ArticlePDF Available

Age, JAK2V617F and SF3B1 mutations are the main predicting factors for survival in refractory anaemia with ring sideroblasts and marked thrombocytosis

Authors:

Abstract and Figures

Refractory anaemia with ring sideroblasts (RARS) and marked thrombocytosis (RARS-T) is a provisional entity in the World Health Organisation 2008 classification and has previously been shown to have a high proportion of JAK2V617F (Janus Kinase 2) and SF3B1 (Splicing Factor 3B subunit 1) mutations. The purpose of the present study was to analyse the frequency of SF3B1 mutations in a large cohort of 111 patients with RARS-T and 33 patients with RARS and to explore the prognostic impact of SF3B1 mutational status on RARS-T. The frequency of SF3B1 mutations in RARS-T (96/111, 86.5%) and RARS (28/33, 84.8%) was similar. In RARS-T, median survival was better in SF3B1-mutated patients than in SF3B1-non-mutated patients (6.9 and 3.3 years, respectively, P=0.003). RARS can be differentiated from RARS-T by the frequency of JAK2V617F (0% vs 48.6%). In RARS-T patients, SF3B1 (P=0.021) and JAK2 mutations (P=0.016) were independent factors for a better prognosis. Altogether, our results confirm that RARS-T is an independent entity that should be recognised by the next World Health Organisation classification. The assessment of SF3B1 mutations is of prognostic interest in RARS-T patients. Younger age, JAK2V617F and SF3B1 mutations are the main predicting factors for survival in RARS-T.
Content may be subject to copyright.
ORIGINAL ARTICLE
Age, JAK2
V617F
and SF3B1 mutations are the main predicting
factors for survival in refractory anaemia with ring sideroblasts
and marked thrombocytosis
J Brose´us
1
, T Alpermann
2
, M Wulfert
3
, L Florensa Brichs
4,10
, S Jeromin
2
, E Lippert
5
, M Rozman
6,10
, F Lifermann
7
, V Grossmann
2
,
T Haferlach
2
, U Germing
3
, E Lun˜o
8,10
, F Girodon
9
and S Schnittger
2
for the MPN and MPNr-EuroNet (COST Action BM0902)
Refractory anaemia with ring sideroblasts (RARS) and marked thrombocytosis (RARS-T) is a provisional entity in the World Health
Organisation 2008 classification and has previously been shown to have a high proportion of JAK2
V617F
(Janus Kinase 2) and SF3B1
(Splicing Factor 3B subunit 1) mutations. The purpose of the present study was to analyse the frequency of SF3B1 mutations in a
large cohort of 111 patients with RARS-T and 33 patients with RARS and to explore the prognostic impact of SF3B1 mutational
status on RARS-T. The frequency of SF3B1 mutations in RARS-T (96/111, 86.5%) and RARS (28/33, 84.8%) was similar. In RARS-T,
median survival was better in SF3B1-mutated patients than in SF3B1-non-mutated patients (6.9 and 3.3 years, respectively,
P¼0.003). RARS can be differentiated from RARS-T by the frequency of JAK2
V617F
(0% vs 48.6%). In RARS-T patients, SF3B1
(P¼0.021) and JAK2 mutations (P¼0.016) were independent factors for a better prognosis. Altogether, our results confirm that
RARS-T is an independent entity that should be recognised by the next World Health Organisation classification. The assessment of
SF3B1 mutations is of prognostic interest in RARS-T patients. Younger age, JAK2
V617F
and SF3B1 mutations are the main predicting
factors for survival in RARS-T.
Leukemia (2013) 27, 1826–1831; doi:10.1038/leu.2013.120
Keywords: refractory anaemia with ring sideroblasts and marked thrombocytosis; SF3B1; prognostic impact; survival
INTRODUCTION
Refractory anaemia with ring sideroblasts (RARS) and marked
thrombocytosis (RARS-T) has been proposed in the World Health
Organisation 2001 classification of tumours of haematopoietic
and lymphoid tissues and retained as a provisional entity in the
classification of 2008.
1
RARS-T is classified in the myelodysplastic/
myeloproliferative (MDS/MPN) disorders group, because it
presents with the dysplastic features of RARS
2
and the
myeloproliferative features of essential thrombocythemia (ET).
3
RARS-T is characterised by a high rate of JAK2
V617F
(Janus Kinase 2)
mutations
4–15
and/or the presence of the mutation MPL
W515L/R
(MyeloProliferative Leukemia).
16,17
The classification of RARS-T as an
entity that is independent from RARS or ET is currently a matter of
debate. Several specialists favour the hypothesis that RARS-T is a
form of ET with Z15% of ring sideroblasts in the bone marrow
18
while others think that RARS-T develops from RARS with
secondary thrombocytosis accompanying the acquisition of the
JAK2
V617F
mutation.
19
Recent publication from our group in a European retrospective
multicentre collaborative study demonstrated that RARS-T was
independent from RARS and ET from a clinical and biological as
well as prognostic point of view.
20
Our results have recently been strengthened by the discovery of
the association between myelodysplastic syndromes and muta-
tions involving components of the RNA splicing machinery,
including U2AF35 (U2 small nuclear RNA Auxiliary Factor 35), ZRSR2
(Zinc finger CCCH type,RNA-binding motif and Serine/aRginine
rich 2), SRSF2 (Serine/aRginine-rich Splicing Factor 2) and SF3B1
(Splicing Factor 3B subunit 1). SF3B1 mutations (SF3B1
mut
) are found
in about 20% of total MDS and correlate strongly with the
presence of Z15% of ring sideroblasts (MDS-RS; 64–82.6% in
RARS, 57–76% in refractory cytopenia with multilineage dysplasia
and ring sideroblasts (RCMD-RS) and 66.7–72% in RARS-T).
21–27
On
the other hand, mutations of SF3B1 are found at a lower frequency
in MDS with o15% ring sideroblasts, which confirms the
specificity of SF3B1
mut
in MDS-RS. SF3B1 mutations are rare in
myeloproliferative neoplasms and particularly in ET (0–3%).
22,28
The high frequency of SF3B1 mutations suggests that these
mutations have a pathophysiological role in these diseases,
probably through perturbations of RNA splicing. The link
between SF3B1-mutated status and ring sideroblasts has been
confirmed in a recent experimental study on murine models.
29
About one quarter of MDS-RS are SF3B1
wt
and somatic mutations
of SRSF2 or ZRSR2 have been described in about 7% of MDS-RS,
21
1
Haematology Laboratory, University Hospital, Nancy, France;
2
MLL Munich Leukemia Laboratory, Munich, Germany;
3
Department of Haematology, Oncology and Clinical
Immunology, Heinrich-Heine-Universita
¨t, Du¨ sseldorf, Germany;
4
Laboratorio Citologia Hematolo
`gica, Servicio Patologia, Hospital del Mar, Barcelona, Spain;
5
Haematology
Laboratory, University Hospital, Bordeaux, France;
6
Unidad de Hematopatologı
´a, Departamento de Patologı
´a, Hospital Clı
´nic, IDIBAPS, Barcelona, Spain;
7
Department of Internal
Medicine, Hospital of Dax, Dax, France;
8
Servicio de Hematologı
´a, Servicio de Salud del Principado de Asturias, Oviedo, Spain and
9
Haematology Laboratory, University Hospital,
Dijon, France. Correspondence: Professor F Girodon, Haematology Laboratory, University Hospital, Plateau technique de biologie, 2 rue Ange
´lique Ducoudray, Dijon, cedex 21079,
France or Dr S Schnittger, MLL Munich Leukemia Laboratory, Munich, Germany.
E-mail: francois.girodon@chu-dijon.fr or susanne.schnittger@mll.com
10
On behalf of the Spanish Group of Hematological Cytology (GECH).
