Therapy-related myelodysplasia (t-MDS) and acute myeloid leukemia (t-AML) after high-dose chemotherapy (HD-CT) and autologous stem cell transplantation (ASCT) for malignant diseases have become an important problem. The actuarial risk has varied, but has often been high if compared to the risk after conventional therapy. Prior chemotherapy with large cumulative doses of alkylating agents is the most important risk factor. In addition, patient age and previous radiotherapy, particularly the use of total body irradiation (TBI) in the preparative regimen for ASCT, have been identified as risk factors. In 3 studies, patients transplanted with CD34+ cells from peripheral blood after chemotherapy priming showed a higher risk of t-MDS or t-AML than patients transplanted with cells isolated from the bone marrow without priming. To what extent this higher risk relates to the prior therapy with a different contamination with preleukemic, hematopoietic precursors of the CD34+ cells obtained by the 2 methods, or is a direct result of chemotherapy priming, or of an increasing awareness of these complications, remains to be determined. The latent period from ASCT to t-MDS and t-AML has often been short, 12 months or less in 27% of the patients. Bone marrow pathology of early cases of t-MDS after ASCT has often been neither diagnostic nor prognostic, but most patients presented chromosome aberrations, primarily deletions or loss of the long arms of chromosomes 5 and 7. The prognosis was in general poor, although 17% with indolent t-MDS survived more than 18 months from diagnosis, and most of these presented a normal karyotype or a single chromosome aberration.

During the last decade the issue of therapy-related myelodysplasia (t-MDS) and acute myeloid leukemia (t-AML) following high-dose chemotherapy (HD-CT) and autologous stem cell transplantation (ASCT) for malignant diseases has become increasingly important, as discussed in previous reviews.1,2 The number of patients with hematologic malignancies and chemosensitive solid tumors undergoing this new type of treatment has expanded dramatically, and because HD-CT and ASCT are followed by a limited morbidity and low mortality, much attention has been paid to the long-term outcome, including secondary malignancies. t-MDS and t-AML are the predominant types of secondary malignancies observed after HD-CT and ASCT. The actuarial risk of leukemic complications has varied considerably, but has often been very high compared to the results in studies of patients treated for the same diseases by conventional chemotherapy and radiotherapy. Several risk factors, the clinical and cytologic characteristics, the chromosome abnormalities, therapy, and survival of this interesting subgroup of patients with t-MDS and t-AML have been discussed in many studies. An important question has been the extent to which the high risk of leukemic complications relates to prior therapy with reinfusion of preleukemic hematopoietic precursor cells at ASCT, the extent to which the high risk could result directly from the HD-CT and ASCT procedures per se, or finally in a few cases could result from chemotherapy administered after ASCT for late relapse of the primary tumor.

Risk and risk factors

Following a few early reports briefly discussing single cases of t-MDS and t-AML,3-7 at least 16 studies,8-23 3 of which were a follow-up of previous reports,9,11,14 have evaluated the actuarial risk of t-MDS and t-AML following HD-CT and ASCT (Table1). In these studies the cumulative risk of t-MDS and t-AML has varied widely from 1.1% at 20 months16and up to 24.3% 43 months after ASCT.17 Much of this difference possibly relates to the wide confidence intervals, because the calculations have often been based on only a few cases of leukemia. However, major variations in risk factors from study to study may also explain the surprising differences observed in these well-conducted and well-controlled studies.

Table 1.

Risk of MDS and AML after autologous stem cell transplantation—results from the literature

