Allogeneic conventional hematopoietic cell transplantation (HCT) can be curative treatment for lymphoid malignancies, but it has been characterized by high nonrelapse mortality (NRM). Here, we compared outcomes among patients with lymphoma or chronic lymphocytic leukemia given either nonmyeloablative (n = 152) or myeloablative (n = 68) conditioning. Outcomes were stratified by the HCT-specific comorbidity index. Patients in the nonmyeloablative group were older, had more previous treatment and more comorbidities, more frequently had unrelated donors, and more often had malignancy in remission compared with patients in the myeloablative group. Patients with indolent versus aggressive malignancies were equally distributed among both cohorts. After HCT, patients without comorbidities both in the nonmyeloablative and myeloablative cohorts had comparable NRM (P = .74), overall survival (P = .75), and progression-free survival (P = .40). No significant differences were observed (P = .91, P = .89, and P = .40, respectively) after adjustment for pretransplantation variables. Patients with comorbidities experienced lower NRM (P = .009) and better survival (P = .04) after nonmyeloablative conditioning. These differences became more significant (P < .001 and .007, respectively) after adjustment for other variables. Further, nonmyeloablative patients with comorbidities had favorable adjusted progression-free survival (P = .01). Patients without comorbidities could be enrolled in prospective randomized studies comparing different conditioning intensities. Younger patients with comorbidities might benefit from reduced conditioning intensity.

Only limited options for curative treatment are available for patients with relapsed or refractory lymphoid malignancies who either failed autologous hematopoietic cell transplantation (HCT) or lacked stem cells for autologous HCT.1,3  In some patients, prolonged remissions have been achieved with allogeneic HCT after myeloablative conditioning, presumably resulting from tumor cell kill from both high-intensity conditioning and graft-versus-malignancy effects and because of the use of tumor-free grafts.4,6  However, up to 60% nonrelapse mortality (NRM) has been observed even among patients younger than 50 years of age with acceptable performance status.7,,,11  Unfortunately, most patients with lymphoma or chronic lymphocytic leukemia (CLL) are older than 50 and have comorbidities.12  Reduced-intensity conditioning regimens have been developed to decrease NRM and to extend the use of allogeneic HCT to these older patients and to those with comorbidities.13,,,,,,,,,,,,,,,29  Systematic comparisons between reduced-intensity and conventional HCT have been underreported in the literature, particularly those balanced by the impact of important prognostic factors. Accurate risk assessment for the 2 approaches would be useful for patient counseling and stratification in prospective studies. In the present study, we assessed the role of conditioning intensity on outcomes among patients with lymphoma and CLL given allogeneic HCT following either nonmyeloablative or myeloablative conditioning regimens.

The retrospective analysis was approved by the institutional review board of the Fred Hutchinson Cancer Research Center (FHCRC; Seattle, WA). Informed consent was obtained from all patients at time of transplantation in accordance with the Declaration of Helsinki.

Patients

Patients and disease characteristics are described in Table 1. Consecutive patients (n = 220) with lymphoma or CLL given allogeneic HCT at the FHCRC between December 1997 and June 2005 were included in this analysis. December 1997 was chosen as the starting point because it coincided with the initiation of nonmyeloablative conditioning at FHCRC. Most patients (n = 152) received nonmyeloablative conditioning regimens (nonmyeloablative patients), which consisted of 2 Gy total-body irradiation (TBI) alone or preceded by fludarabine, 90 mg/m2; all received postgrafting immunosuppression with mycophenolate mofetil and cyclosporine (CSP).30,32  Sixty-eight patients received myeloablative conditioning regimens (myeloablative patients), which included cyclophosphamide (CY) combined with either at least 12 Gy TBI or busulfan (levels targeted to plasma mean steady-state concentrations of 800-900 ng/mL)33 ; most were given methotrexate/CSP for graft-versus-host disease (GVHD) prophylaxis.34  Overall, nonmyeloablative protocols were offered to patients who were either 50 or older or, if younger than 50, had significant pre-existing medical problems or had failed high-dose autologous HCT. Only 8 patients, 38 to 48 years old, who did not meet these criteria were enrolled in nonmyeloablative protocols based on their own preference (n = 4) or on consensus decisions from the Patient Care Conference of the FHCRC Faculty (n = 4), which was based on the chemosensitive nature of their disease. Nonmyeloablative patients were treated in the outpatient clinic during the first 100 days before returning to their referring physicians and were admitted to the hospital only as required for treatment of complications. All myeloablative patients were hospitalized for 3 to 4 weeks before being discharged to the outpatient clinic.

Patients and donors were matched for HLA-A, -B, and -C antigens by either intermediate resolution DNA typing (to a level at least as sensitive as serology) or by high-resolution techniques. Patients and donors were matched for HLA-DRB1 and DQB1 alleles.35  All patients received infection prophylaxis according to standard institutional guidelines.36,,,40  Aggressive diseases included CLL with Richter transformation, Hodgkin disease (HD) with the exception of nodular lymphocyte-predominant subtype, and aggressive lymphoma as defined by the Physician Data Query Modification of the Revised European American Lymphoma Classification of Lymphoproliferative Diseases (see the link http://www.cancer.gov/cancertopics/pdq/treatment/adult-non-hodgkins/HealthProfessional/page2#Section_17), whereas indolent diseases included all other histologic subtypes.

