Abstract

In 1974, Van den Berghe et al described a distinct hematologic disorder associated with acquired, interstitial deletion of part of the long arm of chromosome 5. This condition is now classified as myelodysplastic syndrome (MDS) with isolated deletion 5q, or MDS-del(5q). The common deletion region 5q32-5q33 contains several genes and microRNAs whose expression levels are reduced in hematopoietic cells, consistent with the loss of 1 allele. Haploinsufficiency production of multiple gene transcripts, primarily involving CSNK1A1, RPS14, MIR145, and MIR146A, results in myelodysplastic hematopoiesis. Lenalidomide can selectively suppress the del(5q)-mutant clone by promoting proteasomal degradation of casein kinase 1A1 and inducing mutant stem cell failure. However, lenalidomide is not a curative treatment, as almost all patients relapse. Molecular profiling studies have significantly improved our understanding of MDS-del(5q). Only a minority of patients have interstitial deletion 5q as their sole genetic lesion, a condition that is associated with an indolent clinical course. Most patients have co-occurring somatic mutations in myeloid genes, including DNMT3A, TET2, ASXL1, SF3B1, TP53, RUNX1, and CSNK1A1. These comutations have independent effects on leukemic transformation and survival, so genomic profiling is required for implementing a precision management approach to MDS-del(5q) in a clinical setting. Accurate assessment of the TP53 allelic state is crucial for distinguishing MDS-del(5q) from TP53-mutant MDS, a myeloid malignancy characterized by TP53 multihit state and very aggressive clinical course. Genomic profiling is also critical for therapeutic decision-making in patients with MDS-del(5q), particularly for assessing a patient’s eligibility for allogeneic transplantation, which remains the only curative treatment.

In a letter to Nature in 1974, Van den Berghe et al reported on 3 patients who presented with a clinically similar hematologic disorder and an apparently identical chromosomal abnormality in the bone marrow.1 These patients had long-standing macrocytic anemia that was refractory to conventional treatments, a low or normal white blood cell count, and a normal or elevated platelet count. Their bone marrow showed dysplastic features, particularly the absence of megakaryocytic nuclei lobulation. The chromosomal abnormality consisted of a deletion of the long arm of 1 chromosome 5, without translocation of the deleted segment. This aberration was detected in cultured bone marrow cells but not in lymphocytes or skin fibroblasts, indicating that it was a somatically acquired genetic lesion.1 Additional investigations on the new hematologic syndrome associated with the 5q− chromosome were reported 1 year later by the same group in Blood.2 

For many years, the hematologic disorder associated with the 5q chromosomal abnormality has been designated the 5q− syndrome.3 In the 2001 World Health Organization (WHO) classification of myeloid neoplasms, this disorder was included as a specific myelodysplastic syndrome (MDS) subtype, termed MDS with isolated deletion 5q, or MDS-del(5q).4 As illustrated in Table 1, the diagnostic criteria for this entity have been subsequently refined, specifically in both the International Consensus Classification (ICC) and the 5th Edition of the WHO classification (WHO 2022) of myeloid neoplasms and acute leukemias.5-8 This report illustrates how genomic profiling of well-characterized populations of patients with MDS has transformed our understanding of MDS-del(5q), whose management now requires a precision-medicine approach.

The deletion of the long arm of chromosome 5 or the loss of the whole chromosome 5 are common occurrences in myeloid neoplasms, including MDS, acute myeloid leukemia (AML), and therapy-related myeloid neoplasms (t-MNs).9 In many of these conditions, del(5q) is typically accompanied by additional chromosomal abnormalities, frequently resulting in a complex karyotype (CK). Conversely, in most patients with the condition identified by Van den Berghe et al, del(5q) is typically the sole identified cytogenetic abnormality, which indicates that it likely represents a founding genetic lesion (Figure 1A).1,2,10 A few patients may exhibit 1 additional chromosomal abnormality in conjunction with del(5q), without experiencing a significant impact on clinical outcomes.10 In contrast, those carrying ≥2 additional chromosomal abnormalities (representing a CK) or an accompanying −7 or del(7q) cytogenetic abnormality have worse survival.10,11 These observations led to the conclusion that a diagnosis of MDS-del(5q) can be made if there is 1 additional cytogenetic abnormality beyond the del(5q), except monosomy 7 or del(7q) , which was reflected in the revised 4th edition of the WHO classification published in 2016 (Table 1).5 

Figure 1.

