5,233
Views
22
CrossRef citations to date
0
Altmetric
Original Article

Cytogenetic abnormalities in myelodysplastic syndrome: an overview

&
Pages 131-138 | Published online: 12 Nov 2013

Abstract

The myelodysplastic syndromes (MDS) are clonal disorders of haematopoietic stem cells characterized by ineffective haematopoiesis leading to blood cytopenias and by high incidence of progression to acute myeloid leukaemia (AML). These disorders generally arise de novo, but may also occur years after exposure to mutagenic chemotherapy. Clonal cytogenetic abnormalities are detected in about 30–50% de novo cases, whereas more than 80% of therapy-related forms harbour such markers. Although in the Western countries, MDS cases are mainly reported in the elderly population and rarely in the paediatric age group; this disease is increasingly seen in young adults in India. Cytogenetic study plays an important role in the diagnosis and is useful for prediction of individual prognosis using the international prognostic scoring system. Specific chromosomal abnormalities, such as −5/5q-, −7/7q-, and complex abnormalities, play an important role in the development of new therapeutic options and clinical management of MDS. In this review, we summarize the cytogenetic abnormalities in MDS from various parts of the world.

Introduction

The myelodysplastic syndromes (MDS) are clonal disorders of haematopoietic stem cells characterized by ineffective haematopoiesis leading to blood cytopenias and by high incidence of progression to acute myeloid leukaemia (AML).Citation1 These disorders generally develop de novo, but may also occur years after exposure to mutagenic chemotherapy.Citation2 Primary and secondary MDS are recognized taking into account the previous history of the patients. In MDS patients, variability is seen in cytogenetics, which may influence the prognostic classification and understanding of the molecular background of the abnormalities seen in MDS. In MDS, cellular dysplasia of blood and marrow cells is linked to functional defects in both neutrophils and platelets, which contribute to infection and bleeding manifestations, even in the absence of substantial reduction in blood cell production. The variable risk of progression to AML which is characteristic of MDS appears to be driven by somatic genomic instability and epigenetic genomic modification. Allelic imbalances arising from structural or numerical chromosomal aberrations, defects in DNA repair,Citation3,Citation4 and linkage to genotoxic exposures including alkylating agents, topoisomerase 2 inhibitors, environmental poisons, or radiation.Citation5,Citation6 In this review, the frequency and type of chromosomal abnormalities found in different populations are considered, which may be helpful in the understanding the aetiology, current stratification, and management of MDS.

Classification of MDS

MDS has been recognized for the past 60 years and there have been attempts at classification. The French–American–British (FAB) classification which was revised and expanded in 1982 is based on the proportion of immature blast cells in the blood and bone marrow, and also on the presence or absence of ringed sideroblast or peripheral monocytosis.Citation7 World Health Organization (WHO) created a new classification system based on FAB, but included new morphological features and cytogenetic abnormalities.Citation8,Citation9 Based on peripheral blood and bone marrow findings, the FAB cooperative groupCitation7 distinguishes fine morphological subtypes of MDS, which include refractory anaemia (RA), refractory anaemia with ringed sideroblasts (RARS), refractory anaemia with excessive blast in transformation (RAEB-t), and chronic myelomonocytic leukaemia (CMMoL). Controversies related to FAB classification were, for example, the heterogeneity of the refractory anaemias, the inclusion and definition of CMMoL, the definition of prognosis of patients with del(5q), and the observation that some groups of MDS patients with medullary blast counts above 20% generally have a prognosis similar to one with AML.Citation5,Citation10

The WHO classification has been more widely accepted by pathologists and clinicians, and was combined with the international prognostic scoring system,Citation11 which utilizes the percentage medullary blasts, the number of cytopenic lines and cytogenetic abnormalities as prognostic factors, signifying an advance in the diagnosis and treatment of patients with MDS ()Citation8.

Table 1. WHO classification for the myelodysplastic syndromes

The main changes of WHO classification refined the definitions of low-grade subtypes (RA and RARS) as being strictly erythrocytic, and added a new category, refractory cytopenia with multilineage dysplasia. The lower limit for the percentage blasts for the diagnosis of AML, in peripheral blood or bone marrow, was reduced from 30 to 20%, which resulted in the elimination of subtype RAEB-t from the WHO classification. Other changes included in WHO classification included RAEB-I with 5–9% medullary blasts, RAEB-II with 10–19% and MDS without classification, and the specific genetic subtype, 5q- syndrome.Citation8 The FAB guidelines for RA and RARS are somewhat ambiguous and result in different interpretations by different research groups. Bennet et al.Citation7 state that, in RA and RARS, morphological abnormalities in the granulocytic and megakaryocytic series identical to those present in the other subtypes of MDS may occasionally be found in varying degrees. They also noted that the erythroid series are mainly affected and the granulocytic and megakaryocytic series almost always appear normal.