Assessment of pretransplantation comorbidities

Comorbidities were assessed by comprehensive review of medical records and computer database systems. Scores were assigned by a single investigator (M.L.S.) using definitions of the 17 comorbidities included in the HCT-specific comorbidity index (HCT-CI).41  The HCT-CI included thresholds for pulmonary, hepatic, and renal function tests to allow for sensitive detection of organ impairments that might compromise the ability of patients to tolerate a given conditioning regimen.

Half of the current patients were included in the original training set used to develop the HCT-CI. Because no direct comparisons of outcomes on the basis of comorbidity scores were made, inclusion of these patients was not thought to bias the comparison of outcomes on the basis of conditioning regimen intensity.

Statistical methods

Survival curves and probabilities were estimated according to the Kaplan-Meier method, whereas cumulative incidence curves and probabilities for NRM were estimated as described.42  Multivariate hazard ratios (HRs) for NRM and survival outcomes were estimated from Cox regression models, treating NRM and relapse or progression of malignancy as competing risks where appropriate. Adjusted survival curves were estimated based on methods described by Makuch.43  Briefly, the adjusted survival curve for the nonmyeloablative group represented a model-based projection of survival for a group of patients with the same baseline hazard function as estimated for those patients, but with the covariate characteristics of the myeloablative group. These estimates were derived from Cox regression models incorporating the adjustment factors as covariates and stratified on conditioning group. Adjusted cumulative incidence curves were estimated by applying the same adjustment procedure to the component cause-specific survival functions and then computing the cumulative incidence curve in the usual way.

Pretransplantation characteristics

In part because of protocol inclusion and exclusion criteria, the following differences were found among pretransplantation characteristics of the 2 groups (Table 1).

Table 1

Patients and disease characteristics

CharacteristicsNonmyeloablative patients, n = 152Myeloablative patients, n = 68
Median age, y (range) 60 (18-70) 46 (10-59) 
Median follow-up, mo (range) 44 (7-90) 61 (21-97) 
Median number of previous regimens (range) 5 (1-13) 3 (1-7) 
Median interval between diagnosis and HCT, mo (range) 7.0 (0.2-254) 6.6 (0.2-157) 
Previous autologous high-dose HCT, %   
    Overall 52 10 
    Planned/failed 11/41 0/10 
Conditioning regimens, %   
    2 Gy TBI 18 
    Flu + 2 Gy TBI 82 
    Cy + ≥12 Gy TBI 87 
    tBu + Cy 13 
Diagnosis, %   
    NHL 54 78 
        Low-grade 14 27 
        High-grade 40 51 
    CLL 26 15 
    HD 20 
Disease nature, %*   
    Indolent 37 41 
    Aggressive 63 59 
Disease status at the time of HCT, %   
    Sensitive 46 25 
    Refractory/untested relapse 40/14 53/21 
Hematopoietic cell source, %   
    G-PBMC 99 69 
    Marrow 31 
Donor type, %   
    HLA-matched sibling 55 68 
    HLA- antigen mismatched related 
    HLA-matched unrelated 39 25 
    HLA- antigen mismatched unrelated 
Patient CMV sero-status, %   
    Negative 43 44 
    Positive 57 56 
CharacteristicsNonmyeloablative patients, n = 152Myeloablative patients, n = 68
Median age, y (range) 60 (18-70) 46 (10-59) 
Median follow-up, mo (range) 44 (7-90) 61 (21-97) 
Median number of previous regimens (range) 5 (1-13) 3 (1-7) 
Median interval between diagnosis and HCT, mo (range) 7.0 (0.2-254) 6.6 (0.2-157) 
Previous autologous high-dose HCT, %   
    Overall 52 10 
    Planned/failed 11/41 0/10 
Conditioning regimens, %   
    2 Gy TBI 18 
    Flu + 2 Gy TBI 82 
    Cy + ≥12 Gy TBI 87 
    tBu + Cy 13 
Diagnosis, %   
    NHL 54 78 
        Low-grade 14 27 
        High-grade 40 51 
    CLL 26 15 
    HD 20 
Disease nature, %*   
    Indolent 37 41 
    Aggressive 63 59 
Disease status at the time of HCT, %   
    Sensitive 46 25 
    Refractory/untested relapse 40/14 53/21 
Hematopoietic cell source, %   
    G-PBMC 99 69 
    Marrow 31 
Donor type, %   
    HLA-matched sibling 55 68 
    HLA- antigen mismatched related 
    HLA-matched unrelated 39 25 
    HLA- antigen mismatched unrelated 
Patient CMV sero-status, %   
    Negative 43 44 
    Positive 57 56 

Flu indicates fludarabine; and tBu, targeted busulfan.

*

Aggressive diseases included CLL with Richter's transformation, HD with the exception of nodular lymphocyte–predominant subtype, and aggressive lymphoma as defined by the Physician Data Query Modification of the Revised European American Lymphoma Classification of Lymphoproliferative Diseases (see http://www.cancer.gov/cancertopics/pdq/treatment/adult-non-hodgkins/HealthProfessional/page2#Section_17), whereas indolent diseases included all other histological subtypes.

Complete or partial remission.

Included 6 nonmyeloablative and 2 myeloablative patient/donor pairs with single allele mismatch.

