Skip to main content
Blood logoLink to Blood
. 2006 Oct 17;109(4):1765–1772. doi: 10.1182/blood-2006-05-022335

Diabetes, hypertension, and cardiovascular events in survivors of hematopoietic cell transplantation: a report from the bone marrow transplantation survivor study

K Scott Baker 1, Kirsten K Ness 1, Julia Steinberger 1, Andrea Carter 2, Liton Francisco 2, Linda J Burns 1, Charles Sklar 3, Stephen Forman 4, Daniel Weisdorf 1, James G Gurney 5, Smita Bhatia 2,4
PMCID: PMC1794046  PMID: 17047152

Abstract

We ascertained the prevalence of self-reported late occurrence of diabetes, hypertension, and cardiovascular (CV) disease in 1089 hematopoietic cell transplantation (HCT) survivors who underwent HCT between 1974 and 1998, survived at least 2 years, and were not currently taking immunosuppressant agents and compared them with 383 sibling controls. All subjects completed a 255-item health questionnaire. The mean age at survey completion was 39.3 years for survivors and 38.6 years for siblings; mean follow-up was 8.6 years. Adjusting for age, sex, race, and body mass index (BMI), survivors of allogeneic HCT were 3.65 times (95% confidence interval [CI], 1.82-7.32) more likely to report diabetes than siblings and 2.06 times (95% CI, 1.39-3.04) more likely to report hypertension compared with siblings but did not report other CV outcomes with any greater frequency. Recipients of autologous HCTs were no more likely than siblings to report any of the outcomes studied. Allogeneic HCT survivors were also more likely to develop hypertension (odds ratio [OR] = 2.31; 95% CI, 1.45-3.67) than autologous recipients. Total body irradiation (TBI) exposure was associated with an increased risk of diabetes (OR = 3.42; 95% CI, 1.55-7.52). Thus, HCT survivors have a higher age- and BMI-adjusted risk of diabetes and hypertension, potentially leading to a higher than expected risk of CV events with age.

Introduction

Improvements in hematopoietic cell transplantation (HCT) methods and supportive-care strategies have resulted in an expanding population of long-term survivors. Several recent review articles have provided comprehensive summaries of established late effects in HCT survivors14; however, there are emerging issues in HCT survivors for which there exists a paucity of information. One such health concern that is prevalent in the general population, and that is becoming an issue of concern in cancer and HCT survivors, is the metabolic syndrome: a constellation of central obesity, insulin resistance, glucose intolerance, dyslipidemia, and hypertension. Metabolic syndrome is associated with a substantially increased risk for type 2 diabetes mellitus and atherosclerotic cardiovascular disease (CVD).115 Data from the third National Cholesterol Education Program Adult Treatment Panel III (ATP III)5,6 show the age-adjusted prevalence of metabolic syndrome in the US population of adults to be 26.7%.7 Additionally, a recent analysis of National Health and Nutrition Examination Survey (NHANES) 1999-2000 data demonstrates an increasing prevalence of the metabolic syndrome among US adolescents, where 6.8% of teens were found to have features suggestive of this syndrome.8

There is evidence to suggest that long-term cancer survivors may be at high risk for premature development of characteristics associated with the metabolic syndrome.911 Cancer survivors are more likely to be significantly overweight, have higher fasting plasma glucose and insulin levels, and significantly decreased serum high-density lipoprotein (HDL) cholesterol levels.9 Furthermore, hyperinsulinemia, impaired glucose tolerance, hypertriglyceridemia, low HDL cholesterol level, and abdominal obesity were more common among the HCT patients than among a non-HCT group of leukemia patients or healthy controls.12 Additionally, certain cancer treatments are known to be cardiotoxic, most notably radiation therapy and common chemotherapeutic agents such as anthracyclines and cyclophosphamide.1318 Thus, the combined effects of metabolic syndrome and direct damage to the myocardium and endothelium by cancer therapy represent a potentially devastating late effect of cancer treatment.11 Existing literature provides very little information regarding long-term cardiovascular outcomes in cancer survivors; however, an analysis of data from the Childhood Cancer Survivor Study has shown that the standardized mortality ratio for cardiac-related deaths was 8.2 (95% confidence interval [CI], 6.4-10.4) among long-term survivors of childhood cancer.19

Furthermore, while hyperglycemia and impaired glucose tolerance are well-recognized acute complications of cancer treatment, such as asparaginase and glucocorticoids,2023 or when steroids are used for prophylaxis or treatment of acute and/or chronic graft-versus-host disease (GVHD), there are minimal data regarding the long-term risk of diabetes in these patients. One study of 748 survivors of pediatric HCT found a higher than expected prevalence of both type 1 and type 2 diabetes in these survivors at a median of 11 years of follow-up.24

The purpose of this analysis was to (1) ascertain the prevalence of obesity and medical late effects including diabetes, hypertension, and cardiovascular events in a cohort of patients treated with HCT for cancer or hematologic disorders; (2) to compare the prevalence of these medical late effects with the prevalence in age- and sex-adjusted siblings; and (3) to evaluate the association between these medical late effects, demographic, and clinical factors.

Patients, materials, and methods

Subjects

This analysis was part of the Bone Marrow Transplant Survivor Study (BMT-SS), a collaborative cohort study established in 2000 between City of Hope Cancer Center and the University of Minnesota. Eligibility criteria included (1) HCT between January 1, 1974, and December 31, 1998, at City of Hope or University of Minnesota; and (2) survival of at least 2 years after hematopoietic cell transplantation (HCT). This analysis was restricted to 1089 HCT survivors who were alive and not taking immunosuppressant agents at the time of the baseline interview. A random sample of 383 siblings of HCT survivors were also recruited for comparison. The informed consent process, study protocols, and documents were approved by the Human Subjects Research Review Committees at the City of Hope Cancer Center and the University of Minnesota, in accordance with the Declaration of Helsinki. Each eligible participant, or his or her parent or legal guardian if younger than age 18 years at the time of interview, provided informed consent for the study.