Incidence

The precise incidence of de novo MDS is not known; however, there are estimates that it exceeds the incidence for acute myeloid leukaemia in the elderly, the risk increasing with the age. In one study, the annual incidence per 100,000 was estimated at 0·5 for people less than 50 years of age compared to 5·3 for the ages 50–59 years, 15 for ages 60–69 years, 49 for ages 70–79 years, and 89 for ages older than 80 years.Citation12 Aul et al.Citation13 reported a crude annual incidence of 4·1/100 000 for MDS. The mean age in most series is approximately 65 years, and in the majority of the cases, there is a male predominance.Citation14 Onset of the disease before the age of 50 years is unusual with the specific exception of mutagen-induced MDS.Citation15

Cytogenetic Features in MDS

The chromosomal aberration frequency varies with the geographic and ethnic changes (). Chromosomal aberrations are present in half of all de novo MDS patients. There are various cytogenetic abnormalities observed in MDS that are also seen in AML, thus supporting a common origin for a proportion of these two diseases.Citation16 Some karyotypic abnormalities are more frequently associated with MDS than AML, mainly del(5q) and del(20q).Citation16 A loss of genetic material in the form of deletions and monosomies is frequently observed in MDS and gain of genetic material in the form of total or partial trisomies is not frequently observed. In consideration into loss or gain of genetic material, it is assumed that main molecular mechanism in MDS is the inactivation of tumour suppressor genes in the form of loss as compared to the activation of oncogenes. Difference between the primary and secondary MDS is complexity of abnormal karyotypes as single chromosomal abnormalities typical for primary MDS, while complex chromosomal abnormalities are seen in secondary MDS.Citation17

Table 2. The frequency of chromosomal abnormalities reported in MDS

Primary MDS and cytogenetics

Most frequent chromosomal abnormalities in MDS

Monosomy 5/del(5q)

Monosomy 5 or deletion of long arm of chromosome 5(5q-) was the most frequent chromosomal abnormality found across the world and have been reported in various populations, including Tunisian 13%, Brazilian 12·5%, USA 5·9%, India 42%, and Germany 30% groups. The chromosome 5q region contains many genes that are involved in the regulation of haematopoiesis, including cytokines and their receptors, cell cycle regulators, transcription factors, and signalling mediators. The haematological genes between 5q13 and 5q33 suggest a significant correlation between the genetic abnormality and the clinical features of 5q- syndrome and other del(5q) MDS subtypes.Citation18Citation20 More than 50% of patients with MDS present with cytogenetic aberrations at diagnosis. Partial or complete deletion of the long arm of chromosome 5 is the most frequent abnormality. Isolated del(5q) is generally associated with refractory anaemia without excess of blasts, macrocytic anaemia, thrombocytosis, and typical mononucleated megakaryocytes, features characteristics of the 5q- syndrome.Citation21 In 5q- syndrome, there are characteristic clinical features, in addition to the above, including mild leukocytopenia, and elevated platelets with less than 5% blasts in bone marrow smears with or without the presence of erythroid and granulocytic dysplasia. Sole 5q- deletion in MDS is of favourable prognosis. In MDS with 5q-, the deletion is always interstitial but is of variable extent. A commonly deleted segment involving 5q31 and 5q32 bands was detected using cytogenetic analysis.Citation22 Boultwood et al.Citation21,Citation23 has described a small commonly deleted segment at 5q31–32 of only 1·5 Mb.

On the basis of haploinsufficiency, it is shown that 41 candidate genes in the critical region of chromosome 5q have been reported and gene for ribosomal subunit protein (RPS41) seems to be a relevant gene in 5q- syndrome.Citation24 In 5q31 region, there are two distinct deleted regions, which is located more centromeric, possibly associated with high risk, complex abnormalities, poor prognosis as well as secondary MDS. Another area which is more telomeric near 5q32 is supposed to be associated with good prognostic 5q- syndrome.Citation25Citation27 Supportive care with red blood cell transfusion is the main option in majority of patients with non complex del(5q-).Citation28Citation30 The use of recombinant erythropoietin and granulocyte colony-stimulating factor has yielded poor results in 5q- syndrome compared to non-5q- MDS.Citation31,Citation32 Modern therapeutic options, such as the immunomodulating agent lenalidomide, give a good response in 5q- syndrome patients.Citation33 Chromosome 5 abnormalities were significantly associated with exposure to inorganic gases and fumes which include exhaust gases, ammonia fumes, hydrogen peroxide, and mineral acids.Citation34

Monosomy 7/del(7q)

Along with −5/5q- abnormality, chromosome 7 abnormalities are also frequently observed in MDS patients.Citation35 The frequency of chromosome 7 abnormalities in various countries such as Tunisia, Brazil, and India were 8, 14, and 32·2%, respectively. Monosomy 7 is characterized by a severe refractory cytopenias, and susceptibility to major infections. Allelotyping studies have identified three distinct loci, 7q22, 7q31·1, and 7q31·3, which are frequently deleted, and a region between 7q22 and 7q36 has been identified as a region harbouring more than one tumour suppressor genes which are inactivated in AML and MDS.Citation36Citation38 No significant prognostic relevance has been observed in monosomy 7/7q-. The German–Austrian study groupCitation39 reported 36% where monosomy 7 was the sole abnormality, 14% with one additional abnormality and 50% as part of complex chromosomal abnormalities. It can be assumed that there must be a cluster of genes with tumour suppressor features present in 7q region associated with MDS.Citation40 Molecular background of the monosomy 7 in MDS is associated with cytogenetic subgroup with RAS mutation, AML1 mutations, and hypermethylation of P15INK4B.Citation41,Citation42

There is no favoured therapeutic option available in the monosomy 7 subgroup. Allogeneic stem cell transplantation can be done if age and clinical conditions are adequate,Citation40 but in monosomy 7 there is a high risk of early relapse. Chromosome 7 abnormalities were significantly associated with exposure to organics, inorganic dusts, and inorganic gases.Citation35 According to Raj et al.,Citation43 MDS and AML patients treated with 5- azacytidine shows a good response in monosomy 7 with continuous complete remission for 10-month follow-up.