Nonmyeloablative patients were older (median age 60 vs 46 years), with 59% versus 18% 50 or older and 21% versus 0% 60 or older, had received more preceding chemotherapy regimens (median of 5 vs 3) and more often received previous high-dose HCT (52% vs 10%) than myeloablative patients. In addition, nonmyeloablative patients more often received grafts from unrelated donors (44% vs 28%) and granulocyte-colony stimulating factor-mobilized peripheral blood mononuclear cell (G-PBMC) grafts (99% versus 69%) compared with myeloablative patients. Diagnoses included non-Hodgkin lymphoma (NHL), HD, and CLL in 53%, 20%, and 27% of nonmyeloablative patients and in 78%, 7%, and 15% of myeloablative patients. Although no differences were found in the distribution of indolent versus aggressive lymphoproliferative malignancies between nonmyeloablative and myeloablative patients, more nonmyeloablative patients had complete or partial remissions at the time of HCT compared with their myeloablative counterparts (46% vs 25%). The median follow-up times of surviving nonmyeloablative and myeloablative patients were 44 and 61 months, respectively.

Lungs, liver, and heart were the organs most frequently affected by comorbidity in both patient cohorts (Figure 1). HCT-CI scores of 0, 1-2, and 3 or more were assigned to 30%, 30%, and 41% of nonmyeloablative patients, respectively, compared with 41%, 26%, and 32% of myeloablative patients (Figure 2; P = .01). The distributions of these comorbidity scores among nonmyeloablative (31%, 26%, and 43%) and myeloablative (46%, 27%, and 27%) patients remained comparable after exclusion of patients whose data were used to develop the HCT-CI.

Figure 1

Distribution of individual comorbidities among nonmyeloablative and myeloablative patients as assessed by the HCT-CI. Other comorbidities included rheumatologic, gastrointestinal, previous malignancy, and obesity. Of note, scores of 2 and 3 were assigned for moderate and severe pulmonary comorbidities, whereas scores of 1 and 3 were assigned for mild and moderate hepatic comorbidities.

Figure 1

Distribution of individual comorbidities among nonmyeloablative and myeloablative patients as assessed by the HCT-CI. Other comorbidities included rheumatologic, gastrointestinal, previous malignancy, and obesity. Of note, scores of 2 and 3 were assigned for moderate and severe pulmonary comorbidities, whereas scores of 1 and 3 were assigned for mild and moderate hepatic comorbidities.

Close modal
Figure 2

Distribution of scores as assigned by the HCT-CI to nonmyeloablative compared with myeloablative patients with lymphoma or CLL.

Figure 2

Distribution of scores as assigned by the HCT-CI to nonmyeloablative compared with myeloablative patients with lymphoma or CLL.

Close modal

Risk factors for HCT outcomes

The 3-year NRM and overall survival rates for all patients were 28% and 51%, respectively. These percentages were 25% and 53% for nonmyeloablative patients versus 35% and 45% for myeloablative patients. Cox regression multivariate models were constructed to define risk factors for NRM and overall mortality for all patients (Table 2). Conditioning type, HCT-CI scores, age, disease diagnosis and status, previous regimens, donor type, stem-cell source type, and cytomegalovirus (CMV) serology results were tested in these analyses as potential risk factors. The 2 most influential risk factors associated with increased HRs for both NRM and overall mortality were HCT-CI scores of 1 or more (P < .001 and .002) and myeloablative conditioning (P < .001 and .007), respectively. Less significant associations were found for age 50 or older for NRM (P = .006), diagnosis of NHL versus HD, versus CLL (P = .05) and previous CMV infection (P = .03) for overall mortality, and use of G-PBMC versus marrow for both NRM and overall mortality (P = .01 and .05, respectively). Effects of other factors did not reach statistical significance.

Table 2

Multivariate analyses of risk factors for NRM and mortality among all patients with lymphoma or CLL treated with allogeneic HCT

Risk factorsNRM
Mortality
HR* (95% CI)PHR* (95% CI)P
Conditioning  <.001  .007 
    Nonmyeloablative 1.0  1.0  
    Myeloablative 3.70 (2.0-10.0)  2.04 (1.25-3.33)  
HCT-CI scores  <.001  .002 
    0 1.0  1.0  
    1-2 2.48 (1.2-5.3)  1.71 (1.0-2.09)  
    ≥3 3.82 (1.9-7.8)  2.41 (1.5-4.0)  
Age  .006  .11 
    <50 years 1.0  1.0  
    ≥50 years 2.58 (1.3-5.1)  1.49 (0.9-2.4)  
Diagnosis  .19  .05 
    CLL 1.0  1.0  
    HD 1.11 (0.45-2.5)  1.16 (0.63-2.0)  
    NHL 1.89 (0.91-3.33)  1.89 (1.11-3.33)  
Previous regimens  .12  .11 
    0-2 1.0  1.0  
    3-4 1.15 (0.7-2.0)  1.38 (0.9-2.1)  
    ≥5 0.38 (0.1-1.3)  0.70 (0.3-1.6)  
Disease status  .83  .17 
    Remission 1.0  1.0  
    Refractory/relapse 1.06 (0.6-1.8)  1.32 (0.9-2.0)  
Donor  .12  .64 
    Matched siblings 1.0  1.0  
    Others 1.55 (0.9-2.7)  1.10 (0.7-1.6)  
Stem-cell source  .01  .05 
    Marrow 1.0  1.0  
    G-PBMC 3.12 (1.11-10.0)  2.0 (1.0-5.0)  
Patient CMV sero-status  .09  .03 
    Negative 1.0  1.0  
    Positive 1.45 (0.8-2.5)  1.43 (1.0-2.1)  
Risk factorsNRM
Mortality
HR* (95% CI)PHR* (95% CI)P
Conditioning  <.001  .007 
    Nonmyeloablative 1.0  1.0  
    Myeloablative 3.70 (2.0-10.0)  2.04 (1.25-3.33)  
HCT-CI scores  <.001  .002 
    0 1.0  1.0  
    1-2 2.48 (1.2-5.3)  1.71 (1.0-2.09)  
    ≥3 3.82 (1.9-7.8)  2.41 (1.5-4.0)  
Age  .006  .11 
    <50 years 1.0  1.0  
    ≥50 years 2.58 (1.3-5.1)  1.49 (0.9-2.4)  
Diagnosis  .19  .05 
    CLL 1.0  1.0  
    HD 1.11 (0.45-2.5)  1.16 (0.63-2.0)  
    NHL 1.89 (0.91-3.33)  1.89 (1.11-3.33)  
Previous regimens  .12  .11 
    0-2 1.0  1.0  
    3-4 1.15 (0.7-2.0)  1.38 (0.9-2.1)  
    ≥5 0.38 (0.1-1.3)  0.70 (0.3-1.6)  
Disease status  .83  .17 
    Remission 1.0  1.0  
    Refractory/relapse 1.06 (0.6-1.8)  1.32 (0.9-2.0)  
Donor  .12  .64 
    Matched siblings 1.0  1.0  
    Others 1.55 (0.9-2.7)  1.10 (0.7-1.6)  
Stem-cell source  .01  .05 
    Marrow 1.0  1.0  
    G-PBMC 3.12 (1.11-10.0)  2.0 (1.0-5.0)  
Patient CMV sero-status  .09  .03 
    Negative 1.0  1.0  
    Positive 1.45 (0.8-2.5)  1.43 (1.0-2.1)  