Interstitial deletion of the long arm of 1 chromosome 5 and molecular pathogenesis of the MDS-del(5q). (A) The karyogram (R-banding) of a patient with MDS-del(5q): the red arrow indicates the chromosome 5 with an interstitial deletion of the q arm involving bands q31 to q34. (B) The role of haploinsufficiency of multiple genes, located in the common deletion region 5q32-5q33, in the pathophysiology of MDS-del(5q). The karyogram is from the UZ Leuven (Belgium), the institution in which Herman Van den Berghe worked for many years. The microphotographs of the peripheral blood and bone marrow are from the Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia (Italy).

Figure 1.

Interstitial deletion of the long arm of 1 chromosome 5 and molecular pathogenesis of the MDS-del(5q). (A) The karyogram (R-banding) of a patient with MDS-del(5q): the red arrow indicates the chromosome 5 with an interstitial deletion of the q arm involving bands q31 to q34. (B) The role of haploinsufficiency of multiple genes, located in the common deletion region 5q32-5q33, in the pathophysiology of MDS-del(5q). The karyogram is from the UZ Leuven (Belgium), the institution in which Herman Van den Berghe worked for many years. The microphotographs of the peripheral blood and bone marrow are from the Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia (Italy).

Close modal

The somatic deletion of MDS-del(5q) is interstitial (supplemental Figure 1, available on the Blood website).3 The prevalent chromosomal alteration is a 5q32-5q33 deletion, with the commonly deleted region (CDR) spanning ∼1.5 megabases and containing 44 genes and several microRNA (miRNA) genes, as described by Boultwood et al.12,13 The 1- to 1.5-megabase CDR at 5q31 identified by Lai et al in patients with AML or more advanced forms of MDS, many of whom have additional chromosomal abnormalities, is distinct from the CDR of MDS-del(5q) because it is more proximal.14,15 

Gene expression analysis of CD34+ cells from patients with MDS-del(5q) unequivocally demonstrated that most genes assigned to the CDR at 5q32-5q33 had reduced expression levels, consistent with the loss of 1 allele.16 This suggested that the pathogenesis of MDS-del(5q) is due to the haploinsufficiency of multiple genes located within the deleted region, a working hypothesis that has been confirmed by subsequent studies and is schematically depicted in Figure 1B.

Mechanisms of anemia

The RPS14 gene maps to chromosome 5q33.1 and encodes a ribosomal protein that is a component of the 40S ribosomal subunit. The reduced expression of this gene was documented in hematopoietic cells from patients with MDS-del(5q).16 Ebert et al then demonstrated that haploinsufficiency of RPS14 is the underlying cause of the erythroid dysfunction.17 RPS14 protein deficiency interferes with the normal formation of the 40S ribosomal subunit, thereby impairing erythroid differentiation. The mechanism is analogous to that of Diamond-Blackfan anemia, an inherited autosomal dominant disorder associated with germ line mutations in ribosomal protein genes, such as RPS19.18 

In both patients with MDS-del(5q) and animal models of the disease, the defective formation of the 40S ribosomal subunit leads to upregulation of the p53 pathway in erythroid cells.19-22 A mouse model of the 5q− syndrome was generated by Barlow et al using large-scale chromosomal engineering.19 Mice with haploinsufficiency of the Cd74-Nid67 interval (syntenic to the CDR of the human 5q− syndrome and including Rps14) recapitulated the key features of the human disease, including macrocytic anemia and megakaryocytes with round or nonlobulated nuclei. This mouse model displayed defective bone marrow progenitor development and an accumulation of p53 protein with increased apoptosis in bone marrow cells. The progenitor cell defect was rescued by intercrossing the “5q− mouse” with p53-deficient mice, providing evidence that a p53-dependent mechanism plays a role in the pathophysiology of the 5q− syndrome.19 

In a murine model with conditional inactivation of Rps14, ribosomal haploinsufficiency was found to induce overexpression of p53 and a related impairment in erythroid differentiation, characterized by defective generation of orthochromatic erythroblasts.22 Patients with MDS-del(5q) typically exhibit relative erythroid hypoplasia, likely because the defective ribosome biogenesis limits erythroid proliferation.23 

Clonal proliferation of del(5q)-mutant hematopoietic cells

The pathophysiologic process that leads to MDS consists of the growth and expansion of a mutated clone, whose selective advantage is provided by a somatic genetic lesion occurring in a self-renewing hematopoietic stem cell.8 In MDS-del(5q), the clonal advantage of mutant hematopoietic cells originates from the haploinsufficiency of ≥1 genes that are located in the deleted region.