Other chromosomal abnormalities in primary MDS

Other chromosome abnormalities have also been frequently observed in MDS after −5/5q- and −7/7q-. The variant chromosome rearrangements such as trisomy 4, trisomy 8, trisomy 9, trisomy 11, trisomy 13, trisomy 14, trisomy 17, trisomy 18, trisomy 19, trisomy 21, −8, −10, −16, −22, −X, −Y, monosomy 19, monosomy 21, del(1)(q32), del(2)(q33-ter), del(8q), del(9)(q31), del(12)(p12p13), del(13q), del(17)(q23), del(18p), del(20q), +1q, +3q, +mar, 11q23 rearrangements, t(6;14)(q23;q32), inv(1)(p22q24), i(1q), i(14)(q11), i(17q), dup(1)(q21q32), t(1;1)(q41;q43), t(1;8)(q21;p12), derivative(1;7)(q10;p10), t(1;11)(q11;q25), t(1;11)(q11;p15), t(1;12)(q10;q10), t(1;19)(q23;p13), t(3;9)(p21;p23), t(5;13)(q21;q31), t(5;16)(q14;p13), t(5;19)(q22;q13), t(1;7)(q10;p10), t(1;17)(q10;q10), t(7;17)(p15;q11), t(X;6)(p10;p10), t(X;11), t(9;12)(q12;q24·3), t(1;2)(q13;q12), t(11;14)(q13;q32), dic(1;16)(q21;p13·3) t(1;22),t(9;12;22)(q34;q15;q11), t(10;14)(q31;q21), der(4)t(1;4)(q21;q35), t(7;13), t(1;X)(p21;q31), t(2;11)(p21;q23), and t(3;3)(q21;q26) were observed to date. These aberrations are reported to be associated with poor prognosis.

In a Tunisian population, chromosome abnormalities in MDS patients were found in 51% and the most frequent karyotypic abnormalities found was del(12p) in 4%, del(20q) and trisomy 8 in each 3%, i(17q) in 21%, and −Y in 0·4% cases. In this population, patients are classified according to prognostic significance as good risk cytogenetics, whose median survival was 30 months, if they possessed specific single chromosomal abnormalities, such as del(5q), del(20q), −Y, and normal karyotype. Poor risk patients, whose median survival was 10 months, possessed −7 and complex karyotypes. Intermediate prognosis is observed with various abnormalities, such as trisomy 8, trisomy 21, del(12)(p12p13), i(17q), t(2;11)(p21;q23), t(3;3)(q21;q26), del(12)(12p13), i(14)(q11), trisomy 19, monosomy 19 +mar, del(5q) del(13q), t(6;14)(q23;q32), +13, and +21.Citation44 In a Brazilian population,Citation45 the frequency of chromosomal alterations was 69% of the patients with primary MDS, and the most frequent chromosomal abnormalities were 11q23 rearrangements in 12·5%, del(12p) in 12·5%, trisomy 8 in 9·4%, and del(20q) in 7·8%. Among the abnormalities detected in adults, the most frequent were rearrangements of 11q23 (16·23%) and del(12p) (14·3%). In paediatric patients, del(5q) (20%) and monosomy 7 (20%) were the most frequent alterations detected. In these studies, some structural aberrations involved rare breakpoints such as 1q24, 1q32, 1q41, 1q43, 9q31, and 17q23 in inv(1)(p22q24), del(1)(q32), t(1;1)(q41;q43), del(9)(q31), and del(17)(q23), respectively. One more study in de Souza Fernandez et al.Citation46 reported lower (32%) chromosomal aberration frequency compared to other studies. The most frequent chromosomal abnormalities found in this series were +8, dup(1)(q21q32), del (20q), t(1;8)(q21;p12), and −8. A large group (968) with primary MDS were studied in a Spanish population and 45% chromosome aberrations were detected. The most common abnormalities found were trisomy 8, rearrangement of 11q, del(12p), i(17q), del(20q), and loss of Y. Complex karyotypes were observed in 107 patients, 23·6% among those patients with abnormal karyotypes. Single chromosomal abnormalities were found to be gain of 1q, involvement of 3q, trisomy 8, trisomy 9, del, or t(11q), trisomy 11, del(12p), rearrangement of 13q, i(17q), trisomy 17, trisomy 18, trisomy 19, del(20q), trisomy 21, monosomy 21, and loss of X and Y chromosomes.Citation47 Pozdnyakova et al.Citation48 conducted a study in the USA on a series of 1029 patients with primary MDS which showed various cytogenetic abnormalities with a frequency of chromosomal aberrations found to be 45% (458 patients) of the single chromosomal abnormalities, including del(20q), −Y, +8, 3q rearrangement, +9, del(11q), +11, del(12p), 13q rearrangements, isochromosome 17q[i(17q)], +17, +18, +19, +21 and −21, −7, del(7q), and derivative(1;7)(q10;p10). The most frequent single chromosomal aberrations in there series were +8 (3·7%) and del(20q) (2·7%). A complex karyotype (mostly chromosome 5 and chromosome 7 or both) was observed in 39·5% among the patients with abnormal karyotype.Citation48