CI indicates confidence interval.

*

Higher HR indicates worse outcomes.

Includes antigen-mismatched related and all unrelated donors.

Risk stratification of HCT outcomes

Since HCT-CI scores and conditioning intensity were the two most influential risk factors, we stratified patients into 4 groups based on nonmyeloablative versus myeloablative conditioning and HCT-CI scores of 0 versus 1 or more.

Patients with HCT-CI score of 0

Nonmyeloablative and myeloablative patients with an HCT-CI score of 0 had 3-year NRM rates of 18% and 15% and overall survival rates of 68% and 60%, respectively. Differences in these outcomes were not statistically significant (the unadjusted HR for NRM and survival were 1.23 [P = .74] and .88 [P = .75], respectively). Likewise, there was no statistically significant difference in risk for relapse (HR: 1.41; P = .43). After adjustment for other risk factors, differences in NRM, relapse, and overall survival among nonmyeloablative and myeloablative patients remained statistically not significant (HR = 0.90, P = .91; HR = 1.94, P = .27; and HR = 0.94, P = .89, respectively). Figure 3A,B shows the observed NRM and overall survivals for nonmyeloablative and myeloablative patients, together with the hypothetical outcome for nonmyeloablative patients after adjustment for pretransplantation differences. Further, the HR for progression-free survival among nonmyeloablative and myeloablative patients with an HCT-CI score of 0 was not statistically significant (1.44; P = .40).

Figure 3

Cumulative incidence estimates of NRM and Kaplan–Meier survival estimates among nonmyeloablative compared with myeloablative patients with lymphoma or CLL and HCT-CI score of 0. No statistically significant differences were found between outcomes among nonmyeloablative and myeloablative patients. Further, there were no statistically significant differences between the observed outcomes among myeloablative patients and the hypothetical outcomes among nonmyeloablative patients after adjustment for pretransplantation variables including age, previous HCT, previous regimens, previous CMV infection, type and stage of malignancy, donor type, and stem-cell source. — indicates observed outcomes for myeloablative patients; , observed outcomes for nonmyeloablative patients; and …, adjusted outcomes for nonmyeloablative patients.

Figure 3

Cumulative incidence estimates of NRM and Kaplan–Meier survival estimates among nonmyeloablative compared with myeloablative patients with lymphoma or CLL and HCT-CI score of 0. No statistically significant differences were found between outcomes among nonmyeloablative and myeloablative patients. Further, there were no statistically significant differences between the observed outcomes among myeloablative patients and the hypothetical outcomes among nonmyeloablative patients after adjustment for pretransplantation variables including age, previous HCT, previous regimens, previous CMV infection, type and stage of malignancy, donor type, and stem-cell source. — indicates observed outcomes for myeloablative patients; , observed outcomes for nonmyeloablative patients; and …, adjusted outcomes for nonmyeloablative patients.

Close modal

We also investigated inpatient hospitalization days, outpatient follow-up days, and outpatient clinic visits among the 2 patient cohorts (Table 3). No differences were observed in the mean number of hospitalization (P = .53) and outpatient follow-up days (P = .06). Nonmyeloablative patients had a statistically significant lower median number of inpatient days (P = .004) and, conversely, a higher median number of total outpatient follow-up days (P < .001). Mean and median numbers of outpatient days associated with clinic visits (P = .02 and .03, respectively) were statistically significantly higher among nonmyeloablative patients. When we calculated the mean and median percentages of outpatient days associated with clinic visits among the total outpatient days for each patient, no statistically significant differences were found between the 2 cohorts (P = .46 and .41, respectively). Overall, 9 nonmyeloablative patients had inpatient hospital stays longer than 30 days because of severe acute GVHD (n = 5), infections (n = 3), and pathologic femur fracture (n = 1).