Received 20 March 2013; accepted 10 April 2013; accepted article preview online 18 April 2013; advance online publication, 14 May 2013
Leukemia (2013) 27, 1826– 1831
&
2013 Macmillan Publishers Limited All rights reserved 0887-6924/13
www.nature.com/leu
which suggests that other mutant genes may have a role in the
appearance of ring sideroblasts. Furthermore, a recent study
showed that RARS-T presented with a particular genetic pattern
with a high frequency of JAK2
V617F
and SF3B1 mutations,
confirming the classification of RARS-T in the category of
myelodysplastic/myeloproliferative neoplasms.
30
Finally, as precedent studies have been performed on little
RARS-T cohorts, the prognostic impact of SF3B1
mut
status remains
controversial,
22,24,25,27,31
and there is a need for a study on a larger
cohort. Our purpose was to analyse the frequency of SF3B1
mutations in a large cohort of 111 RARS-T and to explore the
prognostic impact of SF3B1 mutations in this disorder.
PATIENTS AND METHODS
Patient selection
According to the World Health Organisation 2008 classification, patients
were diagnosed with RARS-T if they fulfilled the following criteria: (i)
anaemia (haemoglobin level o125 g/l for females and o135 g/l for males)
with erythroid dysplasia and Z15% ring sideroblasts; (ii) thrombocytosis of
Z450 10
9
platelets/l; (iii) o5% blast cells in the bone marrow; (iv) the
presence of large atypical megakaryocytes similar to those observed in
BCR-ABL1-negative myeloproliferative neoplasms; (v) no secondary cause
of ring sideroblasts; and (vi) no karyotype abnormalities, such as del(5q),
t(3;3)(q21;q26) or inv(3)(q21q26).
1
To obtain a comprehensive data set of this rare entity, samples from
seven European centres in three European countries were collected. The
total cohort comprised 111 cases of RARS-T and 33 cases of RARS.
Data record
For each patient, demographic (gender, age at diagnosis, date of death or
last follow-up) and biological data (blood cell count, bone marrow
exploration, ring sideroblasts, karyotype, molecular explorations) were
collected.
The SF3B1 mutations were analysed with a sensitive next-generation
amplicon deep-sequencing assay (454 Life Sciences, Branford, CT, USA)
with a median coverage of 500 reads. This approach was able to detect
mutations with a sensitivity o1%.
The JAK2
V617F
mutation was analysed by allele-specific real-time PCR to
estimate allele burden according to methods published by Lippert et al.
32
and Schnittger et al.
33
with a sensitivity of 1%. JAK2
exon12
analysis was
performed according to the method of Schnittger et al.,
34
and the MPL
W515
mutations were analysed by high-resolution melting curve analyses
followed by Sanger sequencing if positive, as previously published by
Schnittger et al.
35
Statistical analyses
Standardised overall survival was estimated by the Kaplan–Meier method
and based on the time from diagnosis to death or last contact. Survival
curves for the different groups were compared using the log rank test.
Multivariate analysis was performed using Cox’s proportional hazards
model.
Approval for the study was obtained from the ethics committee of each
institution, and the procedures were carried out in accordance with the
Helsinki Declaration of 1975, as revised in 2000.
RESULTS
Demographic and biological features
A total of 144 cases (111 RARS-T and 33 RARS including 72 males
and 72 females) were recorded in the study. Median age at
diagnosis was 73.9 years (range: 44.4–96.1 years). The median
follow-up was 37.5 and 55.2 months for the RARS-T and RARS
cohort, respectively (Table 1). Survival data were available in 130
(97 RARS-T and 33 RARS) of the 144 patients.
Frequencies and characterisation of mutations
A karyotype was available in 128 cases. One hundred and ten
(85.9%) patients carried a normal karyotype, whereas 18 (14.1%)
patients showed aberrant karyotypes, which was equally
distributed between RARS-T and RARS patients. Even if the IPSS
(International Prognostic Scoring System) score can only be
applied to MDS de novo, we calculated it to check if we had a
homogeneous group of patients. Most of the patients of the total
cohort (133 out of 144) had an IPSS score of 0.
ASF3B1
mut
was noted in 124 out of the 144 patients (86.1%).
A total of 127 SF3B1 mutations were detected in these 124
patients (28 RARS and 96 RARS-T). Three RARS-T cases carried two
Table 1. Demographic and biological characteristics of RARS-T and
RARS patients
RARS-T patients RARS patients
n111 33
Male (%) 46.8 60.6
Age at diagnosis (years)
Median 74.3 71.1
Range 44.4–92.1 48.4–96.1
20–50 years, n(%) 4 (3.6) 2 (6.1)
50–70 years, n(%) 36 (32.5) 9 (27.2)
470 years, n(%) 71 (63.9) 22 (66.7)
Available survival data (n)97 33
Median follow-up (years) 3.1 4.6
WBC ( 10
9
/l)
Median 7.6 5.2
Range 2.1–60.0 1.6–17.3
Hb (g/l)
Median 96.5 91.0
Range 51.0–131.0 69.0–128.0
Platelets ( 10
9
/l)
Median 646 314
Range 452–1500 61–444
450–600, n(%) 54 (48.6)
4600, n(%) 57 (51.4)
Ring sideroblasts (%)
Median 52 40
Range 16–97 19–85
SF3B1 mutations (%) 86.5 84.8
Tested (n) 111 33
Mutated (n)9628
p.Lys700Glu 51 16
p.Lys666Glu/Thr/Asp/Asn 16 2
p.His662Asp/Gln 11 2
p.Glu622Asp 7 2
p.Arg625Gys/Leu/Gly 5 3
p.Thr663Pro 2 2
p.Met784_Lys785delinsIle 1 0
p.Asp781Gly 0 1
Two different mutations 3 0
JAK2
V617F
mutations (%) 48.6 0
Tested (n) 111 33
MPL mutations (%) 1 0
Tested (n) 102 27
IPSS
0 104 29
0.5 5 3
121
Abbreviations: Hb, haemoglobin; IPSS, International Prognostic Scoring
System; JAK2, Janus Kinase 2; MPL, MyeloProliferative Leukaemia; RARS,
refractory anaemia with ring sideroblasts; RARS-T, refractory anaemia with
ring sideroblasts and marked thrombocytosis; SF3B1, Splicing Factor 3B
subunit 1; WBC, white blood cells.
Impact of SF3B1 mutations on the prognosis of RARS-T
J Brose
´us et al
1827
&2013 Macmillan Publishers Limited Leukemia (2013) 1826 – 1831
different mutations. With the exception of one p.Arg549Cys in
exon 12 and two in exon 16, all mutations were located in exons
14 and 15. All but one del/ins mutations (p.Met784_Lys785del/
insIle) were missense mutations. In detail, the most frequent
mutation was p.Lys700Glu (68/127 53.5%), followed by p.Lys666-
Glu/Thr/Asp/Asn mutations (n¼18, 14.2%), p.His662Asp/Gln
(n¼13, 10.2%), p.Arg625Cys/Leu (n¼10, 7.9%), p.Glu622Asp
(n¼9, 7.1%) and p.Thr663Pro (n¼4, 3.1%). Five further mutations
were detected in single cases only. Frequencies and positions of
mutations are illustrated in Table 1 and Figure 1. Median
mutation/wildtype load was 40% (range: 15–78%). Small sub-
clones with SF3B1
mut
were not detected.
Frequency of mutations in RARS and RARS-T
The frequency of SF3B1 mutations in RARS-T (96/111, 86.5%) was
similar to that in RARS (28/33, 84.8%). By contrast, both entities
differed by the presence of the JAK2
V617F
mutation, which was
detected in 54/144 (37.5%) in the total cohort but in 54/111
(48.6%) in RARS-T compared with none (0/33) in RARS (Po0.001).