Data collection

Participants, or their parents if they were younger than age 18 at the time of interview, completed the BMT-SS questionnaire, a 255-item survey assessing medical late effects, current medical conditions, medication use, health status, health behaviors, pregnancy history, demographic characteristics, socioeconomic indicators, and insurance coverage. The BMT-SS questionnaire asked participants to report impairment of organ systems and requests that the participant indicate the year the impairment was first diagnosed by a doctor or other health-care provider. This instrument was initially developed for use by the Childhood Cancer Survivor's Study25 and has been subsequently modified to address topics related specifically to the HCT population in the current study. The BMT-SS questionnaire was validated in a random sample of 100 HCT survivors. The agreement with medical records was excellent (Kappa > 0.8 agreement adjusted for chance) for musculoskeletal impairments, cardiovascular disease (including myocardial infarction and hypertension), pulmonary conditions, endocrine conditions (including diabetes), and GVHD, and moderate for second cancers, central nervous system disorders, and eye problems (Kappa 0.4-0.7).26 Additionally, self-reported height and weight have also been shown to be accurate and do not contribute significantly to errors in assessing body mass index (BMI).27,28

Outcomes

Five self-reported medical late effects were considered in these analyses: diabetes, hypertension (HTN), arterial disease, myocardial infarction (MI), and stroke. Participants were classified as diabetic if they answered “yes” to a question that asked about current use of a diabetic medication or if they reported onset of diabetes controlled by diet, pills, or insulin 2 or more years after their transplantation. Participants were classified as hypertensive if they reported current use of a medication to treat HTN or if they reported the onset of high blood pressure 2 or more years after transplantation. The presence of arterial disease was based on positive responses to questions that asked if participants had been told by a doctor or other health-care professional that they had hardening of the arteries or arteriosclerosis, coronary artery disease, or angina pectoris (chest pains due to lack of oxygen to heart requiring medication such as nitroglycerin), or if they indicated a coronary artery bypass surgery or angioplasty 2 or more years after their transplantation. MI and stroke were counted as events if participants indicated that they had these events 2 or more years after transplantation. HTN, diabetes, or cardiovascular events reported prior to HCT or within the first 2 years after HCT were excluded because of their likely association with medication use (calcineurin inhibitors and/or steroids for GVHD) or other early transplantation-related complications. Siblings were frequency matched for this analysis.

Independent variables

Case status (HCT survivors vs siblings), conditioning regimen, growth-hormone insufficiency, chronic GVHD, and donor type were considered as risk factors of interest for these analyses. Information regarding therapeutic exposures (conditioning regimens, prophylaxis, and treatment of GVHD), donor type, and presence of chronic GVHD was obtained from the HCT databases at each institution. Growth-hormone insufficiency was classified as positive if the participant indicated that they had growth-hormone insufficiency or received growth-hormone injections 2 or more years after their transplantation date. Age at transplantation, age at interview, race/ethnicity, sex, and BMI (calculated from self-reported height and weight at the time of the interview) were included as possible modifiers or confounders in the case-sibling analyses. Age at transplantation, age at interview, race/ethnicity, sex, BMI, and stem-cell source were included as possible modifiers or confounders in the case-case analyses.

Data analysis

The prevalence of medical late effects was described by tabulating “yes” responses to specific questions presented in the BMT-SS questionnaire. Within-survivor analysis was conducted to assess the role of clinical factors and therapeutic exposures such as conditioning regimen, growth-hormone insufficiency, chronic GVHD, and donor type. The analyses related to GVHD were limited to participants who received an allogeneic transplant. All analyses excluded subjects who reported the use of cyclosporine, steroids, or tacrolimus for at least 30 days in the past 2 years to account for the association between the known side-effect profiles of these medications that may themselves lead to the development of HTN and diabetes.

Descriptive statistics including means, standard deviations, frequencies, percentages, and ranges were calculated for demographic variables for the sibling comparison group and for demographic and treatment variables for the participating and nonparticipating HCT survivors. Two-sample t tests (for continuous variables) and chi-square tests (for dichotomous variables) were used to compare the difference between cases and siblings and between cases who participated and those who did not.

Frequencies and percentages were calculated for medical late effects among siblings and HCT survivors as totals and by conditioning regimen, growth-hormone insufficiency, chronic GVHD, and donor type among cases. The prevalence of each medical late effect, after adjusting for age at interview, age at transplantation, sex, race/ethnicity, and BMI was compared between cases and siblings by calculating odds ratios (ORs) and 95% confidence intervals using generalized estimating equations (GEEs) with a binomial distribution and a logit link. GEE methods were used in all models to account for the possible correlation between survivors and siblings from the same family.29 In an analysis limited to survivors only, the use of total body irradiation (TBI), GVHD status, and donor type were evaluated in relation to the outcome variables in unconditional logistic regression models. These models were adjusted for age at transplantation, age at interview, race/ethnicity, sex, and BMI. Stem-cell source, growth-hormone insufficiency, and institution were not identified as independent predictors of the outcomes, nor did they appreciably alter the estimates, so they were not included in the final models. Interaction terms for age at transplantation, age at interview, sex, and conditioning regimen were also evaluated. Confounding by age at transplantation, age at interview, and sex was examined by looking at the strength and the precision of the estimate in both full and reduced models. SAS version 9.1 was used for all analyses (SAS Institute, Cary, NC).

Results

Study participants

Of the 2091 eligible HCT survivors, 1276 (83.1% of those successfully contacted) became study participants. Of the remaining survivors, 556 (28.8%) refused participation, 236 (13.2%) were lost to follow-up, and 23 (1.1%) enrolled but did not complete the health questionnaire. An additional 187 participants were not included in these analyses, as they reported either current or recent (past 2 years) use of immunosuppressant drugs. Characteristics of the study participants are shown in Table 1. Participants were more likely to be female (44.9% vs 37.6%, P = .003) and older at study participation (mean age, 39.3 vs 34.8 years; P < .001) than nonparticipants. Participants were also older (mean age, 30.8 years; range, 2 months to 68.6 years) than nonparticipants (mean age, 24.9 years; range, 1 month to 68.2 years) when they received their transplant (P < .001) and, on average, had survived 16 months longer than nonparticipants since their HCT (P < .001). Participants were more likely to report their race/ethnicity as white (P = .001), to have received an autologous transplant (P = .006), and to have had peripheral blood as their stem-cell source (P = .01).

Table 1.

