Article

Review Article

Ann Lab Med 2025; 45(2): 133-145

Published online January 8, 2025 https://doi.org/10.3343/alm.2024.0477

Copyright © Korean Society for Laboratory Medicine.

Abnormalities in Chromosomes 5 and 7 in Myelodysplastic Syndrome and Acute Myeloid Leukemia

Tulene S. Kendrick , B.Sc., M.B.B.S., Ph.D.1,2,3, Daria Buic , B.Sc., M.C.P.2, Kathy A. Fuller , B.Sc., Ph.D.2, and Wendy N. Erber, M.D., Ph.D.2,3

1Haematology Department, Royal Perth Hospital, Perth, Australia; 2School of Biomedical Sciences, The University of Western Australia, Crawley, Australia; 3PathWest Laboratory Medicine WA, Perth, Australia

Correspondence to: Wendy N. Erber, M.D., Ph.D.
School of Biomedical Sciences (M504), The University of Western Australia, Crawley, WA 6009, Australia
E-mail: wendy.erber@uwa.edu.au

Received: September 5, 2024; Revised: October 17, 2024; Accepted: December 23, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Chromosomes 5 and 7 are large chromosomes that contain close to 1,000 genes each. Deletions of the long arms or loss of the entire chromosome (monosomy) are common defects in myeloid disorders, particularly MDS and AML. Loss of material from either chromosome 5 or 7 results in haploinsufficiency of multiple genes, with some implicated in leukemogenesis. Abnormalities of one or both occur in up to 15% of MDS and AML cases and co-segregate in half of these. Generally, these chromosomal abnormalities are harbingers of adverse risk in both myeloid disorders. A notable exception is del(5q) in 5q– syndrome, a subtype of MDS. In this review, we describe the pathogenesis and genetic consequences of deletions in chromosomes 5 and 7. Furthermore, we provide an overview of current testing methodologies used in the assessment of these chromosomal defects in hematological malignancies and describe the disease associations and prognostic implications of aberrations in chromosomes 5 and 7 in both MDS and AML.

Keywords: Acute myeloid leukemia, Chromosome 5, Chromosome 7, Monosomy, Myelodysplastic syndrome

Chromosome 5 and 7 abnormalities are observed in myeloid malignancies, including MDS and both de-novo and secondary AML. Chromosome 5, one of the largest human chromosomes, spans 181 million base pairs, representing ~6% of total cellular DNA. It includes numerous intra-chromosomal duplications and 923 protein-coding genes. Despite its size, chromosome 5 has among the lowest gene densities due to gene-poor regions that are evolutionarily conserved in vertebrates, implying functional importance [1]. Chromosome 7 includes 160 million base pairs, representing over 5% of the genome, and contains 1,455 genes [2]. Aberrations within these chromosomes may involve loss of chromosomal material leading to unbalanced deletions, such as interstitial deletion of the short (p) or long (q) arms (described as deletion 5q or del(5q) when involving chromosome 5q). Complete loss of a chromosome (monosomy 5 or 7) or loss of genetic material due to structural aberrations, such as unbalanced translocations with a known partner chromosome, additional material of unknown origin, and isochromosome formation, are also observed [3].

MDS and AML are heterogeneous diseases characterized by somatic gene mutations and chromosomal abnormalities, including balanced genetic abnormalities, such as reciprocal translocations, and unbalanced chromosomal abnormalities, leading to material gain or loss through deletion or aneuploidy. Identifying these genetic abnormalities is crucial for diagnostic sub-classification, predicting therapeutic responses, and guiding treatment choices [46]. Outcomes depend on patient-specific factors, leukemia genetics, and treatment intensity. In this review, we explore the implications of chromosome 5 and 7 alterations within the context of the myeloid malignancies in which they appear and describe the testing methods available to detect these chromosomal changes.

Since the identification of the association between chromosome 5 deletions and myeloid diseases in 1974 [7], extensive research has been conducted to identify commonly deleted regions (CDRs) to characterize the genes involved in leukemogenesis and identify potential therapeutic targets in MDS and AML. Deletions in 5q result in the loss of large chromosome segments, representing ~2% of the haploid genome [8]. Given the substantial size of the deleted regions, identifying key genes with leukemogenic potential is challenging. Despite intensive efforts, no somatic mutations of single candidate tumor suppressor genes have been identified [9]. No single genomic aberration comprehensively explains clone inception or progression to AML. Genes on 5q exhibiting haploinsufficiency in AML are involved in signal transduction, DNA repair, and cell cycle control [10]. Similarly, in myeloid malignancies with –7/del(7q), genes critical to DNA damage response, DNA replication, and chromatin regulation are deleted [11].

Chromosome 5 deletions in myeloid malignancies

Two distinct regions of 5q are commonly deleted in MDS and AML. The distal CDR (CDR-1), located at 5q33, is associated with 5q– syndrome, an MDS entity [12]. These cases have an isolated interstitial deletion of the long arm of chromosome 5, including bands q31-q33, and exhibit a low blast count (<5%) [5]. Single-nucleotide polymorphism array (SNP-A) karyotyping in patients with 5q– syndrome revealed that the CDR-1 boundaries lie between the genes encoding SH3 domain and tetratricopeptide repeats 2 proximally and glycine receptor alpha 1 distally, covering ~2.9 Mb, with deletions between 5q32 and 5q33.2 [13]. The proximal and terminal regions of the long arm are consistently retained, with the commonly retained region (CRR) ending at 5q14.2 proximally and beginning at 5q34 distally [14]. The proximal CDR (CDR-2) centers on bands 5q31.2 and 5q31.3, spanning ~1.92 Mb [14]. This CDR is associated with aggressive MDS and AML and a poor prognosis (Fig. 1). These 5q deletions frequently co-occur with additional cytogenetic abnormalities, most commonly del(7q) and/or a complex karyotype (defined by the presence of three or more chromosomal aberrations) [7, 9,].

Figure 1. CDRs and CCRs of chromosome 5q associated with MDS and AML. The distal CDR (CDR-1) is located at 5q33 and is associated with the deletion of RPS14 and 5q– syndrome. The proximal CDR (CDR-2) at 5q31 encompasses genes, including EGR1, CSNK1A1, and G3BP1, and is associated with aggressive MDS and AML. CRRs are shown in green. This figure was created with BioRender.com.
Abbreviations: MDS, myelodysplastic syndrome; CDR, commonly deleted region; CRR, commonly retained region; RPS14, ribosomal protein S14; EGR1, early growth response 1; CSNK1A1, casein kinase 1 alpha 1; G3BP1, G3BP stress granule assembly factor 1; NPM1, nucleophosmin 1; DDX41, DEAD-box helicase 41.

Larger 5q deletions (median size, 71.4 Mb) also occur and often extend across the proximal and/or telomeric extremes of the q arm, encompassing both CDR-1 and CDR-2. These occur in high-risk MDS and AML, with nearly all affected patients also exhibiting mutations in the CRRs [14]. Other chromosomal defects, including 7q deletions (20% of cases), may co-occur. Unbalanced rearrangements involving 5q are more prevalent in AML than in MDS [15]. The larger 5q deletions (>70 Mb) are associated with tumor protein p53 gene (TP53) deletions [16] because of the deletion or uniparental disomy of 17p [14]. Copy-neutral loss of heterozygosity (CN-LOH) of 17p occurs in ~20% of del(5q) AML cases, all spanning TP53, with most also exhibiting TP53 mutations on the remaining allele [14]. Complex karyotypes with 5q and 7q abnormalities but without del(17p) frequently show CN-LOH of 17p, which may remain undetected unless examined using SNP-A [17]. LOH at 17p only appears in patients whose 5q deletion affects the centromeric and telomeric regions, particularly at band 5q13. Deletion size and CRR involvement influence survival in MDS, with smaller lesions exhibiting lower transformation rates than those affecting the extremes of 5q. Loss of 5q13.3 (within the centromeric CRR) is hypothesized to precede TP53 mutations, conferring a proliferative advantage to the dysplastic clone. Consequently, larger deletions correlate with additional cytogenetic abnormalities and TP53 mutation, contributing to more severe disease in MDS and AML. These findings suggest that the loss of additional genes in the proximal or centromeric extremes of 5q may drive increased genomic instability, favoring AML transformation [14]. Graubert, et al. [18] utilized total exonic resequencing and array comparative genomic hybridization to evaluate all 28 genes in the 5q31.2 region in 46 patients with MDS. None of the del(5q) samples harbored mutations in the remaining wild-type chromosome, and no cytogenetically silent microdeletions were detected in non-del(5q) samples, confirming that loss of one copy of the 5q32.1 genes in de-novo MDS predominantly occurs via large cytogenetic deletions affecting numerous genes. This finding reinforces the pathogenic role of haploinsufficiency of multiple 5q genes in myeloid neoplasms.

