, Inc.; OptumHealth; Orca Biosystems, Inc.; PCORI; Pfizer, Inc.; Phamacyclics, LLC; PIRCHE AG; Regeneron Pharmaceuticals, Inc.; REGiMMUNE Corp.; Sanofi Genzyme; Seattle Genetics; Shire; Sobi, Inc.; Spectrum Pharmaceuticals, Inc.; St. Baldrick’s Foundation; Swedish Orphan Biovitrum, Inc.; Takeda Oncology; The Medical College of Wisconsin; University of Minnesota; University of Pittsburgh; University of Texas-MD Anderson; University of Wisconsin - Madison; Viracor Eurofins and Xenikos BV. The views expressed in this article do not reflect the official policy or position of the National Institute of Health, the Department of the Navy, the Department of Defense, Health Resources and Services Administration (HRSA) or any other agency of the U.S. Government.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Interest

VTH has served as a consultant for Jazz Pharmaceuticals. ASM, WSP, PS, M-JZ, and WS: none. DC was an employee of Pfizer at the time of manuscript development. CJH and FRL are employees of and have equity ownership in Pfizer.

Data Statement

CIBMTR supports accessibility of research in accord with the National Institutes of Health (NIH) Data Sharing Policy and the National Cancer Institute (NCI) Cancer Moonshot Public Access and Data Sharing Policy. These policies hold that data that correspond to and support publications should be made as widely and freely available as possible while safeguarding the privacy of participants and protecting confidential and proprietary data. As of January 1, 2020, and in accordance with journal embargo policy, CIBMTR will make available on its public website de-identified analysis datasets and corresponding data dictionaries.