Among the RARS-T SF3B1
mut
, 46/96 (47.9%) harboured a JAK2
V617F
mutation. JAK2
V617F
allele burden was very heterogeneous with a
median of 49% (range: 1–100%). No JAK2
exon12
mutation (111
tested) was observed, whereas only one case with the MPL
W515L
mutation was noted in a RARS-T (102 tested; Table 1 and Figure 2).
Biological association
The presence of SF3B1
mut
was analysed with respect to age, sex,
white blood cell count, haemoglobin levels, platelet counts, blast
counts, percentage of ring sideroblasts, karyotype and JAK2
V617F
allele burden. In RARS-T, SF3B1 mutations were more frequent in
females (56/59, 94.9%) than in males (40/52, 76.9%) (P¼0.010),
and mean ring sideroblast counts were higher in SF3B1
mut
than in
SF3B1
wt
(55% vs 38%) (P¼0.007). No further correlations were
detected for these parameters.
Impact of mutations on outcome
The difference in survival between RARS-T and RARS was not
statistically significant (median survival 10.7 vs 6.2 years,
respectively, P40.05). On the other hand, in the total cohort,
patients with SF3B1
mut
had longer survival than those with
Figure 1. Positions and characterisation of mutations in the HEAT domains 1–6 (exons 12–16) of the SF3B1 gene. Missense mutations are
indicated in green and the rare ins/del mutation in yellow. HEAT, Huntingtin, elongation factor 3, protein phosphatase 2A, Tor1; PPP1R8,
protein phosphatase 1, regulatory subunit 8; aa, amino acid.
Figure 2. Distribution of SF3B1,JAK2
V617F
and MPL
W515L
mutations in RARS-T and RARS.
Impact of SF3B1 mutations on the prognosis of RARS-T
J Brose
´us et al
1828
Leukemia (2013) 1826 – 1831 &2013 Macmillan Publishers Limited
SF3B1
wt
(8.0 vs 1.8 years, respectively, Po0.001; Figure 3a). When
restricted to RARS-T (85 SF3B1
mut
and 12 SF3B1
wt
), median overall
survival was 6.9 years in SF3B1
mut
vs 3.3 years in SF3B1
wt
(P¼0.003; Figure 3b). In RARS, survival was 10.7 years in the
SF3B1
mut
(n¼28) and 1.8 years in SF3B1
wt
patients (n¼5;
P¼0.001; Figure 3c). In RARS-T patients, the survival rates at 2, 4
and 6 years within the JAK2
V617F
sub-cohort were 94.9, 84.3 and
67.4%, respectively, while within the JAK2
wt
sub-cohort, they were
79.7, 69.6 and 32.2%, respectively. JAK2
V617F
(n¼50) was then
associated with a more favourable outcome compared with
JAK2
wt
(n¼47; P¼0.019; Figure 3d).
Cox regression analysis
In the total cohort including RARS-T and RARS cases in univariate
analysis, age (P¼0.020), ring sideroblast count (P¼0.008) and
SF3B1 mutational status (Po0.001) were prognostically significant,
but SF3B1 mutational status was the only independent prognostic
factor (P¼0.001) in multivariable analysis.
In the RARS-T cohort in univariate analysis, age (P¼0.038), ring
sideroblast count (P¼0.014), JAK2
V617F
(P¼0.024) and SF3B1
mut
(P¼0.006) were prognostically significant but only age (P¼0.044),
JAK2
V617F
(P¼0.016) and SF3B1
mut
(P¼0.021) were independent
prognostic parameters in multivariable analysis. Survival was
better in patients with age r80 years, JAK2
V617F
and SF3B1
mutations. Taking into account these three prognostic factors, a
model for survival in RARS-T patients was constructed in which
each risk factor (that is, age 480 years, SF3B1
wt
and JAK2
wt
) was
worth 1 point and defined two groups of risk: high risk for patients
with a score of 2 or 3 (n¼13) and low risk when the score is 0 or 1
(n¼84). Median survival in the high-risk group was 1.6 vs 8.0 years
in the low-risk group (Po0.001; corresponding survival rates at 2,
4 and 6 years are 31.4, 31.4 and 0% for high-risk patients vs 95.8,
84.4 and 62.8% for low-risk patients; Figure 4).
DISCUSSION
Up to now, SF3B1 mutations have been studied in small series of
RARS-T patients. In this study, we provide data on SF3B1 mutations
in a large cohort of RARS-T patients, which is, to the best of our
knowledge, the largest published to date.
SF3B1 mutations were observed with a high frequency in both
RARS-T and RARS patients (86.5% and 84.8%, respectively). These
proportions are slightly higher than those already published, as
SF3B1 mutations have been found in 64–82.6% of RARS
21–24
and
66.7–72% in RARS-T.
23,24
The slightly higher frequency of SF3B1
mutations in the current study may be due to the larger cohort of
RARS-T patients than in other studies. There may also be
differences in methodology, for example, direct sequencing vs
non-sequencing-based screening strategies.
However, RARS differs from RARS-T in that there are no
JAK2
V617F
mutations in RARS,
4,14
whereas there is a high
frequency in RARS-T.
4–13,15,19
We recently showed that RARS-T
differed from RARS and ET from a clinical, biological and
SF3B1mut
(n=113, median 8.0 yrs)
p<0.001
SF3B1wt
(n=17, median 1.8 yrs)
1
2
4
5
11
17
1010
12
14
2028
4556
7588
113
Patients at risk
1
1
2
3
712
668
9
12
15
31
4156
66
85
Patients at risk
SF3B1mut
(n=85, median 6.9 yrs)
SF3B1wt
(n=12, median 3.3 yrs)
p=0.003
1
2
24
5
444
58
1314
15
19
22
28
Patients at risk
SF3B1mut
(n=28, median 10.7 yrs)
SF3B1wt
(n=5, median 1.8 yrs)
p=0.001
334
5
791822
28
3647
3
34
4
5
7
14
21
3137
50
Patients at risk
JAK2wt
(n=47, median 5.4 yrs;
6-yr survival rate 32.2%)
p=0.019
JAK2V617F
(n=50, median 20.4 yrs;
6-yr survival rate 67.4%)
ab
cd
Figure 3. Kaplan–Meier analysis for survival since diagnosis according to SF3B1 and JAK2
V617F
mutation status. SF3B1 in (a) total cohort, (b)
RARS-T, (c) RARS and (d)JAK2
V617F
in RARS-T.
Impact of SF3B1 mutations on the prognosis of RARS-T
J Brose
´us et al
1829
&2013 Macmillan Publishers Limited Leukemia (2013) 1826 – 1831
prognostic point of view, suggesting that RARS-T could be
considered as a unique entity.
20
The results of our current study
showing the presence of both SF3B1 and JAK2
V617F
mutations in a
high proportion of RARS-T confirm that RARS-T is a unique entity.
Indeed, RARS-T was associated with high rates of SF3B1 mutations
(86.5%) and JAK2
V617F
mutations while ET patients have a low
frequency of SF3B1 mutations, and in RARS, no JAK2
V617F
mutations were detected.
A minority of RARS-T patients (13.5%) presented without SF3B1
mutations. This could be due to another mutation affecting
components of the RNA splicing machinery as mutations of SRSF2,
U2AF35 and ZRSR2 have already been described in 12.4, 7.3 and
3.1% of MDS, respectively,
36
or due to mutations of proteins
associated with SF3B1 (SF3B4 and SF3B14).
These results are in line with our hypothesis that RARS-T is an
independent entity characterised by a particular molecular
combination associating mutations that give a myeloproliferative
advantage (JAK2
V617F
,MPL
W515R/L
or other unknown mutations)
and mutations of components of the splicing machinery
responsible for myelodysplastic features (SF3B1 in most cases,
and possibly SRSF2,U2AF35 or ZRSF2 in the remaining cases).
