Most patients requiring allogeneic bone marrow transplant (allo-BMT) do not have an HLA-matched sibling donor. A phenotypically matched unrelated donor graft has been made available for approximately 50% of Caucasians and less than 10% of ethnic and racial minorities in need. However, almost all patients have a readily available partially mismatched related donor (PMRD). We summarize our experience with 72 patients who ranged from 1 to 50 years of age (median, 16 years) and who were recipients of a PMRD allo-BMT from haploidentical family members following conditioning therapy using total body irradiation (TBI) and multiagent, high-dose chemotherapy. T-cell depletion and post-BMT immunosuppression were combined for graft-versus-host disease (GVHD) prophylaxis. The probability of engraftment was 0.88 at 32 days. Six of 10 patients who failed to engraft achieved engraftment after secondary transplant. Grade II to IV acute GVHD was seen in 9 of 58 (16%) evaluable patients; extensive chronic GVHD was seen in 4 of 48 (8%) evaluable patients. There was a statistically significant difference in 2-year survival probability between low-risk and high-risk patients (0.55 v 0.27, P = .048). Prognostic factors that affected outcomes in multivariate analysis were (1) a lower TBI dose and 3-antigen rejection mismatch decreased stable engraftment (P = .005 and P = .002, respectively); (2) a higher T-cell dose increased acute GVHD (P = .058); (3) a higher TBI dose increased chronic GVHD (P = .016); and (4) a high-risk disease category increased treatment failure from relapse or death (P = .037). A PMRD transplant can be performed with acceptable rates of graft failure and GVHD. Using sequential immunomodulation, the disease status at the time of transplant is the only prognostic factor significantly associated with long-term successful outcome after PMRD allo-BMT. When allogeneic rather than autologous BMT is indicated, progression in disease status before transplant can be avoided using a PMRD with equal inclusion of all ethnic or racial groups.

BONE MARROW transplantation (BMT) from genotypically HLA-matched siblings has improved long-term survival in patients with hematologic malignancies and marrow failure syndromes.1 However, more than 70% of patients who could benefit from allogeneic-BMT (allo-BMT) do not have a matched sibling donor (MSD). Attention has turned, therefore, to alternative donors2-5 primarily, partially mismatched related donors (PMRD),6-9 and phenotypically matched unrelated donors (PMUD).10-16 The chance of receiving a PMUD varies with the race of the patient, ranging from approximately 50% for Caucasians to less than 10% for ethnic minorities, and often requires waiting months to identify the donor and obtain the graft.17 However, allowing for unusual circumstances in which no biological relative is available, there is a greater than 90% chance to promptly identify a haploidentical donor within the family.

Use of alternative donors for allo-BMT involves crossing histocompatibility barriers and, therefore, carries a greater risk of nonengraftment,8,12,13,18,19 severe acute and chronic graft-versus-host disease (aGVHD, cGVHD),7,8,11-14,20-23 and prolonged immunodysregulation increasing the risk of fatal infections and lymphoproliferative disorders.24-26 We sought to reduce these complications through sequential immunomodulation of the recipient, donor marrow, and resultant chimera when using a readily available PMRD for patients with malignant and nonmalignant hematologic conditions.

Clinical Protocol

Between February 1993 and November 1994, 72 patients who lacked an MSD underwent PMRD allo-BMT on a protocol approved by the Richland Memorial Hospital Institutional Review Board (Table 1). Among patients with leukemia, those classified as low risk included patients with acute leukemia in first or second complete remission and chronic myeloid leukemia (CML) in primary chronic phase. Patients classified as high risk had acute leukemia in greater than second remission or in relapse or CML in accelerated or blastic phase. Patients with marrow failure syndromes were considered high risk if they had HLA antibodies, abnormal cytogenetics, or disease for more than 6 months.

Table 1.

Diagnosis, Risk Category, and HLA Disparity at Time of PMRD allo-BMT

PatientsLow RiskHigh Risk
n = 72 20 (28%)* 52 (72%) 
Diagnosis 
ALL 10 (14%) 18 (25%) 
AML 3 (4%) 16 (22%) 
CML/CLL 5 (7%) 14 (19%) 
SAA/MDS 2 (3%) 4 (6%) 
Disease-status linked to degree of HLA disparity 
   
Donor mismatch (rejection direction) 
1 antigen (n = 13) 4 (6%) 9 (13%) 
2 antigen (n = 35) 13 (18%) 22 (31%) 
3 antigen (n = 24) 3 (4%) 21 (29%) 
Recipient mismatch (GVHD direction) 
1 antigen (n = 11) 5 (7%) 6 (8%) 
2 antigen (n = 30) 9 (13%) 21 (29%) 
3 antigen (n = 31) 6 (8%) 25 (35%) 
PatientsLow RiskHigh Risk
n = 72 20 (28%)* 52 (72%) 
Diagnosis 
ALL 10 (14%) 18 (25%) 
AML 3 (4%) 16 (22%) 
CML/CLL 5 (7%) 14 (19%) 
SAA/MDS 2 (3%) 4 (6%) 
Disease-status linked to degree of HLA disparity 
   
Donor mismatch (rejection direction) 
1 antigen (n = 13) 4 (6%) 9 (13%) 
2 antigen (n = 35) 13 (18%) 22 (31%) 
3 antigen (n = 24) 3 (4%) 21 (29%) 
Recipient mismatch (GVHD direction) 
1 antigen (n = 11) 5 (7%) 6 (8%) 
2 antigen (n = 30) 9 (13%) 21 (29%) 
3 antigen (n = 31) 6 (8%) 25 (35%) 

Abbreviations: n, number of patients; ALL, acute lymphoblastic leukemia; AML, acute myelogenous leukemia; CML, chronic myelogenous leukemia; CLL, chronic lymphocytic leukemia; SAA, severe aplastic anemia; MDS, myelodysplastic syndrome; GVHD, graft-versus-host disease.

*

Proportion of the entire patient sample.

P = .035 (one-tailed) for the likelihood of receiving a 3-Ag rejection mismatched graft for high-risk versus low-risk category.

P = .131 (one-tailed) for likelihood of receiving a 3-Ag GVHD mismatched graft for high-risk versus low-risk category.

The conditioning regimen and GVHD prophylaxis administered to all patients (except as noted for nonmalignant disease) are described in Fig 1. Patients were housed in a positive-pressure high efficiency particulate air (HEPA)-filtered BMT unit. All patients received granulocyte colony-stimulating factor (G-CSF ) from day +1 until the white blood cell (WBC) count exceeded 5,000/μL. Prophylactic and therapeutic antimicrobial therapy included broad-spectrum antibiotics, intravenous Ig, inhaled Pentamidine (or trimethoprim-sulfamethoxazole, if required), and antiviral and antifungal agents as indicated.

Fig. 1.

