Allogeneic peripheral blood stem cell grafts contain about 10 times more T and B cells than marrow grafts. Because these cells may survive in transplant recipients for a long time, recipients of blood stem cells may be less immunocompromised than recipients of marrow. Immune reconstitution was studied in 115 patients randomly assigned to receive either allogeneic marrow or filgrastim-mobilized blood stem cell transplantation. Between day 30 and 365 after transplantation, counts of most lymphocyte subsets were higher in the blood stem cell recipients. The difference was most striking for CD4 T cells (about 4-fold higher counts for CD45RAhigh CD4 T cells and about 2-fold higher counts for CD45RAlow/−CD4 T cells;P < .05). On assessment using phytohemagglutinin and herpesvirus antigen-stimulated proliferation, T cells in the 2 groups of patients appeared equally functional. Median serum IgG levels were similar in the 2 groups. The rate of definite infections after engraftment was 1.7-fold higher in marrow recipients (P = .001). The rate of severe (inpatient treatment required) definite infections after engraftment was 2.4-fold higher in marrow recipients (P = .002). The difference in the rates of definite infections was greatest for fungal infections, intermediate for bacterial infections, and lowest for viral infections. Death associated with a fungal or bacterial infection occurred between day 30 and day 365 after transplantation in 9 marrow recipients and no blood stem cell recipients (P = .008). In conclusion, blood stem cell recipients have higher lymphocyte-subset counts and this appears to result in fewer infections.

Allogeneic bone marrow transplantation is a recognized treatment for certain hematologic malignant diseases, aplastic anemia, and inborn errors of cells originating from hematopoietic stem cells.1,2 Immune deficiency leading to increased susceptibility to infections follows transplantation and lasts for more than a year.3-8 Although infections that occur in the first month after grafting likely result from a deficiency of both granulocytes and mononuclear cell (MNC) subsets, postengraftment infections are probably due to a deficiency in MNC subsets, primarily CD4 T cells and B cells.9-11Postengraftment infections cause substantial morbidity and mortality. Atkinson et al3 showed that 87% of allograft recipients had at least one postengraftment infection and 48% had at least 3 (excluding upper respiratory tract infections) during a 2-year period. A similar incidence of postengraftment infections was observed by Ochs et al,7 who also noted that occurrence of postengraftment infection was the dominant independent factor associated with increased nonrelapse mortality (relative risk = 5.5;P = .0001).

Recipients of blood stem cells receive at least 10 times more lymphocytes than recipients of marrow.12,13 The lifespan of human T cells is months to years.14-16 Therefore, we hypothesized that the large lymphocyte inoculum given to blood stem cell recipients might result in higher T-cell counts in the first year after transplantation. Results of small nonrandomized studies of blood stem cell recipients compared with marrow recipients support this hypothesis.12,13 We also hypothesized that higher lymphocyte counts in blood stem cell recipients could lead to lower infection rates. Thus, the purpose of this randomized study was to analyze MNC-subset recovery and postengraftment infection rates in marrow grafting compared with blood stem cell grafting.

Patients

Of 140 patients who were randomly assigned to undergo either marrow or filgrastim-mobilized blood stem cell allografting, treated at the Fred Hutchinson Cancer Research Center (FHCRC) as reported previously,17 and followed for at least a year, 128 patients agreed to participate in immune reconstitution studies approved by the institutional review board of the FHCRC. One of these patients received marrow instead of blood stem cells, and 12 patients (7 marrow recipients and 5 blood stem cell recipients) died by day 30 after transplantation. Thus, 115 patients were available for MNC-subset testing between day 30 and 365 and could be evaluated for postengraftment infections. Their demographic and clinical characteristics, including infection prophylaxis given, are shown in Table 1. Because the goal of the study was to evaluate transplantation-associated immune deficiency and not malignancy-associated immune deficiency, patients who had a relapse after transplantation were studied only before the diagnosis of relapse. The interval between day 30 and day 365 or the day of death or relapse is referred to in this paper as the interval between “day 30 and day 365.”

Table 1.

Characteristics of patients given marrow as compared with those given blood as the source of hematopoietic cells for transplantation

Patients given marrow (n = 60)Patients given blood (n = 55)P
Patients' median (range) age at Tx, y 43 (22-56) 43 (15-56) .57  
Donors' median (range) age at Tx, y 41 (18-65) 40 (13-63) .34 
Patients' sex (M/F) 39/21 35/20 .97 
Donors' sex (M/F) 32/28 31/24 .89 
Donor-patient histocompatibility    
 HLA-A, B, and DR-matched sibling 60 (100) 54 (98) .97  
 HLA-A, B, and DR-matched child 0 (0) 1 (2)  
Disease/disease stage at Tx*    
 Good risk 34 (57) 31 (56) .88  
 Poor risk 26 (43) 24 (44)  
First Tx 60 (100) 55 (100) 1.00  
CMV serostatus before Tx    
 Donor positive/recipient positive 22 (37) 15 (27) .63  
 Donor negative/recipient positive 11 (18) 14 (25)  
 Donor positive/recipient negative 10 (17) 11 (20)  
 Donor negative/recipient negative 17 (28) 14 (25)  
 Unknown or equivocal 0 (0) 1 (2)  
HSV serostatus before Tx    
 Patient positive 44 (73) 35 (64) .73 
 Patient negative 11 (18) 14 (25)  
 Unknown or equivocal 5 (8) 6 (11)  
VZV serostatus before Tx    
 Patient positive 58 (97) 53 (96) .45 
 Patient negative 0 (0) 2 (4)  
 Unknown or equivocal 2 (3) 0 (0)  
Patients with splenectomy 3 (5) 1 (2) .67  
Myeloablative conditioning    
 Chemotherapy only 30 (50) 27 (49) .33  
 Chemotherapy and total-body irradiation 29 (48) 24 (44)  
 Chemotherapy and total-marrow irradiation 1 (2) 4 (7)  
GVHD prophylaxis with methotrexate (day 1, 3, 6, and 11) and cyclosporine (daily for 6 months)58 60 (100) 55 (100) 1.00  
Acute GVHD    
 Grade 0-1 16 (27) 16 (29) .94 
 Grade 2-4 44 (73) 39 (71)  
Chronic GVHD diagnosed by day 365    
 None or clinically limited 25 (42) 18 (33) .16  
 Clinically extensive 23 (38) 32 (58)  
 Not applicable (relapse or death before day 100) 12 (20) 5 (9)  
Chimerism status    
 Full chimera (≥ 90% marrow cells of donor origin
by day 80) 
52 (87) 47 (85) 1.00 
 Unknown 8 (13) 8 (15)  
Median (range) day of neutrophil engraftment
(> 0.5 × 109/L) 
20 (18-28) 16 (11-28) < .01 
Neutropenia (< 0.5 × 109/L) between day 30 day and 3651-153 7 (12) 4 (7) .63  
Relapse between day 30 and day 3651-155 8 (13) 8 (15) .94  
Death without relapse between day 30 and day 365 18 (30) 6 (11) .02 
Glucocorticoid treatment between day 30 and day 365 51 (86) 45 (82) .84  
CMV prophylaxis    
 Ganciclovir if pp65-antigenemia positive59 53 (88) 51 (93) .63  
 Infusion of ex vivo–expanded CMV-specific T cells601-154 7 (12) 4 (7)  
Viral prophylaxis with acyclovir/valacyclovir/famciclovir beyond standard1-159    
 Yes 22 (37) 14 (25) .20 
 No 33 (55) 39 (71)  
 Unknown 5 (8) 2 (4)  
Bacterial/Pneumocystis/fungal prophylaxis with sulfamethoxazole and trimethoprim and/or fluconazole/itraconazole beyond standard*,*    
 Yes 24 (40) 29 (53) .46 
 No 19 (32) 15 (27)  
 Not applicable (early relapse or death) or unknown 17 (28) 11 (20)  
Prophylaxis with intravenous immunoglobulin, 8 (13) 6 (11) .91 
Patients given marrow (n = 60)Patients given blood (n = 55)P
Patients' median (range) age at Tx, y 43 (22-56) 43 (15-56) .57  
Donors' median (range) age at Tx, y 41 (18-65) 40 (13-63) .34 
Patients' sex (M/F) 39/21 35/20 .97 
Donors' sex (M/F) 32/28 31/24 .89 
Donor-patient histocompatibility    
 HLA-A, B, and DR-matched sibling 60 (100) 54 (98) .97  
 HLA-A, B, and DR-matched child 0 (0) 1 (2)  
Disease/disease stage at Tx*    
 Good risk 34 (57) 31 (56) .88  
 Poor risk 26 (43) 24 (44)  
First Tx 60 (100) 55 (100) 1.00  
CMV serostatus before Tx    
 Donor positive/recipient positive 22 (37) 15 (27) .63  
 Donor negative/recipient positive 11 (18) 14 (25)  
 Donor positive/recipient negative 10 (17) 11 (20)  
 Donor negative/recipient negative 17 (28) 14 (25)  
 Unknown or equivocal 0 (0) 1 (2)  
HSV serostatus before Tx    
 Patient positive 44 (73) 35 (64) .73 
 Patient negative 11 (18) 14 (25)  
 Unknown or equivocal 5 (8) 6 (11)  
VZV serostatus before Tx    
 Patient positive 58 (97) 53 (96) .45 
 Patient negative 0 (0) 2 (4)  
 Unknown or equivocal 2 (3) 0 (0)  
Patients with splenectomy 3 (5) 1 (2) .67  
Myeloablative conditioning    
 Chemotherapy only 30 (50) 27 (49) .33  
 Chemotherapy and total-body irradiation 29 (48) 24 (44)  
 Chemotherapy and total-marrow irradiation 1 (2) 4 (7)  
GVHD prophylaxis with methotrexate (day 1, 3, 6, and 11) and cyclosporine (daily for 6 months)58 60 (100) 55 (100) 1.00  
Acute GVHD    
 Grade 0-1 16 (27) 16 (29) .94 
 Grade 2-4 44 (73) 39 (71)  
Chronic GVHD diagnosed by day 365    
 None or clinically limited 25 (42) 18 (33) .16  
 Clinically extensive 23 (38) 32 (58)  
 Not applicable (relapse or death before day 100) 12 (20) 5 (9)  
Chimerism status    
 Full chimera (≥ 90% marrow cells of donor origin
by day 80) 
52 (87) 47 (85) 1.00 
 Unknown 8 (13) 8 (15)  
Median (range) day of neutrophil engraftment
(> 0.5 × 109/L) 
20 (18-28) 16 (11-28) < .01 
Neutropenia (< 0.5 × 109/L) between day 30 day and 3651-153 7 (12) 4 (7) .63  
Relapse between day 30 and day 3651-155 8 (13) 8 (15) .94  
Death without relapse between day 30 and day 365 18 (30) 6 (11) .02 
Glucocorticoid treatment between day 30 and day 365 51 (86) 45 (82) .84  
CMV prophylaxis    
 Ganciclovir if pp65-antigenemia positive59 53 (88) 51 (93) .63  
 Infusion of ex vivo–expanded CMV-specific T cells601-154 7 (12) 4 (7)  
Viral prophylaxis with acyclovir/valacyclovir/famciclovir beyond standard1-159    
 Yes 22 (37) 14 (25) .20 
 No 33 (55) 39 (71)  
 Unknown 5 (8) 2 (4)  
Bacterial/Pneumocystis/fungal prophylaxis with sulfamethoxazole and trimethoprim and/or fluconazole/itraconazole beyond standard*,*    
 Yes 24 (40) 29 (53) .46 
 No 19 (32) 15 (27)  
 Not applicable (early relapse or death) or unknown 17 (28) 11 (20)  
Prophylaxis with intravenous immunoglobulin, 8 (13) 6 (11) .91 

