You are on page 1of 14

Seminar

Myelodysplastic syndromes
Lionel Adès, Raphael Itzykson, Pierre Fenaux

Myelodysplastic syndromes are clonal marrow stem-cell disorders, characterised by ineffective haemopoiesis leading Lancet 2014; 383: 2239–52
to blood cytopenias, and by progression to acute myeloid leukaemia in a third of patients. 15% of cases occur after Published Online
chemotherapy or radiotherapy for a previous cancer; the syndromes are most common in elderly people. The March 21, 2014
http://dx.doi.org/10.1016/
pathophysiology involves cytogenetic changes with or without gene mutations and widespread gene hypermethylation
S0140-6736(13)61901-7
at advanced stages. Clinical manifestations result from cytopenias (anaemia, infection, and bleeding). Diagnosis is
Service d’hématologie, Hôpital
based on examination of blood and bone marrow showing blood cytopenias and hypercellular marrow with dysplasia, St Louis (Assistance Publique
with or without excess of blasts. Prognosis depends largely on the marrow blast percentage, number and extent of Hôpitaux de Paris) and Paris 7
cytopenias, and cytogenetic abnormalities. Treatment of patients with lower-risk myelodysplastic syndromes, University, Paris, France
(L Adès MD, R Itzykson MD,
especially for anaemia, includes growth factors, lenalidomide, and transfusions. Treatment of higher-risk patients is
Prof P Fenaux MD)
with hypomethylating agents and, whenever possible, allogeneic stem-cell transplantation.
Correspondence to:
Prof Pierre Fenaux, Service
Introduction syndromes, acute myeloid leukaemia, or aplastic anaemia. d’hématologie, Hôpital St Louis
Myelodysplastic syndromes are clonal stem-cell Environmental factors include previous use of (Assistance Publique Hôpitaux
disorders predominantly occurring in elderly people. chemotherapy,11 especially of alkylating agents and purine de Paris) and Paris 7 University,
Paris 75475, France
The pathophysiology is a multistep process involving analogues,12,13 radiotherapy,14,15 and tobacco smoking.16 pierre.fenaux@sls.aphp.fr
cytogenetic changes, gene mutations, or both,1 with Recognised occupational factors include exposure to
widespread gene hypermethylation at advanced stages.2–4 benzene and its derivatives,17 and an excess of cases is
The syndromes are characterised by ineffective reported in agricultural and industrial workers.16,18 These
haemopoiesis leading to blood cytopenias and by secondary myelodysplastic syndromes, especially cases
progression to acute myeloid leukaemia in a third of occurring after chemotherapy, generally have poor
patients.5 prognosis as a result of complex cytogenetic findings.
Diagnosis of myelodysplastic syndromes is still based
on examination of the blood and bone marrow, which Biology
shows blood cytopenias and hypercellular marrow with Ineffective haemopoiesis
dysplasia, with or without excess of marrow immature Ineffective haemopoiesis in myelodysplastic syndromes
cells (blasts).6 Prognosis depends mainly on the marrow results from the increased susceptibility of clonal
blast percentage, number and extent of cytopenias, and myeloid progenitors to apoptosis, which leads to
cytogenetic abnormalities.7,8 Treatment, varying from cytopenias despite a generally hypercellular marrow
symptomatic treatment of cytopenias, especially by (figure).19 Progression to acute myeloid leukaemia is
transfusions, to allogeneic stem-cell transplantation, has thought to result from a subsequent shift from
improved in the past few years. apoptosis to proliferation of these clonal progenitors.20,21
In many cases expansion is oligoclonal, rather than
Incidence and cause monoclonal; expansion of minor subclones can also
Myelodysplastic syndromes are generally diseases of older contribute to transformation to acute myeloid
people, with a median age at diagnosis of 65–70 years; less leukaemia.22 Apoptosis of myeloid progenitors can be
than 10% of the patients are younger than 50 years. The triggered by intrinsic factors, such as BCL2-related
disorder shows a slight male predominance except for in proteins,23 for instance resulting from mitochondrial
the form with isolated 5q deletion in which women depolarisation due to iron retention in ringed
predominate.9 The annual incidence of myelodysplastic
syndromes is about four cases per 100 000 people
(reaching 40–50 per 100 000 after age 70 years).9 There are Search strategy and selection criteria
no known ethnic differences in the incidence, but in We searched Medline for articles published from January,
Asian populations the syndromes tend to occur at an 1981, to April, 2013, with the term “myelodysplastic
earlier age, are more likely to include hypocellular marrow, syndrome”. We largely selected publications from the past
and less commonly show isolated 5q deletion than do 5 years, but we did not exclude commonly referenced and
those in western populations.10 highly regarded older publications. We also searched the
The cause of myelodysplastic syndromes is known in reference lists of articles identified by this search strategy and
only 15% of cases (panel). Inherited predisposition to the selected those that we judged relevant. Several review articles
disorder is evident in a third of paediatric cases, including were included because they provide comprehensive
in children with Down’s syndrome, Fanconi’s anaemia, overviews that are beyond the scope of this Seminar. The
and neurofibromatosis. In adults, inherited predisposition reference list was modified during the peer-review process on
is less common but should be investigated in young adults the basis of comments from reviewers.
or in families with other cases of myelodysplastic

www.thelancet.com Vol 383 June 28, 2014 2239


Seminar

Myelodysplastic syndromes with complex karyotypes


Panel: Causes of myelodysplastic syndromes tend to have larger 5q deletions than in the 5q syndrome;35
Antineoplastic agents this more aggressive phenotype could involve
Alkylating agents haploinsufficient tumour suppressor genes from the
• Busulfan neighbouring regions, including CTTNA136 or EGR1.37
• Carboplatin The selective clonal suppression of del(5q) cells by
• Carmustine lenalidomide seems to preserve del(5q) haemopoietic
• Chlorambucil stem cells29 and to be independent of the pathways
• Cisplatin mentioned above.38 A similar haploinsufficiency model
• Cyclophosphamide might also apply for other recurrent interstitial deletions
• Dacarbazine in myelodysplastic syndromes, including del(7q)39 and
• Lomustine del(20q).40,41
• Melphalan
Somatic gene mutations
Topoisomerase II inhibitors
High-throughput genomic methods have enabled the
• Daunorubicin
characterisation of many recurrently mutated genes in
• Doxorubicin
myelodysplastic syndromes (table 1), coding for
• Etoposide
transcription factors (TP53 or ETV6), but mostly for
• Mitoxantrone
epigenetic regulators involved in the methylation
• Razoxane
(DNMT3A) or hydroxymethylation (TET2, IDH1,
Purine analogues IDH2) of cytosines, or in covalent modifications of
• Fludarabine and others histones (EZH2, UTX, ASXL1). Mutations in those
Radiotherapy genes can be dominant-negative (DNMT3A), gain of
Environmental factors function (IDH1, IDH2), or loss of function (TET2,
• Tobacco ASXL1). They seem to result in global changes in gene
• Ionising radiation expression patterns and can also affect genomic
• Benzene exposure (and industrial hydrocarbons) stability.42 Since these mutations also probably arise in
• Agricultural compounds (pesticides, herbicides, fertilisers) haemopoietic stem cells43 and can be found in other
myeloid neoplasms, they are thought to affect clonal
dominance and progression of myelodysplastic
sideroblasts,24 and by external cues, including death syndromes1,44 rather than to determine the specific
signals (tumour necrosis factor,25 Fas26), or myelo- phenotype.45,46
suppressive cytokines (eg, transforming growth Recurrent mutations in genes encoding components of
factor β27). Intrinsic apoptotic signals seem to reflect the the splicing machinery, including SF3B1, SRSF2, U2AF1,
role of clonal chromosomal changes, gene mutations, and ZRSR2, have also been reported in myelodysplastic
or epigenetic changes, and extrinsic signals are related syndromes.47,48 They alter gene splicing, with many
to immune and microenvironmental insults. potential functional outcomes.48,49 Some of these
spliceosomal components are also involved in the
5q syndrome maintenance of genomic stability.50 By contrast with
Of the subtypes of myelodysplastic syndrome, mutations in epigenetic regulators, spliceosome
5q syndrome has the best understood pathogenesis. The mutations are highly characteristic of myelodysplastic
interstitial deletion del(5)(q31q33) originates in syndromes (and myelodysplastic syndrome with
haemopoietic stem cells28,29 and leads to myeloproliferative feature neoplasms) among myeloid
haploinsufficiency (ie, loss of one allele resulting in malignancies. SF3B1 mutations are closely associated
reduced gene expression, without gene mutation) of a with ring sideroblastic subtypes of myelodysplastic
variable number of genes, according to the extent of the syndromes;47 they could contribute directly to abnormal
deleted region. The shortest deleted segment identified iron retention in the mitochondria of erythroid
so far to result in the 5q phenotype encompasses about precursors, and thus formation of ringed sideroblasts.24,49
40 genes included in a common deleted region.30
Haploinsufficency for the ribosomal subunit RPS14 Epigenetic changes
induces a P53-dependent block in erythroid proliferation Epigenetic changes are particularly seen during
and differentiation,31,32 whereas haploinsufficiency of progression of myelodysplastic syndromes; they include
two micro-RNAs, mir-145 and mir-146a, leads to aberrant gene promoter methylation—eg, in the tumour
dysmegakaryopoiesis and thrombocytosis.33 Subclonal suppressor gene P15 INK4B.51 Aberrant methylation
mutations in the gene TP53, possibly promoted by the also arises in haemopoietic stem cells,3,4 but its relation
constant activation of P53, could contribute to to gene mutations in epigenetic regulators is unclear.52
progression to higher-risk disease.34 The prognostic contribution of aberrant methylation

2240 www.thelancet.com Vol 383 June 28, 2014


Seminar

Immune attack Environmental exposure Ageing Radiotherapy and chemotherapy

(Micro-) environmental changes Somatic gene mutations Epigenetic changes Combined haploinsufficiency
(Osteoblast dysfunction?), (Epigenetic regulators, spliceosome (Promoter methylation) (Interstitial deletion)
immune deregulation genes)

Clonal expansion Increased progenitor apoptosis,


differentiation defect

HSC MDS-initiating MDS progenitors Cytopenias


? cells

Mesenchymal cells

AML
Risk of progression to AML
Clonal selection
(±additional genetic lesions)