Conflicting results on the prognostic impact of SF3B1 mutations
in MDS have been reported as several studies noted a good
prognostic impact in MDS, whereas others hypothesised that SF3B1
mutations were associated with good-prognosis MDS subgroups
but lost their prognostic impact in RARS and in RCMD-RS.
27
In our large cohort of RARS-T patients, SF3B1 mutations were
associated with female sex, higher ring sideroblast counts and a
longer overall survival than in SF3B1
wt
patients. In a multivariable
analysis, age 480 years at diagnosis, SF3B1
wt
as well as JAK2
wt
were independent factors of a worse prognosis. Based on these
three independent parameters, a prognostic score for RARS-T
patients was created to define two risk groups: high risk when
there were two or three risk factors, low when there was only one
or no risk factor. Median survival was 1.6 vs 8.0 years in the high-
and low-risk group, respectively, underlying the relevance of such
score in RARS-T patients.
Exploring SF3B1 mutations in MDS associated with ring
sideroblasts is of interest from a prognostic point of view,
particularly as specific treatment will be available.
37,38
Also,
allele-specific PCR have been designed, and these could be
useful for monitoring minimal residual disease in MDS.
39
In summary, this study confirms that RARS-T should be
considered an independent entity. In RARS-T patients, age o80
years at diagnosis, SF3B1 and JAK2 mutations are independent
factors for better survival and may be used to stratify patients.
CONFLICT OF INTEREST
SS and TH declare part ownership of the MLL Munich Leukemia Laboratory. TA, SJ
and VG are employed by the MLL Munich Leukemia Laboratory. All the other authors
declare no conflict of interest.
ACKNOWLEDGEMENTS
We thank the Medical Doctors from the Haematology Department and Laboratory of
the University Hospital of Dijon, the Spanish Group of Hematological Cytology
(GECH), as well as Philip Bastable for revising the manuscript. EL and FL are grateful to
the Tumor Bank of the CHU of Bordeaux. This work was supported by grants from the
association ‘Tulipes contre le cancer’ (Cha
ˆlon s/Sao
ˆne, Burgundy, France) and from
FEHH (Spain) and 2009 SGR 541 (Generalitat de Catalunya).
REFERENCES
1 Vardiman JW, Bennett JM, Bain BJ, Baumann I, Thiele J, Orazi A. Myelodysplastic/
myeloproliferative neoplasms, unclassifiable. In: Swerdlow SH, Campo E, Lee
Harris N, Jaffe ES, Pileri SA, Stein H et al. (eds) WHO Classification of Tumours
of Haematopoietic and Lymphoid Tissue. 4th edn. IARC: Lyon, France, 2008,
pp 85–86.
2 Hasserjian RP, Gatterman N, Bennett JM, Brunning RD, Thiele J. Refractory anemia
with ringed sideroblasts. In: Swerdlow SH, Campo E, Lee Harris N, Jaffe ES, Pileri
SA, Stein H et al. (eds) WHO Classification of Tumours of Haematopoietic and
Lymphoid Tissue. 4th edn. IARC: Lyon, France, 2008, pp 96–97.
3 Thiele J, Kvasnicka HM, Orazi A, Tefferi A, Gisslinger H. Essential thrombo-
cythaemia. In: Swerdlow SH, Campo E, Lee Harris N, Jaffe ES, Pileri SA, Stein H et al.
(eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue.4th
edn. IARC: Lyon, France, 2008, pp 48–50.
4 Ceesay MM, Lea NC, Ingram W, Westwood NB, Gaken J, Mohamedali A et al. The
JAK2 V617F mutation is rare in RARS but common in RARS-T. Leukemia 2006; 20:
2060–2061.
5 Boissinot M, Garand R, Hamidou M, Hermouet S. The JAK2-V617F mutation and
essential thrombocythemia features in a subset of patients with refractory anemia
with ring sideroblasts (RARS). Blood 2006; 108: 1781–1782.
6 Flach J, Dicker F, Schnittger S, Kohlmann A, Haferlach T, Haferlach C. Mutations of
JAK2 and TET2, but not CBL are detectable in a high portion of patients with
refractory anemia with ring sideroblasts and thrombocytosis. Haematologica
2010; 95: 518–519.
7 Gattermann N, Billiet J, Kronenwett R, Zipperer E, Germing U, Nollet F et al. High
frequency of the JAK2 V617F mutation in patients with thrombocytosis (platelet
count4600 109/L) and ringed sideroblasts more than 15% considered as MDS/
MPD, unclassifiable. Blood 2007; 109: 1334–1335.
8 Hellstrom-Lindberg E, Cazzola M. The role of JAK2 mutations in RARS and other
MDS. Hematology Am Soc Hematol Educ Program 2008; 52–59.
9 Raya JM, Arenillas L, Domingo A, Bellosillo B, Gutierrez G, Luno E et al. Refractory
anemia with ringed sideroblasts associated with thrombocytosis: comparative
analysis of marked with non-marked thrombocytosis, and relationship with JAK2
V617F mutational status. Int J Hematol 2008; 88: 387–395.
10 Remacha AF, Nomdedeu JF, Puget G, Estivill C, Sarda MP, Canals C et al. Occur-
rence of the JAK2 V617F mutation in the WHO provisional entity: myelodysplastic/
myeloproliferative disease, unclassifiable-refractory anemia with ringed
sideroblasts associated with marked thrombocytosis. Haematologica 2006; 91:
719–720.
11 Renneville A, Quesnel B, Charpentier A, Terriou L, Crinquette A, Lai JL et al. High
occurrence of JAK2 V617 mutation in refractory anemia with ringed sideroblasts
associated with marked thrombocytosis. Leukemia 2006; 20: 2067–2070.
12 Schmitt-Graeff AH, Teo SS, Olschewski M, Schaub F, Haxelmans S, Kirn A et al.
JAK2V617F mutation status identifies subtypes of refractory anemia with ringed
sideroblasts associated with marked thrombocytosis. Haematologica 2008; 93:
34–40.
1112713
High risk
668912153142576684
Lowrisk
Patients at risk
Low risk group
(n=84, median 8.0 yrs;
6-yr survival rate 62.8%)
High risk group
(n=13, median 1.6 yrs;
6-yr survival rate 0%)
p<0.001
Figure 4. Kaplan–Meier analysis for survival according to two risk
groups. High risk comprising patients with at least two of the
following risk factors: age 480 years, SF3B1wt, JAK2wt; and low risk:
one or less of the risk factors.
Impact of SF3B1 mutations on the prognosis of RARS-T
J Brose
´us et al
1830
Leukemia (2013) 1826 – 1831 &2013 Macmillan Publishers Limited
13 Szpurka H, Tiu R, Murugesan G, Aboudola S, Hsi ED, Theil KS et al. Refractory
anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T),
another myeloproliferative condition characterized by JAK2 V617F mutation.
Blood 2006; 108: 2173–2181.
14 Steensma DP, Tefferi A. JAK2 V617F and ringed sideroblasts: not necessarily
RARS-T. Blood 2008; 111: 1748.
15 Wang SA, Hasserjian RP, Loew JM, Sechman EV, Jones D, Hao S et al. Refractory
anemia with ringed sideroblasts associated with marked thrombocytosis harbors
JAK2 mutation and shows overlapping myeloproliferative and myelodysplastic
features. Leukemia 2006; 20: 1641–1644.
16 Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, Wadleigh M et al. MPL515
mutations in myeloproliferative and other myeloid disorders: a study of 1182
patients. Blood 2006; 108: 3472–3476.