Table 3

Inpatient and outpatient (OP) days and actual clinic visits among nonmyeloablative compared with myeloablative patients with lymphoma or CLL and HCT-CI scores of 0

ParametersNonmyeloablative patients, mean days plus or minus SDMyeloablative patients, mean days plus or minus SDP*Nonmyeloablative patients, median no. days (range)Myeloablative patients, median no. days (range)P*
Inpatient hospitalization 19.0 ± 43.2 23.4 ± 15.7) .53 3.5 (0-232) 22 (0-61) .004 
OP follow-up 106 ± 46.6 73.6 ± 82.0 .06 97 (19-241) 64 (0-470) <.001 
OP actual clinic visits 42.8 ± 21.0 27.9 ± 13.3 .02 45 (7-94) 31 (2-50) .03 
Clinic visits per OP follow-up days 44.4 ± 16.5 38.9 ± 20.8 .46 43.9 (14.8-71.4) 39.5 (7.2-80.6) .41 
ParametersNonmyeloablative patients, mean days plus or minus SDMyeloablative patients, mean days plus or minus SDP*Nonmyeloablative patients, median no. days (range)Myeloablative patients, median no. days (range)P*
Inpatient hospitalization 19.0 ± 43.2 23.4 ± 15.7) .53 3.5 (0-232) 22 (0-61) .004 
OP follow-up 106 ± 46.6 73.6 ± 82.0 .06 97 (19-241) 64 (0-470) <.001 
OP actual clinic visits 42.8 ± 21.0 27.9 ± 13.3 .02 45 (7-94) 31 (2-50) .03 
Clinic visits per OP follow-up days 44.4 ± 16.5 38.9 ± 20.8 .46 43.9 (14.8-71.4) 39.5 (7.2-80.6) .41 
*

Student t test or Wilcoxon rank sum test.

Patients with HCT-CI scores of 1 or more

Among patients with HCT-CI scores of 1 or more, those receiving nonmyeloablative conditioning had a statistically significantly lower incidence of NRM (28% versus 50%; HR: .47, P = .009) and a better overall survival rate (47% versus 35%; HR: 0.63; P = .04) compared with myeloablative patients (Figure 4A,B). After adjustment for other pretransplantation risk factors, differences became more significant for NRM (HR: 0.19; P < .001) and overall survival (HR: 0.33; P = .007). Relapse risks were not statistically significantly different among the 2 cohorts before (HR: 1.54; P = .22) or after adjustment for other risk factors (HR: 1.96; P = .26). As a result, HR for progression-free survival was better among nonmyeloablative patients when we adjusted for other risk factors (unadjusted HR: 0.78, P = .26 and adjusted HR: 0.47, P = .01).

Figure 4

Cumulative incidence estimates of NRM and Kaplan–Meier survival estimates among nonmyeloablative compared with myeloablative patients with lymphoma or CLL and HCT-CI score of 1 or more. Nonmyeloablative patients had statistically significantly better outcomes compared with those of myeloablative patients. Differences between groups increased after adjustment for pretransplantation variables including age, previous HCT, previous regimens, previous CMV infection, type and stage of malignancy, donor type, and stem-cell source. — indicates observed outcomes for myeloablative patients; , observed outcomes for nonmyeloablative patients; and …, adjusted outcomes for nonmyeloablative patients.

Figure 4

Cumulative incidence estimates of NRM and Kaplan–Meier survival estimates among nonmyeloablative compared with myeloablative patients with lymphoma or CLL and HCT-CI score of 1 or more. Nonmyeloablative patients had statistically significantly better outcomes compared with those of myeloablative patients. Differences between groups increased after adjustment for pretransplantation variables including age, previous HCT, previous regimens, previous CMV infection, type and stage of malignancy, donor type, and stem-cell source. — indicates observed outcomes for myeloablative patients; , observed outcomes for nonmyeloablative patients; and …, adjusted outcomes for nonmyeloablative patients.

Close modal

Outcomes of patients with indolent versus aggressive diseases

We adjusted the previous comparisons for indolent versus aggressive diseases and found no change in the association between conditioning intensity and any of the outcomes. We also evaluated the interaction between indolent versus aggressive diseases and conditioning intensity. Patients with indolent diseases had higher NRM (HR: 3.16; P = .02) and higher mortality (HR: 2.02; P = .07) after myeloablative compared with nonmyeloablative conditioning. Similarly, patients with aggressive diseases had higher NRM (HR: 4.46; P = .007) and mortality (HR: 2.21; P = .01) after myeloablative conditioning. Conversely, relapse risks were lower after myeloablative conditioning among patients with indolent (HR: 0.56; P = .33) or aggressive diseases (HR: 0.51; P = .11), although these differences were not statistically significant. There were no statistically significant interactions between disease histology and conditioning intensity for NRM (P = .56), mortality (P = .84), or relapse (P = .88)

We also assessed the impact of grade of NHL on outcomes after myeloablative and nonmyeloablative HCT. Relapse risks were not statistically significantly different between the 2 cohorts among patients with indolent (HR: 1.97; P = .37) or aggressive NHL (HR: 0.72; P = .50). Conversely, statistically significant higher NRM was observed after myeloablative conditioning among patients with both indolent (HR: 3.35; P = .07) and aggressive NHL (HR: 6.95; P = .006). Consequently, patients with both indolent and aggressive NHL had statistically significant higher mortality after myeloablative than nonmyeloablative conditioning (HR: 3.34, P = .02 and HR: 3.02, P = .003), respectively.