Authors/year/
institution
Primary tumor
No. t-MDS/ No. t-AML/ No. treated Actuarial
risk/relative
risk (RR)
Risk factors
and remarks
Miller et al 19948 Minnesota  Hodgkin Non-Hodgkin  3/0/68  3/1/138  14.5%/4 y  PBSC/BMSC 31% vs. 10.5%;P = .0035  
Bhatia et al 19969 (follow-up) Hodgkin Non-Hodgkin  9/1/258  13.5%/6 y PBSC/BMSC; P = .004 Age >35 y;P = .07  
Traweek et al 199410 City of Hope  Hodgkin Non-Hodgkin  2/2/108 5/1/167  9%/3 y  5 patients with chromosome aberration: no signs of MDS  
Krishnan et al 199811 (follow-up) Hodgkin Non-Hodgkin  24/612* 9.4%/9 y PBSC/BMSC; P = .05 Topo II inhibitors—11q23/21q22 translocations  
Darrington et al 199412 Nebraska  Hodgkin Non-Hodgkin  4/2/249 6/0/262  4%/5 y  Age ≥40 y; P = .05 TBI; P = .06  
Stone et al 199413 Dana Farber  Non-Hodgkin  18/2/262  18%/6 y  Prolonged interval diagnosis to ASCT; P = .003 Duration of prior CT; P = .019 Radiation therapy; P = .032 Pelvic irradiation; P = .003 Age >38 y with low platelet counts  
Friedberg et al 199914 (follow-up)  Non-Hodgkin  41/0/552 19.8%/10 y  Fewer no. of cells reinfused;P = .0003  
Wheeler et al 199715 Dana Farber  Hodgkin Non-Hodgkin  6/300* 4.2%/5 y  Prior relapses; P = .009 Prior radiotherapy; P = .05  
Taylor et al 199716 Newcastle  Hodgkin Non-Hodgkin  0/1/52 0/0/62  1.1%/20 mo (+1 CML)  60% transplant. in 1. CR 
Pedersen-Bjergaard et al 199717 Copenhagen Hodgkin Non-Hodgkin  1/1/27 3/1/49  24.3%/43 mo RR = 357  Antecedent CT risk factor  
André et al 199818 French Cooperative  Hodgkin  5/3/467  4.3%/5 y  Splenectomy;P = .067 PBSC/BMSC;P = .077  
Harrison et al 199919British Cooperative  Hodgkin  8/595* 3.1%/5 y Quantity of prior therapy; P < .0001 MOPP; P = .0009 Lomustine;P = .001 Transplant. versus non-transplant; P = .25  
Milligan et al 199920 EMBT Cooperative  Hodgkin Non-Hodgkin  51/15/4998  4.6%/5 y 3.0%/5 y Age at transplantation; P < .001 TBI at conditioning; P < .001 Number of transplants; P < .002 Years from diagnosis to transplantation;P < .001  
Govindarajan et al 199621Little Rock, Arkansas  Multiple myeloma (tandem-transpl.) 0/0/71 7/0/117 0% 12%/4 y  ASCT in 1. CR versus ASCT in multitreated P = .02  
Laughlin et al 199822 Duke University  Breast cancer  3/2/8641-153 1.6%/4 y  —  
Kollmannsberger et al 199823 German/French Cooperative  Germ cell tumors (advanced stage)  2/4/302  1.3%/52 mo1-155 RR: 160 Previously etoposide >2.4 g/m2 + Cis + Ifos/Ctx 
Authors/year/
institution
Primary tumor
No. t-MDS/ No. t-AML/ No. treated Actuarial
risk/relative
risk (RR)
Risk factors
and remarks
Miller et al 19948 Minnesota  Hodgkin Non-Hodgkin  3/0/68  3/1/138  14.5%/4 y  PBSC/BMSC 31% vs. 10.5%;P = .0035  
Bhatia et al 19969 (follow-up) Hodgkin Non-Hodgkin  9/1/258  13.5%/6 y PBSC/BMSC; P = .004 Age >35 y;P = .07  
Traweek et al 199410 City of Hope  Hodgkin Non-Hodgkin  2/2/108 5/1/167  9%/3 y  5 patients with chromosome aberration: no signs of MDS  
Krishnan et al 199811 (follow-up) Hodgkin Non-Hodgkin  24/612* 9.4%/9 y PBSC/BMSC; P = .05 Topo II inhibitors—11q23/21q22 translocations  
Darrington et al 199412 Nebraska  Hodgkin Non-Hodgkin  4/2/249 6/0/262  4%/5 y  Age ≥40 y; P = .05 TBI; P = .06  
Stone et al 199413 Dana Farber  Non-Hodgkin  18/2/262  18%/6 y  Prolonged interval diagnosis to ASCT; P = .003 Duration of prior CT; P = .019 Radiation therapy; P = .032 Pelvic irradiation; P = .003 Age >38 y with low platelet counts  
Friedberg et al 199914 (follow-up)  Non-Hodgkin  41/0/552 19.8%/10 y  Fewer no. of cells reinfused;P = .0003  
Wheeler et al 199715 Dana Farber  Hodgkin Non-Hodgkin  6/300* 4.2%/5 y  Prior relapses; P = .009 Prior radiotherapy; P = .05  
Taylor et al 199716 Newcastle  Hodgkin Non-Hodgkin  0/1/52 0/0/62  1.1%/20 mo (+1 CML)  60% transplant. in 1. CR 
Pedersen-Bjergaard et al 199717 Copenhagen Hodgkin Non-Hodgkin  1/1/27 3/1/49  24.3%/43 mo RR = 357  Antecedent CT risk factor  
André et al 199818 French Cooperative  Hodgkin  5/3/467  4.3%/5 y  Splenectomy;P = .067 PBSC/BMSC;P = .077  
Harrison et al 199919British Cooperative  Hodgkin  8/595* 3.1%/5 y Quantity of prior therapy; P < .0001 MOPP; P = .0009 Lomustine;P = .001 Transplant. versus non-transplant; P = .25  
Milligan et al 199920 EMBT Cooperative  Hodgkin Non-Hodgkin  51/15/4998  4.6%/5 y 3.0%/5 y Age at transplantation; P < .001 TBI at conditioning; P < .001 Number of transplants; P < .002 Years from diagnosis to transplantation;P < .001  
Govindarajan et al 199621Little Rock, Arkansas  Multiple myeloma (tandem-transpl.) 0/0/71 7/0/117 0% 12%/4 y  ASCT in 1. CR versus ASCT in multitreated P = .02  
Laughlin et al 199822 Duke University  Breast cancer  3/2/8641-153 1.6%/4 y  —  
Kollmannsberger et al 199823 German/French Cooperative  Germ cell tumors (advanced stage)  2/4/302  1.3%/52 mo1-155 RR: 160 Previously etoposide >2.4 g/m2 + Cis + Ifos/Ctx 
*

No discrimination between t-MDS and t-AML.

Previously received only 1 cycle of CT.

Previously received more than 1 cycle of CT.

F1-153

One case of ALL with t(1;11) included.

F1-155

Only 4 cases of overt AML are included, whereas 2 cases of MDS are excluded in the calculation.

Abbreviations: PBSC indicates peripheral blood stem cells; BMSC, bone marrow stem cells; TBI, total body irradiation; CT, chemotherapy; CR, complete remission; Cis, cisplatin; Ifos, ifosphamide; Ctx, cyclophosphamide; MOPP, mechlorethamine, vincristine, procarbazine, and prednisone.

In several previous studies of t-MDS and t-AML following conventional chemotherapy, the risk has been shown to increase with patient age and with the exposure to alkylating agents.24-29 More precisely, in patients with Hodgkin disease the risk has been shown to increase by the square of age and is almost directly proportional to the cumulative dose of alkylating agents.30 In patients treated with HD-CT and ASCT, the risk of t-MDS and t-AML seems also to increase with age and with the burden of alkylating agents administered before the transplantation procedure (Table 1). In the study from Nebraska12 and in the follow-up study from Minnesota9 of patients transplanted for Hodgkin disease and non-Hodgkin lymphomas, ages over 40 and 35 years, respectively, were risk factors (Table 1). In the EBMT cooperative study20 of the same type of patient, age at transplantation was an independent and significant risk factor. In the French cooperative study of patients transplanted for Hodgkin disease,18 age over 40 years was a significant risk factor for the development of all types of secondary malignancy.

Increased duration of previous chemotherapy and treatment with alkylating agents were significant risk factors in 1 study from the Dana Farber Institute of patients with non-Hodgkin lymphomas13 (Table 1), and in a later study from the same institution,15 the number of prior relapses, also reflecting the burden of previous chemotherapy, was a significant risk factor. In the EBMT cooperative study,20 the interval from diagnosis of lymphoma to ASCT, likewise reflecting the burden of previous therapy, was a significant risk factor. In the British cooperative study of patients transplanted for Hodgkin disease,19 the quantity of previous therapy and chemotherapy with MOPP and with lomustine were all significant and independent risk factors for t-MDS and t-AML. In the study from Arkansas21 of patients transplanted for multiple myeloma, all patients who developed t-MDS belonged to a subgroup of 117 patients initially treated with more than 1 cycle of conventional chemotherapy including an alkylating agent, whereas 71 patients who underwent ASCT after only 1 cycle of chemotherapy did not develop leukemic complications (P = .02). In a study from Newcastle16 of patients transplanted for Hodgkin disease and non-Hodgkin lymphomas, in most cases early in the course of their disease, the cumulative risk of t-MDS and t-AML was only 1.1% at 20 months, whereas in our simultaneously published study from Copenhagen17 of multitreated patients of the same type, a very high risk of 24.3% at 43 months was observed. However, this high risk was at the same level as in our previous studies of patients multitreated for the same diseases with conventional chemotherapy.30 These observations all indicate a marked dose-response effect of prior chemotherapy with alkylating agents on the risk of t-MDS and t-AML following ASCT.