Figure: Pathogenesis of myelodysplastic syndromes


Age-induced genetic, epigenetic, and immune-mediated changes in haemopoietic stem cells (HSC) lead to oligoclonal expansion of myelodysplastic stem cells, with
defective differentiation, characterised by increased apoptosis of erythroid and myeloid progenitors. Microenvironmental changes and immune deregulation
contribute to this differentiation defect. MDS=myelodysplastic syndromes. AML=acute myeloid leukaemia.

profiles, and their potential use as biomarkers in


Frequency of Gene function Prognosis
patients treated with hypomethylating agents, also mutations (%)
remain unclear.2
SF3B1 15–30% Spliceosome Favourable?
TET2 15–25% DNA hydroxymethylation Neutral
Microenvironment and immune cells
ASXL1 10–20% Histone modifications Unfavourable
Xenotransplantation assays and in-vitro experiments 25,53
RUNX1 5–15% Transcription factor Unfavourable
have shown the crucial role of microenvironmental
TP53 5–10% Transcription factor Unfavourable
cues in the maintenance and clinical expression of
DNMT3A 5–10% DNA methylation Unfavourable?
myelodysplastic syndromes.54 The haemopoietic stem-
NRAS, KRAS 5–10% Signal transduction Unfavourable (low-risk syndromes)
cell niche could contribute to the pathogenesis of these
disorders.55,56 Immune deregulation is shown by the SRSF2 5–10% Spliceosome Unfavourable

finding of clonal T-cell expansions in some patients, U2AF1 5–10% Spliceosome Unfavourable (low-risk syndromes)
especially those with hypoplastic syndromes. Expansion BCOR-L1 5–6% Transcription repressor Unfavourable
of distinct T-cell subpopulations could occur at different ZRSR2 5% Spliceosome Neutral?
disease stages; inflammatory Th17 cells could contribute EZH2 3–7% Histone modifications Unfavourable
to ineffective haemopoiesis in lower-risk myelo- ETV6 3% Transcription factor Unfavourable
dysplastic syndromes57 and regulatory T cells contribute JAK2 3–4% Signal transduction Favourable?
to evasion from antitumoral immunity in higher-risk
disease.58 Patients with higher-risk myelodysplastic IDH1, IDH2 4–5% DNA hydroxymethylation Unfavourable
and histone modifications
syndromes also have higher than normal numbers of
UTX 1–2% Histone modifications Unfavourable?
T regulatory cells,58 which suggests that progression is
facilitated by immune suppression. The mode of action Table 1: Recurrent somatic gene mutations in myelodysplastic syndromes
of several drugs in myelodysplastic syndromes,
including the immunomodulatory agent lenalidomide microenvironmental and immune modifications59
and hypomethylating agents, which cannot eradicate beyond their specific action on the myelodysplastic
myelodysplastic stem cells,3,29 could involve syndrome clone.60

www.thelancet.com Vol 383 June 28, 2014 2241


Seminar

Diagnosis mitochondria around the nucleus) are counted after


Clinical findings staining with Prussian blue.
Clinical features are non-specific and mainly result from Generally, marrow aspirates are sufficient. The more
cytopenias, especially anaemia, which is symptomatic in invasive trephine biopsy is, however, important to assess
many patients, leading to fatigue, poor quality of life, and marrow fibrosis, present in 15% of cases, and is useful
destabilisation of underlying cardiovascular disease. when the marrow is hypocellular, to differentiate
Thrombocytopenia is commonly associated with platelet myelodysplastic syndromes from aplastic anaemia or
dysfunction, potentially leading to bleeding symptoms acute myeloid leukaemia.64 It can also enable detection, in
even in moderate thrombocytopenia. Similarly, infections the absence of excess blasts, of the abnormal localisation
(especially with gram-negative bacilli, gram-positive cocci, of immature precursors, which is associated with worse
and fungi) can occur with only moderate neutropenia due prognosis.65 In most patients, aspirates are sufficient for
to frequent neutrophil function defects.61 follow-up including for assessment of treatment efficacy.
Many patients with myelodysplastic syndromes also
have immune disorders, including relapsing poly- Differential diagnosis
chondritis, vasculitis, and seronegative polyarthritis.62,63 Diagnosis of myelodysplastic syndromes is generally
The two disorders tend to be diagnosed almost simul- made readily on blood and marrow examination. All
taneously, which suggests a pathophysiological relation. other causes of cytopenias must be carefully excluded;
the list includes, but is not limited to, vitamin deficiencies
Blood and marrow examination (ruled out by serum testing), autoimmune disease, liver
90% of patients have anaemia that is typically, but not disease, hypersplenism, drug intake, exposure to
always, macrocytic and non-regenerative. Neutropenia environmental toxins, aplastic anaemia, paroxysmal
and thrombocytopenia are seen at diagnosis in about a nocturnal haemoglobinuria, bone-marrow infiltration by
third of patients, and rarely without anaemia. Small malignancy, viral infections, and rare forms of hereditary
numbers of circulating blasts can be found, rarely anaemias (such as congenital dyserythropoetic
exceeding 5%. However, on the peripheral blood smear, anaemias). Diagnosis can be difficult when cytopenias
200 cells should be counted to ensure correct are moderate and marrow dysplasia is mild. In such
classification that depends partly on the number of patients, detection of a chromosomal abnormality (and
peripheral blasts (>1% or not). Leucocytosis and possibly of a somatic mutation or abnormal flow
immature granulocytes are rare on the differential count cytometry features)1,44,66 in marrow cells can point to a
except in chronic myelomonocytic leukaemia, now clonal disorder, and therefore in the context to an early
classified among myelodysplastic–myeloproliferative myelodysplastic syndrome. In the absence of such
neoplasms and defined by blood monocytosis of more abnormalities, the diagnosis must be idiopathic cytopenia
than 1 × 10⁹ cells per L.6 of undetermined significance.67
Analysis of bone marrow is central to the diagnosis of
myelodysplastic syndromes. The bone marrow is Cytogenetic findings
generally hypercellular and shows dysplastic features in An abnormal karyotype is shown by conventional
one or several myeloid series, generally differing from cytogenetic analysis in 40–50% of cases at diagnosis.68,69 By
those seen in deficiency of vitamin B12 or folate.6 The contrast with acute myeloid leukaemia, in which balanced
marrow blast percentage (including agranular blasts and translocations are frequent, myelodysplastic syndromes
myeloblasts, but not promyelocytes) should be assessed are characterised by partial or complete loss or gain of
on at least 500 nucleated cells. Ring sideroblasts chromosomes, the most frequent findings being
(erythroblasts with abnormal localisation of iron in deleted 5q, −7 or deleted 7q, +8, deleted 20q, and
deleted 17p; complex cytogenetic findings are common in
Proportion Karyotype Median Time to 25% patients with a major excess of marrow blasts or with
of patients survival AML evolution therapy-related myelodysplastic syndromes (table 2).
(%) (years) (years) Fluorescence in-situ analysis with specific probes
Very good 4% –Y, del(11q) 5·4 NR (especially for chromosomes 5, 7, 8, 17, and 20) can be
Good 72% Normal, del(5q), del(12p), del(20q), double 4·8 9·4 useful when fewer than 20 mitoses have been obtained for
including del(5q) conventional analysis or when the karyotype is complex,
Intermediate 13% del(7q), +8, +19, i(17q), any other single or 2·7 2·5 to identify the chromosomes involved more accurately.
double independent clones
Cytogenetic analysis has major prognostic value for
Poor 4% −7, inv(3)/t(3q)/del(3q), double including 1·5 1·7
−7/del(7q); complex: 3 abnormalities
myelodysplastic syndromes but can also enable diagnosis
Very poor 7% Complex >3 abnormalities 0·7 0·7
in difficult cases. Such cases include patients with
unexplained isolated thrombocytopenia (deleted 20q),
AML=acute myeloid leukaemia. NR=not reached. elderly women with mild anaemia (deleted 5q), and
younger patients with moderate cytopenias (−7 or +8),70
Table 2: Cytogenetic findings in patients with myelodysplastic syndromes, by their prognostic value8,68
confirming the clonal nature of the disease.

2242 www.thelancet.com Vol 383 June 28, 2014


Seminar

Other diagnostic tests defined categories are refractory anaemia with excess
Other tests can be useful for diagnosis, in rare cases of blasts (type 1 or 2 based on the marrow blast count being
undefined cytopenias. The presence of a somatic mutation below or above 10%), refractory anaemia with or without
other than TET2 mutation, reported in some healthy ringed sideroblasts, refractory cytopenias with multilineage
individuals,71 can allow a diagnosis of a myelodysplastic dysplasia, and myelodysplastic syndromes with isolated
syndrome. Flow cytometry takes advantage of the frequent deleted 5q. Categories of refractory cytopenia with
abnormal expression of markers on marrow myeloid cells unilineage dysplasia other than refractory anaemia have
in these syndromes. Consensus workshops have proved less reproducible in clinical practice.72 Therapy-
established combinations of markers for which related myelodysplastic syndromes are classified
abnormalities strongly suggest myelodysplastic separately, in a group that also includes therapy-related
syndromes, but experience is needed for this approach.66 acute myeloid leukaemia.
Patients whose diagnosis remains uncertain after Boundaries include the myelodysplastic–myelo-
marrow examination and cytogenetic analysis generally proliferative neoplasms group, in which at least 50% of
have moderate cytopenias and slowly evolving disease. patients have chronic myelomonocytic leukaemias.
No therapeutic intervention at this stage has shown any However, except for some cases of chronic myelomonocytic
efficacy; if a very early diagnosis of myelodysplastic leukemia, these disorders have generally limited pro-
syndromes is missed, the consequences are unlikely to liferative features and evolve mostly like typical
be severe except in younger patients. myelodysplastic syndromes.6 Patients who have refractory
anaemia with ringed sideroblasts and thrombocytosis,
Classification also in this group, generally carry SF3B1 mutations (as do
Morphological classification of myelodysplastic syndromes those with refractory anaemia with ringed sideroblasts
is based on the WHO 2008 criteria6 (table 3). The best alone); the disease is also mainly characterised by