Values are numbers (percentages) of patients unless otherwise indicated. The significance of differences between patients given marrow and those given blood stem cells was assessed by eithert test (age and neutrophil engraftment) or χ2 square test (all other variables).

Tx indicates transplantation; CMV, cytomegalovirus; HSV, herpes simplex virus; VZV, varicella zoster virus; GVHD, graft-versus-host disease.

*

Good risk was defined as the presence of chronic myelogenous leukemia (CML) in first chronic or accelerated phase, acute leukemia in first remission, refractory anemia, or myelofibrosis. Poor risk was defined as the presence of CML in blast crisis, acute leukemia beyond first remission, refractory anemia with excess blasts, lymphoma, or multiple myeloma.

Typically treated with prednisone (1-2 mg/kg per day given orally for 10 to 14 days with subsequent tapering during 50 days).

Typically treated with prednisone (0.5-1.0 mg/kg given orally every other day) and cyclosporine (approximately 6 mg/kg given orally every other day) for at least 9 months.

F1-153

Values are numbers of episodes. There was one episode per patient. No episode was due to graft rejection. Episodes were typically associated with a viral infection and lasted for several days.

F1-155

For patients with CML, detection of bcr/abl transcript by polymerase chain reaction in the absence of cytogenetic or hematologic relapse was not considered a relapse.

F1-154

Values were typically 1 × 109 CD4 cells/m2 and 2 × 109 CD8 cells/m2 of body-surface area.

F1-159

According to standard practice, all patients seropositive for herpes simplex virus received acyclovir (800 mg twice a day given orally) during the first month only. Some patients or physicians chose prophylaxis with acyclovir, valacyclovir, or famciclovir between day 30 and day 365 after transplantation; the patients are included here.

F1-160

According to standard practice, all patients received prophylactic sulfamethoxazole and trimethoprim until day 180 after transplantation (800 mg sulfamethoxazole and 160 mg trimethoprim given orally twice a day every Monday and Tuesday) and fluconazole until day 75 (400 mg given orally every day). Itraconazole was given instead of fluconazole to 5 marrow recipients and 2 blood stem cell recipients. After the end of the routine bacterial/Pneumocystis/fungal prophylaxis, some patients or physicians chose to continue prophylactic therapy; the patients are counted here. Typically, these were patients with clinically extensive chronic GVHD who were treated with immunosuppressive drugs, sulfamethoxazole and trimethoprim (800 mg and 160 mg, respectively, given orally twice a day every Monday and Tuesday), penicillin (500 mg given orally twice a day every day), and rarely, fluconazole or itraconazole.

F1-164

Typically, 200 mg/kg was given weekly before day 100 after transplantation, and 500 mg/kg was given monthly after day 100.

Enumeration of MNC subsets

Blood specimens were drawn from donors before transplantation (before filgrastim was used) and from recipients on about day 30 after transplantation (median, day 31 for marrow and day 32 for blood stem cell recipients), day 80 (median, day 78 and day 79), day 180 (median, day 182 and day 188), and day 365 (median, day 377 and day 376). The days after transplantation on which blood was drawn were not significantly different between the 2 groups of recipients (byt test). With use of density gradient (Ficoll) centrifugation, MNCs were separated from blood specimens and not separated from marrow or blood stem cell grafts. Cells were stained with fluorochrome-conjugated monoclonal antibodies and analyzed by using 3-color flow cytometry as described previously.18,19The antigenic phenotypes defining each MNC subset are detailed in Table2. The rationales for defining the B- and T-cell subsets in Table 2 were as follows. Naive B cells are represented by IgD+ B cells, since most IgD+ B cells lack somatic mutations.20-22 Naive CD4 T cells were defined as CD45RAhigh CD4 T cells, since this subset contains thymic emigrants and nearly all cord blood CD4 T cells are CD45RAhigh.23-25 Naive CD8 T cells were defined as CD11alow CD8 T cells because virtually all cord blood CD8 T cells are CD11alow and become CD11ahigh after activation.26,27CD28+ T cells represent cells that can receive both the signal mediated by the T-cell receptor and the CD28-mediated costimulatory signal.28 Each absolute MNC-subset count was calculated as the absolute MNC count multiplied by the percentage of the MNC subset divided by 100. The absolute MNC count represented the sum of the absolute lymphocyte count and the absolute monocyte count determined by a clinical hematology laboratory. For grafts, total nucleated cells were gated on forward x side-scatter plots and each absolute cell-subset count was calculated as the absolute nucleated-cell count multiplied by the percentage of the cell subset among total nucleated cells divided by 100.

Table 2.