Conditioning therapy and GVHD prophylaxis in 72 recipients of PMRD allo-BMT. In an effort to improve engraftment, the total dose of TBI was increased from 1,332 cGy (administered to the first 45 patients) to 1,500 cGy (administered to the subsequent 27 patients). Cyclosporin and prednisone were gradually tapered and discontinued in patients free of acute and/or chronic GVHD. Abbreviations: TBI , total body irradiation administered twice daily via AP/PA fields with ∼50% pulmonary transmission with lung shielding and electron beam boosting of chest wall, and testicles (ALL, biphenotypic) using an instantaneous dose rate of 13 to 22 cGy/min and interfraction interval of ∼8 hours; VP-16 ✶, etoposide administered once (omitted for nonmalignant disease); Ara-C ⬇, cytosine arabinoside administered twice daily × 6 doses; CTX ✳, cyclophosphamide administered daily × 2 doses; H-D MPD ⇩, high-dose methylprednisolone administered every 12 hours × 4 doses; T10B9 + C BMT ▴, marrow graft T-cell depleted with T10B9 and complement, L-D CYS ▹, low-dose cyclosporin started day −1 at 3 mg/kg constant infusion and maintained at levels between 100 to 200 as measured by monoclonal antibody technique, switched to orally after day +21 and weaned gradually through the first year post-BMT; M-D MPD ⇩, moderate-dose methylprednisolone administered before ATG; ATG ♦, antithymocyte globulin administered daily × 12 doses on day +5 to +16; ⇩ MPD/Pred; steroid dose tapered 10% weekly and switched to prednisone orally after day +21; ❁BMT➭, days before and after BMT; →, expanded time period.

Fig. 1.

Conditioning therapy and GVHD prophylaxis in 72 recipients of PMRD allo-BMT. In an effort to improve engraftment, the total dose of TBI was increased from 1,332 cGy (administered to the first 45 patients) to 1,500 cGy (administered to the subsequent 27 patients). Cyclosporin and prednisone were gradually tapered and discontinued in patients free of acute and/or chronic GVHD. Abbreviations: TBI , total body irradiation administered twice daily via AP/PA fields with ∼50% pulmonary transmission with lung shielding and electron beam boosting of chest wall, and testicles (ALL, biphenotypic) using an instantaneous dose rate of 13 to 22 cGy/min and interfraction interval of ∼8 hours; VP-16 ✶, etoposide administered once (omitted for nonmalignant disease); Ara-C ⬇, cytosine arabinoside administered twice daily × 6 doses; CTX ✳, cyclophosphamide administered daily × 2 doses; H-D MPD ⇩, high-dose methylprednisolone administered every 12 hours × 4 doses; T10B9 + C BMT ▴, marrow graft T-cell depleted with T10B9 and complement, L-D CYS ▹, low-dose cyclosporin started day −1 at 3 mg/kg constant infusion and maintained at levels between 100 to 200 as measured by monoclonal antibody technique, switched to orally after day +21 and weaned gradually through the first year post-BMT; M-D MPD ⇩, moderate-dose methylprednisolone administered before ATG; ATG ♦, antithymocyte globulin administered daily × 12 doses on day +5 to +16; ⇩ MPD/Pred; steroid dose tapered 10% weekly and switched to prednisone orally after day +21; ❁BMT➭, days before and after BMT; →, expanded time period.

Close modal

When patients required regrafting, the same or an alternative PMRD donor was used. Second grafts were processed using a single incubation with complement to limit the efficiency of T-cell depletion effectively increasing the T-cell dose. Reconditioning therapy included the use of cyclophosphamide, antithymocyte globulin, and methylprednisolone. Patients who developed greater than grade I acute GVHD were treated with high-dose pulsed methylprednisolone (MPD; 500 mg/m2 every 12 hours for 2 doses and repeated every 48 to 72 hours for up to 4 pulses). Second-line therapy for recurrence of GVHD included a second course of antithymocyte globulin (ATG) and/or azathioprine.

Donors

Family members were assessed for degree of mismatch by HLA serologic typing.27 When necessary, molecular techniques were used to define class II antigens (Ag).28 Donors were prioritized on the basis of the greatest HLA matching, cytomegalovirus (CMV) seronegativity for seronegative recipients, younger age, same sex, nonparity, and better health.29 

Marrow Graft Preparation

Donors were harvested by aspirating 4.5 to 6 × 108 nucleated cells per kilogram of recipient body weight. Grafts were depleted of mature T lymphocytes using T10B9.1A-31 (T10B9 courtesy of Dr John Thompson, University of Kentucky, Lexington, KY), an IgM murine monoclonal antibody (MoAb) directed against the αβ chains of the T-cell receptor heterodimer, and rabbit complement as described previously.30 The degree of T-cell depletion was assessed by limiting dilution analysis.31 Recovery of hematopoietic precursors was assessed by predepletion and postdepletion colony-forming unit–granulocyte-macrophage (CFU-GM) and CD34+ cell assays. Marrow grafts were cultured for possible microbial contaminants.

Statistical Design

This study was conducted as a single-arm prospective trial in which consecutive patients who met inclusion criteria were entered. Data were collected prospectively on case report forms and retrospectively by medical record review. Graft characteristics were calculated excluding second/third transplants. Patients were monitored for toxicity, engraftment, acute and chronic GVHD, relapse, and survival.

Endpoints.Toxicity was graded according to standard criteria for organ systems. All patients were considered evaluable for engraftment. The day of engraftment was taken as the first of three consecutive days on which the WBC count was 1,000/μL. Patients were considered to have achieved stable engraftment if they maintained their blood counts with DNA evidence of donor-derived hematopoiesis. Patients who failed to establish engraftment within 30 days of BMT, as evidenced by a WBC count less than 200/μL and absence of donor-specific DNA properties, were considered to have graft failure. Achievement of a WBC count of 500/μL with subsequent decline was criteria for rejection, which was considered to have occurred if a corresponding marrow was mostly acellular. Patients were considered evaluable for aGVHD if they engrafted successfully and were considered evaluable for cGVHD if they also survived at least 80 days post-BMT. Acute GVHD was graded according to accepted criteria during the first 100 days post-BMT.32 Time-to-GVHD was defined as the time interval from allo-BMT to onset of any grade of aGVHD. Patients were monitored for cGVHD beginning at day 80 post-BMT, and cases were graded according to accepted definitions of limited and extensive disease.33 Tissue biopsies were obtained to distinguish toxicity, infection, and GVHD whenever possible. All leukemia patients were considered evaluable for relapse and disease-free survival.

Statistical methods.Distributions for time-to-engraftment, time-to-acute/chronic GVHD, time-to-relapse, survival, and disease-free survival (DFS) were evaluated using the method of Kaplan and Meier.34 Such variables were measured beginning on day 0. Values were censored for engraftment at time of death if the patient did not engraft, for GVHD at time of death or at time of second transplant, and for survival and relapse at last follow-up. Comparisons were implemented using the log-rank test. Cox proportional hazards multivariate regression was used to investigate relationships with prognostic variables including patient age, donor age, racial group, disease status at transplant, sex mismatch, CMV status, rejection and GVHD Ag disparities, total body irradiation (TBI) dose, acute/chronic GVHD, and chronic leukemia/other diagnosis. For second transplants, values for prognostic variables were those associated with the initial transplant, except that all donors were considered jointly for CMV seropositivity. Frequencies for categorical variables were compared using Fisher's exact test. Unless otherwise specified, all reported P values are for two-sided hypothesis tests. Confidence intervals (CI) were computed using standard techniques and reflect a confidence coefficient of 95% in each case.