The tumor suppressor gene CSNK1A1, which encodes casein kinase 1A1, exhibits a reduction in gene expression of ∼50% in hematopoietic cells from patients with MDS-del(5q).16 In a murine model, Csnk1a1 haploinsufficiency was found to induce hematopoietic stem cell expansion and a competitive repopulation advantage.24 Additionally, recurrent somatic mutations in the nondeleted CSNK1A1 allele were detected in patients with MDS-del(5q).24-26 Collectively, these observations indicate that CSNK1A1 haploinsufficiency leads to clonal expansion of del(5q)-mutant hematopoietic cells (Figure 1B). This conclusion has been recently corroborated by studies of genetic barcoding in murine models.27 

Dysmegakaryopoiesis and increased platelet production

In the report by Sokal et al, the striking morphologic abnormality concerned megakaryocytes, whose nuclei “were generally small, round or oval, and nonlobulated”; additionally, affected patients showed “normal to elevated platelets (300 000 to 500 000).”2 

These morphologic and clinical findings likely reflect, at least in part, the dysregulated miRNA activity caused by the 5q deletion. MiRNAs are noncoding RNAs that regulate biologic processes by silencing gene expression posttranscriptionally.28 Their haploinsufficiency is therefore expected to increase the expression of targeted genes. Starczynowski et al observed that deletion of chromosome 5q32-5q33 is associated with the loss of 2 miRNAs, MIR145 and MIR146A.29 Using animal models, they demonstrated that the loss of these miRNAs in hematopoietic cells results in activation of innate immune signaling, dysmegakaryopoiesis, and thrombocytosis.29 Similarly, Kumar et al showed that the combined loss of MIR145 and RPS14 in murine models leads to alterations of the megakaryocytic differentiation that are analogous to those of patients with MDS-del(5q).30 

Hematopoietic cell of origin of MDS-del(5q)

In a study of patients with lower-risk MDS, including MDS-del(5q), Woll et al investigated whether the underlying genetic lesion occurred in hematopoietic stem cells capable of self-renewal or more differentiated progenitor cells.31 Collectively, the findings of this study indicate that MDS is propagated by rare and distinct cancer stem cells capable of self-renewal.31 In a portion of patients with MDS-del(5q), no recurrent driver mutation could be identified through targeted or exome sequencing, confirming that del(5q) represented the initiating genetic lesion. At variance, in patients with MDS with del(5q) associated with CK and advanced disease, del(5q) was a secondary hit, often preceded by TP53 mutation.32 

Lenalidomide is currently used as a targeted therapy for patients with MDS-del(5q), capable of suppressing the mutant clone and restoring normal hematopoiesis.8 However, its initial development was for a more limited purpose, namely the amelioration of anemia that had been previously observed with thalidomide in patients with MDS.33 

Clinical studies that demonstrated the efficacy of lenalidomide in transfusion-dependent patients with lower-risk MDS-del(5q)

The first clinical trial on the use of this thalidomide analogue in patients with MDS showed cytogenetic responses in patients with del(5q).34 Therefore, a subsequent phase 2 trial investigated whether lenalidomide could reduce the transfusion requirement by selectively targeting the mutant clone in patients with MDS-del(5q).35 Patients were risk-stratified using the International Prognostic Scoring System (IPSS) and 94% of patients had lower-risk MDS.36 This study demonstrated that lenalidomide can indeed target the del(5q) clone, eliminating or reducing the need for transfusions. A phase 3 randomized trial subsequently established that lenalidomide treatment (5 or 10 mg per day) can achieve transfusion independence in ∼60% of patients with MDS-del(5q) (supplemental Table 1).37 However, less than one-third of patients achieved complete cytogenetic remission, and after a median duration of response of ∼2 years, most patients experienced reappearance or reexpansion of the del(5q) clone and recurrence of transfusion dependence.8,38 Additionally, lenalidomide treatment involved grade 3 to 4 adverse effects in most patients, primarily severe neutropenia (supplemental Table 1). Ultimately, there was no statistically significant difference in the median overall survival rates between the placebo group and the 2 lenalidomide groups.