Blood findings Bone-marrow findings


Myelodysplastic syndrome
Refractory cytopenia with unilineage dysplasia One or two cytopenias; no or rare blasts (<1%) One lineage dysplasia ≥10% of cells in one myeloid lineage; <5% blasts;
(refractory anaemia; refractory neutropenia; <15% of erythroid precursors ring sideroblasts
refractory thrombocytopenia)
Refractory anaemia with ring sideroblasts Anaemia; no blasts ≥15% of erythroid precursors ring sideroblasts; erythroid dysplasia only;
<5% blasts
Refractory cytopenia with multilineage dysplasia Cytopenia(s); no or rare blasts (<1%); no auer rods; Dysplasia in ≥10% of cells in at least two myeloid lineages (neutrophil,
<1 × 10⁹ cells per L monocytes erythroid precursors, or megakaryocytes); <5% blasts in marrow; no auer
rods; with or without 15% ring sideroblasts
Refractory anaemia with excess blasts 1 Cytopenia(s); <5% blasts; no auer rods; <1 × 10⁹/L monocytes Dysplasia in one or several lineages; 5–9% blasts; no auer rods
Refractory anaemia with excess blasts 2 Cytopenia(s); 5–19% blasts; with or without auer rods; Dysplasia in one or several lineages ; 10–19% blasts; with or without auer
<1 × 10⁹/L monocytes rods
Myelodysplastic syndrome unclassified Cytopenias; <1% blasts Unequivocal dysplasia in <10% of cells in one or more myeloid lineages
accompanied by a cytogenetic abnormality is presumptive evidence for
diagnosis; <5% blasts
MDS associated with isolated del(5q) Anaemia; normal or increased platelet count in most cases; Normal to increased megakaryocytes with hypolobated nuclei; <5% blasts;
no or rare blasts (<1%) isolated del(5q) cytogenetic abnormality; no auer rods
Myelodysplastic–myeloproliferative neoplasms
Chronic myelomonocytic leukaemia 1 Persistent peripheral blood monocytosis (>1 × 10⁹/L); Dysplasia in one or more cell lines; <10% blasts
no Philadelphia chromosome or BCR-ABL1 fusion gene;
<5% blasts
Chronic myelomonocytic leukaemia 2 Persistent peripheral blood monocytosis (>1 × 10⁹/L); Dysplasia in one or more cell lines; 10–19% blasts
no Philadelphia chromosome or BCR-ABL1 fusion gene;
<19% blasts
Myelodysplastic or myeloproliferative disease, Morphological features of myelodysplastic syndrome; ··
unclassifiable prominent myeloproliferative features (platelets >600× 10⁹
cells per L, leucocytes >13 × 10⁹/L, splenomegaly);
no Philadelphia chromosome or BCR-ABL1 fusion gene;
no del(5q), t(3;3)(q21;q26), inv3(q21;q26); no underlying
myeloproliferative disease
Provisional entity: refractory anaemia with ring Similar to refractory anaemia with ring sideroblasts; platelet Similar to refractory anaemia with ring sideroblasts; without del(5q)
sideroblasts and thrombocytosis >600×10⁹ cells per L
Therapy-related neoplasm
Acute myeloid leukaemia or myelodysplastic ·· ··
syndrome in individuals exposed to cytotoxic agents

Table 3: WHO 2008 classification6

www.thelancet.com Vol 383 June 28, 2014 2243


Seminar

anaemia.6,73 The 30% marrow blast threshold (subsequently prognostic scoring system (WPSS), which takes into
lowered to 20%) to distinguish myelodysplastic syndromes account WHO classification and requirements for
from acute myeloid leukaemia should be critically transfusion of red blood cells.74 The most important is
interpreted in regards to treatment, because acute myeloid the revised IPSS (table 6),8 which also uses cytogenetic
leukaemia is generally considered for chemotherapy and abnormalities, cytopenias, and blast count for scoring
higher-risk myelodysplastic syndromes for hypo- but with new thresholds. With the revised IPSS, 27% of
methylating agents. For example, a younger patient with lower-risk myelodysplastic syndromes according to the
15% marrow blasts and normal karyotype—judged to original IPSS are reclassified as having a higher risk
have a myelodysplastic syndrome—would probably (table 7), and patients potentially need more intensive
benefit from chemotherapy, whereas an elderly patient treatment, whereas 18% of high-risk patients defined by
with 25% marrow blasts and complex karyotype—judged the original IPSS are reclassified as low risk.8
to have acute myeloid leukaemia— might benefit more
from hypomethylating agents. Other prognostic factors
Age is an important indicator of poor prognosis; however,
Prognostic factors it is not included in the original or revised IPSS because it
International prognostic scoring system (IPSS) and its could allow underestimation of the severity of
update myelodysplastic syndromes in young patients. Indeed, the
Many prognostic factors have been identified in loss of years of life expectancy is greater in younger than
myelodysplastic syndromes but international efforts have for older adults with these disorders.76 However, age-
concentrated on identification of a small number of related factors, especially non-haematological comorbidity,
features with independent prognostic value, routinely also clearly affects survival and must be taken into account
available in all centres, which can be grouped in scoring in decisions on intensive treatments. In a large study of
systems. A high percentage of marrow blasts, increasing 600 patients, the assessment of comorbidities by the Adult
number and importance of cytopenias, and abnormal Comorbidity Evaluation-27 showed that patients with
karyotype are the most important independent factors of severe comorbidity had 50% lower survival than did those
poor prognosis in myelodysplastic syndromes; the IPSS7 without comorbidity, independently of age and IPSS risk
allows classification of patients in four subgroups with group.77 Similarly, the haemopoietic-stem-cell trans-
largely differing risks of progression to acute myeloid plantation-specific comorbidity index and the Charlson
leukaemia and survival (table 4, table 5). comorbidity index are independent prognostic factors in
Several attempts have been made to refine IPSS, still patients with lower-risk myelodysplastic syndromes,78
used for drug approval by most health authorities, which suggests that these factors should be taken into
especially in lower-risk myelodysplastic syndromes in account in treatment decisions.
which it cannot distinguish between patients with longer Bone-marrow fibrosis (grade ≥2) is independently
survival and those with only intermediate outcome.74,75 associated with poorer survival, in both lower-risk and
These refined systems include the WHO-based higher-risk myelodysplastic syndromes, so systematic
bone-marrow biopsy is justified at least at diagnosis.64
Somatic mutations have a strong effect on survival. The
0 points 0·5 points 1·0 point 1·5 points 2·0 points
presence of any of the mutations TP53, RUNX1, TP53,
Bone-marrow blasts (%) <5% 5–10% ·· 11–20% 21–30% EZH2, or ETV6 worsens prognosis,1,44 independently of
Number of cytopenias* 0–1 2–3 ·· ·· ·· IPSS. ASXL1 and SRSF2 mutations also seem to be
Cytogenetics Good: Intermediate: Poor: complex ·· ·· associated with a poor prognosis.79,80
normal, Y, other ≥3 abnormalities,
del(5q), abnormalities chromosome 7
Flow cytometry features of marrow cells have
del(20q) abnormalities prognostic value with experienced researchers, but this
test cannot yet be generalised.81 High serum
*Platelet count <100 × 10⁹/L; haemoglobin <100 g/L; absolute neutrophil count <1·8 × 10⁹/L.
concentrations of ferritin and lactate dehydrogenase are
Table 4: The international pronostic scoring sytem (IPSS)7 score values also associated with poorer survival.82,83

Predictive factors for response to treatment


Low Intermediate 1 Intermediate 2 High Most prognostic factors in myelodysplastic syndromes
have been assessed in patients receiving supportive
Risk score 0 0·5–1·0 1·5–2·0 ≥2·5
care only. Because more effective treatments are
Proportion of patients (%) 33% 38% 22% 7%
emerging, prognosis is increasingly analysed in terms
Median survival (years) 5·7 3·5 1·2 0·4
of such treatments. For example, low baseline serum
Time to 25% AML evolution (years) 9·4 3·3 1·1 0·2
concentrations of erythropoietin can predict response
IPSS=international prognostic scoring system. AML=acute myeloid leukaemia. to erythropoiesis-stimulating drugs,84 whereas early
resistance to these drugs in lower-risk myelodysplastic
Table 5: IPSS prognostic risk category clinical outcomes
syndromes is associated with poorer outcome.85

2244 www.thelancet.com Vol 383 June 28, 2014


Seminar

0 points 0·5 points 1·0 point 1·5 points 2·0 points 3·0 points 4·0 points
Cytogenetics* Very good ·· Good ·· Intermediate Poor Very poor
Bone-marrow blasts (%) ≤2% ·· >2 to <5% ·· 5–10% >10% ··
Haemoglobin (g/L) ≥100 ·· 80 to <100 <80 ·· ·· ··
Platelet count (×109/L) ≥100 50 to <100 <50 ·· ·· ·· ··
Absolute neutrophil count (×109/L) ≥0·8 <0·8 ·· ·· ·· ·· ··

*As in table 2.

Table 6: Revised international prognostic scoring system prognostic score values

Response to hypomethylating agents can be predicted


Very low Low Intermediate High Very high
by conventional factors (including performance status,
karyotype, erythrocyte transfusion requirements, Risk score ≤1·5 >1·5–3·0 >3·0–4·5 >4·5–6·0 >6·0
presence of circulating blasts),86 but possibly also Proportion of patients (%) 19% 38% 20% 13% 10%
somatic mutations; TP53 mutations predict poor Median survival (years) 8·8 5·3 3·0 1·6 0·8
response, whereas TET2, EZH2, or DNMT3A Time to 25% evolution to AML (years) NR 10·8 3·2 1·4 0·73
mutations, associated with neutral or unfavourable IPSS=international prognostic scoring system. AML=acute myeloid leukaemia. NR=not reached.
outcome in general, seem to be associated with
favourable response, although these findings remain Table 7: Revised IPSS prognostic risk category clinical outcomes
unconfirmed.87–90 Presence of deleted 5q is clearly
associated with erythroid response to lenalidomide; outcome.93 Various comorbidity scores can be used to
however, in patients with deleted 5q, a complex predict post-transplantation outcomes in patients with
karyotype, TP53 mutation, and a blast count above 5% myelodysplastic syndromes.93–95
predict poorer response to this drug.34,91 The optimum timing of allogeneic stem-cell
transplantation is a balance between a risk of transplant-
Treatment related mortality in patients who might expect long-term
Strategy survival without transplantation and a risk of relapse after
Treatment of myelodysplastic syndromes has improved transplantation in patients with advanced disease. In
lately but remains challenging. The therapeutic strategy higher-risk patients, both those younger than 60 years
remains largely based on the IPSS. In patients classified receiving myeloablative conditioning transplantation and
as high or intermediate 2 on the IPSS (higher risk) with older patients treated with non-myeloablative regimens,96–99
median survival if untreated of only about 12 months, early stem-cell transplantation was associated with a
treatment should aim to modify the disease course, survival advantage compared with other therapeutic
avoiding progression to acute myeloid leukaemia, and options.92 By contrast, early stem-cell transplantation had
extending survival. By contrast, in those classified as low an adverse effect on survival in lower-risk patients.92,100
or intermediate 1 on the IPSS (lower risk), survival is A high bone-marrow blast percentage before stem-cell
longer and many patients die from causes other than transplantation (especially if >10%) is associated with a
myelodysplastic syndromes. Therefore, their treatment higher risk of relapse, so the procedure is generally done
mainly aims to ameliorate the consequences of after a cytoreductive regimen (with chemotherapy or
cytopenias and transfusions and improve quality of life. perhaps hypomethylating agents) in patients with an
However, some lower-risk patients can benefit from excess of marrow blasts, although no evidence from
treatments generally applied to those at higher risk. prospective studies is available to support this approach.