Characteristics of the study participants

Characteristic Patients Nonparticipants P
N 1089 792
Sex, n (%) .003
    Female 489 (44.9) 298 (37.6)
    Male 600 (55.1) 494 (62.4)
Race/ethnicity, no. (%)
    White 879 (80.7) 554 (69.9) .001
    Hispanic 135 (12.4) 128 (16.2)
    Other 75 (6.9) 110 (13.9)
Institution, no. (%)
    COH 523 (48.0) 362 (45.7) .45
    UMN 566 (52.0) 430 (54.3)
Mean age at transplantation, y (SD) 30.8 (16.7) 24.9 (15.7) < .001
Mean age at interview, y (SD) 39.3 (15.4) 34.8 (14.7) < .001
Mean time since transplantation, y (SD) 8.6 (5.1) 9.9 (5.5) < .001
Diagnosis, no. (%) .001
    Acute leukemia 357 (32.8) 283 (35.7)
    Lymphoma 331 (30.4) 190 (24.0)
    Chronic leukemia 204 (18.7) 135 (17.0)
    Aplastic anemia 64 (5.9) 83 (10.5)
    Metabolic disease 38 (3.5) 44 (5.6)
    Immune disorder 28 (2.6) 14 (1.8)
    Breast cancer 8 (0.7) 15 (1.9)
    Sarcoma 8 (0.7) 0 (0.0)
    Other 49 (4.5) 28 (3.5)
Stem-cell source, no. (%) .01
    Bone marrow 745 (68.4) 615 (77.7)
    Peripheral-blood stem cells 324 (29.8) 165 (20.8)
    Cord blood 20 (1.8) 12 (1.5)
Donor type, no. (%) .006
    Autologous 499 (45.8) 268 (33.8)
    Allogeneic 590 (54.2) 524 (66.2)
Conditioning regimen, no. (%) .22
    Chemotherapy 285 (26.2) 210 (26.5)
    TBI and chemotherapy 804 (73.8) 582 (73.5)
Chronic GVHD, allogeneic transplants only, no. (%) .48
    Yes 241 (40.8) 214 (40.8)
    No 349 (59.2) 310 (59.2)

SD indicates standard deviation; TBI, total body irradiation; and GVHD, graft-versus-host disease.

P values are for comparison with patients nonparticipants.

When compared with the sibling comparison group, HCT survivors were more likely to be male (55.1% vs 38.9%, P < .001) and of nonwhite race/ethnicity (19.3% vs 6.0%, P < .001) but comparable in age at study participation (HCT survivors: mean = 39.3 years, standard deviation [SD] = 15.4 years; siblings: mean = 38.6 years, SD = 16.5 years) and in age at transplantation or the sibling's age at the time their brother or sister received a transplant (cases: mean = 30.8 years, SD = 16.7; siblings: mean = 29.4 years, SD = 17.5).

Among the participating HCT survivors, acute leukemia was the most common diagnosis (32.8%), and bone marrow was the most prevalent (68.4%) stem-cell source. Fifty-four percent of the survivors received an allogeneic transplant, and 73.8% received TBI as part of their conditioning regimen. Among those who received an allogeneic transplant, 40.8% developed chronic GVHD. Overall, 4.5% of the HCT survivors reported growth-hormone insufficiency or received growth-hormone injections.

Outcomes associated with metabolic syndrome

Comparison with siblings.

The prevalence of medical late effects associated with metabolic syndrome by patient characteristics is shown in Tables 23. Diabetes was reported by 7.6% of HCT survivors and by 3.1% of siblings. While the specific type of diabetes could not be ascertained from data available in this study, of survivors with diabetes, 39.8% were obese (BMI ≥ 30 kg/m2) compared with 75% of siblings with diabetes who were obese (Table 4). The majority of both survivors and siblings with diabetes were treated by diet or oral agents (66.3% and 58.3%, respectively), and 26.5% of survivors and 33.3% of siblings were receiving insulin. HTN was reported by 18.5% of HCT survivors and by 14.9% of siblings. The prevalence of arterial disease, MI, or stroke was lower than 2% in the study population. The prevalence of obesity was 16.3% among HCT survivors and 19.8% among siblings.

Table 2.

Means and standard deviations of patient disease and transplant characteristics in those with diabetes, hypertension, and cardiovascular events

Characteristic Diabetes
Hypertension
Arterial disease
Myocardial infarction
Stroke
Mean SD Mean SD Mean SD Mean SD Mean SD
Age at transplantation, y 36.3 15.7 39 14.6 45.4 9.0 45.0 13.3 30.8 16.7
Age at interview, y 46.9 12.6 47.5 12.9 55.9 6.5 54.0 10.2 39.3 15.4
Time since transplantation, y 10.6 6.3 8.5 5.5 10.5 5.4 9.0 4.8 8.5 5.1
BMI 28.8 5.9 27.3 5.1 26.5 4.9 25.7 4.6 25.4 5.5
Table 3.

Frequencies and percentages of patient disease and characteristics in those with diabetes, hypertension, and cardiovascular events

Diabetes
Hypertension
Arterial disease
Myocardial infarction
Stroke
No. % No. % No. % No. % No. %
Siblings 12 3.1 57 14.9 6 1.6 3 0.8 1 0.3
Survivors 83 7.6 201 18.5 14 1.3 9 0.8 8 0.7
    Sex
        Female 30 36.1 86 42.8 1 7.1 4 44.4 4 50.0
        Male 53 63.9 115 57.2 13 92.9 5 55.6 4 50.0
    Race/ethnicity
        White 54 65.1 171 85.1 14 100.0 8 88.9 8 100.0
        Hispanic 20 24.1 20 10.0 0 0.0 0 0.0 0 0.0
        Other 9 10.8 10 5.0 0 0.0 1 11.1 0 0.0
    Diagnosis
        Acute leukemia 33 39.8 56 27.9 5 35.7 4 44.4 4 50.0
        Chronic leukemia 24 28.9 51 25.4 3 21.4 0 0.0 2 25.0
        Lymphoma 21 25.3 64 31.8 6 42.9 5 55.6 1 12.5
        Aplastic anemia 2 2.4 11 5.5 0 0.0 0 0.0 1 12.5
        Metabolic diseases 0 0.0 5 2.5 0 0.0 0 0.0 0 0.0
        Immune disorder 0 0.0 2 1.0 0 0.0 0 0.0 0 0.0
        Breast cancer 0 0.0 1 0.5 0 0.0 0 0.0 0 0.0
        Sarcoma 0 0.0 1 0.5 0 0.0 0 0.0 0 0.0
        Other 3 3.6 9 4.5 0 0.0 0 0.0 0 0.0
    Transplant type
        Autologous 31 37.3 88 43.8 6 42.9 5 55.6 3 37.5
        Allogeneic 52 62.7 113 56.2 8 57.1 4 44.4 5 62.5
    Stem-cell source
        Bone marrow 58 69.9 132 65.7 10 71.4 4 44.4 6 75.0
        Peripheral-blood stem cells 24 28.9 67 33.3 4 28.6 5 55.6 2 25.0
        Cord blood 1 1.2 2 1.0 0 0.0 0 0.0 0 0.0
    Conditioning regimen
        Chemotherapy 9 10.8 53 26.4 2 14.3 2 22.2 1 12.5
        TBI and chemotherapy 74 89.2 148 73.6 12 85.7 7 77.8 7 87.5
    Chronic GVHD
        No 61 73.5 145 72.1 8 57.1 7 77.8 6 75.0
        Yes 22 26.5 56 27.9 6 42.9 2 22.2 2 25.0
Growth-hormone deficiency
    No 80 96.4 197 98.0 14 100.0 9 100.0 6 75.0
    Yes 3 3.6 4 2.0 0 0.0 0 0.0 2 25.0

For siblings, N = 383; for survivors, N = 1089.

