Clonal cytogenetic abnormalities are a major risk factor for relapse after hematopoietic cell transplantation (HCT) for myelodysplastic syndrome (MDS). We determined the impact of the recently established 5-group cytogenetic classification of MDS on outcome after HCT. Results were compared with the impact of the International Prognostic Scoring System (IPSS) 3 cytogenetic risk groups, and the additional effect of a monosomal karyotype was assessed. The study included data on 1007 patients, 1-75 years old (median 45 years), transplanted from related (n = 547) or unrelated (n = 460) donors. Various conditioning regimens were used, and marrow, peripheral blood, or cord blood served as stem cell source. Both IPSS and 5-group cytogenetic risk classifications were significantly associated with post-HCT relapse and mortality, but the 5-group classification discriminated more clearly among the lowest- and highest-risk patients. A monosomal karyotype tended to further increase the rates of relapse and mortality, even after considering the IPSS or 5-group classifications. In addition, the pathologic disease category correlated with both relapse and mortality. Mortality was also impacted by patient age, donor type, conditioning regimen, platelet count, and etiology of MDS. Although mortality declined significantly in recent years, novel strategies are needed to overcome the barrier of high-risk cytogenetics.

The prognosis of patients with myelodysplastic syndromes (MDSs) is dependent on disease and patient characteristics at the time of presentation.1  MDS tends to progress more rapidly in older than in younger patients, in those with greater comorbidities as determined by the ACE-27 instrument,1  and in patients with higher scores based on the International Prognostic Scoring Stem (IPSS)2  or the World Health Organization (WHO)–based prognostic scoring system.3,4  These factors also impact survival with various treatment modalities, including hematopoietic cell transplantation (HCT),5-7  currently the only therapy with proven curative potential.

A major cause of failure after HCT for MDS is disease relapse, profoundly affected by the clonal karyotype and the proportion of marrow myeloblasts at the time of HCT. Many recent studies have applied the 3 cytogenetic risk categories defined by the IPSS (good, intermediate, and poor2 ; Table 1) to the analysis of HCT results,6,7  although it has been suggested that categorization of cytogenetics into 2 risk groups (IPSS good/intermediate vs poor) is sufficient.8  However, a recent joint report analyzing the impact of cytogenetics on survival in nearly 3000 nontransplanted patients with MDS indicated that a distinction of 5 cytogenetic risk groups (very good, good, intermediate, poor, and very poor; Table 1) provided for more accurate prognostication than the 3 IPSS categories, identifying patients with median life expectancies that ranged from 61 months (very good group) to 6 months (very poor group).9 

Table 1

Cytogenetic classification according to the IPSS and the new 5-group classification2,10 

Classification/prognostic groupAbnormalities
SingleDoubleComplex
IPSS    
    Good Normal; −Y; — — 
 del(5q); del(20q)   
    Intermediate Other Any — 
    Poor 7* — ≥ 3 
5-group    
    Very good −Y; del(11q) — — 
    Good Normal; del(5q); del (20q); del(12p) Including del(5q) — 
    Intermediate del(7q); +8; i(17q); +19; any other Any other — 
    Poor −7; Inv(3)/t(3q)/del(3q) Including −7/ del(7q) 3 
    Very poor — — > 3 
Classification/prognostic groupAbnormalities
SingleDoubleComplex
IPSS    
    Good Normal; −Y; — — 
 del(5q); del(20q)   
    Intermediate Other Any — 
    Poor 7* — ≥ 3 
5-group    
    Very good −Y; del(11q) — — 
    Good Normal; del(5q); del (20q); del(12p) Including del(5q) — 
    Intermediate del(7q); +8; i(17q); +19; any other Any other — 
    Poor −7; Inv(3)/t(3q)/del(3q) Including −7/ del(7q) 3 
    Very poor — — > 3 

— indicates not applicable.

*

Any chromosome 7 abnormality.

Number of clonal abnormalities.

In the present analysis, we determined the impact of this 5-group classification along with that of a monosomal karyotype on transplant outcome in a large cohort of patients with MDS who had undergone HCT at our center.

Patients

Through June 2010, 1007 patients with MDS (including patients whose disease had transformed to acute myeloid leukemia [tAML]) underwent HCT at the Fred Hutchinson Cancer Research Center; 3 patients were transplanted before 1980, and 89% of HCT were carried out since 1990. Patient and disease characteristics are summarized in Table 2. All protocols were approved by the Fred Hutchinson Cancer Research Center Institutional Review Board.

Table 2

Patient and disease characteristics

VariableNo. (%) of patients
No. of patients 1007 
Patient age, y, range (median) 1-75 (45) 
Sex (male/female) 588 (58)/419 (42) 
De novo MDS 771 (77) 
Secondary MDS 236 (23) 
    Antecedent disorders* 228 (23) 
    Prior therapy 374 (37) 
FAB/WHO classification  
    RA 377 (37.4) 
    RCMD 82 (8.1) 
    MDS-U 25 (2.5) 
    RARS 15 (1.5) 
    RAEB§ 304 (30.2) 
        RAEB-1 46 (4.5) 
        RAEB-2 42 (4.2) 
    RAEB-T 91 (9.1) 
    tAML 113 (11.2) 
Cytogenetics  
    IPSS  
        Good 434 (43.1) 
        Intermediate 182 (18.1) 
        Poor 304 (30.2) 
    5-group  
        Very good 13 (1.3) 
            MK+/MK−/unknown 0/13/0 
        Good 440 (43.7) 
            MK+/MK−/unknown 0/440/0 
        Intermediate 175 (17.4) 
            MK+/MK−/unknown 7/167/1 
        Poor 148 (14.7) 
            MK+/MK−/unknown 60/88/0 
        Very poor 97 (9.6) 
            MK+/MK−/unknown 89/8/0 
        Data incomplete 134 (13.3) 
VariableNo. (%) of patients
No. of patients 1007 
Patient age, y, range (median) 1-75 (45) 
Sex (male/female) 588 (58)/419 (42) 
De novo MDS 771 (77) 
Secondary MDS 236 (23) 
    Antecedent disorders* 228 (23) 
    Prior therapy 374 (37) 
FAB/WHO classification  
    RA 377 (37.4) 
    RCMD 82 (8.1) 
    MDS-U 25 (2.5) 
    RARS 15 (1.5) 
    RAEB§ 304 (30.2) 
        RAEB-1 46 (4.5) 
        RAEB-2 42 (4.2) 
    RAEB-T 91 (9.1) 
    tAML 113 (11.2) 
Cytogenetics  
    IPSS  
        Good 434 (43.1) 
        Intermediate 182 (18.1) 
        Poor 304 (30.2) 
    5-group  
        Very good 13 (1.3) 
            MK+/MK−/unknown 0/13/0 
        Good 440 (43.7) 
            MK+/MK−/unknown 0/440/0 
        Intermediate 175 (17.4) 
            MK+/MK−/unknown 7/167/1 
        Poor 148 (14.7) 
            MK+/MK−/unknown 60/88/0 
        Very poor 97 (9.6) 
            MK+/MK−/unknown 89/8/0 
        Data incomplete 134 (13.3) 

MDS-U indicates myelodysplastic syndrome, unclassified.

*

Among these, 54 had aplastic anemia and 8 had Fanconi anemia or other constitutional marrow failure states; 20 had Crohn disease, juvenile rheumatoid arthritis, or other autoimmune disorders; 9 had myeloproliferative neoplasms that evolved to MDS, 56 had Hodgkin or non-Hodgkin lymphoma, and 5 had multiple myeloma. Seventeen had previously been treated for other lymphoid or myeloid leukemias and 57 for solid tumors; 4 had received solid organ transplants, 2 had been accidentally exposed to radiation, and in 4 patients the etiology could not be ascertained.

Unknown in 34.

WHO categories defined only in more recent patients; RA includes patients with RCMD transplanted in earlier years.

§

In 139 patients with RAEB (categorized by FAB), a breakdown into RAEB-1 and RAEB-2 was not possible.

Cytogenetic findings were categorized according to the IPSS (information complete in 918 patients) as well as the 5-group classification recently presented by Schanz et al10  (information complete in 871 patients). In addition, we categorized patients by monosomal and nonmonosomal karyotypes, as originally defined for patients with AML. Thus, patients who had 2 (or more) autosomal monosomies or one monosomy in combination with other structural abnormalities were considered as having a monosomal karyotype; marker chromosomes were not considered in this assessment11  (information complete in 905 patients). Cytogenetic classifications were applied both to patients with de novo and with secondary MDS. The reason for “incomplete data” was 2-fold: (1) in some instances, cultures to induce cell division for banding analysis were unsuccessful; and (2) others were categorized as intermediate or poor according to the IPSS without providing the specific clonal abnormalities.