17 Schnittger S, Bacher U, Haferlach C, Dengler R, Krober A, Kern W et al. Detection of
an MPLW515 mutation in a case with features of both essential thrombocyt hemia
and refractory anemia with ringed sideroblasts and thrombocytosis. Leukemia
2008; 22: 453–455.
18 Wardrop D, Steensma DP. Is refractory anaemia with ring sideroblasts and
thrombocytosis (RARS-T) a necessary or useful diagnostic category? Br J Haematol
2009; 144: 809–817.
19 Malcovati L, Della Porta MG, Pietra D, Boveri E, Pellagatti A, Galli A et al. Molecular
and clinical features of refractory anemia with ringed sideroblasts associated with
marked thrombocytosis. Blood 2009; 114: 3538–3545.
20 Broseus J, Florensa L, Zipperer E, Schnittger S, Malcovati L, Richebourg S et al.
Clinical features and course of refractory anemia with ring sideroblasts associated
with marked thrombocytosis. Haematologica 2012; 97: 1036–1041.
21 Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y, Yamamoto R et al. Frequent
pathway mutations of splicing machinery in myelodysplasia. Nature 2011; 478:
64–69.
22 Papaemmanuil E, Cazzola M, Boultwood J, Malcovati L, Vyas P, Bowen D et al.
Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med
2011; 365: 1384–1395.
23 Visconte V, Makishima H, Jankowska A, Szpurka H, Traina F, Jerez A et al. SF3B1, a
splicing factor is frequently mutated in refractory anemia with ring sideroblasts.
Leukemia 2012; 26: 542–545.
24 Malcovati L, Papaemmanuil E, Bowen DT, Boultwood J, Della Porta MG, Pascutto C
et al. Clinical significance of SF3B1 mutations in myelodysplastic syndromes
and myelodysplastic/myeloproliferative neoplasms. Blood 2011; 118: 6239–6246.
25 Damm F, Thol F, Kosmider O, Kade S, Loffeld P, Dreyfus F et al. SF3B1 mutations in
myelodysplastic syndromes: clinical associations and prognostic implications.
Leukemia 2012; 26: 1137–1140.
26 Damm F, Kosmider O, Gelsi-Boyer V, Renneville A, Carbuccia N, Hidalgo-Curtis C et al.
Mutations affecting mRNA splicing define distinct clinical phenotypes and
correlate with patient outcome in myelodysplastic syndromes. Blood 2012; 119:
3211–3218.
27 Patnaik MM, Lasho TL, Hodnefield JM, Knudson RA, Ketterling RP, Garcia-Manero
Get al. SF3B1 mutations are prevalent in myelodysplastic syndromes with ring
sideroblasts but do not hold independent prognostic value. Blood 2012; 119:
569–572.
28 Visconte V, Makishima H, Maciejewski JP, Tiu RV. Emerging roles of the
spliceosomal machinery in myelodysplastic syndromes and other hematological
disorders. Leukemia 2012; 26: 2447–2454.
29 Visconte V, Rogers HJ, Singh J, Barnard J, Bupathi M, Traina F et al. SF3B1
haploinsufficiency leads to formation of ring sideroblasts in myelodysplastic
syndromes. Blood 2012; 120: 3173–3186.
30 Jeromin S, Haferlach T, Grossmann V, Alpermann T, Kowarsch A, Haferlach C et al.
High frequencies of SF3B1 and JAK2 mutations in refractory anemia with ring
sideroblasts associated with marked thrombocytosis strengthen the assignment
to the category of myelodysplastic/myeloproliferative neoplasms. Haematologica
2013; 98:1517.
31 Cazzola M, Rossi M, Malcovati L. Biologic and clinical significance of somatic
mutations of SF3B1 in myeloid and lymphoid neoplasms. Blood 2013; 121:
260–269.
32 Lippert E, Boissinot M, Kralovics R, Girodon F, Dobo I, Praloran V et al. The JAK2-
V617F mutation is frequently present at diagnosis in patients with essential
thrombocythemia and polycythemia vera. Blood 2006; 108: 1865–1867.
33 Schnittger S, Bacher U, Kern W, Schroder M, Haferlach T, Schoch C. Report on two
novel nucleotide exchanges in the JAK2 pseudokinase domain: D620E and E627E.
Leukemia 2006; 20: 2195–2197.
34 Schnittger S, Bacher U, Haferlach C, Geer T, Muller P, Mittermuller J et al. Detection
of JAK2 exon 12 mutations in 15 patients with JAK2V617F negative polycythemia
vera. Haematologica 2009; 94: 414–418.
35 Schnittger S, Bacher U, Haferlach C, Beelen D, Bojko P, Burkle D et al.
Characterization of 35 new cases with four different MPLW515 mutations and
essential thrombocytosis or primary myelofibrosis. Haematologica 2009; 94:141144.
36 Thol F, Kade S, Schlarmann C, Loffeld P, Morgan M, Krauter J et al. Frequency and
prognostic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with
myelodysplastic syndromes. Blood 2012; 119: 3578–3584.
37 Rymond B. Targeting the spliceosome. Nat Chem Biol. 2007; 3: 533–535.
38 Webb TR, Joyner AS, Potter PM. The development and application of small
molecule modulators of SF3b as therapeutic agents for cancer. Drug Discov Today
2013; 18:4349.
39 Matsuda K, Ishida F, Ito T, Nakazawa H, Miura S, Taira C et al. Spliceosome-related
gene mutations in myelodysplastic syndrome can be used as stable markers
for monitoring minimal residual disease during follow-up. Leuk Res 2012; 36:
1393–1397.
Impact of SF3B1 mutations on the prognosis of RARS-T
J Brose
´us et al
1831
&2013 Macmillan Publishers Limited Leukemia (2013) 1826 – 1831
... MDS/MPN-RS-T is characterized by ≥15% bone marrow ring sideroblasts (RS) and sustained thrombocytosis [6]. The median age at diagnosis is~75 years [8][9][10][11][12][13]. Approximately 80% of patients have normal cytogenetics, and approximately one-half have SF3B1 and JAK2 mutations [14]. ...
... The median survival of people with MDS/MPN-RS-T is 75 months [8,10,11]. Previous studies reported that age >80 years, abnormal cytogenetics, hemoglobin concentration (Hb) ≤ 100 g/L, and ASXL1 and/or SETBP1 mutations were correlated with worse survival [9][10][11]. There is no widely accepted survival prediction model. ...
... Previous studies of MDS/MPN-RS-T were based on the 2008 or 2016 revision of the WHO classification and were restricted to people with ≥15% bone marrow RS [8][9][10][11][12]20]. We aimed to describe the clinical and molecular landscapes of MDS/MPN-SF3B1-T patients, compare the prognoses of people with < or ≥15% bone marrow RS, and develop prognostic models. ...
Article
Full-text available
We investigated data from 180 consecutive patients with myelodysplastic/myeloproliferative neoplasms with SF3B1 mutation and thrombocytosis (MDS/MPN-SF3B1-T) who were diagnosed according to the 2022 World Health Organization (WHO) classification of myeloid neoplasms to identify covariates associated with survival. At a median follow-up of 48 months (95% confidence interval [CI] 35–61 months), the median survival was 69 months (95% CI 59–79 months). Patients with bone marrow ring sideroblasts (RS) < 15% had shorter median overall survival (OS) than did those with bone marrow RS ≥ 15% (41 months [95% CI 32–50 months] versus 76 months [95% CI 59–93 months]; P < 0.001). According to the univariable analyses of OS, age ≥ 65 years (P < 0.001), hemoglobin concentration (Hb) < 80 g/L (P = 0.090), platelet count (PLT) ≥ 800 × 10E + 9/L (P = 0.087), bone marrow RS < 15% (P < 0.001), the Revised International Prognostic Scoring System (IPSS-R) cytogenetic category intermediate/poor/very poor (P = 0.005), SETBP1 mutation (P = 0.061) and SRSF2 mutation (P < 0.001) were associated with poor survival. Based on variables selected from univariable analyses, two separate survival prediction models, a clinical survival model, and a clinical-molecular survival model, were developed using multivariable analyses with the minimum value of the Akaike information criterion (AIC) to specifically predict outcomes in patients with MDS/MPN-SF3B1-T according to the 2022 WHO classification.
