A user-friendly, frequently updated reference guide that aligns with international guidelines and protocols.
Myelodysplastic Neoplasms (MDS)
Introduction:
It is a group of clonal hematological disorders characterized by moderate to marked dysplasia of one or more of 3 lineages (erythroid / myeloid / megakaryocytic), generally involving at least 10% of the cells of respective lineage in consideration, which results in cytopenias due to ineffective blood cell production.
Defining cytopenia:
Anemia: Hb <13 g/dL in males and <12 g/dL in females
Leukopenia: ANC <1800/cmm
Thrombocytopenia: Platelets <1,50,000/cmm
Diagnostic evaluation must include:
Peripheral smear
Bone marrow
Karyotyping,
Mutation analysis by next-generation sequencing
Multiparameter flow cytometry
Epidemiology:
It is usually seen after age of 50 years.
Median age of diagnosis is 70 years.
Prevalence is 3-5 per 1 lakh in general population.
In age group of more than 70 years prevalence is 20 per 1 lakh.
Etiology: In most of the cases cause is not known
Exposure to radiation.
Use of radiomimetic alkylating agents, topoisomerase inhibitors
Smoking.
Benzene, agricultural chemicals and solvents
Congenital disorders like Down syndrome, Fanconi anaemia, Neurofibromatosis, Ataxia telangiectasia, xeroderma pigmentosa etc.
Abnormalities in enzymes that metabolize carcinogens.
Pathogenesis:
Increased apoptosis
Mutations of proto-oncogenes such as MYC, TNER, tumour growth factor β &IL1 lead to increased apoptosis.
BCL2 expression is also low in MDS.
Increased apoptosis is seen in early phase of MDS especially in refractory anaemia & refractory anaemia with ringed sideroblasts.
Increase in apoptosis is responsible for cytopenias in spite of hypercellular marrow.
Apoptosis may be triggered by variety of factors such as.
Intrinsic DNA damage.
Mitochondrial dysfunction.
Cytotoxic T cells.
Over expression of cytokine TNF- with subsequent upregulation of the FAS/FAS L pathway.
Abnormality of marrow stromal cells.
Erythroid precursors in MDS spontaneously release cytochrome C from mitochondria, leading to activation of caspase 9.
Immunological abnormalities in MDS
These also explain presence of cytopenias in MDS
Common features in aplastic anaemia and hypocellular MDS include.
Clonal expansion of T-cells.
Over representation of HLA-DR 15.
Presence of mutations in PLG-A gene which is characteristic of PNH.
Clinical response to immunosuppressive therapy.
Decreased colony formation by hematopoietic stem cells
Due to decreased production of GM-CSF, IL-3, M-CSF, and IL6.
Epigenetic alterations:
Methylation of DNA (especially of cytosine residues) leads to formation of CpG pair. Increased number of CpG islands proximal to gene promoter region causes decreased gene expression (gene silencing).
Silencing of tumour suppressor genes such as CTNNA1, P15INK4B lead to increased proliferation of cells.
Formation of ringed sideroblasts.
Mitochondrial DNA mutations are seen in 50% of MDS cases.
Impaired function of mitochondrial respiratory chain
↓
Oxidation of ferrous ion to ferric form
↓
Accumulation of ferric ions in mitochondria
↓
Formation of ringed sideroblasts
Change from MDS to AML occurs due to accumulation of additional mutations which block the maturation at blast level.
New (2024) WHO Classification: (Details given at the end)
Disease
Blasts
Cytogenetics
Mutations
MDS with defining genetic abnormality
MDS with low blasts and 5q deletion
<5% BM and <2% PB
5q deletion alone, or with 1 other abnormality other than monosomy 7 or 7q deletion
Any except multi-hit TP53
MDS with low blasts
and SF3B1 mutation
(MDS with low blasts and ringed sideroblasts)
<5% BM and <2% PB
Absence of 5q deletion,
monosomy 7, or complex karyotype
SF3B1
(Detection of ≥15% ring sideroblasts may substitute for SF3B1 mutation)
MDS with biallelic TP53 inactivation
<20% BM and PB
Usually complex
Multihit TP53 alterations (Two or more TP53 mutations, or 1 mutation with evidence
of TP53 copy number loss or copy neutral Loss of Heterozygosity)
MDS, morphologically defined
MDS with low blasts (MDS-LB)
<5% BM and <2% PB
Any
Any except multihit TP53 or SF3B1
MDS, hypoplastic (MDS-h)
<5% BM and <2% PB
Any
Any except multihit TP53 or SF3B1
MDS with increased blasts- 1
5-9% BM or 2-4% PB
Any
Any except multihit TP53 or SF3B1
MDS with increased blasts- 2
10-19% BM or 5-19% PB or Auer rods
Any
Any except multihit TP53 or SF3B1
MDS with fibrosis
5-19% BM; 2-19% PB
Any
Any except multihit TP53 or SF3B1
Old Classification:
Disease
Blood Findings
Bone Marrow Findings
Refractory cytopenia with unilineage dysplasia
Uni/Bicytopenia
No or rare blasts
<5% blasts
<15% ringed sideroblasts
Dysplasia in only one lineage of cells.