Real-world studies on the use of lenalidomide in patients with (MDS)-del(5q) have yielded worse results. In a Mayo Clinic study, the proportion of patients showing complete cytogenetic remission was very low (12%), whereas most individuals (65%) eventually discontinued treatment.39 Of 118 patients enrolled in the HARMONY Alliance study, 70 (59%) had discontinued lenalidomide because of intolerance.40 

Lenalidomide in patients who are not transfusion dependent

The SintraREV trial aimed to investigate whether an early intervention of low doses of lenalidomide (5 mg per day) for 2 years could delay transfusion dependency in patients with anemia with MDS-del(5q) who were not already transfusion dependent.41 Patients were risk-stratified using the revised IPSS (IPSS-R), and all but 1 had very low or low IPSS-R risk.42 The study was conducted at 22 centers and enrolled 61 patients over 8 years; 40 patients were assigned to receive lenalidomide, and 21 were given placebo. Lenalidomide treatment was found to delay the time to transfusion dependency but had no impact on overall survival. Complete cytogenetic response was observed in 28 of 40 patients receiving lenalidomide. Approximately 50% of patients discontinued the study medication prematurely during the study period; as would have been anticipated, a significantly greater number of treatment-related adverse events were observed in patients receiving lenalidomide.

The mechanism of action of lenalidomide in MDS-del(5q)

In vitro studies clearly showed that lenalidomide had selective cytotoxic effects on del(5q) hematopoietic progenitor cells, resulting, in part, from inhibition of the haplodeficient Cdc25C and PP2Acα phosphatases.43 The major mechanism through which lenalidomide suppresses the del(5q) clone has been elucidated more recently. This compound promotes ubiquitination and proteasomal degradation of casein kinase 1A1, inducing stem cell failure.24,44 In patients with MDS-del(5q), hematopoietic cells harboring del(5q) have a single copy of the CSNK1A1 gene and reduced production of casein kinase 1A1, which renders these cells more sensitive to the effects of lenalidomide compared with normal cells.44 

Despite lenalidomide targeting mutant hematopoietic cells, this is not a curative treatment.38 Almost all patients who achieve complete or partial cytogenetic response subsequently experience recurrence or expansion of the del(5q) clone, and relapsing patients are at elevated risk of disease progression and leukemic transformation.

Phenotypically distinct cancer stem cells that are resistant to lenalidomide

In light of the preceding observation that MDS-del(5q) is propagated by rare cancer stem cells, Tehranchi et al tested the hypothesis that these stem cells might be selectively resistant to lenalidomide treatment.31,45 They identified a subset of mutant stem cells (phenotypically defined as CD34+, CD38−/low, CD90+) that persisted under treatment and subsequently expanded at the time of relapse.

Co-occurring somatic mutations may be associated with poor response to lenalidomide and disease progression

In a study of 55 patients with lower-risk MDS-del(5q), Jadersten et al detected somatic mutations of TP53 in approximately a fifth of cases.46 These mutations were associated with poor response to lenalidomide, lower probability of complete cytogenetic response, and an increased risk of leukemic transformation. This initial observation has been corroborated by several subsequent reports.47-52 Additional studies indicated that the presence of somatic mutations in CSNK1A1 or RUNX1 also predicted a worse prognosis for patients with MDS-del(5q) treated with lenalidomide.26,53 

Exposure to lenalidomide and development of therapy-related myeloid neoplasms

To elucidate the molecular pathogenesis of t-MNs, Sperling et al analyzed the association between genotype and previous exposure to chemotherapy and/or radiotherapy in a cohort of 416 patients with t-MNs.54 Approximately two-thirds of the patients had a primary diagnosis of solid tumors, whereas the remaining third had nonmyeloid hematologic cancers. A somatic mutation in TP53 was identified in 37% of patients, and most cases exhibited a TP53 multihit state. A significant association was established between TP53-mutant t-MNs and previous exposure to lenalidomide. Using mouse models, the authors subsequently demonstrated that lenalidomide treatment provides a selective advantage to Trp53-mutant hematopoietic stem and progenitor cells in vitro and in vivo.54 

TP53 allelic state and outcomes of lenalidomide treatment in patients with MDS-del(5q)

The observation that lenalidomide treatment confers a selective advantage to TP53-mutant hematopoietic cells is relevant to the potential risks posed by lenalidomide treatment of patients diagnosed with MDS-del(5q), because some of these patients may carry a TP53 mutation.55,56 