Patients with CLL or lymphoma with poor prognostic features44  or resistance to salvage chemotherapy and patients with recurrent or progressive malignancy after autologous HCT1,2  have no potentially curative treatment except allogeneic HCT. Conventional allogeneic HCT with high-dose conditioning has been restricted to younger and medically fit patients out of concern for high NRM. This restriction has limited therapeutic options because patients with lymphoid malignancies have median ages of 65 to 70 years45  at diagnoses, and the prevalence of comorbidity often exceeds 50%.46  The advent of reduced-intensity conditioning regimens, which promised less NRM, has expanded the pool of patients with lymphoid malignancies offered allogeneic HCT. Efforts are needed to risk-stratify patient care and determine the optimal conditioning regimens for individual patients with lymphoid malignancies. Our results of this retrospective study suggest that increased conditioning dose intensity does not lower relapse rates but does result in higher NRM, particularly among patients with comorbidities.

The finding of comparable relapse rates is complicated by differences in pretransplantation characteristics, including more complete or partial remissions among nonmyeloablative patients compared with their myeloablative counterparts (46% vs 25%). However, relapse risks among the 2 cohorts were not statistically significantly different even after adjustment for differences in disease status and other variables. These findings persisted even after stratifying patients into indolent versus aggressive lymphoma/CLL. This suggests that graft-versus-leukemia effects are the most important in controlling disease relapse among patients with CLL or lymphoma. However, although relapse rates were comparable, patients with high comorbidity scores given high conditioning intensity had increased NRM and worse overall survival.

Patients with an HCT-CI score of 0 tolerated either conditioning regimen equally well and had comparable outcomes. Further, they did not differ with respect to median and mean percentages of clinic visits in relation to their total outpatient stays. Nonmyeloablative patients were generally older and more intensively pretreated than myeloablative patients. It was unknown whether there would be less frequent clinic visits and possible survival benefits if young, less frequently treated patients were given nonmyeloablative and not myeloablative conditioning. Within the limitations of this retrospective analysis, adjustment for pretransplantation differences did not demonstrate a statistically significant survival benefit for nonmyeloablative conditioning among patients with an HCT-CI score of 0. Results suggested that younger patients (< 60 years of age) with an HCT-CI score of 0 could be enrolled in prospective randomized trials designed to test which conditioning regimen would be appropriate.

In our series, myeloablative patients with CLL or lymphoma who had an HCT-CI score of 0 experienced relatively low NRM (15% at 3 years) compared with the overall historical experience, which showed NRM ranging from 40% to 61%.7,,,11  Conversely, a 3-year NRM of 50% was observed among myeloablative patients with HCT-CI scores of 1 or more, which was consistent with the reported experience after conventional HCT for lymphoid malignancies.7,,,11  The high NRM in these reports might have been partly attributable to the effects of unidentified comorbidities. Nonmyeloablative patients with similar HCT-CI scores had a 3-year NRM of 28%, which agreed with published reports by us13,,,17  and others.18,,,,,,,,,,29  The survival benefits for nonmyeloablative compared with myeloablative patients with HCT-CI scores of 1 or more gained additional statistical significance after adjustment for pretransplantation differences, in particular age, suggesting that young patients with comorbidities could further benefit from nonmyeloablative conditioning.

The HCT-CI was designed to reliably identify even minimal degrees of organ impairment.41  HCT-CI scores of 1 or more were assigned to 70% and 59% of nonmyeloablative and myeloablative patients, respectively, and this difference persisted even after excluding those patients who had contributed to the development of the index (69% vs 54%, respectively). More than half of myeloablative patients had comorbidities because subclinical organ impairments detected by the HCT-CI (eg, pulmonary function impairments in the range of 66%-80% of predicted values) were not considered as exclusion criteria when these patients were enrolled. Thus, future use of the HCT-CI might allow for more appropriate assignment of patients to different intensity conditioning regimens

In conclusion, patients without comorbidities (34% of our total population) tolerated nonmyeloablative and myeloablative conditioning regimens equally well, whereas those with comorbidities (whether young or old) had better survival after reduced-intensity conditioning. Our results from this retrospective study suggested that conditioning regimen intensity played a minor role in disease control while contributing to excessive NRM for patients with CLL or lymphoma who have comorbidities. For patients with no comorbidities and indolent or aggressive diseases, a randomized study might answer the question whether nonmyeloablative or myeloablative conditioning would be the most appropriate regimen.

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

The authors thank data coordinators Gary Schoch, Chris Davis, Jennifer Freese, and Heather Hildebrant and study nurses Mary Hinds, John Sedgwick, Michelle Bouvier, and Joanne Greene for their invaluable help in making the study possible. Bonnie Larson, Helen Crawford, Karen Carbonneau, and Sue Carbonneau provided assistance with manuscript preparation. We also thank all the transplantation teams of the Seattle Cancer Care Alliance.

This work was supported in part by grants CA78902, CA18029, CA15704, and HL088021 from the National Institutes of Health, Bethesda, MD, and in part by the Paros Family Fund.

National Institutes of Health

Contribution: M.L.S. designed the overall study, collected data, analyzed and interpreted the data, and wrote the manuscript. B.E.S. performed the statistical analysis. D.G.M. contributed study patients, contributed to data interpretation, and edited the manuscript. B.M.S. contributed study patients and contributed to study design. P.J.M. contributed to study design and edited the manuscript. R.S. contributed to study design and data interpretation and edited the manuscript.

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

Correspondence: Mohamed Sorror, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D1-100, PO Box 19024, Seattle, WA 98109-1024; e-mail: msorror@fhcrc.org.