3 subtypes:
1. Refractory anemia- Dysplasia in erythroids
2. Refractory neutropenia- Dysplasia in myeloid cells
3. Refractory thrombocytopenia- Dysplasia in megakaryocytes (>10% dysplastic megakaryocytes of at least 30 megakaryocytes evaluated)
Refractory anemia with ringed sideroblasts (RARS)
Anemia
No blasts
>15% ringed sideroblasts
Erythroid dysplasia only
<5% blasts
Refractory cytopenia with multilineage dysplasia (RCMD)
Cytopenias (bicytopenia or pancytopenia)
No or rare blasts
No Auer rods
< 1 x 109/L monocytes
Dysplasia in >10% of the cells of two or more myeloid cells lines
<5% blasts in marrow
No Auer rods
MDS with excess blasts -1
Cytopenias
<5% blasts
No Auer rods
< 1 x 109/L monocytes
Unilineage or multilineage dysplasia
5-9% blasts
No Auer rods
MDS with excess blasts -2
Cytopenias
5-19% blasts
Auer rods +/-
< 1 x 109/L monocytes
Presence of Auer rods in blasts in PB/BM qualifies a case as RAEB-2 irrespective of blast percentage.
Unilineage or multilineage dysplasia
10-19% blasts
Auer rods +/-
Myelodysplastic syndrome unclassified (MDS-U)
Cytopenias
No or rare blasts
No Auer rods
Unilineagedysplasia in<10% of cells in one or more myeloid cell lines
<5% blasts
No Auer Rods
MDS associated with isolated del(5q)
Macrocytic anemia
Usually normal or increased platelet count
<1% blasts
Normal to increased megakaryocytes, which are slightly smaller in size. Nucleus is hypolobated and eccentrically placed.
Dysmorphic erythropoiesis is seen in some cases (Lobulated erythroblast nuclei)
<5% blasts
Isolated del(5q) cytogenetic abnormality
No Auer rods
If SF3B1 mutation is present, then presence of >5% sideroblasts are enough to classify it as MDS with ringed sideroblasts
Note: Presence of MDS related mutations alone (with absence of morphologic dysplasia), even in the presence of clinically significant cytopenias, is not diagnostic of MDS.
Clinical Features:
Asymptomatic
Anaemia
Recurrent infections due to leukopenia. Bacterial pneumonia and skin abscess are most common.
Haemorrhagic manifestation – Occurs due to thrombocytopenia or abnormal platelet function.
Wide range of auto immune disorders seen in 10% cases- Cutaneous vasculitis, relapsing polychondritis, polymyalgia rheumatica, necrotizing panniculitis, Coomb’s positive auto immune haemolytic anaemia, seronegative synovitis and arthritis etc
Hepatomegaly in 5% cases
Splenomegaly in 10% cases
Investigations:
Hemogram:
Normocytic / macrocytic hypochromic anaemia
Anisopoikilocytosis, oval macrocytes
Howell – jolly bodies may be seen which indicate accelerated erythropoiesis.
Basophilic stripling.
Nucleated RBCS seen.
Sideroblasts may be seen.
Reticulocytopenia may be noted.
Neutropenia may be noted
Neutrophils are hypogranular, hyposegmented; contain Dohle bodies, sometimes Auer rods.
Pseudo Pelger Huet anomaly– They are neutrophils with bilobed nuclear configuration
Nuclear fragmentation/ ring shaped nuclei / nuclear sticks may be seen
Monocytosis may be seen in some cases
Rarely circulating myeloblasts may be seen
Thrombocytopenia / thrombocytosis
Platelets are large and lack granules
Circulating micromegakaryocytes may be present. They characteristically contain cytoplasmic blebs and sometimes 1-2 platelets are attached to the surface.
Percentage of blasts must include all nucleated cells except nRBCs.
Reticulocyte count- Inappropriately low
Serum iron, ferritin level – Increased
Serum cobalamin and folic acid levels – Increased
Bone marrow aspiration
Cellularity – Normal / increased / decreased (If hypocellular, dysplasia must be present in myeloid precursors and megakaryocytes for diagnosis of MDS, as erythroid dysplasia is often present in aplastic anaemia)
Cytoplasm – vacuolation, defective haemoglobinisation, and basophilic stripling are noted.
Giant, multinucleated erythroid precursors may be noted
Ringed sideroblasts are present – They are the normoblasts in which mitochondrial iron deposits. By definition, they have 5 or more iron granules, that encircle one third or more of the nucleus in an iron stained (Prussian blue reaction) smear.
Abnormal staining of primary granules in promyelocyte and myelocyte. Sometimes these granules are larger (Pseudo- Chediak- Higashi granules).
Secondary granules may be absent in myelocyte and mature neutrophils.
Irregular cytoplasmic basophilia with a dense rim of peripheral basophilia is also characteristic.
Auer rods are sometimes seen.
Nuclear hypolobulation or hypersegmentation may be noted.
Increased number of marrow monocytes.
In case of del (17p)- Hypolobated neutrophil nuclei are seen
Blast Cells- Maximum number of blast cells compatible with a diagnosis of MDS is 20%. Blast count is an important prognostic indicator in MDS
Blasts have central nucleus with fine nuclear chromatin, prominent nucleoli, high nuclear – cytoplasmic ratio, deeply basophilic and a granular cytoplasm. Auer rods may be seen in some blasts.