Comorbidity scores as determined by the HCT comorbidity index (HCT-CI)12  were available in 440 patients (44%), representing the most recently transplanted subcohort.

Donors and sources of stem cells

Donors were related in 547 patients (54%), including 461 HLA-identical siblings (46%), 9 syngeneic donors (1%), and 77 (8%) HLA nonidentical family members, and unrelated in 460 patients (46%), including 21 patients (2%) who received cord blood as a source of stem cells. The level of HLA typing varied over the reporting period, ranging from serologic typing and mixed leukocyte cultures in the earlier transplant recipients to high resolution typing (DNA sequencing) in the more recently transplanted patients. Among the 439 patients with unrelated donors (noncord blood), 266 (61%) were determined (prospectively or retrospectively) to be matched by high-resolution typing at HLA-A, -B, -C, -DRB1, and -DQB1.13  There were 147 (33%) who were mismatched for at least one allele, and for 26 patients (6%) high resolution typing was not performed.

The source of hematopoietic stem cells was bone marrow in 502 patients (50%), G-CSF mobilized peripheral blood progenitor cells (PBPCs) in 480 patients (48%), a combination of marrow and PBPCs in 4 patients (< 0.5%) and cord blood in 21 patients (2%).

Complete information on CMV immune status in both patient and donor was available for 916 transplants (91%), whereas the information was incomplete for 91 (9%; Table 3).

Table 3

Donor and transplant characteristics

VariableNo. of patients (%)
Donors, related 547 (54) 
    HLA-matched sibling* 470 (47) 
    HLA-mismatched/nonsibling 77 (8) 
    Unrelated 460 (46) 
    HLA-matched§ 266 (26) 
    HLA mismatched 158 (16) 
Source of stem cells  
    Marrow 502 (50) 
    PBPC 484 (48) 
    Cord blood 21 (2) 
Donor/patient CMV status**  
    D+/P+ 280 (28) 
    D+/P− 112 (11) 
    D−/P− 272 (27) 
    D−/P+ 252 (25) 
Conditioning regimens  
    BU + CY (± ATG) 413 (41) 
    FLU + TBI (2–4.5 Gy) 101 (10) 
    BU + TBI (12 Gy) 72 (7) 
    FLU + BU 47 (5) 
    BU + CY + TBI (12 Gy) 44 (4) 
    131I-CD45 + FLU + TBI (2 Gy) 23 (2) 
    FLU + treosulfan 22 (2) 
    CY + TBI (≥ 12 Gy) ± ATG 216 (21) 
    Other 69 (7) 
    GVHD prophylaxis††  
        CSP + MTX 659 (65) 
        Tacrolimus + MTX 136 (14) 
        MMF + CSP/tacrolimus 101 (10) 
        CSP ± steroids 58 (6) 
        Other 44 (4) 
        None‡‡ 9 (1) 
VariableNo. of patients (%)
Donors, related 547 (54) 
    HLA-matched sibling* 470 (47) 
    HLA-mismatched/nonsibling 77 (8) 
    Unrelated 460 (46) 
    HLA-matched§ 266 (26) 
    HLA mismatched 158 (16) 
Source of stem cells  
    Marrow 502 (50) 
    PBPC 484 (48) 
    Cord blood 21 (2) 
Donor/patient CMV status**  
    D+/P+ 280 (28) 
    D+/P− 112 (11) 
    D−/P− 272 (27) 
    D−/P+ 252 (25) 
Conditioning regimens  
    BU + CY (± ATG) 413 (41) 
    FLU + TBI (2–4.5 Gy) 101 (10) 
    BU + TBI (12 Gy) 72 (7) 
    FLU + BU 47 (5) 
    BU + CY + TBI (12 Gy) 44 (4) 
    131I-CD45 + FLU + TBI (2 Gy) 23 (2) 
    FLU + treosulfan 22 (2) 
    CY + TBI (≥ 12 Gy) ± ATG 216 (21) 
    Other 69 (7) 
    GVHD prophylaxis††  
        CSP + MTX 659 (65) 
        Tacrolimus + MTX 136 (14) 
        MMF + CSP/tacrolimus 101 (10) 
        CSP ± steroids 58 (6) 
        Other 44 (4) 
        None‡‡ 9 (1) 

ATG indicates antithymocyte globulin; CSP, cyclosporine; MTX, methotrexate; and MMF, mycophenolate mofetil.

*

Including 9 syngeneic twins.

HLA mismatches typically involved one class I or class II antigen.

In 36 patients, high-resolution typing was not available.

§

HLA-matched at the allele level by high-resolution typing.

HLA-mismatched for one antigen, 1 or 2 alleles, or 1 antigen plus 1 allele.

Including 4 patients who receive PBPC + marrow.

**

Information incomplete in 91 cases.

††

In addition, 15 patients received ATG and 28 sirolimus in combination with various regimens.

‡‡

Patients were transplanted from syngeneic twin donors.

Transplant procedure

Patients were prepared for HCT with various conditioning regimens (Table 3), the spectrum reflecting the evolution of transplant regimens over the duration of the study. The analysis of the impact of regimens was based on treatment components and dose intensities, rather than using classification into 2 or 3 subgroups as proposed elsewhere.14 

Statistical analysis

Stepwise Cox regression models were fit to assess the association of various factors with the time-to-event outcomes relapse, overall mortality, and the earliest of relapse or mortality (failure of relapse-free survival) with entry and exit P values of .05. For variables with more than 2 categories, a global test was conducted using the score test. Kaplan-Meier estimates were used to determine the probability of overall and relapse-free survival for listed categories, and cumulative incidence estimates were used to summarize the probability of relapse and nonrelapse mortality (NRM), where NRM was regarded as a competing risk for relapse, and relapse as a competing risk for NRM.15  The analysis included patients transplanted as of December 31, 2010.

Changes in cohort composition over time

Patient age increased from a median of 31.7 years before 1990 to a median of 54.3 years in the interval 2006-2010. Concurrently, the proportion of unrelated donor HCT increased from 6% to 57%, and the proportion of patients who received PBPCs rather than marrow increased from 0% before 1990 to 84% from 2006-2010.

The proportion of patients with refractory anemia (RA) decreased from 35% before 1990 to 14% in the period from 2006-2010 with a concurrent increase in more advanced cases of MDS.

Cytogenetic risk groups

The 5-group cytogenetic risks “good” and “intermediate” largely coincided with the IPSS cytogenetic risk groups “good” (99%) and “intermediate” (79%), respectively (Table 1). The IPSS group, “poor,“ was split into the categories “intermediate” (12%), “poor” (47%), and “very poor” (32%) in the novel 5-group classification (in the remaining 9% of patients classified as IPSS risk “poor,” details were insufficient to subcategorize them in the 5-group classification).

Within the 5 risk groups, secondary MDS accounted for 0 of 13, 69 of 440 (16%), 46 of 175 (26%), 46 of 148 (31%), and 28 of 97 (29%) in the very good, good, intermediate, poor, and very poor groups, respectively. Cytogenetic information for 5-group classification was incomplete in 134 patients, 47 (35%) of whom had secondary MDS (Table 4).

Table 4

IPSS categories and distribution by 5-group risk classification

IPSS category5-group classification
Very goodGoodIntermediatePoorVery poorData incomplete
Good 431 
Intermediate 10 139 19 
Poor 36 143 97 28 
Data incomplete 87 
IPSS category5-group classification
Very goodGoodIntermediatePoorVery poorData incomplete
Good 431 
Intermediate 10 139 19 
Poor 36 143 97 28 
Data incomplete 87 

Considering only patients with complete cytogenetic information, there were 167 patients with monosomal karyotypes, 59 (30%) of 200 with secondary MDS, and 108 (15%) of 705 with de novo MDS (P < .0001). All but 4 monosomal karyotypes were within the IPSS poor-risk cytogenetic group, and all but 7 within the poor-risk and very poor-risk groups in the 5-group classification. The proportion of patients with monosomal karyotype was stable across time, ranging from 19% before 1990 to 20% after 2006.

Disease stage and pre-HCT therapy

There were 371 patients with RA (by French-American-British [FAB] criteria), 15 with RA and ring sideroblasts (RARS), 82 with refractory cytopenia with multilineage dysplasia (RCMD; including 9 with RS), 302 with RA with excess blasts (RAEB, including 46 who were classified by WHO as RAEB-1 and 41 classified as RAEB-2), 89 with RAEB in transformation (RAEB-T), and 107 in whom the disease had transformed to tAML. Thus, by WHO criteria, 196 patients had AML. In 41 patients, MDS could not be further classified.