Definitions of subsets of mononuclear cells

MNC subsetDefinition
B cells MNCs expressing CD19 or CD20 and not brightly expressing CD3, CD13, CD14, CD16, CD56, CD10, or CD34 
 Naive (IgD+B cells (defined as above) expressing surface IgD  
 Memory (IgDB cells (defined as above) not expressing surface IgD  
CD4 T cells (total) CD3+ CD4+ CD8 MNCs 
 Naive (CD45RAhighMNCs expressing CD4, brightly expressing CD45RA, and not expressing CD8, CD13, CD14, or CD16 
 Memory or effector (CD45RAlow/−MNCs expressing CD4, dimly expressing or not expressing CD45RA, and not expressing CD8, CD13, CD14, or CD16  
 Costimulation competent (CD28+MNCs expressing CD4, expressing CD28, and not expressing CD8, CD13, CD14, or CD16  
 Costimulation incompetent (CD28MNCs expressing CD4, not expressing CD28, and not expressing CD8, CD13, CD14, or CD16  
CD8 T cells (total) CD3+ CD4 CD8+ MNCs 
 Naive (CD11alowMNCs brightly expressing CD8, dimly expressing CD11a, and not expressing CD4, CD13, CD14, CD16, or CD19  
 Memory or effector (CD11ahighMNCs brightly expressing CD8, brightly expressing CD11a, and not expressing CD4, CD13, CD14, CD16, or CD19  
 Costimulation competent (CD28+MNCs brightly expressing CD8, expressing CD28, and not expressing CD4, CD13, CD14, or CD16  
 Costimulation incompetent (CD28MNCs brightly expressing CD8, not expressing CD28, and not expressing CD4, CD13, CD14, or CD16 
CD4 CD8 T cells CD3+CD4 CD8 MNCs  
CD8+CD8+ T cells CD3+ CD4+CD8+ MNCs  
Natural killer cells MNCs expressing CD16 or CD56 and not expressing CD3 or CD14 
Monocytes CD14+ MNCs  
“Stem cells” CD34+ CD14 cells  
B-cell progenitors Cells expressing CD19 or CD20 together with CD10 or CD34 and not brightly expressing CD3, CD13, CD14, CD16, or CD56  
Plasma cells Cells expressing CD38 very brightly and not expressing CD3, CD10, CD13, CD14, CD16, CD34, or CD56 
MNC subsetDefinition
B cells MNCs expressing CD19 or CD20 and not brightly expressing CD3, CD13, CD14, CD16, CD56, CD10, or CD34 
 Naive (IgD+B cells (defined as above) expressing surface IgD  
 Memory (IgDB cells (defined as above) not expressing surface IgD  
CD4 T cells (total) CD3+ CD4+ CD8 MNCs 
 Naive (CD45RAhighMNCs expressing CD4, brightly expressing CD45RA, and not expressing CD8, CD13, CD14, or CD16 
 Memory or effector (CD45RAlow/−MNCs expressing CD4, dimly expressing or not expressing CD45RA, and not expressing CD8, CD13, CD14, or CD16  
 Costimulation competent (CD28+MNCs expressing CD4, expressing CD28, and not expressing CD8, CD13, CD14, or CD16  
 Costimulation incompetent (CD28MNCs expressing CD4, not expressing CD28, and not expressing CD8, CD13, CD14, or CD16  
CD8 T cells (total) CD3+ CD4 CD8+ MNCs 
 Naive (CD11alowMNCs brightly expressing CD8, dimly expressing CD11a, and not expressing CD4, CD13, CD14, CD16, or CD19  
 Memory or effector (CD11ahighMNCs brightly expressing CD8, brightly expressing CD11a, and not expressing CD4, CD13, CD14, CD16, or CD19  
 Costimulation competent (CD28+MNCs brightly expressing CD8, expressing CD28, and not expressing CD4, CD13, CD14, or CD16  
 Costimulation incompetent (CD28MNCs brightly expressing CD8, not expressing CD28, and not expressing CD4, CD13, CD14, or CD16 
CD4 CD8 T cells CD3+CD4 CD8 MNCs  
CD8+CD8+ T cells CD3+ CD4+CD8+ MNCs  
Natural killer cells MNCs expressing CD16 or CD56 and not expressing CD3 or CD14 
Monocytes CD14+ MNCs  
“Stem cells” CD34+ CD14 cells  
B-cell progenitors Cells expressing CD19 or CD20 together with CD10 or CD34 and not brightly expressing CD3, CD13, CD14, CD16, or CD56  
Plasma cells Cells expressing CD38 very brightly and not expressing CD3, CD10, CD13, CD14, CD16, CD34, or CD56 

Cells with forward-scatter compared with side-scatter characteristics of lymphocytes and monocytes.

MNC indicates mononuclear cell; IgD, immunoglobulin D.

Lymphoproliferation assays

Herpes simplex virus (HSV)–specific, varicella zoster virus (VZV)–specific T-helper-cell function and phytohemagglutinin (PHA)-stimulated T-cell proliferation were assessed as described previously.29,30 Briefly, MNCs separated by a Ficoll device were suspended at a concentration of 2 × 106cells/mL in RPMI with 10% human AB serum, glutamine (4 mM), penicillin (100 U/mL), streptomycin (0.1 mg/mL), and amphotericin B (250 ng/mL). One hundred microliters of the peripheral blood mononuclear cell suspension was dispensed into wells of 96 round-bottomed plates. VZV and HSV antigens were prepared by sonication and heat inactivation of virus grown on foreskin fibroblasts. HSV or VZV antigen or PHA (final concentration, 10 μg/mL) was added to triplicate wells containing MNCs on day zero. The plates were incubated at 37°C in a humidified 5% carbon dioxide atmosphere for 4 days. Eighteen to 24 hours before harvest, tritium thymidine (0.074 MBq [2 μCi]) was added to each well. Cells were harvested by using a semiautomated harvester, and counts per minute (cpm) were determined in a β-scintillation counter. The mean cpm of cells exposed to the antigen minus the mean cpm of cells incubated with medium alone (Δ cpm) was calculated. Because the percentage of CD4 cells, as well as total T cells, among MNCs was higher in the blood stem cell recipients than in the marrow recipients, results were expressed as Δ cpm per 1000 CD4 T cells (HSV and VZV assessment) or per 1000 T cells (PHA assessment).

IgG levels

Serum IgG levels were determined by standard rate nephelometry.

Enumeration of infections

The goal of this study was to determine whether higher lymphocyte counts after blood stem cell grafting would be associated with an infection rate lower than that after marrow grafting. Because blood stem cell grafting is associated with earlier neutrophil engraftment,17 only postengraftment infections were counted. These were defined as infections diagnosed between day 30 and day 365, since both marrow recipients and blood stem cell recipients achieved neutrophil engraftment by day 30 (Table 1) and the minimum follow-up period was 365 days.

Definite infection was defined as an illness with symptoms and signs consistent with an infection and microbiological documentation of a pathogen, except in cases of dermatomal zoster, for which a clinical diagnosis was considered sufficient to classify an infection as definite. Microbiological documentation of an infection consisted of isolation of the pathogen by culture from a sterile or nonsterile site (if from a nonsterile site, the organism had to be clinically judged to be pathogenic) or histologic or immunohistologic evidence. Culture-documented viremia, bacteremia, or fungemia was considered a definite infection even if the patient had no symptoms or signs of infection. Oral candidiasis was not considered a definite infection because it could not be reliably distinguished from oral graft-versus-host disease (GVHD) with candidal colonization.

Clinical infection (no microorganism identified) was defined as illness with symptoms and signs consistent with an infection. Presumed oral, gastrointestinal, conjunctival, and respiratory tract infections were not included, however, because they could not be reliably distinguished from GVHD or allergy. Hemorrhagic cystitis was not included because it could not be differentiated from conditioning-regimen–induced cystitis. Fever of presumed infectious cause was included only if the patient's body temperature was above 38.5°C and the condition responded to antibiotic therapy within 3 days. Sinusitis and pneumonia were included only if documented radiologically.

A chronic infection was counted as one infection. A recurrent infection was counted as multiple infections only if episodes were clearly separated by an asymptomatic period of longer than 4 weeks. A polymicrobial infection of one organ or several adjacent organs was considered one infection (due to the organism judged to be the major pathogen). Infections with one microorganism in 2 nonadjacent organs were counted as 2 infections. The respiratory tract was considered adjacent to the paranasal sinuses and lungs. Lungs and paranasal sinuses were considered nonadjacent. An organ infection with viremia, bacteremia, or fungemia was counted only as the organ infection. Severe infections were defined as infections treated in a hospital. Nonsevere infections were treated in an outpatient setting.

Death associated with a definite infection was defined as (1) autopsy findings consistent with an infection and detection of the pathogen in an autopsy specimen, or (2) death after a definite infection that was judged to have caused the death either directly (eg, pneumonia) or indirectly (eg, sepsis with subsequent adult respiratory distress syndrome).

Statistical analysis

The significance of differences between marrow recipients and blood stem cell recipients in MNC-subset counts, Δ cpm, and serum IgG levels at each time point was tested by the Mann-Whitney rank sum test. The significance of differences in infection rates was determined by the likelihood ratio test. The number of infections was treated as a Poisson random variable. Regression models were fit with the SAS Genmod (SAS Institute, Cary, NC) procedure using the log of the number of days at risk as a fixed predictor (offset). Days at risk were calculated as 365 or the day after transplantation of death or relapse (whatever occurred first) minus 30.