Percentage of blasts must be percentage of all nucleated cells cells in BM (including erythroid precursors)
Dysmegakaryopoiesis
Micromegakaryocytes with nuclear hypolobation
Megakaryocytes with small, multiple, separated nuclei
Large mononuclear megakaryocytes
Granules may be absent or may be large and abnormal.
Trephine Biopsy
It is done to detect "Abnormal localization of immature precursors" (ALIP).
Presence of ALIP indicates increased risk of transformation to leukaemia
Normally immature myeloid precursors are seen close to bony trabeculae and around blood vessels, whereas developing erythroid cells and megakaryocytes are seen in inter trabecular spaces
In ALIP, myeloid precursors are displaced from trabecular margins and small clusters of myeloblasts and promyelocytes are seen in inter trabecular spaces
5 cell clusters and 3 clusters in single HPF are significant.
Hypocellularity can be better appreciated in BM biopsy sample. <10% cellularity indicates hypocellular MDS. Always rule out toxic myelopathy and autoimmune disorders in such patients.
Cytochemistry
Peroxidase and Sudan black B- Positive in blasts of myeloid origin
Iron stains- Helps in identification of ringed sideroblasts
PAS- Positive in erythroid precursors
Flow cytometry abnormalities
Low side scatter due to cytoplasmic hypogranularity. Differentiated from myeloblasts by lack of CD34, CD117 and/or HLA-DR. MDS cells are often positive for CD10 and CD16.
Down regulation of antigens that are normally expressed on myeloid cells
Abnormal patterns of cell marker expression
Lineage infidelity in antigen expression.
Increased ratio of CD34+/CD33+
Premature phenotype of myeloid cells- Increased expression of CD33 and CD13 with loss of CD16.
NAT 9- Decreased expression
Large granular lymphocyte population may be identified with flow.
Evaluation of PNH clone and HLA-DR15can be done with flow. If they are positive, patients are responsive to immunosuppressive therapy.
Flow helps to know antibody combinations that characterize the blasts.
Flow percentage of CD34+ cells cannot replace differential count on smears while calculating percentage of blasts.
Cytogenetics
Abnormalities are found in 50% of cases.
Essential investigation in MDS, as it helps to prove clonality and important in prognostication.
Should be performed on at least 25 metaphases.
If there is a dry tap, do FISH on peripheral blood for monosomy 7, del5q and trisomy 8.
Common abnormalities include:
Deletion of 5q/ Monosomy 5
Deletion of 7q / Monosomy 7
Trisomy 8
t (11q23)
del 17p
del 20q
del 18q
Complex karyotyping abnormalities
Molecular studies by next generation sequencing: Mutation of following genes may be seen
Platelet function test – Reduced aggregation with adrenaline and collagen
Platelet associated immunoglobulin – May be raised in the absence of immune mediated thrombocytopenia
Immunoglobulin study
Polyclonal gammopathy in 30%
Hypogammaglobulinemia in 20%
Paraproteins in 5-10%
Copper level- Deficiency may mimic MDS
LDH and Uric acid levels- Increased
Beta 2 microglobulin level- May be increased
MRI of femoral head- Hyperintensity
S. Erythropoietin levels- Helpful in planning the treatment in refractory anaemia
TSH- To rule out hypothyroidism as cause of anemia
HLA typing- If transplant is planned
Genetic screening for Fanconi anaemia, dyskeratosis congenita, RUNX1 and GATA2 in young patients
HbF level-Elevated in 5% cases
Acquired HbH disease in some cases leading to microcytic hypochromic anaemia
Criteria for Diagnosis:
To establish diagnosis of MDS, careful morphologic review and correlation with patient's clinical features are important, as number of medications and viral infections may cause morphologic changes in marrow cells similar to MDS.
Diagnosis is established when
Other causes of dysplasia are excluded
Cytopenia persists for at least 6 months
To call as significant dysplasia is present, ≥10% cells of respective lineage must show features of dysplasia or Blast count is 5-19% or an MDS associated karyotype must be present
MDS-defining abnormalities (by conventional cytogenetics): −7 or del(7q), t(11;16)(q23;p13.3), −5 or del(5q), t(3;21)(q26.2;q22.1), i(17q) or t(17p), t(1;3)(p36.3;q21.1), −13 or del(13q), t(2;11)(p21;q23), del(11q), inv(3)(q21q26.2), del(12p) or t(12p), t(6;9)(p23;q34), del(9q), idic(X)(q13), or complex karyotype (three or more chromosomal abnormalities involving one or more of the above).
Prognosis:
International Prognostic scoring system (IPSS)
Score value
0
0.5
1.0
1.5
2.0
BM blasts (%)
<5
5-10
-
11-20
21-30
Karyotypes*
Good
Intermediate
Poor
Cytopenias
0/1
2/3
*Good: Normal, -Y alone, del (5q) alone, del (20q) alone
Poor: complex (≥3 abnormalities) or chromosome 7 anomalies
Intermediate: other abnormalities.
Cytopenia defined as hemoglobin concentration <10 g/dL, neutrophils<1.5 x 109/L and platelets < 100 x 109/L
Median survival of primary myelodysplastic syndrome using the IPSS score.