Montoro et al retrospectively investigated the clinical implications of TP53 mutations and their allelic state in a large cohort of patients with MDS-del(5q).57 They adopted the 2016 WHO definition of MDS-del(5q), which did not exclude patients with TP53 multihit state.5 Of the 682 patients examined, 129 (19%) carried a somatic mutation in TP53: 98 patients had monoallelic TP53 mutation and 31 had TP53 multihit state.57 Most of these patients received lenalidomide treatment at some point during their disease. The presence of either TP53 multihit state or monoallelic mutation with a variant allele frequency (VAF) of >20% was associated with inferior survival and higher risk of leukemic transformation.57 

Over the past 15 years, the application of massively parallel DNA sequencing has elucidated the genetic basis of MDS, thereby enabling the development of innovative diagnostic and prognostic methods.58 The International Working Group for the Prognosis of MDS (IWG-PM) has made significant contributions to this field.55,56,59 We have now conducted computational analyses using the database that was created for the IWG-PM studies. The findings of these analyses definitively support a precision medicine approach to the management of patients with MDS-del(5q), as outlined hereafter.

The molecular taxonomy of MDS and the del(5q) subtype

The recent IWG-PM study was conducted to investigate whether genomic alterations allow for the delineation of specific MDS molecular subtypes (supplemental Methods).56 Eighteen distinct groups of MDS were identified and 1 of them was the del(5q) group (supplemental Figure 2).55,56 

Within the IWG-PM cohort, 388 patients with MDS carried a deletion 5q as a chromosomal abnormality. Supplemental Figure 3 reports an oncoplot describing the gene mutations in these patients, with TP53 mutations observed in 54% of cases. By using a Sankey diagram, we investigated how these patients were classified according to the last 3 classifications of MDS (WHO 2016, WHO 2022, and ICC 2022) and the recent molecular taxonomy.5-7,56 As illustrated in supplemental Figure 4, most patients with MDS with a deletion 5q were ultimately included in the TP53 multihit/CK group or the del(5q) group of the molecular taxonomy. As illustrated in Figure 2, these 2 subsets were associated with significantly different clinical outcomes, consistent with substantially different disease biology.

Figure 2.

Kaplan-Meier analysis of overall survival and leukemia-free survival of patients with MDS with a deletion 5q as a chromosomal abnormality. Patients are subdivided into 2 groups according to the molecular taxonomy of MDS illustrated in supplemental Figure 2: the TP53-complex group (defined by the presence of TP53 multihit state and/or CK) or the del(5q) group (defined by the presence of del(5q) as the sole cytogenetic abnormality or with 1 additional abnormality excluding −7/7q, and by the absence of TP53 multihit state). Patients belonging to the TP53-complex group had significantly worse overall survival and higher risk of leukemic transformation, consistent with a substantially different disease biology. These observations substantiate the current diagnostic criteria for MDS-del(5q), which exclude the presence of TP53 multihit state and CK.6,7 P values are from the log-rank test.

Figure 2.

Kaplan-Meier analysis of overall survival and leukemia-free survival of patients with MDS with a deletion 5q as a chromosomal abnormality. Patients are subdivided into 2 groups according to the molecular taxonomy of MDS illustrated in supplemental Figure 2: the TP53-complex group (defined by the presence of TP53 multihit state and/or CK) or the del(5q) group (defined by the presence of del(5q) as the sole cytogenetic abnormality or with 1 additional abnormality excluding −7/7q, and by the absence of TP53 multihit state). Patients belonging to the TP53-complex group had significantly worse overall survival and higher risk of leukemic transformation, consistent with a substantially different disease biology. These observations substantiate the current diagnostic criteria for MDS-del(5q), which exclude the presence of TP53 multihit state and CK.6,7 P values are from the log-rank test.

Close modal

Approximately one-fifth of patients in the del(5q) group of the molecular taxonomy had excess blasts (≥5% in the bone marrow). As shown in supplemental Figure 5, patients with excess blasts had worse clinical outcomes compared with those with a blast count of <5%. This observation supports excluding cases with excess blasts from the MDS-del(5q) category in the current ICC and WHO classifications.6,7 However, the molecular taxonomy of MDS suggests a novel paradigm in which genetic subtypes represent the overarching basis of classification, whereas blast count designates a disease stage within specific molecular groups.56 Table 2 illustrates this concept by distinguishing between the initial and advanced stages of MDS-del(5q). The initial stage is characterized by a blast count of <5%, whereas the advanced stage is characterized by a blast count ranging from 5% to 19%. Of note, the phase 2 clinical trial on the use of lenalidomide in MDS-del(5q) included patients with excess blasts, and most of them responded to treatment, which resulted in the normalization of their blast count.34 