For 973 patients, the extent of prior therapy (other than transfusion support, hematopoietic growth factors, or antibiotics) could be ascertained. Among these, 599 had received no therapy, 329 had been given induction-type chemotherapy, 41 hypomethylating drugs, and 4 both. In 34 patients, prior therapy could not be verified.

Secondary MDS

MDS was considered to be secondary in 236 patients, the majority of whom had antecedent hematologic disorders or lymphoid neoplasms. Details are provided in Table 2. Among 236 patients with secondary MDS, 65 (28%) advanced to tAML, similar to 214 (28%) of 771 patients with de novo MDS.

Transplant outcome

Overall, 650 patients had died by the time of last contact. The follow-up among the 357 surviving patients was 0.5 to 27.5 years (median, 7.1 years). There were 254 patients in whom the disease progressed or relapsed, and 222 (87%) of these had died at the time of last contact; 428 patients died from nonrelapse causes. The 5-year estimates of relapse, NRM, overall survival, and relapse-free survival were 25%, 40%, 38%, and 35%, respectively. Grades 2-4 acute GVHD occurred in 663 of 964 evaluable patients (69%), and 248 patients (26%) had grades 3-4 disease. Chronic GVHD occurred in 406 patients (40%).

Univariate analysis

Relapse.

As summarized in Table 5, disease stage/risk category (by FAB or WHO), marrow myeloblast count, cytogenetic risk group, patient age, year of HCT, presence of monosomal karyotype, conditioning, etiology, and pre-HCT treatment were each associated with the rate of relapse. The cumulative incidence of relapse by karyotype as determined by IPSS and 5-group criteria is shown in Figure 1A and B. The relapse rate was nonsignificantly increased in patients with secondary MDS compared with patients with de novo MDS (hazard rate [HR] = 1.27). However, as shown in Table 5, the relapse rate was dependent on the antecedent disorders and was significantly lower than with de novo MDS only in patients with prior aplastic anemia (HR = 0.26). When year of HCT was modeled as a continuous linear variable, there was an increase in relapse as the transplant year increased (P = .03). However, the relapse rate from 1996 onward was quite stable (compared with 1996-2000, the HRs for the years 2001-2005 and 2006-2010 were 1.03 and 1.04, respectively).

Table 5

Univariate regression model for relapse

VariableGroupHR95% CIPGlobal P
Marrow myeloblasts*  1.02 1.01-1.03 < .0001  
Hemoglobin*  0.96 0.89-1.04 .31  
Platelet count*  0.98 0.95-1.03 .50  
Patient age*  1.10 1.02-1.19 .01  
Disease category MDS    
 tAML 2.51 1.96-3.22  < .0001 
IPSS cytogenetic risk Good  < .0001  
 Intermediate 1.10 0.75-1.62 .62  
 Poor 2.76 2.08-3.66 < .0001  
5-group cytogenetic risk Good   < .0001 
 Very good 0.82 0.20-3.34 .78  
 Intermediate 1.23 0.84-1.80 .28  
 Poor 1.74 1.21-2.51 .003  
 Very poor 6.68 4.67-9.56 < .0001  
 Data incomplete 1.72 1.15-2.55 .008  
Donor type Matched sibling   .15 
 Nonsibling relative 0.69 0.39-1.22 .20  
 URD matched 0.76 0.56-1.02 .07  
 URD mismatched 0.83 0.58-1.21 .34  
 URD unknown 0.46 0.19-1.11 .09  
Year of HCT*  1.02 1.00-1.04 .03  
Monosomal karyotype No    
 Yes 3.70 2.78-4.92 < .0001  
Secondary MDS No    
 Yes 1.27 0.95-1.68 .10  
Conditioning regimen BU + CY   .001 
 FLU + LD TBI 2.38 1.64-3.45 < .0001  
 FLU + BU 2.03 1.21-3.41 .008  
 131I + FLU + TBI 2.34 1.18-4.64 .02  
 FLU + TREO 0.39 0.10-1.59 .19  
 CY + TBI (≥ 12 Gy) (± ATG) 1.56 1.12-2.18 .009  
FAB/WHO classification RA/RARS/RCMD   < .0001 
 RAEB. 2.09 1.56-2.79 < .0001  
 RAEB-T 2.18 1.41-3.37 .0005  
 tAML 2.70 1.82-3.99 < .0001  
Etiology de novo   .002 
 Aplastic anemia 0.26 0.10-0.70 .008  
 Lymphoma 1.70 1.09-2.67 .02  
 Solid tumor 1.92 1.22-3.01 .005  
 Other leukemias 2.57 1.31-5.01 .006  
Treatment pre-HCT None   < .0001 
 IC 2.09 1.61-2.71 < .0001  
 IC + hypomethylation 0.00 0.00-**** .97  
 Hypomethylation 2.53 1.51-4.23 .0004  
VariableGroupHR95% CIPGlobal P
Marrow myeloblasts*  1.02 1.01-1.03 < .0001  
Hemoglobin*  0.96 0.89-1.04 .31  
Platelet count*  0.98 0.95-1.03 .50  
Patient age*  1.10 1.02-1.19 .01  
Disease category MDS    
 tAML 2.51 1.96-3.22  < .0001 
IPSS cytogenetic risk Good  < .0001  
 Intermediate 1.10 0.75-1.62 .62  
 Poor 2.76 2.08-3.66 < .0001  
5-group cytogenetic risk Good   < .0001 
 Very good 0.82 0.20-3.34 .78  
 Intermediate 1.23 0.84-1.80 .28  
 Poor 1.74 1.21-2.51 .003  
 Very poor 6.68 4.67-9.56 < .0001  
 Data incomplete 1.72 1.15-2.55 .008  
Donor type Matched sibling   .15 
 Nonsibling relative 0.69 0.39-1.22 .20  
 URD matched 0.76 0.56-1.02 .07  
 URD mismatched 0.83 0.58-1.21 .34  
 URD unknown 0.46 0.19-1.11 .09  
Year of HCT*  1.02 1.00-1.04 .03  
Monosomal karyotype No    
 Yes 3.70 2.78-4.92 < .0001  
Secondary MDS No    
 Yes 1.27 0.95-1.68 .10  
Conditioning regimen BU + CY   .001 
 FLU + LD TBI 2.38 1.64-3.45 < .0001  
 FLU + BU 2.03 1.21-3.41 .008  
 131I + FLU + TBI 2.34 1.18-4.64 .02  
 FLU + TREO 0.39 0.10-1.59 .19  
 CY + TBI (≥ 12 Gy) (± ATG) 1.56 1.12-2.18 .009  
FAB/WHO classification RA/RARS/RCMD   < .0001 
 RAEB. 2.09 1.56-2.79 < .0001  
 RAEB-T 2.18 1.41-3.37 .0005  
 tAML 2.70 1.82-3.99 < .0001  
Etiology de novo   .002 
 Aplastic anemia 0.26 0.10-0.70 .008  
 Lymphoma 1.70 1.09-2.67 .02  
 Solid tumor 1.92 1.22-3.01 .005  
 Other leukemias 2.57 1.31-5.01 .006  
Treatment pre-HCT None   < .0001 
 IC 2.09 1.61-2.71 < .0001  
 IC + hypomethylation 0.00 0.00-**** .97  
 Hypomethylation 2.53 1.51-4.23 .0004  

CMV status and source of stem cells had no significant impact on outcome.

URD indicates unrelated donor; and ATG, antithymocyte globulin.

*

Modeled as continuous linear variables; HR for platelet count represents increase in hazard associated with increase in platelets by 50 000; HR for age represents increase in hazard associated with increase in age by 10 years; HR for year of HCT represents increase in hazard associated with increase in year of HCT by one year; HR for hemoglobin represents increase in hazard associated with increase in hemoglobin by 1 g/dL; HR for blasts represents increase in hazard associated with increase in blasts by 1%.

Other regimens used (CY + TMI; FLU + TBI + CY; CY + BU; BU + TBI; BU + CY + TBI; other regimens) did not result in a significant differences.

Other antecedent conditions (myeloproliferative neoplasms; constitutional marrow failure syndromes; multiple myeloma; autoimmune disoders; prior solid organ transplantation or accidental radiation exposure) had no significant impact.

Figure 1

Impact of karyotype on posttransplant relapse. (A) Cumulative incidence (CI) of relapse by 5-group cytogenetic classification. (B) CI of relapse by IPSS cytogenetic classification. (C) CI of relapse among patients in the IPSS group “poor,” reclassified by 5-group criteria.