1
Vose
JM
Bierman
PJ
Anderson
JR
et al
Progressive disease after high-dose therapy and autologous transplantation for lymphoid malignancy: clinical course and patient follow-up.
Blood
1992
80
2142
2148
2
Shamash
J
Lee
SM
Radford
JA
et al
Patterns of relapse and subsequent management following high-dose chemotherapy with autologous haematopoietic support in relapsed or refractory Hodgkin's lymphoma: a two centre study.
Ann Oncol
2000
11
715
719
3
Pavletic
ZS
Bierman
PJ
Vose
JM
et al
High incidence of relapse after autologous stem-cell transplantation for B-cell chronic lymphocytic leukemia or small lymphocytic lymphoma.
Ann Oncol
1998
9
1023
1026
4
Khouri
IF
Keating
MJ
Saliba
RM
et al
Long-term follow-up of patients with CLL treated with allogeneic hematopoietic transplantation.
Cytotherapy
2002
4
217
221
5
Anderson
JE
Litzow
MR
Appelbaum
FR
et al
Allogeneic, syngeneic, and autologous marrow transplantation for Hodgkin's disease: the 21 year Seattle experience.
J Clin Oncol
1993
11
2342
2350
6
Bierman
PJ
Sweetenham
JW
Loberiza
FRJ
et al
Syngeneic hematopoietic stem-cell transplantation for non-Hodgkin's lymphoma: a comparison with allogeneic and autologous transplantation–the Lymphoma Working Committee of the International Bone Marrow Transplant Registry and the European Group for Blood and Marrow Transplantation [see comment].
J Clin Oncol
2003
21
3744
3753
7
Michallet
M
Archimbaud
E
Bandini
G
et al
HLA-identical sibling bone marrow transplantation in younger patients with chronic lymphocytic leukemia. European Group for Blood and Marrow Transplantation and the International Bone Marrow Transplant Registry.
Ann Intern Med
1996
124
311
315
8
Milpied
N
Fielding
AK
Pearce
RM
et al
Allogeneic bone marrow transplant is not better than autologous transplant for patients with relapsed Hodgkin's disease.
J Clin Oncol
1996
14
1291
1296
9
Gajewski
JL
Phillips
GL
Sobocinski
KA
et al
Bone marrow transplants from HLA-identical siblings in advanced Hodgkin's disease.
J Clin Oncol
1996
14
572
578
10
Verdonck
LF
Dekker
AW
Lokhorst
HM
et al
Allogeneic versus autologous bone marrow transplantation for refractory and recurrent low-grade non-Hodgkin's lymphoma.
Blood
1997
90
4201
4205
11
Gribben
JG
Zahrieh
D
Stephans
K
et al
Autologous and allogeneic stem cell transplantations for poor-risk chronic lymphocytic leukemia.
Blood
2005
106
4389
4396
12
van Spronsen
DJ
Janssen-Heijnen
ML
Lemmens
VE
et al
Independent prognostic effect of co-morbidity in lymphoma patients: results of the population-based Eindhoven Cancer Registry.
Eur J Cancer
2005
41
1051
1057
13
Maris
MB
Sandmaier
BM
Storer
BE
et al
Allogeneic hematopoietic cell transplantation after fludarabine and 2 Gy total body irradiation for relapsed and refractory mantle cell lymphoma.
Blood
2004
104
3535
3542
14
Sorror
ML
Maris
MB
Sandmaier
BM
et al
Hematopoietic cell transplantation after nonmyeloablative conditioning for advanced chronic lymphocytic leukemia.
J Clin Oncol
2005
23
3819
3829
15
Rezvani
AR
Sandmaier
BM
Storer
B
et al
Outcomes of allogeneic hematopoietic stem cell transplantation (HCT) after non-myeloablative conditioning in relapsed, refractory, or transformed indolent non-Hodgkin lymphoma (NHL) [abstract].
Blood
2006
108
Part 1
891a
#3124
16
Norasetthada
L
Maris
MB
Sandmaier
BM
et al
HLA-matched related (MRD) or unrelated donor (URD) non-myeloablative hematopoietic cell transplantation (HCT) for patients (pts) with refractory and relapsed aggressive non-Hodgkin lymphoma [abstract].
Blood
2004
104
Part 1
634a
17
Burroughs
LM
Maris
MB
Sandmaier
BM
et al
HLA-matched related (MRD) or unrelated donor (URD) non-myeloablative conditioning and hematopoietic cell transplant (HCT) for patients with advanced Hodgkin disease (HD) [abstract].
Biol Blood Marrow Transplant
2004
10
Suppl. 1
73
18
Khouri
IF
Saliba
RM
Giralt
SA
et al
Nonablative allogeneic hematopoietic transplantation as adoptive immunotherapy for indolent lymphoma: low incidence of toxicity, acute graft-versus-host disease, and treatment-related mortality.
Blood
2001
98
3595
3599
19
Robinson
SP
Goldstone
AH
Mackinnon
S
et al
Chemoresistant or aggressive lymphoma predicts for a poor outcome following reduced-intensity allogeneic progenitor cell transplantation: an analysis from the Lymphoma Working Party of the European Group for Blood and Bone Marrow Transplantation.
Blood
2002
100
4310
4316
20
Morris
E
Thomson
K
Craddock
C
et al
Outcomes after alemtuzumab-containing reduced-intensity allogeneic transplantation regimen for relapsed and refractory non-Hodgkin lymphoma.
Blood
2004
104
3865
3871
21
Faulkner
RD
Craddock
C
Byrne
JL
et al
BEAM-alemtuzumab reduced-intensity allogeneic stem cell transplantation for lymphoproliferative diseases: GVHD, toxicity, and survival in 65 patients.
Blood
2004
103
428
434
22
Kusumi
E
Kami
M