Recently, a multicentre study was carried out in Chinese population and high frequency (67·5%) of chromosomal aberrations was detected. Ninety-nine had copy number changes alone (41·7%), 70 had structural abnormalities alone (29·5%), and 68 displayed both of these changes (28·8%). Chromosomal abnormalities frequently detected in these series were +8,−20/20q-/del(20)(q11–13), −18, −11/11q-/del(11)q23–25), +21, −Y, −21, −10, −16, −22, +9, and del(12)(p12). Besides these aberrations, they also found some rare chromosomal translocations such as t(1;11)(q11;q25), t(1;11)(q11;p15), t(1;12)(q10;q10), t(1;19)(q23;p13), t(3;9)(p21;p23), t(5;13)(q21;q31), t(5;16)(q14;p13), t(5;19)(q22;q13), t(1;7)(q10;p10), t(1;17)(q10;q10), t(7;17)(p15;q11), and t(X;6)(p10;p10).Citation49 Panani et al.Citation50 carried out a study on Greek population on 239 primary MDS patients and 63 (26·36%) cases were found with an abnormal karyotype. Among the cytogenetically abnormal cases, 46 patients presented single chromosomal abnormalities (73·01%). These aberrations were according to frequency +8 (28·57%), +14 (6·35% each), i(17q) and del(11)(q13) (4·76% each), +11, −Y, i(1q), del(8q), del(18p), and t(X;11) (1·59% each). The incidence of chromosomal abnormalities in Greek MDS patients was lower (26·36%) than that reported in the literature.Citation50

In Cardiff, UK clonal karyotype abnormalities in 124 patients with MDS were reviewed which found that 45(36·3%) showed chromosomal abnormalities. Trisomy 8 represented the second most frequent abnormality in all subtypes but most commonly in RAEB; chromosome 7 abnormalities like −7, +der(1)t(1;7), del(20q), and trisomy 21 were also observed. They carried out sequential studies in 77 patients of whom 12 showed karyotypic evolution. Leukaemic transformation most commonly occurred in patients with an abnormal karyotype, particularly those with multiple abnormalities, and in patients with evolving karyotype.Citation51 In China,Citation52 chromosomal analysis was successfully carried out in 424 patients, of which 164 (38·7%) were found to have clonal chromosomal abnormalities. The greatest frequency of abnormal karyotypes was found in RAEB subtypes. The most common chromosomal abnormality was trisomy 8 occurring either singly or in combination with other abnormalities. Various other chromosomal abnormalities are also observed, such as −5/5q-, −7/7q-, 20q-, 9q-, 11q, 12p-, 17p, −Y, translocations, and complex karyotypes.Citation52 In Germany, Haase et al.Citation53 carried out sequential study on 2124 myelodysplastic syndrome patients which reveals clonal chromosomal abnormalities in 1084 (52·3%) patients. Other frequent chromosomal abnormalities including −7/7q-, +8, −18/18q-, 20q-, −5, −Y, −17/17p- [including isochromosome (17q)], +mar, +21, inv/t(3q), −13/13q-, +1/+1q, −21, +11, 12p-, t(5q), 11q-, and t(7q) were present in various proportions in German populations.Citation53

The position in India in MDS appears quite different from the rest of the world as revealed by a number of studies. One studyCitation54 showed chromosomal abnormalities in 64·5% of patients. The most frequent chromosomal abnormalities were del(5q/5-) (42%), −7/7q- (32·2%), +8, and 20q- (19·3%) each and i(17)(q10), −Y, trisomy 4, and trisomy 14 (3·2%) each. A further study conducted from India reported 54·48% of chromosomal abnormalities including novel chromosomal aberrations,Citation55 frequency of chromosomal abnormalities was increased with the advancing age (>30 years) and chromosomal abnormalities are significantly associated with pesticide exposure. Chromosomal deletions (19%) are more frequent as compared to other chromosomal aberrations (12%). Novel chromosomal abnormalities such as t(9;12)(q12;q24·3), t(1;2)(q13;q12), and t(11;14)(q13;q32) in RA, dic(1;16)(q21;p13·3), t(1;22), t(9;12;22)(q34;q15;q11), and t(10;14)(q31;q21) in RAEB-t. der(4)t(1;4)(q21;q35) +mar, t(7;13), t(8;21)(q22;q22), t(1;X)(p21;q31), and −7 in RA with RAEB-t, del(2)(q33-ter) in CMMoL), and trisomy 14 in RA with RARS have been reported.Citation55