Figure 1

Impact of karyotype on posttransplant relapse. (A) Cumulative incidence (CI) of relapse by 5-group cytogenetic classification. (B) CI of relapse by IPSS cytogenetic classification. (C) CI of relapse among patients in the IPSS group “poor,” reclassified by 5-group criteria.

Close modal

There was a trend to lower rates of relapse with HLA-matched unrelated compared with HLA-matched sibling donors (HR = 0.76). As summarized in Table 5, the relapse rate varied dependent on the type of conditioning regimen; it was highest among patients conditioned with fludarabine (FLU) plus low-dose (2-4.5 Gy) total body irradiation (TBI; HR = 2.38) and in patients conditioned with 131I-CD45 radioimmunotherapy (HR = 2.34) in a phase 1 or 2 study enrolling patients with refractory disease, not eligible for other protocols. The lowest rate of relapse was observed in a cohort of patients conditioned with a treosulfan plus FLU combination.

The incidence of post-HCT relapse was higher among patients given induction chemotherapy (HR = 2.09) or hypomethylating agents (HR = 2.53) before HCT than in those who were untreated.

Mortality.

Overall mortality was impacted by the same factors that were associated with relapse, and, in addition, by platelet count, type of donor, and the presence of secondary MDS (Table 6). The probability of survival by karyotype is shown in Figure 2. The rate of mortality increased as patient age increased (P = .005, modeling age as a continuous linear variable). If patient age was categorized as 0-50, 50-60, 60-65, 65-70, and > 70 years, the rate of mortality increased with each category (relative to ≤ 50 years, HR = of 1.09, 1.14, 1.24, and 1.62, respectively).

Table 6

Univariate regression model for overall mortality

VariableGroupHR95% CIPGlobal P
Marrow myeloblasts*  1.02 1.01-1.02 < .0001  
Hemoglobin*  0.95 0.90-1.01 .08  
Platelet count*  0.93 0.90-0.97 .0005  
Patient age*  1.07 1.02-1.12 .005  
Disease category MDS    
 tAML 1.57 1.34-1.86 < .0001  
IPSS cytogenetic risk Good   < .0001 
 Intermediate 1.16 0.93-1.45 .19  
 Poor 1.77 1.47-2.12 < .0001  
5-group cytogenetic risk Good   < .0001 
 Very good 1.15 0.57-2.32 .71  
 Intermediate 1.13 0.90-1.43 .28  
 Poor 1.29 1.01-1.63 .04  
 Very poor 3.08 2.40-3.94 < .0001  
 Data incomplete 1.88 1.50-2.36 < .0001  
Donor type Matched sibling   .01 
 Nonsibling relative 1.40 1.06-1.85 .02  
 URD matched 0.88 0.72-1.07 .19  
 URD mismatched 1.22 0.98-1.52 .07  
 URD unknown§ 0.89 0.57-1.40 .61  
Year of HCT*  0.97 0.96-0.99 < .0001  
Monosomal karyotype No    
 Yes 2.44 2.01-2.96 < .0001  
Secondary MDS No    
 Yes 1.33 1.11-1.58 .002  
Conditioning regimen BU + CY (± ATG)   < .0001 
 FLU + LD TBI 1.49 1.14-1.96 .004  
 BU + TBI (12 Gy) 2.21 1.65-2.95 < .0001  
 FLU + BU 1.46 1.01-2.10 .04  
 BU + CY + TBI (12 Gy) 1.86 1.30-2.67 .0007  
 131I + FLU + TBI (2 Gy) 1.86 1.15-3.00 .01  
 FLU + TREO 0.43 0.18-1.04 .06  
 CY + TBI (≥ 12 Gy) (± ATG) 1.66 1.35-2.03 < .0001  
 Other 1.69 1.11-2.57 .01  
FAB/WHO classification RA/RARS/RCMD   < .0001 
 RAEB 1.63 1.36-1.96 < .0001  
 RAEB-T 2.02 1.56-2.63 < .0001  
 tAML 2.37 1.86-3.02 < .0001  
Etiology de novo  < .0001  
 MPN 2.36 1.17-4.75 .03  
 Lymphoma 1.50 1.11-2.02 .008  
 Multiple myeloma 3.12 1.29-7.55 .01  
 Autoimmune disorders 1.76 1.08-2.85 .02  
 Organ transplant 2.64 0.85-8.22 .09  
 Accidental radiation exposure 11.4 2.83-46.1 .0006  
Treatment pre-HCT No   .01 
 IC 1.27 1.08-1.50 .004  
 IC + hypomethylation 0.51 0.07-3.63 .50  
 Hypomethylation 0.84 0.53-1.34 .47  
VariableGroupHR95% CIPGlobal P
Marrow myeloblasts*  1.02 1.01-1.02 < .0001  
Hemoglobin*  0.95 0.90-1.01 .08  
Platelet count*  0.93 0.90-0.97 .0005  
Patient age*  1.07 1.02-1.12 .005  
Disease category MDS    
 tAML 1.57 1.34-1.86 < .0001  
IPSS cytogenetic risk Good   < .0001 
 Intermediate 1.16 0.93-1.45 .19  
 Poor 1.77 1.47-2.12 < .0001  
5-group cytogenetic risk Good   < .0001 
 Very good 1.15 0.57-2.32 .71  
 Intermediate 1.13 0.90-1.43 .28  
 Poor 1.29 1.01-1.63 .04  
 Very poor 3.08 2.40-3.94 < .0001  
 Data incomplete 1.88 1.50-2.36 < .0001  
Donor type Matched sibling   .01 
 Nonsibling relative 1.40 1.06-1.85 .02  
 URD matched 0.88 0.72-1.07 .19  
 URD mismatched 1.22 0.98-1.52 .07  
 URD unknown§ 0.89 0.57-1.40 .61  
Year of HCT*  0.97 0.96-0.99 < .0001  
Monosomal karyotype No    
 Yes 2.44 2.01-2.96 < .0001  
Secondary MDS No    
 Yes 1.33 1.11-1.58 .002  
Conditioning regimen BU + CY (± ATG)   < .0001 
 FLU + LD TBI 1.49 1.14-1.96 .004  
 BU + TBI (12 Gy) 2.21 1.65-2.95 < .0001  
 FLU + BU 1.46 1.01-2.10 .04  
 BU + CY + TBI (12 Gy) 1.86 1.30-2.67 .0007  
 131I + FLU + TBI (2 Gy) 1.86 1.15-3.00 .01  
 FLU + TREO 0.43 0.18-1.04 .06  
 CY + TBI (≥ 12 Gy) (± ATG) 1.66 1.35-2.03 < .0001  
 Other 1.69 1.11-2.57 .01  
FAB/WHO classification RA/RARS/RCMD   < .0001 
 RAEB 1.63 1.36-1.96 < .0001  
 RAEB-T 2.02 1.56-2.63 < .0001  
 tAML 2.37 1.86-3.02 < .0001  
Etiology de novo  < .0001  
 MPN 2.36 1.17-4.75 .03  
 Lymphoma 1.50 1.11-2.02 .008  
 Multiple myeloma 3.12 1.29-7.55 .01  
 Autoimmune disorders 1.76 1.08-2.85 .02  
 Organ transplant 2.64 0.85-8.22 .09  
 Accidental radiation exposure 11.4 2.83-46.1 .0006  
Treatment pre-HCT No   .01 
 IC 1.27 1.08-1.50 .004  
 IC + hypomethylation 0.51 0.07-3.63 .50  
 Hypomethylation 0.84 0.53-1.34 .47  

CMV status and source of stem cells had no significant impact on outcome.

URD indicates unrelated donor; ATG, antithymocyte globulin; LD, low-dose TBI (2-4.5 Gy); TREO, treosulfan; MPN, myeloproliferative neoplasm; and IC, induction chemotherapy.

*

Modeled as continuous linear variables (as specified in Table 5);

Other regimens used (CY + TMI; FLU + TBI + CY; CY + BU) did not result in a significant difference.

Other antecedent conditions (aplastic anemia; constitutional marrow failure syndromes; solid tumors; other leukemias) had no significant impact.

§

Allele level typing was not performed.

Figure 2

Survival by 5-group cytogenetic classification.

Figure 2

Survival by 5-group cytogenetic classification.

Close modal

Overall mortality was higher in patients with secondary compared with patients with de novo MDS (HR = 1.33) and was dependent on the type of antecedent disorder (Table 6). Mortality was also higher in patients who had received induction type chemotherapy before HCT (HR = 1.27) compared with patients without chemotherapy. The pre-HCT use of hypomethylating agents had no significant effect on mortality.