TBI indicates total body irradiation.

Table 4.

Characteristics of cases and siblings who report having diabetes

Survivors
Siblings
n % n %
Body mass index
    Less than 18.5 kg/m2 2 2.4 0 0.0
    18.5-24.9 kg/m2 19 22.9 1 8.3
    25-29.9 kg/m2 29 34.9 2 16.7
    Greater than 30 kg/m2 33 39.8 9 75.0
Treatment for diabetes
    Diet 11 13.3 1 8.3
    Oral agent(s) 44 53.0 6 50.0
    Insulin 22 26.5 4 33.3
    Not indicated 6 7.2 1 8.3

In models adjusted for age at interview, age at transplantation, BMI, race/ethnicity, and sex (Table 5), HCT survivors of allogeneic HCT were 3.65 times (95% CI, 1.82-7.32) more likely to report diabetes than siblings, but the risk was not increased in autologous HCT survivors. Other factors that increased the risk of developing diabetes included Hispanic race (OR, 1.99; 95% CI, 1.12-3.55), older age at interview (OR, 1.14; 95% CI, 1.08-1.19), and BMI (OR, 1.13; 95% CI, 1.09-1.18). Interestingly, for each year older at the time of HCT, the odds of having diabetes decreased by 8% (OR, 0.92; 95% CI, 0.88-0.96). The odds of HTN were also significantly elevated in allogeneic HCT survivors (OR, 2.06; 95% CI,1.39-3.04), in those survivors who were older at the time of interview (OR, 1.07; 95% CI, 1.04-1.11), and for those who had a higher BMI (OR, 1.07; 95% CI, 1.04-1.10). As with diabetes, autologous HCT survivors were no more likely to report HTN than were siblings, and survivors of either allogeneic or autologous HCT were not more likely than siblings to report arterial disease, MI, or stoke.

Table 5.

Multiple-variable model comparing patients to siblings by body mass index, sex, race/ethnicity, age at transplantation (or sibling's transplantation), and age at interview

Diabetes
Hypertension
Arterial disease
MI
CVA
OR 95% CI P OR 95% CI P OR 95% CI P OR 95% CI P OR 95% CI P
Case status
    Allogeneic 3.65 1.82-7.32 < .001 2.06 1.39-3.04 < .001 1.16 0.33-4.02 .82 1.21 0.24-6.04 .82 3.48 0.40-30.61 .26
    Autologous 2.03 0.98-4.21 .06 0.96 0.65-1.44 .86 0.42 0.12-1.48 .18 0.92 0.20-4.10 .91 2.62 0.26-26.76 .42
    Sibling Ref Ref Ref Ref Ref
Sex
    Female 0.80 0.51-1.25 .33 0.88 0.65-1.17 .37 0.11 0.02-0.49 .004 0.55 0.16-1.91 .35 1.32 0.34-5.09 .30
    Male Ref Ref Ref Ref Ref
Race/ethnicity
    Hispanic 1.99 1.12-3.55 .02 0.62 0.37-1.03 .06 NE NE NE
    Other 2.18 0.99-4.79 .06 0.83 0.44-1.57 .57 NE NE NE
    White Ref Ref Ref Ref Ref
Age at transplantation, y 0.92 0.88-0.96 < .001 0.98 0.96-1.01 .25 0.91 0.83-0.99 .03 0.95 0.85-1.06 .37 0.96 0.85-1.08 .50
Age at interview, y 1.14 1.08-1.19 < .001 1.07 1.04-1.11 < .001 1.26 1.29-1.41 < .001 1.16 1.01-1.32 .04 1.06 0.93-1.20 .38
BMI 1.13 1.09-1.18 < .001 1.07 1.04-1.10 < .001 0.98 0.87-1.09 .66 0.99 0.87-1.12 .83 0.92 0.79-1.08 .32

NE indicates not estimable because no outcome in one or more categories; and Ref, reference category.

Within-survivor cohort analysis.

In this analysis, models were adjusted for transplant type, sex, race/ethnicity, age at transplantation, age at interview, BMI, TBI exposure, and history of chronic GVHD, and these data are shown in Table 6. Compared with autologous transplant recipients, allogeneic transplant recipients were not more likely to report diabetes but were 2.31 times (95% CI, 1.45-3.67) more likely to report HTN. Exposure to TBI was associated with a 3.42-fold (95% CI, 1.55-7.52) increased risk of reporting diabetes but did not increase the risk for developing any of the other outcomes. Nonwhite race/ethnicity was associated with an over 2-fold increased risk for diabetes although a reduced risk for hypertension.

Table 6.

Multiple-variable model comparing cases only by transplant type, current immunosuppressant status, body mass index, sex, race/ethnicity, age at transplantation, and age at interview

Diabetes
Hypertension
Arterial disease
MI
Stroke
OR 95% CI P OR 95% CI P OR 95% CI P OR 95% CI P OR 95% CI P
Transplant type
    Autologous Ref Ref Ref Ref Ref
    Allogeneic 1.60 0.83-3.11 .16 2.31 1.45-3.67 < .001 1.79 0.26-12.56 .56 1.42 0.20-10.03 .72 0.84 0.13-5.25 .85
Sex
    Male Ref Ref Ref Ref Ref
    Female 0.72 0.44-1.18 .20 0.92 0.66-1.28 .61 0.10 0.01-0.81 .03 1.04 0.27-4.05 .95 1.19 0.29-4.90 .81
Race/ethnicity
    White Ref Ref Ref Ref Ref
    Hispanic 2.52 1.38-4.57 .003 0.57 0.33-0.96 .04 NE NE NE
    Other 2.86 1.26-6.50 .01 0.65 0.32-1.32 .23 NE NE NE
Age at transplantation, y 0.92 0.88-0.96 < .001 1.00 0.96-1.03 .81 0.92 0.83-1.03 .15 0.96 0.84-1.10 .55 0.95 0.84-1.08 .46
Age at interview, y 1.14 1.08-1.20 < .001 1.06 1.02-1.10 .001 1.26 1.10-1.44 < .001 1.16 0.98-1.36 .08 1.04 0.91-1.19 .58
BMI 1.14 1.08-1.18 < .001 1.05 1.01-1.08 .004 0.97 0.84-1.12 .70 0.96 0.82-1.11 .55 0.94 0.80-1.11 .49
Conditioning
    Chemotherapy Ref Ref Ref Ref Ref
    TBI and chemotherapy 3.42 1.55-7.52 .002 0.71 0.48-1.06 .10 1.56 0.30-8.09 .59 1.18 0.22-6.28 .85 2.40 0.27-21.07
Chronic GVHD
    No Ref Ref Ref Ref Ref
    Yes 0.71 0.38-1.34 .29 1.09 0.70-1.71 .70 2.44 0.46-12.94 .30 0.99 0.13-7.3 .99 1.16 0.18-7.45 .87