Genomic landscape of MDS-del(5q) and its clinical implications

As illustrated in supplemental Figure 6, 85% of patients with MDS belonging to the del(5q) group of the molecular taxonomy of MDS had at least 1 co-occurring somatic mutation, most commonly in DTA (DNMT3A, TET2, or ASXL1) genes, SF3B1, TP53 (monoallelic mutation), CSNK1A1, and RUNX1. Monoallelic TP53 mutations were significantly enriched in the del(5q) group, along with mutations in CSNK1A1, IRF1, and RAD50, the latter 3 genes being all located on 5q.56 

Of 195 patients classified in the del(5q) group of the molecular taxonomy of MDS, 29 did not have any other chromosomal aberrations or somatic gene mutations. Thus, they met the criteria for MDS with truly isolated del(5q), a condition in which del(5q) is the sole identified genetic lesion. Figure 3A shows that these patients had significantly better overall survival and lower risk of leukemic transformation than those with additional somatic genetic lesions. These differences in clinical outcomes persisted, although less notably, when the comparison was limited to patients with co-occurring DTA mutations, which are likely an expression of age-related clonal hematopoiesis (supplemental Figure 7).

Figure 3.

Kaplan-Meier analysis of overall survival and leukemia-free survival in patients with MDS belonging to the del(5q) group of the molecular taxonomy, subdivided into distinct subsets. (A) Patients with truly isolated del(5q) (as indicated by the absence of any additional chromosomal abnormality or somatic gene mutation) vs patients with at least 1 co-occurring genetic lesion. (B) Patients with truly isolated del(5q) vs those with an SF3B1 comutation. (C) Patients with truly isolated del(5q) vs those with RUNX1 comutation. P values are from the log-rank test.

Figure 3.

Kaplan-Meier analysis of overall survival and leukemia-free survival in patients with MDS belonging to the del(5q) group of the molecular taxonomy, subdivided into distinct subsets. (A) Patients with truly isolated del(5q) (as indicated by the absence of any additional chromosomal abnormality or somatic gene mutation) vs patients with at least 1 co-occurring genetic lesion. (B) Patients with truly isolated del(5q) vs those with an SF3B1 comutation. (C) Patients with truly isolated del(5q) vs those with RUNX1 comutation. P values are from the log-rank test.

Close modal

As illustrated in Figure 3B, within the del(5q) group, patients carrying a concomitant mutation in SF3B1 exhibited significantly worse clinical outcomes than those lacking an SF3B1 mutation. The deleterious clinical consequences of this comutation have been previously reported in patients diagnosed with MDS-del(5q).60 As SF3B1 mutations are the most frequent co-occurring mutations in MDS-del5q, identified in approximately one-fifth of patients (supplemental Figure 6), it is important to include this information as a molecular classifier within the diagnostic criteria (Table 2). A co-occurring mutation in RUNX1 was detected in only 6% of patients classified within the del(5q) molecular group but this concomitant genetic lesion was associated with very poor overall survival and very high risk of leukemic transformation (Figure 3C).

By contrast, there was no significant relationship between a monoallelic mutation in TP53, found in 17% of patients, and clinical outcomes (supplemental Figure 8). In the Mayo-Moffitt study, TP53 mutations occurred in ∼15% of patients with chronic phase disease and their prevalence increased significantly at the time of leukemic transformation.50,51 In the recent study by Montoro et al, the overall survival of patients with MDS del(5q) patients with co-occurring monoallelic TP53 mutation (14% of the entire cohort) did not differ from that of patients without TP53 mutation.57 Zampini et al performed longitudinal studies on patients with MDS who carried a TP53 mutation at diagnosis and then progressed to AML.61 Among 23 patients with the monoallelic mutation, 18 progressed to a multihit TP53 state before leukemic evolution.

Taken together, the above data indicate that comutation patterns may have different implications for clinical outcomes and treatment. Co-occurring mutations in SF3B1 or RUNX1 are associated with a more aggressive clinical course. In contrast, monoallelic TP53 mutations do not inherently affect clinical outcomes; however, they may evolve into TP53 multihit lesions under lenalidomide treatment, thereby leading to a t-MN.