The risk of t-MDS and t-AML was low in the 2 studies of ASCT for solid tumors22,23 (Table 1). In both studies patients were apparently less pretreated than patients in the lymphoma studies, and patients with germ cell tumors were in general young. In addition, in the study of germ cell tumors, 2 cases of t-MDS were excluded from the risk estimate because the authors considered them as unrelated to therapy.23 Thus, different factors may have contributed to the low risk of t-MDS and t-AML observed in the 2 studies of HD-CT and ASCT for breast cancer and germ cell tumors.

In previous studies of patients treated traditionally with chemotherapy, including alkylating agents, for lymphomas and solid tumors, the leukemogenic potential of additional high-voltage radiotherapy has been controversial, because no increase in risk was observed in several studies.25,26,28-30 The risk of t-MDS or t-AML after radiotherapy alone, observed in 1 study, was approximately 10-fold lower than the risk following treatment with alkylating agents.27 In 2 independent studies from the Dana Farber Institute,13,15 the first a study of patients treated with ASCT for non-Hodgkin lymphomas, the second of patients treated with ASCT for Hodgkin disease and non-Hodgkin lymphomas, previous radiotherapy, in particular irradiation toward the pelvic region, was a significant risk factor for development of t-MDS and t-AML (Table 1). Perhaps even more important, in the study from Nebraska12 total body irradiation (TBI), used in the preparative regimen for ASCT, reached almost significance as a risk factor. A comparison of the 2 studies from the Dana Farber Institute supports that TBI could be a risk factor. In patients transplanted for non-Hodgkin lymphoma, the use of TBI in the preparative regimen for ASCT resulted in a very high actuarial risk of 18% at 6 years and 19.8% at 10 years,13,14 whereas in the parallel study from the same institution of patients transplanted for Hodgkin disease and non-Hodgkin lymphomas conditioned solely by chemotherapy,15the risk was only 4.2% at 5 years. Finally, in the recently published EBMT cooperative study,20 radiotherapy used in the conditioning regimen for ASCT was a significant and independent risk factor. Low-dose whole-body or hemibody irradiation was used many years ago as therapy for indolent lymphoid malignancies but was abandoned because it resulted in many cases of AML,31 and an actuarial risk of 17% after 15 years was observed.32 As a result of this experience, TBI in the preparative regimen for ASCT is now in many centers used more selectively in patients with malignancies that are highly sensitive to radiotherapy.

Among other risk factors under discussion for development of t-MDS and t-AML was prior splenectomy. In the French cooperative study18 of patients with Hodgkin disease, splenectomy was a risk factor with borderline significance. However, in the British cooperative study19 of the same type of patient, splenectomy was not a significant and independent risk factor. In the follow-up study from the Dana Farber Institute14 a lower number of CD34+ cells reinfused at ASCT was a risk factor. In the study of patients with multiple myeloma from Arkansas21 a high risk of 12% at 4 years was observed in the subgroup of 117 patients transplanted late in the course of their disease and 73% of these had received tandem transplantation. A specific effect on the risk of leukemia of repeated transplantations was confirmed by the EBMT cooperative study,20 in which the number of transplants received by each patient was a significant risk factor.

In previous studies of patients with malignant diseases treated conventionally with chemotherapy, often combined with radiotherapy, major variations in the risk estimates of t-MDS and t-AML have been observed. In patients treated with HD-CT and ASCT, similar variations are observed, likewise with an often much higher risk of t-MDS and t-AML in many smaller single-institute studies8-11,13,14,17,21 in which cytogenetic data are provided for each patient, as compared to a lower risk in many of the larger cooperative studies.18-20,23 These obviously benefit from more statistical power, and in the smaller studies a selection for publication of series presenting a very high risk, and the delimitation of the cohorts, could both contribute to a relatively high risk. However, a difference in case ascertainment should also be considered. The systematic use of bone marrow cytogenetics as an important diagnostic tool in most single-institute studies, especially in patients with refractory cytopenia but without an excess of blasts in the blood or the bone marrow, and a more careful clinical follow-up, even of patients with relapse of their primary tumor after ASCT, could also contribute to the difference in risks observed between the 2 types of study. Only rather few undiagnosed cases of t-MDS may lead to a marked underestimate of the actuarial risk of t-MDS and t-AML.

Some years ago, hematopoietic stem cell support for ASCT was always obtained as direct aspirates from the iliac crest without chemotherapy priming. Subsequently, because of a number of potential advantages, the harvesting procedures were changed, and CD34-enriched cells were obtained from peripheral blood after chemotherapy priming and administration of hematopoetic growth factors. The effects of this change on the outcome of the primary malignancy and on the risk of t-MDS and t-AML after ASCT have never been studied prospectively. A retrospective, matched-pair comparison of patients transplanted for Hodgkin disease treated with stem cells from the 2 sources has, as expected, demonstrated an equivalent survival outcome.33However, 3 independent retrospective studies have so far suggested,18 or even significantly demonstrated,8,9,11 a higher risk of t-MDS and t-AML following ASCT with CD34-enriched cells isolated from peripheral blood after chemotherapy priming and growth factors, as compared to ASCT using CD34+ cells from the bone marrow without pretreatment. There could be at least 3 reasons for this important difference. First, hematopoietic precursor cells critically damaged by chemotherapy priming, harvested before DNA repair is completed, reinfused and thereby forced to a marked proliferation and self-renewal, could explain the excess of t-MDS and t-AML with the use of cells isolated from peripheral blood. Second, CD34-enriched cells from peripheral blood could, for some reason, be more contaminated with preleukemic, clonogenic cells originating from the prior treatment, than cells isolated from the bone marrow. Third, an increased awareness of the risk of t-MDS and t-AML after the switch from harvesting cells from bone marrow to harvesting cells from peripheral blood, is also a consideration.