... Median survival of people with MDS/MPN-RS-T is about 75 months 8, 10,11 . Previous studies reported age > 80 years, abnormal cytogenetics, haemoglobin concentration ≤ 100g/L, ASXL1 and/or SETBP1 mutations are correlated with worse survival [9][10][11] . There is no widely-accepted survival prediction model. ...
... Prior studies of MDS/MPN-RS-T were based on 2008 or 2016 revision of WHO classi cation restricted to people with ≥ 15% bone marrow RS [8][9][10][11][12]20 . We set out to describe the clinical and molecular landscapes of MDS/MPN-SF3B1-T, compare prognosis of people with < or ≥ 15% bone marrow RS and to develop a prognostic model. ...
... The SF3B1 mutation is the most common mutation in the disease entity and more than 90% of patients with sequencing information in our study harbored the mutation. The speci c mutation sites of SF3B1 in our study mainly distributed in the 700th, 666th, 625th and 662nd amino acid sites, which was similar to those reported in other studies on RARS-T or MDS/MPN-RS-T [9][10][11] . Previous studies on RARS-T or MDS/MPN-RS-T reported that about 30-75% of patients have both SF3B1 and JAK2 mutations 9-11, 20, 27 . ...
Preprint
Full-text available
We interrogated data from 180 consecutive subjects with myelodysplastic/myeloproliferative neoplasm with SF3B1 mutation and thrombocytosis (MDS/MPN- SF3B1 -T) diagnosed according to the 2022 World Health Organization (WHO) classification of myeloid neoplasms to identify co-variates associated with survival. At a median follow-up of 45 months (Inter-Quartile Range [IQR], 19–73 months), median survival was 70 months (95% Confidence Interval [CI], 57, 84 months). Subjects with bone marrow ring sideroblasts < 15% had briefer median survival compared with those in whom it was ≥ 15% (41 months [32, 50 months] versus 80 months [63, 97 months]; P = 0.003). In multi-variable analyses, age ≥ 65 years ( P = 0.005), haemoglobin concentration < 80 g/L ( P = 0.008), platelet concentration ≥ 800×10E + 9/L ( P = 0.02), bone marrow ring sideroblasts < 15% ( P = 0.007), complex cytogenetics ( P = 0.04) and SETBP1 mutation ( P = 0.004) were independently associated with worse survival. Using these data we divided subjects into 3 risk cohorts: (1) low- ( N = 77; median 96 months [72, 120 months]); (2) intermediate- ( N = 70; median 56 months [32, 80 months]); and (3) high-risk ( N = 7; median 17 months [15, 19 months]; All pairwise comparisons P < 0.0001). Our model, if validated, should help physicians estimate survival in persons with MDS/MPN- SF3B1 -T in the 2022 WHO classification.
... Accepted treatment options, such as cytotoxic chemotherapy and DNMTi, provide small, if any, survival benefits [5][6][7][8]. In general, patients with CMML (median survival 20-40 months [9][10][11][12]), atypical CML (median survival 12-37 months [13][14][15][16][17]), and MDS/MPN-U (median survival of 22-33 months [16,18]) have inferior survivals relative to patients with MDS/MPN-RS-T (median survival 76-128 months [19][20][21]). ...
... Although many prognostic tools have been validated in CMML, only one has been developed for atypical CML [17], and none have been developed for other MDS/MPN subtypes, and the applicability of prognostic tools developed for MDS or CMML has not been fully explored in other MDS/MPN entities. In patients with MDS/MPN-RS-T, younger age and presence of SF3B1 and/or JAK2 mutations have been associated with a more favorable outcome [19]. In patients with aCML, age > 65 years, female sex, WBC > 50 × 10 9 /L, thrombocytopenia, and/or hemoglobin < 10 g/dL have been reported to be adverse prognostic findings [13,14,17]. ...
Article
Full-text available
Background Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) comprise several rare hematologic malignancies with shared concomitant dysplastic and proliferative clinicopathologic features of bone marrow failure and propensity of acute leukemic transformation, and have significant impact on patient quality of life. The only approved disease-modifying therapies for any of the MDS/MPN are DNA methyltransferase inhibitors (DNMTi) for patients with dysplastic CMML, and still, outcomes are generally poor, making this an important area of unmet clinical need. Due to both the rarity and the heterogeneous nature of MDS/MPN, they have been challenging to study in dedicated prospective studies. Thus, refining first-line treatment strategies has been difficult, and optimal salvage treatments following DNMTi failure have also not been rigorously studied. ABNL-MARRO ( A B asket study of N ove l therapy for untreated M DS/MPN a nd R elapsed/ R efractory O verlap Syndromes) is an international cooperation that leverages the expertise of the MDS/MPN International Working Group (IWG) and provides the framework for collaborative studies to advance treatment of MDS/MPN and to explore clinical and pathologic markers of disease severity, prognosis, and treatment response. Methods ABNL MARRO 001 (AM-001) is an open label, randomly allocated phase 1/2 study that will test novel treatment combinations in MDS/MPNs, beginning with the novel targeted agent itacitinib, a selective JAK1 inhibitor, combined with ASTX727, a fixed dose oral combination of the DNMTi decitabine and the cytidine deaminase inhibitor cedazuridine to improve decitabine bioavailability. Discussion Beyond the primary objectives of the study to evaluate the safety and efficacy of novel treatment combinations in MDS/MPN, the study will (i) Establish the ABNL MARRO infrastructure for future prospective studies, (ii) Forge innovative scientific research that will improve our understanding of pathogenetic mechanisms of disease, and (iii) Inform the clinical application of diagnostic criteria, risk stratification and prognostication tools, as well as response assessments in this heterogeneous patient population. Trial registration This trial was registered with ClinicalTrials.gov on August 19, 2019 (Registration No. NCT04061421).
... Cytogenetic anomalies are demonstrated in about 70% of MPNs patients relying on conventional cytogenetic and single nucleotide polymorphism array karyotyping testing (5). The approach to clonal markers starts with JAK2-V617F first (96% sensitivity for PV and 60% for ET or PMF), then CALR (present in 20-25% patients with ET or PMF), followed by MPL (3-7% of patients with ET or PMF) (6). A bone marrow examination holds importance to morphologically distinguish clonal expansion and make accurate diagnosis of myeloid anomalies. ...
Article
Full-text available
Chronic myeloproliferative neoplasms (MPN) include polycythemia vera (PV), primary myelofibrosis, essential thrombocythemia (ET) and chronic myeloid leukemia (CML). Overlapping MPNs are rare; however, they can occur in the same individual. The present case report describes a patient with both triple-negative ET and CML. A 64-year-old woman was followed-up at our hematology clinic at Feist Weiller Cancer Center, Louisiana State University Health Shreveport (Shreveport, LA, USA) since 2000 after she was diagnosed with JAK2V617F-negative ET. The patient remained stable on hydroxyurea until 2012, when they underwent a bone marrow biopsy for progressively increasing white blood cell counts, and the pathology was consistent with CML; PCR for BCR-ABL was positive for both P210 and P190 transcripts. The patient was then initiated on dasatinib. After dasatinib, they were given a trial of imatinib, and were later transitioned to nilotinib and finally to bosutinib (2019) due to unchanged thrombocytosis. Next-generation sequencing from a bone marrow biopsy in 2019 demonstrated an EZH2 mutation that may be associated with triple-negative ET. CML was in major molecular response at that time. The patient was continued on bosutinib with hydroxyurea, after which hydroxyurea was changed to anagrelide due to worsening anemia and persistent thrombocytosis. However, bosutinib and anagrelide were discontinued due to worsening pulmonary hypertension. The patient was noted to have peripheral blasts of 14% by flow cytometry, after which they underwent a repeat bone marrow biopsy in 2022, which showed extensive myelofibrosis. BCR-ABL transcripts were undetectable. Given their accelerated myelofibrosis, the patient was started on a hypomethylating agent, decitabine/cedazuridine, along with darbepoetin for anemia in June 2022. Given their persistent thrombocytosis, the patient was also started on peginterferon α. Most studies reporting two clonal processes in the same patient have been for PV and CML. To the best of our knowledge, this is the first reported case of triple-negative ET with double transcript CML in the same individual.