Secondary MDS

Therapy-related myelodysplasia and myeloid leukaemia (t-MDS/t-AML) is a distinctive clinical syndrome occurring after exposure to chemotherapy or radiotherapy. Most patients with t-MDS/t-AML have clonal chromosomal abnormalities in their bone marrow cells at diagnosis. In Chicago, clonal chromosomal abnormalities observed in 282 (92%) patients having one or two detectable abnormal clones and 24 (8%) patients were karyotypically normal. The most common abnormalities involved loss of a whole chromosome 5, 7, or both (−5 and −7) or a deletion of the long arm of these chromosomes [del(5q) or del(7q)] in 214 (70%) patients. Monosomy 7 (−7) was present in 102 (33%) patients and monosomy 5 (−5) in 36 (12%) patients. The most common structural chromosomal abnormality was a del(5q) in 59 (19%) patients.Citation56 Increasing copy number of chromosome 8/8q may be a marker of advanced clonal evolution and toxic-exposure myelodysplasia, and both these features were associated with a poor prognosis. In one such therapy, related MDS due to cyclophosphomide therapy for fibrosing alveolitis showed complex karyotype: 45–47,XX,der(5)t(5;12)(q14;q13)[21],del(5)(q13)[3],i(8)(q10)[20],+i(8)(q10)[20], inv(9)(p13q13)[20],−12[20],+r[2][cp21] with a normal clone of 46,XX[3].Citation57

Impact of fluorescence in situ hybridization (FISH) in detection of chromosomal aberrations in MDS

FISH may add value to karyotype non-informative results and occasionally reveals cryptic aberrations not recognized by conventional cytogenetics. According to Yang et al.,Citation58 FISH testing is informative only in those cases with karyotype failure and intermediate–high grade MDS with normal karyotype and has limited utility in cases that have normal G-band karyotypes and morphological features of low-grade MDS or in cases with abnormal G-band karyotypes. G-banded karyotyping has limited resolution power in the range of few megabases of DNA. FISH has a much higher resolution power and can detect chromosomal aberrations in the order of few hundred kilobases of DNA.Citation59 Hence, FISH is more sensitive in detecting chromosomal aberrations in clones with low mitotic index, in detecting cryptic or submicroscopic chromosomal aberrations, and in detecting minor clones.

Cytogenetics and prognostic significance

Karyotype plays an important role in the prognosis of patients with MDS which is helpful for the development of therapeutic strategies based on individual risk assessment. According to World Health Organization classification-based prognostic scoring system, cytogenetics is of immense importance ().Citation60

Table 3. Chromosome aberrations and prognosis in MDS

Good prognosis

According to Knapp et al.,Citation61 cytogenetic findings with a normal karyotype is favourable and complex abnormalities are linked to a poor prognosis, in contrast to the AML patients where a normal karyotype is associated with intermediate prognosis. Isolated del(20q) as the sole abnormality had improved outcome.Citation10,Citation62 A recent study reported most MDS patients had complex abnormalities with del(20q) as part of abnormal karyotype, and associated with poor prognosis.Citation63 Altogether del(20q) as sole abnormality is associated with favourable outcome, but less favourable when present with a part of complex chromosomal abnormalities. This phenomenon is similar with the del(5q). According to a few studies, loss of Y- chromosome is also of good prognosis, but this abnormality has been noted in bone marrow samples from haematologically normal elderly men.Citation64

The Spanish group identified new cytogenetic abnormalities with a good prognosis (12p- and 11q-)Citation47 according to German–Austrian study; the following abnormalities linked with good prognosis are t(1q), 5q-, t(7q), 9p-, 12q-, t(15q), t(17q), 20q-, +21, −21, −X, and −Y. Prognosis was favourable only when there are not more than one additional abnormality present.Citation53

Intermediate prognosis

Patients with trisomy 8 represent an intermediate clinical course. Data presented by the Spanish group on more than 900 patients give new abnormalities which are associated with an intermediate clinical course, such as 3q abnormalities, trisomy 9, 11q translocations, and 17p deletions.Citation47 Recently, a German–Austrian study group revealed several infrequent abnormalities with an intermediate prognosis. In trisomy 8 and 11q-, a median survival time of 23–26 months was observed which is termed as intermediate 1, and with 11q23 translocations, chromosome 3q aberrations, trisomy 19, 7q deletions, and complex abnormalities with three different chromosomal alterations and monosomy 7 are associated with worse prognosis with a median survival time between 20 and 14 months, which is termed as intermediate 2.Citation65

Poor prognosis

Complex chromosomal abnormalities in MDS are associated with a bad prognosis and median survival time significantly below 1 year.Citation66 Median survival in MDS is significantly reduced only when more than three cytogenetic abnormalities are present.Citation53 Most of the publications feature two to three abnormalities as ‘complex’, but in the Medical Research Council AML trials, poor prognosis was associated to cases with five or more abnormalities.Citation67 The monosomy 5 and chromosome 7 abnormalities have been associated with poor prognosis in MDS and AML.Citation68,Citation69 The cytogenetic findings of Toyama et al. who observed unfavourable outcome in trisomy 8 and del(20q) were not confirmed by other group study.