Compared with patients conditioned with busulfan/cyclophosphamide (BU/CY), mortality was increased to various degrees, particularly in patients conditioned with regimens, including high-dose TBI. Mortality was nonsignificantly reduced in patients conditioned with a treosulfan plus FLU regimen (HR = 0.43) compared with BU/CY (Figure 3).

Figure 3

Survival by conditioning regimen.

Figure 3

Survival by conditioning regimen.

Close modal

Relapse-free survival.

Because most patients (87%) who experienced post-HCT disease progression or relapse subsequently died, the associations of all variables with relapse or mortality were very similar to the associations with mortality (supplemental Table 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Multivariable analysis

Relapse.

In multivariable analysis, the 5-group cytogenetic risk categories, FAB/WHO classification, conditioning regimen, and progression of MDS to tAML were significant risk factors for relapse (Table 7). Patients with very poor cytogenetics had a much higher rate of relapse than patients with good-risk cytogenetics (HR = 5.26). Patients with RAEB-T were significantly more likely to relapse than were RA patients (HR = 2.14). Patients who received FLU plus low-dose TBI had a higher relapse rate than patients conditioned with BU/CY (HR = 2.01). Patients in whom MDS transformed to tAML had a higher rate of relapse compared with those who did not progress (HR = 2.39).

Table 7

Multivariable regression model for relapse

VariableGroupHR95% CIPGlobal P
5-group cytogenetic risk Good   < .0001 
 Very good 0.75 0.18-3.09 .70  
 Intermediate 1.19 0.81-1.76 .38  
 Poor 1.82 1.24-2.67 .002  
 Very poor 5.26 3.59-7.71 < .0001  
 Data incomplete 1.60 1.03-2.47 .04  
FAB/WHO classification RA/RARS/RCMD   .001 
 RAEB 1.81 1.33-2.47 .0002  
 RAEB-T 2.14 1.35-3.40 .001  
 tAML 1.29 0.81-2.05 .28  
Disease category MDS    
 tAML 2.39 1.77-3.23 < .0001  
Conditioning regimen* BU + CY   .02 
 FLU + LD TBI 2.01 1.36-2.96 .0004  
 CY + TMI 2.62 0.94-7.28 .06  
 FLU + TREO 0.34 0.08-1.37 .13  
VariableGroupHR95% CIPGlobal P
5-group cytogenetic risk Good   < .0001 
 Very good 0.75 0.18-3.09 .70  
 Intermediate 1.19 0.81-1.76 .38  
 Poor 1.82 1.24-2.67 .002  
 Very poor 5.26 3.59-7.71 < .0001  
 Data incomplete 1.60 1.03-2.47 .04  
FAB/WHO classification RA/RARS/RCMD   .001 
 RAEB 1.81 1.33-2.47 .0002  
 RAEB-T 2.14 1.35-3.40 .001  
 tAML 1.29 0.81-2.05 .28  
Disease category MDS    
 tAML 2.39 1.77-3.23 < .0001  
Conditioning regimen* BU + CY   .02 
 FLU + LD TBI 2.01 1.36-2.96 .0004  
 CY + TMI 2.62 0.94-7.28 .06  
 FLU + TREO 0.34 0.08-1.37 .13  

LD indicates low dose; TMI, total marrow irradiation (with lung and liver shielding); and TREO, treosulfan.

*

Other regimens (CY + TMI; CY + BU; FLU + TBI + CY; FLU + BU; BU + TBI; BU + CY + TBI;131I + FLU + TBI; FLU + LD TBI + CY; CY + BU; CY + TBI [≥ 12 Gy]) had no significant impact.

Mortality.

Mortality was similarly impacted by each of the factors significant for relapse and, in addition, by etiology, platelet count, patient age, type of donor, and year of HCT (Table 8). Patients with very poor cytogenetics were 2.68 times more likely to die than patients with good-risk cytogenetics. Mortality also increased as patient age increased (P < .0001). The rates of mortality for patients with RAEB or RAEB-T were approximately 1.4 times as high as the rate among patients with RA. The HR was similar (1.41) for patients who transformed to tAML (including myeloblast counts ≥ 30%). Among patients with secondary MDS, the rate of mortality was dependent on the type of antecedent disease (Table 8). Patients transplanted from unrelated donors who were HLA mismatched by high-resolution typing had a higher mortality rate than patients transplanted from HLA-matched siblings (HR = 1.58). A delay of HCT resulting from a prolonged donor search might have been a contributing factor.16 

Table 8

Multivariable regression model for overall mortality

VariableGroupHR95% CIPGlobal P
5-group cytogenetic risk Good   < .0001 
 Very good 1.15 0.54-2.47 .72  
 Intermediate 1.04 0.82-1.32 .75  
 Poor 1.18 0.91-1.52 .21  
 Very poor 2.68 2.05-3.50 < .0001  
 Data incomplete 1.74 1.34-2.28 < .0001  
FAB/WHO classification RA/RARS/RCMD   .008 
 tAML 1.44 1.04-2.00 .03  
 RAEB 1.42 1.15-1.74 .001  
 RAEB-T 1.45 1.08-1.94 .01  
Disease category MDS    
 tAML 1.41 1.13-1.76 .003  
Platelet count*  0.92 0.88-0.96 .0002  
Patient age*  1.18 1.10-1.26 < .0001  
Year of HCT*  0.96 0.94-0.98 .0002  
Donor type Matched sibling   .003 
 Nonsibling relative 1.35 0.99-1.85 .06  
 URD matched 1.19 0.95-1.48 .14  
 URD mismatched 1.58 1.24-2.01 .0002  
 URD unknown 0.98 0.60-1.61 .93  
Conditioning regimen BU + CY   .003 
 FLU + LD TBI 1.56 1.14-2.15 .006  
 BU + TBI 1.81 1.31-2.51 .0003  
 BU + CY + TBI 1.53 1.01-2.34 .05  
 131I + FLU + TBI 1.77 1.07-2.93 .03  
 FLU + TREO 0.55 0.22-1.36 .20  
 CY + TBI (≥ 12 Gy) (± ATG) 1.29 0.97-1.73 .08  
 Other 1.90 1.09-3.30 .02  
Etiology§ de novo   < .0001 
 Aplastic anemia 1.45 0.97-2.17 .07  
 MPN 2.19 1.04-4.60 .04  
 Multiple myeloma 9.00 3.24-25.0 < .0001  
 Solid tumor 1.67 1.16-2.40 .006  
 Autoimmune disorders 1.60 0.97-2.62 .06  
 Organ transplantation 23.1 6.53-81.8 < .0001  
 Accidental radiation exposure 9.66 1.29-72.2 .03  
VariableGroupHR95% CIPGlobal P
5-group cytogenetic risk Good   < .0001 
 Very good 1.15 0.54-2.47 .72  
 Intermediate 1.04 0.82-1.32 .75  
 Poor 1.18 0.91-1.52 .21  
 Very poor 2.68 2.05-3.50 < .0001  
 Data incomplete 1.74 1.34-2.28 < .0001  
FAB/WHO classification RA/RARS/RCMD   .008 
 tAML 1.44 1.04-2.00 .03  
 RAEB 1.42 1.15-1.74 .001  
 RAEB-T 1.45 1.08-1.94 .01  
Disease category MDS    
 tAML 1.41 1.13-1.76 .003  
Platelet count*  0.92 0.88-0.96 .0002  
Patient age*  1.18 1.10-1.26 < .0001  
Year of HCT*  0.96 0.94-0.98 .0002  
Donor type Matched sibling   .003 
 Nonsibling relative 1.35 0.99-1.85 .06  
 URD matched 1.19 0.95-1.48 .14  
 URD mismatched 1.58 1.24-2.01 .0002  
 URD unknown 0.98 0.60-1.61 .93  
Conditioning regimen BU + CY   .003 
 FLU + LD TBI 1.56 1.14-2.15 .006  
 BU + TBI 1.81 1.31-2.51 .0003  
 BU + CY + TBI 1.53 1.01-2.34 .05  
 131I + FLU + TBI 1.77 1.07-2.93 .03  
 FLU + TREO 0.55 0.22-1.36 .20  
 CY + TBI (≥ 12 Gy) (± ATG) 1.29 0.97-1.73 .08  
 Other 1.90 1.09-3.30 .02  
Etiology§ de novo   < .0001 
 Aplastic anemia 1.45 0.97-2.17 .07  
 MPN 2.19 1.04-4.60 .04  
 Multiple myeloma 9.00 3.24-25.0 < .0001  
 Solid tumor 1.67 1.16-2.40 .006  
 Autoimmune disorders 1.60 0.97-2.62 .06  
 Organ transplantation 23.1 6.53-81.8 < .0001  
 Accidental radiation exposure 9.66 1.29-72.2 .03  

URD indicates unrelated donor; LD, low dose; TREO, treosufan; ATG, antithymocyte globulin; MPN, myeloproliferative neoplasm; and TMI, total marrow irradiation (with lung and liver shielding).