Biologically, CD34-enriched cells isolated from bone marrow without priming, and cells obtained from peripheral blood after chemotherapy priming, differ in several respects. Most importantly, they possess a different capacity to restore bone marrow function after myeloablative therapy. The more effective recovery of hematopoiesis, particularly of thrombopoiesis, by using cells isolated from peripheral blood, possibly relates to a higher content of clonogenic, normal hematopoietic precursor cells. This source of cells could likewise preferentially contain clonogenic preleukemic cells.

The CD34-enriched cells from the 2 sources show other important differences. As an example, in leukemia, cells isolated from peripheral blood after HD-CT may contain fewer or no cytogenetically abnormal cells, although such cells are still present in the bone marrow.34,35 Although this finding does not unambiguously indicate reasons for a higher risk of t-MDS and t-AML after ASCT with CD34-enriched cells from peripheral blood, it emphasizes the major biologic differences between the cells obtained by the 2 methods.

The possibility that the higher risk of t-MDS and t-AML observed following the shift of stem cell harvest from bone marrow to peripheral blood could actually be due to an increasing awareness of the problem has recently been supported by data from the EBMT cooperative study.20 In this study, the risk of leukemic complications increased significantly in time from 1978, whereas no difference in the risk was observed in relation to the procedure for stem cell harvest.

The cellular origin of t-MDS and t-AML following HD-CT and ASCT has been subject to much debate. The significant impact of primary chemotherapy with alkylating agents on the risk of t-MDS and t-AML after transplantation, outlined in different ways in several studies,13,15,17,19-21 with an actual risk similar to that of patients treated intensively with traditional types of chemotherapy,30 points toward a disease origin in events before transplantation, but perhaps triggered by a reinfusion of preleukemic hematopoietic precursors. Such an origin is supported by the very short latent period from HD-CT and ASCT to t-MDS or t-AML observed in many patients (see below), and by a recent study36 in which specific cytogenetic abnormalities observed in t-MDS or t-AML after transplantation were shown to be present already in the stem cell harvest in 9 of 12 patients.

Other findings, however, support the view that t-MDS and t-AML could be a direct result of the transplant procedure. The facts that the use of TBI in the preparative regimen for ASCT, and the use of CD34-enriched cells from peripheral blood after chemotherapy priming, have both been reported to increase the risk of t-MDS and t-AML, indicate that at least some cases of t-MDS and t-AML could be directly initiated or triggered by the transplantation procedure. A leukemogenic effect of TBI could seem intriguing because its purpose is a complete bone marrow ablation. However, in allogeneic bone marrow transplantation it has been demonstrated that residual recipient stem cells may often survive a preparative regimen of high-dose cyclophosphamide and TBI, as mixed chimerism was observed in 51% of such patients.37 Analogously, surviving hematopoietic precursors could be the origin of t-MDS and t-AML after ASCT. Also, the significantly increasing risk of t-MDS and t-AML with the number of transplants observed in the EBMT-conducted study,20 and, in cases of AML de novo, a shift in cytogenetic characteristics from balanced translocations to deletions or loss of chromosome arms 5q and 7q at relapse after conditioning with busulfan and cyclophosphamide,38 could be taken into account as a leukemogenic effect of the HD-CT and ASCT procedure itself.

The extent to which the origin of t-MDS and t-AML predominantly relates to prior chemotherapy or to the ASCT procedure has recently been addressed by the major British cooperative study19 of 4576 patients treated for Hodgkin disease. In this study, the general risk of t-MDS and t-AML was closely related to prior therapy with alkylating agents, and in a multivariate analysis, when taking the extent of this therapy into account, HD-CT and ASCT treatment in a subgroup of 595 patients was not associated with a significantly increased risk of leukemic complications (RR 1.83, CI 0.66-5.11). Thus, most cases of t-MDS and t-AML following HD-CT and ASCT are probably initiated by therapy prior to transplantation, whereas a minor part possibly is the result of the transplantation procedure.

The latent period from HD-CT and ASCT to development of t-MDS and t-AML has been specified for at total of 100 patients (Table2). Four cases were observed within 4 months, an additional 6 cases were observed within 8 months, and as many as 26 of 102 cases of t-MDS and t-AML were observed up to only 12 months after ASCT. Because previous studies have shown that a latent period of 12 months or less is rather rare in patients treated with conventional chemotherapy including alkylating agents,24-27,30these data likewise, as discussed, support an origin of the disease as being mainly prior to ASCT. Five cases of t-MDS were observed more than 5 years after ASCT. All 5 patients had previously received chemotherapy with alkylating agents, all 5 had characteristic defects of chromosome 5 or 7 or both, and there was no information on chemotherapy for relapse of their primary malignancy after ASCT. These cases emphasize that, although the general risk of t-MDS and t-AML decreases markedly 5 to 7 years after cessation of therapy,30 cases of late occurrence may occasionally be observed.

Table 2.

Latent period for the development of t-MDS or t-AML after autologous stem cell transplantation—review of the literature

Latent period (mo) t-MDS (no. of patients)t-AML (no. of patients)
1-4  3  1  
5-8  5  
9-12  12  4  
13-24  20  6  
25-36  12  
37-60  23  4  
>60  5  —  
Total  80 20 
Latent period (mo) t-MDS (no. of patients)t-AML (no. of patients)
1-4  3  1  
5-8  5  
9-12  12  4  
13-24  20  6  
25-36  12  
37-60  23  4  
>60  5  —  
Total  80 20 

In MDS and AML de novo, most cases can be diagnosed and subgrouped morphologically according to the French-American-British (FAB) classification.39,40 In MDS de novo, this classification is an important and independent prognostic factor that can contribute substantially to a prognostic score.41 Whereas many cases of overt t-AML and t-MDS with a high percentage of blasts can be classified according to the FAB criteria, cases of t-MDS with a low percentage of blasts after ASCT often do not meet the FAB criteria for classification.42-44 The bone marrow is often hypoplastic and fibrotic in such patients, and dysplastic changes are observed regularly, also in cases without evidence of t-MDS. For this reason neither the diagnosis nor a prediction of the outcome can be based on bone marrow pathology in such patients, but must rely on the presence of refractory cytopenia as well as clonal chromosome abnormalities in the bone marrow. The course of the disease may differ even in cases with chromosome abnormalities, and an aggressive type as well as a more indolent type of t-MDS have been described.42 

The disease presented as t-MDS in 132 patients and as overt t-AML in only 36 patients in well-documented reports. This large excess of t-MDS is characteristic for leukemic complications following alkylating agents45-47 and can be taken into account for a generally high diagnostic accuracy in these studies.