NE indicates not estimable because no outcome in one or more categories; and Ref, reference category.

Other factors that increased the risk of developing diabetes and hypertension included older age at interview (OR, 1.14; 95% CI, 1.08-1.20 for diabetes; and OR, 1.06; 95% CI, 1.02-1.10 for HTN) and higher BMI (OR, 1.14; 95% CI, 1.08-1.18 for diabetes; and OR, 1.05; 95% CI, 1.01-1.08 for HTN). As seen with the case-sibling analyses, for each year older at the time of HCT, the odds of having diabetes decreased by 8% (OR, 0.92; 95% CI, 0.88-0.96). A history of chronic GVHD was not found to be associated with an increased risk for diabetes or hypertension.

With the exception of older age being associated with a higher risk of arterial disease (OR, 1.26; 95% CI, 1.10-1.44), we did not find an increased risk of arterial disease, MI, or stroke in association with TBI exposure, chronic GVHD history, or type of HCT. Participants who reported a history of growth-hormone insufficiency were 7.5 times (95% CI, 1.1-52.9) more likely to report stroke than those who did not report a history of growth-hormone insufficiency. However, the risk for other late effects did not vary by the presence or absence of growth-hormone insufficiency (data not shown).

Discussion

This study provides long-term follow-up data on the largest cohort of HCT survivors ever examined for outcomes related to diabetes, HTN, and cardiovascular events. Our findings suggest that HCT survivors have an elevated risk of diabetes and HTN but that this population is not at an increased risk for stroke, MI, or arterial disease when compared with the sibling control group.

In this cohort, diabetes was more than 3 times as likely to occur in allogeneic transplant recipients as in a sibling comparison group, and when examined further, this risk was associated with exposure to TBI. The higher risk of diabetes in the allogeneic transplantation group, compared with autologous recipients, was also influenced by the fact that a higher proportion of those patients received TBI in their conditioning regimens. Previous studies indicate that radiation exposure to the abdomen may be associated with the development of diabetes in a small number of patients, but overall the evidence is quite limited.3032 Another study that compared 34 children and adolescents after either autologous or allogeneic HCT to 21 age-/sex-matched controls found that the 18 patients who received TBI had a significantly higher first-phase insulin response and insulinemia-glycemia ratio on glucose-tolerance testing compared with patients who received only lymphoid radiation, no radiation, or controls.33 These findings suggest that TBI may play a role in the development of insulin resistance, which may predispose patients to the subsequent development of diabetes. Insulin resistance is also linked in the causal pathway leading to the cardiovascular effects of the metabolic syndrome because of its clinical association with increased rates of macrovascular disease and subsequent cardiovascular morbidity and mortality.34,35 The only other large study in HC transplant recipients also found the prevalence of diabetes (6%, 35/578) to be higher after HCT than in the general population, and this prevalence was very similar to what was found in our study (7.6%, 83/1089).24

HTN was not found to be influenced by TBI, but a significantly higher risk of HTN was associated with allogeneic transplantation. Interestingly, the risk of HTN was not increased in subjects who had a history of chronic GVHD where exposure to calcineurin inhibitors and steroids was likely more prolonged. Hypertension is a well-known side effect of calcineurin inhibitors while the exposure is ongoing, but whether this translates into a higher risk years after discontinuation of these medications is unknown but does not appear to be so based on our data.

Other mechanisms related to the immunologic and inflammatory effects of allogeneic HCT may play a role in the development of diabetes (or insulin resistance) and hypertension. Inflammation has been shown to be an integral component of the atherosclerotic process.36 Elevated levels of C-reactive protein (CRP) predict coronary events in population studies and in individuals with angina,3739 and elevated levels of TNF-α and of IL-6 are correlated with worse outcomes.4042 Recent studies have shown a significant increase in inflammatory markers in association with both atherosclerotic disease and the insulin-resistance syndrome.43,44 Inflammatory- and cytokine-mediated mechanisms that contribute to insulin resistance and development of features of the metabolic syndrome are of particular interest in individuals who have undergone an allogeneic HCT because the immunologic effects of these proinflammatory cytokines are mediators of the development and progression of both acute and chronic GVHD.4548 For individuals with chronic GVHD, this cytokine-mediated inflammatory process may continue for months or even years, and therapeutic maneuvers using cytokine inhibitory or blocking agents have recently been used successfully in treating chronic GVHD.4952 Interestingly, however, in our data the increased risk of diabetes and hypertension was not found to be directly associated with a history of chronic GVHD but was associated with receiving an allogeneic HC transplant regardless of whether the patients had a history of chronic GVHD or not.

The low prevalence of specific cardiovascular outcomes, such as arterial disease, MI, or stroke, can be explained by the fact that the study participants, both HCT survivors and siblings, are still relatively young with a relatively short mean follow-up of 8.3 years. Additionally, we did not identify risk factors for these events, with the exception of an increased risk of stroke in subjects who had growth-hormone insufficiency. Although details regarding pre-HCT therapeutic exposures, in particular, cranial radiation, are not available, it is possible that these subjects may have received additional cranial radiation that may have increased the risk of growth-hormone insufficiency as well as stroke.