Candidate genes in the pathogenesis of del(5q) in myeloid malignancies

Early growth response 1 (EGR1), a gene located on chromosome 5, functions as a tumor suppressor in various cancers, including breast cancer, fibrosarcoma, and glioblastoma. In mouse models, EGR1 mediates cellular responses to growth factors, mitogens, and stress, acting as a direct transcriptional regulator of tumor suppressor genes, including TP53, transforming growth factor beta, and phosphatase and tensin homolog [19]. EGR1-knockout mouse embryonic fibroblasts bypass senescence and become immortalized, suggesting a gatekeeper role in p53-dependent growth regulation [20]. Heterozygous and homozygous loss of EGR1, coupled with alkylating agent-induced secondary mutations, leads to myeloid neoplasms in mice [21]. EGR1 was universally deleted in a large cohort of 1,200 patients with MDS or AML with del(5q) [15]. Indicating that loss of EGR1 function contributes to the pathogenesis of these myeloid neoplasms by permitting uncontrolled cell growth and impairing cellular differentiation and apoptosis.

Hosono, et al. [22] employed SNP-A and next-generation sequencing (NGS) to investigate pathogenic molecular features in patients with MDS and AML with del(5q), either as the sole abnormality or part of a complex karyotype. Several genes, including those encoding casein kinase 1 alpha 1 (CSNK1A1) and G3BP stress granule assembly factor 1 (G3BP1) within the distal CDR (5q31.1–5q33.1), and DEAD-box helicase 41 (DDX41) within CRR-2, were frequently mutated and exhibited consistent haploinsufficiency in deletion cases. Low expression or mutations in G3BP1 or DDX41 are associated with poor survival. CSNK1A1 is a putative tumor suppressor, whereas G3BP1 regulates p53 activity. DDX41 is involved in RNA splicing, and germline mutations in this gene predispose individuals to MDS and AML.

Chromosome 5 and/or 7 deletions are also found in therapy-related AML (t-AML) following prolonged azathioprine treatment [23, 24], presumably owing to defective DNA mismatch repair, leading to leukemic clone formation. Microsatellite instability occurs in patients with t-AML and those >60 yrs old (but not in younger patients with de-novo AML) and is associated with chromosome 5 and/or 7 abnormalities. This provides functional evidence for DNA mismatch repair defects in t-AML [24]. Deficient expression of the MutS homolog 2 gene (MSH2), involved in DNA mismatch repair, is observed in adult AML cases, predominantly in patients aged >60 yrs and those with t-AML, and is associated with TP53 mutations and microsatellite instability. Therefore, defective DNA mismatch repair because of reduced MSH2 expression may play a key role in leukemogenesis, particularly in older patients and those with prior chemotherapy exposure [25].

The nucleophosmin 1 gene (NPM1), which encodes nucleophosmin, a chaperone protein that shuttles between the nucleus and cytoplasm, is located at 5q35. NPM1 has roles in TP53 regulation, ribosome biogenesis, and maintaining genomic stability [26]. NPM1 is the most frequently mutated gene in AML, occurring in ~30% of cases, including at least 50% of AML with a normal karyotype [27]. NPM1 is lost in 10% of MDS cases because of large 5q deletions and is associated with a poor clinical course and increased likelihood of progression to AML [28, 29]. NPM1 haploinsufficiency is hypothesized to offer a proliferative advantage to myeloid clones [30]. Mice heterozygous for NPM1 have an increased rate of spontaneous malignancy, with 75% developing myeloid and lymphoid neoplasms [31]. NPM1 knockout is embryonically lethal. Conditional NPM1-knockout mice develop an MDS-like phenotype with aging, whereas combined heterozygous knockout of NPM1 and TP53 results in myeloid leukemia in >90% of mice. NPM1 loss appears to cause an MDS-like phenotype, which promotes AML when TP53 is lost [32]. Therefore, aneuploidy with loss of chromosome 5 and/or 7 in the context of mutated TP53 may promote genetic instability because of NPM1 loss, promoting transformation to AML.

Chromosome 7 deletions in myeloid malignancies

The CDR in –7/del(7q) myeloid neoplasms is located in the 7q22 region [11, 33]. Mapping with SNP-A and whole-genome sequencing has identified deletion of 7q21.13–36.3 in >70% of cases and deletions of 7q31.33, 7q34, and 7q35–7q36.1 in over 90% of cases. These areas comprise 304 genes, including those encoding sterile alpha motif domain-containing 9 (SAMD9), cut-like homeobox 1 (CUX1), dedicator of cytokinesis 4 (DOCK4), cullin 1 (CUL1), enhancer of zeste 2 polycomb-repressive complex 2 subunit (EZH2), lysine methyltransferase 2C (KMT2C), and LUC7-like 2, pre-mRNA splicing factor (LUC7L2) [11] (Fig. 2). Analyses of somatic mutations in chromosome 7 in cases with –7/del(7q) have identified 16 recurrently mutated genes. EZH2 mutations are the most common (5% of cases) and are associated with hemizygous deletion of EZH2 on one allele and EZH2 mutation on the other in 16% of cases. In isolated –7/del(7q), the most recurrently mutated genes are those encoding Tet methylcytosine dioxygenase 2 (TET2), DNA methyltransferase 3 alpha (DNMT3A), ASXL transcriptional regulator 1 (ASXL1), RUNX family transcription factor 1 (RUNX1), and serine and arginine-rich splicing factor 2 (SRSF2), and EZH2. In contrast, in –7/del(7q) associated with a complex karyotype, TP53 is the most commonly mutated (57%), followed by del(5q) (>50%) and del(17p) (25%) [11].

Figure 2. CDRs of chromosome 7q associated with MDS and AML. The CDR on chromosome 7q spans 7q21.13–7q36.3, with regions corresponding to bands 7q31.33, 7q34, and 7q35–7q36.1 deleted in the majority of cases [15]. Genes of interest in the pathogenesis of –7/del(7q) myeloid neoplasms include SAMD9/SAMD9L, CUX1, and EZH2. This figure was created with BioRender.com.
Abbreviations: MDS, myelodysplastic syndrome; CDR, commonly deleted region; SAMD9/SAMD9L, sterile alpha motif domain-containing 9/sterile alpha motif domain-containing 9-like; CUX1, cut-like homeobox 1; EZH2, enhancer of zeste 2 polycomb-repressive complex 2 subunit.

Candidate genes in the pathogenesis of deletion 7 in myeloid malignancies

The genes within the CDRs have been analyzed to elucidate leukemogenesis associated with –7/del(7q). Theories include LOH, haploinsufficiency of tumor suppressor genes, and somatic rescue of suppressive germline variants located within the deleted regions. Additionally, tumor suppressor genes may modulate genes on other chromosomes through downstream pathways or compensatory mechanisms [11].