Conclusions

Cytogenetic study plays an important role in diagnosis and prognosis of MDS patients. Although common chromosome aberrations −5/5q- and −7/7q- are reported worldwide, MDS shows considerable heterogeneity in karyotypic pattern. The complex chromosome abnormalities are seen in significant number of MDS and associated with a poor prognosis. FISH investigation has a major impact on identification of common chromosome aberrations; however, conventional cytogenetics has its own position in identification of complex karyotypic abnormalities. The advancement in cytogenetic technology using multicolour FISH or spectral karyotyping essentially helps in identification of unidentified cytogenetic abnormalities or complex translocations. The identification of complex gene rearrangements may help in the development of new therapeutic options for the management of the disease.

References

  • Greenberg PL. The smoldering myeloid leukemic states. Clinical and biologic features. Blood 1983;61:1035–44.
  • Levine EG, Bloomfield CD. Leukemias and myelodysplastic syndromes secondary to drug, radiation and environmental exposure. Semin Oncol 1992;19:47–84.
  • Ohyashiki JH, Ohyashiki K, Aizawa S, Kawakubo K, Shimamoto T, Iwama H, et al.. Replication errors in hematological neoplasias: genomic instability in progression of disease is different among different types of leukemia. Clin Cancer Res 1996;2:1583–9.
  • Sieber OM, Heinimann K, Tomlinson IP. Genomic instability—the engine of tumorigenesis? Nat Rev Cancer 2003;3:701–8.
  • Steensma DP, Tefferi A. The myelodysplastic syndrome(s): a perspective and review highlighting current controversies. Leuk Res 2003;27:95–120.
  • Heaney ML, Golde DW. Myelodysplasia. N Engl J Med 1999;340:1649–60.
  • Bennet JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR, et al.. Proposals for the classification of the myelodysplastic syndromes.Br J Haematol 1982;51:189–99.
  • Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002;100:2292–302.
  • Harris NL, Jaffe ES, Diebold J, Flandrin G, Muller-Hermelink HK, Vardiman J, et al.. World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting—Airlie House, Virginia, November 1997. J Clin Oncol 1999;17:3835–49.
  • Germing U, Gattermann N, Strupp C, Aivado M, Aul C. Validation of the WHO proposals for a new classification of primary myelodysplastic syndromes: a retrospective analysis of 1600 patients. Leuk Res 2000;24:983–92.
  • Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, et al.. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997;89:2079–88
  • Williamson PJ, Kruger AR, Reynolds PJ, Hamblin TJ, Oscier DG. Establishing the incidence of myelodysplastic syndrome. Br J Haematol 1994;87:743–5.
  • Aul C, Gatterman N, Schneider W. Age-related incidence and other Epidemiological aspects of myelodysplastic syndromes. Br J Haematol 1992;82:358–67.
  • Anonymous French registry of acute leukemia and myelodysplastic syndromes. Age distribution and hemogram analysis of the 4496 cases recorded during 1982–1983 and classified according to FAB criteria. Groupe Francais de Morphologie Hematologique. Cancer 1987;60:1385–94.
  • Michels SD, Mckenna RW, Arthur DC, Brunning RD. Therapy related acute myeloid leukemia and myelodysplastic syndrome: a clinical and morphologic study of 65 cases. Blood 1985;65:1364–72.
  • Mauritzson N, Albin M, Rylander L, Billström R, Ahlgren T, Mikoczy Z, et al.. Pooled analysis of clinical and cytogenetic features in treatment related and de novo adult acute myeloid leukemia and myelodysplastic syndromes based on a consecutive series of 761 patients analyzed 1976–1993 and on 5098 unselected cases reported in the literature 1974–2001. Leukemia 2002;16:2366–78.
  • Third MIC Cooperative Study Group. Recommendations for a morphologic, immunologic, and cytogenetic (MIC) working classification of the primary myelodysplastic syndromes and therapy-related myelodysplastic disorders. Report of the workshop held in Scottsdale, Arizona, USA, on February 23–25, 1987. Cancer Genet Cytogenet 1988;32:1.
  • Boultwood J, Fidler C, Strickson AJ, Watkins F, Kostrzewa M, Jaju RJ, et al.. Transcription mapping of the 5q- syndrome critical region: cloning of two novel genes and sequencing, expression, and mapping of a further six novel cDNAs. Genomics 2000;66:26–34.
  • van den Berghe H, Cassiman JJ, David G, Fryns JP, Michaux JL, Sokal G. Distinct hematological disorder with the deletion of long arm of no 5. chromosome. Nature 1974;251:437–8.
  • van den Berghe H, Michaux L. 5q-, twenty-five years later: a synopsis. Cancer Genet Cytogenet 1997;94:1–7.
  • Boultwood J, Fidler C, Lewis S, Kelly S, Sheridan H, Littlewood TJ, et al.. Molecular mapping of uncharacteristically small 5q deletions in two patients with the 5q- syndrome: delineation of the critical region on 5q and identification of a 5q- breakpoint. Genomics 1994;19:425–32.