*

Modeled as a continuous linear variables (as specified in Table 5).

HLA allele level typing was not carried out.

Other regimens (CY + TMI; CY + BU; FLU + TBI + CY; FLU + BU) had no significant impact.

§

Other antecedent conditions had no significant impact.

Patients conditioned with FLU plus low-dose TBI (HR = 1.56) or BU/TBI (HR = 1.81) had significantly higher death rates than patients who received BU/CY, and although not statistically significantly different from BU/CY, patients conditioned with treosulfan plus FLU had the lowest rate of death (HR = 0.55, P = .20) of all regimens.

Overall, the rate of death decreased with more recent years of HCT (P = .0002).

Relapse-free survival.

Factors affecting relapse-free survival (supplemental Table 2) were similar to those impacting overall survival (Table 8) and, in addition, cytogenetics, disease stage, platelet count, etiology of the disease, patient age, donor type, conditioning regimen, and year of HCT.

Impact of monosomal karyotype on outcome.

Because there was a strong correlation between 5-group cytogenetics and the presence of monosomal karyotypes (Table 2), we carried out an additional analysis of the impact of monosomal karyotype in patients with poor-risk cytogenetics by the 5-group classification (which contained a mix of patients with and without monosomal karyotype). The incidence of relapse was higher in patients with monosomal karyotype compared with those without (HR = 1.56; confidence interval, 0.85-2.87), although the difference was not statistically significant (P = .15), presumably because of insufficient power (Figure 4). Yet, the rate of overall mortality was significantly higher among patients with monosomal karyotype than in those without (HR = 2.02; confidence interval, 1.34-3.04; P = .0007), an expression of significantly increased NRM, possibly related to a more severe disease pathophysiology.

Figure 4

Impact of monosomal karyotype (MK) on posttransplant relapseand overall survival (OS). Incidence among patients with poor-risk karyotype (by 5-group classification), without or with concurrent MK.

Figure 4

Impact of monosomal karyotype (MK) on posttransplant relapseand overall survival (OS). Incidence among patients with poor-risk karyotype (by 5-group classification), without or with concurrent MK.

Close modal

Impact of comorbidity.

HCT-CI scores were not included in the overall analysis because they were available for only 441 patients (44%). Among these, increasing scores were associated with an increased rate of mortality (P < .0001) and relapse (P < .0001); however, inclusion into the multivariable model did not qualitatively change the associations with the other variables, probably because comorbidity was associated with disease stage at HCT.17 

Comparison of IPSS and 5-group cytogenetic classifications.

There was almost complete agreement for patients categorized as “good” or intermediate” risk between the IPSS and the 5-group cytogenetic classifications. However, patients within the IPSS “poor” risk group were separated by the 5-group classification into 3 subcohorts, “very poor,” “poor,” and “intermediate.“ Among these subcohorts, the rate of relapse was similar for those with intermediate- and poor-risk cytogenetics (HR = 0.86; confidence interval, 0.46-1.61; P = .65) but was significantly higher in patients with very poor cytogenetics (HR = 2.92; confidence interval, 1.58-5.42; P = .0007; Figure 1C).

The risk of mortality relative to patients with intermediate cytogenetics was significantly higher for patients with very poor cytogenetics (HR = 2.59; confidence interval, 1.62-4.15; P < .0001) but did not differ significantly for those with poor cytogenetics (HR = 1.13; confidence interval, 0.71-1.79; P = .62). The 5-group category “very good” was too small to draw firm conclusions.

Clonal cytogenetic abnormalities, determined by classic banding technique or by FISH, are strongly associated with the prognosis of myeloid malignancies. The present data in a large cohort of patients with MDS transplanted at a single center underscore the profound effect of cytogenetics on relapse and mortality after HCT, currently the only treatment modality with curative potential. Moreover, results indicate that the newly proposed 5-group cytogenetic risk classification, developed for nontransplanted patients,9  distinguishes patients with different post-HCT prognosis more clearly than do the 3 risk groups of the original IPSS classification,2  particularly among patients with the lowest and highest risk of post-HCT relapse. Although retrospective data in a rather heterogeneous group of patients must be interpreted with caution, it is of note that the small cohort of patients in the very good risk group had a 5-year probability of relapse of 8% with 48% survival, compared with 50% and 8%, respectively, in the very poor risk group. By IPSS criteria, the relapse incidence was similar for the cytogenetic groups good and intermediate, and significantly lower than for the poor group, supporting the proposal by others that a separation into 2 risk categories by IPSS criteria was sufficient to distinguish patients with different post-HCT prognosis.18  However, patients with IPSS poor-risk cytogenetics, by 5-group criteria, was composed of 3 subcohorts, intermediate, poor, and very poor, with relapse incidence rates of 30% and 50% for the poor and very poor subcohorts, respectively. If, in addition, the presence of a monosomal karyotype is considered, this combination may select a subcohort of patients for whom HCT efforts with currently used modalities prove to be futile. Those results confirm reports by others on the negative impact of a monosomal karyotype19-21  in patients with AML; only one previous report22  failed to show a significant contribution of monosomy to prognosis beyond that of a complex karyotype. It is of note, however, that the present analysis of transplant outcomes indicates a negative impact of monosomal karyotype not only on relapse, but even more so on NRM. Further investigations into this effect of monosomal karyotype are warranted.

In addition to cytogenetics, myeloblast count (categorized according to FAB or WHO), proved significant for outcome, the former reflecting, presumably, the biologic risk profile of the disease, and the latter underscoring primarily the impact of tumor burden. Based on current clinical opinion, pre-HCT debulking therapy should reduce the incidence of relapse after HCT.23-25  The present results, however, revealed a negative impact of pre-HCT therapy on post-HCT outcome, either reflecting disease progression while on therapy26,27  or a lack of response to induction therapy,23,28  although our dataset did not allow to analyze results by depth of response (to either hypomethylating or induction type therapy).25,28  Alternatively, inferior outcome in these patients may have been related to intrinsic characteristics of the disease, such as high-risk cytogenetics, which had led to the decision to administer pre-HCT therapy in the first place; however, there was no statistical evidence of a preferential use of pre-HCT therapy in patients with high-risk cytogenetics (not shown).

Other risk factors for post-HCT outcome included HCT from donors other than HLA-identical siblings, although the relapse rate was lower in patients transplanted from unrelated donors, implying a clinically relevant graft-versus-leukemia effect. Secondary MDS was significant only for mortality, but not for relapse after adjusting for cytogenetics, in agreement with previous studies.29,30  The negative impact of lower platelet counts was thought to be related to the disease stage and underlying disease biology.

As expected, older patient age was associated with a higher probability of death, although it had no significant impact on relapse in multivariable analysis. Several studies reported that transplant outcome in “older” patients was not significantly different from results in younger persons.31,32  However, the definition of “older” (and “younger”) has varied from study to study, the methods used to quantify age (linear vs categorical) have been inconsistent, and a selection bias (for HCT) is highly probable.33  The present analysis included patients up to 75 years of age, many of whom were conditioned with high-intensity regimens as used in younger patients, suggesting that those patients were considered biologically younger than their chronologic age. The inclusion of comorbidities did not further improve the model, but comorbidity scores were available only in the most recently transplanted patients. The rate of mortality was not lower with the use of a low-intensity conditioning regimen (FLU + low-dose TBI). Indeed, there was evidence of higher mortality and relapse rates than observed with more intensive regimens, most likely related to patient selection17 ; however, the effect remained significant in multivariable analysis.

The finding that more recent transplant years were associated with a reduced rate of mortality is consistent with the report from our center in a large cohort of patients with various diagnoses,34  reflecting progressive improvement in supportive care measures.35  These results are encouraging, particularly because the median age of transplanted patients has increased by more than 2 decades from before 1990 to 2005, and the proportion of unrelated donor transplants has risen from 6% to 57%. The concurrent trend of an increase in relapse appears to be related to the declining proportion of patients with low-grade/early MDS (RA) and the increasing numbers of patients with advanced MDS/tAML. In addition, many MDS patients are currently being treated with hypomethylating agents as standard therapy36  and are being referred for HCT with greater delay after diagnosis.27  Prébet et al recently reported that, although the most promising option for patients with disease progression on azacitidine was HCT, the median survival after HCT was only 17 months.26  The present analysis failed to show a significant advantage of any particular regimen over a BU/CY regimen that uses BU targeting.6,37  Nevertheless, there was a trend for a treosulfan plus FLU combination38  to lead to improved survival. The high rate of failure in the small cohort of patients conditioned with radioimmunotherapy very likely was related to the enrollment of high-risk patients in a phase 1 or 2 study.39 

Both acute and chronic GVHD occurred with frequencies as reported in previous studies.40  GVHD did not represent a focus of the present analysis, and the interactions of cytogenetics with GVHD, the source of stem cells and relapse, will be the subject of a separate report.