Mori, et al. [11] used NGS to analyze clonal hierarchy and define the initial molecular events in the pathogenesis of –7/del(7q). These events varied based on associated genomic complexity. In the majority of isolated –7/del(7q) cases, the chromosome 7 abnormality was secondary, and mutations in ASXL1 or TET2 were the most common dominant somatic alterations. Conversely, in del(7q) myeloid neoplasms with complex karyotypes, 73% exhibited dominant TP53 mutations. Single-cell DNA sequencing indicated loss of chromosome 7 as a secondary event following DNMT3A and TET2 mutations in a case of AML with isolated –7 and subsequent to TP53 mutation and del(5q) in another case with subclonal del(7q). This suggests that –7/del(7q) may not be a primary event in leukemogenesis and can occur secondary to other genomic defects.

Bioinformatics analysis of haploinsufficiency within the 7q21–7q36 CDR in –7/del(7q) cases identified 199 haploinsufficient genes, including tumor suppressors, genes involved in DNA damage response, DNA replication, and chromatin regulation. CUX1 and EZH2 exhibited haploinsufficiency in >90% of cases and were associated with lethality in knockout mice. Expression analysis of genes outside the defined CDRs on chromosome 7 helped identify 19 significantly upregulated genes and seven downregulated genes, including a tumor suppressor gene (ribosomal protein S6 kinase A1) and two oncogenes (lysosomal protein transmembrane 4 beta and Ras homolog family member C). These findings suggest impaired DNA damage repair may contribute to the genesis of myeloid neoplasms with –7/del(7q) and align with the known association of –7/del(7q) in AML with genotoxic exposure (i.e., t-AML) [11].

Mono- or biallelic EZH2 mutations at 7q35 are among the most frequent mutations in myeloid malignancies with chromosome 7 deletions, affecting 10% of MDS and t-AML). EZH2 encodes a methyltransferase that acts as an epigenetic regulator and contributes to gene silencing via histone modification [34]. Somatic EZH2 mutations confer an unfavorable prognosis, including the risk of transformation to AML [35]. Biallelic mutations are often associated with CN-LOH of the distal portion of 7q, suggesting that EZH2 haploinsufficiency may promote leukemogenesis. However, EZH2 deletion in mice resulted in T-cell leukemia rather than myeloid neoplasms [36]. EZH2 mutations are commonly associated with TET2 mutations in myeloid neoplasms. In in-vitro TET2- or RUNX1-knockdown models, the incidence of aggressive myeloid disease increased when EZH2 was deleted, suggesting that additional loss of EZH2 contributes to myeloid neoplasia development [37].

CUX1, located at 7q22.1, encodes a homeobox transcription factor and is implicated in –7/del(7q) malignancies and may fiunction as a role as a haploinsufficient tumor suppressor. CUX1–/– mice do not survive past weaning and exhibit myeloid hyperplasia [38, 39].

SAMD9 and SAMD9L (SAMD9-like), at 7q21.2, encode endosomal proteins crucial for endosomal trafficking, including cytokine receptor metabolism. Mutations in SAMD9/SAMD9L are associated with hereditary diseases predisposing to MDS with monosomy 7 [40, 41]. Germline gain-of-function (GOF) mutations have been identified in children and adolescents with inherited bone marrow failure syndromes and MDS [42]. The transcription factor EVI1, frequently associated with therapy-resistant myeloid malignancies bearing inv(3)/t(3;3) abnormalities [43], suggests that haploinsufficiency of SAMD9L at 7q21 and over-function of EVI1 may cooperatively promote myeloid leukemogenesis [40].

Karyotyping

ELN prognostic groups are defined on conventional karyotypic analysis, which is mandated in the diagnostic evaluation of AML [4]. Giemsa staining of metaphase chromosomes after short-term culture (G-banding) enables detection of numerical changes and structural variations (SVs), including deletions, translocations, inversions, and duplications (Fig. 3A). This method requires dividing cells, and failure rates for AML samples can reach up to 20% [44]. While G-banding offers a comprehensive view of the chromosome complement of the cell, its resolution is limited—deletions <5–10 Mb and rare events such as mosaicism may be missed. Furthermore, this technique cannot detect CN-LOH or acquired segmental uniparental disomy.

Figure 3. Examples of chromosome abnormalities via karyotyping and FISH. (A) Karyotype of AML with a monosomal karyotype, with losses of multiple chromosomes, including monosomy 7. (B) del(5q) in a case of AML. FISH using a dual color probe set for EGR1 (5q31) and D5S23 (5p15.2) (one orange signal for EGR1 and two green signals for D5S23).
Abbreviations: EGR1, early growth response 1; D5S23, human D5S23 control region on 5p.

FISH

FISH uses fluorescently labeled DNA probes to interrogate chromosomal sequences of interest and can identify gene fusions, deletions, or aneuploidies using various probe designs, including deletion/duplication probe sets or whole-chromosome paints [45]. In AML, prognostically significant abnormalities routinely targeted by FISH include recurrent translocations (e.g., RUNX1::RUNX1 partner transcriptional co-repressor 1, core-binding factor subunit beta::myosin heavy chain 11) and gene fusions, and myelodysplasia-related chromosome abnormalities [e.g., del(5q)/add(5q), –7/del(7q), and –17/add(17p)/del(17p)] [4]. FISH can be performed on non-dividing interphase cells, making it applicable when karyotyping fails [46]. Deletion/duplication probe sets can help detect del(5q) and monosomy 5 (Fig. 3B). 5q deletions are detectable with FISH probes targeting 5q31.2 (EGR1) and 5q33.1 (ribosomal protein S14 [RPS14]), corresponding to CDR-2 and CDR-1, respectively, with a concurrent 5p probe enabling monosomy 5 detection [47]. EGR1 FISH is more sensitive than G-band karyotyping for detecting del(5q) in MDS (6%). Therefore, FISH studies for del(5q) are recommended in cases with insufficient cell growth or <20 normal metaphases in karyotyping and when rapid results are needed to guide treatment choice (e.g., lenalidomide for 5q– syndrome) [46].

Probes targeting the 7q CDR, including 7q22 and 7q36, have been used to identify del(7q) and a centromeric probe for monosomy 7 in MDS and AML [48]. FISH studies have helped detect a high monosomy 7 rate in cytogenetically normal MDS and AML cases. The frequency of occult monosomy 7 was up to 30% [49, 50]. MDS patients with monosomy 7 detected using FISH are more likely to progress to AML [49].

SNP-A

Newer genomic technologies such as chromosome arrays and NGS give more precise characterization of chromosomal abnormalities. SNP-A enables high-resolution whole-genome scanning for unbalanced defects, such as deletions, by assessing individual base pairs. Fragmented single-stranded DNA is fluorescently labeled and hybridized to a microarray chip containing hundreds of thousands of unique nucleotide probes. The fluorescence intensity measured at each probe location helps quantify gene dosage and highlights cryptic changes, such as small deletions. SNP-A can identify CN-LOH, which is undetectable with metaphase karyotyping and common in myeloid malignancies [51]. CN-LOH may result in the duplication of oncogenic mutations in genes such as Janus kinase 2, KIT proto-oncogene, receptor tyrosine kinase, and FMS-related receptor tyrosine kinase 3 (FLT3), with concurrent loss of the normal allele, or loss of tumor suppressor genes such as c-CBL and TET2 [52]. When applied to MDS and AML cases with del(5q), SNP-A has facilitated precise breakpoint identification and revealed additional cryptic lesions in >50% of cases and retention of chromosome 5q material in marker chromosomes [14]. In AML cases with a normal karyotype, SNP-A has revealed chromosomal rearrangements in 32% of cases and CN-LOH in 23% [51]. The presence and number of abnormal SNP lesions were independent predictors of poorer overall survival (OS) in MDS and AML [53].