Patient-Donor Characteristics

Twenty patients were considered low risk and 52 were high risk at the time of transplant (Table 1). Frequencies of HLA disparities in the donor and recipient show a positive association between high-risk disease category and the use of a 3-Ag mismatched graft. Table 2 shows characteristics of recipients and donors.

Table 2.

Recipient and Donor Characteristics

ParametersValues*
Median age in yr (range) 
Recipient 16 (1-50) 
Donor 27 (4-55) 
Recipient/donor CMV status 
Seropositive/seropositive 34 (47%) 
Seropositive/seronegative 12 (17%) 
Seronegative/seropositive 13 (18%) 
Seronegative/seronegative 13 (18%) 
Recipient race 
Caucasian 50 (69%) 
African American 16 (22%) 
Asian 3 (4%) 
Indian 3 (4%) 
Sex 
Recipient: M/F 49 (68%)/23 (32%) 
Donor: M/F 33 (46%)/39 (54%) 
Donor-recipient sex pairs 
Same: F → F/M → M 15 (21%)/25 (35%) 
Opposite: F → M/M → F 8 (11%)/24 (33%) 
Donor relationship to recipient 
Parent 32 (44%) 
Sibling 27 (37%) 
Child 11 (16%) 
Cousin 2 (2%) 
ParametersValues*
Median age in yr (range) 
Recipient 16 (1-50) 
Donor 27 (4-55) 
Recipient/donor CMV status 
Seropositive/seropositive 34 (47%) 
Seropositive/seronegative 12 (17%) 
Seronegative/seropositive 13 (18%) 
Seronegative/seronegative 13 (18%) 
Recipient race 
Caucasian 50 (69%) 
African American 16 (22%) 
Asian 3 (4%) 
Indian 3 (4%) 
Sex 
Recipient: M/F 49 (68%)/23 (32%) 
Donor: M/F 33 (46%)/39 (54%) 
Donor-recipient sex pairs 
Same: F → F/M → M 15 (21%)/25 (35%) 
Opposite: F → M/M → F 8 (11%)/24 (33%) 
Donor relationship to recipient 
Parent 32 (44%) 
Sibling 27 (37%) 
Child 11 (16%) 
Cousin 2 (2%) 

Abbreviation: CMV, cytomegalovirus.

*

Values are rounded to nearest whole number.

Graft Characteristics

The median degree of T-cell depletion was 1.8 logs (range, 1.2 to 2.8). Patients received a median of 1.5 × 108 per kilogram of mononuclear cells (range, 0.6 to 3.7) and 7.5 × 104 per kilogram of T cells (range, 0.02 to 1.61). Median CFU-GM and CD34+ doses were 7.0 × 104 per kilogram (range, 1.3 to 40.5) and 1.36 × 106 per kilogram (range, 0.14 to 8.92), respectively. Median CFU-GM dose in patients who engrafted after the first BMT was 7.26 × 104 versus 5.11 × 104 in those who did not (P = .391), whereas median CD34+ dose was 1.3 × 106 and 1.51 × 106, respectively (P = .229). Only T-cell dose was shown to have a significant effect on transplant outcomes in multivariate analysis (Table 3).

Table 3.

Prognostic Factors Associated With Outcomes Following PMRD Allo-BMT

OutcomePrognostic Factor3-150ReferenceRisk LevelRR3-15195% CIP Value
Level(unfavorable)
(favorable)
WBC 1,000 engraftment3-152 TBI dose 1,500 cGy 1,332 cGy 0.33 (0.18, 0.59) <.001 
 Rejection mismatch <3-Ag 3-Ag 0.29 (0.14, 0.57) <.001 
 Degree of TCDρ Less More 0.45 (0.21, 0.97) .043 
Stable engraftment TBI dose 1,500 cGy 1,332 cGy 0.46 (0.27, 0.80) .005 
 Rejection mismatch <3-Ag 3-Ag 0.38 (0.29, 0.70) .002 
Acute GVHD III-IV3-154 T-cell doseρ Lower Higher 7.17 (0.94, 54.98) .058 
Chronic GVHD3-154 TBI dose 1,332 cGy 1,500 cGy 3.54 (1.26, 9.89) .016 
Relapse3-154 Disease category Low risk High risk 3.57 (1.04, 12.20) .043 
Treatment failure¶# Disease category Low risk High risk 2.17 (1.05, 4.51) .037 
Death3-154 Disease category Low risk High risk 2.10 (1.01, 4.35) .046 
OutcomePrognostic Factor3-150ReferenceRisk LevelRR3-15195% CIP Value
Level(unfavorable)
(favorable)
WBC 1,000 engraftment3-152 TBI dose 1,500 cGy 1,332 cGy 0.33 (0.18, 0.59) <.001 
 Rejection mismatch <3-Ag 3-Ag 0.29 (0.14, 0.57) <.001 
 Degree of TCDρ Less More 0.45 (0.21, 0.97) .043 
Stable engraftment TBI dose 1,500 cGy 1,332 cGy 0.46 (0.27, 0.80) .005 
 Rejection mismatch <3-Ag 3-Ag 0.38 (0.29, 0.70) .002 
Acute GVHD III-IV3-154 T-cell doseρ Lower Higher 7.17 (0.94, 54.98) .058 
Chronic GVHD3-154 TBI dose 1,332 cGy 1,500 cGy 3.54 (1.26, 9.89) .016 
Relapse3-154 Disease category Low risk High risk 3.57 (1.04, 12.20) .043 
Treatment failure¶# Disease category Low risk High risk 2.17 (1.05, 4.51) .037 
Death3-154 Disease category Low risk High risk 2.10 (1.01, 4.35) .046 

Abbreviations: PMRD, partially mismatched related donor; allo-BMT, allogeneic bone marrow transplant; RR, relative risk; WBC, white blood cell; TBI, total body irradiation; Ag, antigen; TCD, T-cell depletion; GVHD, graft-versus-host disease.

F3-150

Only prognostic factors that were significant at the 0.05 level in multivariate analysis are shown.

F3-151

Relative risk (RR) applies to the unfavorable level of the prognostic factor compared with the favorable level used as the reference group.

F3-152

Engraftment to WBC count of ≥1,000/μL uses data from initial transplants only. RR < 1 indicates lower probability of engraftment and/or delayed engraftment for patients with an unfavorable level of the given prognostic factor.

ρ Degree of T-cell depletion and T-cell dose were entered as continuous variables in log10 units.