... It is defined by mutations, cytopenia, morphological dysplasia (with or without ring sideroblasts), bone marrow blasts <5%, peripheral blasts <1%, an indolent clinical course (25,47), and less frequent progression to AML (26,47,60,61). In MDS/MPN, JAK2 and SF3B1 mutations are predictors of better survival (36,62). SF3B1mutated CMML was also found to be a subtype with significant dysplastic features, lower ASXL1 mutation frequency, higher JAK2 V61F mutation frequency, and a more favorable AML-free survival (63). ...
Article
Background/aim: Mutations in the ASXL transcriptional regulator 1 (ASXL1) and splicing factor 3b subunit 1(SF3B1) genes are commonly observed in myeloid neoplasms and are independent predicative factors for overall survival (OS). Only a few contradictory reports exist on the clinical significance of concurrent ASXL1 and SF3B1 mutations. Previous studies also did not exclude patients with mutations of other genes, which could be confounding factors. Materials and methods: We identified 69 patients with mutation of only ASXL1, 89 patients with mutation of only SF3B1, and 17 patients with mutations exclusively of both ASXL1 and SF3B1 from our database of 8,285 patients and compared their clinical features and outcomes. Results: Patients with ASXL1 mutations more frequently had acute myeloid leukemia (22.47%) or clonal cytopenia of unknown significance than patients with SF3B1 mutations (1.45%) or with ASXL1/SF3B1 mutations (11.76%). Patients with SF3B1 or ASXL1/SF3B1 mutations were more frequently diagnosed with myelodysplastic syndrome (75.36% and 64.71%, respectively) than patients with ASXL1 mutations (24.72%). Patients with ASXL1/SF3B1 (23.53%) mutations more frequently had myelodysplastic/myeloid proliferative neoplasm than did patients with ASXL1 mutations (5.62%) or with SF3B1 mutations (15.94%). OS of the ASXL1 mutation-only group was worse than that of the SF3B1 mutation-only group with a hazard ratio of 5.83 (p=0.017). Finally, and most importantly, the OS of the ASXL1/SF3B1 co-mutation group was poorer than that of both single-mutation groups (p=0.005). Conclusion: ASXL1/SF3B1 co-mutations portend worse OS than isolated ASXL1 or SF3B1 mutations, which might be due to abnormalities in both the epigenetic-regulatory and RNA-splicing pathways or because two genes instead of one are mutated.
... Specifically, the acquired heterozygous missense alleles of the SF3B1 (splicing factor 3B, subunit 1) component of the splicing machinery are present in up to 85% of patients with MDS-RS-SLD, MDS-RS-MLD, and MDS/MPN-RS-T. 5,6 On the contrary, non-clonal SA could be secondary to drugs, heavy metal poisoning (Lead, Arsenic), copper deficiency, alcohol use, hypothermia, or chronic neoplastic disease. 7 Herein, we report a challenging case of sideroblastic anemia secondary to pyridoxin deficiency presented as pregnancy-associated severe, recurrent anemia. ...
Article
Full-text available
Sideroblastic anemia is a heterogeneous group of disorders typified by the presence of ring sideroblasts in the bone marrow and has congenital and acquired types. Sideroblastic anemia is a rare event in pregnancy. We report a case of a 32‐year‐old female patient, gravida 4 para 3, 27th weeks pregnant, who presented to the emergency department complaining of palpitation and generalized weakness for 2 weeks. She was found to have severe normochromic normocytic anemia, with hemoglobin of 4.2 g/dl, and low reticulocytes count of 13 × 10³/μl. She gave a history of recurrent anemia, which had only occurred during pregnancy. Her bone marrow aspirate showed many ring sideroblasts concluding the diagnosis of sideroblastic anemia (SA). Further investigation revealed a significantly low pyridoxine level (vitamin B6) of (8 nmol/L). The Hb level improved with vitamin B6 replacement, without any transfusion support.
... The median overall survival of patients with MDS/MPN-RS-T is 76 months, which is shorter than that of patients with ET but longer than in MDS-RS [53]. Given that MDS/MPN-RS-T is associated with mutations in the spliceosome gene SF3B1 in 85% of cases, which in turn are associated with the presence of ring sideroblasts, the ICC approach supports recognition of MDS/MPN with SF3B1 mutation and thrombocytosis (MDS/MPN-SF3B1-T) as a distinct entity [54][55][56]. In the presence of SF3B1 mutation at ≥ 10% VAF, the identification of ring sideroblasts, although common, is no longer required. ...
Article
Full-text available
The myelodysplastic syndrome/myeloproliferative neoplasms (MDS/MPN) category includes a heterogeneous group of diseases characterized by the co-occurrence of clinical and pathologic features of both myelodysplastic and myeloproliferative neoplasms. The recently published International Consensus Classification of myeloid neoplasms revised the entities included in the MDS/MPN category as well as criteria for their diagnosis. In addition to the presence of one or more increased peripheral blood cell counts as evidence of myeloproliferative features, concomitant cytopenia as evidence of ineffective hematopoiesis is now an explicit requirement to diagnose the diseases included in this category. The increasing availability of modern gene sequencing has allowed better understanding of the biologic characteristics of these myeloid neoplasms. The presence of specific mutations in the appropriate clinicopathologic context is now included in the diagnostic criteria for some of MDS/MPN entities. In this review, we highlight what has changed in the diagnostic criteria of MDS/MPN from the WHO 2016 classification while providing practical guidance in diagnosing these diseases.
Article
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Article
Myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) is a rare disease, which presents with features of myelodysplastic syndromes with ring sideroblasts and essential thrombocythemia, as well as anemia and marked thrombocytosis. SF3B1 and JAK2 mutations are often found in patients, and are associated with their specific clinical features. This study was a retrospective analysis of 34 Japanese patients with MDS/MPN-RS-T. Median age at diagnosis was 77 (range, 51-88) years, and patients had anemia (median hemoglobin: 9.0 g/dL) and thrombocytosis (median platelet count: 642 × 109/L). Median overall survival was 70 (95% confidence interval: 68-not applicable) months during the median follow-up period of 26 (range: 0-91) months. A JAK2V617F mutation was detected in 46.2% (n = 12) of analyzed patients (n = 26), while an SF3B1 mutation was detected in 87.5% (n = 7) of analyzed patients (n = 8). Like those with myelodysplastic syndromes or myeloproliferative neoplasms, patients often received erythropoiesis-stimulating agents and aspirin to improve anemia and prevent thrombosis. This study, which was the largest to describe the real-world characteristics of Japanese patients with MDS/MPN-RS-T, showed that the patients had similar characteristics to those in western countries.