The resolution of SNP-A is limited by the finite number of probes, and smaller CNVs or SNPs may be missed because of probe spacing. As SNP-A uses a reference genome, variants that are absent in the reference genome may be overlooked, and population-specific variations under-represented. Furthermore, SNP-A can be expensive, and high-throughput analysis of large datasets requires substantial resources and bioinformatics experience. Thus, SNP-A is not routinely used in diagnostic practice to analyze chromosome 5 and 7 integrity.

NGS

In NGS, nucleic acids are fragmented and attached to a solid surface (library preparation), and the entire library is sequenced simultaneously, generating large quantities of complex data consisting of short DNA reads. Bioinformatics software is used to annotate variants. NGS offers higher resolution than karyotyping and FISH and does not require dividing cells or prior knowledge of abnormalities. NGS is primarily used in AML diagnostics to screen targeted panels covering a limited number of driver gene mutations, including in FLT3, NPM1, DNMT3A, and TP53, as recommended by international guidelines [4, 54]. Of the 36 genes currently recommended for screening by the ELN-2022 guidelines, four are located on chromosomes 5 and 7, including NPM1 and EZH2 at 7q36.1 within the 7q CDR. Another important gene in myeloid neoplasms is the B-Raf proto-oncogene, serine/threonine kinase gene (BRAF), located at 7q34 within the 7q22–q34 CDR [11]. This gene encodes a Raf family serine/threonine protein kinase that regulates the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway, which affects cell division and differentiation. BRAF mutations in AML are associated with poor prognosis regardless of clonal burden and are enriched in MDS-related AML (AML-MR) [55]. Detection of these mutations by NGS may indicate the presence of 5q or 7q deletions. Mate-pair sequencing (MPseq), an NGS variant involving a unique library preparation method with circularization of long input DNA (2–5 kb), enables paired-end sequencing of reads that map 2–5 kb apart in the genome and facilitates whole-genome sequencing optimized for the detection of SVs and CNVs [56]. MPseq allows comprehensive CNV evaluation of a sample in a single assay. The reduced sequencing depth makes it more cost-effective than traditional NGS [57]. MPseq demonstrates utility in high-resolution detection and improved characterization of chromosome 5 and 7 abnormalities, genomic complexity, and TP53 mutation detection in AML [58] but does not improve risk stratification [59].

Genetic material is commonly lost from chromosomes 5 and 7 in MDS [16, 60]. Deletion of 5q, 7q, or monosomy 7 are defining cytogenetic abnormalities in the WHO classification and recognized by the International Consensus Classification (ICC) as sufficient for the diagnosis of MDS/AML or AML with MDS-related cytogenetic abnormalities in the absence of other recurrent genetic abnormalities or gene mutations [4, 5]. These chromosomal defects are also present in other myeloid malignancies, including myeloproliferative neoplasms (MPN) and MDS/MPN albeit at lower frequencies [16, 60].

Chromosome 5 and 7 deletions in MDS

Interstitial chromosome 5q deletion is the most common karyotypic abnormality in de-novo MDS, occurring in up to 20% of cases and chromosome 7 abnormalities in 10% [61]. Chromosome 5 and 7 abnormalities may appear as the sole abnormality or as part of a complex karyotype [61]. Detection of these is important as they provide critical prognostic information regarding outcomes and the risk of transformation to AML [62]. The cytogenetic risk classification system identifies five groups: very good, good, intermediate, poor, and very poor [62, 63].

Deletion 5q confers a good prognosis, with a median survival of 4.8 yrs, and a low risk of evolution to AML [63]. Although monosomy 5 is not specifically addressed in the MDS prognostic scoring system, patient survival is worse than that of patients with sole del(5q) [64]. Deletion 7q alone is of intermediate risk, with a 2.7-yr median survival. Monosomy 7 with or without del(7q) confers a poor risk, with a 1.5-yr survival and a median survival of 1.7 yrs for the 25% of cases that evolve to AML [63]. Monosomy 7 is associated with a high rate of progression to AML relative to del(7q) [63, 65], and the presence of chromosome 7 anomalies is a potential indicator of leukemic evolution in MDS [66].

5q– syndrome

5q deletion was the first chromosomal deletion associated with a specific hematological phenotype [7]. The WHO has now names this entity “myelodysplastic neoplasm with low blasts and 5q deletion.” Deletion of 5q frequently is the sole cytogenetic abnormality in patients with 5q– syndrome. Patients are predominantly female and present with refractory macrocytic anemia and thrombocytosis. The bone marrow exhibits erythroid hyperplasia, small megakaryocytes with non-lobated nuclei, and relatively preserved granulocytic activity (Fig. 4). The WHO criteria include <5% blasts, absence of Auer rods, no more than one other cytogenetic abnormality, and absence of –7/del(7q). Notably, most patients with MDS and del(5q) do not meet these criteria for “MDS with isolated del(5q).” Patients respond to lenalidomide therapy but some are transfusion-dependent. The prognosis is generally favorable, with low rates of transformation to AML.

Figure 4. Bone marrow morphology of a myelodysplastic neoplasm with low blasts and isolated del(5q). (A) Bone marrow aspirate showing a small megakaryocyte with a non-lobated eccentric nucleus (Romanowsky staining, 1,000×). (B) Section of the bone marrow trephine showing megakaryocytic hyperplasia with a predominance of characteristic small forms with hypolobated nuclei (hematoxylin and eosin staining, 400×).

Additional CNVs involving other chromosomes, including 17p, are rare in del(5q), as are large areas of CN-LOH [14]. Other leukemia-associated mutations, such as in the NRAS proto-oncogene, GTPase gene (NRAS), and FLT3, are also uncommon [13]. Therefore, the favorable prognosis for patients with 5q– syndrome may relate to their relative genetic stability. The consistent clinical phenotype of 5q– syndrome suggests that the loss of one or more genes within CDR-1 is pathogenic. This area includes 40 genes encoding proteins and microRNAs, with numerous genes encoding hematopoietic growth factors and receptors [12]. Studies using Sanger and NGS technologies, SNP-A, and gene expression profiling have failed to identify a tumor suppressor gene on 5q with both alleles inactivated, supporting the notion that haploinsufficiency for one or more genes on 5q contributes to the molecular pathophysiology of MDS [67]. Functional studies using RNA interference have identified critical genes. Research using short hairpin RNAs targeting RPS14 demonstrated that partial loss of RPS14 function results in anemia because of reduced erythroid production, whereas RPS14 overexpression in MDS patient-derived hematopoietic cells rescued the phenotype [68]. The erythroid defect was analogous to that observed in Diamond–Blackfan anemia, a congenital bone marrow failure syndrome caused by heterozygous mutations in ribosomal proteins [69]. Defective ribosomal biogenesis may induce high p53 expression by interfering with the ubiquitin ligase mouse double minute 2 homolog, which targets p53 for proteasomal destruction [70]. Bone marrow-derived CD34-positive stem cells from patients with 5q– syndrome show marked downregulation of the microRNAs miR-145 and miR-146a. Loss of one allele of these microRNAs results in increased megakaryocyte production and megakaryocytic dysplasia via activation of the nuclear factor kappa-light-chain-enhancer of activated B cells pathway. MicroRNA-145 and RPS14 co-localize within 1 Mb in CDR-1; thus, their combined loss may be sufficient to explain the clinical features of 5q– syndrome [10].