The present analysis was based on clonal chromosomal abnormalities as defined by classic banding technique or FISH. It is likely, however, that differences in outcome within a given cytogenetic risk group as currently defined are related to gene mutations not reflected in cytogenetic findings41  and possibly other factors, such as DNA methylation, histone methylation, or acetylation or elevated ferritin levels.42-45  Further, recent studies show that mutations in genes, such as TP53, ASXL1, EZH2, or RUNX1, or genes involved in transcription, such as SF3B1, significantly affect prognosis of nontransplanted patients with MDS.41,46  Such mutations are also present in approximately half of all patients with a normal conventional karyotype and are correlated with significantly shortened survival in nontransplanted patients.41  As molecular mutation data are being validated, it will be necessary to test their relevance for HCT outcome. It is likely that there is a population of patients who, despite normal cytogenetics, should be considered high risk and, possibly, should be transplanted earlier in the disease course. Conversely, patients with normal cytogenetics may include a subcohort without mutations, which very likely has a better prognosis than the “lumped” data on patients with normal karyotype would suggest.

Thus, this analysis shows that the 5-group cytogenetic risk classification has greater discriminating power for post-HCT relapse and mortality than the IPSS cytogenetic risk classification, by separating subgroups within the IPSS cytogenetic poor-risk category. The analysis further shows that advances in the management of HCT patients in recent years has resulted in considerable improvements in survival, overcoming otherwise negative impact factors, such as older age, more advanced MDS at the time of HCT, and the increasing use of unrelated donors. This finding indicates that considerable progress has been made with the overall approach to HCT. However, future trials must focus on conditioning regimens that lead to reduced NRM in patients with low risk and reduced relapse incidence in patients with high-risk cytogenetics. Such improvement may not be achievable with conventional modalities but will require innovative strategies, involving, possibly, immunotherapeutic approaches, the use of novel compounds directed at mutated gene products, the signaling pathways dependent on those genes, and other adjuvant strategies before and after HCT.

The online version of this article contains a data supplement.

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 Helen Crawford and Bonnie Larson for help with manuscript preparation; Carlos Breton, Gary Schoch, and Elizabeth Soll for updating and maintaining the database; Dr Michael Boeckh for providing comments on the analysis; all physicians and physician assistants involved in the clinical care of patients; the referring physicians; and all patients who agreed to participate in clinical research.

This work was supported by The National Institutes of Health (grants HL036444, CA018029, CA015704, HL095999, CA078902, and HL062946).

National Institutes of Health

Contribution: H.J.D. and T.G. designed and carried out the analysis and wrote the manuscript; M.F., H.M.S., and D.M. provided expertise on cytogenetics and hematopathology; W.A.W. verified and collated data for analysis; and B.L.S., B.G., J.M.P., U.P., A.R., J.P.R., B.M.S., D.L.S., M.S., R.S., and F.R.A. read and provided critique for the manuscript.

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

Correspondence: H. Joachim Deeg, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D1–100, Seattle, WA 98109-1024; e-mail: jdeeg@fhcrc.org.

1
Naqvi
 
K
Garcia-Manero
 
G
Sardesai
 
S
, et al. 
Association of comorbidities with overall survival in myelodysplastic syndrome: development of a prognostic model.
J Clin Oncol
2011
, vol. 
29
 
16
(pg. 
2240
-
2246
)
2
Greenberg
 
P
Cox
 
C
LeBeau
 
MM
, et al. 
International scoring system for evaluating prognosis in myelodysplastic syndromes [erratum appears in Blood. 1998;91(3):1100].
Blood
1997
, vol. 
89
 
6
(pg. 
2079
-
2088
)
3
Malcovati
 
L
Germing
 
U
Kuendgen
 
A
, et al. 
A WHO classification-based prognostic scoring system (WPSS) for predicting survival in myelodysplastic syndromes [abstract].
Blood
2005
, vol. 
106
 
11
(pg. 
232a
-
233a
Abstract 788
4
Malcovati
 
L
Germing
 
U
Kuendgen
 
A
, et al. 
Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes.
J Clin Oncol
2007
, vol. 
25
 
23
(pg. 
3503
-
3510
)
5
Alessandrino
 
EP
Della Porta
 
MG
Bacigalupo
 
A
, et al. 
WHO classification and WPSS predict posttransplantation outcome in patients with myelodysplastic syndrome: a study from the Gruppo Italiano Trapianto di Midollo Osseo (GITMO).
Blood
2008
, vol. 
112
 
3
(pg. 
895
-
902
)
6
Deeg
 
HJ
Storer
 
B
Slattery
 
JT
, et al. 
Conditioning with targeted busulfan and cyclophosphamide for hemopoietic stem cell transplantation from related and unrelated donors in patients with myelodysplastic syndrome.
Blood
2002
, vol. 
100
 
4
(pg. 
1201
-
1207
)
7
Nevill
 
TJ
Shepherd
 
JD
Sutherland
 
HJ
, et al. 
IPSS poor-risk karyotype as a predictor of outcome for patients with myelodysplastic syndrome following myeloablative stem cell transplantation.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
2
(pg. 
205
-
213
)
8
Armand
 
P
Kim
 
HT
DeAngelo
 
DJ
, et al. 
Impact of cytogenetics on outcome of de novo and therapy-related AML and MDS after allogeneic transplantation.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
6
(pg. 
655
-
664
)
9
Schanz
 
J
Tuchler
 
H
Sole
 
F
, et al. 
New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge.
J Clin Oncol
2012
, vol. 
30
 
8
(pg. 
820
-
829
)
10
Schanz
 
J
Steidl
 
C
Fonatsch
 
C
, et al. 
Coalesced multicentric analysis of 2351 patients with myelodysplastic syndromes indicates an underestimation of poor-risk cytogenetics of myelodysplastic syndromes in the international prognostic scoring system.
J Clin Oncol
2011
, vol. 
29
 
15
(pg. 
1963
-
1970
)
11
Breems
 
DA
van Putten
 
WL
De Greef
 
GE
, et al. 
Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4791
-
4797
)
12
Sorror
 
ML
Giralt
 
S
Sandmaier
 
BM
, et al. 
Hematopoietic cell transplantation-specific comorbidity index as an outcome predictor for patients with acute myeloid leukemia in first remission: combined FHCRC and MDACC experiences.
Blood
2007
, vol. 
110
 
13
(pg. 
4608
-
4613
)
13
Petersdorf
 
EW
Gooley
 
T
Malkki
 
M
Horowitz
 
M
Clinical significance of donor-recipient HLA matching on survival after myeloablative hematopoietic cell transplantation from unrelated donors.
Tissue Antigens
2007
, vol. 
69
 
suppl 1
(pg. 
25
-
30
)
14
Bacigalupo
 
A
Ballen
 
K
Rizzo
 
D
, et al. 
Defining the intensity of conditioning regimens: working definitions.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
12
(pg. 
1628
-
1633
)
15
Gooley
 
TA
Leisenring
 
W
Crowley
 
J
Storer
 
BE
Estimation of failure probabilities in the presence of competing risks: new representations of old estimators.
Stat Med
1999
, vol. 
18
 
6
(pg. 
695
-
706
)
16
Jurado
 
M
Deeg
 
HJ
Storer
 
B
, et al. 
Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome after conditioning with busulfan and fractionated total body irradiation is associated with low relapse rate but considerable nonrelapse mortality.
Biol Blood Marrow Transplant
2002
, vol. 
8
 
3
(pg. 
161
-
169
)
17
Sorror
 
ML
Sandmaier
 
BM
Storer
 
BE
, et al. 
Comorbidity and disease status-based risk stratification of outcomes among patients with acute myeloid leukemia or myelodysplasia receiving allogeneic hematopoietic cell transplantation.
J Clin Oncol
2007
, vol. 
25
 
27
(pg. 
4246
-
4254
)
18
Armand
 
P
Deeg
 
HJ
Kim
 
HT
, et al. 
Multicenter validation study of a transplantation-specific cytogenetics grouping scheme for patients with myelodysplastic syndromes.
Bone Marrow Transplant
2010
, vol. 
45
 