Risk Group
IPSS score
Median Survival (in years)
<60 years
> 60 years
<70 years
> 70 years
Low
0
11.8
4.8
9
3.9
Intermediate 1
0.5-1.0
5.2
2.7
4.4
2.4
Intermediate 2
1.5-2.0
1.8
1.1
1.3
1.2
High
>2.5
0.3
0.5
0.4
0.4
Revised IPSS scoring considers anaemia, neutropenia and thrombocytopenia as separate entities. Also, classification of cytogenetic abnormalities is different.
Other scoring systems include Düsseldorf, Varela, Sanz and Bournemouth
Adverse prognostic factors in MDS
Severe cytopenias
Raised LDH or beta 2– microglobulin
Increased blasts
Trilineage dysplasia
Presence of ALIPs
Chromosome abnormalities
Loss of chromosome 5 or 7
Deletion of chromosome 3q, 5q (excluding 5q syndrome), 7q, 17p
Structural abnormality of chromosome 11q23
Complex chromosome abnormalities
P53, RAS mutations
Over-expression of WT1
P15 hypermethylation
Mutations of ASXL1, RUNX1, TP53, EZH-2, NRAS, ETV6 (High risk of leukemic transformation)
Immunophenotype- CD7-Positive blasts
Telomere shortening
In general
Median survival for all MDS is less than 2 years
Leukemic transformation occurs in 10-40% cases
Differential Diagnosis: Other causes for marrow dysplasia
(Slides for assessing marrow dysplasia should be of very high quality. They should be prepared from freshly obtained specimen. Presence of clonality, rules out all below mentioned possibilities and establishes diagnosis of MDS. Hence doing cytogenetics is must in all suspected cases of MDS)
Healthy elderly individuals.
Vitamin B-12 and folic acid deficiency
Heavy metal- Especially arsenic and zinc
Alcohol abuse
HIV and Parvo virus infection
Antituberculous therapy
Treatment with G-CSF
Congenital dyserythropoieticanaemia
Drugs and chemicals- Cotrimoxazole, mycophenolate, chemotherapy agents, valproic acid
Molecular testing for MDS related genes (selected cases)
RUNX1 OR GATA 2 in familial cases
Prognostic score
HLA typing for HSCT candidate
HLA-DR 15 typing (for hypo MDS)
Chemotherapy consent after informing about disease, prognosis, cost of therapy, side effects, hygiene, food and contraception
Fertility preservation
PICC line insertion and Chest X ray after line insertion
Tumor board meeting and decision
Attach supportive care drug sheet
Inform primary care physician
Treatment Plan:
For following category of patients consider ATG with Cyclosporine therapy:
<60 years with hypocellular BM with <5% blasts
PNH clone positivity
STAT-3 mutant cytotoxic T cell clones
International Working Group Modified Response Criteria:
CR Marrow- ≤5% blasts in BM and normal maturation of all cell lines
CR Peripheral blood
Hemoglobin ≥ 11gm/dL
ANC ≥ 1000/cmm
Platelets ≥ 1lac/cmm
No blasts in peripheral smear
Criteria for hematological improvement:
Rise in hemoglobin by 1.5gm/dL
Platelet count >30,000/cmm
ANC >500/cmm
About Each Modality of Treatment:
Hypomethylating agents:
Disease modifying agents which offer survival advantage but not cure
Induce demethylation of hypermethylated genes.
Usually 4-6 cycles are given, then if there is response, then hypomethylating agents are continued till there is loss of response
30% patients show haematological response.
Results in improved quality of life.
They also delay transformation to AML.
No survival advantage over supportive care
Azacytidine is preferred over Decitabine
Patients failing response to one type of hypomethylating agent, may respond to other.
High dose vitamin C can improve response to Azacytidine.
Hypomethylating agents can be used as interim therapy till transplant arrangements are made. They can used to induce remission as well, in patients who are not fit for 7+3 induction
Decitabine
Inj. Decitabine- 20mg/m2- in 300ml NS over 3 hrs- From Day 1 to Day 5
Frequency: 28 days
Dose adjustments:
S. Creatinine- >2mg/dL- Hold until recovery of toxicity
Bilirubin- >2gm/dL- Hold until recovery
ANC <1000/cmm or platelet count <50,000/cmm- Hold until CBC values increase to more than cut off values (this applies only if, baseline ANC was >1500/cmm and baseline platelet count was >75,000/cmm)
Active infection- Hold until recovery
Azacytidine
Inj. Azacytidine- 75mg/m2- SC- OD- Divide doses if volume is >4ml.- From Day 1 to Day 7
Frequency: 28 days
Dose adjustments:
S. Creatinine- >3mg/dL- Hold until recovery to normal and then restart with 50% of dose.
Hepatic impairment- Use with caution
ANC <1000/cmm or platelet count <50,000/cmm- Hold untill CBC values increase to more than cut off values and then give 50% of dose. (this applies only if, baseline ANC was >1500/cmm and baseline platelet count was >75,000/cmm)
Active infection- Hold until recovery
Much lower doses have also been tried which has shown less toxicity and more effectiveness
Decitabine- 20mg/m2 OD for 3 days in 46 days cycle
Azacytidine- 20-45mg/m2-OD 3-5 days, every 4-6 weeks
Lenalidomide:
Dose- 10mg- OD for 21 days in 28 days cycle. (Dose of 5mg- OD is tolerated by many patients)
Response is assessed at 2-4 months
No response is <1.5gm/dL rise in haemoglobin or transfusion dependency at 12-16 weeks of therapy.