Chromosome 5 and 7 deletions occur frequently, with each present in 5–15% of cases of de-novo AML [7174]. In half of these cases, chromosome 5 and 7 abnormalities co-occur and represent the most common chromosomal abnormalities to co-segregate [8]. The relative frequency of these abnormalities increases with age, prior chemotherapeutic exposure, and a history of myeloid disease [73, 74].

t-AML

t-AML is increasing in incidence, representing 10%–15% of all AML cases, and occurs more commonly in older patients [73]. Two types of t-AML with distinct cytogenetic profiles are recognized: those caused by alkylating agents or ionizing radiation and those induced by topoisomerase II inhibitors [75]. Alkylating agent-related t-AML accounts for 75% of cases and typically presents with cytopenia and multilineage dysplasia 4–7 yrs after exposure to the mutagen [76].

The prevalence of chromosome 5 or 7 abnormalities in t-AML is estimated at 20%–30%, with 70%–80% harboring unbalanced chromosomal deletions of either chromosome [77]. del(5q) in t-AML is frequently associated with additional changes, usually in the context of a complex or monosomal karyotype [78, 79]. TP53 mutation is found in 80% of del(5q) but is not significantly associated with del(7q) or cases with balanced chromosomal translocations [79]. TP53 mutations in t-AML are associated with a highly complex genetic profile (mean number of genomic alterations, 7.5) and genomic instability [78]. Chromosome 5 and/or 7 deletions, TP53 mutation, and a complex karyotype are hallmarks of alkylator-associated t-AML.

AML, myelodysplasia-related (AML-MR)

AML-MR describes a subset of AML with features overlapping with those of MDS [5]. AML-MR represents up to 50% of adult AML and is most common age >60 yrs [80, 81]. These cases may arise following a history of MDS or MDS/MPN, or de novo and genetic characteristics, rather than clinical history, have the highest biological relevance [4, 5, 78, 82]. The most frequent abnormalities are a complex karyotype, del(5q), del(7q), or monosomy 7 [4, 5]. The increase in chromosome 5 and 7 abnormalities with age likely reflects a complex interplay between aging-related genetic changes, prior therapy, and myeloid disease progression.

AML with complex karyotype (AML-CK)

Chromosome 5 and 7 abnormalities often co-exist and in association with other chromosomal abnormalities in the context of a complex or monosomal karyotype. AML-CK is defined by the presence of three or more unrelated chromosomal aberrations and the absence of favorable cytogenetic rearrangements. AML-CK comprises 10%–12% of AML, and its incidence increases with age [83]. The median number of chromosomal aberrations ranges from 6 to 10, show a non-random pattern, with a relative paucity of balanced translocations and a predominance of monosomies, deletions, and unbalanced translocations, leading to loss of chromosome material, resulting in a hypodiploid karyotype [8386]. Approximately 80% of patients show loss of genomic material from 5q, and 50% have 7q deletions and 17p abnormalities andthese abnormalities often co-occur [73, 83]. In AML, –7/del(7q) may be the sole abnormality or as part of a complex karyotype. In contrast –5/del(5q) tends to occur only as part of the complex karyotyp; however the chromosome is rarely lost entirely and parts of the chromosome are frequently found in marker and/or ring chromosomes [83]. However, the net result is still loss of chromosome 5 material, primarily from 5q.

AML with monosomal karyotype

AML with monosomal karyotype (AML-MK) is defined by the presence of at least two autosomal monosomies or a single autosomal monosomy in combination with at least one structural abnormality [87]. AML-MK comprises 20%–30% of cases in patients aged >60 yrs (cf <5% in <30 yrs) and is more common with preceding MDS or t-AML [71, 87]. Chromosome 5 and 7 abnormalities are frequently observed with AML-MK. Monosomy 5 occurs in 55% and –7 in 45% of adult AML cases, commonly in association with other changes, including del(12p), del(17p), –18/del(18q), –20/del(20q) and inv(3)/t(3;3), a complex karyotype, and MDS-related cytogenetic abnormalities [88]. NPM1 and FLT3 mutations are less frequent in AML-MK, whereas TP53 alterations occur in 70% of patients [88]. Furthermore, they appear to be more frequent in patients with both complex and monosomal karyotypes than in those with a complex karyotype alone [89].

TP53-mutated AML

The transcription factor p53 is pivotal in normal hematopoiesis, and aberrations are early events in myeloid leukemogenesis. TP53, on the short arm of chromosome 17 (17p), can lose functionality through deletion of chromosome 17 or 17p, or through gene mutation. TP53 mutations may result in LOF or GOF and have been detected in founding clones in clonal hematopoiesis of indeterminate potential, MDS, AML with a complex karyotype, and t-AML [82, 90]. Most TP53-mutant cases exhibit complex karyotypes, and in ~50%, TP53 mutations occur without other AML-associated gene mutations. “AML with TP53 mutations, chromosomal aneuploidies, or both” occurs in up to 15% of cases, are generally older patients, and have an association with chromosome 5 and/or 7 deletions in a complex karyotype [82]. Pitel et al. used mate-pair sequencing (MPseq) to explore the relationships among 5q and 7q deletions, genomic complexity, and TP53 mutations in MDS and AML [56]. They demonstrated that del(5q) and, particularly, del(5q)/(7q) subtypes showed higher genomic complexity than AML-MK and del(7q) subtypes. Cases with del(5q)/(7q) exhibited significantly higher genome-wide copy number gains and losses and SVs (median, 14.0, 24.0, and 60.0, respectively) than those with a normal karyotype. TP53 deletions were identified in 55% of del(5q)/(7q), 44% of del(5q) cases, and only 9% of those with del(7q), with no deletions found in AML with a normal karyotype. Monoallelic and biallelic TP53 SNVs were significantly increased in del(5q) and del(5q)/(7q) cases and rare in del(7q) and normal karyotype cases [22, 78, 82]. A history of prior myeloid disorder or chemotherapy, increasing age, and the presence of chromosome 5 and/or 7 abnormalities imply the probable presence of TP53 mutation in AML.

Chromosome 5 and 7 abnormalities in pediatric AML

Chromosome 5 and 7 abnormalities are also highly prevalent in pediatric AML, particularly when they evolve from a prior bone marrow disorder [91]. Chromosome 7 deletions are commonly implicated in bone marrow failure syndromes, such as Fanconi anemia, severe congenital neutropenia, Shwachman–Diamond syndrome, and aplastic anemia [92, 93]. Germline SAMD9/9L and GATA-binding protein 2 mutations have been reported in pediatric MDS with –7 [94]. Complex and monosomal karyotypes are less common in pediatric patients.

Prognostic impact of chromosome 5 and 7 abnormalities in MDS and AML

Karyotype and mutation status are the most important independent prognostic factors of AML and determine the likelihood of achieving complete remission, relapse risk, and OS [4, 71, 72, 82, 95]. Therefore, guidelines mandate cytogenetic analysis at diagnosis to stratify patients into risk groups [4, 96]. The ELN-2022 AML classification includes del(5q), –5, and –7, as well as complex and monosomal karyotypes, monosomy 17/abnormal 17p, and mutated TP53 in the adverse risk category.

AML clinical trials have shown that chromosome 5 abnormalities [del(5q), add(5q), and –5] or monosomy 7 are associated with a low complete remission rate (53%–58%) and a 10-yr OS of 0%–12%. Deletion of 7q is less adverse, with a 10-yr OS of 26%–30% [95]. Most chromosome 5 and 7 abnormalities occur in the context of a complex karyotype, and additional cytogenetic abnormalities likely adversely influence the outcome [72]. Evidence suggests that chromosome 7 deletion is a highly unfavorable prognostic factor conferring a worse prognosis than a complex karyotype without –7/del(7q) [11]. A monosomal karyotype also confers a dismal prognosis, which may be partly attributed to monosomy 7 [87]. TP53 mutations, which frequently co-occur with complex and monosomal karyotypes involving chromosomes 5 and 7, further adversely impact prognosis [15, 82]. Current guidelines recommend offering allogeneic HSCT to patients with unfavorable-risk AML [4]. However, long-term outcomes remain poor, with few achieving long-term survival [97].