Allogeneic stem-cell transplantation Chemotherapy


This approach remains, with few exceptions, the only Intensive chemotherapy mainly uses combinations of
curative treatment of higher-risk myelodysplastic anthracyclines and cytarabine, as for acute myeloid
syndromes, with prolonged disease-free survival of leukaemia,101–103 but it yields only 40–60% complete
35–50%.34,85 However, it can generally be offered only to a remissions in patients with myelodysplastic syndromes
few patients with myelodysplastic syndromes—younger or those who progress to acute myeloid leukaemia and
patients can be offered allogeneic stem-cell shorter durations of complete remission (median
transplantation for myeloablation and older patients (up <12 months) than in de-novo acute myeloid
to 70 years or even older if they are very fit) can be offered leukaemia.101–103 Patients with unfavourable karyotype
reduced-intensity stem-cell transplantation from an have fewer complete remissions and shorter durations of
HLA-identical donor.92 remission.101 No drug combination (including fludarabine,
Many patients have comorbidities and impaired topotecan, and gemtuzumab ozogamicin, with cytarabine,
functional status, which can lead to poor treatment with or without granulocyte colony-stimulating factor

www.thelancet.com Vol 383 June 28, 2014 2245


Seminar

[G-CSF]) has shown any survival advantage over classic care.114,115 This difference in survival benefit between
anthracycline–cytarabine combinations.102,104–106 Thus, decitabine and azacitidine suggests intrinsic differences
intensive chemotherapy has limited indication in higher- between them, because the mechanisms of action are
risk myelodysplastic syndromes and is generally offered not identical. It could also be the result of the low
to patients younger than 65 years with favourable number of cycles given (median three and four cycles,
cytogenetics, especially as a bridge to allogeneic stem-cell in the two phase 3 trials), and possibly to a non-
transplantation. optimum decitabine schedule. In an adjusted analysis
Low-dose cytarabine (20 mg/m² daily, 14–21 days every between azacitidine-treated and decitabine-treated
month) yields complete remission and partial remission patients, no survival difference was reported for
rates of 15–20% in higher-risk myelodysplastic patients younger than 65 years, but azacitidine showed
syndromes, but no proven survival advantage.107,108 an advantage in older patients.115,116 A 5-day regimen of
Response is seen only in patients without an unfavourable decitabine (20 mg/m² daily intravenously) yielded a
karyotype.108,109 In a randomised study, low-dose cytarabine 39% complete remission rate and was superior to two
was associated with significantly fewer responses, shorter other regimens,117,118 but the effect of this regimen on
responses, and lower survival than was azacitidine, survival is unknown.
irrespective of WHO classification and karyotype, Since median survival is only 2 years with azacitidine
although it was more myelosuppressive.110 in high-risk myelodysplastic syndromes, combinations
of drugs with azacitidine are under investigation. Results
Hypomethylating agents of some phase 2 trials combining azacitidine and valproic
These drugs have become the first-line treatment in most acid,119 vorinostat,120 entinostat,121 lenalidomide,122
higher-risk myelodysplastic syndromes. After clinical thalidomide,123 and gemtuzumab ozogamicin124 have
trials established clear efficacy of azacitidine in shown promising response rates, but no randomised
myelodysplastic syndromes,111 a survival benefit with trial so far has shown any response or survival advantage
azacitidine (75 mg/m² daily subcutaneously, 7 days every of these combinations over azacitidine alone.
4 weeks) was shown over conventional care regimens, Hypomethylating agents are increasingly being tested
including best supportive care, low-dose cytarabine, or before allogeneic stem-cell transplantation in patients
intensive chemotherapy, in a randomised trial in with excess blasts or an unfavourable karytotype, with
356 higher-risk patients,109 with median survival of the aim to reduce the risk of relapse after transplantation,
24·4 months versus 15 months. Progression to acute and avoid the toxic effects of intensive chemotherapy. A
myeloid leukaemia was delayed and independence from large retrospective analysis showed similar outcome fot
erythrocyte transfusion was significantly more common. allogeneic stem-cell transplantation after treatment with
The survival advantage with azacitidine was apparent azacitidine and intensive chemotherapy, which needs to
irrespective of age, marrow blast percentage, and be confirmed prospectively.125
karyotype.109 Response to azacitidine was delayed in
many cases, with a median time to response of Treatment of lower-risk myelodysplastic
2–3 months; some patients responded after only six syndromes
cycles.112 These findings suggest that patients should not Treatment approaches mainly focus, at least initially, on
be classed as resistant before six cycles, unless they the treatment of cytopenias. Anaemia is the predominant
clearly progress. Response was seen in 51% patients (17% cytopenia in most patients with lower-risk myelodysplastic
complete remission, 12% partial remission) and most syndromes. Chronic anaemia leads to increased
responses were only haematological improvement morbidity and lower quality of life,126,127 and many patients
(achievement of erythrocyte-transfusion dependence and need repeated erythrocyte transfusions, which might
increase in platelet counts). Achievement of (although this notion is disputed) result in potentially
haematological improvement, even in the absence of deleterious iron overload in various organs.126
complete or partial remission, was associated with better High-dose erythropoiesis-stimulating agents, including
survival.113 The median duration of response to azacitidine recombinant erythropoietin or darbepoietin alfa, with or
was 13·6 months. The median number of cycles was 15 without G-CSF, are generally the first-choice treatment
in responders, which suggests that long-term treatment for anaemia in lower-risk myelodysplastic syndromes (at
is necessary for survival improvement. The current least in the absence of deleted 5q) yielding 30–50%
recommendation is to continue azacitidine until erythroid responses of median duration 2 years. Response
progression or unacceptable toxicity, but optimum rates are higher in patients with no or limited requirement
duration of treatment in responders remains unknown. for erythrocyte transfusions and low serum erythropoietin
Two phase 3 trials comparing decitabine (15 mg/m² concentrations (below 200–500 IU/L) than in patients
three times a day for 3 days every 6 weeks) with best without these characteristics.128 Both retrospective studies
supportive care showed to have response rates similar and a prospective study show that erythropoiesis-
to those observed with azacitidine, but with no stimulating agents have no effect on progression to acute
significant survival advantage over best supportive myeloid leukaemia and that they could improve survival

2246 www.thelancet.com Vol 383 June 28, 2014


Seminar

in low-risk myelodysplastic syndromes compared with Despite these treatments, many patients with lower-
erythrocyte transfusions alone.129–131 risk myelodysplastic syndromes eventually need long-
Erythropoiesis-stimulating agents are less effective in term erythrocyte transfusions. The adverse effect of iron
lower-risk patients with deleted 5q than in those without overload remains disputed, as does the usefulness of
this feature.132 Lenalidomide yields erythrocyte iron chelation in these patients. In particular, although
transfusion independence in two-thirds of such patients, cardiac iron overload is a well-documented cause of
for a median duration of 2–3 years, and cytogenetic heart failure in children with thalassaemia,146 its
responses in 50–70%; it has been approved, in the USA incidence and clinical consequences are less clear in
and other countries, and in the European Union for patients transfused for myelodysplastic syndromes,
transfusion-dependent anaemia.133,134 TP53 mutations, especially when they have other causes of cardiac
present in 20% of lower-risk patients with deleted 5q, are morbidity. Clinically significant iron overload associated
associated with primary resistance to lenalidomide and a with decreased cardiac function is, however, quite
high risk of progression to acute myeloid leukaemia;34 common in patients who have received 100 or more red-
other therapeutic options might be better for these blood-cell concentrates.147
patients. Because stem cells with deleted 5q can persist, In the absence of prospective studies, retrospective
even in patients who achieve a complete cytogenetic studies have suggested that adequate iron chelation can
response to lenalidomide,29 long-term treatment could be improve survival in these patients;148–150 such studies can,
needed irrespective of some reports of sustained however, be subject to selection bias. Expert opinion151,152
responses after drug discontinuation.135 Two retrospective suggests that chelation should be started in patients with
analyses comparing the long-term outcome of lower-risk lower-risk myelodysplastic syndromes who have received
patients with deleted 5q treated with and without 20–40 red-blood-cell concentrates, or if serum ferritin
lenalidomide found no excess risk of acute myeloid concentration rises above 1500–2500 U/L. However, even
leukaemia with the drug.136,137 moderate iron overload is associated with increased
Treatment of anaemia after primary resistance to transplant-related mortality, so iron chelation therapy
erythropoiesis-stimulating agents (or lenalidomide in should probably be offered early in patients who are
patients with deleted 5q) or after relapse remains a candidates for allogeneic stem-cell transplantation.153,154
challenge. Antithymocyte globulins, with or without Neutropenia and thrombocytopenia are less frequent
ciclosporin, can yield an erythroid response, with than is anaemia in lower-risk myelodysplastic syndromes,
response to other cytopenias, especially thrombocytopenia and rarely occur alone or severely. Absolute neutrophil
in 25–40% of patients, but in selected younger patients counts are below 1·5 × 10⁹ cells per L in only 5–10% of
with low-risk MDS,138–140 with limited erythrocyte low-risk patients,75 and neutropenia is rarely associated
transfusion history, normal karyotype (or possibly with life-threatening infection if no drugs that worsen
trisomy 8), no excess marrow blasts or HLA DR15 neutropenia are used. G-CSF and granulocyte-
genotype, no deleted 5q, limited exposure to previous macrophage colony-stimulating factor can improve
treatments, and possibly hypocellular marrow, although neutropenia in 60–75% of cases,155 but long-term use has
this finding is disputed.141 no proven effect on survival. Broad-spectrum antibiotics
Hypomethylating agents are approved in the treatment should be used immediately in neutropenic patients with
of lower-risk myelodysplastic syndromes in several myelodysplastic syndromes when fever or other signs of
countries, can lead to transfusion independence in up to infection occur.
40% of patients,111,142 and are effective against other Platelet counts below 50 × 10⁹ cells per L are seen in
cytopenias. In a phase 2 trial of azacitidine alone or with 30–35% of low-risk patients,75 and severe bleeding is rare
erythropoiesis-stimulating agents in patients with unless drugs that interfere with haemostasis are used.
erythrocyte-dependent lower-risk myelodysplastic High-dose androgens can improve thrombocytopenia in
syndromes who showed resistance to erythropoiesis- about a third of affected patients, but response is
stimulating agents, transfusion independence was generally transient.156
achieved in only 17% of the patients, with no difference The thrombopoietin-receptor agonist romiplostin in
between the two randomised groups.143 high dose (500–1500 μg per week) yielded 55% platelet
Lenalidomide enables transfusion independence in responses in a phase 2 trial in lower-risk patients with
25–30% of patients with lower-risk myelodysplastic thrombocytopenia.157 However, a transient rise in marrow
syndromes who do not have deleted 5q and who are blasts occurred in 15% of patients; this was reversible
resistant to erythropoiesis-stimulating agents,144 but it is after drug discontinuation. In a phase 3 placebo-
not approved for this indication and should be used only controlled study in patients with lower-risk
in clinical trials. Preliminary results suggest that the myelodysplastic syndromes and thrombocytopenia,
combination of lenalidomide and erythropoiesis- romiplostin transiently increased peripheral blasts in a
stimulating agents can lead to high rates of independence proportion of patients, although the rate of progression
from erythrocyte transfusion in patients resistant to an to acute myeloid leukaemia did not differ significantly
erythropoiesis-stimulating agent alone.145 between the study groups.158 Thrombopoietin-receptor