Interestingly, HCT survivors were not found to be any more likely than the sibling comparison group to be obese, as might have been expected in view of their exposure to radiation, steroids, and prolonged periods of physical inactivity. A recent report from the Childhood Cancer Survivor Study (CCSS) examined obesity in 1765 adult survivors of childhood acute lymphoblastic leukemia compared with 2565 sibling controls. The age-/race-adjusted odds ratio for being obese in survivors treated with cranial radiation at doses of at least 20 Gy in comparison to the siblings was 2.59 for females (95% CI, 1.88-3.55; P < .001) and 1.86 for males (95% CI, 1.33-2.57; P < .001). Obesity was not associated with treatment consisting of chemotherapy only or with cranial radiation doses of 10 to 19 Gy.53 Therefore, since the total dose of radiation exposure in HCT protocols is typically in the range of 10 Gy to 14 Gy,54,55 it appears that HCT survivors who have not been exposed to additional cranial radiation might not acquire an additional risk of obesity. This is despite the fact that the dose rate is higher and the total treatment is given over 1 to 4 days, which may have a greater impact at the cellular level than a higher dose given over a longer time period.5658

The results of the study must be interpreted in the context of potential limitations. Participation rate was 61% of those presumed eligible and 83.1% of those successfully contacted, and there were several differences between participants and nonparticipants as detailed previously. As a result, our analysis may have slightly overestimated or underestimated the prevalence of the outcomes of interest in the post-HCT period. We also acknowledge the fact that this study is based upon self-reported medical information and despite the fact that a validation study showed very good agreement between questionnaire answers and medical records, there is still the possibility that some subjects incorrectly reported conditions that they did not have or that they failed to report conditions that they did actually have. However, it is also important to note that the comparison group (ie, siblings) also provided self-reported data, thus there should not have been any systematic differences in bias by case or control status. Finally, there are other known risk factors for several outcomes in this study such as family history, smoking history, and physical activity that we did not have data available on and were not able to account for in the analysis that may have had an impact on the development of some of these complications.

Taken together, the findings presented here support the need for systematic and comprehensive research into the presence of early indicators of metabolic syndrome, or insulin resistance in HCT survivors, especially those who have received an allogeneic HC transplant.

Additionally, further studies that more precisely examine the mechanisms leading to the development of these outcomes will be important. HCT survivors should be monitored for the development of HTN and diabetes and counseled regarding health behaviors that might modify these outcomes, such as diet and exercise. Longer follow-up will be required to determine the true prevalence of serious cardiovascular events in this survivor population.

Acknowledgments

This study was supported in part by grants from the National Cancer Institute (R01 CA078938; S.B.), the Leukemia Lymphoma Society (2192; S.B.), and the National Institutes of Health (K23 CA85503-01; K.S.B.).

Footnotes

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

Authorship

Contribution: K.S.B., as corresponding author, had full access to all the data in the study, had final responsibility for the decision to submit for publication and participated in the study design, analysis, interpretation of results, manuscript preparation, review, and submission; K.K.N. and S.B. participated in the study design, analysis, interpretation of results, and manuscript preparation, review, and submission; J.S., L.J.B., C.S., S.F., D.W., and J.G.G. participated in the interpretation of study results and manuscript preparation, review, and submission; A.C. and L.F. participated in the conduct of the study, data collection and preparation, and review of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: K. Scott Baker, Department of Pediatrics, University of Minnesota, Minneapolis MN, 55455; e-mail: baker084@umn.edu.