Chromosome 5 and 7 deletions are prevalent in MDS and AML, involving large genomic areas that encompass numerous genes. These deletions are markers of increased genomic complexity, aneuploidy, TP53 mutations, and adverse risk; therefore, their identification is crucial for prognostic stratification and treatment planning. Currently, testing relies on karyotyping and FISH; however, emerging laboratory techniques promise enhanced resolution, accuracy, and high-throughput genomic characterization of leukemic cells.

  1. Schmutz J, Martin J, Terry A, Couronne O, Grimwood J, Lowry S, et al. The DNA sequence and comparative analysis of human chromosome 5. Nature 2004;431:268-74.
    Pubmed CrossRef
  2. Scherer SW, Cheung J, MacDonald JR, Osborne LR, Nakabayashi K, Herbrick JA, et al. Human chromosome 7: DNA sequence and biology. Science 2003;300:767-72.
    Pubmed KoreaMed CrossRef
  3. Mrózek K, Heinonen K, Bloomfield CD. Clinical importance of cytogenetics in acute myeloid leukaemia. Best Pract Res Clin Haematol 2001;14:19-47.
    Pubmed CrossRef
  4. Döhner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022;140:1345-77.
    Pubmed CrossRef
  5. Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al. The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia 2022;36:1703-19.
    Pubmed KoreaMed CrossRef
  6. DiNardo CD, Erba HP, Freeman SD, Wei AH. Acute myeloid leukaemia. Lancet 2023;401:2073-86.
    Pubmed CrossRef
  7. Van den Berghe H, Cassiman JJ, David G, Fryns JP, Michaux JL, Sokal G. Distinct haematological disorder with deletion of long arm of no. 5 chromosome. Nature 1974;251:437-8.
    Pubmed CrossRef
  8. Castro PD, Liang JC, Nagarajan L. Deletions of chromosome 5q13.3 and 17p loci cooperate in myeloid neoplasms. Blood 2000;95:2138-43.
    Pubmed CrossRef
  9. Lai F, Godley LA, Joslin J, Fernald AA, Liu J, Espinosa R 3rd, et al. Transcript map and comparative analysis of the 1.5-Mb commonly deleted segment of human 5q31 in malignant myeloid diseases with a del(5q). Genomics 2001;71:235-45.
    Pubmed CrossRef
  10. Barlow JL, Drynan LF, Trim NL, Erber WN, Warren AJ, McKenzie AN. New insights into 5q- syndrome as a ribosomopathy. Cell Cycle 2010;9:4286-93.
    Pubmed CrossRef
  11. Mori M, Kubota Y, Durmaz A, Gurnari C, Goodings C, Adema V, et al. Genomics of deletion 7 and 7q in myeloid neoplasm: from pathogenic culprits to potential synthetic lethal therapeutic targets. Leukemia 2023;37:2082-93.
    Pubmed KoreaMed CrossRef
  12. 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.
    Pubmed CrossRef
  13. Wang L, Fidler C, Nadig N, Giagounidis A, Della Porta MG, Malcovati L, et al. Genome-wide analysis of copy number changes and loss of heterozygosity in myelodysplastic syndrome with del(5q) using high-density single nucleotide polymorphism arrays. Haematologica 2008;93:994-1000.
    Pubmed CrossRef
  14. Jerez A, Gondek LP, Jankowska AM, Makishima H, Przychodzen B, Tiu RV, et al. Topography, clinical, and genomic correlates of 5q myeloid malignancies revisited. J Clin Oncol 2012;30:1343-9.
    Pubmed KoreaMed CrossRef
  15. Volkert S, Kohlmann A, Schnittger S, Kern W, Haferlach T, Haferlach C. Association of the type of 5q loss with complex karyotype, clonal evolution, TP53 mutation status, and prognosis in acute myeloid leukemia and myelodysplastic syndrome. Genes Chromosomes Cancer 2014;53:402-10.
    Pubmed CrossRef
  16. Stengel A, Kern W, Haferlach T, Meggendorfer M, Haferlach C. The 5q deletion size in myeloid malignancies is correlated to additional chromosomal aberrations and to TP53 mutations. Genes Chromosomes Cancer 2016;55:777-85.
    Pubmed CrossRef
  17. Jasek M, Gondek LP, Bejanyan N, Tiu R, Huh J, Theil KS, et al. TP53 mutations in myeloid malignancies are either homozygous or hemizygous due to copy number-neutral loss of heterozygosity or deletion of 17p. Leukemia 2010;24:216-9.
    Pubmed KoreaMed CrossRef
  18. Graubert TA, Payton MA, Shao J, Walgren RA, Monahan RS, Frater JL, et al. Integrated genomic analysis implicates haploinsufficiency of multiple chromosome 5q31.2 genes in de novo myelodysplastic syndromes pathogenesis. PLoS One 2009;4:e4583.
    Pubmed KoreaMed CrossRef
  19. Baron V, Adamson ED, Calogero A, Ragona G, Mercola D. The transcription factor Egr1 is a direct regulator of multiple tumor suppressors including TGFbeta1, PTEN, p53, and fibronectin. Cancer Gene Ther 2006;13:115-24.
    Pubmed KoreaMed CrossRef
  20. Krones-Herzig A, Adamson E, Mercola D. Early growth response 1 protein, an upstream gatekeeper of the p53 tumor suppressor, controls replicative senescence. Proc Natl Acad Sci U S A 2003;100:3233-8.
    Pubmed KoreaMed CrossRef
  21. Joslin JM, Fernald AA, Tennant TR, Davis EM, Kogan SC, Anastasi J, et al. Haploinsufficiency of EGR1, a candidate gene in the del(5q), leads to the development of myeloid disorders. Blood 2007;110:719-26.
    Pubmed KoreaMed CrossRef
  22. Hosono N, Makishima H, Mahfouz R, Przychodzen B, Yoshida K, Jerez A, et al. Recurrent genetic defects on chromosome 5q in myeloid neoplasms. Oncotarget 2017;8:6483-95.
    Pubmed KoreaMed CrossRef
  23. Leone G, Pagano L, Ben-Yehuda D, Voso MT. Therapy-related leukemia and myelodysplasia: susceptibility and incidence. Haematologica 2007;92:1389-98.
    Pubmed CrossRef
  24. Das-Gupta EP, Seedhouse CH, Russell NH. Microsatellite instability occurs in defined subsets of patients with acute myeloblastic leukaemia. Br J Haematol 2001;114:307-12.
    Pubmed CrossRef
  25. Zhu YM, Das-Gupta EP, Russell NH. Microsatellite instability and p53 mutations are associated with abnormal expression of the MSH2 gene in adult acute leukemia. Blood 1999;94:733-40.
    Pubmed CrossRef
  26. Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer 2006;6:493-505.
    Pubmed CrossRef
  27. Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci L, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med 2005;352:254-66.
    Pubmed CrossRef
  28. Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 2014;28:241-7.
    Pubmed KoreaMed CrossRef
  29. Nazha A, Bejar R. Molecular Data and the IPSS-R: how mutational burden can affect prognostication in MDS. Curr Hematol Malig Rep 2017;12:461-7.
    Pubmed CrossRef
  30. Di Matteo A, Franceschini M, Chiarella S, Rocchio S, Travaglini-Allocatelli C, Federici L. Molecules that target nucleophosmin for cancer treatment: an update. Oncotarget 2016;7:44821-40.
    Pubmed KoreaMed CrossRef
  31. Sportoletti P, Grisendi S, Majid SM, Cheng K, Clohessy JG, Viale A, et al. Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse. Blood 2008;111:3859-62.
    Pubmed KoreaMed CrossRef
  32. Zarka J, Short NJ, Kanagal-Shamanna R, Issa GC. Nucleophosmin 1 mutations in acute myeloid leukemia. Genes (Basel) 2020;11:649.