MNC subsets

Compared with marrow recipients, blood stem cell recipients received significantly higher numbers of all subsets of MNCs with the graft, except for B-cell progenitors and plasma cells (Table3). In the first year after grafting, counts of the following MNC subsets were significantly higher in blood stem cell recipients at 3 or more time points (Figure1): total CD4 T cells and their CD45RAhigh (naive), CD45RAlow/−(memory/effector) and CD28+ subpopulations, CD8 T cells and their CD11alow (naive) and CD28+subpopulations, and CD4CD8 T cells. Subsets not significantly different at any time point after transplantation included CD28CD8 T cells and monocytes. MNC subsets with significantly greater numbers in marrow recipients than in blood stem cell recipients early (on day 30 or day 80) but not late (on day 180 or day 365) after transplantation included total B cells and their IgD+ (naive) and IgD (memory) subsets, CD11ahigh (memory/effector) CD8 T cells, and natural killer (NK) cells. Conversely, CD28CD4 T-cell counts and CD4+CD8+ T cell counts were similar in the 2 groups of recipients early after transplantation but higher in blood stem cell recipients later. MNC-subset counts in marrow donors and those in blood stem cell donors (before use of filgrastim) were similar (P > 0.1 for all the subsets). The data from donors were pooled and are shown in Figure 1 as the normal reference values.

Table 3.

Quantities of cells infused into patients, according to mononuclear cell subset and source of hematopoietic cells (marrow or blood)

MNC subsetMarrowBlood3-150Fold increase3-151
B cells (total) 4 (1-25) 71 (6-141) 18 
 Naive (IgD+3 (1-22) 62 (22-129) 20 
 Memory (IgD0.8 (0.1-3.1) 9.6 (0.9-33.8) 12 
CD4 T cells (total) 14 (2-29) 174 (64-414) 12 
 Naive (CD45RAhigh6 (0-16) 60 (19-233) 10 
 Memory or effector (CD45RAlow/−8 (1-25) 115 (1-204) 14 
 Costimulation competent (CD28+14 (1-30) 156 (57-348) 11 
 Costimulation incompetent (CD280.4 (0.0-12.8) 4.0 (0.8-50.7) 10 
CD8 T cells (total) 10 (1-31) 82 (22-254) 
 Naive (CD11alow4 (0-13) 34 (15-114) 
 Memory or effector (CD11ahigh7 (1-17) 34 (7-106) 
 Costimulation competent (CD28+7 (1-20) 55 (16-142) 
 Costimulation incompetent (CD283 (0-8) 11 (2-49) 
CD4CD8 T cells 0.6 (0.1-2.9) 4.8 (0.9-19.5) 
CD8+CD8+ T cells 1.0 (0.1-7.9) 6.5 (0.8-101.6) 
Natural killer cells 3 (0-13) 30 (13-71) 10 
Monocytes3-152 26 (0-111) 547 (150-2142) 21 
“Stem cells” 2.5 (1.0-8.0) 7.4 (1.0-17.5) 
B-cell progenitors 4 (0-22) 0 (0-2) 
Plasma cells 0.5 (0.0-4.7) 0.0 (0.0-0.7) 
MNC subsetMarrowBlood3-150Fold increase3-151
B cells (total) 4 (1-25) 71 (6-141) 18 
 Naive (IgD+3 (1-22) 62 (22-129) 20 
 Memory (IgD0.8 (0.1-3.1) 9.6 (0.9-33.8) 12 
CD4 T cells (total) 14 (2-29) 174 (64-414) 12 
 Naive (CD45RAhigh6 (0-16) 60 (19-233) 10 
 Memory or effector (CD45RAlow/−8 (1-25) 115 (1-204) 14 
 Costimulation competent (CD28+14 (1-30) 156 (57-348) 11 
 Costimulation incompetent (CD280.4 (0.0-12.8) 4.0 (0.8-50.7) 10 
CD8 T cells (total) 10 (1-31) 82 (22-254) 
 Naive (CD11alow4 (0-13) 34 (15-114) 
 Memory or effector (CD11ahigh7 (1-17) 34 (7-106) 
 Costimulation competent (CD28+7 (1-20) 55 (16-142) 
 Costimulation incompetent (CD283 (0-8) 11 (2-49) 
CD4CD8 T cells 0.6 (0.1-2.9) 4.8 (0.9-19.5) 
CD8+CD8+ T cells 1.0 (0.1-7.9) 6.5 (0.8-101.6) 
Natural killer cells 3 (0-13) 30 (13-71) 10 
Monocytes3-152 26 (0-111) 547 (150-2142) 21 
“Stem cells” 2.5 (1.0-8.0) 7.4 (1.0-17.5) 
B-cell progenitors 4 (0-22) 0 (0-2) 
Plasma cells 0.5 (0.0-4.7) 0.0 (0.0-0.7) 

Values are median (range) number of cells × 106/kg of body weight of recipient. The difference between the marrow and the blood stem cell graft was significant for all the cell subsets (P < .001).

For abbreviations, see Table 2.

F3-150

Eighteen recipients of blood stem cells received 2 MNC apheresis products. The values given represent the sum of the 2 products.

F3-151

Median for blood stem cell grafts compared with median for marrow grafts.

F3-152

When monocytes were defined as CD14highinstead of CD14+ MNCs, median values were 2 × 106/kg for marrow cells and 221 × 106/kg for blood stem cells (a 111-fold increase).

Fig. 1.

Median MNC-subset counts in recipients of marrow and blood stem cells on about day 30, 80, 180, and 365 after transplantation.

Gray circles indicate marrow; black triangles, stem cells. Error bars indicate the 25th to 75th percentiles. Stars indicate a significant difference (P < .05). Normal values are shown as horizontal lines (thick solid line for the 10th and 90th percentiles and broken line for the median). Days after transplantation are shown on all x-axes. On all y-axes, values for cell counts are per microliter. The numbers of marrow recipients and blood stem cell recipients studied were 43 and 45, respectively, on day 30; 40 and 40 on day 80; 29 and 26 on day 180; and 25 and 30 on day 365.

Fig. 1.

Median MNC-subset counts in recipients of marrow and blood stem cells on about day 30, 80, 180, and 365 after transplantation.

Gray circles indicate marrow; black triangles, stem cells. Error bars indicate the 25th to 75th percentiles. Stars indicate a significant difference (P < .05). Normal values are shown as horizontal lines (thick solid line for the 10th and 90th percentiles and broken line for the median). Days after transplantation are shown on all x-axes. On all y-axes, values for cell counts are per microliter. The numbers of marrow recipients and blood stem cell recipients studied were 43 and 45, respectively, on day 30; 40 and 40 on day 80; 29 and 26 on day 180; and 25 and 30 on day 365.

Close modal

Lymphoproliferation in vitro

To assess whether the functional capacity of T cells was different in blood stem cell recipients compared with marrow recipients, PHA-stimulated proliferation of MNCs corrected for the percentage of T cells among MNCs was evaluated. The median corrected Δ cpm was 1.5 times higher in blood stem cell recipients on day 30 (P = .04) and not significantly different on day 80, 180, or 365 (Figure 2). This suggests that during most of the first year after transplantation, the functional capacity of single T cells is similar in marrow recipients and blood stem cell recipients.

Fig. 2.

Results of lymphoproliferation assays using stimulation with PHA, HSV, and VZV.

The symbols are the same as in Figure 1. Only HSV (VZV)–infected patients with HSV (VZV)–infected donors were included in the analysis of HSV (VZV)–induced proliferation. The numbers of marrow recipients and blood stem cell recipients studied with PHA were 41 and 41, respectively, on day 30; 38 and 42 on day 80; 31 and 33 on day 180; and 27 and 30 on day 365. For HSV, the numbers of marrow recipients and blood stem cell recipients studied were 13 and 22, respectively, on day 30; 13 and 20 on day 80; 8 and 16 on day 180; and 6 and 14 on day 365. For VZV, the numbers of marrow recipients and blood stem cell recipients studied were 16 and 19, respectively, on day 30; 15 and 17 on day 80; 11 and 14 on day 180; and 10 and 12 on day 365.

Fig. 2.

Results of lymphoproliferation assays using stimulation with PHA, HSV, and VZV.

The symbols are the same as in Figure 1. Only HSV (VZV)–infected patients with HSV (VZV)–infected donors were included in the analysis of HSV (VZV)–induced proliferation. The numbers of marrow recipients and blood stem cell recipients studied with PHA were 41 and 41, respectively, on day 30; 38 and 42 on day 80; 31 and 33 on day 180; and 27 and 30 on day 365. For HSV, the numbers of marrow recipients and blood stem cell recipients studied were 13 and 22, respectively, on day 30; 13 and 20 on day 80; 8 and 16 on day 180; and 6 and 14 on day 365. For VZV, the numbers of marrow recipients and blood stem cell recipients studied were 16 and 19, respectively, on day 30; 15 and 17 on day 80; 11 and 14 on day 180; and 10 and 12 on day 365.