www.thelancet.com Vol 383 June 28, 2014 2247


Seminar

agonists are not yet available for routine practice. 11 Pedersen-Bjergaard J, Daugaard G, Hansen SW, Philip P,
Antithymocyte globulins and hypomethylating agents Larsen SO, Rørth M. Increased risk of myelodysplasia and
leukaemia after etoposide, cisplatin, and bleomycin for germ-cell
give platelet responses in 35–40% of patients with lower- tumours. Lancet 1991; 338: 359–63.
risk myelodysplastic syndromes, as well as erythroid 12 McLaughlin P, Estey E, Glassman A, et al. Myelodysplasia and acute
responses, and can be used in such patients.159,160 myeloid leukemia following therapy for indolent lymphoma with
fludarabine, mitoxantrone, and dexamethasone (FND) plus
The classic IPSS,7 at diagnosis, can wrongly classify rituximab and interferon alpha. Blood 2005; 105: 4573–75.
some patients in lower-risk myelodysplastic syndromes, 13 Morrison VA, Rai KR, Peterson BL, et al. Therapy-related myeloid
because it does not incorporate marrow multilineage leukemias are observed in patients with chronic lymphocytic
leukemia after treatment with fludarabine and chlorambucil: results
dysplasia,74 erythrocyte transfusion dependence,7 severity of an intergroup study, cancer and leukemia group B 9011.
of thrombocytopenia,8 myelofibrosis, and somatic J Clin Oncol 2002; 20: 3878–84.
mutations, which are associated with poorer prognosis. 14 Iwanaga M, Hsu WL, Soda M, et al. Risk of myelodysplastic
syndromes in people exposed to ionizing radiation: a retrospective
The revised IPSS8 and specific scores designed for lower- cohort study of Nagasaki atomic bomb survivors. J Clin Oncol 2011;
risk myelodysplastic syndromes address some of these 29: 428–34.
weaknesses.8,44,64,75 Patients classed as lower risk by the 15 Cardis E, Vrijheid M, Blettner M, et al. Risk of cancer after low
doses of ionising radiation: retrospective cohort study in
IPSS who remain at lower risk, but have early resistance 15 countries. BMJ 2005; 331: 77.
to erythropoiesis-stimulating agents (patients with 16 Nisse C, Haguenoer JM, Grandbastien B, et al. Occupational and
non-deleted 5q)85 or resistance to lenalidomide (patients environmental risk factors of the myelodysplastic syndromes in the
North of France. Br J Haematol 2001; 112: 927–35.
with deleted 5q)134 also have poor survival.
17 Aksoy M, Ozeriş S, Sabuncu H, Inanici Y, Yanardağ R. Exposure to
Although prospective studies are scarce, hypo- benzene in Turkey between 1983 and 1985: a haematological study
methylating agents are increasingly used in some of on 231 workers. Br J Ind Med 1987; 44: 785–87.
these IPSS-defined lower-risk patients on the basis of 18 Rigolin GM, Cuneo A, Roberti MG, et al. Exposure to myelotoxic
agents and myelodysplasia: case-control study and correlation with
their effect on survival in higher-risk myelodysplastic clinicobiological findings. Br J Haematol 1998; 103: 189–97.
syndromes. Allogeneic stem-cell transplantation is also 19 Raza A, Gezer S, Mundle S, et al. Apoptosis in bone marrow biopsy
sometimes proposed in patients younger than 60–70 years samples involving stromal and hematopoietic cells in 50 patients
with myelodysplastic syndromes. Blood 1995; 86: 268–76.
in the absence of major response to hypomethylating
20 Raza A, Galili N. The genetic basis of phenotypic heterogeneity in
agents (or subsequent relapse). myelodysplastic syndromes. Nat Rev Cancer 2012; 12: 849–59.
Contributors 21 Corey SJ, Minden MD, Barber DL, Kantarjian H, Wang JC,
LA wrote the classification and treatment sections, RI wrote the biology Schimmer AD. Myelodysplastic syndromes: the complexity of
section, and PF wrote the incidence, diagnosis, and prognosis sections. stem-cell diseases. Nat Rev Cancer 2007; 7: 118–29.
22 Walter MJ, Shen D, Ding L, et al. Clonal architecture of secondary
Declaration of interests acute myeloid leukemia. N Engl J Med 2012; 366: 1090–98.
LA has received honoraria from Celgene, Novartis, Janssen, and Roche. 23 Parker JE, Mufti GJ, Rasool F, Mijovic A, Devereux S, Pagliuca A.
RI has received honoraria from Celgene. PF has received honoraria from The role of apoptosis, proliferation, and the Bcl-2-related proteins in
Celgene, Novartis, Janssen, and Roche. the myelodysplastic syndromes and acute myeloid leukemia
secondary to MDS. Blood 2000; 96: 3932–38.
References
1 Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point 24 Tehranchi R, Invernizzi R, Grandien A, et al. Aberrant
mutations in myelodysplastic syndromes. N Engl J Med 2011; mitochondrial iron distribution and maturation arrest characterize
364: 2496–506. early erythroid precursors in low-risk myelodysplastic syndromes.
Blood 2005; 106: 247–53.
2 Shen L, Kantarjian H, Guo Y, et al. DNA methylation predicts
survival and response to therapy in patients with myelodysplastic 25 Zang DY, Goodwin RG, Loken MR, Bryant E, Deeg HJ. Expression
syndromes. J Clin Oncol 2010; 28: 605–13. of tumor necrosis factor-related apoptosis-inducing ligand, Apo2L,
and its receptors in myelodysplastic syndrome: effects on in vitro
3 Will B, Zhou L, Vogler TO, et al. Stem and progenitor cells in hemopoiesis. Blood 2001; 98: 3058–65.
myelodysplastic syndromes show aberrant stage-specific expansion
and harbor genetic and epigenetic alterations. Blood 2012; 26 Claessens YE, Bouscary D, Dupont JM, et al. In vitro proliferation
120: 2076–86. and differentiation of erythroid progenitors from patients with
myelodysplastic syndromes: evidence for Fas-dependent apoptosis.
4 Jiang Y, Dunbar A, Gondek LP, et al. Aberrant DNA methylation is Blood 2002; 99: 1594–601.
a dominant mechanism in MDS progression to AML. Blood 2009;
113: 1315–25. 27 Bhagat TD, Zhou L, Sokol L, et al. miR-21 mediates hematopoietic
suppression in MDS by activating TGF-β signaling. Blood 2013;
5 Tefferi A, Vardiman JW. Myelodysplastic syndromes. N Engl J Med 121: 2875–81.
2009; 361: 1872–85.
28 Nilsson L, Astrand-Grundström I, Arvidsson I, et al. Isolation and
6 Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the characterization of hematopoietic progenitor/stem cells in
World Health Organization (WHO) classification of myeloid 5q-deleted myelodysplastic syndromes: evidence for involvement at
neoplasms and acute leukemia: rationale and important changes. the hematopoietic stem cell level. Blood 2000; 96: 2012–21.
Blood 2009; 114: 937–51.
29 Tehranchi R, Woll PS, Anderson K, et al. Persistent malignant stem
7 Greenberg P, Cox C, LeBeau MM, et al. International scoring cells in del(5q) myelodysplasia in remission. N Engl J Med 2010;
system for evaluating prognosis in myelodysplastic syndromes. 363: 1025–37.
Blood 1997; 89: 2079–88.
30 Boultwood J, Fidler C, Strickson AJ, et al. Narrowing and genomic
8 Greenberg PL, Tuechler H, Schanz J, et al. Revised international annotation of the commonly deleted region of the 5q- syndrome.
prognostic scoring system for myelodysplastic syndromes. Blood Blood 2002; 99: 4638–41.
2012; 120: 2454–65.
31 Ebert BL, Pretz J, Bosco J, et al. Identification of RPS14 as a
9 Neukirchen J, Schoonen WM, Strupp C, et al. Incidence and 5q- syndrome gene by RNA interference screen. Nature 2008;
prevalence of myelodysplastic syndromes: data from the Düsseldorf 451: 335–39.
MDS-registry. Leuk Res 2011; 35: 1591–96.
32 Barlow JL, Drynan LF, Hewett DR, et al. A p53-dependent
10 Matsuda A, Germing U, Jinnai I, et al. Difference in clinical mechanism underlies macrocytic anemia in a mouse model of
features between Japanese and German patients with refractory human 5q- syndrome. Nat Med 2010; 16: 59–66.
anemia in myelodysplastic syndromes. Blood 2005; 106: 2633–40.