Luspatercept: Details are available in drugs section. Click here
Erythropoiesis stimulating agents (ESA)
Erythropoietin- 40,000 units- SC- Once a week, if no response at 8 weeks, increase dose to 60,000 units per week for further period of 8 weeks.
Darbepoetin- 300mcg SC once in 2 weeks, if no response at 8 weeks, increase the dose to 300mcg- SC- Once a week for further period of 8 weeks.
If there is no response (complete/partial) after 16 weeks of therapy then stop erythropoietin. Complete response is achievement of hemoglobin >11gm/dL and transfusion independence. Partial response is rise in hemoglobin by >2gm/dL from baseline (but less than 11gm/dL) and transfusion independence.
Rule out hemolysis and deficiency status prior to starting erythropoietin
Good response in following situation
Low serum EPO levels - < 500 U/L
Non RARS subtype
No / Low transfusion requirements
RARS Subtype respond well when EPO is combined with G-CSF
In those with durable complete response the dose of ESA should be slowly reduced, to a lowest dose that maintains the response.
If response is lost, consider functional iron deficiency.
Avoid rise of haemoglobin to >12gm/dL, as there is high risk of thrombosis (especially if there is previous stroke or history of DM2/ Hypertension)
G-CSF
90% show – Dose dependent neutrophilia and improvement in neutrophil function
Given to patients with severe sepsis
Adjust the dose to maintain TLC between 6000 - 1000/cmm.
Start with 300mcg SC once a week, if there is no response increase the dose up to 300mcg SC 3 times a week.
It does not affect the risk of leukemic transformation
Eltrombopag
It is useful in following conditions
Positive for PNH clone
Age <60 years
Hypoplastic MDS
HLA-DR 15 positive patients
Intensive chemotherapy
It is given to reduce the tumour burden, so that risk of relapse is less after transplant.
Useful if stem cell transplant is planned following cytoreduction.
Donor must be identified prior to starting induction chemotherapy, if time allows.
Usually 7+3 induction chemotherapy is given
Remissions are seen in less than 20% of patients
It should be avoided in patients with poor performance score, as this therapy worsens patient’s condition.
It should be considered in patients less than 50 years of age.
Factors association with higher chances of CR
Early treatment (within 3 months of diagnosis)
Young age (<50 years)
Normal Karyotype
Presence of Auer rods
Allogeneic Stem cell transplantation
It is the only curative treatment
Conditioning is usually done with Busulfan and Cyclophosphamide.
PBSC is better than BM harvest
Overall disease-free survival- 30-40%
Given after induction of CR by hypomethylating agents or induction chemotherapy. If marrow blast count is <5% then, prior chemotherapy is not required.
Good results with
Patients < 40 years old
Short disease duration
<5% BM blasts
Platelet count >1lac/cmm
Poor prognostic factors for outcome
Old age
Poor risk cytogenetics
High IPSS score
Advanced disease (RAEB, RAEBt)
Treatment related MDS
Prolonged disease duration
Marrow fibrosis
Reduced intensity conditioning:
Least day 100 mortality
2-year event free survival- 56%
With RIC, it is possible to transplant patients aged >70 years
Disease control depends on graft vs. MDS effect
For relapse after Allo SCT following can be done:
Salvage with DLI
Second transplant
Chemotherapy
Results with MSD and MUD are same. Hence if familial bone marrow failure syndromes are suspected, MUD is preferred over MSD.
Autologous transplant has problem of contamination of stem cell product with tumour cells. There is no GvL effect as well. Hence there are high chances of relapse. There is 10% peritransplant mortality. With advent of RIC, it is not recommended now.
Immunosuppression with ATG and Cyclosporine
Better response in case of – Hypocellular MDS and HLA – DR15 positive cases
Response rate in unselected MDS- 30 – 50%
ATG must be restricted to fit, relatively young patients.
Dose and method of administration is similar to that in aplastic anaemia.
Ciclosporine is continued for at least 6 months aiming a trough level of 100-200 microgm/L and then slowly tapered
Supportive Care:
Red cell transfusion of correct anaemia. Trigger depends on patient’s activity level. Generally, it is better to maintain haemoglobin level of 7-8gm/dL.
Chelation is given once patient has received 5g iron (25 units of red cells) & those who have a good prognosis according to IPSS score and reasonable life expectancy.
Platelet transfusions for thrombocytopenia
Antibiotics to prevent infections in case of neutropenia
Other Treatment Options:
Low dose cytarabine
May be useful in intermediate 2 and high risk patients
5-20mg/m2/day- SC-BD- for up to 8-16 weeks
Remission is seen in about 20% patients, which remains for about 10 months
Survival is same as supportive care alone
Danazol- Useful in increasing platelet count in some cases
Hydroxyurea and Low dose etoposide- Control leukemic cell population but does not influence survival duration
Maturation enhancing agents
Retinoids
Vitamin D derivatives
Arsenic trioxide
Azacytidine with Lenalidomide
Histone deacetylase inhibitors- Vorinostat and others
Luspatercept, Sotatercept, Roxadustat
Low dose Melphalan- 2mg/day for 50 days. CR observed in 30% patients which remains for approximately 1 year.