  • Pedersen B, Jensen IM. Clinical and prognostic implications of chromosome 5q deletions: 96 high resolution studied patients. Leukemia 1991;5:566–73.
  • Boultwood J, Fidler C, Strickson AJ, Watkins F, Gama S, Kearney L, et al.. Narrowing and genomic annotation of the commonly deleted region of the 5q- syndrome. Blood 2002,99:4638–41.
  • Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, et al.. Identification of RPS14 as the 5q- syndrome gene by RNA interference screen. Nature 2008;451:335–40.
  • Nagarajan L. Molecular anatomy of the 5q- chromosome. Leuk Lymphoma 1995;17:361–6.
  • Olney HJ, LeBeau MM. The myelodysplastic syndromes. Pathobiology and clinical management. In: , Bennett J M, ed, editor. The cytogenetics and molecular biology of myelodysplastic syndromes. New York: Marcel Dekker; 2002. p. 89–119.
  • Pellagatti A, Cazzola M, Giagounidis AA, Malcovati L, Della Porta MG, Killick S, et al.. Gene expression profiles of CD34+ cells in myelodysplastic syndromes: involvement of interferon-stimulated genes and correlation to FAB subtype and karyotype. Blood 2006;108:337–45.
  • Mathew P, Tefferi A, Dewald GW, Goldberg SL, Su J, Hoagland HC, et al.. The 5q- syndrome: a single-institutional study of 43 consecutive patients. Blood 1993;81:1040–5.
  • Bowen D, Culligan D, Jowitt S, Kelsey S, Mufti G, Oscier D, et al.. Guidelines for the diagnosis and therapy of adult myelodysplastic syndrome. Br J Haematol 2003;120:187–200.
  • Giagounidis AA, Germing U, Haase S, Hildebrandt B, Schegelberger B, Schoch C, et al.. Clinical: morphological, cytogenetic, and prognostic features of patients with myelodysplastic syndromes and del(5q) including band q31. Leukemia 2004;18:113–9.
  • Hellstrom-Lindberg E. Efficacy of erythropoietin in the myelodysplastic syndromes: a meta-analysis of 205 patients from 17 studies. Br J Haematol 1995;89:67–71.
  • Hellstrom-Lindberg E, Ahlgren T, Beguin Y, Carlsson M, Carneskog J, Dahl M, et al.. Treatment of anemia in myelodysplastic syndromes with granulocyte colony-stimulating factor plus erythropoietin: results from a randomized phase II study and long-term follow-up of 71 patients. Blood 1998;92:68–75.
  • List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, et al.. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 2005;352:549–57.
  • West RR, Stafford DA, White AD, Bowen DT, Padua RA. Cytogenetic abnormalities in the myelodysplastic syndromes and occupational or environmental exposure. Blood 2000;95:2093–7.
  • Abdelrazik HN, Farawila HM, Sherif MA, AlAnsary M. Molecular characterization of chromosome 7 in AML and MDS patients. Afr J Health Sci 2006;13:33–42.
  • Chen Z, Pasquini M, Hong B, Heikens M, Tsai S. The human Penumbra gene is mapped to a region on chromosome 7 frequently deleted in myeloid malignancies. Cancer Genet Cytogenet 2005;162:95–98.
  • Liang H, Fairman J, Claxton DF, Nowell PC, Green ED, Nagarajan L. Molecular anatomy of chromosome 7q deletions in myeloid neoplasms: evidence for multiple critical loci. Proc Natl Acad Sci USA 1998;95:3781–5.
  • Le Beau MM, Espinosa R, Davis EM, Eisenbart JD, Larson RA, Green ED. Cytogenetic and molecular delineation of a region of chromosome 7 commonly deleted in malignant myeloid diseases. Blood 1996;88:1930–5.
  • Haase D, Germing U, Schanz J, Pfeilstocker M, Nosslinger T, Hildebrandt B, et al.. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood 2007;110:4385–95.
  • Detlef haase: Cytogenetic features in myelodysplastic syndromes. Ann Hematol 2008;87:515–26.
  • Christiansen DH, Andersen MK, Pedersen-Bjergaard J. Methylation of p15INK4B is common, is associated with deletion of genes on chromosome arm 7q and predicts a poor prognosis in therapy-related myelodysplasia and acute myeloid leukemia. Leukemia 2003;17:1813–9.
  • Christiansen DH, Andersen MK, Pedersen-Bjergaard J. Mutations of AML1 are common in therapy-related myelodysplasia following therapy with alkylating agents and are significantly associated with deletion or loss of chromosome arm 7q and with subsequent leukemic transformation. Blood 2004;104:1474–81.
  • Raj K, John A, Ho A, Chronis C, Khan S, Samuel J, et al.. CDKN2B methylation status and isolated chromosome 7 abnormalities predict responses to treatment with 5-azacytidine. Leukemia 2007;21:1937–44.
  • Gmidène A, Sennana H, Fenaux P, Laatiri A, Zarrouk M, Bouaziz H, et al.. Cytogenetic abnormalities in Tunisian de novo myelodysplastic syndrome: a comparison with other populations. Leuk Res 2008;32:1824–9.
  • Borgonovo T, Ribeiro EMSF, Cornelio DA, Schmid-Braz AT, Jamur VR, Wuicik L, et al.. Cytogenetic study of Brazilian patients with myelodysplastic syndrome (MDS). Genet Mol Biol 2005;28:654–60.