References

  • 1.Brennan BM, Shalet SM. Endocrine late effects after bone marrow transplant. Br J Haematol. 2002;118:58–66. doi: 10.1046/j.1365-2141.2002.03527.x. [DOI] [PubMed] [Google Scholar]
  • 2.Leiper AD. Non-endocrine late complications of bone marrow transplantation in childhood: part II. Br J Haematol. 2002;118:23–43. doi: 10.1046/j.1365-2141.2002.03471.x. [DOI] [PubMed] [Google Scholar]
  • 3.Leiper AD. Non-endocrine late complications of bone marrow transplantation in childhood: part I. Br J Haematol. 2002;118:3–22. doi: 10.1046/j.1365-2141.2002.03470.x. [DOI] [PubMed] [Google Scholar]
  • 4.Socie G, Salooja N, Cohen A, et al. Nonmalignant late effects after allogeneic stem cell transplantation. Blood. 2003;101:3373–3385. doi: 10.1182/blood-2002-07-2231. [DOI] [PubMed] [Google Scholar]
  • 5.Park YW, Zhu S, Palaniappan L, Heshka S, Carnethon MR, Heymsfield SB. The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988-1994. Arch Intern Med. 2003;163:427–436. doi: 10.1001/archinte.163.4.427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287:356–359. doi: 10.1001/jama.287.3.356. [DOI] [PubMed] [Google Scholar]
  • 7.Ford ES, Giles WH, Mokdad AH. Increasing prevalence of the metabolic syndrome among U.S adults. Diabetes Care. 2004;27:2444–2449. doi: 10.2337/diacare.27.10.2444. [DOI] [PubMed] [Google Scholar]
  • 8.Cook S, Auinger P, Weitzman M. Increasing prevalence of the metabolic syndrome among US adolescents [abstract]. Pediatr Res. 2004;55:592a. Abstract 3360. [Google Scholar]
  • 9.Talvensaari KK, Lanning M, Tapanainen P, Knip M. Long-term survivors of childhood cancer have an increased risk of manifesting the metabolic syndrome. J Clin Endocrinol Metab. 1996;81:3051–3055. doi: 10.1210/jcem.81.8.8768873. [DOI] [PubMed] [Google Scholar]
  • 10.Talvensaari K, Knip M. Childhood cancer and later development of the metabolic syndrome. Ann Med. 1997;29:353–355. doi: 10.3109/07853899708999360. [DOI] [PubMed] [Google Scholar]
  • 11.Nuver J, Smit AJ, Postma A, Sleijfer DT, Gietema JA. The metabolic syndrome in long-term cancer survivors, an important target for secondary preventive measures. Cancer Treat Rev. 2002;28:195–214. doi: 10.1016/s0305-7372(02)00038-5. [DOI] [PubMed] [Google Scholar]
  • 12.Taskinen M, Saarinen-Pihkala UM, Hovi L, Lipsanen-Nyman M. Impaired glucose tolerance and dyslipidaemia as late effects after bone- marrow transplantation in childhood. Lancet. 2000;356:993–997. doi: 10.1016/S0140-6736(00)02717-3. [DOI] [PubMed] [Google Scholar]
  • 13.Lipshultz SE, Lipsitz SR, Mone SM, et al. Female sex and drug dose as risk factors for late cardiotoxic effects of doxorubicin therapy for childhood cancer. N Engl J Med. 1995;332:1738–1743. doi: 10.1056/NEJM199506293322602. [DOI] [PubMed] [Google Scholar]
  • 14.Sorensen K, Levitt G, Bull C, Chessells J, Sullivan I. Anthracycline dose in childhood acute lymphoblastic leukemia: issues of early survival versus late cardiotoxicity. J Clin Oncol. 1997;15:61–68. doi: 10.1200/JCO.1997.15.1.61. [DOI] [PubMed] [Google Scholar]
  • 15.Matthys D, Verhaaren H, Benoit Y, Laureys G, De Naeyer A, Craen M. Gender difference in aerobic capacity in adolescents after cure from malignant disease in childhood. Acta Paediatr. 1993;82:459–462. doi: 10.1111/j.1651-2227.1993.tb12722.x. [DOI] [PubMed] [Google Scholar]
  • 16.Colvin M, Hilton J. Pharmacology of cyclophosphamide and metabolites. Cancer Treat Rep. 1981;65(suppl 3):89–95. [PubMed] [Google Scholar]
  • 17.Erselcan T, Kairemo KJ, Wiklund TA, et al. Subclinical cardiotoxicity following adjuvant dose-escalated FEC, high-dose chemotherapy, or CMF in breast cancer. Br J Cancer. 2000;82:777–781. doi: 10.1054/bjoc.1999.0998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Morandi P, Ruffini PA, Benvenuto GM, La Vecchia L, Mezzena G, Raimondi R. Serum cardiac troponin I levels and ECG/Echo monitoring in breast cancer patients undergoing high-dose (7 g/m(2)) cyclophosphamide. Bone Marrow Transplant. 2001;28:277–282. doi: 10.1038/sj.bmt.1703132. [DOI] [PubMed] [Google Scholar]
  • 19.Mertens AC, Yasui Y, Neglia JP, et al. Late mortality experience in five-year survivors of childhood and adolescent cancer: the Childhood Cancer Survivor Study. J Clin Oncol. 2001;19:3163–3172. doi: 10.1200/JCO.2001.19.13.3163. [DOI] [PubMed] [Google Scholar]
  • 20.Pandit MK, Burke J, Gustafson AB, Minocha A, Peiris AN. Drug-induced disorders of glucose tolerance. Ann Intern Med. 1993;118:529–539. doi: 10.7326/0003-4819-118-7-199304010-00008. [DOI] [PubMed] [Google Scholar]
  • 21.Pui CH, Burghen GA, Bowman WP, Aur RJ. Risk factors for hyperglycemia in children with leukemia receiving L-asparaginase and prednisone. J Pediatr. 1981;99:46–50. doi: 10.1016/s0022-3476(81)80955-9. [DOI] [PubMed] [Google Scholar]
  • 22.Capizzi RL, Bertino JR, Skeel RT, et al. L-asparaginase: clinical, biochemical, pharmacological, and immunological studies. Ann Intern Med. 1971;74:893–901. doi: 10.7326/0003-4819-74-6-893. [DOI] [PubMed] [Google Scholar]
  • 23.Underwood TW, Frye CB. Drug-induced pancreatitis. Clin Pharm. 1993;12:440–448. [PubMed] [Google Scholar]
  • 24.Hoffmeister PA, Storer BE, Sanders JE. Diabetes mellitus in long-term survivors of pediatric hematopoietic cell transplantation. J Pediatr Hematol Oncol. 2004;26:81–90. doi: 10.1097/00043426-200402000-00003. [DOI] [PubMed] [Google Scholar]
  • 25.Robison LL, Mertens AC, Boice JD, et al. Study design and cohort characteristics of the Childhood Cancer Survivor Study: a multi-institutional collaborative project. Med Pediatr Oncol. 2002;38:229–239. doi: 10.1002/mpo.1316. [DOI] [PubMed] [Google Scholar]
  • 26.Louie AD, Robison LL, Bogue M, Hyde S, Forman SJ, Bhatia S. Validation of self-reported complications by bone marrow transplantation survivors. Bone Marrow Transplant. 2000;25:1191–1196. doi: 10.1038/sj.bmt.1702419. [DOI] [PubMed] [Google Scholar]
  • 27.Stevens J, Keil JE, Waid LR, Gazes PC. Accuracy of current, 4-year, and 28-year self-reported body weight in an elderly population. Am J Epidemiol. 1990;132:1156–1163. doi: 10.1093/oxfordjournals.aje.a115758. [DOI] [PubMed] [Google Scholar]
  • 28.Willett WC, Dietz WH, Colditz GA. Guidelines for healthy weight. N Engl J Med. 1999;341:427–434. doi: 10.1056/NEJM199908053410607. [DOI] [PubMed] [Google Scholar]
  • 29.Zeger SL, Liang KY. Longitudinal data analysis for discrete and continuous outcomes. Biometrics. 1986;42:121–130. [PubMed] [Google Scholar]
  • 30.Teinturier C, Tournade MF, Caillat-Zucman S, et al. Diabetes mellitus after abdominal radiation therapy. Lancet. 1995;346:633–634. doi: 10.1016/s0140-6736(95)91461-7. [DOI] [PubMed] [Google Scholar]