2248 www.thelancet.com Vol 383 June 28, 2014


Seminar

33 Starczynowski DT, Kuchenbauer F, Argiropoulos B, et al. 56 Muguruma Y, Matsushita H, Yahata T, et al. Establishment of a
Identification of miR-145 and miR-146a as mediators of the xenograft model of human myelodysplastic syndromes.
5q- syndrome phenotype. Nat Med 2010; 16: 49–58. Haematologica 2011; 96: 543–51.
34 Jädersten M, Saft L, Smith A, et al. TP53 mutations in low-risk 57 Kordasti SY, Afzali B, Lim Z, et al. IL-17-producing CD4(+) T cells,
myelodysplastic syndromes with del(5q) predict disease pro-inflammatory cytokines and apoptosis are increased in low risk
progression. J Clin Oncol 2011; 29: 1971–79. myelodysplastic syndrome. Br J Haematol 2009; 145: 64–72.
35 Jerez A, Gondek LP, Jankowska AM, et al. Topography, clinical, and 58 Kordasti SY, Ingram W, Hayden J, et al. CD4+CD25high Foxp3+
genomic correlates of 5q myeloid malignancies revisited. regulatory T cells in myelodysplastic syndrome (MDS). Blood 2007;
J Clin Oncol 2012; 30: 1343–49. 110: 847–50.
36 Liu TX, Becker MW, Jelinek J, et al. Chromosome 5q deletion and 59 Costantini B, Kordasti S, Kulasekararaj AG, et al. The effects of
epigenetic suppression of the gene encoding alpha-catenin 5-azacytidine on the function and number of regulatory T-cells and
(CTNNA1) in myeloid cell transformation. Nat Med 2007; 13: 78–83. T-effectors in myelodysplastic syndrome. Haematologica 2013;
37 Joslin JM, Fernald AA, Tennant TR, et al. Haploinsufficiency of 98: 1196–205.
EGR1, a candidate gene in the del(5q), leads to the development of 60 Tsai HC, Li H, Van Neste L, et al. Transient low doses of
myeloid disorders. Blood 2007; 110: 719–26. DNA-demethylating agents exert durable antitumor effects on
38 Wei S, Chen X, Rocha K, et al. A critical role for phosphatase hematological and epithelial tumor cells. Cancer Cell 2012;
haplodeficiency in the selective suppression of deletion 5q MDS by 21: 430–46.
lenalidomide. Proc Natl Acad Sci USA 2009; 106: 12974–79. 61 Toma A, Fenaux P, Dreyfus F, Cordonnier C. Infections in
39 McNerney ME, Brown CD, Wang X, et al. CUX1 is a haploinsufficient myelodysplastic syndromes. Haematologica 2012; 97: 1459–70.
tumor suppressor gene on chromosome 7 frequently inactivated in 62 Hebbar M, Kozlowski D, Wattel E, et al. Association between
acute myeloid leukemia. Blood 2013; 121: 975–83. myelodysplastic syndromes and inflammatory bowel diseases.
40 Gurvich N, Perna F, Farina A, et al. L3MBTL1 polycomb protein, Report of seven new cases and review of the literature. Leukemia
a candidate tumor suppressor in del(20q12) myeloid disorders, is 1997; 11: 2188–91.
essential for genome stability. Proc Natl Acad Sci USA 2010; 63 Hebbar M, Brouillard M, Wattel E, et al. Association of
107: 22552–57. myelodysplastic syndrome and relapsing polychondritis: further
41 Aziz A, Baxter EJ, Edwards C, et al. Cooperativity of imprinted evidence. Leukemia 1995; 9: 731–33.
genes inactivated by acquired chromosome 20q deletions. 64 Della Porta MG, Malcovati L, Boveri E, et al. Clinical relevance of
J Clin Invest 2013; 123: 2169–82. bone marrow fibrosis and CD34-positive cell clusters in primary
42 Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of myelodysplastic syndromes. J Clin Oncol 2009; 27: 754–62.
mutations in epigenetic regulators in myeloid malignancies. 65 Verburgh E, Achten R, Maes B, et al. Additional prognostic value of
Nat Rev Cancer 2012; 12: 599–612. bone marrow histology in patients subclassified according to the
43 Damm F, Fontenay M, Bernard OA. Point mutations in International Prognostic Scoring System for myelodysplastic
myelodysplastic syndromes. N Engl J Med 2011; 365: 1154–55, author syndromes. J Clin Oncol 2003; 21: 273–82.
reply 155. 66 Westers TM, Ireland R, Kern W, et al. Standardization of flow
44 Bejar R, Stevenson KE, Caughey BA, et al. Validation of a prognostic cytometry in myelodysplastic syndromes: a report from an
model and the impact of mutations in patients with lower-risk international consortium and the European LeukemiaNet Working
myelodysplastic syndromes. J Clin Oncol 2012; 30: 3376–82. Group. Leukemia 2012; 26: 1730–41.
45 Moran-Crusio K, Reavie L, Shih A, et al. Tet2 loss leads to increased 67 Wimazal F, Fonatsch C, Thalhammer R, et al. Idiopathic cytopenia
hematopoietic stem cell self-renewal and myeloid transformation. of undetermined significance (ICUS) versus low risk MDS:
Cancer Cell 2011; 20: 11–24. the diagnostic interface. Leuk Res 2007; 31: 1461–68.
46 Quivoron C, Couronné L, Della Valle V, et al. TET2 inactivation 68 Haase D, Germing U, Schanz J, et al. New insights into the
results in pleiotropic hematopoietic abnormalities in mouse and is prognostic impact of the karyotype in MDS and correlation with
a recurrent event during human lymphomagenesis. Cancer Cell subtypes: evidence from a core dataset of 2124 patients. Blood 2007;
2011; 20: 25–38. 110: 4385–95.
47 Papaemmanuil E, Cazzola M, Boultwood J, et al, and the Chronic 69 Schanz J, Steidl C, Fonatsch C, et al. Coalesced multicentric
Myeloid Disorders Working Group of the International Cancer analysis of 2,351 patients with myelodysplastic syndromes indicates
Genome Consortium. Somatic SF3B1 mutation in myelodysplasia an underestimation of poor-risk cytogenetics of myelodysplastic
with ring sideroblasts. N Engl J Med 2011; 365: 1384–95. syndromes in the international prognostic scoring system.
48 Yoshida K, Sanada M, Shiraishi Y, et al. Frequent pathway mutations J Clin Oncol 2011; 29: 1963–70.
of splicing machinery in myelodysplasia. Nature 2011; 478: 64–69. 70 Braun T, de Botton S, Taksin AL, et al. Characteristics and outcome
49 Visconte V, Rogers HJ, Singh J, et al. SF3B1 haploinsufficiency of myelodysplastic syndromes (MDS) with isolated 20q deletion:
leads to formation of ring sideroblasts in myelodysplastic a report on 62 cases. Leuk Res 2011; 35: 863–67.
syndromes. Blood 2012; 120: 3173–86. 71 Busque L, Patel JP, Figueroa ME, et al. Recurrent somatic TET2
50 Xiao R, Sun Y, Ding JH, et al. Splicing regulator SC35 is essential mutations in normal elderly individuals with clonal hematopoiesis.
for genomic stability and cell proliferation during mammalian Nat Genet 2012; 44: 1179–81.
organogenesis. Mol Cell Biol 2007; 27: 5393–402. 72 Maassen A, Strupp C, Giagounidis A, et al. Validation and
51 Khan H, Vale C, Bhagat T, Verma A. Role of DNA methylation in proposals for a refinement of the WHO 2008 classification of
the pathogenesis and treatment of myelodysplastic syndromes. myelodysplastic syndromes without excess of blasts. Leuk Res
Semin Hematol 2013; 50: 16–37. 2013; 37: 64–70.
52 Pérez C, Martínez-Calle N, Martín-Subero JI, et al. TET2 mutations 73 Malcovati L, Papaemmanuil E, Bowen DT, et al, for the Chronic
are associated with specific 5-methylcytosine and Myeloid Disorders Working Group of the International Cancer
5-hydroxymethylcytosine profiles in patients with chronic Genome Consortium and of the Associazione Italiana per la Ricerca
myelomonocytic leukemia. PLoS One 2012; 7: e31605. sul Cancro Gruppo Italiano Malattie Mieloproliferative. Clinical
significance of SF3B1 mutations in myelodysplastic syndromes and
53 Marcondes AM, Mhyre AJ, Stirewalt DL, Kim SH, Dinarello CA,
myelodysplastic/myeloproliferative neoplasms. Blood 2011;
Deeg HJ. Dysregulation of IL-32 in myelodysplastic syndrome and
118: 6239–46.
chronic myelomonocytic leukemia modulates apoptosis and
impairs NK function. Proc Natl Acad Sci USA 2008; 105: 2865–70. 74 Malcovati L, Germing U, Kuendgen A, et al. Time-dependent
prognostic scoring system for predicting survival and leukemic
54 Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor
evolution in myelodysplastic syndromes. J Clin Oncol 2007;
dysfunction induces myelodysplasia and secondary leukaemia.
25: 3503–10.
Nature 2010; 464: 852–57.
75 Garcia-Manero G, Shan J, Faderl S, et al. A prognostic score for
55 Blau O, Baldus CD, Hofmann WK, et al. Mesenchymal stromal cells
patients with lower risk myelodysplastic syndrome. Leukemia 2008;
of myelodysplastic syndrome and acute myeloid leukemia patients
22: 538–43.
have distinct genetic abnormalities compared with leukemic blasts.
Blood 2011; 118: 5583–92.