Subtypes of Myelodysplastic neoplasms:
MDS with defining genetic abnormalities
MDS with low basts and 5q deletion
It is a myeloid neoplasm with cytopenia and dysplasia characterized by chromosome 5q deletion occurring in isolation or with one other cytogenetic abnormality (other than monosomy 7 or 7q deletion).
Account for 2.5% of all MDS
Median age- 67 years
Common in women
Present with anemia requiring regular transfusions
Peripheral smear:
Macrocytic anemia
Usually normal or increased platelet count
<1% blasts
Bone marrow
Normocellular or hypercellular
Erythropoiesis is suppressed
Dysmorphic erythropoiesis is seen in some cases (Lobulated erythroblast nuclei)
Granulocytic dysplasia is uncommon
Normal to increased megakaryocytes, which are slightly smaller in size. Nucleus is hypolobated and eccentrically placed.
<5% blasts
No Auer rods
Presence of ringed sideroblasts/ SF3B1 mutation does not exclude the diagnosis of MDS with 5q deletion
Prognosis:
Most patients belong to IPSS- low risk
TP53 mutation is associated with decreased response to lenalidomide and increased risk of transformation to AML
MDS with low blasts and SF3B1 mutation
(MDS with low blasts and ringed sideroblasts)
It is a myeloid neoplasm with cytopenia and dysplasia characterized by SF3B1 mutation and ring sideroblasts.
Account for 17% of all MDS cases
Median age- 70- 75 years
Present with chronic anemia
Peripheral smear
Macrocytic or normocytic normochromic RBCs
Rarely neutropenia and thrombocytopenia
Blasts are rarely seen
Granulocytes may show dysplastic features
Bone marrow
Hypercellular
Increased erythropoiesis with dysplastic features
Ringed sideroblasts are seen in iron stain
Dysplastic changes in myeloid cells may be present
Megakaryocytes are normal
Blasts - <5%
Cytogenetics: Usually normal
Criteria for diagnosis: Essential
Cytopenia involving one or more lineages, without thrombocytosis
Erythroid lineage dysplasia;
Blasts <5% in the bone marrow and <2% in the peripheral blood;
Detection of SF3B1 mutation (High- >5% VAF is necessary). If SF3B1 mutation analysis is not available, demonstration of ring sideroblasts comprising ≥15% of erythroid precursors
Not fulfilling diagnostic criteria of AML, MDS with low blasts and 5q deletion, MDS with biallelic TP53 inactivation, MDS with increased blasts, or any MDS/MPN type.
Prognosis: Best among all MDS subtypes. Most belong to IPSS-Low risk.
MDS with biallelic TP53 inactivation
It is a myeloid neoplasm with cytopenia, dysplasia and less than 20% blasts or 30% erythroblasts, characterized by two or more TP53 mutations or one TP53 mutation and concurrent evidence of TP53 copy loss or copy neutral loss of heterozygosity
Account for 7-11% of all MDS
Present with pancytopenia
Bone marrow
Dysplasia noted in all 3 lineages
Blast count is usually high
BM fibrosis if often present
Biallelic alterations can be identified by NGS. TP53 VAF of >40% is considered as significant.
Cytogenetics: Usually complex abnormalities are found
Criteria for diagnosis:
Essential:
Cytopenia involving one or more lineages
Dysplasia involving one or more lineages
Blasts constitute <20% in PB and BM
Detection of one or more TP53 mutations
In the presence of one TP53 mutation, evidence of TP53 copy loss or copy neutral LOH
Desirable:
Complex karyotype (at least 3 abnormalities)
Prognosis: Risk of leukemic transformation and hence death is high.
MDS, morphologically defined
MDS with low blasts (MDS-LB)
(MDS with single lineage dysplasia and MDS with multilineage dysplasia)
It is a myeloid neoplasm with cytopenia and dysplasia but without defining genetic abnormalities, defined by having <5% bone marrow blasts and <2% peripheral blood blasts.
Accounts for 15- 20% of all MDS cases
Most of them present with anemia
PS and BM findings: As described above
Cytogenetics: Usually normal
Criteria for diagnosis:
Essential:
Cytopenia involving one or more lineages.
Dysplastic changes in one or more lineages, involving at least 10% of cells.
<5% bone marrow blasts and <2% peripheral blood blasts
Exclusion of folate and vitamin B12 deficiency.
No fulfilling diagnostic criteria of MDS with defining genetic alterations or hypoplastic MDS.
Desirable:
Hypercellular bone marrow for age.
Detection of clonal cytogenetic and/or molecular abnormality.
Prognosis: Depends on type and degree of cytopenia
MDS, hypoplastic (MDS-h)
It is a myeloid neoplasm with cytopenia and dysplasia, characterized by significantly decreased age-adjusted bone marrow cellularity as determined on a trephine biopsy.
Accounts for 10-15% of all MDS.
Significant overlap with PNH and aplastic anemia is noted.
They have more profound cytopenia, compared to other MDS.
Bone marrow:
Cellularity: <30% in patients younger than 70 years and below 20% in patients aged 70 and older.
Dysplastic features are identified in one or more haematopoietic lineages.
Cytogenetics: Abnormal in 25-40% cases. Commonly trisomy 8 and del (20q).