Close modal

T-helper-cell function was assessed by observing proliferation on stimulation with HSV and VZV proteins. We focused on recipients with a latent or active infection whose donors had an infection because uninfected recipients with uninfected donors are not expected to mount a lymphoproliferative response. Infection can be assessed indirectly by detection of antiviral antibodies or, in healthy persons, detection of antiviral T cells, by lymphoproliferation.31 HSV-specific lymphoproliferation was evaluated in HSV-seropositive patients with HSV-lymphoproliferation–positive donors (13 marrow recipients and 21 blood stem cell recipients), and VZV-specific lymphoproliferation was evaluated in VZV-seropositive patients with VZV-lymphoproliferation–positive donors (17 marrow recipients and 19 blood stem cell recipients). For both HSV and VZV, the median corrected Δ cpm in marrow recipients and blood stem cell recipients at all the time points were not significantly different (Figure 2). Groups of HSV (VZV)-seronegative recipients with HSV (VZV)-lymphoproliferation–positive donors and HSV (VZV)-seropositive recipients with HSV (VZV)-lymphoproliferation–negative donors were too small for a formal statistical analysis; however, there appeared to be no differences between marrow recipients and blood stem cell recipients. The results of the HSV- and VZV-induced proliferation assessment suggest that the functional capacity of single CD4 T cells is similar in marrow recipients and blood stem cell recipients.

Interestingly, when uncorrected Δ cpm in marrow recipients were compared with those in blood stem cell recipients, there were no significant differences in PHA-, HSV-, or VZV-stimulated responses at any time point (data not shown) except day 30, when the median Δ cpm PHA-stimulated response was 1.5 times higher in blood stem cell recipients (P = .03).

Serum IgG

Measurements of total serum IgG levels on day 80 after transplantation were available for 44 marrow recipients and 44 blood stem cell recipients who did not have multiple myeloma and did not receive intravenous immunoglobulin between day 0 and day 80. The levels were similar (median, 5.70 g/L in marrow recipients and 5.45 g/L in blood stem cell recipients; P = .99). Determinations of day 365 IgG levels were available for 31 marrow recipients and 39 blood stem cell recipients who did not have multiple myeloma and had not received intravenous immunoglobulin within 2 months before the evaluation on day 365. Levels were also similar at that time (median, 6.32 g/L in marrow recipients and 5.65 g/L in blood stem cell recipients; P = .56). Our normal range (5th-95th percentile) is 6.94 to 15.18 g/L. Thus, the degree of IgG deficiency was similar in the 2 groups of patients.

Infections

The rate of total infections was 1.4 times higher in marrow recipients than in blood stem cell recipients (P ≤ .01; Table 4). The rate of total definite infections was 1.7 times higher in marrow recipients (P ≤ .001). Importantly, the difference was more striking for severe definite infections (2.4 times higher rate in marrow recipients) than for nonsevere definite infections (1.4 times higher rate in marrow recipients). The difference was more striking for bacterial infections (1.7 times higher rate) and fungal infections (5.5 times higher rate) than for viral infections (1.4 times higher rate). The rate of clinical infections was similar in the 2 groups. Details on the infections are shown in Table 5. There was no difference in the posttransplantation day of diagnosis of the infections (median, day 78 for marrow recipients [25th-75th percentile, day 52-day 168] and day 98 for blood stem cell recipients [25th-75th percentile, day 50-day 182];P = .43; Figure 3).

Table 4.

Infection rates between day 30 and day 365 in patients given marrow compared with those given blood stem cells

Marrow4-150Blood4-150Rate ratio for marrow/blood (95% CI)PAdjusted rate ratio (95% CI)4-151P
Total infections (definite and clinical) 0.88 0.62 1.42 (1.09-1.85) .01 1.48 (1.13-1.94) .004 
Total definite infections 0.68 0.40 1.70 (1.24-2.34) .001 1.79 (1.29-2.47) .0005 
Severe definite infections 0.30 0.13 2.36 (1.39-4.02) .002 2.46 (1.44-4.21) .001 
Nonsevere definite infections 0.38 0.27 1.39 (0.93-2.08) .11 1.46 (0.97-2.21) .07 
Viral infections (definite) 0.33 0.23 1.44 (0.93-2.24) .10 1.59 (1.02-2.48) .04 
Bacterial infections (definite) 0.28 0.16 1.76 (1.07-2.90) .03 1.73 (1.04-2.88) .04 
Fungal infections (definite) 0.071 0.013 5.50 (1.20-25.1) .03 5.55 (1.20-25.7) .03 
Total clinical infections 0.21 0.23 0.91 (0.56-1.49) .71 0.94 (0.57-1.55) .81 
Severe clinical infections 0.09 0.08 1.19 (0.54-2.61) .66 1.34 (0.61-2.97) .47 
Nonsevere clinical infections 0.11 0.15 0.76 (0.40-1.45) .41 0.74 (0.39-1.41) .36 
Marrow4-150Blood4-150Rate ratio for marrow/blood (95% CI)PAdjusted rate ratio (95% CI)4-151P
Total infections (definite and clinical) 0.88 0.62 1.42 (1.09-1.85) .01 1.48 (1.13-1.94) .004 
Total definite infections 0.68 0.40 1.70 (1.24-2.34) .001 1.79 (1.29-2.47) .0005 
Severe definite infections 0.30 0.13 2.36 (1.39-4.02) .002 2.46 (1.44-4.21) .001 
Nonsevere definite infections 0.38 0.27 1.39 (0.93-2.08) .11 1.46 (0.97-2.21) .07 
Viral infections (definite) 0.33 0.23 1.44 (0.93-2.24) .10 1.59 (1.02-2.48) .04 
Bacterial infections (definite) 0.28 0.16 1.76 (1.07-2.90) .03 1.73 (1.04-2.88) .04 
Fungal infections (definite) 0.071 0.013 5.50 (1.20-25.1) .03 5.55 (1.20-25.7) .03 
Total clinical infections 0.21 0.23 0.91 (0.56-1.49) .71 0.94 (0.57-1.55) .81 
Severe clinical infections 0.09 0.08 1.19 (0.54-2.61) .66 1.34 (0.61-2.97) .47 
Nonsevere clinical infections 0.11 0.15 0.76 (0.40-1.45) .41 0.74 (0.39-1.41) .36 

CI indicates confidence interval.

F4-150

Number of infections/100 patient days, ie, number of infections in all recipients of marrow or blood stem cells divided by the number of days at risk and multiplied by 100. The number of days at risk (day 30 through day 365 or the day of death or relapse) was 14 131 for recipients of marrow and 15 537 for recipients of blood stem cells.

F4-151

Adjusted for pretransplantation variables known to influence the rate of posttransplantation infections, ie, donor/recipient cytomegalovirus (CMV) serostatus before transplantation (+/+, +/−, −/+, or −/−), disease/disease stage (good risk or poor risk), and splenectomy (yes or no).

Table 5.

Infections occurring between day 30 and day 365, according to source of hematopoietic cells (marrow or blood)

Type of infectionMarrowBlood
Viral (total n) 46 35  
 Herpes simplex   
  Oral/perioral 12 
  Anogenital 2  
 Zoster 10 
 CMV   
  Viremia only5-150 11 
  Pneumonia 3  
  Gastroenteritis 
 Respiratory virus (influenza A or B, parainfluenza, or respiratory syncytial virus)   
  Upper respiratory tract infection only 4  
  Sinusitis 
  Pneumonia 1  
 BK virus cystitis and/or prostatitis 1  
Bacterial (total n) 40 25 
 Gram-positive   
  Bacteremia only, with or without catheter-site infection 17 10  
  Sinusitis 
  Bronchitis 2  
  Pneumonia 
  Esophagitis 0  
  Urinary system infection 0  
 Gram-negative   
  Bacteremia only, with or without catheter site infection 
  Bronchitis 0  
  Pneumonia 
  Urinary system infection 3  
Clostridium difficile colitis 1  
Legionellapneumonia 0  
Fungal (total n) 10 
Aspergillus pneumonia 2  
 Candidemia only 0  
Candida esophagitis 
Clinical (total n) 29 34  
 Brain abscess 
 Otitis media 1  
 Parotitis 0  
 Lip infection 3  
 Sinusitis 
 Pneumonia 85-151 
 Diverticulitis 
 Colitis with pericolic phlegmon/abscess 1  
 Urinary tract infection or prostatitis 1  
 Vaginitis 
 Venous catheter site infection without bacteremia/fungemia 1  
 Superficial phlebitis 
 Cellulitis 1  
 Warts 2  
 Other skin infections (paronychia, tinea, folliculitis) 6  
 Fever of presumed infectious cause 
Type of infectionMarrowBlood
Viral (total n) 46 35  
 Herpes simplex   
  Oral/perioral 12 
  Anogenital 2  
 Zoster 10 
 CMV   
  Viremia only5-150 11 
  Pneumonia 3  
  Gastroenteritis 
 Respiratory virus (influenza A or B, parainfluenza, or respiratory syncytial virus)   
  Upper respiratory tract infection only 4  
  Sinusitis 
  Pneumonia 1  
 BK virus cystitis and/or prostatitis 1  
Bacterial (total n) 40 25 
 Gram-positive   
  Bacteremia only, with or without catheter-site infection 17 10  
  Sinusitis 
  Bronchitis 2  
  Pneumonia 
  Esophagitis 0  
  Urinary system infection 0  
 Gram-negative   
  Bacteremia only, with or without catheter site infection 
  Bronchitis 0  
  Pneumonia 
  Urinary system infection 3  
Clostridium difficile colitis 1  
Legionellapneumonia 0  
Fungal (total n) 10 
Aspergillus pneumonia 2  
 Candidemia only 0  
Candida esophagitis 
Clinical (total n) 29 34  
 Brain abscess 
 Otitis media 1  
 Parotitis 0  
 Lip infection 3  
 Sinusitis 
 Pneumonia 85-151 
 Diverticulitis 
 Colitis with pericolic phlegmon/abscess 1  
 Urinary tract infection or prostatitis 1  
 Vaginitis 
 Venous catheter site infection without bacteremia/fungemia 1  
 Superficial phlebitis 
 Cellulitis 1  
 Warts 2  
 Other skin infections (paronychia, tinea, folliculitis) 6  
 Fever of presumed infectious cause 