 Stable engraftment refers to achievement of WBC count of ≥1,000/μL using data for all patients receiving one or more transplants. RR < 1 indicates lower probability of engraftment and/or delayed engraftment for patients with an unfavorable level of the given prognostic factor.

F3-154

RR > 1 indicates higher risk of the adverse event for patients with an unfavorable level of the indicated prognostic factor.

F3-167

Treatment failure refers to relapse or death.

Engraftment

Fifty-nine patients established successful engraftment after initial transplantation at a median of 20 days, resulting in an estimate of the probability of engraftment at 32 days of 0.88 (CI, 0.80 to 0.97). Patients receiving the higher TBI dose engrafted more quickly than those receiving the lower dose (median, 16.5 v 20.0 days, respectively; P = .003; Fig 2). Engraftment rates did not differ by sex mismatch between donor and recipient (P = .880). Patients receiving 3-Ag rejection mismatched grafts had diminished engraftment compared with those with less than 3-Ag rejection mismatched grafts (P = .002; Fig 3). Of those who failed to engraft or maintain engraftment, 10 received a second transplant, and 6 of these engrafted. Thus, a total of 62 patients (87%) achieved stable engraftment. The estimate of the probability of engraftment by day 70 for all patients was 0.96 (CI, 0.90 to 1.00).

Fig. 2.

Estimated probability of engraftment to WBC count of 1,000/μL for patients receiving 1,332 cGy (n = 46) and 1,500 cGy (n = 26) of TBI. Patients receiving the higher TBI dose experienced earlier engraftment with an estimated probability of engraftment by 32 days of 0.90 compared with 0.86 in patients receiving 1,332 cGy (P = .003). Data are derived from first transplants only.

Fig. 2.

Estimated probability of engraftment to WBC count of 1,000/μL for patients receiving 1,332 cGy (n = 46) and 1,500 cGy (n = 26) of TBI. Patients receiving the higher TBI dose experienced earlier engraftment with an estimated probability of engraftment by 32 days of 0.90 compared with 0.86 in patients receiving 1,332 cGy (P = .003). Data are derived from first transplants only.

Close modal
Fig. 3.

Estimated probability of engraftment to WBC count of 1,000/μL for patients with 3-Ag rejection mismatch (n = 24) and those with less than 3-Ag rejection mismatch (n = 48). Delayed engraftment and lower probability of engraftment by 32 days occurred in the 3-Ag mismatched group (P = .002) in which the median time to engraftment was 25 days versus 18 days in the less than 3-Ag mismatched group. Data are derived from first transplants only.

Fig. 3.

Estimated probability of engraftment to WBC count of 1,000/μL for patients with 3-Ag rejection mismatch (n = 24) and those with less than 3-Ag rejection mismatch (n = 48). Delayed engraftment and lower probability of engraftment by 32 days occurred in the 3-Ag mismatched group (P = .002) in which the median time to engraftment was 25 days versus 18 days in the less than 3-Ag mismatched group. Data are derived from first transplants only.

Close modal

Acute and Chronic GVHD

Grade I to IV aGVHD occurred in 18 of 58 (31%) evaluable patients. Grade II to IV aGVHD was seen in 9 (16%) patients between days 15 and 38 (median, 20 days), whereas grades III to IV occurred in 4 (7%) patients. The estimated risk of greater than grade I aGVHD by 100 days was 0.16 (CI, 0.06 to 0.25), and 0.07 for greater than grade II (CI, 0.00 to 0.14; Fig 4). There was not a statistically significant difference in probabilities of aGVHD among patients receiving 1-, 2-, or 3-Ag GVHD mismatched grafts, the number of cases being 2 of 11, 3 of 25, and 4 of 22, respectively, for greater than grade I (P = .852), and 1 of 11, 1 of 25, and 2 of 22, respectively, for greater than grade II (P = .771).

Fig. 4.

Estimated risks of grade II to IV (9 cases) and grade III to IV (4 cases) aGVHD among 58 evaluable patients; probability of aGVHD by 100 days was 0.16 and 0.07, respectively.

Fig. 4.

Estimated risks of grade II to IV (9 cases) and grade III to IV (4 cases) aGVHD among 58 evaluable patients; probability of aGVHD by 100 days was 0.16 and 0.07, respectively.

Close modal

Chronic GVHD occurred in 17 (35%) of 48 evaluable patients 3 to 17 months post-BMT, with an estimated risk of 0.51 (CI, 0.33 to 0.68) within 18 months. Four patients developed extensive cGVHD, all with onset times of less than 6 months, resulting in a 6-month risk of 0.10 (CI, 0.01 to 0.20; Fig 5).

Fig. 5.

Estimated risks of limited and extensive chronic GVHD. By 2 years, the cumulative risks were 44% and 10%, respectively.

Fig. 5.

Estimated risks of limited and extensive chronic GVHD. By 2 years, the cumulative risks were 44% and 10%, respectively.

Close modal

Complications and Regimen-Related Toxicity

Nonhematologic grade 4 toxicity involved the gastrointestinal (25%), hepatic (19%) renal (10%), pulmonary (8%), cardiovascular (6%), and central nervous (4%) systems. There was a 7% risk of regimen-related early (<60 days) mortality, which occurred exclusively in high-risk patients.

Survival

Of 72 patients, 25 are surviving at a median follow-up of 24 months (range, 14 to 31 months). The overall median survival was 5.9 months (CI, 3.4 to 9.6), with an estimated 2-year survival probability of 0.35 (CI, 0.24 to 0.46). There was a statistically significant difference between survival distributions for risk groups in which there were 9 deaths among 20 low-risk patients compared with 38 of 52 high-risk patients with a 2-year survival probability of 0.55 (CI, 0.33 to 0.77) and 0.27 (CI, 0.15 to 0.39), respectively (P = .048; Fig 6). The relative risk (RR) of death for high-risk versus low-risk patients unadjusted for other factors was 2.0 (CI, 1.0 to 4.2; P = .053). An analysis to compare patients who received a 1-Ag GVHD mismatched graft with those who received a 2- or 3-Ag GVHD mismatched graft showed no statistically significant difference in the probability of survival (0.45 v 0.33, respectively; P = .272; Fig 7). There was also not a significant difference in survival according to TBI dose (P = .450), patient age ≤18 versus >18 years (P = .974), diagnosis (P = .994), racial group (P = .091), sex mismatch (P = .538), or CMV seropositivity (P = .860). Among surviving patients, 23 of 25 had Karnofsky clinical performance scores ≥90. The primary cause of death seen most frequently was relapse, occurring in 16 of 47 (34%) patients, followed by engraftment failure (n = 10), fungal infection (n = 6), nonfungal infection (n = 6), interstitial pneumonitis (n = 3), GVHD (n = 2), Epstein-Barr virus lymphoma (n = 1), veno-oclusive disease (n = 1), and other (n = 2).

Fig. 6.

Probability of survival for patients with low-risk and high-risk disease status. Estimated survival at 2 years was 0.55 and 0.27, respectively (P = .048).

Fig. 6.