Criteria for diagnosis:
Essential:
Cytopenia involving one or more lineages
Hypocellular bone marrow (assessed on a trephine core biopsy, adjusted for age of the patient) not explained by drug/toxin exposure or pertinent nutritional deficiency or PNH or inherited BM failure syndromes
<5% blasts in bone marrow and <2% blasts in peripheral blood
Not meeting criteria for MDS with defining genetic abnormalities or MDS with increased blasts.
Desirable:
Detection of clonal cytogenetic and/or molecular abnormality.
Prognosis: Significantly worse compared to aplastic anemia, but better, when compared with other MDS.
Treated usually with immunosuppressive therapy
MDS with increased blasts
It is a myeloid neoplasm with cytopenia and dysplasia but without defining genetic abnormalities, defined by having 5-19% bone marrow blasts and/or 2-19% peripheral blood blasts.
2 subtypes:
MDS with increased blasts- 1: Blasts: 5-9% BM or 2-4% PB
MDS with increased blasts- 2: Blasts: 10-19% BM or 5-19% PB or Auer rods
MDS with increased blasts and fibrosis (MDS-F): 5-19% blasts in the bone marrow and/or 2-19% blasts in the peripheral blood, with significant bone marrow fibrosis (defined as grade 2 or 3).
Accounts for 29-38% of all MDS cases
Most present with pancytopenia
Cytogenetics: Abnormal in 50- 70% cases
Criteria for diagnosis:
Essential:
Cytopenia involving one or more lineages
Dysplastic changes in one or more lineages, involving at least 10% of cells
5-19% bone marrow blasts and 2-20% peripheral blood blasts
Not fulfilling diagnostic criteria of MDS with biallelic TP53 inactivation or AML.
Desirable:
Detection of clonal cytogenetic and/or molecular abnormality.
Childhood Myelodysplastic Neoplasm
(Refractory cytopenia of childhood)
It is a myeloid neoplasm with cytopenia and dysplasia arising in children and adolescents (<18 years of age)
3 subtypes:
Childhood myelodysplastic neoplasm with low blasts: <5% bone marrow blasts and <2% peripheral blood blasts.
Childhood myelodysplastic neoplasm with increased blasts: 5-19% bone marrow blasts and/or 2-19%% peripheral blood blasts
Childhood myelodysplastic neoplasm with low blasts- hypocellular
Incidence-
1-4 cases/Million population/ year
Median age- 6.8 years
Strongly associated with congenital disorders such as
Congenital aplastic anaemia syndromes
Neurofibromatosis- type I
Bloom syndrome
Noonan syndrome
Dubowitz syndrome
Cytogenetic changes
Monosomy 7 is seen in 30% cases
Trisomy 8 and 21
Differential diagnosis
Aplastic anaemia and congenital marrow failure syndromes
Acute myeloid leukaemia
Megaloblastic anaemia
Infections- HIV, Parvovirus, EBV, CMV, HHV6
Toxins- Insecticides, chemotherapy, arsenic
ALPS
Pearson syndrome
Radiation
Criteria for diagnosis: Essential:
Cytopenia involving one or more lineages
Dysplastic changes in one or more lineages, involving at
Blast percentage as described above
At least 1 of following 2 criteria
Detection of clonal cytogenetic and/or molecular abnormality
Exclusion of other causes of cytopenia
Non-neoplastic and some germline mutations in cases with hypocellular bone marrow
Down syndrome, juvenile myelomonocytic leukaemia, and AML with defining genetic abnormalities in cases with hypercellular marrow
Treatment
HSCT is the only curative option
With HSCT- 5year DFSR- 50%
Myeloablative regimens are used
No intensive chemotherapy is needed for inducing remission
Figure 3.1.6- Megakaryocyte in 5q deletion syndrome
Figure 3.1.7- 5q deletion syndrome- Bone marrow biopsy
Recent advances:
Longer-term benefit of luspatercept in transfusion-dependent lower-risk myelodysplastic syndromes with ring sideroblasts
Nearly 50% of patients with lower-risk MDS require RBC transfusions within 2 years of diagnosis. Chronic RBC transfusions are associated with decreased quality of life, increased risk of iron overload, and reduced survival. In the present study, during the entire treatment phase, a significantly greater proportion of patients receiving luspatercept vs placebo achieved ≥75% reduction in RBC transfusion burden over ≥24 weeks. Luspatercept had a generally acceptable and predictable safety profile.
Eltrombopag and azacitidine in patients with high-risk myelodysplastic syndromes
In present study (the ELASTIC study) patients with baseline platelets of <150 × 109/l received eltrombopag ranging from 25 to 300 mg. Marrow response rates after three and six treatment cycles were 32% and 29% respectively. Study concluded that eltrombopag/azacitidine is safe in terms of conventional measures defined by adverse-event reporting.
https://doi.org/10.1111/bjh.18389
Lenalidomide treatment of Japanese patients with myelodysplastic syndromes with 5q deletion
Lenalidomide was approved in Japan for treating myelodysplastic syndromes with a 5q deletion (del 5q-MDS) in 2010. A post-marketing surveillance study followed 173 patients (mean age 72.4 ± 9.0 years) receiving lenalidomide between 2010 and 2011, up to 6 cycles or 6 months. Adverse reactions (ADRs) occurred in 78.0%, with common ADRs being decreased platelet and neutrophil counts, and rash. Among transfusion-dependent patients, 34.2% achieved independence. Acute myeloid leukemia (AML) progression occurred in 11.0% during the study and 17.6% in a 3-year follow-up of 68 patients.