Kanda
Y
et al
Reduced-intensity hematopoietic stem-cell transplantation for malignant lymphoma: a retrospective survey of 112 adult patients in Japan.
Bone Marrow Transplant
2005
36
205
213
23
Anderlini
P
Saliba
R
Acholonu
S
et al
Reduced-intensity allogeneic stem cell transplantation in relapsed and refractory Hodgkin's disease: low transplant-related mortality and impact of intensity of conditioning regimen.
Bone Marrow Transplant
2005
35
943
951
24
Peggs
KS
Hunter
A
Chopra
R
et al
Clinical evidence of a graft-versus-Hodgkin's-lymphoma effect after reduced-intensity allogeneic transplantation.
Lancet
2005
365
1934
1941
25
Alvarez
I
Sureda
A
Caballero
MD
et al
Nonmyeloablative stem cell transplantation is an effective therapy for refractory or relapsed Hodgkin lymphoma: results of a Spanish prospective cooperative protocol.
Biol Blood Marrow Transplant
2006
12
172
183
26
Dreger
P
Brand
R
Hansz
J
et al
Treatment-related mortality and graft-versus-leukemia activity after allogeneic stem cell transplantation for chronic lymphocytic leukemia using intensity-reduced conditioning (review).
Leukemia
2003
17
841
848
27
Schetelig
J
Thiede
C
Bornhauser
M
et al
Evidence of a graft-versus-leukemia effect in chronic lymphocytic leukemia after reduced-intensity conditioning and allogeneic stem-cell transplantation: the Cooperative German Transplant Study Group.
J Clin Oncol
2003
21
2747
2753
28
Khouri
IF
Lee
MS
Saliba
RM
et al
Nonablative allogeneic stem cell transplantation for chronic lymphocytic leukemia: impact of rituximab on immunomodulation and survival.
Exp Hematol
2004
32
28
35
29
Delgado
J
Thomson
K
Russell
N
et al
Results of alemtuzumab-based reduced-intensity allogeneic transplantation for chronic lymphocytic leukemia: a British Society of Blood and Marrow Transplantation Study.
Blood
2006
107
1724
1730
30
Maloney
DG
Molina
AJ
Sahebi
F
et al
Allografting with nonmyeloablative conditioning following cytoreductive autografts for the treatment of patients with multiple myeloma.
Blood
2003
102
3447
3454
31
McSweeney
PA
Niederwieser
D
Shizuru
JA
et al
Hematopoietic cell transplantation in older patients with hematologic malignancies: replacing high-dose cytotoxic therapy with graft-versus-tumor effects.
Blood
2001
97
3390
3400
32
Maris
MB
Niederwieser
D
Sandmaier
BM
et al
HLA-matched unrelated donor hematopoietic cell transplantation after nonmyeloablative conditioning for patients with hematologic malignancies.
Blood
2003
102
2021
2030
33
Clift
RA
Buckner
CD
Thomas
ED
et al
Marrow transplantation for chronic myeloid leukemia: a randomized study comparing cyclophosphamide and total body irradiation with busulfan and cyclophosphamide.
Blood
1994
84
2036
2043
34
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
1986
314
729
735
35
Petersdorf
EW
Gooley
TA
Anasetti
C
et al
Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient.
Blood
1998
92
3515
3520
36
Boeckh
M
Bowden
RA
Gooley
T
et al
Successful modification of a pp65 antigenemia-based early treatment strategy for prevention of cytomegalovirus disease in allogeneic marrow transplant recipients (Letter to Editor).
Blood
1999
93
1781
1782
37
Marr
KA
Seidel
K
Slavin
M
et al
Prolonged fluconazole prophylaxis is associated with persistent protection against candidiasis-related death in allogeneic marrow transplant recipients: long-term follow-up of a randomized, placebo-controlled trial.
Blood
2000
96
2055
2061
38
Boeckh
M
Marr
KA
Rubin
RH
Young
LS
Infection in hematopoietic stem cell transplantation.
Clinical approach to infection in the compromised host
2002
New York
Kluwer Academic/Plenum Publishers
527
571
39
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
1996
88
4063
4071
40
Kanda
Y
Mineishi
S
Saito
T
et al
Long-term low-dose acyclovir against varicella-zoster virus reactivation after allogeneic hematopoietic stem cell transplantation.
Bone Marrow Transplant
2001
28
689
692
41
Sorror
ML
Maris
MB
Storb
R
et al
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT.
Blood
2005
106
2912
2919
42
Kalbfleisch
JD
Prentice
RL
The statistical analysis of failure time data.
1980
New York
John Wiley & Sons
43
Makuch
RW
Adjusted survival curve estimation using covariates.
J Chronic Dis
1982
35
437
443
44
Zent
CS
Call
TG
Hogan
WJ
et al
Update on risk-stratified management for chronic lymphocytic leukemia (review).
Leuk Lymphoma
2006
47
1738
1746
45
Molina
AJ
Storb
RF
Rowe
JM
Lazarus
HM
Carella
AM
Hematopoietic stem cell transplantation in older adults.
Handbook of bone marrow transplantation
2000
London, UK
Martin Dunitz
111
137
46
van Spronsen
DJ
Janssen-Heijnen
ML
Breed
WP
et al
Prevalence of co-morbidity and its relationship to treatment among unselected patients with Hodgkin's disease and non-Hodgkin's lymphoma, 1993-1996.
Ann Hematol
1999
78
315
319
Sign in via your Institution