CMV indicates cytomegalovirus.

F5-150

On culture (not by pp65-antigen detection only).

F5-151

In one case, pneumonia was reported to be due toTrichomonas, but no written confirmation or clinical details were obtained.

Fig. 3.

Cumulative number of day 30 to day 365 infections after allogeneic transplantation of bone marrow or peripheral blood stem cells.

Both definite and clinical infections are included. BM, bone marrow; PBSCs, peripheral blood stem cells.

Fig. 3.

Cumulative number of day 30 to day 365 infections after allogeneic transplantation of bone marrow or peripheral blood stem cells.

Both definite and clinical infections are included. BM, bone marrow; PBSCs, peripheral blood stem cells.

Close modal

Although all patients had a sustained absolute neutrophil count of 0.5 × 109/L by day 28 after transplantation (Table 1), absolute neutrophil counts were significantly higher in blood stem cell recipients compared with marrow recipients until day 48. To eliminate any influence of the different neutrophil counts, we compared infection rates between day 60 and day 365. The rate of total infections was significantly higher in marrow recipients (unadjusted rate ratio, 1.48 [P = .02], and adjusted rate ratio, 1.53 [P = .01]). The rate of total definite infections was also significantly higher in marrow recipients (unadjusted rate ratio, 1.90 [P = .002], and adjusted rate ratio, 1.99 [P = .001]). Therefore, the differences in infection rates between day 30 and day 365 in the 2 groups of patients were most likely due to the differences in lymphocyte-subset counts and not neutrophil counts.

Death associated with a definite infection diagnosed between day 30 and day 365 occurred between day 30 and day 365 in 9 marrow recipients and 3 blood stem cell recipients (P = .17 by Χ2test). Details on these infections are provided in Table6. There were 9 deaths associated with a bacterial or fungal infection among marrow recipients and none among blood stem cell recipients (P = .008 by Χ2test).

Table 6.

Deaths (occurring between day 30 and day 365 after transplantation) associated with a definite infection diagnosed between day 30 and day 365

Tx type/infectionOther conditions contributing to deathPost-Tx day of diagnosis of infection/death
Marrow   
Haemophilus influenzaepneumonia None 156/210 
Enterobacter cloacae pneumonia Chronic GVHD 145/153  
 Sepsis due to gram-negative rod (unspeciated) leading to ARDS None 221/234  
Legionella micdadei/CMV pneumonia Chronic GVHD 65/108 
Staphylococcus epidermidis sepsis leading to ARDS Chronic GVHD 224/241  
Staphylococcus epidermidis sepsis leading to ARDS None 31/37 
Aspergillus fumigatus pneumonia Acute GVHD, VOD 38/38  
Aspergillus flavus pneumonia Chronic GVHD 68/121  
Aspergillus fumigatus/CMV pneumonia Chronic GVHD 253/272  
Blood stem cells   
 RSV pneumonia Acute GVHD, VOD 33/34  
 CMV pneumonia Acute GVHD, MOF 34/34  
 CMV pneumonia Chronic GVHD 130/158 
Tx type/infectionOther conditions contributing to deathPost-Tx day of diagnosis of infection/death
Marrow   
Haemophilus influenzaepneumonia None 156/210 
Enterobacter cloacae pneumonia Chronic GVHD 145/153  
 Sepsis due to gram-negative rod (unspeciated) leading to ARDS None 221/234  
Legionella micdadei/CMV pneumonia Chronic GVHD 65/108 
Staphylococcus epidermidis sepsis leading to ARDS Chronic GVHD 224/241  
Staphylococcus epidermidis sepsis leading to ARDS None 31/37 
Aspergillus fumigatus pneumonia Acute GVHD, VOD 38/38  
Aspergillus flavus pneumonia Chronic GVHD 68/121  
Aspergillus fumigatus/CMV pneumonia Chronic GVHD 253/272  
Blood stem cells   
 RSV pneumonia Acute GVHD, VOD 33/34  
 CMV pneumonia Acute GVHD, MOF 34/34  
 CMV pneumonia Chronic GVHD 130/158 

ARDS indicates adult respiratory distress syndrome; VOD, venocclusive disease of the liver; RSV, respiratory syncytial virus; MOF, multiorgan failure; for other abbreviations, see Table 1.

The important findings of this study were that (1) compared with marrow recipients, blood stem cell recipients had higher counts of T cells (particularly CD4 T cells); (2) T cells were equally functional in the 2 patient groups; and (3) improving the quantitative lymphocyte deficiency in recipients of hematopoietic transplants may result in fewer infections.

The higher counts of certain lymphocyte subsets in blood stem cell recipients were anticipated because of the results of smaller nonrandomized studies and have been attributed to the larger lymphocyte inoculum.12,13 It is reasonable to attribute the higher posttransplantation counts of T cells, particularly naive T cells, to the large inoculum because T cells, especially naive T cells, are extremely long-lived.14-16 Also, few T cells are produced from stem cells in adult transplant recipients in the first year after transplantation because of age- and allografting-associated thymic atrophy; therefore, most T cells in patients in this year originate from the T cells infused with the graft.8,32-35 This idea is in agreement with the observation that naive T-cell counts but not CD34 cell counts in the grafts correlate with naive T-cell counts after transplantation (J. S. et al, unpublished data, March 2001). We found that even though blood stem cell grafts contained more monocytes, NK cells, and B cells than marrow grafts, reconstitution of these cell populations was similar in marrow recipients and blood stem cell recipients. The numbers of monocytes and NK cells had already reached the normal range by day 30, suggesting that reconstitution of these populations occurs rapidly and is not limited by the number of cells in the graft. B-cell counts were higher during the first 3 months after blood stem cell transplantation compared with marrow transplantation. This finding is consistent with the presence of much higher numbers of naive and memory B cells in blood stem cell grafts than in marrow grafts. However, the rate of B-cell recovery was faster in marrow recipients, as indicated by the steeper slope during the first 3 months after grafting (Figure 1). This might have been due to the higher number of B-cell progenitors in the marrow grafts (Table 3).

Total IgG levels were similar in the 2 patient groups, suggesting that humoral immunity is not improved after blood stem cell grafting compared with marrow grafting. After marrow grafting, IgG is frequently composed of monoclonal or oligoclonal immunoglobulins of unclear specificity (not against infectious agents)36-50 and therefore the protective value of these immunoglobulins in marrow recipients is low. Qualitative comparisons of immunoglobulins will be needed to assess the contribution of humoral immunity to preventing infection after blood stem cell transplantation. The presence of higher numbers of plasma cells in the marrow inoculum (Table 3) raises the question of whether the immunoglobulins produced by the infused plasma cells markedly contribute to the total IgG levels. If they do, this could be why IgG levels are not higher in blood stem cell recipients than in marrow recipients, even though B-cell counts are higher.