Probability of survival for patients with low-risk and high-risk disease status. Estimated survival at 2 years was 0.55 and 0.27, respectively (P = .048).

Close modal
Fig. 7.

Probability of survival for recipients of a 1-Ag versus 2- or 3-Ag GVHD mismatched graft. Estimated survival at 2 years was 0.45 and 0.33, respectively (P = .272).

Fig. 7.

Probability of survival for recipients of a 1-Ag versus 2- or 3-Ag GVHD mismatched graft. Estimated survival at 2 years was 0.45 and 0.33, respectively (P = .272).

Close modal

Relapse

Twenty-two of 68 evaluable patients (32%) relapsed, which occurred before 16 months in all but 1 patient, who relapsed at 27 months; estimated risk of relapse at 2 years was 0.47 (CI, 0.31 to 0.62). Three of 19 evaluable patients in the low-risk group (16%) relapsed, compared with 19 of 49 in the high-risk group (39%), with estimated risks of relapse at 2 years of 0.21 and 0.58, respectively. Thus, there was a significant difference between risk groups (P = .031), but not between acute lymphoblastic leukemia (11/28), acute myelogenous leukemia (6/18), and chronic myelogenous leukemia (3/17) patients (P = .495).

DFS

Of 67 evaluable patients, 20 (30%) were alive and free of disease at median follow-up of 21.5 months. Median DFS was 4.2 months (CI, 3.0 to 6.2), and estimated 2-year DFS probability was 0.31 (CI, 0.20 to 0.42). There was a significant difference in 2-year DFS between low-risk and high-risk groups (0.53 and 0.23, respectively; P = .032). There was not a significant difference in DFS by diagnosis (P = .964).

Multivariate Analysis

Table 3 shows the results of multivariate analysis. Relative risk values less than 1.0 correspond to lower probability of engraftment, and values greater than 1.0 correspond to increased rates of aGVHD, cGVHD, relapse, treatment failure, and death. Lower TBI dose, 3-Ag rejection mismatch, and higher degree of T-cell depletion adversely affected engraftment. Higher T-cell dose was associated with severe aGVHD and higher TBI dose with cGVHD. High-risk disease category increased treatment failure from relapse and death; other factors were not significant after adjustment for risk category.

Almost a decade ago, for patients with leukemia it was shown that major HLA barriers would significantly interfere with success of allo-BMT from other than a 1-Ag mismatched haploidentical familial donor.7,18,21 However, in their series comparing PMRD and MSD recipients, Beatty et al7 found no difference in survival when patients were transplanted in remission. Nonetheless, rates of rejection and severe aGVHD, both of which carried a high mortality risk, were regarded as unacceptable, and transplants from a PMRD with more than one major HLA disparity were not generally recommended. Therefore, the attention of most investigators turned to using PMUDs. Unfortunately, this approach is costly, time-consuming, and has been attained for less than half of the patients who seek an alternative donor, usually excluding patients from ethnic and racial minorities. Our study defines techniques that circumvent major HLA barriers, thus expanding access to allo-BMT for almost all patients. We estimate that greater than 90% of patients will have a haploidentical family member who is immediately available.

During this past decade, investigations into new approaches for immunomodulation after PMRD transplantation have achieved higher rates of engraftment and control of aGVHD.9,35-37 Henslee-Downey et al9 explored a strategy that combined ex vivo and in vivo T-cell lysis using two experimental MoAb products targeted to T-cell–specific antigens for sequential immunotherapy. For marrow T-cell depletion, T10B9 was used,38 which was shown by Ash et al39 to be effective in reducing the risk of severe aGVHD after PMUD transplants. An immunotoxin, H65-RTA,40 was used for in vivo T-cell lysis. Patients experienced a 93% engraftment rate and a 36% risk of grade II to IV aGVHD. In our larger study, T10B9 was again used for T-cell depletion, ATG replaced H65-RTA, and low-dose cyclosporin was added in an effort to improve outcome using full haplodisparate PMRDs.

Engraftment kinetics significantly improved after more intensive host conditioning from an increase in the dose of TBI. This may be the result of creating a more empty marrow space and/or decreasing immunologic resistance to engraftment. We did not detect a significant difference in engraftment based on the CD34+ cell dose, contrary to a report from Aversa et al36 showing improved engraftment in haploidentical recipients receiving T-cell–depleted, G-CSF–mobilized peripheral blood preparations in combination with T-cell–depleted bone marrow. However, it is possible that our patients benefited from a similar mechanism through systemic administration of G-CSF immediately after graft infusion. Both studies indicate the need for sufficient immunologic ablation of the recipient to achieve successful engraftment, the toxicity of which must be carefully evaluated and constrained.

In our study, a more highly T-cell–depleted marrow graft was associated with diminished engraftment in multivariate analysis, which agrees with other studies41,42 and cautions against potent depletion methods. Consistent with Anasetti et al18 and others,8 our data showed that the degree of major HLA mismatch for rejection also had an adverse effect on engraftment. Because there are often multiple familial donors available for any given patient, one could preferentially choose donors who exhibit the least mismatch and, when possible, are homozygous for major HLA antigens.

Moderate-to-severe aGVHD was reduced in our study to an estimated risk of 16%, which is lower than that often published for major HLA “compatible” grafts from a PMUD or even from an MSD, which ranges between 33% to 78% and 23% to 42%, respectively.11-16,20-22,39,43-48 Furthermore, the aGVHD rate was not associated with the degree of recipient major HLA mismatch, which differs from previous experience with alternative donors.7,8,39,43 This could be caused by the low overall incidence of aGVHD as a result of highly effective prophylaxis. Another factor may include the role that nonmajor HLA and/or minor histocompatibility antigens plays in the genesis of aGVHD.49 Most PMUDs are not molecularly identical for major HLAs50 and are unlikely to share other histocompatibility antigens, whereas PMRDs share genotypic major histocompatibility complex (MHC) haplotype identity, including HLA variant and MHC non-HLAs. In addition, PMRDs may share genotypic identity for minor histocompatibility antigens, which could result in a broader immunologic compatibility.5,51,52 Our results challenge the requisite for major HLA “matching” to define an acceptable allogeneic alternative donor.

In multivariate analysis, a greater number of T cells in the donor marrow graft was associated with a higher risk of severe aGVHD, supporting the concept that minimizing the dose of T cells administered at the time of graft infusion is an important step in the control of aGVHD. It remains a challenge to produce a graft with optimal quantitative and qualitative properties that will ensure engraftment while minimizing the risk of severe aGVHD. Our study suggests that depletion should not exceed approximately 2.8 logs if a high probability of engraftment is to be attained and that post-BMT immunotherapy must then be used for control of aGVHD.9 For this purpose, the combination of lympholytic and suppressive agents used in our study was successful.