  • 31.Cicognani A, Cacciari E, Mancini AF, et al. Abnormal insulin response to glucose following treatment for Wilms' tumor in childhood. Eur J Pediatr. 1997;156:371–375. doi: 10.1007/s004310050617. [DOI] [PubMed] [Google Scholar]
  • 32.Hawkins MM, Robertson CM, Edge JA, Neil HA. Is risk of diabetes mellitus increased after abdominal radiotherapy? Lancet. 1996;347:539–540. [PubMed] [Google Scholar]
  • 33.Lorini R, Cortona L, Scaramuzza A, et al. Hyperinsulinemia in children and adolescents after bone marrow transplantation. Bone Marrow Transplant. 1995;15:873–877. [PubMed] [Google Scholar]
  • 34.Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 1988;37:1595–1607. doi: 10.2337/diab.37.12.1595. [DOI] [PubMed] [Google Scholar]
  • 35.Reaven GM, Laws A. Insulin resistance, compensatory hyperinsulinaemia, and coronary heart disease. Diabetologia. 1994;37:948–952. doi: 10.1007/BF00400953. [DOI] [PubMed] [Google Scholar]
  • 36.Pearson TA, Mensah GA, Alexander RW, et al. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: a statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation. 2003;107:499–511. doi: 10.1161/01.cir.0000052939.59093.45. [DOI] [PubMed] [Google Scholar]
  • 37.Liuzzo G, Biasucci LM, Gallimore JR, et al. The prognostic value of C-reactive protein and serum amyloid A protein in severe unstable angina. N Engl J Med. 1994;331:417–424. doi: 10.1056/NEJM199408183310701. [DOI] [PubMed] [Google Scholar]
  • 38.Ridker PM, Buring JE, Shih J, Matias M, Hennekens CH. Prospective study of C-reactive protein and the risk of future cardiovascular events among apparently healthy women. Circulation. 1998;98:731–733. doi: 10.1161/01.cir.98.8.731. [DOI] [PubMed] [Google Scholar]
  • 39.Koenig W, Sund M, Frohlich M, et al. C-reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from the MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Cohort Study, 1984 to 1992. Circulation. 1999;99:237–242. doi: 10.1161/01.cir.99.2.237. [DOI] [PubMed] [Google Scholar]
  • 40.Biasucci LM, Vitelli A, Liuzzo G, et al. Elevated levels of interleukin-6 in unstable angina. Circulation. 1996;94:874–877. doi: 10.1161/01.cir.94.5.874. [DOI] [PubMed] [Google Scholar]
  • 41.Miyao Y, Yasue H, Ogawa H, et al. Elevated plasma interleukin-6 levels in patients with acute myocardial infarction. Am Heart J. 1993;126:1299–1304. doi: 10.1016/0002-8703(93)90526-f. [DOI] [PubMed] [Google Scholar]
  • 42.Ridker PM, Rifai N, Pfeffer M, Sacks F, Lepage S, Braunwald E. Elevation of tumor necrosis factor-alpha and increased risk of recurrent coronary events after myocardial infarction. Circulation. 2000;101:2149–2153. doi: 10.1161/01.cir.101.18.2149. [DOI] [PubMed] [Google Scholar]
  • 43.Festa A, D'Agostino R, Jr, Howard G, Mykkanen L, Tracy RP, Haffner SM. Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS). Circulation. 2000;102:42–47. doi: 10.1161/01.cir.102.1.42. [DOI] [PubMed] [Google Scholar]
  • 44.McLaughlin T, Abbasi F, Lamendola C, et al. Differentiation between obesity and insulin resistance in the association with C-reactive protein. Circulation. 2002;106:2908–2912. doi: 10.1161/01.cir.0000041046.32962.86. [DOI] [PubMed] [Google Scholar]
  • 45.Schots R, Kaufman L, Van Riet I, et al. Proinflammatory cytokines and their role in the development of major transplant-related complications in the early phase after allogeneic bone marrow transplantation. Leukemia. 2003;17:1150–1156. doi: 10.1038/sj.leu.2402946. [DOI] [PubMed] [Google Scholar]
  • 46.Sakata N, Yasui M, Okamura T, Inoue M, Yumura-Yagi K, Kawa K. Kinetics of plasma cytokines after hematopoietic stem cell transplantation from unrelated donors: the ratio of plasma IL-10/sTNFR level as a potential prognostic marker in severe acute graft-versus-host disease. Bone Marrow Transplant. 2001;27:1153–1161. doi: 10.1038/sj.bmt.1703060. [DOI] [PubMed] [Google Scholar]
  • 47.Deeg HJ. Cytokines in graft-versus-host disease and the graft-versus-leukemia reaction. Int J Hematol. 2001;74:26–32. doi: 10.1007/BF02982546. [DOI] [PubMed] [Google Scholar]
  • 48.Ferrara JL. Pathogenesis of acute graft-versus-host disease: cytokines and cellular effectors. J Hematother Stem Cell Res. 2000;9:299–306. doi: 10.1089/15258160050079407. [DOI] [PubMed] [Google Scholar]
  • 49.Krenger W, Ferrara JL. Dysregulation of cytokines during graft-versus-host disease. J Hematother. 1996;5:3–14. doi: 10.1089/scd.1.1996.5.3. [DOI] [PubMed] [Google Scholar]
  • 50.Reimold AM. New indications for treatment of chronic inflammation by TNF-alpha blockade. Am J Med Sci. 2003;325:75–92. doi: 10.1097/00000441-200302000-00005. [DOI] [PubMed] [Google Scholar]
  • 51.Jacobsohn DA, Hallick J, Anders V, McMillan S, Morris L, Vogelsang GB. Infliximab for steroid-refractory acute GVHD: a case series. Am J Hematol. 2003;74:119–124. doi: 10.1002/ajh.10392. [DOI] [PubMed] [Google Scholar]
  • 52.Bruner RJ, Farag SS. Monoclonal antibodies for the prevention and treatment of graft-versus-host disease. Semin Oncol. 2003;30:509–519. doi: 10.1016/s0093-7754(03)00250-1. [DOI] [PubMed] [Google Scholar]
  • 53.Oeffinger KC, Mertens AC, Sklar CA, et al. Obesity in adult survivors of childhood acute lymphoblastic leukemia: a report from the childhood cancer survivor study. J Clin Oncol. 2003;17:1359–1365. doi: 10.1200/JCO.2003.06.131. [DOI] [PubMed] [Google Scholar]
  • 54.Sobecks RM, Daugherty CK, Hallahan DE, Laport GF, Wagner ND, Larson RA. A dose escalation study of total body irradiation followed by high-dose etoposide and allogeneic blood stem cell transplantation for the treatment of advanced hematologic malignancies. Bone Marrow Transplant. 2000;25:807–813. doi: 10.1038/sj.bmt.1702230. [DOI] [PubMed] [Google Scholar]
  • 55.Shank B. Total body irradiation for marrow or stem-cell transplantation. Cancer Invest. 1998;16:397–404. doi: 10.3109/07357909809115780. [DOI] [PubMed] [Google Scholar]
  • 56.Evans RG. Radiobiological considerations in magna-field irradiation. Int J Radiat Oncol Biol Phys. 1983;9:1907–1911. doi: 10.1016/0360-3016(83)90361-9. [DOI] [PubMed] [Google Scholar]
  • 57.Vriesendorp HM. Aims of conditioning. Exp Hematol. 2003;31:844–854. doi: 10.1016/s0301-472x(03)00229-7. [DOI] [PubMed] [Google Scholar]
  • 58.Down JD, Boudewijn A, van Os R, Thames HD, Ploemacher RE. Variations in radiation sensitivity and repair among different hematopoietic stem cell subsets following fractionated irradiation. Blood. 1995;86:122–127. [PubMed] [Google Scholar]

Articles from Blood are provided here courtesy of The American Society of Hematology

RESOURCES