Article
Full-text available
Whole exome/genome sequencing has been fundamental in the identification of somatic mutations in the spliceosome machinery in myelodysplastic syndromes (MDSs) and other hematologic disorders. SF3B1, splicing factor 3b subunit 1 is mutated in 60%-80% of refractory anemia with ring sideroblasts (RARS) and RARS associated with thrombocytosis (RARS-T), 2 distinct subtypes of MDS and MDS/myeloproliferative neoplasms (MDSs/MPNs). An idiosyncratic feature of RARS/RARS-T is the presence of abnormal sideroblasts characterized by iron overload in the mitochondria, called RS. Based on the high frequency of mutations of SF3B1 in RARS/RARS-T, we investigated the consequences of SF3B1 alterations. Ultrastructurally, SF3B1 mutants showed altered iron distribution characterized by coarse iron deposits compared with wild-type RARS patients by transmission electron microscopy. SF3B1 knockdown experiments in K562 cells resulted in down-regulation of U2-type intron-splicing by RT-PCR. RNA-sequencing analysis of SF3B1 mutants showed differentially used genes relevant in MDS pathogenesis, such as ASXL1, CBL, EZH, and RUNX families. A SF3B pharmacologic inhibitor, meayamycin, induced the formation of RS in healthy BM cells. Further, BM aspirates of Sf3b1 heterozygous knockout mice showed RS by Prussian blue. In conclusion, we report the first experimental evidence of the association between SF3B1 and RS phenotype. Our data suggest that SF3B1 haploinsufficiency leads to RS formation.
Article
460 Background SF3B1 mutations were recently reported to occur in myelodysplastic syndromes (MDS), especially in the presence of ring sideroblasts (RS) (Papaemmanuil et al. Leukemia Research 2011, Volume 35, Supplement 1, page S18). We sought to accurately define the interaction between SF3B1 mutations, bone marrow morphology, karyotype and prognosis in MDS with ≥15% RS (MDS-RS). Methods Mayo Clinic databases and cell banks were queried to identify patients with MDS and ≥15% RS (MDS-RS). All study patients were required to have undergone bone marrow examination and cytogenetic evaluation at diagnosis. Pathology slides, including iron stains, were centrally re-reviewed to accurately quantify bone marrow RS percentage and confirm World Health Organization (WHO) morphologic categories. Detailed analysis of clinical and cytogenetic parameters was performed to risk stratify patients according to the revised International Prognostic Scoring System (IPSS-R) (Greenberg et al. Leukemia Research 2011, Volume 35, Supplement 1, page S6). All patients were annotated for their mutational status involving JAK2, MPL and IDH. PCR sequencing was used to identify SF3B1 mutations. Results I: Baseline patient information The study is currently ongoing; to date, results are availab1e in 88 patients (median age 72 years, 69% males) including 36 (41%) patients with refractory anemia with RS (RARS), 36 (41%) with refractory cytopenia with multilineage dysplasia and ≥15% RS (RCMD-RS), 11 (12%) with refractory anemia with excess blasts (RAEB)-1 and ≥15% RS (RAEB1-RS) and 5 (6%) with RAEB2-RS. Median (range) values were 9.1 g/dL (5.8–15.7) for hemoglobin, 4.1 × 109/L (1.2–35.2) for leukocytes and 164 × 109/L (6–585) for platelets. IPSS-R risk categories were 20% very good, 49% good, 11% intermediate, 3% poor and 16% very poor. Karyotype was normal in 60%, other very good/good risk 16%, intermediate-risk 2%, and poor/very poor in 22%; 14% had monosomal karyotype (MK). Six (7%), four (5%) and zero (0%) patients displayed IDH, JAK2 or MPL mutations. Red cell transfusion need was documented in 27 (31%) patients. Results II: Prevalence and distribution of SF3B1 mutations and clinical correlates SF3B1 mutations (all heterozygous) were detected in 40 (∼46%) of all 88 study patients: 25 (63%) K700E, 6 (15%) K666N/Q/R, 3 (8%) E622D, 2 (5%) H662D/Q, 2 (5%) Y623C, 1 (3%) R625C, and 1 (3%) T663I. One patient each displayed both SF3B1K700E and IDH2R140Q or SF3B1K700E and JAK2V617F. SF3B1 mutational frequencies were 69% for RARS, 36% for RCMD-RS, 18% for RAEB1-RS and 0% for RAEB2-RS (p=0.0007). SF3B1 mutations clustered with normal (59%) and other very good/good risk (57%) karyotype and were infrequent in poor/very poor karyotype (5%; p=0.0004). Additional significant correlations were noted between SF3B1 mutations and lower IPSS-R risk category (p=0.0004), advanced age (p=0.006), higher platelet count (p=0.0003). Results II: SF3B1 mutations and prognostic relevance in MDS-RS To date, 68 (∼77%) deaths and 10 (∼11%) leukemic transformations have been documented. Median follow-up for living patients was 82 months. Univariate survival analysis considering all 88 study patients identified the following as strongly significant risk factors: IPSS-R (p<0.0001), WHO morphologic categories (p<0.0001), cytogenetic risk categories (p<0.0001), and transfusion need (p=0.0003). In multivariable analysis, only IPSS-R and transfusion need remained significant. In univariate analysis, the presence of SF3B1 mutations was significantly associated with better overall (p=0.04) and leukemia-free (p=0.03) survival; however, in both instances, significance was completely accounted for by WHO morphologic risk categorization and the morphology-adjusted p values were 0.6 and 0.2, respectively. In other words, when RARS and RCMD were analyzed separately, there was no additional prognostic value from the presence or absence of SF3B1 mutations (Figures 1 and 2; p=0.67 and 0.44, respectively). Conclusions SF3B1 mutations are prevalent in RARS and RCMD-RS but do not provide additional, WHO morphology-independent or IPSS-R-independent, prognostic information. Disclosures No relevant conflicts of interest to declare.
Article
Precursor mRNA splicing is catalyzed by the spliceosome, a macromolecule composed of small nuclear RNAs associated with proteins. The SF3B1 gene encodes subunit 1 of the splicing factor 3b, which is important for anchoring the spliceosome to precursor mRNA. In 2011, whole exome sequencing studies showed recurrent somatic mutations of SF3B1 and other genes of the RNA splicing machinery in patients with myelodysplastic syndrome or myelodysplastic/myeloproliferative neoplasm. SF3B1 mutations had a particularly high frequency among conditions characterized by ring sideroblasts, consistent with a causal relationship. SF3B1 mutants were detected at lower frequency also in a variety of other tumor types. In chronic lymphocytic leukemia, SF3B1 was found to be the second most frequently mutated gene. In myelodysplastic syndromes, SF3B1 mutations appear to be founding genetic lesions and are associated with a low risk of leukemic evolution. By contrast, SF3B1 mutations have a lower incidence in early stages of chronic lymphocytic leukemia, are more common in advanced disease, and tend to be associated with poor prognosis, suggesting that they occur during clonal evolution of the disease. In perspective, assessment of SF3B1 mutation status may become part of innovative diagnostic and prognostic tools, while the availability of spliceosome modulators opens novel therapeutic prospects.
Article
Various gene mutations have been reported in patients with myelodysplastic syndrome (MDS). Serial studies of mutations during follow-up are important for investigating the stability of the mutations for use as minimal residual disease (MRD) markers. Sequential quantitative analyses of 5 patients with spliceosome-related gene mutations by allele-specific quantitative polymerase chain reaction revealed that the U2AF1 S34F and SF3B1 K666N were persistently retained during the disease progression. The spliceosome-related gene mutations appear to be stable during disease progression and may be useful as potential markers for MRD monitoring in MDS patients that usually lack established specific MRD markers.
Article
The identification of potent spliceosome modulators that demonstrate antitumor activity indicates that this complex may be a target for drug development. Several natural products have been demonstrated to bind to the SF3b1 subunit of this macromolecule and these agents modulate alternative RNA splicing. In this article we describe their biological properties, discuss the validity of the spliceosome as a therapeutic target, and propose that alteration of alternative splicing represents a viable approach for inducing tumor-selective cytotoxicity.