Detailed cytogenetic characteristics have been published for at least 75 patients with t-MDS or t-AML after HD-CT and ASCT.5,7,8,10,12,13,16,17,21-23,36,38,44Only 4 of these patients presented a normal karyotype, whereas 17 patients (23%) had a complex karyotype with more than 5 different cytogenetic abnormalities (Table 3). As in patients with t-MDS and t-AML in general,45-47 deletions or loss of the long arms or loss of the whole chromosomes 5 or 7 were most common, confirming that the majority of these cases are a result of prior therapy with alkylating agents. Interestingly, 11 patients presented balanced translocations to chromosome bands 11q23 or 21q22 and 2 patients a t(9;22). Twelve of these 13 patients had previously received chemotherapy with DNA topoisomerase II inhibitors, confirming the association between this type of chemotherapy and t-AML with balanced chromosome aberrations.48-51 Likewise, in the follow-up study from City of Hope,11 exposure to DNA topoisomerase II inhibitors before ASCT and priming with high-dose etoposide were significantly associated with the development of t-MDS and t-AML with balanced translocations to chromosome bands 11q23 and 21q22.

Table 3.

Cytogenetic characteristics of t-MDS and t-AML after autologous stem cell transplantation—review of the literature

Cytogenetics t-MDS (no. of patients) t-AML (no. of patients)
Normal  4  —  
1 Aberration  21  
2-3 Aberrations  14  4  
4-5 Aberrations  —  
>5 Aberrations  17  —  
Total 63  12  
−5 or del(5q)  26  4  
−7 or del(7q) 28  5  
−17 or del(17p)  5  —  
del(20q) 9  —  
Balanced transl. to 11q23  2  33-150 
Balanced transl. to 21q22  3  3  
Other unspecific balanced 13  —  
t(9;22)  1  13-151 
Cytogenetics t-MDS (no. of patients) t-AML (no. of patients)
Normal  4  —  
1 Aberration  21  
2-3 Aberrations  14  4  
4-5 Aberrations  —  
>5 Aberrations  17  —  
Total 63  12  
−5 or del(5q)  26  4  
−7 or del(7q) 28  5  
−17 or del(17p)  5  —  
del(20q) 9  —  
Balanced transl. to 11q23  2  33-150 
Balanced transl. to 21q22  3  3  
Other unspecific balanced 13  —  
t(9;22)  1  13-151 

Data extracted from references 5, 7, 8, 10, 12, 13, 16, 17, 21-23, 36, 38, and 44.

F3-150

One of these patients had acute lymphoblastic leukemia.

F3-151

This patient had chronic myeloid leukemia.

The presence of a cytogenetically abnormal clone in the bone marrow after HD-CT and ASCT is considered by most investigators as diagnostic for a leukemic complication, also in cases without an excess of blasts or other cytologic abnormalities characteristic of t-MDS or t-AML. A few cases of this type with uncharacteristic cytogenetic abnormalities have been shown to relate to bone marrow involvement of the primary tumor, detected due to the presence of cytogenetically unrelated clones.52 In other cases, cytogenetically abnormal clones, with uncharacteristic chromosome aberrations, have been observed in some patients without other evidence of MDS.10,11,13,42,53The origin and the significance of these findings have been discussed. In some cases the abnormalities have persisted for prolonged periods of time; in other cases refractory cytopenia and t-MDS have gradually developed. In our experience of t-MDS and t-AML54 as well as in other series, a few patients turn out to be long-term survivors. These are often patients with t-MDS and a single cytogenetic abnormality, mainly monosomy 7, and they may survive for many years before the disease, in all cases, shows progression.

The survival of patients with t-MDS and t-AML after HD-CT and ASCT was in general poor, as demonstrated in Table4. Median survival was 6 months and thereby identical to the survival of patients with t-MDS and t-AML in general.45,54 However, 11 patients were still alive more than 18 months after diagnosis, without further treatment. Five of these 11 long-term survivors, whose disease was discussed in more detail, presented defects of chromosome number 7, in 3 cases as the only cytogenetic abnormality present. Another 3 patients presented a normal karyotype.

Table 4.

Survival of patients from the diagnosis of t-MDS and t-AML after autologous stem cell transplantation—review of the literature

Survival (mo) Died of t-MDS (no. of patients)Died of t-AML (no. of patients) Still alive (no. of patients)
0-6  24  8  5  
7-12  12  5  
13-18  1  2  —  
>18  —  114-150 
Total  39  13  21 
Survival (mo) Died of t-MDS (no. of patients)Died of t-AML (no. of patients) Still alive (no. of patients)
0-6  24  8  5  
7-12  12  5  
13-18  1  2  —  
>18  —  114-150 
Total  39  13  21 
F4-150

Three of these patients presented a normal karyotype, another 5 patients a del(7q) or −7, in 3 cases as the only aberration, whereas 1 patient presented a del(11q).

Because of the generally poor response to conventional, intensive, antileukemic chemotherapy of patients with t-AML in general and after ASCT, allogeneic bone marrow transplantation has been attempted. In one study of patients with t-AML in general, the actuarial disease-free survival was 24% at 3 years.55 In this study, however, only 5 of 18 patients presented characteristic abnormalities of chromosomes 5 and 7, and only 2 a complex karyotype. In a study from Seattle,56 the 5-year disease-free survival of patients transplanted for t-AML in general was only 7.8% with no difference between untreated patients and patients brought into remission with intensive antileukemic chemotherapy before transplantation. Finally, in the study from the Dana Farber Institute all 13 patients who underwent allogeneic bone marrow transplantation for t-AML after ASCT have died.14 For this reason intensive antileukemic chemotherapy with subsequent harvest of karyotypically normal progenitor cells followed by HD-CT and ASCT has been attempted, and so far demonstrated a clinical effect in a few cases.35 To what extent this type of therapy or other regimens will be a future option, remains to be determined.

As demonstrated in an important joint study from Stanford and City of Hope, cytogenetic screening of the bone marrow before harvesting cells for ASCT is of major importance, particularly in heavily pretreated patients, to avoid transplantation in patients with an already established t-MDS.57 

Because of the poor prognosis, attempts have been made to predict the development of t-MDS and t-AML following ASCT. This has so far been possible to some extent in women by studying clonality, as revealed by the X-chromosome inactivation pattern of the human androgen receptor gene.58,59 To what extent this type of analysis will find a place in clinical practice remains to be determined.

Although t-MDS and t-AML after HD-CT and ASCT do not seem to differ from t-MDS and t-AML after conventional chemotherapy and radiotherapy as far as clinical characteristics, phenotype, and chromosome abnormalities are concerned, the leukemic complications following ASCT present specific problems. Because of the high risk observed in many studies and the dramatically increasing number of patients undergoing this type of therapy, efforts to decrease the risk of t-MDS and t-AML should have a high priority together with a search for more effective types of therapy in patients who have acquired a leukemic complication. Among the possibilities of decreasing the risk of t-MDS and t-AML in patients suffering from a primary tumor with poor prognostic indices is transplantation early in first complete remission. This may improve the outcome with regard to the primary tumor as well as reduce the risk of t-MDS and t-AML. Such measures are already undertaken in many centers, primarily in patients with malignant lymphomas. Likewise, a restrictive use of TBI in the preparative regimens for ASCT must be considered if the risk of leukemic complications should be shown to outweigh the potential benefit of radiotherapy.

Because of its many advantages, harvesting of CD34+ cells from peripheral blood after chemotherapy priming has become the dominant standard procedure in obtaining stem cells for transplantation. In consequence, it is now impossible prospectively to solve the question as to what extent this procedure is particularly leukemogenic. Instead, a modification of the priming procedure should be considered. Most rational would be to substitute alkylating agents and DNA topoisomerase II inhibitors in the preparative regimens for ASCT by cytostatic drugs without leukemogenic potential, such as the antimetabolites. Also, the methods for an early detection of clonality before and after transplantation will possibly improve, and if clonality is observed, purging of the CD34+ cells harvested for ASCT could be a possibility.

The generally poor results of intensive antileukemic chemotherapy in patients with advanced t-MDS and t-AML after HD-CT and ASCT is analogous to the experience in t-MDS and t-AML following conventional chemotherapy.47-49 They relate primarily to an initial phase of t-MDS observed in most patients and to the cytogenetic characteristics with deletion or loss of the long arms of chromosomes 5 and 7 and a complex karyotype. In addition, in t-MDS and t-AML after HD-CT and ASCT a reduced pool of hematopoietic stem cells may explain the many treatment-related deaths due to persistent bone marrow hypoplasia after intensive antileukemic therapy. Although allogeneic bone marrow transplantation in some studies offers rather poor results, it may at present represent the best choice of therapy in younger patients with advanced t-MDS or t-AML following HD-CT and ASCT, but also new therapeutic approaches should be considered, such as autologous tandem transplantations.

Note added in proof. Since submission of this manuscript, 3 important studies have been published discussing t-MDS and t-AML after HD-CT and ASCT.60-62 

Micallef et al reported an actuarial risk of t-MDS and t-AML of 14.2% and 36.5% at 5 and at 10 years after HD-CT and ASCT in 230 patients transplanted for non-Hodgkin lymphomas.60 Prior therapy with fludarabine and older age were significant risk factors. Twenty-two out of 24 patients with cytogenetic data available showed monosomy of chromosome arms 5q or 7q. The study confirms the high risk of t-MDS and t-AML observed in other single institute studies using cytogenetics as a diagnostic tool.

Yakoub-Agha et al demonstrated a 2-year overall survival of 30% in 70 patients with t-MDS or t-AML after HD-CT and ASCT following allogeneic bone marrow transplantation.61 Age 37 years or younger, female sex, negative CMV serology, and the use of an intensive conditioning regimen were predictive for a more favorable outcome, and patients with abnormalities of chromosome 7 and a complex karyotype had an inferior survival rate. The study confirms that allogeneic bone marrow transplantation must be considered in younger patients with t-MDS or t-AML after HD-CT and ASCT.

Krishnan et al reported a follow-up study of patients transplanted for Hodgkin disease and non-Hodgkin lymphomas.62 The actuarial risk of t-MDS and t-AML was 8.6% at 6 years. An increased risk was observed in 61 out of 612 patients primed with VP-16 for stem cell harvest (P = .002), which likewise was significantly associated with development of t-AML with 11q23/21q22 chromosome abnormalities (P = .006). The study for the first time demonstrates that the type of priming may be of importance.

The authors are indebted to Professor Flemming Skovby, MD, to Mr Harry Cowan, BSc, and to Hanne Nielsen for their help in preparing the manuscript.

Supported by grants from the Danish Cancer Association and H:S Forskningspulje 1997.

Reprints:J. Pedersen-Bjergaard, Department of Clinical Genetics, Section 4052, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.

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

1
Stone
RM
Myelodysplastic syndrome after autologous transplantation for lymphoma: the price of progress?
Blood.
83
1994
3437
2
Deeg
HJ
Socié
G
Malignancies after hematopoietic stem cell transplantation: many questions, some answers.
Blood.
91
1998
1833
3
Wheeler
C
Antin
JH
Churchill
WH
et al
Cyclophosphamide, carmustine and etoposide with autologous bone marrow transplantation in refractory Hodgkin's disease and non-Hodgkin's lymphoma: a dose-finding study.
J Clin Oncol.
8
1990
648
4
Biermann
PJ
Bagin
RG
Jagannath
S
et al
High dose chemotherapy followed by autologous hematopoietic rescue in Hogkin's disease: long term follow-up in 128 patients.
Ann Oncol.
4
1993
767
5
Marolleau
JP
Brice
P
Morel
P
Gisselbrecht
C
Secondary acute myeloid leukemia after autologous bone marrow transplantation for malignant lymphomas.
J Clin Oncol.
11
1993
590
6
Rohatiner
AZS
Johnson
PWM
Price
CGA
et al
Myeloablative therapy with autologous bone marrow transplantation as consolidation therapy for recurrent follicular lymphoma.
J Clin Oncol.
12
1994
1177
7
Roman-Unfer
S
Bitran
JD
Hanauer
S
et al
Acute myeloid leukemia and myelodysplasia following intensive chemotherapy for breast cancer.
Bone Marrow Transplant.
16
1995
163
8
Miller
JS
Arthur
DC
Litz
CE
Neglia
JP
Miller
WJ
Weisdorf
DJ
Myelodysplastic syndrome after autologous bone marrow transplantation: an additional late complication of curative cancer therapy.
Blood.
83
1994
3780
9
Bhatia
S
Ramsay
NKC
Steinbuch
M
et al
Malignant neoplasms following bone marrow transplantation.
Blood.
87
1996
3633
10
Traweek
ST
Slovak
ML
Nademanee
AP
Brynes
RK
Niland
JC
Forman
SJ
Clonal karyotypic hematopoietic cell abnormalities occurring after autologous bone marrow transplantation for Hodgkin's disease and non-Hodgkin's lymphoma.
Blood.
84
1994
957
11
Krishnan
A
Bhatia
S
Bhatia
R
et al
Risk factors for development of therapy-related leukemia (t-MDS/t-AML) following autologous transplantation (ABMT) for lymphoma [abstract].
Blood.
92
1998
493a
12
Darrington
DL
Vose
JM
Anderson
JR
et al
Incidence and characterization of secondary myelodysplastic syndrome and acute myelogenousleukemia following high-dose chemoradiotherapy and autologous stem-cell transplantation for lymphoid malignancies.
J Clin Oncol.
12
1994
2527
13
Stone
RM
Neuberg
D
Soiffer
R
et al
Myelodysplastic syndrome as a late complication following autologous bone marrow transplantation for non-Hodgkin's lymphoma.
J Clin Oncol.
12
1994
2535
14
Friedberg
JW
Neuberg
D
Stone
RM
et al
Outcome in patients with myelodyplastic syndrome after autologous bone marrow transplantation for non-Hodgkin's lymphoma.
J Clin Oncol.
17
1999
3128
15
Wheeler
C
Khurshid
A
Ibrahim
J
Elias
A
Mauch
P
Antin
J
Low incidence of post-transplant myelodysplasia/acute leukemia (MDS/AML) in NHL patients autotransplanted after cyclophosphamide, carmustine and etoposide (CBV) [abstract].
Blood.
90
1997
385b
16
Taylor
PRA
Jackson
GH
Lennard
AL
Hamilton
PJ
Proctor
SJ
Low incidence of myelodysplastic syndrome following transplantation using autologous non-cryopreserved bone marrow.
Leukemia.
11
1997
1650
17
Pedersen-Bjergaard
J
Pedersen
M
Myhre
J
Geisler
C
High risk of therapy-related leukemia after BEAM chemotherapy and autologous stem cell transplantation for previously treated lymphomas is mainly related to primary chemotherapy and not to the BEAM-transplantation procedure.
Leukemia.
11
1997
1654
18
André
M
Henry-Amar
M
Blaise
D
et al
Treatment-related deaths and second cancer risk after autologous stem-cell transplantation for Hodgkin's disease.
Blood.
92
1998
1933
19
Harrison
CN
Gregory
W
Hudson
GV
et al
High-dose BEAM chemotherapy with autologous haemopoietic stem cell transplantation for Hodgkin's disease is unlikely to be associated with a major increased risk of secondary MDS/AML.
Br J Cancer.
81
1999
476
20
Milligan
DW
Ruiz De Elvira
MC
Kolb
H-J
et al
Secondary leukemia and myelodysplasia after autografting for lymphoma: results from the EBMT.
Br J Haematol.
106
1999
1020
21
Govindarajan
R
Jagannath
S
Flick
JT
et al
Preceding standard therapy is the likely cause of MDS after autotransplants for multiple myeloma.
Br J Haematol.
95
1996
349
22
Laughlin
MJ
McGaughey
DS
Crews
JR
et al
Secondary myelodysplasia and acute leukemia in breast cancer patients after autologous bone marrow transplant.
J Clin Oncol.
16
1998
1008
23
Kollmannsberger
C
Beyer
J
Droz
J-P
et al
Secondary leukemia following high cumulative doses of etoposide in patients treated for advanced germ cell tumors.
J Clin Oncol.
16
1998
3386
24
Valagussa
P
Santoro
A
Bellani
FF
Franchi
F
Banfi
A
Bonadonna
G
Absence of treatment-induced second neoplasms after ABVD in Hodgkin's disease.
Blood.
59
1982
488
25
Boice
JD
Greene
MH
Killen
JY
et al
Leukemia and preleukemia after adjuvant treatment of gastrointestinal cancer with semustine (methyl-CCNU).
N Engl J Med.
309
1983
1079
26
Greene
MH
Harris
EL
Gershenson
DM
et al
Melphalan may be a more potent leukemogen than cyclophosphamide.
Ann Intern Med.
105
1986
360
27
Tucker
MA
Coleman
CN
Cox
RS
Varghese
A
Rosenberg
SA
Risk of second cancers after treatment for Hodgkin's disease.
N Engl J Med.
318
1988
76
28
Kaldor
JM
Day
NE
Pettersson
F
et al
Leukemia following chemotherapy for ovarian cancer.
N Engl J Med.
322
1990
1
29
van Leeuwen
FE
Chorus
AMJ
van den Belt-Dusebout
AW
et al
Leukemia risk following Hodgkin's disease: relation to cumulative dose of alkylating agents, treatment with teniposide combinations, number of episodes of chemotherapy, and bone marrow damage.
J Clin Oncol.
12
1994
1063
30
Pedersen-Bjergaard
J
Specht
L
Larsen SO, et al. Risk of therapy-related leukaemia and preleukaemia after Hodgkin's disease.
Lancet.
I
1987
83
31
O'Donnell
JF
Brereton
HD
Greco
FA
Gralnick
HR
Johnson
RE
Acute nonlymphocytic leukemia and acute myeloproliferative syndrome following radiation therapy for non-Hodgkin's lymphoma and chronic lymphocytic leukemia: clinical studies.
Cancer.
44
1979
1930
32
Travis
LB
Weeks
J
Curtis
RE
et al
Leukemia following low-dose total body irradiation and chemotherapy for non-Hodgkin's lymphoma.
J Clin Oncol.
14
1996
565
33
Perry
AR
Peniket
AJ
Watts
MJ
Leverett
D
Goldstone
AH
Linch
DC
Peripheral blood stem cell versus autologous bone marrow transplantation for Hodgkin's disease: equivalent survival outcome in a single-centre matched-pair analysis.
Br J Haematol.
105
1999
280
34
Carella
AM
Frassoni
F
Pollicardo
N
et al
Philadelphia-chromosome-negative peripheral blood stem cells can be mobilized in the early phase of recovery after a myelosuppressive chemotherapy in Philadelphia-chromosome-positive acute lymphoblastic leukaemia.
Br J Haematol.
89
1995
535
35
Carella
AM
Dejana
A
Lerma
E
et al
In vivo mobilization of karyotypically normal peripheral blood progenitor cells in high-risk MDS, secondary or therapy-related acute myelogenous leukaemia.
Br J Haematol.
95
1996
127
36
Abruzzese
E
Radford
JE
Miller
JS
et al
Detection of abnormal pretransplant clones in progenitor cells of patients who developed myelodysplasia after autologous transplantation.
Blood.
94
1999
1814
37
Roy
DC
Tantravahi
R
Murray
C
et al
Natural history of mixed chimerism after bone marrow transplantation with CD6-depleted allogeneic marrow: a stable equilibrium.
Blood.
75
1990
296
38
Rege
KP
Janes
SL
Saso
R
et al
Secondary leukemia characterized by monosomy 7 occurring post-autologous stem cell transplantation for AML.
Bone Marrow Transplant.
21
1998
853
39
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposals for the classification of the acute leukemias: French-American-British (FAB) co-operative group.
Br J Haematol.
33
1976
451
40
Bennett
JM
Catovsky
D
Daniel
MT
et al
Proposals for the classification of the myelodysplastic syndromes.
Br J Haematol.
51
1982
189
41
Greenberg
P
Cox
C
Le Beau
MM
et al
International scoring system for evaluating prognosis in myelodysplastic syndromes.
Blood.
89
1997
2079
42
Wilson
CS
Traweek
T
Slovak
ML
Niland
JC
Forman
SJ
Brynes
RK
Myelodysplastic syndrome occurring after autologous bone marrow transplantation for lymphoma: morphologic features.
Am J Clin Pathol.
108
1997
369
43
Amigo
ML
del Cañizo
MC
Rios
A
et al
Diagnosis of secondary myelodysplastic syndromes (MDS) following autologous transplantation should not be based only on morphological criteria used for diagnosis of de novo MDS.
Bone Marrow Transplant.
23
1999
997
44
Sobecks
RM
LeBeau
MM
Anastasi
J
Williams
SF
Myelodysplasia and acute leukemia following high-dose chemotherapy and autologous bone marrow or peripheral blood stem cell transplantation.
Bone Marrow Transplant.
23
1999
1161
45
Le Beau
MM
Albain
KS
Larson
RA
et al
Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosomes no. 5 and 7.
J Clin Oncol.
4
1986
325
46
Johansson
B
Mertens
F
Heim
S
Kristoffersson
U
Mitelman
F
Cytogenetics of secondary myelodysplasia (sMDS) and acute nonlymphocytic leukemia (sANLL).
Eur J Haematol.
47
1991
17
47
Pedersen-Bjergaard
J
Pedersen
M
Roulston
D
Philip
P
Different genetic pathways in leukemogenesis for patients presenting with therapy-related myelodysplasia and therapy-related acute myeloid leukemia.
Blood.
86
1995
3542
48
Pui
C-H
Behm
FG
Raimondi
SC
et al
Secondary acute myeloid leukemia in children treated for acute lymphoid leukemia.
N Engl J Med.
321
1989
136
49
Pedersen-Bjergaard
J
Philip
P
Balanced translocations involving chromosome bands 11q23 and 21q22 are highly characteristic of myelodysplasia and leukemia following therapy with cytostatic agents targeting at DNA-topoisomerase II.
Blood.
78
1991
1147
50
Larson
RA
Le Beau
MM
Ratain
MJ
Rowley
JD
Balanced translocations involving chromosome bands 11q23 and 21q22 in therapy-related leukemia.
Blood.
79
1992
1892
51
Pedersen-Bjergaard
J
Andersen
MK
Johansson
B
Balanced chromosome aberrations in leukemias following chemotherapy with DNA-topoisomerase II inhibitors.
J Clin Oncol.
16
1998
1897
52
Pedersen-Bjergaard
J
Timshel
S
Andersen
MK
Andersen
A-ST
Philip
P
Cytogenetically unrelated clones in therapy-related myelodysplasia and acute myeloid leukemia: experience from the Copenhagen series updated to 180 consecutive cases.
Genes Chromosomes Cancer.
23
1998
337
53
Imrie
KR
Dubé
I
Prince
HM
Girouard
C
Crump
M
Keating
A
New clonal karyotypic abnormalities acquired following autologous bone marrow transplantation for acute myeloid leukemia do not appear to confer an adverse prognosis.
Bone Marrow Transplant.
21
1998
395
54
Pedersen-Bjergaard
J
Philip
P
Larsen
SO
Jensen
G
Byrsting
K
Chromosome aberrations and prognostic factors in therapy-related myelodysplasia and acute nonlymphocytic leukemia.
Blood.
76
1990
1083
55
Ballen
KK
Gilliland
DG
Guinan
EC
et al
Bone marrow transplantation for therapy-related myelodysplasia: comparison with primary myelodysplasia.
Bone Marrow Transplant.
20
1997
737
56
Anderson
JE
Gooley
TA
Schoch
G
et al
Stem cell transplantation for secondary acute myeloid leukemia: evaluation of transplantation as initial therapy or following induction chemotherapy.
Blood.
89
1997
2578
57
Chao
NJ
Nademanee
AP
Long
GD
et al
Importance of bone marrow cytogenetic evaluation before autologous bone marrow transplantation for Hodgkin's disease.
J Clin Oncol.
9
1991
1575
58
Gale
RE
Bunch
C
Moir
DJ
Patterson
KG
Goldstone
AH
Linch
DC
Demonstration of developing myelodysplasia/acute myeloid leukemia in haematologically normal patients after high-dose chemotherapy and autologous bone marrow transplantation using X-chromosome inactivation patterns.
Br J Haematol.
93
1996
53
59
Mach-Pascual
S
Legare
RD
Lu
D
et al
Predictive value of clonality assays in patients with Non-Hodgkin's lymphoma undergoing autologous bone marrow transplant: a single institution study.
Blood.
91
1998
4496
60
Micallef
INM
Lillington
DM
Apostolidis
J
et al
Therapy-related myelodysplasia and secondary acute myelogenous leukemia after high-dose therapy with autologous hematopoietic progenitor-cell support for lymphoid malignancies.
J Clin Oncol.
18
2000
947
61
Yakoub-Agha
I
de La Salmonière
P
Ribaud
P
et al
Allogenic bone marrow transplantation for therapy-related myelodysplastic syndrome and acute myeloid leukemia: a long-term study of 70 patients—Report of the French Society of Bone Marrow Transplantation.
J Clin Oncol.
18
2000
963
62
Krishnan
A
Bhatia
S
Slovak
ML
et al
Predictors of therapy-related leukemia and myelodysplasia following autologous transplantation for lymphoma: an assessment of risk factors.
Blood.
95
2000
1588
Sign in via your Institution