The majority of our patients were in a high-risk disease category, which, in multivariate analysis, was the only significant prognostic factor associated with treatment failure from relapse or death. This association is well established after all types of BMT using either autologous or allogeneic stem cells. Avoidance of haploidentical donors has been based primarily on concerns regarding transplant-related mortality, and yet the leading cause of death in our patients was relapse, particularly among those at high risk. One might ask whether the successful avoidance of aGVHD activity in our patients might have interfered with a graft-versus-leukemia effect shown to be operative in the cure of leukemic, allo-BMT recipients.53 However, the risk of relapse falls within the range reported from large series of similar-risk patients.54 Estimates of 2-year survival and DFS, based on disease-risk category, are comparable to results after either PMUD11-16,20,22,39,43-46 or MSD BMT.54-58 

Prospective randomized trials comparing PMRDs, PMUDs, and cord blood stem cells59-62 are needed to determine whether there is a preference among available alternative donor sources. In the absence of differences, pragmatic and logistic issues surface, those being primarily cost and time. In this respect, the use of a PMRD becomes advantageous by creating immediate donor availability and reducing costs associated with obtaining the graft, including substantial federal and private funding required to develop and maintain donor registries and banks. The timing of transplant, preferably when patients are in remission, is the one factor most likely to improve long-term outcome. The ability to use haploidentical family donors provides near-unrestricted access to allo-BMT.

The authors thank the BMT program staff, nursing service, and laboratory team for the dedicated and skilled care given to the patients and their families. We are indebted to the clinical research data management team, biostatistics group, and secretarial staff for their invaluable service in the preparation of this manuscript.

Supported in part by grants from the Center for Cancer Treatment and Research Board of Directors and the South Carolina Cancer Center.

Address reprint requests to P. Jean Henslee-Downey, MD, Division of Transplantation Medicine, 7 Richland Medical Park, Columbia, SC 29203.