REFERENCES

  • 1.Richardson PG, Ho VT, Cutler C, Glotzbecker B, Antin JH, Soiffer R. Hepatic veno-occlusive disease after hematopoietic stem cell transplantation: novel insights to pathogenesis, current status of treatment, and future directions. Biol Blood Marrow Transplant. 2013;19:S88–90. doi: 10.1016/j.bbmt.2012.10.023. [DOI] [PubMed] [Google Scholar]
  • 2.Jones RJ, Lee KS, Beschorner WE, et al. Venoocclusive disease of the liver following bone marrow transplantation. Transplantation. 1987;44:778–783. [DOI] [PubMed] [Google Scholar]
  • 3.Coppell JA, Richardson PG, Soiffer R, et al. Hepatic veno-occlusive disease following stem cell transplantation: incidence, clinical course, and outcome. Biol Blood Marrow Transplant. 2010;16:157–168. doi: 10.1016/j.bbmt.2009.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Carreras E, Diaz-Beya M, Rosinol L, Martinez C, Fernandez-Aviles F, Rovira M. The incidence of veno-occlusive disease following allogeneic hematopoietic stem cell transplantation has diminished and the outcome improved over the last decade. Biol Blood Marrow Transplant. 2011;17:1713–1720. doi: 10.1016/j.bbmt.2011.06.006. [DOI] [PubMed] [Google Scholar]
  • 5.Lee JH, Choi SJ, Lee JH, et al. Decreased incidence of hepatic veno-occlusive disease and fewer hemostatic derangements associated with intravenous busulfan vs oral busulfan in adults conditioned with busulfan + cyclophosphamide for allogeneic bone marrow transplantation. Ann Hematol. 2005;84:321–330. doi: 10.1007/s00277-004-0982-4. [DOI] [PubMed] [Google Scholar]
  • 6.Bornhauser M, Storer B, Slattery JT, et al. Conditioning with fludarabine and targeted busulfan for transplantation of allogeneic hematopoietic stem cells. Blood. 2003;102:820–826. doi: 10.1182/blood-2002-11-3567. [DOI] [PubMed] [Google Scholar]
  • 7.Pfizer Inc. Mylotarg (gemtuzumab ozogamicin) prescribing information. 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761060lbl.pdf (accessed September 11, 2019).
  • 8.McKoy JM, Angelotta C, Bennett CL, et al. Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project. Leuk Res. 2007;31:599–604. doi: 10.1016/j.leukres.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 9.Giles FJ, Kantarjian HM, Kornblau SM, et al. Mylotarg (gemtuzumab ozogamicin) therapy is associated with hepatic venoocclusive disease in patients who have not received stem cell transplantation. Cancer. 2001;92:406–413. [DOI] [PubMed] [Google Scholar]
  • 10.Ehninger A, Kramer M, Rollig C, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014;4:e218. doi: 10.1038/bcj.2014.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wadleigh M, Richardson PG, Zahrieh D, et al. Prior gemtuzumab ozogamicin exposure significantly increases the risk of veno-occlusive disease in patients who undergo myeloablative allogeneic stem cell transplantation. Blood. 2003;102:1578–1582. doi: 10.1182/blood-2003-01-0255. [DOI] [PubMed] [Google Scholar]
  • 12.Castaigne S, Pautas C, Terre C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379:1508–1516. doi: 10.1016/S0140-6736(12)60485-1. [DOI] [PubMed] [Google Scholar]
  • 13.Lambert J, Pautas C, Terre C, et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica. 2019;104:113–119. doi: 10.3324/haematol.2018.188888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Pautas C, Raffoux E, Lambert J, et al. Outcomes following hematopoietic stem cell transplantation in patients treated with chemotherapy with or without gemtuzumab ozogamicin for acute myeloid leukemia. Blood. 2018;132:28–28. doi: 10.1182/blood-2018-99-111251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bross PF, Beitz J, Chen G, et al. Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin Cancer Res. 2001;7:1490–1496. [PubMed] [Google Scholar]
  • 16.Shulman HM, Hinterberger W. Hepatic veno-occlusive disease--liver toxicity syndrome after bone marrow transplantation. Bone Marrow Transplant. 1992;10:197–214. [PubMed] [Google Scholar]
  • 17.Battipaglia G, Labopin M, Candoni A, et al. Risk of sinusoidal obstruction syndrome in allogeneic stem cell transplantation after prior gemtuzumab ozogamicin treatment: a retrospective study from the Acute Leukemia Working Party of the EBMT. Bone Marrow Transplant. 2017;52:592–599. doi: 10.1038/bmt.2016.302. [DOI] [PubMed] [Google Scholar]
  • 18.Iravani M, Evazi MR, Mousavi SA, et al. Fludarabine and busulfan as a myeloablative conditioning regimen for allogeneic stem cell transplantation in high- and standard-risk leukemic patients. Bone Marrow Transplant. 2007;40:105–110. doi: 10.1038/sj.bmt.1705685. [DOI] [PubMed] [Google Scholar]
  • 19.Bornhauser M, Illmer T, Oelschlaegel U, et al. Gemtuzumab ozogamicin as part of reduced-intensity conditioning for allogeneic hematopoietic cell transplantation in patients with relapsed acute myeloid leukemia. Clin Cancer Res. 2008;14:5585–5593. doi: 10.1158/1078-0432.CCR-08-0894. [DOI] [PubMed] [Google Scholar]
  • 20.Mohty M, Malard F, Abecassis M, et al. Sinusoidal obstruction syndrome/veno-occlusive disease: current situation and perspectives-a position statement from the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 2015;50:781–789. doi: 10.1038/bmt.2015.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ossenkoppele GJ, Janssen JJ, van de Loosdrecht AA. Risk factors for relapse after allogeneic transplantation in acute myeloid leukemia. Haematologica. 2016;101:20–25. doi: 10.3324/haematol.2015.139105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Craddock CF. Full-intensity and reduced-intensity allogeneic stem cell transplantation in AML. Bone Marrow Transplant. 2008;41:415–423. doi: 10.1038/sj.bmt.1705975. [DOI] [PubMed] [Google Scholar]
  • 23.Barrett AJ, Battiwalla M. Relapse after allogeneic stem cell transplantation. Expert Rev Hematol. 2010;3:429–441. doi: 10.1586/ehm.10.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pautas C, Raffoux E, Lambert J, et al. Outcomes following hematopoietic stem cell transplantation in patients treated with chemotherapy with or without gemtuzumab ozogamicin for acute myeloid leukemia American Society of Hematology (ASH). San Diego, CA: 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990;75:555–562. [PubMed] [Google Scholar]
  • 26.Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–4383. doi: 10.1182/blood-2008-03-077974. [DOI] [PubMed] [Google Scholar]
  • 27.Weiden PL, Flournoy N, Thomas ED, et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med. 1979;300:1068–1073. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
  • 28.Saad A, Mahindra A, Zhang MJ, et al. Hematopoietic cell transplant comorbidity index is predictive of survival after autologous hematopoietic cell transplantation in multiple myeloma. Biol Blood Marrow Transplant. 2014;20:402–408 e401. doi: 10.1016/j.bbmt.2013.12.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid leukemia. Blood. 2006;107:3481–3485. doi: 10.1182/blood-2005-09-3724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood. 2000;96:4075–4083. [PubMed] [Google Scholar]
  • 31.Michallet M, Thomas X, Vernant JP, et al. Long-term outcome after allogeneic hematopoietic stem cell transplantation for advanced stage acute myeloblastic leukemia: a retrospective study of 379 patients reported to the Societe Francaise de Greffe de Moelle (SFGM). Bone Marrow Transplant. 2000;26:1157–1163. doi: 10.1038/sj.bmt.1702690. [DOI] [PubMed] [Google Scholar]
  • 32.Zhang MJ, Davies SM, Camitta BM, et al. Comparison of outcomes after HLA-matched sibling and unrelated donor transplantation for children with high-risk acute lymphoblastic leukemia. Biol Blood Marrow Transplant. 2012;18:1204–1210. doi: 10.1016/j.bbmt.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Strouse C, Zhang Y, Zhang MJ, et al. Risk score for the development of veno-occlusive disease after allogeneic hematopoietic cell transplant. Biol Blood Marrow Transplant. 2018;24:2072–2080. doi: 10.1016/j.bbmt.2018.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Duncan C, St Martin A, Perez W, et al. Veno-occlusive disease characteristics in pediatric patients with acute myeloid leukemia receiving gemtuzumab ozogamicin before allogeneic stem cell transplant. Biol Blood Marrow Transplant. 2018;24:S302. doi: 10.1016/j.bbmt.2017.12.347. [DOI] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES