You are on page 1of 23

Achievements in Understanding and Treatment of

Myelodysplastic Syndromes
Eva Hellström-Lindberg (Chair), Cheryl Willman, A. John Barrett, and Yogen Saunthararajah

The myelodysplastic syndromes (MDS) constitute a cially in those with low blast counts. In Section III
challenge for the biologist as well as for the treat- Dr. Hellström-Lindberg presents results of alloge-
ing physician. In Section I, Dr. Willman reviews the neic and autologous stem cell transplantation
current classifications and disease mechanisms (SCT), intensive and low-dose chemotherapy. The
involved in this heterogeneous clonal hematopoi- results of allogeneic SCT in MDS are slowly improv-
etic stem cell disorder. A stepwise genetic progres- ing but are still poor for patients with unfavorable
sion model is proposed in which inherited or cytogenetics and/or a high score according to the
acquired genetic lesions promote the acquisition of International Prognostic Scoring System. A recently
“secondary” genetic events mainly characterized published study of patients between 55–65 years
by gains and losses of specific chromosome old showed a disease-free survival (DFS) at 3 years
regions. The genetic risk to develop MDS is likely of 39%. Consolidation treatment with autologous
multifactorial and dependent on various constella- SCT after intensive chemotherapy may result in
tions of risk-producing and -protecting alleles. In long-term DFS in a proportion of patients with high-
Section II Dr. Barrett with Dr. Saunthararajah risk MDS. Low-dose treatment with 5-azacytidine
addresses the immunologic factors that may act as has been shown to significantly prolong the time to
important secondary events in the development of leukemic transformation or death in patients with
severe pancytopenia. T cells from patients with high-risk MSA. Erythropoietin and granulocyte
MDS may suppress autologous erythroid and colony-stimulating factor may synergistically
granulocytic growth in vitro, and T cell suppression improve hemoglobin levels, particularly in
by antithymocyte globulin or cyclosporine may sideroblastic anemia. Recent therapeutic advances
significantly improve cytopenia, especially in have made it clear that new biological information
refractory anemia. Recent studies have also dem- may lead to new treatment modalities and, in
onstrated an increased vessel density in MDS bone combination with statistically developed predictive
marrow, and a phase II trial of thalidomide showed models, help select patients for different therapeu-
responses in a subgroup of MDS patients espe- tic options.

I. BIOLOGIC AND GENETIC FEATURES OF THE history of these syndromes ranges from more indolent
MYELODYSPLASTIC SYNDROMES forms of disease spanning years to those with a rapid
Cheryl L. Willman, M.D.* evolution to AML. Thus, MDS is best considered a pre-
leukemic disorder in which the neoplastic clone that has
Recent scientific advances have provided new insights been established may or may not fully progress to acute
into the etiology and pathogenesis of the myelodysplastic leukemia. Although the relationship between MDS and
syndromes (MDS). Despite heterogeneous morphologic, de novo AML has been controversial and current disease
genetic, biologic, and clinical features, all forms of MDS classification systems (Table 1) are considered unsatis-
are clonal hematopoietic stem cell disorders character- factory, most hematologists now consider MDS and AML
ized by ineffective hematopoiesis and peripheral cyto- as part of the same continuous disease spectrum rather
penias. Although a substantial proportion of MDS cases than as distinct disorders. This review will briefly high-
evolve to acute myeloid leukemia (AML), the natural light current controversies in the classification of MDS
and AML, the cytogenetic and molecular genetic features
* University of New Mexico Cancer Center, 2325 Camino de Salud of MDS, the biologic features that characterize MDS in-
NE, Room 101, Albuquerque NM 87131 cluding abnormal apoptosis and an altered marrow mi-

110 American Society of Hematology


Table 1. MDS Classification Systems.1

IPSS Risk-Based
FAB Classification System2 WHO Classification System3 Classification System4

Refractory Anemia (RA): Cytopenia of one Myelodysplastic Syndromes Overall IPSS Risk Score Based On:
PB lineage; normo- or hypercellular marrow with
dysplasias; < 1% PB Blasts and < 5% BM Blasts. Refractory Anemia (RA) Marrow Blast Percentage
With ringed sideroblasts (RARS) Blast % IPSS Score
Refractory Anemia with Ringed Sideroblasts Without ringed sideroblasts <5 0
(RARS): Cytopenia, dysplasia and the same % 5-10 0.5
blast involvement in BM and PB as RA. Ringed Refractory Cytopenia (MDS) with 11-20 1.5
sideroblasts account for >15% of nucleated Multilineage Dysplasia (RCMD) 21-30 2.0
cells in marrow.
Refractory Anemia with Cytogenetic Features5
Refractory Anemia with Excess Blasts Excess Blasts (RAEB) Karyotype IPSS Score
(RAEB): Cytopenia of two or more PB lineages; Good prognosis 0
dysplasia involving all 3 lineages; < 5% PB 5q- Syndrome (-Y, 5q-,20q-)
blasts and 5-20% BM Blasts. Intermediate prognosis 0.5
Myelodysplastic syndrome, Poor prognosis 1.0
Refractory Anemia with Excess Blasts in unclassifiable (abn. 7; Complex)
Transformation (RAEB-T): Hematologic features
identical to RAEB. > 5% Blasts in PB or 21-30% Myelodysplastic/Myeloproliferative Cytopenias6
Blasts in BM or the presence of Auer rods Diseases Cytopenia IPSS Score
in the blasts. None or 1 Type 0
Chronic Myelomonocytic 2 or 3 Types 0.5
Chronic Myelomonocytic Leukemia (CMML): Leukemia (CMML)
Monocytosis in PB (>1x109 per liter); < 5% blasts Overall IPSS Score and Survival
in PB and up to 20% BM blasts Atypical Chronic Myelogenous
Leukemia (aCML) Overall Score Median Survival
Low (0) 5.7 Yrs.
Juvenile Myelomonocytic Intermediate
Leukemia (JMML) 1 (0.5 or 1.0) 3.5 Yrs
2 (1.5 or 2.0) 1.2 Yrs.
High (> 2.5) 0.4 Yrs.

1 Abbreviations: PB, peripheral blood; BM, bone marrow; abn, abnormality


2 References 3-4.
3 Reference 21.
4 Reference 30.
5 IPSS Cytogenetic Classification28: Good prognosis: -Y only, normal, del(5q)
only, del(20q) only; Intermediate prognosis: +8, Single
miscellaneous abnormality, double abnormalities; Poor prognosis: Complex (i.e. > 3 abnormalities), any chromosome 7 abnormality.
6 IPSS Types of Cytopenia28: Hemoglobin <10g per deciliter; Absolute neutrophil count <1500 per cubic millimeter; Platelet count
< 100,000 per cubic millimeter.

croenvironment, and new and highly interesting insights Epidemiology, and End Results (SEER) program
into the complex genetic predisposition to MDS. Excel- (www.seer.cancer.gov). This has greatly impeded stud-
lent, well-referenced reviews are also available.1,2 ies of the true incidence, natural history, and epidemiol-
ogy of MDS in the US. Importantly, however, European
MDS and AML Disease Classification Systems: epidemiologic studies suggest that the incidence of MDS
Unresolved Controversies is at least as high as that of AML, particularly AML cases
The French-American-British (FAB) Classification, pro- that arise in older individuals.5 In the US, the age-spe-
posed in 1977, provided hematologists with the first con- cific incidence rate for AML in males aged 50 years at
sistent framework for morphologic classification of MDS diagnosis is 3.5 per 100,000, increasing dramatically to
(Table 1), the myeloproliferative disorders, and the acute 15 at age 70 and 35 at age 90. 6 (See also
leukemias.3,4 However, the separation of MDS as a dis- www.seer.cancer.gov.) With the exponential increase in
tinct disorder from AML in the FAB classification scheme the incidence of AML with age and the aging of our popu-
has been perceived by many to have scientifically im- lations, the median age at diagnosis of AML in the US is
peded our understanding of the full spectrum of leuke- currently 63 years. Thus, the majority of AML cases, like
mic progression.1 Indeed, the initial failure to recognize MDS, occur in older individuals. Further linking MDS
and classify MDS as a “neoplastic” pre-leukemic disor- and AML, several studies have noted that the biologic,
der and part of the same disease spectrum as AML re- morphologic, and genetic features of AML arising in older
sulted in the exclusion of MDS cases from virtually all individuals are similar to 1) primary MDS; 2) AML aris-
US cancer registries and the NCI-sponsored Surveillance, ing secondary to antecedent MDS; 3) AML arising sec-

Hematology 2000 111


ondary to prior therapy, particularly alkylating agent ex- the evolution of MDS to AML as proposed in the new
posure; and 4) AML cases that arise from documented WHO Classification21 (Table 1) has been perceived by
environmental or occupational exposures to agents such many hematologists to be even more problematic.26,27
as benzene, petroleum, organic solvents, and arsenical While RAEB-T and AML arising clinically as “de novo”
pesticides.7-13 disease in older patients share highly similar cytogenetic
In the FAB Classification (Table 1), the two primary features,13,26 they have differing clinical and biologic fea-
distinguishing features between the various MDS sub- tures and therapeutic responsiveness.26-28 Although not
types, chronic myelomonocytic leukemia (CMML) and directly tested in a randomized fashion, in several, if not
AML, are blast cell percentage and the presence of dys- all, studies RAEB-T patients appear to have a worse re-
plastic features. CMML is now considered a myelopro- sponse to intensive chemotherapy when compared his-
liferative/leukemic-like disorder and frequently associ- torically to AML cases with similar biologic and cytoge-
ated with t(5;12)(q33;p13),1,14 and AML is defined as > netic features.28,29 Thus, it will be particularly important
30% marrow blasts with the various MDS subtypes rang- to retain the distinct RAEB-T MDS subtype in order to
ing from < 5% to < 30% blasts. However, the previous compare future therapeutic advances in AML/MDS to
distinction between MDS and AML has become blurred historical controls. Additional concerns that have arisen
with the recognition of several common features of the with the proposed WHO Classification (Table 1) include29:
two diseases: 1) MDS is now recognized to be a clonal 1) the proposal that a diagnosis of RA and RARS be re-
pre-leukemic hematopoietic stem cell disorder frequently stricted to patients who have abnormalities solely involv-
associated with specific recurrent cytogenetic abnormali- ing the erythroid lineage, even though a diagnosis of MDS
ties15-18; 2) multi-lineage dysplasia is now recognized to requires dysplasia in at least two hematopoietic lineages;
occur in the majority of AML cases presenting clinically 2) the creation of vague new MDS diagnostic categories
as “de novo” disease in older individuals7,19-21; 3) AML (“refractory cytopenias with multilineage dysplasia
arising in older individuals and primary, secondary, or (RCMD)” and “MDS, unclassifiable”) which have no
therapy-induced MDS are now known to share strikingly biologic, clinical, or genetic basis; and 3) the general lack
similar biologic and genetic features7-13; 4) de novo AML of clinical and prognostic relevance in the proposed WHO
cases such as those with t(8;21) and inv(16) may present classification scheme. Unfortunately for clinicians and
clinically with less than 30% marrow blasts and may have diagnosticians alike, these controversies will likely con-
dysplastic features21; and 5) transgenic and “knock-in” tinue until our knowledge has increased to the degree
murine models of leukemia made with fusion genes from that disease classification systems can be developed on
translocations associated primarily with de novo AML clinical features, genetics/genomics, and functional biol-
[t(15;17), t(8;21), inv(16)] are often characterized by ogy. And as our knowledge continues to evolve, classifica-
hematopoietic dysplasia or an MDS-like disease ante- tion systems will necessarily require constant revision.
cedent to the development of AML.22-25 These more re- Taking an alternative approach, others have worked
cent clinical and biologic studies indicate that disorders to develop risk-based classification systems for MDS in
previously classified as MDS are part of the same dis- order to facilitate clinical decision-making.30 The Inter-
ease continuum as AML and that MDS is best consid- national Scoring System for Evaluating Prognosis (IPSS)
ered a pre-leukemic disorder with variable frequencies in MDS assigns IPSS scores to varying degrees of those
and rates of progression to AML. clinical and biologic features that today provide the most
As we now recognize that MDS and AML are part prognostic significance in MDS: marrow blast cell per-
of the same continuous biologic and genetic spectrum of centage, karyotype, and degree of cytopenia (Table 1).29
disease, the use of arbitrary “thresholds” for the distinc- An overall IPSS score developed using these variables
tion of AML from MDS for the purposes of disease clas- has a strong correlation with predicted median survival.29
sification and therapeutic decision making has become The IPSS system has proven to be a highly useful method
particularly problematic. At what blast cell percentage for evaluating prognosis in MDS patients, has achieved
should a clinician institute AML-based therapies in an international acceptance, and is being used to design clini-
MDS patient progressing to RAEB-T and from RAEB-T cal trials.
to AML? Should AML-based therapies be instituted in a
patient whose marrow has dysplastic morphologic fea- Genetic Features of MDS: Models for Genetic
tures, a blast cell percentage < 20%, and a t(8;21)-con- Progression and Clues to Etiology
taining clonal population of cells? While the threshold
of 30% blasts used by the FAB Classification to distin- MDS is a clonal hematopoietic stem cell disorder
guish AML from MDS (Table 1) is clearly arbitrary, a characterized by step-wise genetic progression
reduction in this threshold to 20% blasts and the result- Initially demonstrated by studies of expression of the vari-
ant elimination of RAEB-T as a distinct clinical stage in ous isoforms of the X-linked gene G6PD and more re-

112 American Society of Hematology


cently by molecular methods that detect non-random pat- primarily characterized by stepwise gains and losses of
terns of X-inactivation, evidence for clonality has been specific chromosomal regions (particularly chromosome
found in all forms of MDS, even in their very earliest 3p-, 3q-, 5q-, 7q-, 12p-, -17, -18, 20q11-12, +8). Such
stages.17,18 Interestingly, cytogenetic and molecular data gross chromosomal changes are ultimately accompanied
provide evidence, in some MDS patients, for the exist- by sub-microscopic DNA mutations of genes such as p53,
ence of a clonal phase of disease prior to the acquisition FLT3, or RAS, methylation of specific gene promoters,
of the characteristic cytogenetic abnormalities associated and in some cases by the reciprocal translocations and
with MDS.18 Similarly, MDS patients who evolve to acute inversions more frequently associated with AML. This
leukemia may after therapy revert to a cytogenetically model of step-wise genetic progression for MDS and re-
normal but persistently clonal remission. Such findings lated AML is strikingly reminiscent of those proposed
have led to the hypothesis that the recurrent cytogenetic for human solid tumors, such as colon cancer. Three lines
abnormalities associated with MDS, previously consid- of evidence support this model and the existence of a
ered the “primary” cause of disease, are actually “sec- genetic predisposition to MDS: 1) the occurrence of AML
ondary” cytogenetic abnormalities that arise due to cyto- and MDS in families with inherited defects in DNA re-
genetically undetectable initiating lesions in a clonal he- pair or neurofibromatosis-type I33-37; 2) genetic mapping
matopoietic stem cell population.30 Such initiating events studies in the rare families with “familial” MDS and
are likely to be heterogeneous and could either be inher- AML38-41; and 3) studies of the association of various
ited or result from acquired somatic DNA damage, ge- genetic polymorphisms with AML and MDS.42-48
nomic instability, defective DNA repair, or perturbations
in cell signal transduction pathways that give rise to stem An inherited genetic predisposition to MDS
cell clones that have a growth or survival advantage. In Support for an inherited predisposition to MDS and re-
contrast to the de novo acute leukemias that occur pri- lated AML has long been evident from studies of inher-
marily in younger patients (particularly those associated ited constitutional genetic defects (such as Schwachman-
with balanced translocations and inversions such as Diamond syndrome, the defective DNA repair of Fanconi
t(8;21), t(15;17), inv(16), t(9;11), etc. lacking dysplastic anemia, or deregulation of the RAS signal transduction
features), MDS and AML arising in older individuals pathway in neurofibromatosis-type 1) that are present in
appear to have a different model of genetic progression a large proportion of children who develop MDS and
(Figure 1).31-32 In this proposed model, initiating genetic AML.33-37 Indeed, recent studies indicate that up to 30%
lesions (which may be inherited or acquired) promote of children affected by MDS and related myeloprolifera-
the acquisition of “secondary” genetic events that are tive disorders have an inherited constitutional genetic

Figure 1. Proposed model for genetic progression of MDS to AML.

Hematology 2000 113


disorder.34 The original studies by Shannon and col- studies also further cement a genetic and etiologic link
leagues35,36 of the genetic basis of familial MDS with chro- between MDS and AML (and even CML). Not unexpect-
mosome 7q abnormalities (the “monosomy 7 syndrome”) edly, a low percentage of sporadic AML, ALL, and CML
are important in that they first revealed that abnormali- cases (5–8%) have recently been reported to have simi-
ties of chromosome 7q were not the “primary” cause of lar AML1 mutations50; whether such AML1 mutations
the syndrome; indeed, these investigators concluded that can be observed in primary “sporadic” MDS cases is
the predisposition locus mapped to some other as yet currently under investigation. Whether AML and MDS
unidentified chromosomal location. Thus, the foundation cases with AML1 mutations are indeed “sporadic” or rep-
was laid for the putative existence of genetic loci that resent AML and MDS cases that have arisen in individu-
could “predispose” to chromosomal instability, second- als with inherited AML1 mutations is as yet unknown.
ary loss of specific chromosomal regions (such as 5q, Given the functional role and association of CBF (map-
7q, and 20q), and the ultimate development of MDS and ping to chromosome 16q22) with AML1 in normal and
AML in adults and children. This original hypothesis was neoplastic hematopoiesis,51 it is tempting to further specu-
recently supported the findings of Gilliland and colleagues late that CBF might be the causative gene for the sec-
who determined the genetic basis of familial platelet dis- ond predisposition locus in AML and MDS in those fa-
order with leukemia (FPD/AML), an autosomal-domi- milial cases where the predisposition has been mapped
nant congenital thrombocytopenia characterized by plate- to chromosome 16q21-23.2.40
let aggregation abnormalities.38-39 Affected individuals in
the seven pedigrees studied to date all have a striking Models for the development of sporadic MDS:
propensity to develop MDS, AML, and more rarely, Cumulative environmental exposures in genetically
chronic myelogenous leukemia (CML). Interestingly, the predisposed individuals
MDS and AML cases that develop in these pedigrees have While genetic and familial mapping studies have clearly
the cytogenetic abnormalities classically associated with demonstrated that mutations in a specific gene, such as
MDS, particularly abnormalities of chromosomes 5q and AML1, NF1, or genes mediating DNA repair, can pre-
7q and complex abnormalities. After mapping the FDP/ dispose to the acquisition of secondary cytogenetic ab-
AML predisposition locus to chromosome 21q22 in 1998, normalities and MDS, it is likely that such inherited ge-
Gilliland and colleagues went on to determine that the netic mutations will account for only a minority of MDS
causative gene for this disorder was CBFA2 (AML1), cases. How the majority of “sporadic” MDS cases arise
the gene whose function is most frequently disrupted in is as yet undetermined. However, epidemiologic case-
the acute leukemias by various reciprocal translocations control studies of MDS (and related AML) have demon-
and inversions, including the t(8;21), inv(16), t(3;21), and strated associations between MDS and smoking, expo-
t(12;21).49 Heterogeneous point mutations and small de- sure to chemical compounds (particularly petroleum prod-
letions of a single AML1 allele were found in these dif- ucts and diesel derivatives, exhausts, organic solvents,
ferent pedigrees.39 Despite this molecular heterogeneity, fertilizers, and nitro-organic explosives), semi-metals
each mutation characteristic of each pedigree affected the (arsenic and thallium), stone dusts (such as silica), and
DNA-binding domain of one AML1 allele, particularly cereal dusts.52-53 In light of these epidemiologic studies, it is
targeting the two arginine residues at positions 166 and interesting that evidence is increasing for a complex ge-
201 that bind to DNA. The change of arginine to netic predisposition to MDS involving naturally occurring
glutamine resulted in a loss of DNA-binding activity.38 DNA polymorphisms in genes that mediate DNA repair
These data thus support the hypothesis that AML1 may and metabolize environmental carcinogens.42-48 These stud-
surprisingly function as a tumor suppressor gene and that ies are leading to a model, also diagrammed in Figure 1,
loss of one AML1 allele (hemizygous loss) is sufficient in which MDS arises as a result of cumulative environ-
to initiate tumorigenesis and establish a neoplastic clone mental exposures in genetically predisposed individuals.
in affected individuals. This loss of function of a single In this case, the genetic predisposition is a more com-
AML1 allele appears to also confer a susceptibility to plex genetic trait: a constellation of genetic variants in
the acquisition of secondary mutations and the gain and/ critical polymorphic genes. The initial attempts to dis-
or loss of the chromosomal regions frequently associ- sect this complex genetic predisposition have focused on
ated with AML and MDS. This discovery has led to a the association of MDS with naturally occurring poly-
particularly attractive model for MDS/AML whereby morphisms in genes that mediate carcinogen metabo-
AML1 mutations predispose to chromosome instability lism.42-48 Following initial reports of the association of a
leading to the eventual loss of chromosomes 5q, 7q; such non-functioning 609C➝T polymorphic allele of the
models are currently being developed in mice in which NAD(P)H:Quinone Oxidoreductase (NQO1) gene that
the mutated AML1 allele has been introduced (Downing plays a critical role in detoxifying benzene metabolites
and Gilliland, personal communication). These pivotal with an increased incidence of hematologic malignan-

114 American Society of Hematology


cies in Chinese workers exposed to benzene,43 several normalities are likely no less critical for disease progres-
groups have attempted to determine the incidence of this sion, and identification of the gene(s) involved in these
polymorphism in primary and secondary leukemia regions remains very important. Unfortunately, despite
cases.44,45 Larson and colleagues first reported an in- years of mapping and definition of minimally deleted
creased frequency of the NQ01 609C➝T polymorphism chromosomal regions on chromosomes 5, 7, and 20, no
in patients with therapy-related AML, particularly in AML investigator has yet succeeded in identifying the “single”
patients with abnormalities involving chromosomes 5 and tumor suppressor gene that is responsible for MDS and
7, 88% of whom were homozygous for the non-func- AML on any of these chromosomes. Recent detailed cy-
tional allele.44 Interestingly, studies of de novo AML in togenetic and molecular mapping studies reveal that re-
children45 and adults (C.L. Willman et al, manuscript in arrangements and deletions involving these chromosomes
preparation and M.A. Smith et al, personal communica- are very complex and that multiple distinct regions may
tion) have failed to demonstrate an association of this contribute to the disease phenotype or progression: at least
NQO1 polymorphism with abnormalities of chromosome two different regions are implicated on chromosome 7q
5 and 7, but have instead demonstrated strong associa- (7q22 and 7q32-34) and more than four different regions
tions with balanced translocations and inversions, par- may be involved on chromosome 5q (5q11, 5q13-q21,
ticularly involving MLL and chromosome 11q23. While 5q31, and distal 5q33-35).54-61 One issue with many map-
it is tempting to speculate that the NQO1 609C➝T poly- ping studies is that in most instances little attention was
morphism could predispose to the development of MDS, paid to “phenotype” rather than “genotype.” In other
no such studies focusing on MDS cases have been re- words, patient samples were selected for molecular stud-
ported; our own limited studies of 120 primary MDS cases ies based on the presence of a specific cytogenetic ab-
have failed to reveal such an association. Similar com- normality (such as a 5q- or a 7q- with or without addi-
plexities have arisen in studies of the association of MDS tional cytogenetic abnormalities) without regard to the
and polymorphisms in the glutathione S-transferases specific form of disease or mode of disease presentation
(GST) that mediate exposure to cytotoxic and genotoxic (primary MDS, secondary AML, de novo AML, or t-
agents, specifically the “null” variant allele GST theta 1 AML/MDS). Both cytogenetic and molecular genetic
(GSTT1).46-48 Chen and colleagues initially reported that studies are not only revealing the tremendous heteroge-
the frequency of the GSTT1 null genotype was higher neity in different breakpoints but also the need to focus
among MDS cases than controls.46 However, Fenaux and on a pure genotype and phenotype for mapping studies.
colleagues47 and other groups48 did not confirm these ini- Very detailed cytogenetic studies by Pederson in 1996
tial observations and actually reported that the incidence revealed that while chromosome region 5q31 was deleted
of the GSTT1 null genotype tended to be higher in unex- in all patients with MDS, other chromosome 5 regions
posed MDS patients and controls. Thus, while the hy- were deleted more often than 5q31 in AML patients; the
pothesis and model that MDS arises due to cumulative chromosome 5q13-q21 region was particularly involved
environmental exposures in genetically predisposed in- in the genetic progression of RA to RAEB and 5q22-
dividuals is indeed attractive, these studies of natural 5q33 for further progression to AML.61 Recent studies
human genetic variation and disease association are only by Westbrook and colleagues have focused on a single
in their infancy. Moreover, it is likely that true genetic AML patient with a very small deletion in the 5q31 re-
risk will not be simply determined through studies of one gion. In this patient, the D5S500-D5S594 region was
gene, but through the constellation of risk-producing and identified to be the minimal deletion interval, and this
risk-protecting alleles present in each individual. Thus, interval was shown to contain nine transcriptional units
it will ultimately be necessary to study polymorphic vari- with five unknown expressed sequence tags (ESTs) and
ants in many human genes in a large number of affected the genes CDC25, HSPA9, EGR1, and CTNNA1.58 While
individuals and controls and carefully monitor environ- all of these sequences are interesting candidates and are
mental exposures in order to dissect what is likely to be a expressed in hematopoietic cells, none has yet been iden-
very complex genetic predisposition. tified as “critical” for disease. It also remains possible
that loss of more than one gene in this region, as well as
Cloning and identification of genes disrupted by the the other distinct regions on chromosome 5, could actu-
recurrent cytogenetic abnormalities associated with MDS ally be responsible for the disease phenotype. Boultwood
The identification of potential “initiating” genetic lesions and colleagues56,57 have focused on the identification of
in MDS and related AML patients has lead to the hy- the gene(s) deleted in MDS patients who have the iso-
pothesis that the cytogenetic abnormalities traditionally lated “5q- syndrome,” a clinically distinct form of RA
associated with MDS (involving chromosomes 7q, 5q, associated with more indolent disease and a lower rate of
20q11-12, trisomy 8, 12p, and 3q) are “secondary” ge- progression to AML, chronic macrocytic anemia, throm-
netic events. However, these secondary cytogenetic ab- bocytosis, and dysplastic megakaryocytes. Interestingly,

Hematology 2000 115


the region on chromosome 5 specifically associated with Biologic Features of MDS
this disease presentation appears to be more distal to 5q31;
novel transcriptional units have also been recently iden- An abnormal marrow microenvironment:
tified in this distinct region.57 Cytokines, adhesion, apoptosis
In light of the complexity of this cytogenetic and mo- While there is strong evidence supporting the view that
lecular data, it is attractive to hypothesize that in MDS, MDS arises from an intrinsic or acquired genetic defect
an initiating abnormality gives rise to genome instability in hematopoietic stem cells leading to clonal expansion
leading to the deletion and rearrangement of particularly of a stem cell population, it is also clear that other epige-
susceptible chromosomal regions, such as those on chro- netic abnormalities such as aberrant cytokine production,
mosome 5q and 7q. Cytogenetic studies have revealed altered stem cell adhesion, or an abnormal marrow mi-
the continued instability of these regions during disease croenvironment contribute to the biology of the disease
progression in individual patients.62 While it may be that and may provide important therapeutic targets (Figure
loss of function of a single gene in each of these rela- 2). While studies reporting aberrant cytokine expression
tively large regions is responsible for disease progres- profiles in MDS patients have been criticized for their
sion, more recent studies have given more credence to lack of controls, lack of a suitable ex vivo system to study
the possibility that hemizygous loss of the function of the true clonogenic hematopoietic stem cell and marrow
several genes in each of these regions could contribute to stromal cell interactions, and the failure to precisely iden-
the disease phenotype. tify the cellular origin of particular cytokines, abnormali-
ties and elevations in tumor necrosis factor- (TNF),
Molecular mutations and genome methylation in MDS transforming growth factor- (TGF), and interleukin-
In addition to the complex cytogenetic abnormalities seen 1 (IL-1) have all been reported.2 In particular, an over-
in MDS, several molecular defects have also been re- abundance of IL-1 and a relative lack of its antagonist
ported. Studies of MDS in children, with or without Neu- IL-1(ra) have been hypothesized to support the clonal
rofibromatosis-type 1, have provided evidence that ab- expansion of aberrant hematopoietic stem cells.2
normalities in the RAS signal transduction pathway co- Several recent studies have revealed that in its early
operate with a loss of genes on chromosome 7 to pro- stages, MDS is characterized by accelerated apoptosis of
mote myeloid leukemogenesis.63 In adults with MDS, hematopoietic progenitor cells.71-72 While some contro-
disease progression has been associated with the muta- versy remains, most studies of MDS samples using vari-
tions in genes such as RAS, p53, and FLT364-67 and with ous techniques have demonstrated a lowered apoptotic
progressive methylation and transcriptional inactivation threshold of marrow CD34+ cells to TNF-, interferon-
of critical cell cycle regulatory genes such as p15INK4b  (IFN-), and anti-FAS antibodies.70,71 Excessive apopto-
that normally function to inhibit cyclin-dependent kinase sis is an attractive explanation for how a clonal expan-
activity at the G1 phase of the cell cycle.68-69 MDS pa- sion of marrow progenitor cells could result in ineffec-
tients have also been shown to have defective activation tive hematopoiesis and marrow failure. However, how
of signal transduction pathways, particularly involving such accelerated apoptosis is initiated or acquired is not
STAT5, in response to erythropoietin,70 which may ac- yet understood. Apoptosis may be triggered in cells by
count in part for the defective erythropoiesis and persis- intrinsic DNA damage or in cells that have an abnormal
tent anemia. These molecular abnormali-
ties associated with MDS all further con-
tribute to an escape from normal cell cycle
regulation, a disruption of the faithful
maintenance of DNA integrity and repair
leading to continued genetic instability,
and the altered activation of various sig-
nal transduction pathways.

Figure 2. MDS—An altered marrow microenvironment.

Cytokines, adhesion defects, angiogenesis, accelerated apoptosis, adhesion


dependence of proliferation and survival.

116 American Society of Hematology


growth factor milieu; both mechanisms could contribute II. IMMUNE MECHANISMS AND MODULATION IN MDS
to the accelerated apoptosis observed in MDS. Another A. John Barrett, M.D., FACP,* and Yogen
biologic mechanism that can induce apoptosis is a lack Saunthararajah*
of appropriate adhesive interactions between cells and
their stromal support (Figure 2). In the case of MDS, Despite improvements in the supportive care of patients
perturbed adhesive interactions between clonogenic he- with MDS with transfusions and antibiotics, nearly 50%
matopoietic stem cells and the underlying marrow stroma of patients die before transformation to acute leukemia
or endothelium could clearly trigger excessive apopto- from complications of thrombocytopenic bleeding or in-
sis. The migration, retention, and survival of hematopoi- fection. Treatment of the marrow failure alone should
etic stem cells in the marrow microenvironment is con- therefore bring survival advantages to a significant pro-
trolled by critical adhesion receptors including CD34, portion of patients with MDS. To improve the treatment
CD44, selectins, and integrins; 1 integrins play a par- of the cytopenia associated with MDS requires an under-
ticularly critical role in this process and virtually all he- standing of the pathophysiology of the disease. It is well
matopoietic stem cells express high levels of 4, 5, 6, accepted that MDS is a clonal stem cell disorder, charac-
and 1 integrins.73-74 Unfortunately, this is a relatively terized by cytopenia, progressive de-differentiation of
unexplored area of MDS and is worthy of continued in- myeloid lineages and ultimately unregulated blast pro-
vestigation. Interestingly, while MDS is characterized by liferation. Recent research reveals a further dimension to
excessive apoptosis, further genetic progression in this this pathophysiology, with extrinsic immunological and
disease and the transition to AML appears to be associ- microenvironmental factors compounding the intrinsic
ated with a loss of functional apoptosis, though additional stem cell defect and contributing to the pancytopenia and
studies documenting this progressive change in individual possibly to leukemic progression. In this section, patho-
patients with disease progression are clearly necessary. physiological findings in MDS that underpin experimental
trials of immune suppression, cytokine regulation and
Summary and Future Scientific Questions inhibition of angiogenesis are reviewed.
Exciting scientific studies have provided phenomenal
insights into the striking biologic and genetic heteroge- Dysplasia, Apoptosis and Cytokines
neity of MDS and related AML. As these scientific stud- Dysplasia of erythroid, granulocytic and megakaryocytic
ies lead to the further refinement of genetic models for lineages are the diagnostic hallmarks of MDS. Despite
the development of MDS, we may gain new insights that increased proliferation of the marrow, there is an increased
will ultimately lead to the design of more effective pre- rate of programmed cell death. Indeed some of the dys-
vention strategies and therapeutic regimens. Our ongo- plastic appearance may be explained by apoptotic
ing attempts to improve our knowledge, classification, changes.1,2 Kinetically the apoptosis prevails over the in-
and therapy of MDS and AML would be best served by creased proliferation, causing the peripheral cytopenia.3,4
appreciating that these disorders are linked clonal dis- Cytokines derived from unselected marrow mononuclear
eases and represent stages in the progressive transforma- cells are believed to be extrinsic factors predisposing to
tion of clonal neoplastic hematopoietic stem cells. Par- apoptosis in MDS. 5-10 In MDS many studies link
ticularly fruitful areas for future investigation include the overexpression of TNF- to cell death.7,11,12 TNF- pro-
application of genomic and proteomic technologies to duced by MDS mononuclear cells is inhibitory to both
understand alterations in the global patterns of gene ex- normal and MDS colony growth indicating that residual
pression and protein function in MDS and the alterations normal hematopoiesis can also be blocked in MDS.7,8
in these patterns during the variable progression of MDS IFN-, IL-1, and TGF-4,13 as well as undefined factors
to AML, studies of global genome methylation and aber- produced by stromal cells have also been implicated in
rant methylation in the development of MDS, continued causing apoptosis14 but their role in causing marrow fail-
studies of the complex genetic predisposition to MDS ure is not well established. The identification of TNF-
and how such genetic predispositions and environmental as a key cytokine in cell death regulation and the increased
exposures contribute to disease etiology and pathogen- susceptibility of MDS cells to TNF- is the basis for sev-
esis, studies of the adhesive interactions of hematopoi- eral clinical trials of TNF- inhibitors.4
etic stem cells and their underlying stroma and how these
interactions are perturbed in MDS, and the role of neo-
angiogenesis in the development of MDS. *Hematology Branch, National Heart, Lung and Blood Institute,
National Institutes of Health, 9000 Rockville Pike, Bldg. 10 Room
7C103, Bethesda MD 20892-0003

Hematology 2000 117


Immune Dysfunction in MDS karyocytes, and investigators have found an increased
Hamblin and others have pointed out an association of density of blood vessels and an association of increasing
MDS with an autoimmune process, noting the occurrence marrow vascularity with leukemic transformation.28,29
of autoantibody production, and monoclonal gammopathy This is the basis for using angiogenesis inhibitors to cor-
in some patients with MDS.15 The occasional finding of rect abnormalities in MDS and possibly to retard disease
T cell clonality in MDS has been interpreted as evidence progression.
of T cell involvement in the stem cell disorder16; how-
ever, more recent evidence suggests that clonal T cell A Model of Pathophysiological Changes of MDS
expansion is a feature of autoimmunity: we have observed A hypothetical model of MDS pathophysiology and the
a high frequency of T cell receptor (TCR) V- repertoire role of experimental treatments is presented in Figure
skewing in MDS patients indicating the presence of a 3.1 Transformation of normal stem cells induces danger
persisting clonally-expanded T cell population, charac- signals or antigenic change and hence an autoimmune T
teristic of an autoimmune process. Interestingly, it is not cell response directed against the marrow. Both MDS and
uncommon to find MDS in association with T-cell large normal marrow cells at various stages of differentiation
granular lymphocyte (T-LGL) leukemia, a condition char- are directly inhibited by CD8+ CTL causing varying de-
acterized by oligoclonal or clonal T cell expansions aris- grees of stem cell failure. The MDS cells may be rela-
ing in a background of autoimmune disease.17 MDS also tively resistant to this immune attack than normal stem
shares some of the features of acquired aplastic anemia cells, and consequently the abnormal immune environ-
(AA), a disease with an established autoimmune pedi- ment may provide selective pressure in favor of the ab-
gree.18 Both in AA and MDS, plasma TNF- and IFN- normal cells.2 T cell expansions are clonal and can be-
levels are high and T cell-mediated myelosuppression come autonomous in some individuals as illustrated by
occurs.19 Three groups studying MDS identified T cells the coincidence of MDS with T cell LGL.3 The persist-
inhibitory to autologous granulocyte20, 21 or erythroid22 ing autoimmune attack results in chronic overproduction
colony growth. These observations strongly suggest that, of pro-apoptotic cytokines, especially TNF-. This af-
as in AA, an autoimmune T cell-mediated myelosuppres- fects cells differentiated at or beyond the CD34+ stage
sion contributes to the cytopenia of MDS. The use of of differentiation and may contribute to a dysplastic mor-
immunosuppressive treatment to restore marrow func- phology and increased apoptosis in the marrow. Despite
tion in patients with AA has been the stimulus to con- the increased cell proliferation in MDS, the marrow fails
sider similar immunosuppressive treatment in MDS.23- to export sufficient cells into the blood because intramed-
25,32
Evidence for immune-based myelosuppression in ullary apoptosis mechanism prevails over proliferation.
MDS is summarized in Table 2. Increased TNF- levels lead to changes in the MDS cells:
increased FAS, downregulation of Fap-1, and an increase
Marrow Microenvironment and Angiogenesis in caspases causing apoptosis. Non-hematopoietic cells
In MDS the marrow shows a non-homogeneous distri- in the marrow microenvironment contribute to the pro-
bution of cell types, seen as islands of pure erythroid cells, cess in two ways: (1) stromal cells induce apoptosis by a
granulocytes, megakaryocytes26 or blast cells termed “ab- non TNF-dependent mechanism; (2) VEGF production
normally localized immature precursors” (ALIPS).27 This by the MDS cells may stimulate angiogenesis that could
appearance may reflect an underlying abnormality in the favor disease progression by altering the microenviron-
marrow stroma. In MDS, vascular endothelial growth ment to favor unregulated cell growth.
factor (VEGF) is strongly expressed especially in mega-
Experimental Treatments for MDS
Previous research in MDS has focused on increasing
Table 2. Evidence for an immune-mediated suppression of marrow cell proliferation and differentiation by growth
the marrow in myelodysplastic syndrome (MDS). factors and differentiating agents. Perhaps because of the
importance of inhibitory factors extrinsic to the MDS stem
• T cells inhibit MDS CFU-E
cell itself, these treatment approaches have produced only
• CD8+ cells inhibit CFU-GM
partial beneficial effects.
• Immunosuppressive agents improve cytopenia in MDS and The results of experimental treatments that affect
eliminate autosuppressive T cells
extrinsic mechanisms of cytopenia and disease progres-
• T cells are activated in MDS
sion in MDS are outlined below.
• T cells show a skewed T cell receptor V- repertoire It is important to recognize that MDS represents a
• HLA DR15 over representation in MDS and aplastic anemia diversity of subtypes with different degrees of severity
of cytopenia and different rates of progression to leuke-
Abbreviations: CFU-E, colony-forming units-erythroid; CFU-GM,
colony-forming units-granulocyte/macrophage
mia. Determining if a novel treatment has altered the natu-

118 American Society of Hematology


Figure 3. Mechanism of action of immune modulators in MDS.

Abbreviations: TNF, tumor necrosis factor; LGL, large granular lymphocytes; ATG, antithymocyte globulin

ral disease progression requires a knowledge of the rate Immunosuppressive Treatments


of progression and survival probability of MDS subtypes
with comparable prognostic features. Currently the IPSS Prednisone
provides the most widely accepted yardstick to measure Bagby et al reported in 1980 that prednisone alone im-
disease progression and survival.30 Numerical changes proved low blood counts in a minority of patients with
in cell counts are frequently used to describe responses MDS and that the response could be predicted in vitro by
to cytopenia. However, even the finding of a 50% in- enhancement of granulocyte macrophage colony-form-
crease in neutrophils or platelets is only of clinical rel- ing units (CFU-GM) growth.31 However, although ste-
evance if treatment raised the count from a level associ- roids have frequently been used in treatment, low response
ated with a high risk of infection or bleeding to a safe rates and increased risk fo infection make corticoster-
level. Furthermore, only sustained responses, not peak oids unattractive agents in MDS.
counts achieved, correlate with prolonged clinical ben-
efit. Reliable end-points of major clinical significance Antithymocyte globulin
are independence from transfusion of blood and plate- Because of the similarity of hypoplastic MDS to SAA,
lets and a recovery of neutrophils from below 500 to antithymocyte globulin (ATG) has been used occasion-
> 1000/mm3. ally to treat hypoplastic MDS with severe cytopenia. A
summary of case reports and small series reveals hema-
tological responses in 8/13 hypoplastic MDS patients.23,24
Based on these reports, unpublished observations, and
the hypothesis that a T cell-mediated process may cause

Hematology 2000 119


pancytopenia, we evaluated ATG as immunosuppressive lularity was observed and cytogenetic abnormalities,
treatment to improve marrow function in MDS.25 This present in four, persisted after treatment. Three relapsed
study, now completed, involved 61 patients. Study entry with transfusion-dependence but one regained red cell
criteria were red cell or platelet transfusion-dependence transfusion-independence after a second course of ATG.
and no concurrent treatment with immunosuppressives, Of the 40 non-responders 21 died, 15 from cytopenia and
chemotherapy or growth factors. Thirty-seven had refrac- 7 from progression to AML.
tory anemia (RA), 12 had RA with excess of blasts
(RAEB) and 10 RA with ring-sideroblasts (RARS). Mar- Factors Predicting a Response to Immunosuppression
row cellularity varied from hypo- to hyper-cellular. Most In a multivariate analysis of 82 RA, RARS or RAEB
patients (75%) had failed previous treatment with single patients treated with either ATG alone or cyclosporine
or multiple agents, including cyclosporine, amifostine, alone, younger age, shorter duration of red cell transfu-
and growth factors. The interval between diagnosis and sion dependence and the presence of HLA DRB1 15 pre-
entry to the study was 3–300 months, with a median of dicted a response to immunosuppression. In other words,
24 months. Twenty-three cases had hypoplastic MDS, to maximize the probability of response to immunosup-
seven had the PNH abnormality by flow cytometry, and pression, patients should be treated sooner rather than
23 had karyotypic abnormalities characteristic of MDS. later and this strategy appears to be particularly appro-
Patients received ATG at 40 mg/kg/day for 4 days and priate and effective in RA or RAEB patients who are
were periodically evaluated for 38 (range 20–58) months. young and/or have HLA DRB1 15. Pretreatment vari-
The main criterion for response was independence from ables associated with a response to immunosuppression
the requirement for red cell transfusions. Twenty-one are summarized in Table 3. Responses were associated
(33%) patients became red cell transfusion-independent with a significant survival benefit at 4 years (95% versus
within 8 months of treatment (median 75 days). Transfu- 38% for non-responders, p = 0.006). In the subset of 41/
sion-independence was maintained in 76% of respond- 61 patients with INT-1 IPSS given ATG, responders had
ing patients for a median of 32 months (range 20–58). 100% survival at 3 years and no disease progression ver-
Twenty-three of 41 (56%) severely thrombocytopenic sus 45% survival (p < 0.0004) and 51% probability of
patients had sustained platelet count increases between disease progression in non-responders (p = 0.02). Since
25,000–290,000/µl and 18/41 (44%) severely neutropenic we previously found correlation between response to ATG
patients achieved sustained neutrophil counts > 1000/µl. treatment and normalization of the T cell repertoire, we
At last follow-up 39 patients were alive with an actuarial studied 15 patients with MDS who received immuno-
survival of 64% at a median of 34 (range 18–72) months. suppression for biological markers of response. Abnor-
Of the 21 responders, 20 survived and one died follow- malities in the TCR V repertoire were common and
ing progression to AML (Figure 4). In the others no sig- occurred both in responders and non-responders, render-
nificant alteration in the bone marrow appearance or cel- ing V analysis of no prognostic value.

Cyclosporine
Jonasova and co-workers reported a
high rate of hematological responses
following cyclosporine treatment in
MDS. Sixteen patients with RA and one
with RAEB were given standard doses
of cyclosporine for 5–31 months. Sub-
stantial hematological responses were
observed,32 mostly occurring around 3
months. Transfusion independence was
achieved in all 12 patients requiring red
cells before cyclosporine administra-
tion, and significant increases in leuco-
cyte and platelet counts also occurred.
Responses occurred in RAEB as well
as RA patients and in hyper- or normo-
cellular MDS as well as in hypoplastic
MDS (6/8 and 7/9 responders respec-
tively). Of six patients with abnormal
Figure 4. Survival of 61 patients with myelodysplastic syndrome given
antithymocyte globulin (ATG) treatment.
karyotype, three responded (all were

120 American Society of Hematology


Table 3. Pretreatment variables correlating with a response to immunosuppression.

Significant Significant
Correlation in Correlation in
Univariate Multivariate
Analysis Analysis
Favorable Factors (p < 0.05) (p < 0.05) Odds in Multivariate Predictive Model

Younger age yes yes 2X less likely to respond with each 5 year increase in age
Shorter duration of red cell transfusion yes yes 1.5X less likely to respond with each 3 month increase
dependence (RCTD) in RCTD
Positive for HLA DRB1 15 yes yes 9.9X more likely to respond if HLA DRB1 15 positive
Decreased cellularity of bone marrow yes no
Increased number of cytopenias yes no
< 5% marrow blasts yes no
Normal cytogenetics yes no
PNH present by flow yes no
Low IPSS score no no

Table 4. Ongoing trials of immunosuppression in myelodysplastic syndromes.

Site Type of Immunosuppression Study Design

Hanover, Germany rATG single arm (n > 20, ongoing)


Basel, Switzerland rATG +CSA randomized, multicenter (new)
London, United Kingdom rATG single arm (n = 30, stopped)
Houston, USA hATG, vs. hATG+CSA vs. hATG+fludarabine randomized (n = 21, ongoing)
Scandinavian MDS Group hATG +CSA single arm (n = 13, ongoing)
Multicenter, USA rATG, supportive care randomized, multicenter (new)
Bethesda, USA hATG +CSA single arm (new)

Abbreviations: rATG, rabbit antithymocyte globuline; hATG, horse antithymocyte globulin

5q-). These clinical studies provide further strong evi- amifostine reduces apoptotic marrow cell death follow-
dence to support an immune mechanism of marrow sup- ing chemotherapy it may also reduce apoptosis in MDS.33
pression in patients with MDS. Indeed, incubation of MDS marrow cells with amifostine
was found to improve colony growth.34 In a study reported
New trials of immunosuppressive agents in MDS by List et al, 35 15 of 18 patients with MDS given
Based on these preliminary findings, several trials evalu- amifostine had single or multilineage hematological re-
ating immunosuppressive treatment for MDS are under- sponses. These results were recently updated for 75 pa-
way in the US and in Europe (Table 4). Collectively these tients; there were 40% platelet responses, 24% ANC re-
studies should better define the potential benefit for im- sponses and 20% with > 50% reduction in need for trans-
munosuppression by comparing ATG treatment with a fusions.36 There are ongoing, not yet reported studies on
control arm of conventionally supported patients. Pro- the use of amifostine in combination with growth factors
spective randomized trials will evaluate the relative ef- and chemotherapeutic agents that should provide more
fects of different ATG types and different immunosup- information about the usefulness of amifostine.
pressive combinations in improving marrow function and
prolonging survival. Pentoxifylline
The observation that the three pro-inflammatory cyto-
Cytokine inhibition kines TNF-, TGF- and IL-1 are implicated in the in-
Amifostine is a phosphorylated organic thiol that is me- creased apoptosis of MDS4 led Raza et al to treat MDS
tabolized to intermediates with antioxidant activity. The patients with pentoxifylline (PTX), a xanthine derivative
drug has two actions: (1) It protects cells from oxidative known to interfere with the lipid-signalling pathway used
stress after exposure to cytokines including TNF-; (2) by these cytokines.37 Ciprofloxacin was added because it
it suppresses inflammatory cytokine release. Since reduced the hepatic degradation of PTX, and dexametha-

Hematology 2000 121


sone was used to further downregulate TNF- produc- had fewer blast cells in the marrow. These findings sug-
tion by reducing translation of TNF- mRNA. Eighteen gest that thalidomide can improve marrow function in
of 43 patients had hematopoietic responses that corre- some patients with MDS.
lated with a reduction in blood levels of TNF-. In a sub-
sequent study a four-drug combination of amifostine, Future directions
pentoxyfylline, ciprofloxacin and dexamethasone was We still know very little about the interaction of the im-
used to optimize the anti-cytokine effect.38 Of 29 evalu- mune system, the marrow microenvironment and the
able patients given this combination (evaluable because MDS stem cell. The antigens evoking the T cell response
they survived 12 or more weeks), 22 (76%) showed par- are unknown and the mechanism of T cell-mediated my-
tial responses, 19 with improvement in neutrophils, 11 elosuppression is not defined. The etiology of MDS re-
with a reduction in transfusion requirement or a rise in mains unclear. Similarities between MDS and AA, in-
hemoglobin, and 7 with improvements in platelet counts. cluding the response to immunosuppressive treatment,
However, these improvements were not sustained or sta- raise questions about the relationship between AA and
tistically significant at 24 weeks and are similar to those MD.20 Are they different diseases sharing a common au-
reported for amifostine alone. There were no complete toimmune pathology or are they two ends of a spectrum
responses or conversions to transfusion independence. of a marrow failure syndrome differing only in their ten-
In contrast, in a study of 14 patients given PTX and dency to evolve to acute leukemia? Provocatively, it has
ciproflovacin, Neumatis et al found a trend to lower TNF- been proposed that the abnormal cytokine milieu in AA
 levels with treatment but failed to demonstrate a hema- is the initiator of genetic instability in the aplastic stem
tological response in any patient.39 cell leading to clonal evolution and leukemia.46 Also, this
abnormal milieu may act as selective pressure in favor of
Soluble TNF- receptor (Enbrel) the abnormal MDS clone. In these scenarios immune
Excessive amounts of soluble receptor can effectively modulation at an early stage of clonal evolution might be
block the function of TNF- by competitive binding. In expected to maintain disease stability. Finally the rela-
a recent report the soluble receptor Enbrel was reported tionship between the MDS stem cell and the marrow
to be well tolerated and to reduce plasma TNF- con- microenvironment deserves further study. In this regard
centrations. However, clinical responses were modest.40 it will be important to determine whether thalidomide
exerts its beneficial effect through inhibition of angio-
Thalidomide genesis as well as through immunosuppression.
Thalidomide is an immunosuppressive drug. It switches
T helper cells from a Th1 to a Th2 cytokine profile, in- III. THERAPEUTIC ADVANCES IN MDS
hibiting production of TNF-.41 It selectively inhibits Eva Hellström-Lindberg, M.D., Ph.D.*
TNF- production by monocytes.42 The drug has also
been found to strongly inhibit angiogenesis in animal Increased biological understanding of different subtypes
experiments and in in vitro cultures of human cells.43,44 of MDS has resulted in new therapeutic alternatives for
Thalidomide has produced surprisingly substantial re- groups of patients in whom, hitherto, only conservative
sponses in patients with myeloma, possibly because of treatment was available. Achievements in the techniques
its effects on marrow angiogenesis. Both the immuno- for stem cell transplantation (SCT) have made it possible
suppression and the angiogenesis inhibition of thalido- to cure an increasing, but still small proportion of the
mide might be beneficial in MDS. Raza and colleagues patients. Allogeneic SCT results for patients in older age
have recently completed a study of thalidomide in 83 groups are constantly improving, which will increase the
patients with MDS.45 The drug was started at a dose of proportion of MDS patients eligible for this treatment. A
100 mg daily and increased to a maximum, when toler- large phase II trial suggests that a proportion of patients
ated, of 400 mg daily. Despite this approach, 26 patients with high-risk MDS with complete remission after high-
stopped treatment within 12 weeks because of intoler- dose chemotherapy may benefit from consolidation with
ance. Twenty-one of the 57 patients who continued treat- autologous stem cell transplantation. Growth factors may
ment responded, 15 showing an erythroid, 13 a platelet support hemoglobin levels and neutrophil counts in sub-
and 7 a neutrophil response, with a median time to re- groups of patients, but is an ineffective and expensive
sponse of 10 weeks. By intention-to-treat analysis, 37% treatment for others, which encourages the development
of patients became responders. Of note, eight patients of of decision models for this type of treatment. While eryth-
the erythroid responders became transfusion-independent.
There was no evidence that thalidomide favorably or
* Department of Medicine, Division of Hematology, Karolinska
unfavorably affected disease progression, but follow-up
Institutet, Huddinge University Hospital, S-141 86 Stockholm,
was short. Patients most likely to respond to thalidomide Sweden

122 American Society of Hematology


ropoietin (Epo) as monotherapy has shown efficacy almost 60%, DFS in patients > 50 years of age was be-
mainly in RA patients with low serum Epo-levels, the low 20%, mostly due to high transplant-related mortal-
combination of G-CSF + Epo may induce long-lasting ity. Increasing disease duration before transplant signifi-
normalization of hemoglobin levels, in particular in pa- cantly increased the risk for non-relapse mortality but
tients with RARS. For RA, again, new therapeutic ap- did not influence disease-free survival. A Canadian study
proaches, such as ATG (reviewed in Section II), seem reported the outcome of 60 adult patients with MDS.4
promising. Low-dose chemotherapy may improve periph- The 7-year event-free survival was 29% for all patients,
eral blood counts, but has previously been shown not to > 60% for patients with RA/RARS, 20% for patients with
improve long-term outcome. Two DNA-hypomethylating > 5% blasts, and 6% for patients in the poor cytogenetic
agents, 5-azacytidine and 2,5-deoxycytidine may how- subgroup. An update of the EBMT experience of SCT in
ever change the role of low-dose chemotherapy, since 5- 1378 patients with MDS has recently been published.5
azacytidine has been shown to prolong time to leukemic Estimated DFS and relapse risk at 3 years were both 36%
transformation or death, compared to untreated patients. for 885 patients transplanted with stem cells from matched
The IPSS scoring system is at present the most use- siblings. DFS and relapse rate in RA/RARS was 55%
ful tool to predict long-term outcome in cohorts of un- and 13%, respectively, while the corresponding figures
treated patients and should be included as a variable in for more advanced MDS was 28% and 43%. DFS in pa-
studies describing the effects of different treatment ap- tients with advanced MDS treated to CR was 44%. This
proaches.1 A new MDS classification has recently been analysis did not indicate a significantly better DFS in
proposed, but its capacity to improve the clinical charac- patients transplanted < 1 year from diagnosis.
terization and management of MDS patients is under Recently, Deeg et al reported results on 50 MDS
debate.2 patients, aged 55–66, receiving allogeneic BMT with stem
cells from matched siblings (36), unrelated volunteers
Allogeneic Transplantation (6), HLA-nonidentical family members (4), and identi-
Allogeneic bone marrow transplantation (allo-BMT) is a cal twins (4).6 The Kaplan-Meier (KM) estimate of re-
curative therapeutic option for younger patients with MDS lapse-free survival at 3 years was 39% for all patients
(Table 5). The outcome of treatment is highly dependent and 47% for patients with primary MDS transplanted with
on the selection of patients, and it is therefore difficult to stem cells from an HLA-identical sibling. As in previous
evaluate the effect of different conditioning regimens and studies, cytogenetic risk group and IPSS score were
other treatment approaches. In a recent review from Fred highly predictive for the outcome of treatment. More-
Hutchinson Cancer Research Center, the results of allo- over, conditioning regimen with cyclophosphamide +
geneic BMT in 251 patients with MDS were reported.3 targeted busulfan showed an advantage compared to other
The overall median disease-free survival (DFS) was 40% conditioning regimens. The study shows that results in
after a median follow-up of 6 years. Important predictors selected groups of older patients are beginning to im-
for long-term survival were age, morphology and cyto- prove.
genetics. While patients below 20 years of age (i.e. pedi- Although the results from allogeneic SCT for MDS
atric MDS and young secondary MDS) showed a DFS of seem to improve over time, the outcome for patients with

Table 5. Large or new allogeneic stem cell transplantation studies in myelodysplastic syndromes.

Author (Year)Ref. Age Donor Outcome


No. pts (period) M (range) Related/VUD DFS Relapse Comments

Appelbaum (1998)3 38 (1-66) 59% / 28% 40% at 6y 18% Age, IPSS highly predictive for DFS
251 (1981-96) (60% < 20y,
20% > 50y)

Nevill (1998)4 40 (15-55) 38/22 29% at 7y 42% 6% DFS in poor cytogenetic risk group
60 (1986-96)

de Witte (2000)5 not given 885/198 36% at 3 y36% (all) Relapse rate in patients with >5% blasts 42–43%,
1378 (1997) 25% (VUD) in MDS-AML 49%

Deeg (2000)6 59 (55-66) 36/6 42% at 3y 19% DFS in RA, 53%. In high cytogenetic risk group 21%
50 (1989-98)*

*Two patients transplanted 1978 and 1983


Abbreviations: DFS, disease-free survival; RA, refractory anemia; VUD, volunteer unrelated donor; IPSS, International Scoring System for
Evaluating Prognosis

Hematology 2000 123


MDS is still worse than for those with other myeloid di- within the EORTC-EBMT working group compares the
agnoses, mostly due to a high transplant-related mortal- effect of APSCT with the effect of a second consolida-
ity and relapse rate. Future studies will need to focus on tion with high-dose cytosine arabinoside. Careful stud-
optimizing pre-treatment schedules, conditioning regi- ies of residual clonality in responding patients will serve
mens and post-transplant immune-modulation. Case re- as a tool to predict the usefulness of this treatment in
ports have shown an effect of donor lymphocyte infu- individual patients. Although the relapse rate after APSCT
sion also in MDS.7 Slavin reported one patient with MDS is higher than for de novo AML, the results suggest that
who entered complete remission after non-myeloablative there is a subset of patients with MDS that may be cured
stem cell transplantation,8 and approximately 60 patients by intensive chemotherapy in combination with stem cell
with MDS and secondary leukemia have been included support.
in the protocols from Jerusalem, Seattle and NIH (Shimon
Slavin, Rainer Storb, Ghulam Mufti, and John Barrett, Intensive Chemotherapy
personal communication), and at the Huddinge transplan- The results of AML-type induction regimens in high-risk
tation unit. Complete remissions lasting for 1–2 years MDS have improved during recent years, but are still
have been observed, which encourages further investiga- poorer than for de novo AML. Estey et al compared 158
tions. patients with MDS with a cohort of patients with AML.14
The CR rates were comparable in the MDS and AML
Autologous Stem Cell Transplantation for MDS groups (62–66%), but patients with RAEB showed sig-
Conventional high-dose chemotherapy for MDS may lead nificantly shorter event-free survival (EFS) than both
to complete remission, but only to cure in extremely rare those with RAEB-t and AML. Poor prognostic karyo-
cases. For patients without a suitable donor, or with age type, length of MDS phase and age predicted for a poorer
and medical conditions making allogeneic stem cell trans- outcome to treatment. A French group compared high-
plantation unsuitable, alternative methods to maintain CR dose chemotherapy given with or without quinine.15 In
and obtain cure need to be developed. The presence of patients expressing P-glycoprotein (PGP) the response
polyclonal peripheral blood stem cells after high-dose rate to chemotherapy + quinine was 52% compared to
chemotherapy constitutes the theoretical basis for autolo- only 18% in those treated with chemotherapy only, indi-
gous stem cell transplantation,9,10 and several European cating that one important reason for the lower CR rates
groups have evaluated the effect of ABMT/APSCT in in MDS is functional drug resistance. Bernasconi et al
high-risk MDS.11,12 Encouraged by early findings that randomized 105 patients to chemotherapy, with or with-
DFS seemed to enter a plateau phase approximately 3 out G-CSF.16 The G-CSF-treated group showed a better
years after transplantation, more studies have followed. response rate (74% vs 52%, p < 0.05), but no effect on
The EBMT and EORTC working groups have conducted long-term survival could be observed unless an allo-BMT
a large phase II trial, in which 184 evaluable patients with was given. In another study, 112 patients with RAEB-t,
high-risk MDS and AML following MDS were given MDS-AML and secondary AML (median age 58 years,
induction therapy to obtain CR. In CR, patients with a range 22–75) were treated with intensive chemotherapy
histocompatible sibling donor were candidates for allo- in combination with G-CSF.17 Overall response rate was
geneic transplantation, while the remaining patients were 62%, but median duration of response was only 8 months.
planned for autologous stem cell transplantation after DFS was 15% at 28 months. The CR rate was higher in
consolidation therapy and stem cell harvest (Th. de Witte patients < 60 years (68% vs 55%), and patients who ob-
et al, submitted). The complete remission rate was 54%, tained CR showed a DFS of 25% at 36 months. Outcome
with a median remission duration of 12 months, and DFS was better compared with a previous trial in which pa-
was 29% at 4 years. Thirty-five of 57 patients without a tients were treated with the same chemotherapy, but with-
donor received APSCT in first CR, and 13 of these (37%) out G-CSF.
were in continuous complete remission at follow-up. The Topotecan, a topoisomerase I inhibitor, has been used
benefit of APSCT compared to conventional high-dose as single therapy in high-risk MDS. In a cohort of 47
chemotherapy was also supported by a study in which patients with RAEB/RAEB-t and CMML, the CR rate
184 patients from the EORTC-EBMT trials were com- was 28%, with a response rate in previously untreated
pared with 216 patients from M.D. Anderson Cancer patients of 31–38%.18 Toxicity, however, was significant
Research Center receiving only combined chemo- with 19% of patients dying during induction treatment.
therapy.13 Preliminary results indicate a possible benefit In a recent review by Estey, the effects of topotecan, alone
of using autologous transplantation as post-remission and in combination with cytosine arabinoside (ara-C),
consolidation, since the survival curve of the transplanted was compared with the effect of ara-C in other combina-
patients showed a plateau, while that of the chemotherapy- tions, such as the FLAG regimen.19 In RAEB/RAEB-t,
treated patients continued to decline. An ongoing study but not in CMML, ara-C seemed to add to the effect of

124 American Society of Hematology


topotecan as single therapy. Ara-C + topotecan did not nia. Other advantages are the extremely low cost of the
show any overall benefit compared to high-dose ara-C drug and its simple administration.
alone, but in patients with high-risk (5/7) chromosomal
aberrations the combination of ara-C + topotecan showed 5-azacytidine and 5–aza-2´-deoxycytidine
better results than both topotecan alone, high-dose ara- Several trials have investigated the effect of low doses of
C, and the FLAG regimen. None of these differences two DNA hypomethylating pyrimidine analogues, 5-
translated into a difference in survival. azacytidine (5-aza) and 5-aza-2´-deoxycytidine (DAC),
Thus, high-dose results are slowly improving over in high-risk MDS. In spite of the similar structure of these
time but long-term survival is still very poor for the ma- drugs, their clinical effects differ in several ways. In phase
jority of patients. It is, however, important to continue to II studies, treatment with 5-aza led to improved periph-
aim for better response rates, since CR is the starting point eral blood values and reduction of bone marrow blasts in
for curative approaches. 49–54% of the patients.26 Inspired by this relatively high
response rate and moderate toxicity, 5-aza was evaluated
Low-Dose Chemotherapy by CALGB in a large randomised phase III study in which
Non-curative treatments must aim at improving quality 5-aza, given subcutaneously 7/28 days, was studied
of life and reducing morbidity, but they may also carry against observation in a cross-over model.27 The overall
hope for improved survival and reduced risk of leukemic response rate was 63% (6% CR, 10% PR, 47% improved).
transformation. Low-dose chemotherapy may improve This is not better than for other chemotherapeutic com-
peripheral blood values and reduce blast counts, and can pounds, but the median time to leukemic transformation
be an effective and inexpensive alternative for certain or death was 22 months in the 5-aza arm versus 12 months
patients with MDS. for those on supportive care (p = 0.003). Median sur-
The drug that has been most extensively used in low vival for 5-aza and observation was 18 and 14 months,
doses is ara-C. Low-dose ara-C may induce complete or respectively (p = 0.1). Moreover, treatment-related mor-
partial remissions in approximately 30% of patients with tality was low, quality of life was enhanced in respond-
RAEB, RAEB-t and MDS-AML.20,21 However, the only ing patients, and toxic side effects few.28 This promising
prospective randomized phase III trial comparing low- study suggests that 5-aza may alter the natural course of
dose ara-C with supportive care failed to show a differ- MDS. However, confirmatory studies are warranted.
ence in survival between the two alternatives.22 More- DAC, given intravenously, has been evaluated in two
over, side effects (mainly bone marrow hypoplasia and pilot trials and a large recently published multi-center
pancytopenia) can be pronounced, with therapy-related phase II trial, in which primary endpoints were response
deaths having a frequency of 7–19%.20 The addition of rate and toxicity.29 The overall response rate was 49%,
growth factors, mostly GM-CSF, to ara-C has not been with 64% response in patients with an IPSS high-risk
successful.23 Based upon a large phase II trial, pretreat- score. The actuarial median survival time from start of
ment bone marrow cellularity, chromosome aberrations therapy was 15 months. Myelosuppression was common,
and ringed sideroblasts were used to formulate a predic- and 7% therapy-related deaths were reported. Noticeable
tive model for the use of low-dose ara-C in MDS and was the rapid and distinct platelet response, which oc-
MDS-AML.21 This model identified patient groups with curred after the first course in the responding patients.
a > 50% probability of response and those with no re- Compared to 5-aza, DAC seems to be more efficient in
sponse to treatment. inducing CR, it has more severe toxicity, and has not yet
been investigated in a trial with survival as endpoint. It
Melphalan is, however, likely that both 5-aza and DAC will add to
Two recently published studies have reported the effects the arsenal of clinically useful therapies for MDS.
of low-dose melphalan (2 mg/day until progression/tox-
icity or response) in patients with high-risk MDS. In the Growth factor treatment
first study, 8 of 21 patients with RAEB or RAEB-t (38%) The bone marrow of patients with RA and RARS is
achieved a complete (7) or partial (1) response with a mainly characterized by ineffective hematopoiesis. This
median survival of 27 months for CR patients and 6.5 is paralleled by reduced clonogenic growth in vitro, and
months for the rest.24 No severe side effects were ob- the presence of an increased proportion of apoptotic bone
served in any patients. Recently, these results were con- marrow precursors.30-32 Recent studies suggest that the
firmed by a European group,25 who reported a response underlying mechanisms behind apoptosis in MDS may
rate of 40% and suggested a better response in patients differ between the subtypes of MDS.33,34 Colony culture
with hypocellular MDS. The difference from other low- studies of myelodysplastic bone marrow have shown that
dose regimens seems to be that melphalan relatively fre- the impaired erythroid growth may respond to Epo alone,
quently causes improvement without preceding cytope- but that it may be further improved if Epo is combined

Hematology 2000 125


Table 6. Larger studies on erythropoietin (Epo) alone or Epo in combination with G-CSF.

Inclusion Erythroid Favorable Variables/


Author Year Treatment No. Pts. Criteria Response Comments

Hellstrom-Lindberg40 1995 Epo 205 Various, all 16%* S-Epo < 200 U/L
meta-analysis studies since 1995 (8% RARS) RA/RAEB, no transfusions
Rose41 1995 Epo 116 Hb < 80 g/L, 28%** S-Epo < 100 U/L, RA
phase II S-Epo < 500 U/L (18% red. transf)
Italian Study Group42 1998 Epo / placebo 87 Hb < 90 g/L 37% vs 11%** Response significant in RA
phase III Blasts < 10% (p = 0.007) and in non-transfused pts
Negrin44 1996 Epo + G-CSF 55 Hb < 100 g/L 48%**, duration S-Epo
phase II median 10 mo.
Hellstrom-Lindberg45 1998 Epo + G-CSF 71 Hb < 100 g/L 38%*, duration S-Epo, transfusion need
phase II 24 months
Remacha47 1999 Epo + G-CSF 32 Hb < 100 g/L 50%** Predictive model (Figure 5)
phase II S-Epo < 250 U/L
Mantovani49 2000 Epo + G-CSF 33 Blasts < 20% 61%* (12 weeks) 50% response after 2 years
80% (36 weeks)

* The definition of response required a total stop in transfusion need in transfusion-dependent patients.
** The definition of response required a 50% reduction in transfusion need in transfusion-dependent patients.
Abbreviations: S-Epo, serum Epo level; G-CSF, granulocyte colony-stimulating factor

with other growth factors.35 The ineffective hematopoie- ful to characterize groups of patients with different out-
sis and cytopenia in RA, RARS and RAEB with < 10% comes to treatment. Patients with RARS responded sig-
blasts is considered to respond poorly to chemotherapy, nificantly less well to treatment than the other subgroups
and a number of clinical trials have therefore addressed (8% versus 21% in non-RARS). The response rates in
the use of growth factors in these subtypes (Table 6). patients with S-Epo below or above 200 U/l, were 21%
and 10%, respectively, and 10% in patients with pretreat-
GM-CSF and G-CSF ment transfusion need. Rose et al treated 116 patients
GM-CSF and G-CSF are both effective in increasing pe- with Epo.41 The response rate was 28%, S-Epo levels <
ripheral neutrophil counts in MDS. In a preliminary re- 100 U/l predicited for a better response, and patients with
ported clinical phase III trial, patients were randomised RA had a better response than those with RARS (39%
to either GM-CSF or observation.36,37 Granulocyte counts versus 17.5%). The Italian cooperative group performed
were significantly increased, while hemoglobin levels, a placebo-controlled randomized study, which showed
survival, and the incidence of leukemic transformation that Epo significantly improved hemoglobin levels in
were not affected. G-CSF, used in phase II studies, showed MDS. However, the response was significant only in pa-
similar effects on neutrophil counts, and favorable long- tients with RA and in patients who did not need transfu-
term results with maintained response for up to 16 months sion before treatment.42 Finally, Hast et al reported a se-
were reported.38 A randomized study between G-CSF and ries of 18 patients responding to Epo, who continued on
observation showed a significant G-CSF-induced increase maintenance treatment and showed a median response
in neutrophil counts but no effect on hemoglobin lev- duration of 15 months. The dose of Epo could be safely
els.39 After correction for risk factors, survival or evolu- reduced or withdrawn, since the majority of patients who
tion to AML was not significantly influenced by treat- then relapsed with anemia responded well to reintroduc-
ment. Both these randomised studies showed that G-CSF, tion of Epo or to an increase of the dose.43
as well as GM-CSF, had an overall negative effect on
platelet counts. Thus, there is at present no evidence that Erythropoietin in combination with other
supports the use of G-CSF and GM-CSF as single therapy growth factors
in MDS. Several clinical trials have studied the combination of
Epo and G-CSF, GM-CSF or IL-3. The response rate to
Erythropoietin Epo + G-CSF has been shown to be around 40% in MDS
Erythropoietin (Epo) may improve the anemia in MDS. patients with < 10% bone marrow blasts, i.e. twice as
All 17 studies (205 patients) published before 1995 were much as for Epo alone. A large US phase II study and the
included in a meta-analysis, which showed an overall trials within the Scandinavian MDS Group have demon-
response rate of 16%.40 Information about FAB group, strated evidence of a clear synergistic effect between the
transfusion need and serum Epo levels (S-Epo) were use- drugs in vivo.44,45 The US study showed that 50% of the

126 American Society of Hematology


patients with a response lost it when G-CSF was with- cant number of patients showed late responses (12–36
drawn and regained it when G-CSF was reintroduced, weeks after start of treatment).
and the Scandinavian study showed that addition of G- These studies indicate a central role for G-CSF in
CSF could induce erythroid responses in Epo-resistant the treatment of RARS, but the mechanism has not yet
patients. The synergistic effect was most pronounced in been clarified. Histopathological studies have shown that
patients with RARS, since this subgroup responded patients who respond to treatment have a significant re-
poorly to Epo alone and showed the best response to the duction of apoptotic cells in the bone marrow. Thus, it
combination (around 50%). A study of predictive factors seems likely that growth factors affect one of the major
for a response to treatment identified S-Epo and the level pathogenetic mechanisms in MDS, ineffective hemato-
of pre-treatment transfusion need as significant variables poiesis.32 Recent laboratory studies49 have shown that the
and these were combined in a predictive model for treat- erythroid apoptosis in RARS is initiated at the CD34+ level,
ment with G-CSF + Epo (Figure 5).46 The value of this is mediated by increase caspase activity, but not by endog-
model was later confirmed in independent patient mate- enous stimulation of the Fas receptor, and can be signifi-
rial from a Spanish study,47 and in a third prospective cantly reduced by G-CSF incubation of CD34+ cells.
study within the Scandinavian MDS Group (data not re- GM-CSF and EPO have also been used in combina-
ported). This study included 52 evaluable patients from tion as treatment for MDS.50-52 One pilot study showed
the high (n = 31) and the intermediate (n = 21) predictive that the combination could induce erythroid responses in
groups (for explanation, see Figure 5). The response rate patients not responding to Epo alone. A phase II trial us-
was 61.3% in group 1 and 14.3% in group 2 (p = 0.004), ing GM-CSF + Epo resulted in an erythroid response rate
and no other pre-treatment variables improved the model. of 34.6%, but the authors questioned whether a synergis-
Quality of life (QLQ 30) improved in patients with a re- tic effect in vivo was obvious. Finally, a randomized phase
sponse to treatment (p < 0.05 for fatigue and global QOL). II trial showed that GM-CSF + Epo was more effective
A simplified and clinically useful decision model will be in reducing transfusion need than GM-CSF + placebo, in
developed based on these results. While waiting for the patients with S-Epo < 500 U/l. Thus, there is some evi-
results of ongoing prospective randomized studies, we dence that the combination of GM-CSF + Epo may also
designed a case-control study in which 135 patients offer an advantage over Epo alone, but the experience is
treated with G-CSF + Epo were compared with control more limited than with to G-CSF + Epo, and GM-CSF
cases from the data base generated by the International has more side effects. Lastly, Epo has been combined
MDS Risk Analysis Workshop (also used to create the with IL-3, but in spite of promising in vitro data, clinical
IPSS score). The median duration of response to G-CSF results have not encouraged a continued development of
+ Epo (defined as transfusion independence or normal this treatment.53
hemoglobin levels) was 10 months (range 0–72+) in the
whole group and 25 months in patients with RARS. Concluding Remarks
Lastly, a recently published study showed a response rate To improve treatment for patients with MDS, better tools
of 61% in a cohort of 33 patients with MDS.48 A signifi- will be required to define the underlying pathogenetic
mechanisms in individual patients. It is
thus important to estimate the risk for leu-
kemic transformation, the degree of inef-
fective hematopoiesis, signs of immune-
mediated myelosuppression, and overall
prognosis including age and performance
status, before making a therapeutic deci-
sion. Recent therapeutic advances have
made it clear that new biological informa-
tion in combination with statistically de-
veloped predictive models may help us to
better select patients for different thera-
peutic options and to develop new treat-
ment modalities. The future thus lies in
developing new insights into the biology
> of MDS, in new therapeutic approaches
and well-conducted clinical trials, and in
the search for better tools to use existing
Figure 5. Predictive model for treatment with G-CSF + erythropoietin for the
anemia of myelodysplastic syndrome (MDS).
therapeutic options.

Hematology 2000 127


REFERENCES dysplastic hematopoiesis: cytogenetic and prognostic signifi-
cance. Leukemia. 1996;10:946.
21. Harris NL, Jaffe ES, Diebold J, et al. The World Health
I. Biologic and Genetic Features of the MDS Organization classification of the hematologic malignancies
1. Heaney ML, Golde DW. Myelodysplasia. N Engl J Med. report of the clinical advisory committee meeting, Airlie House,
1999;340:1649-1660. Virginia, November 1997. J Clin Oncol. 1999;17:3835-3849.
2. Rosenfeld C, List A. A hypothesis for the pathogenesis of 22. He LZ, Triboli C, Rivi R, et al. Acute leukemia with
myelodysplastic syndromes: implications for new therapies. promyelocytic features in PML/RAR transgenic mice. Proc
Leukemia. 2000;14:2-8. Natl Acad Sci USA. 1997;94:5302-5307.
3. Classification of acute leukemia. Ann Intern Med. 1977;87:740- 23. Warrell RP, Barrett AJ, Pandolfi PP, Willman CL. Acute
753. myelocytic leukemia: I. Transgenic models of acute myeloid
4. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the leukemias. In McArthur JR, Schechter GP, Platt OS, eds.
classification of the myelodysplastic syndromes. Br J Haematol. Hematology–1997: Education Program of the American Society
1982;51:189-99. of Hematology. 1997;120-137.
5. Aul C, Cattermann N, Schneider W. Age-related incidence and 24. Okuda T, Cai Z, Yang S, et al. Expression of a knocked-in
other epidemiological aspects of myelodysplastic syndromes. Br AML1-ETO leukemia gene inhibits the establishment of normal
J Haematol. 1993;82:358-362. definitive hematopoiesis and directly generates dysplastic
6. Ries LAG, Kosary CL, Hankey BF, Miller BA, Clegg LX, hematopoietic progenitors. Blood. 1998;91:3134-3143, 1998.
Edwards BK, eds. SEER Cancer Statistics Review, 1973-1996, 25. Castilla LH, Wijmenga C, Wang Q, et al. Failure of embryonic
National Cancer Institute. NIH Pub. No. 99-2789. Bethesda, hematopoiesis and lethal hemorrhages in mouse embryos
MD, 1999. heterozygous for a knocked-in leukemia gene CBFb-MYH11.
7. Head DR. Revised classification of acute myeloid leukemia. Cell. 1996;87:687-696.
Leukemia. 1996;10:1826. 26. Albitar M, Beran M, O’Brien, et al. Differences between
8. Rowley JD, Alimena G, Garson MO, et al. A collaborative study refractory anemia with excess blasts in transformation and acute
of the relationship of the morphologic type of acute non- leukemia (letter). Blood. 2000;96:372-374.
lymphocytic leukemia with patient age and karyotype. Blood. 27. Wattel E, De Botton S, Luc Lai J, et al. Long term follow up of
1982;59:1013. de novo myelodysplastic syndromes treated with intensive
9. Dastugue N, Payen C, Lafage-Pochitaloff, MM, et al. Prognostic chemotherapy: incidence of long term survivors and outcome of
significance of karyotype in adult de novo acute myeloid partial responders. Br J Haematol. 1997;98:983-981.
leukemia. Leukemia. 1995;9:1291. 28. Estey E, Thall P, Beran M, Kantarjian H, Pierce S, Keating M.
10. Leith CP Leith, KJ Kopecky, J et al. Acute myeloid leukemia in Effect of diagnosis (refractory anemia with excess blasts
the elderly: Assessment of multidrug resistance (MDR1) and refractory anemia with excess blasts in transformation or acute
cytogenetics distinguishes biologic subgroups with remarkably myeloid leukemia) on outcome of AML-type chemotherapy.
distinct responses to standard chemotherapy: A Southwest Blood. 1997;90:2969-2977.
Oncology Group Study. Blood. 1997;89:3323. 29. Greenberg P, Anderson J, de Witte T, et al. Problematic WHO
11. Willman CL. The prognostic significance of the expression and re-classification of myelodysplastic syndromes: commentary
function of multidrug resistance transporter proteins in acute from the International MDS Study Group (letter). J Clin Oncol.
myeloid leukemia: Studies of the Southwest Oncology Group In press.
Leukemia Research Program. Semin Hematol. 1997;34(Suppl. 30. Greenberg P, Cox C, LeBeau MM, et al. International prognostic
5):25-33. scoring system for evaluating prognosis in myelodysplastic
12. Lancet JE, Willman CL, Bennett JM. Acute myelogenous syndromes. Blood. 1997;89:2079-2088.
leukemia in the elderly. Hematol/Oncol Clin N Am. 31. Johansson B, Mertens F, Mitelman F. Primary versus secondary
2000;14:251-267. neoplasia associated chromosomal abnormalities—balanced
13. Rossi G, Pelizzari AM, Bellotti, et al. Cytogenetic analogy rearrangements vs. genomic imbalance? Genes Chrom Cancer
between myelodysplastic syndrome and acute myeloid leukemia 1996;16:155.
of elderly patients. Leukemia. 2000;14:636-641. 32. Pedersen-Bjergaard J, Rowley JD. The balanced and unbalanced
14. Carroll M, Tomasson MH, Barker GF, et al. The tel/platelet- chromosome aberrations of acute myeloid leukemia may
derived growth factor beta receptor (PDGF) fusion in chronic develop in different ways and may contribute differently to
myelomonocytic leukemia is a transforming protein that self- malignant transformation. Blood. 1994;83:2780.
associates and activates PDGF R kinase-dependent signaling 33. D’Andrea AD and Grompe M. Molecular biology of Fanconi
pathways. Proc Natl Acad Sci USA. 1996;93:14845-14850. anemia: implications for diagnosis and therapy. Blood.
15. Third MIC Cooperative Study Group. Morphologic, immuno- 1997;90:1725.
logic, and cytogenetic working classification of the primary 34. Luna-Fineman S, Shannon KM, Atwater SK, et al.
myelodysplastic syndromes and therapy-related myelodysplasia Myelodysplastic and myeloproliferative disorders of childhood:
and leukemias. Cancer Genet Cytogenet. 1988;32:1. a study of 167 patients. Blood. 1999;93:459-466.
16. Mecucci M. Molecular features of primary MDS with cytoge- 35. Shannon KM, Turhan AG, Chang SS, et al. Familial bone
netic changes. Leuk Research. 1998;22:293. marrow monosomy 7. Evidence that the predisposing locus is
17. Busque L, Gilliland DG. X-inactivation analysis in the 1990s: not on the long arm of chromosome 7. J Clin Invest.
promise and potential problems. Leukemia. 1998;12:128-135. 1989;84:984-989.
18. Raskind WH, Steinmann L, and Najfeld V. Clonal development 36. Luna-Fineman S, Shannon KM, and Lange BJ. Childhood
of myeloproliferative disorders: clues to hematopoietic monosomy 7: epidemiology, biology, and mechanistic implica-
differentiation and multistep pathogenesis of cancer. Leukemia. tions. Blood. 1995;85:1985-99.
1998;12:108-116. 37. Bollag G, Clapp DW, Shih S, Adler F, Zhang YY, Thompson P,
19. Kuriyama K, Tomonaga M. Matsuo T, et al. Poor response to Lange BJ, Freedman MH, McCormick F, Jacks T, Shannon KM.
intensive chemotherapy in de novo myeloid leukemia with Loss of NF1 results in activation of the RAS pathway and leads
trilineage myelodysplasia. Br J Haematol. 1994;86:767-773. to aberrant growth in hematopoietic cells. Nat Genet.
20. Gahn B, Haase D, Unterhalt M, et al. De novo AML with 1996;12:144.

128 American Society of Hematology


38. Ho CY, et al. Linkage of a familial platelet disorder with a novel cDNAs. Genomics. 2000;15:26-34.
propensity to develop myeloid malignancies to human chromo- 58. Horrigan SK, Arbieva ZH, Yan Xie H, et al. Delineation of a
some 21q22.1-22.2. Blood. 1996;87:5218-5224. minimal interval and identification of 9 candidates for a tumor
39. Song W-J, Sullivan MG, Legare RD, et al. Haploinsufficiency of suppressor gene in malignant myeloid disorders on 5q31. Blood.
CBFA2 (AML1) causes familial thrombocytopenia with 2000;95:2372-2377.
propensity to develop acute myelogenous leukemia (FDP/ 59. Johnson EJ, Scherer SW, Osborne L, et al. Molecular definition
AML). Nat Genet. 1999;23:166-174. of a narrow interval at 7q22.1 associated with MDS. Blood.
40. Horwitz M, Benson KF, Feng-Qian L, et al. Genetic heterogene- 1995;86:270a, 1995.
ity in familial acute myelogenous leukemia: evidence for a 60. Le Beau MM, Espinoza R, Davis EM, et al. Cytogenetic and
second locus at chromosome 16q21-23.2. Am J Human Genet. molecular delineation of a region of chromosome 7 commonly
1997;61:873-881. deleted in malignant myeloid disease. Blood. 1996;88:1930.
41. Olopade O, Roulston D, Baker T, et al. Familial myeloid 61. Pederson B. Anatomy of the 5q- deletion: different sex ratios
leukemia associated with loss of the long arm of chromosome 5. and deleted 5q bands in MDS and AML. Leukemia.
Leukemia. 1996;10:669. 1996;10:1883-1890.
42. Perera FA. Environment and cancer: who are susceptible? 62. Foucar K. Bone Marrow Pathology. Ch. 12 Myelodysplasia.
Science. 1997;278:1068. ASCP Press. 1995.
43. Rothman N, Smith MT, Hayes RB, et al. Benzene poisoning, a 63. Bollag G, Clapp DW, Shih S, et al. Loss of NF1 results in
risk factor for hematological malignancy, is associated with the activation of the RAS pathway and leads to aberrant growth in
NQ01 609 C➝T mutation and rapid fractional excretion of hematopoietic cells. Nat Genet. 1996;12:144.
chlorzoxazone. Cancer Res. 1997;57:2839. 64. Kaneko H, Misawa S, Horiike S, et al. TP53 mutations emerge
44. Larson RA, Wang Y, Banerjee M, et al. Prevalence of the at an early phase of Myelodysplastic syndrome and are
inactivating 609C➝T polymorphism in the NAD(P)H: quinone associated with complex chromosomal abnormalities. Blood.
oxido-reductase (NQ01) gene in patients with primary and 1995;85:2189-2193.
therapy-related myeloid leukemias. Blood. 1999;94:803-807. 65. Padua RA, Guinn BA, Al-Sabah A, et al. RAS, FMS, and p53
45. Wiemels JL, Pagnamenta A, Taylor GM. et al. A lack of mutations and poor clinical outcome in myelodysplasias: a 10-
functional NAD(P)H:quinone oxireductase allele is selectively year follow-up. Leukemia. 1998;12:887-892.
associated with pediatric leukemias that have MLL fusions. 66. Lai JL, Preudhomme C, Zandecki M, et al. Myelodysplastic
Cancer Res. 1999;59:4095-4099. syndromes and acute myeloid leukemia with 17p deletion. An
46. Chen H, Sandler DP, Taylor JA, et al. Increased risk for entity characterized by specific dysgranulopoiesis and a high
myelodysplastic syndromes in individuals with glutathione incidence of p53 mutations. Leukemia. 1995;9:370.
transferase theta 1 (GSTT1) gene defect. Lancet. 1996;347:295. 67. Yokota S, Kiyoi H, Nakao M, et al. Internal tandem duplication
47. Preudhomme C, Nisse C, Hebbar M, et al. Glutathione S of the FLT3 gene is preferentially seen in acute myeloid
transferase theta 1 gene defects in myelodysplastic syndromes leukemia and Myelodysplastic syndrome among various
and their correlation with karyotype and exposure to potential hematologic malignancies. A study of a large series of patients
carcinogens. Leukemia. 1997;11:1580. and cell lines. Leukemia. 1997;11:1605-1609.
48. Atoyebi W, Kusec R, Fidler C, et al. Glutathione S-transferase 68. Uchida T, Kinoshita T, Nagai H, et al. Hypermethylation of the
gene deletions in myelodysplasia. Lancet. 1997;349:1450-51. p15INK4b gene in myelodysplastic syndromes. Blood.
49. Roulston D, Espinosa R, Nucifora G, et al. CBFA2 (AML1) 1997;90:1403-1409.
translocations with novel partner chromosomes in myeloid 69. Quesnel B, Guillerm GM, Vereecque R, et al. Methylation of the
leukemias: association with prior therapy. Blood. 1998;92:2879- p15INK4b gene in Myelodysplastic syndromes is frequent and
2885. acquired during disease progression. Blood. 1998;91:2985-2990.
50. Osato M, Asou N, Abdalla, E, et al. Biallelic and heterozygous 70. Hoefsloot LH, Amelsvoort MP, Broeders LCAM, et al.
point mutations in the runt domain of the AML1/PEBP2aB gene Erythropoietin-induced activation of STAT5 is impaired in the
associated with myeloblastic leukemias. Blood. 1999;93:1817- Myelodysplastic syndromes. Blood. 1997;89:1690-1700.
1824. 71. Raza A, Gezer S, Mundle S, et al. Apoptosis in bone marrow
51. Willman CL. Molecular evaluation of the acute myeloid biopsy samples involving stromal and hematopoietic cells in 50
leukemias. Semin Hematol. 1999;36:390-400. patients with Myelodysplastic syndrome. Blood. 1995;86:268-
52. Nisse C, Lorthois C, Dorp V, et al. Exposure to occupational and 275.
environmental factors in myelodysplastic syndromes. Prelimi- 72. Mundle SD, Venugopal P, Cartlidge JD, et al. Indication of an
nary results of a case-control study. Leukemia. 1995;9:693-699. involvement of interleukin-1b converting enzyme-like protease
53. West RR, Stafford DA, White, AD, et al. Cytogenetic abnormali- in intramedullary apoptotic cell death in the bone marrow of
ties in the myelodysplastic syndromes and occupational or patients with myelodysplastic syndromes. Blood. 1996;88:2640-
environmental exposure. Blood. 2000;95:2093-2097. 2647.
54. Le Beau MM, Espinoza III R, Neuman WL, et al. Cytogenetic 73. Voura EB, Billia F, Iscove NN et al. Expression mapping of
and molecular genetic delineation of the smallest commonly adhesion receptor genes during differentiation of individual
deleted region of chromosome 5 in malignant myeloid diseases. hematopoietic precursors. Exp Hematol. 1997;25:1172-1179.
Proc Natl Acad Sci USA. 1993;90:5484. 74. Potocnik AJ, Brakenbusch C, Fassler R. Fetal and adult
55. Willman CL, Sever CE, Pallavicini MG, et al. Deletion of IRF- hematopoietic stem cells require b1 integrin function for
1, mapping to chromosome 5q31.1 in human leukemia and pre- colonizing fetal liver, spleen, and bone marrow. Immunity.
leukemic myelodysplasia. Science 1993;259:968. 2000;12:653-663.
56. Boultwood J, Fidler C, Lewis S, et al. Molecular mapping of
uncharacteristically small 5q deletions in two patients with the II. Immune Mechanisms and Modulation in MDS
5q- syndrome: delineation of the critical region on 5q and 1. Hatfill SJ, Fester ED, Steyler JG. Apoptotic megakaryocyte
identification of the breakpoint. Genomics. 1994;19:425-432. dysplasia in myelodysplastic syndrome. Hematol Pathol.
57. Boultwood J, Fidler C, Strickson AJ, et al. Transcription 1993;8:87-93.
mapping of the 5q- syndrome critical region: cloning of two 2. Clark DM, Lampert I. Apoptosis is a common histopathological
genes and sequencing, expression, and mapping of a further six finding in myelodysplasia; The correlate of ineffective

Hematology 2000 129


hematopoiesis. Leuk Lymphoma. 1990;2:415-418. receptor V beta profiles. Br J Haematol. 1998;102:1314-1322.
3. Raza A, Gezer S, Mundle S, et al. Apoptosis in bone marrow 22. Sugarawa T, Endo K, Shishido T, et al. T-cell mediated
samples involving stromal and hematopoietic cells in 50 patients inhibition of erythropoiesis in myelodysplastic syndromes. Am J
with myelodysplastic syndromes. Blood. 1995;86:268-276. Hematol. 1992;41:304-305.
4. Raza A, Mundle S, Shetty V, et al. Novel insights into the 23. Tichelli A, Gratwohl A, Wuersch A, et al. Antilymphocyte
biology of myelodysplastic syndromes: excessive apoptosis and globulin for myelodysplastic syndrome? Br J Haematol.
the role of cytokines. Int J Hematol. 1996;63:265-278. 1988;68:139-140
5. Lepelley P, Grardel N, Erny O, et al. Fas/APO-1 (CD95) 24. Biesma DH, Van den Tweel JG, Verdonck LF, et al. Immunosup-
expression in myelodysplastic syndromes. Leuk Lymphoma. pressive therapy for hypoplastic myelodysplastic syndrome.
1998;30:307-312. Cancer. 1997;79:1548-51.
6. Bouscary D, De Vos J, Guesnu M, et al. Fas/Apo-1 (CD95) 25. Molldrem JJ, Caples M, Mavroudis D, et al. Antithymocyte
expression and apoptosis in patients with myelodysplastic globulin for patients with myelodysplastic syndrome. Br J
syndromes. Leukemia. 1997;11:839-845. Haematol. 1997;99:699-705.
7. Kitagawa M, Saito I, Kuwata T, et al. Overexpression of tumor 26. Mangi MH, Mufti GJ. Primary myelodysplastic syndromes:
necrosis factor (TNF)-alpha and interferon (IFN)-gamma by diagnostic and prognostic significance of immunohistochemical
bone marrow cells from patients with myelodysplastic syn- assessment of bone marrow biopsies. Blood. 1992;79:198-205.
dromes. Leukemia. 1997;11:2049-2054. 27. Tricot G, De Wolf-Peeters C, Vlietinck R, et al. Bone marrow
8. Deeg HJ, Beckham C, Loken MR, et al. Negative regulators of histology in myelodysplastic syndromes. II. Prognostic value of
hemopoiesis and stroma function in patients with myelo- abnormal localization of immature precursors in MDS. Br J
dysplastic syndrome. Leuk Lymphoma. 2000;37:405-414 Haematol. 1984;58:217-25.
9. Verhoef GEG, De Schouwer P. Ceuppens JL, Van Damme J, 28. Pruneri G, Bertolini F, Soligo D, et al. Angiogenesis in
Goossens W, Boogaerts MA. Measurement of serum cytokines myelodysplastic syndromes. Br J Cancer. 1999;81:1398-1401.
levels in patients with myelodysplastic syndromes. Leukemia. 29. Bellamy WT, Richter L, Frutiger Y, Grogan TM. Expression of
1992;2:1268-1272. vascular endothelial growth factor and its receptors in hemato-
10. Gersuk GM, Beckham C, Loken MR, Deeg HJ. A role for poietic malignancies. Cancer Res. 1999;59:728-733.
tumour necrosis factor-alpha, Fas and Fas-Ligand in marrow 30. Greenberg PL, Cox C, Le Beau M, et al. International survival
failure associated with myelodysplastic syndrome. Br J score for evaluating prognosis in myelodysplastic syndrome.
Haematol. 1998;103:176-188. Blood. 1997;89:2079-2088.
11. Peddie CM, Wolf CR, Mclellan LI, Collins AR, Bowen DT. 31. Bagby GC Jr, Gabourel JD, Linman JW. Glucocorticoid therapy
Oxidative DNA damage in CD34+ myelodysplastic cells is in the preleukemic syndrome (hemopoietic dysplasia):
associated with intracellular redox changes and elevated plasma identification of responsive patients using in-vitro techniques.
tumour necrosis factor-a concentration. Br J Haematol. Ann Intern Med. 1980;92:55-58.
1997;99:625-631. 32. Jonasova A, Neuwirtova R, Cermak J, et al. Cyclosporin A
12. Ganser A, Ottmann OG, Seipelt G, et al. Effect of long-term therapy in hypoplastic MDS patients and certain refractory
treatment with recombinant human interleukin-3 in patients with anaemias without hypoplastic bone marrow. Br J Haematol.
myelodysplastic syndromes. Leukemia. 1993;7:696-701. 1998;100:304-309.
13. Mundle SD, Venugopal P, Cartlidge JD, et al. Indication of an 33. List AF, Heaton R, Glinsmann-Gibson B, Capizzi RL.
involvement of interleukin-1 converting enzyme-like protease in Amifostine protects primitive hematopoietic progenitors aganist
intramedullary apoptotic cell death in the bone marrow of chemotherapy cytotoxicity. Semin Oncol. 1996;23:58.
patients with myelodysplastic syndromes. Blood. 1996;88:2640- 34. Kemp G, Rose P, Lurain J, et al. Amifostine pretreatment for
2647. protection against cyclophosphamide-induced and cisplatin-
14. Aizawa S, Hiramoto M, Hoshi H, Toyama K, Shima D, Handa induced toxicities: results of a randomized controlled trial in
H. Establishment of stromal cell line from an MDS RA patient patients with advanced ovarian cancer. J Clin Oncol.
which induced an apoptotic change in hematopoietic and 1996;14:2101.
leukemic cells in vitro. Exp Hematol. 2000;28:148-55. 35. List AF, Brasfield F, Heaton R, et al. Stimulation of hematopoie-
15. Hamblin TJ. Immunological abnormalities in myelodysplastic sis by amifostine in patients with myelodysplastic syndrome.
syndromes. Semin Hematol. 1996;33:150-162. Blood. 1997;90:3364-3369.
16. Culligan DJ, Cachia P, Whittaker J, Jacobs A, Padua RA. Clonal 36. List AF, Holmes H, Greenberg PL, Bennett JM, Oster W. Phase
lymphocytes are detectable in only some cases of MDS. Br J II study of amifostine in patients with myelodysplastic
Haematol. 1992;81:346-352. syndromes (MDS) [abstract]. Blood. 1999;94:305a
17. Dhodapkar MV, Li CY, Lust JA, Tefferi A, Phyliky RL. Clinical 37. Raza A, Venugopal P, Gezer S, et al. Pilot study of
spectrum of clonal proliferations of T-large granular lympho- pentoxifylline and ciprofloxacin with or without dexamethasone
cytes: a T-cell clonopathy of undetermined significance? Blood. produces encouraging results in myelodysplastic syndromes. In:
1994;84:1620-1627. W Hiddemann, Buchner T, Wormann J, et al, eds. Acute
18. Young NS, Maciejewski J. The pathophysiology of acquired leukemias VII: Experimental Approaches and Novel Therapies.
aplastic anemia. N Engl J Med. 1997;336:1365-12372. New York, NY: Springer Verlag; 1998:42-51.
19. Barrett AJ, Saunthararajah Y. Molldrem J. Myelodysplastic 38. Raza A, Qawi H, Lisak L, et al. Patients with myelodysplastic
syndrome and aplastic anemia; distinct entities or diseases syndromes benefit from palliative therapy with amifostine,
linked by a common pathophysiology? Semin Hematol. pentoxifylline, and ciprofloxacin with or without dexametha-
2000;37:15-29. sone. Blood. 2000;95:1580-1587.
20. Smith MA, Smith JG. The occurrence subtype and significance 39. Nemunaitis J, Rosenfeld C, Getty L, et al. Rentoxifylline and
of hematopoietic inhibitory T cells (HIT cells) in myelodyspla- ciproloxacin in patients with myelodysplastic syndrome. A
sia: an in vitro study. Leuk Res. 1991;5:597-601. phase II trial. Am J Clin Oncol. 1995;18:189-93.
21. Molldrem JJ, Jiang YZ, Stetler-Stevenson MA, et al. Haemato- 40. Raza A, Allampallam K, Shetty V, et al. Biologic and clinical
logical response of patients with myelodysplastic syndrome to response to recombinant human soluble tumor necrosis factor
antithymocyte globulin is associated with a loss of lymphocyte- receptor (Enbrel) in patients with myelodysplastic syndromes
mediated inhibition of CFU-GM and alterations in T-cell (MDS) [abstract]. Blood. 1999;94;171a.

130 American Society of Hematology


41. McHugh SM, Rifkin IR, Deighton J, et al. The immunosuppres- 11. De Witte T, van Biezen A, Hermans J, et al. Autologous bone
sive drug thalidomide induces T-helper cell type 2 (Th2) and marrow transplantation for patients with myelodysplastic
concomitantly inhibits Th1 cytokine production in mito-gen and syndrome (MDS) or acute myeloid leukemia following MDS.
antigen stimulated human peripheral blood mononuclear cell Blood. 1997:90;3853-3857.
cultures. Clin Exp Immunol. 1995;99:160-167. 12. Wattel E, Solary E, Leleu X, et al. A prospective study of
42. Sampaio EP, Sarno EN, Galilly R, Cohn ZA, Kaplan G. autologous bone marrow or peripheral blood stem cell transplan-
Thalidomide selectively inhibits tumor necrosis factor alpha tation after intensive chemotherapy in myelodysplastic
production by stimulated human monocytes. J Exp Med. syndromes. Leukemia. 1999;13:524-529.
1991;173:699-703. 13. Oosterveld M, Estey E, Muus P, et al. Chemotherapy only versus
43. Klausner JD, Freedman VH, Kaplan G. Thalidomide as an anti- chemotherapy followed by transplantation in high-risk MDS and
TNF-a inhibitor: Implications for clinical use. Clin Immunol s-AML; two parallel studies adjusted for various prognostic
Immunopathol. 1996;81:219-223. factors. Blood. 1999;94 (Suppl 1), abs 2933.
44. Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is 14. Estey E, Thall P, Beran M, Kantarijan H, Pierce S, Keating M.
an inhibitor of angiogenesis. Proc Nat Acad Sci USA. Effect of diagnosis (refractory anemia with excess of blasts,
1994;91:4082-4085. refractory anemia with excess of blasts in transformation, or
45. Raza A, Meyer P, Lisak L, et al. Thalidomide produces acute myeloid leukemia (AML) ) on outcome of AML-type
transfusion independence in long-standing refractory anemias of chemotherapy. Blood. 1997;90:2969-2977.
patients with myelodysplastic syndromes. Blood. In press. 15. Wattel E, Solary E, Hecquet B, et al. Quinine improves the
46. Nissen C, Genitsch A, Sendelov S, Dalle Carbonare V, Wodnar- results of intensive chemotherapy in myelodysplastic syndromes
Filipowicz A. Cell cycling stress in the monocyte line as a risk expressing P glycoprotein: results of a randomised study. Br J
factor for progression of the aplastic anemia/paroxysmal Haematol. 1998;102:1015-1024.
nocturnal hemoglobinuria syndrome to myelodysplastic 16. Bernasconi C, Alessandrini EP, Bernasconi P, et al. Randomized
syndrome. Acta Hematologica. 2000;103:33-40. clinical study comparing aggressive chemotherapy with or
without G-CSF support for high-risk myelodysplastic syn-
III. Therapeutic Advances in MDS dromes or secondary acute myeloid leukemia evolving from
1. Greenberg PL, Cox C, LeBeau M, et al. International scoring MDS. Br J Haematol. 1998;102:678-683.
system for evaluating prognosis in myelodysplastic syndromes. 17. Ganser A, Hofmann WK, Heil G, et al. Intensive chemotherapy
Blood. 1997;89:2079-2088. with idarubicin, ara-C, etoposide, and G-CSF priming yields
2. Harris NL, Jaffe ES, Diebold J, et al. World Health Organization high complete remission rates and is well tolerated in patients
classification of neoplastic diseases of the hemopoietic and with advanced MDS and high-risk AML (AML-MDS.study 02/
lymphoid tissues: report of the Clinical Advisory Committee 95). Blood. 1999:94(Suppl 1): abstr 1336.
meeting – Airlie House, Virginia, November 1997. J Clin Oncol. 18. Beran M, Kantarijan H, O´Brien S et al. Topotecan, a
1999;12:3835-3849. topoisomerase I inhibitor, is active in the treatment of
3. Appelbaum FR, Anderson J. Allogeneic bone marrow transplan- myelodysplastic syndromes and chronic myelomonocytic
tation for myelodysplastic syndromes: outcomes analysis leukemia. Blood. 1996;88:2473-42
according to IPSS score. Leukemia. 1998;12(Suppl 1):25-29. 19. Estey E. Topotecan for myelodysplastic syndromes. Oncology.
4. Nevill TJ, Fung HC, Sheperd JD, et al. Cytogenetic abnormali- 1998;12: NCCN Proc 81-86.
ties in primary myelodysplastic syndromes are highly predictive 20. Cheson BD, Simon R. Low-dose ara-C in acute nonlymphocytic
of outcome after allogeneic bone marrow transplantation. Blood. leukemia and myelodysplastic syndromes: A review of 20 years´
1998;92:1910-1917. experience. Sem Oncol. 1987;14:126-133.
5. DeWitte T, Hermans J, Vossen J, et al. Haematopoietic stem cell 21. Hellström-Lindberg E, Robèrt KH, Gahrton G, et al. A
transplantation for patients with myelo-dysplastic syndromes predictive model for the clinical response to low dose ara-C: a
and secondary acute myeloid leukaemias: a report on behalf of study of 102 patients with myelodysplastic syndromes or acute
the Chronic Leukeamia Working Party of the European Group leukemia. Br J Haematol. 1992;81:503-511.
for Blood and Marrow Transplantation. Br J Haematol. 22. Miller KB, Kyungman K, Morrison FS, et al. The evaluation of
2000;110:620-30. low-dose cytarabine in the treatment of myelodysplastic
6. Deeg HJ, Shulman HM, Andersen JE, et al. Allogeneic and syndromes: a phase III intergroup study. Ann Hematol.
syngeneic marrow transplantation for myelodysplastic syn- 1992;65:162-168.
dromes in patients 55 to 66 years of age. Blood. 2000;95:1188- 23. Gerhartz HH, Walther J, Neuwirtova R, et al. Randomized 3-
1194. armed phase III study of interleukin 3 (IL 3), granulocyte/
7. Collins RH, Shpilberg O, Drobyski WR, et al. Donor leukocyte macrophage colony-stimulating factor as adjuvant to low doses
infusions in 140 patients with relapsed malignancy after of cytosine arabinoside (LD-araC) for the treatment of high-risk
allogeneic bone marrow transplantation. J Clin Oncol. myelodysplastic syndromes (HR-MDS). Blood. 1996;88 (Suppl
1997;15:433-444. 1): abstr 2309.
8. Slavin S, Nagler A, Naparstek E. Nonmyeloablative stem cell 24. Omoto E, Deguchi S, Takaba S, et al. Low-dose melphalan for
transplantation and cell therapy as an alternative to conventional treatment of high-risk myelodysplastic syndromes. Leukemia.
bone marrow transplantation with lethal cytoreduction for the 1996;10:609-614.
treatment of malignant and nonmalignant hematological disease. 25. Denzlinger C, Benz D, Bowen D, Gelly K, Brugger W, Kanz L.
Blood. 1998;91:756-763. Low-dose melphalan induces favourable responses in elderly
9. Demuynck H, Delforge M, Verhoef GEG, et al. Feasibility of patients with high risk myelodysplastic syndromes or secondary
peripheral blood progenitor cell harvest and transplantation in leukemia. Br J Haem. 2000;108:93-95.
patients with poor-risk myelodysplastic syndromes. Br J 26. Silverman LR, Holland JF, Weinberg RS, et al. Effects of
Haematol. 1996;92:351-359. treatment with 5-azacytidine on the in vivo and in vitro
10. Carella AM, Dejana A, Lerma E, et al. In vivo mobilization of hematopoiesis in patients with myelodysplastic syndromes.
karyotypically normal peripheral blood progenitor cells in high- Leukemia. 1993;7 (Suppl 1):21-9.
risk MDS, secondary or therapy-related acute myeloenous 27. Silverman LR, Demakos EP, Peterson B, et al. A randomized
leukemia. Br J Haematol. 1996;95:127-130. controlled trial of subcutaneous azacytidine (AZA C) in patients

Hematology 2000 131


with the myelodysplastic syndrome: a study of the Cancer and 42. Italian Cooperative Study Group for rHuEpo in Myelodysplastic
Leukemia Group B (CALCB). Proc Am Soc Clin Oncol. Syndromes. A randomized double-blind placebo-controlled
1998;17: abstr 14. study with subcutaneous recombinant human erythropoietin in
28. Kornblith AB, Herndon II JE, Silverman LR, et al. The impact patients with low-risk myelodyplastic syndromes. Br J
of 5-azacytidine on the quality of life of patients with the Haematol. 1998;103:1070-1074.
myelodysplastic syndrome (MDS) treated in a randomized 43. Hast R, Wallvik J, Folin A, Bernell P, Stenke L. Long-term
phase III trial of the cancer and leukemia group B (CALGB). follow-up of 18 patients with myelodysplastic syndromes with
Proc Am Soc Clin Oncol. 1998;17: abs 189. myelodysplastic syndromes responding to recombinant
29. Wijermans PW, Lubbert M, Verhoef G, et al. Low-dose 5-aza- erythropoietin treatment. Leukemia Res, accepted for publica-
2´-deoxycytidine, a DNA hypomethylating agent, for the tion.
treatment of high risk myelodysplastic syndrome: a multicenter 44. Negrin RS, Stein R, Doherty K, et al. Maintenance treatment of
phase II study in elderly patients. J Clin Oncol. 2000;18:956-62. the anemia of myelodysplastic syndromes with recombinant
30. Greenberg PL. Biologic and clinical implications of marrow human granulocyte colony-stimulating factor and erythropoietin:
culture studies in the myelodysplastic syndromes. Sem Hematol. evidence for in vivo synergy. Blood. 1996;87:4076-81.
1996;33:163-75. 45. Hellström-Lindberg E, Tangen JM, Grimfors G, et al. Treatment
31. Raza A, Gezer S, Mundle S, et al. Apoptosis in bone marrow of the anemia in myelodysplastic syndromes with granulocyte-
biopsy samples involving stromal and hematopoietic cells in 50 CSF plus erythropoietin: Results from a randomized phase II
patients with myelodysplastic syndromes. Blood. 1995;86:268- study and long-term follow-up on 71 patients. Blood. 1998;
276. 2:68-75.
32. Hellström-Lindberg E, Kanter-Lewensohn L, Öst Å. Morpho- 46. Hellström-Lindberg E, Negrin R, Richard Stein, et al. Erythroid
logical changes and apoptosis in bone marrow from patients response to treatment with G-CSF plus erythropoietin for the
with myelodysplastic syndromes treated with Granulocyte-CSF anemia of patients with myelodysplastic syndromes: proposal
and erythropoietin. Leukemia Res. 1997;21:415-425. for a predictive model. Br J Haematol. 1997;99:344-351.
33. Van de Loosdrecht AA, Vellenga E. Myelodysplasia and 47. Remacha AF, Arrizabalaga B, Villegas A, et al. Erythropoietin
apoptosis: new insights into ineffective erythropoiesis. Medical plus granulocyte colony-stimulating factor in the treatment of
Oncology. 2000;17:573-578. myelodysplastic syndromes. Identification of a subgroup of
34. Gattermann N, Retzlaff S, Wang YL, et al. Heteroplasmic point responders. The Spanish Erythropathology Group.
mutation of mitochondrial DNA affecting subunit I of cyto- Haematologica. 1999;84:1058-64.
chrome c oxidase in two patients with acquired sideroblastic 48. Mantovani L, Lentini G, Hentschel B, et al. Treatment of
anemia. Blood. 1997;90:4961-4972. anaemia in myelodysplastic syndromes with prolonged
35. Backx B, Broeders L, Löwenberg B. Kit ligand improves in administration of recombinant human granulocyte colony-
vitro erythropoiesis in myelodysplastic syndromes. Blood. stimulating factor. Br J Haematol. 2000;109:367-375
1992;80:1213-1217. 49. Hellström-Lindberg E, Schmidt-Mende J, Backman-Johansson
36. Shuster MW, Larson RA, Thompson JA, Coiffier B, Bennett JM, C, Forsblom AM, Fadeel B, Zhivotovsky B. Erythroid apoptosis
Israeil RJ. Granulocyte-macrophage colony-stimulating factor in ringsideroblasic anemia (RARS): influence of Fas antibodies,
(GM-CSF) for myelodysplastic syndrome: Results of a multi- caspase inhibition and G-CSF. Blood. 1999;94 (Suppl 1): abstr
center randomised controlled trial. Blood. 1990;76: abstr 318. 462.
37. Shuster MW, Thompson JA, Larson B, Coiffier B, Bennett J, 50. Bernell P, Stenke L, Wallvik J, Hippe E, Hast R. A sequential
Dugan M. Randomized phase II study of recombinant human erythropoietin and GM-CSF schedule offers clinical benefits in
granulocyte-macrophage colony-stimulating factor (rGM-CSF) the treatment of anaemia in myelodysplastic syndromes.
in patients with neutropenia secondary to myelodysplastic Leukemia Res. 1996;20:693-699.
syndromes. Blood. 1995;86:(Suppl 1): abstr 338. 51. Stasi R, Pagano A, Terzoli E, Amadori S. Recombinant human
38. Negrin RS, Haeuber DH, Nagler A, et al. Maintenance treatment granulocyte-macrophage colony-stimulating factor + erythropoi-
of patients with myelodysplastic syndromes using recombinant etin for the treatment of cytopenias in patients with
human granulocyte colony-stimulating factor. Blood. myelodysplastic syndromes. Br J Haematol. 1999;105:141-148.
1990;76:36-43. 52. Thompson JA, Gilliland DG, Prchal JT, et al. Effect of
39. Greenberg P, Taylor K, Larson R, et al. Phase III randomised recombinant human erythropoietin combined with granulocyte/
multicenter trial of G-CSF vs observation for myelodysplastic macrophage colony-stimulating factor in the treatment of
syndromes (MDS). Blood. 1993; 82: Suppl 1, abs 196. patients with myelodysplastic syndromes. Blood. 2000;95:1175-
40. Hellström-Lindberg E. Efficacy of erythropoietin in the 1179.
myelodysplastic syndromes. An analysis of 205 patients in 17 53. Miller AM, Noyes WE, Taetle R, List AF. Limited erythropoietic
studies. Br J Haematol. 1995;89:67-71. response to combined treatment with recombinant human
41. Rose EH, Abels RI, Nelson RA, McCullough DM, Lessin L. interleukin 3 and erythropoietin in myelodysplastic syndromes.
The use of r-HuEpo in the treatment of anemia related to Leukemia Res. 1999;23:77-83.
myelodysplasia (MDS). Br J Haematol. 1995;89:831-837.

132 American Society of Hematology

You might also like