1
Armitage JO: Medical progress. Bone marrow transplantation. N Engl J Med 330:827 1994
2
Hows
JM
Yin JL; Marsh J
Swirsky
D
Jones
L
Apperley
JF
James
DCO
Smithers
S
Batchelor
JR
Goldman
JM
Gordon-Smith
EC
Histocompatible unrelated volunteer donors compared with HLA non-identical family donors in marrow transplantation for aplastic anemia and leukemia.
Blood
68
1986
1322
3
O'Reilly
RJ
Bone marrow transplant in patients lacking an HLA matched sibling donor.
Pediatr Ann
20
1991
682
4
Trigg
ME
Bone marrow transplantation using alternative donors.
Am J Pediatr Hematol Oncol
15
1993
141
5
Henslee-Downey
PJ
Mismatched bone marrow transplantation.
Curr Opin Oncol
7
1995
115
6
Powles
RL
Morgenstern
GR
Kay
HEM
McElwain
TJ
Clink
HM
Dady
PJ
Barrett
A
Jameson
B
Depledge
MH
Watson
JG
Sloane
J
Leigh
M
Lumley
H
Hedley
D
Lawler
SD
Filshie
J
Robinson
B
Mismatched family donors for bone marrow transplantation as treatment for acute leukemia.
Lancet
8
1983
612
7
Beatty
PG
Cliff
RA
Mickelson
EM
Nisperos
BB
Flournoy N; Martin PJ
Sanders
JE
Stewart
P
Buckner
CD
Storb
R
Thomas
ED
Hansen
JA
Marrow transplantation from related donors other than HLA-identical siblings.
N Engl J Med
313
1985
765
8
Ash
RC
Horowitz
MM
Gale
RP
van Bekkum
DW
Casper
JT
Gordon-Smith
EC
Henslee
PJ
Kolb
H-J
Lowenberg
B
Masaoka
T
McGlave
PB
Rimm
AA
Ringden
O
van Rood
JJ
Sondel
PM
Vowels
MR
Bortin
MM
Bone marrow transplantation from related donors other than HLA-identical siblings: Effect of T-cell depletion.
Bone Marrow Transplant
7
1991
443
9
Henslee-Downey
PJ
Parrish
RS
Macdonald
JS
Romond
EH
Marciniack
E
Coffey
C
Ciocci
G
Thompson
JS
Combined in vitro and in vivo T-lymphocyte depletion for the control of graft-versus-host disease following haploidentical marrow transplant.
Transplantation
61
1996
738
10
Hansen
JA
Clift
RA
Thomas
ED
Buckner
CD
Storb
R
Biblett
ER
Transplantation of marrow from an unrelated donor to a patient with acute leukemia.
N Engl J Med
303
1980
565
11
Gingrich
RD
Ginder
GD
Goeken
NE
Howe
CWS
Wen
B-C
Hussey
DH
Fyfe
MA
Allogeneic marrow grafting with partially mismatched unrelated marrow donor.
Blood
71
1988
1375
12
Hows
J
Bradley
BA
Gore
S
Downie
T
Howard
M
Gluckman
E
Prospective evaluation of unrelated donor bone marrow transplantation.
Bone Marrow Transplant
12
1993
371
13
Kernan
NA
Bartsch
G
Ash
RC
Beatty
PG
Champlin
R
Filipovich
A
Gajewski
J
Hansen
JA
Henslee-Downey
J
McCullough
J
McGlave
P
Perkins
HA
Phillips
GL
Sanders
J
Stroncek
D
Thomas
ED
Blume
KG
Analysis of 462 transplantations from unrelated donors facilitated by the National Marrow Donor Program.
N Engl J Med
328
1993
593
14
Champlin
R
Bone marrow transplantation for leukemia utilizing HLA-matched unrelated donors.
Bone Marrow Transplant
11
1993
74
15
Casper
J
Camitta
B
Truitt
R
Baxter-Lowe
LA
Bunin
N
Lawton
C
Murray
K
Hunter
J
Pietryga
D
Garbrecht
F
Taylor
CK
Drobyski
W
Horowitz
M
Flomenberg
N
Ash
R
Unrelated bone marrow donor transplants for children with leukemia and myelodysplasia.
Blood
85
1995
2354
16
Nademanee
A
Schmidt
GM
Parker
P
Dagis
AC
Stein
A
Snyder
DS
O'Donnell
M
Smith
EP
Stepan
DE
Molina
A
Wong
KK
Margolin
K
Somlo
G
Littrell
B
Woo
D
Sniecinski
I
Niland
JC
Forman
SJ
The outcome of matched unrelated donor bone marrow transplantation in patients with hematologic malignancies using molecular typing for donor selection and graft-versus-host disease prophylaxis regimen of cyclosporin, methotrexate, and prednisone.
Blood
86
1995
1228
17
Beatty
PG
Mori
M
Milford
E
Impact of racial genetic polymorphism on the probability of finding an HLA-matched donor.
Transplantation
60
1995
778
18
Anasetti
C
Amos
D
Beatty
PG
Appelbaum
FR
Bensinger
W
Buckner
CD
Clift
R
Doney
K
Martin
PJ
Mickelson
E
Nisperos
B
O'Quigley
J
Ramberg
R
Sanders
JE
Stewart
P
Storb
R
Sullivan
KM
Witherspoon
RP
Thomas
ED
Hansen
JA
Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma.
N Engl J Med
320
1989
197
19
Davies
SM
Ramsay
NKC
Haake
RJ
Kersey
JH
Weisdorf
DJ
McGlave
PB
Blazar
BR
Comparison of engraftment in recipients of matched sibling or unrelated donor marrow allografts.
Bone Marrow Transplant
13
1994
51
20
Gajewski
JL
Ho
WG
Feig
SA
Hunt
L
Kaufman
N
Champlin
RE
Bone marrow transplantation using unrelated donors for patients with advanced leukemia or bone marrow failure.
Transplantation
50
1990
244
21
Hansen
JA
Anasetti
C
Beatty
PG
Martin
PJ
Sanders
JE
Storb
R
Thomas
ED
Treatment of leukemia by marrow transplantation from HLA incompatible donors: Effect of HLA-disparity on GVHD, relapse and survival.
Bone Marrow Transplant
6
1990
108
22
McGlave
P
Bartsch
G
Anasetti
C
Ash
R
Beatty
P
Gajewski
J
Kernan
NA
Unrelated donor marrow transplantation therapy for chronic myelogenous leukemia: Initial experience of the National Marrow Donor Program.
Blood
81
1993
249
23
Ringden
O
Nilsson
B
Death by graft-vs.-host disease associated with HLA mismatch, high recipient age, low marrow cell dose and splenectomy.
Transplantation
40
1985
39
24
Sullivan
KM
Mori
M
Sanders
J
Siadak
M
Witherspoon
RP
Anasetti
C
Appelbaum
FR
Bensinger
W
Bowden
R
Buckner
CD
Clark
J
Crawford
S
Deeg
HJ
Doney
K
Flowers
M
Hansen
J
Loughran
T
Martin
P
McDonald
G
Pepe
M
Petersen
FB
Schuening
F
Stewart
P
Storb
R
Late complications of allogeneic and autologous marrow transplantation.
Bone Marrow Transplant
10
1992
127
25
Atkinson
K
Farewell
V
Storb
R
Tsoi
M-S
Sullivan
KM
Witherspoon
RP
Fefer
A
Clift
R
Goodell
B
Thomas
ED
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
26
Shapiro
RS
McClain
K
Frizzera
G
Gajl-Peczalska
KJ
Kersey
JH
Blazar
BR
Arthur
DC
Patton
DF
Greenberg
JS
Burke
B
Ramsay
NKC
McGlave
P
Fillipovich
AH
Epstein-Barr virus associated B cell lymphoproliferative disorders following bone marrow transplantation.
Blood
71
1988
1234
27
Vartdal
F
Gaudernack
G
Funderud
S
Bratle
A
Lea
T
Ugelstad
J
Thorsby
E
HLA Class I and II typing using cells positively selected from blood by immunomagnetic isolation — A fast and reliable technique.
Tissue Antigens
28
1986
301
28
Baxter-Lowe LA: HLA testing using molecular genetic tools, in Allen RW, AuBuchon JP (Eds): Molecular Genetics in Diagnosis and Research. Bethesda, MD, American Association of Blood Banks, 1995, p 81
29
Henslee-Downey
PJ
Choosing an alternative donor among available family members.
Am J Pediatr Hematol Oncol
15
1993
150
30
Lee
C
Brouillette
M
Lamb
L
Lamb
L
Pati
A
Brown
S
Thompson
J
Parrish
R
Henslee-Downey
PJ
Gee
AP
Use of a closed system for V αβ-positive T-cell depletion of marrow for use in partially mismatched related donor transplantation.
Prog Clin Biol Res
389
1994
523
31
Kernan NA, Collins NH, Bleau SA, O'Reilly RJ: Evaluation of T-cell depletion: Limiting dilution analysis for clonable T cells-microtechnique, in Areman E, Deeg HJ, Sacher RA (Eds): Bone Marrow and Stem Cell Processing: A Manual of Current Techniques. Philadelphia, PA, FA Davis, 1992, p 423
32
Glucksberg
H
Storb
R
Fefer
A
Buckner
CD
Neiman
PE
Clift
RA
Lerner
KG
Thomas
ED
Clinical manifestations of graft-versus-host disease in human recipients of marrow from HLA-matched sibling donors.
Transplantation
18
1974
295
33
Shulman
HM
Sullivan
KM
Weiden
PL
McDonald
GB
Striker
GE
Sale
GE
Hackman
R
Tsoi
M-S
Storb
R
Thomas
ED