    Pubmed KoreaMed CrossRef
  33. Jerez A, Sugimoto Y, Makishima H, Verma A, Jankowska AM, Przychodzen B, et al. Loss of heterozygosity in 7q myeloid disorders: clinical associations and genomic pathogenesis. Blood 2012;119:6109-17.
    Pubmed KoreaMed CrossRef
  34. Simon JA, Kingston RE. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol 2009;10:697-708.
    Pubmed CrossRef
  35. Jiang L, Ye L, Ma L, Ren Y, Zhou X, Mei C, et al. Predictive values of mutational variant allele frequency in overall survival and leukemic progression of myelodysplastic syndromes. J Cancer Res Clin Oncol 2022;148:845-56.
    Pubmed CrossRef
  36. Simon C, Chagraoui J, Krosl J, Gendron P, Wilhelm B, Lemieux S, et al. A key role for EZH2 and associated genes in mouse and human adult T-cell acute leukemia. Genes Dev 2012;26:651-6.
    Pubmed KoreaMed CrossRef
  37. Sashida G, Harada H, Matsui H, Oshima M, Yui M, Harada Y, et al. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat Commun 2014;5:4177.
    Pubmed CrossRef
  38. Sinclair AM, Lee JA, Goldstein A, Xing D, Liu S, Ju R, et al. Lymphoid apoptosis and myeloid hyperplasia in CCAAT displacement protein mutant mice. Blood 2001;98:3658-67.
    Pubmed CrossRef
  39. Supper E, Rudat S, Iyer V, Droop A, Wong K, Spinella JF, et al. Cut-like homeobox 1 (CUX1) tumor suppressor gene haploinsufficiency induces apoptosis evasion to sustain myeloid leukemia. Nat Commun 2021;12:2482.
    Pubmed KoreaMed CrossRef
  40. Nagamachi A, Matsui H, Asou H, Ozaki Y, Aki D, Kanai A, et al. Haploinsufficiency of SAMD9L, an endosome fusion facilitator, causes myeloid malignancies in mice mimicking human diseases with monosomy 7. Cancer Cell 2013;24:305-17.
    Pubmed CrossRef
  41. Schneider RK, Delwel R. Puzzling pieces of chromosome 7 loss or deletion. Blood 2018;131:2871-2.
    Pubmed CrossRef
  42. Pastor VB, Sahoo SS, Boklan J, Schwabe GC, Saribeyoglu E, Strahm B, et al. Constitutional SAMD9L mutations cause familial myelodysplastic syndrome and transient monosomy 7. Haematologica 2018;103:427.
    Pubmed KoreaMed CrossRef
  43. Hinai AA, Valk PJ. Review: aberrant EVI1 expression in acute myeloid leukaemia. Br J Haematol 2016;172:870-8.
    Pubmed CrossRef
  44. Cox MC, Panetta P, Venditti A, Del Poeta G, Franchi A, Buccisano F, et al. Comparison between conventional banding analysis and FISH screening with an AML-specific set of probes in 260 patients. Hematol J 2003;4:263-70.
    Pubmed CrossRef
  45. Bishop R. Applications of fluorescence in situ hybridization (FISH) in detecting genetic aberrations of medical significance. Int J Stud Res 2010;3:85-95.
    CrossRef
  46. Sun Y, Cook JR. Fluorescence in situ hybridization for del(5q) in myelodysplasia/acute myeloid leukemia: comparison of EGR1 vs. CSF1R probes and diagnostic yield over metaphase cytogenetics alone. Leuk Res 2010;34:340-3.
    Pubmed CrossRef
  47. Galván AB, Mallo M, Arenillas L, Salido M, Espinet B, Pedro C, et al. Does monosomy 5 really exist in myelodysplastic syndromes and acute myeloid leukemia?. Leuk Res 2010;34:1242-5.
    Pubmed CrossRef
  48. Zneimer SM. Cytogenetic Abnormalities: Chromosomal, FISH, and Microarray-Based Clinical Reporting and Interpretation of Result. Wiley, 2014.
    CrossRef
  49. Arif M, Tanaka K, Damodaran C, Asou H, Kyo T, Dohy H, Kamada N. Hidden monosomy 7 in acute myeloid leukemia and myelodysplastic syndrome detected by interphase fluorescence in situ hybridization. Leuk Res 1996;20:709-16.
    Pubmed CrossRef
  50. Brizard F, Brizard A, Guilhot F, Tanzer J, Berger R. Detection of monosomy 7 and trisomies 8 and 11 in myelodysplastic disorders by interphase fluorescent in situ hybridization. Comparison with acute non-lymphocytic leukemias. Leukemia 1994;8:1005-11.
    CrossRef
  51. Huh J, Mun YC, Chung WS, Seong CM. Ring chromosome 5 in acute myeloid leukemia defined by whole-genome single nucleotide polymorphism array. Ann Lab Med 2012;32:307-11.
    Pubmed KoreaMed CrossRef
  52. O'Keefe C, McDevitt MA, Maciejewski JP. Copy neutral loss of heterozygosity: a novel chromosomal lesion in myeloid malignancies. Blood 2010;115:2731-9.
    Pubmed KoreaMed CrossRef
  53. Tiu RV, Gondek LP, O'Keefe CL, Elson P, Huh J, Mohamedali A, et al. Prognostic impact of SNP array karyotyping in myelodysplastic syndromes and related myeloid malignancies. Blood 2011;117:4552-60.
    Pubmed KoreaMed CrossRef
  54. Yang F, Anekpuritanang T, Press RD. Clinical utility of next-generation sequencing in acute myeloid leukemia. Mol Diagn Ther 2020;24:1-13.
    Pubmed CrossRef
  55. Abu-Shihab Y, Nicolet D, Mrózek K, Routbort M, Patel KP, Walker CJ, et al. BRAF-mutated acute myeloid leukemia (AML) represents a Distinct, Prognostically Poor subgroup enriched in myelodysplasia-related (MR-)AML. Blood 2023;142(S1):1575.
    CrossRef
  56. Pitel BA, Zuckerman EZ, Baughn LB. Mate pair sequencing: next-generation sequencing for structural variant detection. Methods Mol Biol 2023;2621:127-49.
    Pubmed CrossRef
  57. Aypar U, Smoley SA, Pitel BA, Pearce KE, Zenka RM, Vasmatzis G, et al. Mate pair sequencing improves detection of genomic abnormalities in acute myeloid leukemia. Eur J Haematol 2019;102:87-96.
    Pubmed KoreaMed CrossRef
  58. Pitel BA, Sharma N, Zepeda-Mendoza C, Smadbeck JB, Pearce KE, Cook JM, et al. Myeloid malignancies with 5q and 7q deletions are associated with extreme genomic complexity, biallelic TP53 variants, and very poor prognosis. Blood Cancer J 2021;11:18.
    Pubmed KoreaMed CrossRef
  59. Pitel BA, Sharma N, Zepeda-Mendoza C, Smadbeck JB, Pearce KE, Smoley SA, et al. Clinical value of next generation sequencing in the detection of recurring structural rearrangements and copy number abnormalities in acute myeloid leukemia. Blood 2020;136(S1):21-2.
    CrossRef
  60. Hartmann L, Haferlach C, Meggendorfer M, Kern W, Haferlach T, Stengel A. Myeloid malignancies with isolated 7q deletion can be further characterized by their accompanying molecular mutations. Genes Chromosomes Cancer 2019;58:698-704.
    Pubmed CrossRef
  61. Haase D, Germing U, Schanz J, Pfeilstöcker M, Nösslinger 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.
    Pubmed CrossRef
  62. Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Solé F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood 2012;120:2454-65.
    Pubmed KoreaMed CrossRef
  63. Schanz J, Tüchler H, Solé F, Mallo M, Luno E, Cervera J, et al. New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. J Clin Oncol 2012;30:820-9.
    Pubmed KoreaMed CrossRef
  64. Kantarjian H, O'Brien S, Ravandi F, Borthakur G, Faderl S, Bueso-Ramos C, et al. The heterogeneous prognosis of patients with myelodysplastic syndrome and chromosome 5 abnormalities: how does it relate to the original lenalidomide experience in MDS?. Cancer 2009;115:5202-9.