5
(pg. 
877
-
885
)
19
Haferlach
 
C
Alpermann
 
T
Schnittger
 
S
, et al. 
Prognostic value of monosomal karyotype in comparison to complex aberrant karyotype in acute myeloid leukemia: a study on 824 cases with aberrant karyotype.
Blood
2012
, vol. 
119
 
9
(pg. 
2122
-
2125
)
20
Hemmati
 
PG
Terwey
 
TH
le Coutre
 
PD
, et al. 
Prognostic impact of a monosomal karyotype on the outcome of patients with acute myeloid leukemia or myelodysplastic syndrome undergoing allogeneic stem cell transplantation [abstract].
Blood
2011
, vol. 
118
 
21
pg. 
2028
 
21
Brands-Nijenhuis
 
AVM
Labopin
 
M
Schouten
 
HC
, et al. 
The influence of monosomal karyotype on survival in patients with acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation: a retrospective survey on behalf of the acute leukemia working party of the European Group for Blood and Marrow Transplantation (EBMT) [abstract].
Blood
2011
, vol. 
118
 
21
pg. 
158
 
22
Valcárcel
 
D
Adema
 
V
Mallo
 
M
, et al. 
Prognostic impact of monosomal karyotype in patients with myelodysplastic syndrome and abnormal karyotype: a report from the Spanish Group of MDS (GESMD) [abstract].
Blood
2011
, vol. 
118
 
21
pg. 
1724
 
23
Yakoub-Agha
 
I
de La Salmonière
 
P
Ribaud
 
P
, et al. 
Allogeneic bone marrow transplantation for therapy-related myelodsyplastic syndrome and acute myeloid leukemia: a long-term study of 70 patients. Report of the French Society of bone marrow transplantation.
J Clin Oncol
2000
, vol. 
18
 
5
(pg. 
963
-
971
)
24
Casper
 
J
Knauf
 
W
Kiefer
 
T
, et al. 
Treosulfan and fludarabine: a new toxicity-reduced conditioning regimen for allogeneic hematopoietic stem cell transplantation.
Blood
2004
, vol. 
103
 
2
(pg. 
725
-
731
)
25
Nakai
 
K
Kanda
 
Y
Fukuhara
 
S
, et al. 
Value of chemotherapy before allogeneic hematopoietic stem cell transplantation from an HLA-identical sibling donor for myelodysplastic syndrome.
Leukemia
2005
, vol. 
19
 
3
(pg. 
396
-
401
)
26
Prébet
 
T
Gore
 
SD
Esterni
 
B
, et al. 
Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure.
J Clin Oncol
2011
, vol. 
29
 
24
(pg. 
3322
-
3327
)
27
Gerds
 
AT
Gooley
 
TA
Estey
 
EH
Appelbaum
 
FR
Deeg
 
HJ
Scott
 
BL
Pre-transplant therapy with azacitidine verus induction chemotherapy and post-transplant outcome in patients with MDS
Biol Blood Marrow Transplant
 
doi: 2012;18(8):1211-1218
28
Scott
 
BL
Storer
 
B
Loken
 
M
Storb
 
R
Appelbaum
 
FR
Deeg
 
HJ
Pretransplantation induction chemotherapy and posttransplantation relapse in patients with advanced myelodysplastic syndrome.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
1
(pg. 
65
-
73
)
29
Chang
 
CK
Storer
 
BE
Scott
 
BL
, et al. 
Hematopoietic cell transplantation in patients with myelodysplastic syndrome or acute myeloid leukemia arising from myelodysplastic syndrome: similar outcomes in patients with de novo disease and disease following prior therapy or antecedent hematologic disorders.
Blood
2007
, vol. 
110
 
4
(pg. 
1379
-
1387
)
30
Litzow
 
MR
Tarima
 
S
Perez
 
WS
, et al. 
Allogeneic transplantation for therapy-related myelodysplastic syndrome and acute myeloid leukemia.
Blood
2010
, vol. 
115
 
9
(pg. 
1850
-
1857
)
31
Sorror
 
ML
Sandmaier
 
BM
Storer
 
BE
, et al. 
Long-term outcomes among older patients following nonmyeloablative conditioning and allogeneic hematopoietic cell transplantation for advanced hematologic malignancies.
JAMA
2011
, vol. 
306
 
17
(pg. 
1874
-
1883
)
32
Lim
 
Z
Brand
 
R
Martino
 
R
, et al. 
Allogeneic hematopoietic stem-cell transplantation for patients 50 years or older with myelodysplastic syndromes or secondary acute myeloid leukemia.
J Clin Oncol
2010
, vol. 
28
 
3
(pg. 
405
-
411
)
33
McClune
 
BL
Weisdorf
 
DJ
Pedersen
 
TL
, et al. 
Effect of age on outcome of reduced-intensity hematopoietic cell transplantation for older patients with acute myeloid leukemia in first complete remission or with myelodysplastic syndrome.
J Clin Oncol
2010
, vol. 
28
 
11
(pg. 
1878
-
1887
)
34
Gooley
 
TA
Chien
 
JW
Pergam
 
SA
, et al. 
Reduced mortality after allogeneic hematopoietic-cell transplantation.
N Engl J Med
2010
, vol. 
363
 
22
(pg. 
2091
-
2101
)
35
Tomblyn
 
M
Chiller
 
T
Einsele
 
H
, et al. 
Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective [erratum appears in Biol Blood Marrow Transplant. 2010;16(2):294].
Biol Blood Marrow Transplant
2009
, vol. 
15
 
10
(pg. 
1143
-
1238
)
36
McCarthy
 
JM
Shickle
 
L
Roberts
 
CH
Candler
 
KS
Chung
 
HM
5Azacytidine prior to allogeneic transplantation effectively reduces relapse, TRM and overall mortality in high-risk myelodysplastic and secondary AML [abstract].
Bone Marrow Transplant
2008
, vol. 
41
 
Suppl
(pg. 
212
-
213
)
37
Slattery
 
JT
Clift
 
RA
Buckner
 
CD
, et al. 
Marrow transplantation for chronic myeloid leukemia: the influence of plasma busulfan levels on the outcome of transplantation.
Blood
1997
, vol. 
89
 
8
(pg. 
3055
-
3060
)
38
Nemecek
 
ER
Guthrie
 
KA
Sorror
 
ML
, et al. 
Conditioning with treosulfan and fludarabine followed by allogeneic hematopoietic cell transplantation for high-risk hematologic malignancies.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
3
(pg. 
341
-
350
)
39
Pagel
 
JM
Gooley
 
TA
Rajendran
 
J
, et al. 
Allogeneic hematopoietic cell transplantation after conditioning with 131I-anti-CD45 antibody plus fludarabine and low-dose total body irradiation for elderly patients with advanced acute myeloid leukemia or high-risk myelodysplastic syndrome.
Blood
2009
, vol. 
114
 
27
(pg. 
5444
-
5453
)
40
Deeg
 
HJ
Storer
 
BE
Boeckh
 
M
, et al. 
Reduced incidence of acute and chronic graft-versus-host disease with the addition of thymoglobulin to a targeted busulfan/cyclophosphamide regimen.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
5
(pg. 
573
-
584
)
41
Bejar
 
R
Stevenson
 
K
Abdel-Wahab
 
O
, et al. 
Clinical effect of point mutations in myelodysplastic syndromes.
N Engl J Med
2011
, vol. 
364
 
26
(pg. 
2496
-
2506
)
42
Cazzola
 
M
Della Porta
 
MG
Travaglino
 
E
Malcovati
 
L
Classification and prognostic evaluation of myelodysplastic syndromes [review].
Semin Oncol
2011
, vol. 
38
 
5
(pg. 
627
-
634
)
43
Armand
 
P
Kim
 
HT
Cutler
 
CS
, et al. 
Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem cell transplantation.
Blood
2007
, vol. 
109
 
10
(pg. 
4586
-
4588
)
44
Bejar
 
R
LeVine
 
R
Ebert
 
BL
Unraveling the molecular pathophysiology of myelodysplastic syndromes [review].
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
504
-
515
)
45
Marcondes
 
AM
Mhyre
 
AJ
Stirewalt
 
DL
Kim
 
S-H
Dinarello
 
CA
Deeg
 
HJ
Dysregulation of IL-32 in myelodysplastic syndrome and chronic myelomonocytic leukemia modulates apoptosis and impairs NK function.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
8
(pg. 
2865
-
2870
)
46
Papaemmanuil
 
E
Cazzola
 
M
Boultwood
 
J
, et al. 
Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts.
N Engl J Med
2011
, vol. 
365
 
15
(pg. 
1384
-
1395
)

Supplemental data

Sign in via your Institution