www.thelancet.com Vol 383 June 28, 2014 2249


Seminar

76 Morel P, Declercq C, Hebbar M, Bauters F, Fenaux P. Prognostic 95 Zipperer E, Pelz D, Nachtkamp K, et al. The hematopoietic stem
factors in myelodysplastic syndromes: critical analysis of the impact cell transplantation comorbidity index is of prognostic relevance for
of age and gender and failure to identify a very-low-risk group using patients with myelodysplastic syndrome. Haematologica 2009;
standard mortality ratio techniques. Br J Haematol 1996; 94: 116–19. 94: 729–32.
77 Naqvi K, Garcia-Manero G, Sardesai S, et al. Association of 96 Anderson JE, Appelbaum FR, Schoch G, et al. Allogeneic marrow
comorbidities with overall survival in myelodysplastic syndrome: transplantation for myelodysplastic syndrome with advanced
development of a prognostic model. J Clin Oncol 2011; 29: 2240–46. disease morphology: a phase II study of busulfan,
78 Sperr WR, Wimazal F, Kundi M, et al. Comorbidity as prognostic cyclophosphamide, and total-body irradiation and analysis of
variable in MDS: comparative evaluation of the HCT-CI and CCI in prognostic factors. J Clin Oncol 1996; 14: 220–26.
a core dataset of 419 patients of the Austrian MDS Study Group. 97 Arnold R, de Witte T, van Biezen A, et al, for the European Blood
Ann Oncol 2010; 21: 114–19. and Marrow Transplantation Group. Unrelated bone marrow
79 Thol F, Friesen I, Damm F, et al. Prognostic significance of ASXL1 transplantation in patients with myelodysplastic syndromes and
mutations in patients with myelodysplastic syndromes. J Clin Oncol secondary acute myeloid leukemia: an EBMT survey.
2011; 29: 2499–506. Bone Marrow Transplant 1998; 21: 1213–16.
80 Thol F, Kade S, Schlarmann C, et al. Frequency and prognostic 98 Sierra J, Pérez WS, Rozman C, et al. Bone marrow transplantation
impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with from HLA-identical siblings as treatment for myelodysplasia. Blood
myelodysplastic syndromes. Blood 2012; 119: 3578–84. 2002; 100: 1997–2004.
81 van de Loosdrecht AA, Westers TM, Westra AH, Dräger AM, 99 Castro-Malaspina H, Harris RE, Gajewski J, et al. Unrelated donor
van der Velden VH, Ossenkoppele GJ. Identification of distinct marrow transplantation for myelodysplastic syndromes: outcome
prognostic subgroups in low- and intermediate-1-risk myelodysplastic analysis in 510 transplants facilitated by the National Marrow Donor
syndromes by flow cytometry. Blood 2008; 111: 1067–77. Program. Blood 2002; 99: 1943–51.
82 Germing U, Hildebrandt B, Pfeilstöcker M, et al. Refinement of the 100 Koreth J, Pidala J, Perez WS, et al. Role of reduced-intensity
international prognostic scoring system (IPSS) by including LDH conditioning allogeneic hematopoietic stem-cell transplantation in
as an additional prognostic variable to improve risk assessment in older patients with de novo myelodysplastic syndromes: an
patients with primary myelodysplastic syndromes (MDS). Leukemia international collaborative decision analysis. J Clin Oncol 2013;
2005; 19: 2223–31. 31: 2662–70.
83 Malcovati L, Della Porta MG, Cazzola M. Predicting survival and 101 Knipp S, Hildebrand B, Kündgen A, et al. Intensive chemotherapy
leukemic evolution in patients with myelodysplastic syndrome. is not recommended for patients aged >60 years who have
Haematologica 2006; 91: 1588–90. myelodysplastic syndromes or acute myeloid leukemia with
84 Hellström-Lindberg E, Negrin R, Stein R, et al. Erythroid response high-risk karyotypes. Cancer 2007; 110: 345–52.
to treatment with G-CSF plus erythropoietin for the anaemia of 102 Kantarjian H, Beran M, Cortes J, et al. Long-term follow-up results
patients with myelodysplastic syndromes: proposal for a predictive of the combination of topotecan and cytarabine and other intensive
model. Br J Haematol 1997; 99: 344–51. chemotherapy regimens in myelodysplastic syndrome. Cancer 2006;
85 Kelaidi C, Park S, Sapena R, et al. Long-term outcome of anemic 106: 1099–109.
lower-risk myelodysplastic syndromes without 5q deletion refractory 103 Wattel E, Solary E, Leleu X, et al. A prospective study of
to or relapsing after erythropoiesis-stimulating agents. Leukemia autologous bone marrow or peripheral blood stem cell
2013; 27: 1283–90. transplantation after intensive chemotherapy in myelodysplastic
86 Itzykson R, Thépot S, Quesnel B, et al, and the Groupe syndromes. Groupe Français des Myélodysplasies. Group
Francophone des Myelodysplasies(GFM). Prognostic factors for Ouest-Est d’étude des Leucémies aiguës myéloïdes. Leukemia
response and overall survival in 282 patients with higher-risk 1999; 13: 524–29.
myelodysplastic syndromes treated with azacitidine. Blood 2011; 104 Estey EH, Thall PF, Cortes JE, et al. Comparison of idarubicin +
117: 403–11. ara-C-, fludarabine + ara-C-, and topotecan + ara-C-based regimens
87 Itzykson R, Kosmider O, Cluzeau T, et al, and the Groupe in treatment of newly diagnosed acute myeloid leukemia, refractory
Francophone des Myelodysplasies (GFM). Impact of TET2 mutations anemia with excess blasts in transformation, or refractory anemia
on response rate to azacitidine in myelodysplastic syndromes and with excess blasts. Blood 2001; 98: 3575–83.
low blast count acute myeloid leukemias. Leukemia 2011; 25: 1147–52. 105 Sekeres MA, Maciejewski JP, Erba HP, et al. A Phase 2 study of
88 Kulasekararaj AG, Mohamedali AM, Smith AE, et al. Polycomb combination therapy with arsenic trioxide and gemtuzumab
Complex Group Gene Mutations and Their Prognostic Relevance In ozogamicin in patients with myelodysplastic syndromes or
5-Azacitidine Treated Myelodysplastic Syndrome Patients. secondary acute myeloid leukemia. Cancer 2011; 117: 1253–61.
ASH Annual Meeting Abstracts 2010; 116: 125-. 106 Estey EH, Thall PF, Giles FJ, et al. Gemtuzumab ozogamicin with
89 Kulasekararaj AG, Smith AE, Mian SA, et al. TP53 mutations in or without interleukin 11 in patients 65 years of age or older with
myelodysplastic syndrome are strongly correlated with aberrations untreated acute myeloid leukemia and high-risk myelodysplastic
of chromosome 5, and correlate with adverse prognosis. syndrome: comparison with idarubicin plus continuous-infusion,
Br J Haematol 2013; 160: 660–72. high-dose cytosine arabinoside. Blood 2002; 99: 4343–49.
90 Traina F, Jankowska AM, Visconte V, et al. Impact of Molecular 107 Cheson BD, Simon R. Low-dose ara-C in acute nonlymphocytic
Mutations on Treatment Response to Hypomethylating Agents in leukemia and myelodysplastic syndromes: a review of 20 years’
MDS. ASH Annual Meeting Abstracts 2011; 118: 461. experience. Semin Oncol 1987; 14 (suppl 1): 126–33.
91 List A, Giagounidis A, Backstrom J, Fu T, Fenaux P. Early 108 Fenaux P, Lai JL, Gardin C, Bauters F. Cytogenetics are a predictive
lenalidomide (LEN) dose intensity and durable RBC-transfusion factor of response to low dose Ara-C in acute myelogenous
independence (RBC-TI) in patients (pts) with low-/int-1-risk leukemia (AML) in the elderly. Leukemia 1990; 4: 312.
myelodysplastic syndromes (MDS) and del5q. J Clin Oncol 2011; 109 Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al, for the
29: 6522. International Vidaza High-Risk MDS Survival Study Group.
92 Cutler CS, Lee SJ, Greenberg P, et al. A decision analysis of Efficacy of azacitidine compared with that of conventional care
allogeneic bone marrow transplantation for the myelodysplastic regimens in the treatment of higher-risk myelodysplastic
syndromes: delayed transplantation for low-risk myelodysplasia is syndromes: a randomised, open-label, phase III study.
associated with improved outcome. Blood 2004; 104: 579–85. Lancet Oncol 2009; 10: 223–32.
93 Sorror ML, Sandmaier BM, Storer BE, et al. Comorbidity and 110 Fenaux P, Gattermann N, Seymour JF, et al. Prolonged survival
disease status based risk stratification of outcomes among patients with improved tolerability in higher-risk myelodysplastic
with acute myeloid leukemia or myelodysplasia receiving allogeneic syndromes: azacitidine compared with low dose ara-C.
hematopoietic cell transplantation. J Clin Oncol 2007; 25: 4246–54. Br J Haematol 2010; 149: 244–49.
94 Sperr WR, Wimazal F, Kundi M, et al. Survival analysis and AML 111 Silverman LR, Demakos EP, Peterson BL, et al. Randomized
development in patients with de novo myelodysplastic syndromes: controlled trial of azacitidine in patients with the myelodysplastic
comparison of six different prognostic scoring systems. syndrome: a study of the cancer and leukemia group B. J Clin Oncol
Ann Hematol 2001; 80: 272–77. 2002; 20: 2429–40.