Results of EQOL-MDS trial: Eltrombopag is safe in Low-Risk Myelodysplastic Syndromes
In this multicenter trial, researchers investigated the long-term efficacy and safety of eltrombopag in low-risk MDS patients with severe thrombocytopenia. Eltrombopag showed promise, with a significant increase in platelet response compared to placebo (42.3% vs. 11.1%). Patients on eltrombopag had a 63.6% thrombocytopenia relapse-free survival at 60 months. Clinically significant bleeding was less common in the eltrombopag group. However, eltrombopag patients experienced more grade 3-4 adverse events. AML evolution and disease progression rates were similar between the eltrombopag and placebo groups.
Risk assessment according to IPSS-M is superior to AML ELN risk classification in MDS/AML overlap patients
This study explores the classification and risk assessment of patients with Myelodysplastic Syndromes (MDS) and Acute Myeloid Leukemia (AML) overlap according to the International Consensus Classification (ICC) versus the WHO 5th edition. The findings suggest that MDS-based risk assessment using IPSS-M is applicable to MDS/AML patients, while AML-based ELN 2022 criteria are not suitable for them. MDS/AML patients classified as adverse risk by ELN 2022 had significantly longer survival than adverse risk AML patients. The study advocates for personalized risk assessment and treatment strategies based on genetic subtypes and progression markers rather than arbitrary blast cell thresholds.
MDS and CMML in Japan: results of JALSG clinical observational study-11
This study conducted a multicenter, prospective observational analysis of acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and chronic myelomonocytic leukemia (CMML) in Japan, enrolling 6568 patients from August 2011 to January 2016. The report focuses on MDS (n = 2747) and CMML (n = 182) patients, with 79.5% and 79.7% aged 65 years or older, respectively. The estimated 5-year overall survival (OS) rates were 32.3% for MDS and 15.0% for CMML. Both diseases were more prevalent in men. Azacitidine was the most common treatment for MDS, used in 45.4% of higher-risk and 12.7% of lower-risk patients. The 5-year OS rate after azacitidine treatment was 12.1% for higher-risk and 33.9% for lower-risk MDS patients.
Oral decitabine–cedazuridine versus intravenous decitabine for MDS and CMML.
The phase 3 trial compared oral decitabine plus cedazuridine with intravenous decitabine in patients with myelodysplastic syndromes or chronic myelomonocytic leukaemia. Results showed equivalent pharmacokinetic exposure between oral and intravenous formulations, with similar safety profiles. The most common adverse events were thrombocytopenia, neutropenia, and anaemia, with a higher incidence of serious adverse events observed with oral decitabine–cedazuridine. However, both formulations demonstrated efficacy, supporting the use of oral decitabine–cedazuridine as a safe and effective alternative to intravenous decitabine in these patients.
Sabatolimab plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes
Sabatolimab is an immunotherapy targeting T-cell immunoglobulin domain and mucin domain-3 expressed on immune cells and leukaemic stem cells. The STIMULUS-MDS1 trial investigated the efficacy and safety of sabatolimab plus a hypomethylating agent compared to placebo plus a hypomethylating agent in previously untreated patients with higher-risk myelodysplastic syndromes (MDS). Despite the addition of sabatolimab, there was no significant improvement in complete response rates or progression-free survival observed.
Prognostic impact of SF3B1 mutation and multilineage dysplasia in myelodysplastic syndromes with ring sideroblasts
This study of 170 Mayo Clinic patients found that MDS-RS-MLD had significantly worse overall survival than MDS-RS-SLD (P<0.01), but the presence of SF3B1 mutation did not impact survival. Multivariable analysis identified multilineage dysplasia, age, transfusion need, and abnormal karyotype as key prognostic factors, while SF3B1 mutations were not prognostically significant. This suggests that an MLD-based classification may be more relevant for predicting outcomes than SF3B1 mutation status in MDS-RS.
Luspatercept versus epoetin alfa in erythropoiesis-stimulating agent-naive, transfusion-dependent, lower-risk myelodysplastic syndromes
The COMMANDS phase 3 trial compared luspatercept and epoetin alfa in ESA-naive patients with transfusion-dependent, lower-risk myelodysplastic syndromes. Luspatercept achieved a significantly higher rate of red blood cell transfusion independence and hemoglobin improvement (60% vs 35%) compared to epoetin alfa. Both treatments had manageable safety profiles, with luspatercept showing a higher incidence of hypertension and anemia.
Disclaimer: Information provided on this website is only for medical education purposes and not intended as medical advice. Although authors have made every effort to provide up-to-date information, the recommendations should not be considered standard of care. Responsibility for patient care resides with the doctors on the basis of their professional license, experience, and knowledge of the individual patient. For full prescribing information, including indications, contraindications, warnings, precautions, and adverse effects, please refer to the approved product label. Neither the authors nor publisher shall be liable or responsible for any loss or adverse effects allegedly arising from any information or suggestion on this website. This website is written for use of healthcare professionals only; hence person other than healthcare workers is advised to refrain from reading the content of this website.