    Pubmed KoreaMed CrossRef
  65. Garcia-Manero G, Chien KS, Montalban-Bravo G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am J Hematol 2020;95:1399-420.
    Pubmed CrossRef
  66. Bănescu C, Tripon F, Muntean C. The genetic landscape of myelodysplastic neoplasm progression to acute myeloid leukemia. Int J Mol Sci 2023;24:5734.
    Pubmed KoreaMed CrossRef
  67. Neuman WL, Rubin CM, Rios RB, Larson RA, Le Beau MM, Rowley JD, et al. Chromosomal loss and deletion are the most common mechanisms for loss of heterozygosity from chromosomes 5 and 7 in malignant myeloid disorders. Blood 1992;79:1501-10.
    Pubmed CrossRef
  68. Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, et al. Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature 2008;451:335-9.
    Pubmed KoreaMed CrossRef
  69. Dutt S, Narla A, Lin K, Mullally A, Abayasekara N, Megerdichian C, et al. Haploinsufficiency for ribosomal protein genes causes selective activation of p53 in human erythroid progenitor cells. Blood 2011;117:2567-76.
    Pubmed KoreaMed CrossRef
  70. Wei S, Chen X, McGraw K, Zhang L, Komrokji R, Clark J, et al. Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion. Oncogene 2013;32:1110-20.
    Pubmed KoreaMed CrossRef
  71. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood 2000;96:4075-83.
    Pubmed CrossRef
  72. Byrd JC, Mrózek K, Dodge RK, Carroll AJ, Edwards CG, Arthur DC, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 2002;100:4325-36.
    Pubmed CrossRef
  73. Grimwade D, Walker H, Harrison G, Oliver F, Chatters S, Harrison CJ, et al. The predictive value of hierarchical cytogenetic classification in older adults with acute myeloid leukemia (AML): analysis of 1065 patients entered into the United Kingdom Medical Research Council AML11 trial. Blood 2001;98:1312-20.
    Pubmed CrossRef
  74. Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G, et al. The importance of diagnostic cytogenetics on outcome in 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.
    Pubmed CrossRef
  75. Pedersen-Bjergaard J, Pedersen M, Roulston D, Philip P. Different genetic pathways in leukemogenesis for patients presenting with therapy-related myelodysplasia and therapy-related acute myeloid leukemia. Blood 1995;86:3542-52.
    Pubmed CrossRef
  76. Bhatia S. Therapy-related myelodysplasia and acute myeloid leukemia. Semin Oncol 2013;40:666-75.
    Pubmed KoreaMed CrossRef
  77. 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.
    Pubmed CrossRef
  78. Lindsley RC, Mar BG, Mazzola E, Grauman PV, Shareef S, Allen SL, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015;125:1367-76.
    Pubmed KoreaMed CrossRef
  79. Christiansen DH, Andersen MK, Pedersen-Bjergaard J. Mutations with loss of heterozygosity of p53 are common in therapy-related myelodysplasia and acute myeloid leukemia after exposure to alkylating agents and significantly associated with deletion or loss of 5q, a complex karyotype, and a poor prognosis. J Clin Oncol 2001;19:1405-13.
    Pubmed CrossRef
  80. Koenig KL, Sahasrabudhe KD, Sigmund AM, Bhatnagar B. AML with myelodysplasia-related changes: development, challenges, and treatment advances. Genes (Basel) 2020;11:845.
    Pubmed KoreaMed CrossRef
  81. Arber DA, Erba HP. Diagnosis and treatment of patients with acute myeloid leukemia with myelodysplasia-related changes (AML-MRC). Am J Clin Pathol 2020;154:731-41.
    Pubmed KoreaMed CrossRef
  82. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 2016;374:2209-21.
    Pubmed KoreaMed CrossRef
  83. Mrózek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol 2008;35:365-77.
    Pubmed KoreaMed CrossRef
  84. Mrózek K, Heinonen K, Theil KS, Bloomfield CD. Spectral karyotyping in patients with acute myeloid leukemia and a complex karyotype shows hidden aberrations, including recurrent overrepresentation of 21q, 11q, and 22q. Genes Chromosomes Cancer 2002;34:137-53.
    Pubmed CrossRef
  85. Schoch C, Haferlach T, Bursch S, Gerstner D, Schnittger S, Dugas M, et al. Loss of genetic material is more common than gain in acute myeloid leukemia with complex aberrant karyotype: a detailed analysis of 125 cases using conventional chromosome analysis and fluorescence in situ hybridization including 24-color FISH. Genes Chromosomes Cancer 2002;35:20-9.
    Pubmed CrossRef
  86. Van Limbergen H, Poppe B, Michaux L, Herens C, Brown J, Noens L, et al. Identification of cytogenetic subclasses and recurring chromosomal aberrations in AML and MDS with complex karyotypes using M-FISH. Genes Chromosomes Cancer 2002;33:60-72.
    Pubmed CrossRef
  87. Breems DA, Van Putten WL, De Greef GE, Van Zelderen-Bhola SL, Gerssen-Schoorl KB, Mellink CH, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol 2008;26:4791-7.
    Pubmed CrossRef
  88. Kayser S, Zucknick M, Döhner K, Krauter J, Köhne CH, Horst HA, et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood 2012;119:551-8.
    Pubmed CrossRef
  89. Rücker FG, Schlenk RF, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012;119:2114-21.
    Pubmed CrossRef
  90. Wong TN, Ramsingh G, Young AL, Miller CA, Touma W, Welch JS, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature 2015;518:552-5.
    Pubmed KoreaMed CrossRef
  91. Simmons HM, Oseth L, Nguyen P, O'Leary M, Conklin KF, Hirsch B. Cytogenetic and molecular heterogeneity of 7q36/12p13 rearrangements in childhood AML. Leukemia 2002;16:2408-16.
    Pubmed CrossRef
  92. Kalra R, Dale D, Freedman M, Bonilla MA, Weinblatt M, Ganser A, et al. Monosomy 7 and activating RAS mutations accompany malignant transformation in patients with congenital neutropenia. Blood 1995;86:4579-86.
    Pubmed CrossRef
  93. Pezeshki A, Podder S, Kamel R, Corey SJ. Monosomy 7/del (7q) in inherited bone marrow failure syndromes: a systematic review. Pediatr Blood Cancer 2017;64:10.1002/pbc.26714.
    Pubmed KoreaMed CrossRef
  94. Sahoo SS, Pastor VB, Goodings C, Voss RK, Kozyra EJ, Szvetnik A, et al. Clinical evolution, genetic landscape and trajectories of clonal hematopoiesis in SAMD9/SAMD9L syndromes. Nat Med 2021;27:1806-17.
    Pubmed KoreaMed CrossRef
  95. Grimwade D, Hills RK, Moorman AV, Walker H, Chatters S, Goldstone AH, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010;116:354-65.
    Pubmed CrossRef
  96. Pollyea DA, Altman JK, Assi R, Bixby D, Fathi AT, Foran JM, et al. Acute myeloid leukemia, version 3.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2023;21:503-13.
    Pubmed CrossRef
  97. Jentzsch M, Bischof L, Ussmann J, Backhaus D, Brauer D, Metzeler KH, et al. Prognostic impact of the AML ELN2022 risk classification in patients undergoing allogeneic stem cell transplantation. Blood Cancer J. 2022;12:170.
    Pubmed KoreaMed CrossRef