The difference in the rates of total definite infections cannot be attributed to differences in clinical patient characteristics because those were balanced between the 2 patient groups (Table 1). Adjustment for minor imbalances in pretransplantation characteristics (splenectomy, disease/disease stage, and CMV serostatus) only strengthened the significance of the differences (Table 4). We did not adjust for minor imbalances in posttransplantation characteristics because those may have been associated with the main treatment (blood stem cell grafting or marrow grafting). Nevertheless, the higher infection rates in the marrow recipients cannot be attributed to a weaker graft, since the incidence of severe neutropenia between day 30 and day 365 was not significantly higher in those patients (Table 1). Also, day 60 to day 365 infection rates, which were not influenced by the higher neutrophil counts in blood stem cell recipients up to day 48, were significantly higher in marrow recipients. Use of glucocorticoids is unlikely to have been responsible for the observed differences in infection rates because the number of patients treated with glucocorticoids was similar in the 2 groups. The differences also cannot be attributed to GVHD, since the incidence of grade 2 to 4 acute GVHD was similar in the 2 patient groups and there was a trend toward a lower incidence of clinical extensive chronic GVHD among the marrow recipients. Infection prophylaxis strategies were also similar in the 2 groups. The only difference was a trend toward a greater use of prophylactic acyclovir in marrow recipients (Table 1), a finding that may explain why the incidence of HSV and VZV infections among blood stem cell recipients was not lower than that among marrow recipients.

The difference in total definite infection rates was attributable primarily to differences in the rates of bacterial and fungal infections. Also, blood stem cell recipients had fewer fatal bacterial and fungal infections. This may seem puzzling because the major difference between the marrow recipients and the blood stem cell recipients appeared to be in their T-cell counts and T cells have traditionally been considered to play a much less important role than neutrophils in host defense against extracellular bacteria and fungi (particularly molds). Perhaps the importance of T cells in host defense against extracellular bacteria and fungi (particularly molds) has been underestimated. Adoptively transferred T cells from immune donors to naive hosts have been found to protect rodents against infection with Bacteroides fragilis, Pseudomonas aeruginosa, Candida albicans, or Aspergillus fumigatus.51-54 

The concurrence of higher T-cell counts and, in part, B-cell and NK cell counts and the lower infection rates in the blood stem cell recipients suggests but does not prove that the higher lymphocyte-subset counts resulted in lower infection rates. However, this is likely because previous studies showed inverse correlations between CD4 T-cell or B-cell counts and postengraftment infection rates.9-11 Moreover, in the marrow recipients in our study, there was a trend toward inverse correlations between day 30, 80, 180, and 365 total CD4 T-cell counts and day 30 to day 365 infection rates; between day 30, 80, 180, and 365 total B-cell counts and day 30 to day 365 infection rates; and between day 30, 80, and 180 NK cell counts and day 30 to day 365 infection rates (by Spearman rank correlation; data not shown). The most striking correlations in marrow recipients were those between day 80 total B-cell counts and total definite infections (r = −0.43; P = .006), day 80 IgD+ B-cell counts and total definite infections (r = −0.50; P = .001), and day 30 IgDB-cell counts and total definite infections (r = −0.42;P = .005). Also, there was a significant correlation in blood stem cell recipients between day 30 NK cell counts and day 30 to day 365 total infection rates (r = −0.38; P = .01). These analyses support the idea that the lower infection rates in blood stem cell recipients compared with marrow recipients probably resulted from the increased counts of T cells, B cells, and possibly NK cells.

It would not be prudent to interpret the finding of lower day 30 to day 365 infection rates in blood stem cell recipients as a recommendation for discontinuing marrow transplantation. Other end points, such as relapse and survival, must also be taken into consideration. Moreover, if chronic GVHD is more prevalent or more severe (requiring more immunosuppressive treatment) after blood stem cell grafting than after marrow grafting,55-57 it is possible that infection rates after day 365 in blood stem cell recipients will be equal to or even higher than those in marrow recipients.

In summary, we found that blood stem cell recipients have fewer day 30 to day 365 infections than marrow recipients. This might be because of improved lymphocyte-subset counts after blood stem cell grafting, an improvement that may result from the high numbers of lymphocytes in blood stem cell grafts.

We thank Patrick Sudour, Kristen White, Erica Ryberg, Amber Wyman, Hana Gage, and the dedicated staff of the FHCRC Cryobiology Laboratory for excellent technical assistance; the study nurse, Terri Cunningham; the patients who agreed to participate in the study; and the staff of the FHCRC Long- Term Follow-Up Department, who diligently gathered clinical information.

Supported by National Institutes of Health grants CA68496, CA18221, CA18029, CA15704, HL36444, and AI46108.