  • de Souza Fernandez T, Silva MLM, de Souza JM, Tabak D, Abdelhay E. Cytogenetic study of 50 Brazilian patients with primary myelodysplastic syndrome. Braz J Genet 1997;20:87–91.
  • Solé F, Luno E, Sanzo C, Espinet B, Sanz GF, Cervera J, et al.. Identification of novel cytogenetic markers with prognostic significance in a series of 968 patients with primary myelodysplastic syndromes. Haematologica 2005;90:1168–1178.
  • Pozdnyakova O, Miron PM, Tang G, Walter O, Raza A, Woda B, et al.. Cytogenetic abnormalities in a series of 1029 patients with primary myelodysplastic syndromes. Cancer 2008;113:3331–40.
  • Li L, Liu XP, Nie L, Yu MH, Zhang Y, Qin TJ, et al.. Unique cytogenetic features of primary myelodysplastic syndromes in Chinese patients. Leuk Res 2009;33:1194–8.
  • Panani A. Cytogenetic findings in adult Greek myelodysplastic syndrome patients: predominance of single trisomy 8. In Vivo 2006;20:115–8.
  • Geddes AD, Bowen DT, Jacobs A. Clonal karyotype abnormalities and clinical progress in the myelodysplastic syndrome. Br J Haematol 1990;76:194–202.
  • Wang XQ, Ryder J, Gross SA, Lin G, Irons RD. Prospective analysis of clinical and cytogenetic features of 435 cases of MDS diagnosed using the WHO (2001) classification: a prognostic scoring system for predicting survival in RCMD. Int J Hematol 2009;890:361–9.
  • Haase D, Germing U, Schanz J, Pfeilstocker M, Nosslinger T, Hildebrandt B, et al.. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood 2007;110:4385–95.
  • Dakshinamurthy AG, Novitzky N, Bharadwaj R, Prakhya BM. Cytogenetic analysis of 52 Indian patients with de novo myelodysplastic syndromes — a comparative analysis of results with reports from Asia. Ann Hematol 2005;84:298–303.
  • Vundinti BR, Kerketta L, Jijina F, Ghosh K. Cytogenetic study of myelodysplasic syndrome from India. Indian J Med Res 2009;130:155–9.
  • Smith SM, Le Beau MM, Huo D, Karrison T, Sobecks RM, Anastasi J, et al.. Clinical–cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: the University of Chicago series. Blood 2003;102:43–52:.
  • Au WY, Ma SK, Wan TS, Man C, Kwong YL. Pentasomy 8q in therapy-related myelodysplastic syndrome due to cyclophosphamide therapy for fibrosing alveolitis. Cancer Genet Cytogenet 2003;141:79–82.
  • Yang W, Stotler B, Sevilla DW, Emmons FN, Murty VV, Alobeid B, et al.. FISH analysis in addition to G-band karyotyping: utility in evaluation of myelodysplastic syndromes? Leuk Res 2010;34:420–5.
  • Cuneo A, Bigoni R, Cavazzini F, Bardi A, Roberti MG, Agostini P, et al.. Incidence and significance of cryptic chromosome aberrations detected by fluorescence in situ hybridization in acute myeloid leukemia with normal karyotype. Leukemia 2002;16:1745–51.
  • Malcovati L, Germing U, Kuendgen A, Della Porta MG, Pascutto C, Invernizzi T, et al.. Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. J Clin Oncol 2007;25:3503–3510.
  • Knapp RH, Dewald GW, Pierre RV. Cytogenetic studies in 174 consecutive patients with preleukemic or myelodysplastic syndromes. Mayo Clin Proc 1985;60:507–16.
  • Wattel E, Lai JL, Hebbar M, Preudhomme C. De novo myelodysplastic syndrome with deletion of the long arm of chromosome 20: a subtype of MDS with distinct hematological and prognostic features? Leuk Res 1993;17:921–6.
  • Campbell LJ, Garson OM. The prognostic significance of deletion of the long Arm of chromosome 20 in myeloid disorders. Leukemia 1994;8:67.
  • United Kingdom Cytogenetics Group (UKCCG). Loss of the Y chromosome from normal and neoplastic bone marrows. Genes Chromosomes Cancer 1992;5:83.
  • Haase D, Germing U, Schanz J, Pfeilstöcker M, Nösslinger T, Hildebrandt B, et al.. New and comprehensive cytogenetic prognostication and categorization in MDS. Blood 2006;108:252.
  • Morel P, Hebbar M, Lai JL, Duhamel A. Cytogenetic analysis has strong Prognostic value in de novo myelodysplastic syndromes and can be incorporated in a new scoring system: a report on 408 cases. Leukemia 1993;7:1315.
  • Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G, et al.. The importance of diagnostic cytogenetics on outcome AML: analysis of 1,612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children’s Leukaemia Working Parties. Blood 1998;92:2322–33.
  • Toyama K, Ohyashiki K, Yoshida Y, Abe T. Clinical implications of chromosomal abnormalities in 401 patients with MDS: a multicentric study in Japan. Leukemia 1993;7:499.
  • Wattel E, Laï JL, Hebbar M, Preudhomme C, Grahek D, Morel P, et al.. De novo myelodysplastic syndrome (MDS) with deletion of the long arm of chromosome 20: a subtype of MDS with distinct hematological and prognostic features? Leuk Res 1993;17:921.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.