Chronic graft-versus-host syndrome in man. A long-term clinicopathological study of 20 Seattle patients.
Am J Med
69
1980
204
34
Kaplan
EL
Meier
P
Non-parametric estimation from incomplete observations.
J Am Stat Assoc
53
1958
457
35
Trigg
ME
Gingrich
R
Goeken
N
deAlarcon
P
Klugman
M
Padley
D
Rumelhart
S
Giller
R
Wen
B-C
Strauss
R
Low rejection rate when using unrelated or haploidentical donors for children with leukemia undergoing marrow transplantation.
Bone Marrow Transplant
4
1989
431
36
Aversa
F
Tabilio
A
Terenzi
A
Velardi
A
Falzetti
F
Giannoni
C
Iacucci
R
Zei
T
Martelli
MP
Gambelunghe
C
Rossetti
M
Caputo
P
Latini
P
Aristei
C
Raymondi
C
Reisner
Y
Martelli
MF
Successful engraftment of T-cell-depleted haploidentical “three-loci” incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum.
Blood
84
1994
3948
37
Fleming
DR
Henslee-Downey
PJ
Romond
EH
Harder
EJ
Marciniak
E
Mann
RK
Messino
MJ
Macdonald
JS
Bishop
M
Rayens
MK
Thompson
JS
Foon
KA
Success of allogeneic bone marrow transplantation with T cell-depleted partially matched related donors for advanced acute lymphoblastic leukemia in children and adults: A comparative matched cohort study.
Bone Marrow Transplant
17
1996
917
38
Marciniak E, Henslee-Downey PJ, Thompson JS, Bailey K: Bone marrow purging of T-lymphocytes with T10B9 monoclonal antibody and compliment, in Areman E, Deeg HJ, Sacher RA (Eds): Bone Marrow and Stem Cell Processing: A Manual of Current Techniques. Philadelphia, PA, FA Davis, 1992, p 423
39
Ash
RC
Casper
JT
Chitambar
CR
Hansen
R
Bunin
N
Truitt
RL
Lawton
C
Murray
K
Hunter
J
Baxter-Lowe
LA
Gottschall
JL
Oldham
K
Anderson
T
Camitta
B
Menitove
J
Successful allogeneic transplantation of T-cell-depleted bone marrow from closely HLA-matched unrelated donors.
N Engl J Med
322
1990
485
40
Kernan
NA
Knowles
RW
Burnes
MJ
Broxmeyer
HE
Lu
L
Lee
HM
Kawahata
RT
Scannon
PJ
Dupont
B
Specific inhibition of in vitro lymphocyte transformation by an anti-pan T-cell (gp67) ricin A chain immunotoxin.
J Immunol
133
1984
137
41
O'Reilly
RJ
Collins
NH
Kernan
N
Brochstein
J
Dinsmore
R
Kirkpatrick
D
Siena
S
Keever
C
Jordan
B
Shank
B
Wolf
L
Dupont
B
Reisner
Y
Transplantation of marrow-depleted T-cells by soybean lectin agglutination and E-rosette depletion: Major histocompatibility complex-related graft resistance in leukemic transplant patients.
Transplant Proc
17
1985
455
42
Martin PJ, Kernan NA: T-cell depletion for the prevention of graft-versus-host disease in man, in Burakoff SJ, Deeg HJ, Ferrara J, Atkinson K (Eds): Graft-vs.-Host Disease: Immunology, Pathophysiology, and Treatment. New York, NY, Marcel Dekker, 1990, p 371
43
Beatty
PG
Anasetti
C
Hansen
JA
Longton
GM
Sanders
JE
Martin
PJ
Mickelson
EM
Choo
SY
Petersdorf
EW
Pepe
MS
Appelbaum
FR
Bearman
SI
Buckner
CD
Clift
RA
Petersen
FB
Singer
J
Stewart
PS
Storb
RF
Sullivan
KM
Tesler
MC
Witherspoon
RP
Thomas
ED
Marrow transplantation from unrelated donors for treatment of hematologic malignancies: Effect of mismatching for one HLA locus.
Blood
81
1993
249
44
Grovas
A
Feig
SA
O'Rouke
S
Valentino
L
Wiley
F
Hunt
L
Landaw
EM
Gayexski
J
Unrelated donor bone marrow transplants in children.
Cell Transplant
3
1994
413
45
Drobyski WR, Ash RC, Casper JT, McAuliffe T, Horowitz MM, Lawton C, Keever C, Baxter-Lowe LA, Camitta B, Garbrecht F, Pietryga D, Hansen R, Chitambar CR, Anderson T, Flomenberg N: Effect of T-cell depletion as graft-versus-host disease prophylaxis on engraftment, relapse, and disease-free survival in unrelated marrow transplantation for chronic myelogenous leukemia. Blood 83:1980, 1994.
46
Balduzzi
A
Gooley
T
Anasetti
C
Sander
JE
Martin
PJ
Petersdorf
EW
Appelbaum
FR
Buckner
CD
Matthews
D
Storb
R
Sullivan
KM
Hansen
JA
Unrelated donor marrow transplantation in children.
Blood
86
1995
3247
47
Deeg HJ, Cottler-Fox M: Clinical spectrum and pathophysiology of acute graft-vs.-host disease, in Burakoff SJ, Deeg HJ, Ferrara J, Atkinson K (Eds): Graft-vs.-Host Disease: Immunology, Pathophysiology, and Treatment. New York, NY, Marcel Dekker, 1990, p 311
48
Ringden
O
Horowitz
MM
Sondel
P
Gale
RP
Biggs
JC
Champlin
RE
Deeg
HJ
Dicke
K
Masaoka
T
Powles
RL
Rimm
AA
Rozman
C
Sobocinski
KA
Speck
B
Zwaan
FE
Bortin
MM
Methotrexate, cyclosporin, or both to prevent graft-versus-host disease after HLA-identical sibling bone marrow transplants for early leukemia?
Blood
81
1993
1094
49
Korngold
B
Sprent
J
Lethal graft-versus-host disease after bone marrow transplantation across minor histocompatibility barriers in mice. Prevention by removing mature T cells from marrow.
J Exp Med
148
1978
1687
50
Petersdorf
EW
Longton
GM
Anasetti
C
Martin
PJ
Mickelson
EM
Smith
AG
Hansen
JA
The significance of HLA-DRB1 matching on clinical outcome after HLA-A, B, DR identical unrelated donor marrow transplantation.
Blood
86
1995
1606
51
Beatty
PG
Results of allogeneic bone marrow transplantation with unrelated or mismatched donors.
Semin Oncol
19
1992
13
52
Beatty
PG
The immunogenetics of bone marrow transplantation.
Transfus Med Rev
8
1994
45
53
Horowitz
MM
Gale
RP
Sondel
PM
Goldman
JM
Kersey
J
Kolb
H-J
Rimm
AA
Ringden
O
Rozman
C
Speck
B
Truitt
RL
Zwaan
FE
Bortin
MM
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
75
1990
555
54
Rowlings
PA
Gale
RP
Horowitz
MM
Bortin
MM
Bone marrow transplantation in leukemia.
J Hematother
3
1994
235
55
Mitus
AJ
Miller
KB
Schenken
DP
Ryan
HF
Parsons
SK
Wheeler
C
Antin
JH
Improved survival for patients with acute myelogenous leukemia.
J Clin Oncol
13
1995
560
56
Brown
RA
Wolff
SN
Fay
JW
Pineiro
L
Collins
RH
Lynch
JP
Stevens
D
Greer
J
Herzig
RH
Herzig
GP
High-dose etoposide, cyclophosphamide, and total body irradiation with allogeneic bone marrow transplantation for patients with acute myeloid leukemia in untreated first relapse: A study by the North American Marrow Transplant Group.
Blood
85
1995
1391
57
Lynch
MH
Petersen
FB
Appelbaum
FR
Bensinger
WI
Clift
RA
Storb
R
Sanders
JE
Hansen
JA
Buckner
CD
Phase II study of busulfan, cyclophosphamide and fractionated total body irradiation as a preparatory regimen for allogeneic bone marrow transplantation in patients with advanced myeloid malignancies.
Bone Marrow Transplant
15
1995
59
58
Dunlop
LC
Powles
R
Singhal
S
Treleaven
JG
Swansbury
GJ
Meller
S
Pinkerton
CR
Horton
C
Mehta
J
Bone marrow transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia.
Bone Marrow Transplant
17
1996
365
59
Broxmeyer
HE
Kurtzberg
J
Gluckman
E
Auerbach
AD
Douglas
G
Cooper
S
Falkenburg
JH
Bard
J
Boyse
EA
Umbilical cord blood hemotopoietic stem cell and repopulating cells in human clinical transplantation.
Blood Cells
17
1991
313
60
Kurtzberg
J
Graham
M
Casey
J
Olson
J
Stevens
CE
Rubinstein
P
The use of umbilical cord blood in mismatched related and unrelated hemopoietic stem cell transplantation.
Blood Cells
20
1994
275
61
Wagner
JE
Kernan
NA
Steinbuch
M
Broxmeyer
HE
Gluckman
E
Allogeneic sibling umbilical cord blood transplantation in children with malignant and non-malignant disease.
Lancet
22
1995
214
62
Kurtzberg
J
Laughlin
M
Graham
ML
Smith
C
Olson
JF
Halperin
EC
Ciocci
G
Carrier
C
Stevens
CE
Rubinstein
P
Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients.
N Engl J Med
335
1996
157
Sign in via your Institution