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

1
Thomas
ED
Blume
KG
Forman
SJ
Hematopoietic Cell Transplantation.
2nd ed.
1999
Blackwell Science
Malden, MA
2
Atkinson
K
Clinical Bone Marrow and Blood Stem Cell Transplantation.
2nd ed.
2000
Cambridge University Press
Cambridge, United Kingdom
3
Atkinson
K
Farewell
V
Storb
R
et al
Analysis of late infections after human bone marrow transplantation: role of genotypic nonidentity between marrow donor and recipient and of nonspecific suppressor cells in patients with chronic graft-versus-host disease.
Blood.
60
1982
714
720
4
Marks
DI
Cullis
JO
Ward
KN
et al
Allogeneic bone marrow transplantation for chronic myeloid leukemia using sibling and volunteer unrelated donors: a comparison of complications in the first 2 years.
Ann Intern Med.
119
1993
207
214
5
Sullivan
KM
Mori
M
Sanders
J
et al
Late complications of allogeneic and autologous marrow transplantation.
Bone Marrow Transplant.
10(suppl 1)
1992
127
134
6
Morrison
VA
Haake
RJ
Weisdorf
DJ
Non-Candida fungal infections after bone marrow transplantation: risk factors and outcome.
Am J Med.
96
1994
497
503
7
Ochs
L
Shu
XO
Miller
J
et al
Late infections after allogeneic bone marrow transplantation: comparison of incidence in related and unrelated donor transplant recipients.
Blood.
86
1995
3979
3986
8
Storek
J
Witherspoon
RP
Immunologic reconstitution after hematopoietic stem cell transplantation.
Clinical Bone Marrow and Blood Stem Cell Transplantation. Cambridge
Atkinson
K
2000
111
146
Cambridge University Press
United Kingdom
9
Small
TN
Avigan
D
Dupont
B
et al
Immune reconstitution following T-cell depleted bone marrow transplantation: effect of age and posttransplant graft rejection prophylaxis.
Biol Blood Marrow Transplant.
3
1997
65
75
10
Storek
J
Gooley
T
Witherspoon
RP
Sullivan
KM
Storb
R
Infectious morbidity in long-term survivors of allogeneic marrow transplantation is associated with low CD4 T cell counts.
Am J Hematol.
54
1997
131
138
11
Storek
J
Espino
G
Dawson
MA
Storer
B
Flowers
MED
Maloney
DG
Low B cell and monocyte counts on day 80 are associated with high infection rates between day 100 and 365 after allogeneic marrow transplantation.
Blood.
96
2000
3290
3293
12
Ottinger
HD
Beelen
DW
Scheulen
B
Schaefer
UW
Grosse-Wilde
H
Improved immune reconstitution after allotransplantation of peripheral blood stem cells instead of bone marrow.
Blood.
88
1996
2775
2779
13
Storek
J
Witherspoon
RP
Maloney
DG
Chauncey
TR
Storb
R
Improved reconstitution of CD4 T cells and B cells but worsened reconstitution of serum IgG levels after allogeneic transplantation of blood stem cells instead of marrow [letter].
Blood.
89
1997
3891
3893
14
Hellerstein
M
Hanley
MB
Cesar
D
et al
Directly measured kinetics of circulating T lymphocytes in normal and HIV-1-infected humans.
Nat Med.
5
1999
83
89
15
McLean
AR
Michie
CA
In vivo estimates of division and death rates of human T lymphocytes.
Proc Natl Acad Sci U S A.
92
1995
3707
3711
16
Michie
CA
McLean
A
Alcock
C
Beverley
PCL
Lifespan of human lymphocyte subsets defined by CD45 isoforms.
Nature.
360
1992
264
265
17
Bensinger
WI
Martin
P
Storer
B
et al
Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers.
N Engl J Med.
344
2001
175
181
18
Storek
J
Ferrara
S
Ku
N
Giorgi
JV
Champlin
RE
Saxon
A
B cell reconstitution after human bone marrow transplantation: recapitulation of ontogeny?
Bone Marrow Transplant.
12
1993
387
398
19
Storek
J
Dawson
MA
Maloney
DG
Comparison of two flow cytometric methods enumerating CD4 T cells and CD8 T cells.
Cytometry.
33
1998
76
82
20
Klein
U
Kuppers
R
Rajewsky
K
Human IgM+IgD+ B cells, the major B cell subset in the peripheral blood, express V-κ genes with no or little somatic mutation throughout life.
Eur J Immunol.
23
1993
3272
3277
21
Suzuki
I
Milner
ECB
Glas
AM
et al
Immunoglobulin heavy chain variable region gene usage in bone marrow transplant recipients: lack of somatic mutation indicates a maturational arrest.
Blood.
87
1996
1873
1880
22
Klein
U
Rajewsky
K
Kuppers
R
Human immunoglobulin (Ig) M+IgD+ peripheral blood B cells expressing the CD27 cell surface antigen carry somatically mutated variable region genes: CD27 as a general marker for somatically mutated (memory) B cells.
J Exp Med.
188
1998
1679
1689
23
Douek
DC
McFarland
RD
Keiser
PH
et al
Changes in thymic function with age and during the treatment of HIV infection.
Nature.
396
1998
690
695
24
Dumont-Girard
F
Roux
E
van Lier
RA
et al
Reconstitution of the T-cell compartment after bone marrow transplantation: restoration of the repertoire by thymic emigrants.
Blood.
92
1998
4464
4471
25
Storek
J
Witherspoon
RP
Storb
R
T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life.
Bone Marrow Transplant.
16
1995
413
425
26
Okumura
M
Fujii
Y
Inada
K
Nakahara
K
Matsuda
H
Both CD45RA+ and CD45RA− subpopulations of CD8+ T cells contain cells with high levels of lymphocyte function-associated antigen-1 expression, a phenotype of primed T cells.
J Immunol.
150
1993
429
437
27
Okumura
M
Fujii
Y
Takeuchi
Y
Inada
K
Nakahara
K
Matsuda
H
Age-related accumulation of LFA-1high cells in a CD8+CD45RAhigh T cell population.
Eur J Immunol.
23
1993
1057
1063
28
Chambers
CA
Allison
JP
Costimulatory regulation of T cell function.
Curr Opin Cell Biol.
11
1999
203
210
29
Meyers
JD
Flournoy
N
Thomas
ED
Infection with herpes simplex virus and cell-mediated immunity after marrow transplant.
J Infect Dis.
142
1980
338
346
30
Meyers
JD
Flournoy
N
Thomas
ED
Cell-mediated immunity to varicella-zoster virus after allogeneic marrow transplant.
J Infect Dis.
141
1980
479
487
31
Wald
A
Zeh
J
Selke
S
et al
Reactivation of genital herpes simplex virus type 2 infection in asymptomatic seropositive persons.
N Engl J Med.
342
2000
844
850
32
George
AJ
Ritter
MA
Thymic involution with ageing: obsolescence or good housekeeping?
Immunol Today.
17
1996
267
272
33
Fukushi
N
Arase
H
Wang
B
et al
Thymus: a direct target tissue in graft-versus-host reaction after allogeneic bone marrow transplantation that results in abrogation of induction of self-tolerance.
Proc Natl Acad Sci U S A.
87
1990
6301
6305
34
Muller-Hermelink
HK
Sale
GE
Borisch
B
Storb
R
Pathology of the thymus after allogeneic bone marrow transplantation in man.
Am J Pathol.
129
1987
242
256
35
Ritter
MA
Ladyman
HM
The effects of cyclosporine on the thymic microenvironment and T cell development.
Thymus Update Series 4. The Thymus in Immunotoxicology.
Kendall
MD
1991
157
176
Harwood Academic Press
Newark, NJ
36
Holmes
JA
Livesey
SJ
Bedwell
AE
Amos
N
Whittaker
JA
Autoantibody analysis in chronic GVHD.
Bone Marrow Transplant.
4
1989
529
531
37
Kier
P
Penner
E
Bakos
S
et al
Autoantibodies in chronic GVHD: high prevalence of antinucleolar antibodies.
Bone Marrow Transplant.
6
1990
93
96
38
Sanmarco
M
Vialettes
B
Maraninchi
D
Bernard
D
Autoantibody formation after bone marrow transplantation: a comparison between autologous and allogeneic grafts.
Autoimmunity.
11
1991
7
12
39
Lortan
JE
Rochfort
NC
el-Tumi
M
Vellodi
A
Autoantibodies after bone marrow transplantation in children with genetic disorders: relation to chronic graft-versus-host disease.
Bone Marrow Transplant.
9
1992
325
330
40
Smith
CIE
Hammarstrom
L
Lefvert
AK
Bone marrow grafting induces acetylcholine receptor antibody formation [letter].
Lancet.
1
1985
978
41
Soderberg
C
Sumitran-Karuppan
S
Ljungman
P
Moller
E
CD13-specific autoimmunity in cytomegalovirus-infected immunocompromised patients.
Transplantation.
61
1996
594
600
42
Soderberg
C
Larsson
S
Rozell
BL
Sumitran-Karuppan
S
Ljungman
P
Moller
E
Cytomegalovirus-induced CD13-specific autoimmunity—a possible cause of chronic GVHD.
Transplantation.
61
1996
600
609
43
Hebart
H
Einsele
H
Klein
R
et al
CMV infection after allogeneic bone marrow transplantation is associated with the occurrence of various autoantibodies and monoclonal gammopathies.
Brit J Haematol.
95
1996
138
144
44
Mitus
AJ
Stein
R
Rappeport
JM
et al
Monoclonal and oligoclonal gammopathy after bone marrow transplantation.
Blood.
74
1989
2764
2768
45
Fischer
AM
Simon
F
LeDeist
F
Blanche
S
Griscelli
C
Fischer
A
Prospective study of the occurrence of monoclonal gammapathies following bone marrow transplantation in young children.
Transplantation.
49
1990
731
735
46
Radl
J
Liu
M
Hoogeveen
CM
et al
Monoclonal gammapathies in long-term surviving rhesus monkeys after lethal irradiation and bone marrow transplantation.
Clin Immunol Immunopathol.
60
1991
305
309
47
Tissot
JD
Helg
C
Chapuis
B
et al
Clonal imbalances of serum immunoglobulins after allogeneic bone marrow transplantation: an analysis by high-resolution two-dimensional gel electrophoresis.
Bone Marrow Transplant.
10
1992
347
353
48
Mink
JG
Radl
J
VanDenBerg
P
VanMuiswinkel
WB
VanOosterom
R
Homogenous immunoglobulins in the serum of irradiated and bone marrow reconstituted mice: the role of thymus and spleen.
Immunology.
37
1979
889
894
49
VanDenBerg
P
Radl
J
Lowenberg
B
Swart
ACW
Homogeneous antibodies in lethally irradiated and autologous bone marrow reconstituted Rhesus monkeys.
Clin Exp Immunol.
23
1976
355
359
50
Gerritsen
EJA
VanTol
MJD
Ballieux
P
et al
Search for the antigen specificity of homogeneous IgG components (H-IgG) after allogeneic bone marrow transplantation.
Bone Marrow Transplant.
17
1996
825
833
51
Crabb
JH
Finberg
R
Onderdonk
AB
Kasper
DL
T cell regulation of Bacteroides fragilis-induced intraabdominal abscesses.
Rev Infect Dis.
12(suppl 2)
1990
S178
S184
52
Powderly
WG
Schreiber
JR
Pier
GB
Markham
RB
T cells recognizing polysaccharide-specific B cells function as contrasuppressor cells in the generation of T cell immunity to Pseudomonas aeruginosa.
J Immunol.
140
1988
2746
2752
53
Sieck
TG
Moors
MA
Buckley
HR
Blank
KJ
Protection against murine disseminated candidiasis mediated by a Candida albicans-specific T-cell line.
Infect Immun.
61
1993
3540
3543
54
Cenci
E
Mencacci
A
Bacci
A
Bistoni
F
Kurup
VP
Romani
L
T cell vaccination in mice with invasive pulmonary aspergillosis.
J Immunol.
165
2000
381
388
55
Schmitz
N
Beksac
M
Hasenclever
D
et al
A randomized study from the European Group for Blood and Marrow Transplantation comparing allogeneic transplantation of filgrastim-mobilized peripheral blood progenitor cells with bone marrow transplantation in 350 patients with leukemia [abstract].
Blood.
96
2000
477a
56
Cutler
C
Giri
S
Jeyapalan
S
Paniagua
D
Viswanathan
A
Antin
J
Incidence of acute and chronic GVHD after allogeneic peripheral blood stem cell and bone marrow transplantation: a meta-analysis [abstract].
Blood.
96
2000
205a
57
Flowers
MED
Matos
A
Storer
B
et al
Clinical manifestation of chronic GVHD after transplantation of peripheral blood stem cells compared to bone marrow [abstract].
Blood.
96
2000
203a
58
Storb
R
Deeg
HJ
Whitehead
J
et al
Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia.
N Engl J Med.
314
1986
729
735
59
Boeckh
M
Gooley
TA
Myerson
D
Cunningham
T
Schoch
G
Bowden
RA
Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study.
Blood.
88
1996
4063
4071
60
Walter
EA
Greenberg
PD
Gilbert
MJ
et al
Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor.
N Engl J Med.
333
1995
1038
1044

Author notes

Jan Storek, FHCRC, D1-100, 1100 Fairview Ave N, Seattle, WA 98109-1024; e-mail: jstorek@fhcrc.org.

Sign in via your Institution