2250 www.thelancet.com Vol 383 June 28, 2014


Seminar

112 Silverman LR, Fenaux P, Mufti GJ, et al. Continued azacitidine 130 Jädersten M, Malcovati L, Dybedal I, et al. Erythropoietin and
therapy beyond time of first response improves quality of response granulocyte-colony stimulating factor treatment associated with
in patients with higher-risk myelodysplastic syndromes. Cancer improved survival in myelodysplastic syndrome. J Clin Oncol 2008;
2011; 117: 2697–702. 26: 3607–13.
113 Gore SD, Fenaux P, Santini V, et al. A multivariate analysis of the 131 Greenberg PL, Sun Z, Miller KB, et al. Treatment of myelodysplastic
relationship between response and survival among patients with syndrome patients with erythropoietin with or without granulocyte
higher-risk myelodysplastic syndromes treated within azacitidine or colony-stimulating factor: results of a prospective randomized
conventional care regimens in the randomized AZA-001 trial. phase 3 trial by the Eastern Cooperative Oncology Group (E1996).
Haematologica 2013; 98: 1067–72. Blood 2009; 114: 2393–400.
114 Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves 132 Kelaidi C, Park S, Brechignac S, et al, and the Groupe
patient outcomes in myelodysplastic syndromes: results of a Francophone des Myélodysplasies (GFM). Treatment of
phase III randomized study. Cancer 2006; 106: 1794–803. myelodysplastic syndromes with 5q deletion before the
115 Lübbert M, Suciu S, Baila L, et al. Low-dose decitabine versus best lenalidomide era; the GFM experience with EPO and thalidomide.
supportive care in elderly patients with intermediate- or high-risk Leuk Res 2008; 32: 1049–53.
myelodysplastic syndrome (MDS) ineligible for intensive 133 List A, Dewald G, Bennett J, et al, for the Myelodysplastic
chemotherapy: final results of the randomized phase III study of Syndrome-003 Study Investigators. Lenalidomide in the
the European Organisation for Research and Treatment of Cancer myelodysplastic syndrome with chromosome 5q deletion.
Leukemia Group and the German MDS Study Group. J Clin Oncol N Engl J Med 2006; 355: 1456–65.
2011; 29: 1987–96. 134 Fenaux P, Giagounidis A, Selleslag D, et al, for the MDS-004
116 Lee YG, Kim I, Yoon SS, et al, for the Korean Society of Lenalidomide del5q Study Group. A randomized phase 3 study of
Haematology AML/MDS working party. Comparative analysis lenalidomide versus placebo in RBC transfusion-dependent patients
between azacitidine and decitabine for the treatment of with low-/intermediate-1-risk myelodysplastic syndromes with
myelodysplastic syndromes. Br J Haematol 2013; 161: 339–47. del5q. Blood 2011; 118: 3765–76.
117 Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a 135 Giagounidis AA, Kulasekararaj A, Germing U, et al. Long-term
randomized study of 3 schedules of low-dose decitabine in transfusion independence in del(5q) MDS patients who discontinue
higher-risk myelodysplastic syndrome and chronic myelomonocytic lenalidomide. Leukemia 2012; 26: 855–58.
leukemia. Blood 2007; 109: 52–57. 136 Adès L, Le Bras F, Sebert M, et al. Treatment with lenalidomide
118 Steensma DP, Baer MR, Slack JL, et al. Multicenter study of does not appear to increase the risk of progression in lower risk
decitabine administered daily for 5 days every 4 weeks to adults with myelodysplastic syndromes with 5q deletion. A comparative
myelodysplastic syndromes: the alternative dosing for outpatient analysis by the Groupe Francophone des Myelodysplasies.
treatment (ADOPT) trial. J Clin Oncol 2009; 27: 3842–48. Haematologica 2012; 97: 213–18.
119 Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B, et al. 137 Kuendgen A, Lauseker M, List AF, et al. Lenalidomide does not
Phase 1/2 study of the combination of 5-aza-2´-deoxycytidine with increase AML progression risk in RBC transfusion-dependent
valproic acid in patients with leukemia. Blood 2006; 108: 3271–79. patients with Low- or Intermediate-1-risk MDS with del(5q):
120 Silverman L, Verma A, Odchimar-Reissig R, et al. A phase I/II study of a comparative analysis. Leukemia 2013; 27: 1072–79.
vorinostat, an oral histone deacetylase inhibitor, in combination with 138 Passweg JR, Giagounidis AA, Simcock M, et al.
azacitidine in patients with the myelodysplastic syndrome (MDS) and Immunosuppressive therapy for patients with myelodysplastic
acute myeloid leukemia (AML). Initial results of the phase I trial: A syndrome: a prospective randomized multicenter phase III trial
New York Cancer Consortium. J Clin Oncol 2008; 26: abstr 7000. comparing antithymocyte globulin plus cyclosporine with best
121 Prebet T, Gore SD, Sun Z, et al. Prolonged Administration of supportive care--SAKK 33/99. J Clin Oncol 2011; 29: 303–09.
Azacitidine with or without Entinostat Increases Rate of 139 Molldrem JJ, Caples M, Mavroudis D, Plante M, Young NS,
Hematologic Normalization for Myelodysplastic Syndrome and Barrett AJ. Antithymocyte globulin for patients with
Acute Myeloid Leukemia with Myelodysplasia-Related Changes: myelodysplastic syndrome. Br J Haematol 1997; 99: 699–705.
Results of the US Leukemia Intergroup Trial E1905. 140 Sloand EM, Wu CO, Greenberg P, Young N, Barrett J. Factors
ASH Annual Meeting Abstracts 2010; 116: 601. affecting response and survival in patients with myelodysplasia
122 Sekeres MA, Tiu RV, Komrokji R, et al. Phase 2 study of the treated with immunosuppressive therapy. J Clin Oncol 2008;
lenalidomide and azacitidine combination in patients with 26: 2505–11.
higher-risk myelodysplastic syndromes. Blood 2012; 120: 4945–51. 141 Saunthararajah Y, Nakamura R, Wesley R, Wang Q J, Barrett AJ.
123 Raza A, Mehdi M, Mumtaz M, Ali F, Lascher S, Galili N. A simple method to predict response to immunosuppressive
Combination of 5-azacytidine and thalidomide for the treatment of therapy in patients with myelodysplastic syndrome. Blood 2003;
myelodysplastic syndromes and acute myeloid leukemia. Cancer 102: 3025–27.
2008; 113: 1596–604. 142 Silverman LR, McKenzie DR, Peterson BL, et al, for the Cancer and
124 Nand S, Godwin J, Smith S, et al. Hydroxyurea, azacitidine and Leukemia Group B. Further analysis of trials with azacitidine in
gemtuzumab ozogamicin therapy in patients with previously patients with myelodysplastic syndrome: studies 8421, 8921, and
untreated non-M3 acute myeloid leukemia and high-risk 9221 by the Cancer and Leukemia Group B. J Clin Oncol 2006;
myelodysplastic syndromes in the elderly: results from a pilot trial. 24: 3895–903.
Leuk Lymphoma 2008; 49: 2141–47. 143 Boehrer S, Beyne-Rauzy O, Prebet T, et al. Interim results of a
125 Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment randomized phase II trial of azacitidine (AZA) +/− epo in lower risk
of imminent relapse in MDS or AML patients after allogeneic myelodysplastic syndrome (MDS) resistant to an erythropoietic
HSCT: results of the RELAZA trial. Leukemia 2012; 26: 381–89. stimulating agent (ESA) alone. Blood 2010; 116: 784.
126 Fenaux P, Rose C. Impact of iron overload in myelodysplastic 144 Raza A, Reeves JA, Feldman EJ, et al. Phase 2 study of lenalidomide
syndromes. Blood Rev 2009; 23 (suppl 1): S15–19. in transfusion-dependent, low-risk, and intermediate-1 risk
127 Crawford J, Cella D, Cleeland CS, et al. Relationship between myelodysplastic syndromes with karyotypes other than deletion 5q.
changes in hemoglobin level and quality of life during Blood 2008; 111: 86–93.
chemotherapy in anemic cancer patients receiving epoetin alfa 145 Komrokji RS, Lancet JE, Swern AS, et al. Combined treatment with
therapy. Cancer 2002; 95: 888–95. lenalidomide and epoetin alfa in lower-risk patients with
128 Hellström-Lindberg E, Gulbrandsen N, Lindberg G, et al, and the myelodysplastic syndrome. Blood 2012; 120: 3419–24.
Scandinavian MDS Group. A validated decision model for treating 146 Davis BA, O’Sullivan C, Jarritt PH, Porter JB. Value of sequential
the anaemia of myelodysplastic syndromes with erythropoietin + monitoring of left ventricular ejection fraction in the management
granulocyte colony-stimulating factor: significant effects on quality of thalassemia major. Blood 2004; 104: 263–69.
of life. Br J Haematol 2003; 120: 1037–46. 147 Jensen PD, Jensen FT, Christensen T, Eiskjaer H, Baandrup U,
129 Park S, Grabar S, Kelaidi C, et al, for the GFM group (Groupe Nielsen JL. Evaluation of myocardial iron by magnetic resonance
Francophone des Myélodysplasies). Predictive factors of response and imaging during iron chelation therapy with deferrioxamine:
survival in myelodysplastic syndrome treated with erythropoietin and indication of close relation between myocardial iron content and
G-CSF: the GFM experience. Blood 2008; 111: 574–82. chelatable iron pool. Blood 2003; 101: 4632–39.

www.thelancet.com Vol 383 June 28, 2014 2251


Seminar

148 Rose C, Brechignac S, Vassilief D, et al, for the GFM (Groupe 155 Hellström-Lindberg E, Ahlgren T, Beguin Y, et al. Treatment of
Francophone des Myélodysplasies). Does iron chelation therapy anemia in myelodysplastic syndromes with granulocyte
improve survival in regularly transfused lower risk MDS patients? colony-stimulating factor plus erythropoietin: results from a
A multicenter study by the GFM (Groupe Francophone des randomized phase II study and long-term follow-up of 71 patients.
Myélodysplasies). Leuk Res 2010; 34: 864–70. Blood 1998; 92: 68–75.
149 Leitch HA. Improving clinical outcome in patients with 156 Wattel E, Preudhomme C, Hecquet B, et al. p53 mutations are
myelodysplastic syndrome and iron overload using iron chelation associated with resistance to chemotherapy and short survival in
therapy. Leuk Res 2007; 31 (suppl 3): S7–9. hematologic malignancies. Blood 1994; 84: 3148–57.
150 Neukirchen J, Fox F, Kündgen A, et al. Improved survival in MDS 157 Kantarjian H, Fenaux P, Sekeres MA, et al. Phase 1/2 study of AMG
patients receiving iron chelation therapy – a matched pair analysis 531 in thrombocytopenic patients (pts) with low-risk
of 188 patients from the Düsseldorf MDS registry. Leuk Res 2012; myelodysplastic syndrome (MDS): update including extended
36: 1067–70. treatment. ASH Annual Meeting Abstracts 2007; 110: 250.
151 Gattermann N, Porter J, Lopes L, Seymour J. Consensus statement 158 Kantarjian HM, Mufti GJ, Fenaux P, et al. Treatment with the
on iron overload in myelodysplastic syndromes. thrombopoietin (TPO)-receptor agonist romiplostim in
Hematol Oncol Clin North Am 2005; 19 (suppl 1): 18–25. thrombocytopenic patients (Pts) with low or intermediate-1 (int-1)
152 Greenberg PL. Myelodysplastic syndromes: iron overload risk myelodysplastic syndrome (MDS): follow-up AML and survival
consequences and current chelating therapies. results of a randomized, double-blind, placebo (PBO)-controlled
J Natl Compr Canc Netw 2006; 4: 91–96. study. ASH Annual Meeting Abstracts 2012; 120: 421.
153 Alessandrino EP, Della Porta MG, Bacigalupo A, et al. Prognostic 159 Lim ZY, Killick S, Germing U, et al. Low IPSS score and bone
impact of pre-transplantation transfusion history and secondary marrow hypocellularity in MDS patients predict hematological
iron overload in patients with myelodysplastic syndrome responses to antithymocyte globulin. Leukemia 2007; 21: 1436–41.
undergoing allogeneic stem cell transplantation: a study from the 160 Broliden PA, Dahl IM, Hast R, et al. Antithymocyte globulin and
Gruppo Italiano Trapianto di Midollo Osseo (GITMO). cyclosporine A as combination therapy for low-risk non-
Haematologica 2010; 95: 476–84. sideroblastic myelodysplastic syndromes. Haematologica 2006;
154 Armand P, Kim HT, Cutler CS, et al. Prognostic impact of elevated 91: 667–70.
pretransplantation serum ferritin in patients undergoing
myeloablative stem cell transplantation. Blood 2007; 109: 4586–88.

2252 www.thelancet.com Vol 383 June 28, 2014

You might also like