As a group, patients diagnosed with MDS-del(5q) have superior overall survival when compared with the entire population of patients with MDS.56 Nevertheless, considerable variability exists in clinical outcomes, which are influenced by several factors, including co-occurring genetic lesions. Thus, 5 decades after its initial identification, management of MDS-del(5q) now requires a precision medicine approach, as illustrated in Figures 4 and 5.

Figure 4.

A precision medicine approach to the diagnosis and prognosis of the MDS-del(5q). The conventional diagnostic workup includes bone marrow aspiration, bone marrow biopsy, and conventional cytogenetic analysis. Genomic profiling is now required for precision diagnosis and prognosis. Images are, in part, from Servier Medical Art (https://smart.servier.com).

Figure 4.

A precision medicine approach to the diagnosis and prognosis of the MDS-del(5q). The conventional diagnostic workup includes bone marrow aspiration, bone marrow biopsy, and conventional cytogenetic analysis. Genomic profiling is now required for precision diagnosis and prognosis. Images are, in part, from Servier Medical Art (https://smart.servier.com).

Close modal
Figure 5.

A precision approach to the treatment of patients with MDS-del(5q). Patients with lower-risk MDS-del(5q) have a median overall survival of ∼5 years, whereas those with higher-risk MDS-del(5q) have a median overall survival of only 2 years. The use of lenalidomide in a patient with a monoallelic TP53 mutation should be carefully discussed with the patient, given the risk of progression to a t-MN. The flowchart does not include patients with MDS with TP53 multihit state; these individuals have TP53-mutant MDS, not MDS-del(5q), as illustrated in Figure 4. Older adult patients with comorbidities and/or frailty may benefit from best supportive care alone, which would represent the initial therapeutic decision, regardless of the IPSS-M. ESAs, erythropoiesis stimulating agents; RBC, red blood cell.

Figure 5.

A precision approach to the treatment of patients with MDS-del(5q). Patients with lower-risk MDS-del(5q) have a median overall survival of ∼5 years, whereas those with higher-risk MDS-del(5q) have a median overall survival of only 2 years. The use of lenalidomide in a patient with a monoallelic TP53 mutation should be carefully discussed with the patient, given the risk of progression to a t-MN. The flowchart does not include patients with MDS with TP53 multihit state; these individuals have TP53-mutant MDS, not MDS-del(5q), as illustrated in Figure 4. Older adult patients with comorbidities and/or frailty may benefit from best supportive care alone, which would represent the initial therapeutic decision, regardless of the IPSS-M. ESAs, erythropoiesis stimulating agents; RBC, red blood cell.

Close modal

Diagnostic/prognostic approach

The diagnostic process is initiated when a patient presents with a cytopenia of unknown origin, typically a macrocytic anemia.62 The identification of megakaryocytes with hypolobulated nuclei is an important diagnostic criterion, whereas conventional chromosome banding cytogenetics is the standard procedure for detecting del(5q) (Figure 1A). However, in a minority of patients, the interstitial deletion remains undetectable using this approach. An ad hoc study revealed that fluorescence in situ hybridization (FISH) of 5q31 can detect the 5q deletion in patients in whom the cytogenetic study fails because of a lack of metaphases, or who display an aberrant karyotype with chromosome 5 involvement that is not clearly del(5q).63 This indicates that FISH of 5q31-5q33 should be performed in patients with suggestive dysmegakaryopoiesis and apparently normal chromosome banding analysis, using a FISH probe for a gene mapping specifically to the MDS-del(5q) CDR, such as CSF1R or SPARC.13,64 In addition, single-nucleotide polymorphism array–based karyotyping can reveal unbalanced defects with high resolution and can also identify segmental uniparental disomy.65 Finally, next-generation sequencing–based methodologies can be effective tools for the detection and mapping of the 5q deletion.32,66 

Genomic profiling is now essential for the precision prognosis of MDS-del(5q).66 Targeted gene panels should encompass a minimum of the 31 genes necessary for the calculation of the molecular IPSS (IPSS-M), and include also CSNK1A1. Genomic profiling approaches should include probes for copy number detection to capture chromosomal abnormalities, especially loss of heterozygosity, which can derive from chromosomal deletion or copy-neutral loss of heterozygosity.66 This is particularly important for detecting TP53 multihit state, because TP53 multihit lesions are associated with genomic instability and high-risk disease, and now exclude MDS-del(5q) whereas defining a specific subtype of MDS, that is, TP53-mutant MDS.55,66,67 Interestingly, single-nucleotide polymorphism array–based copy number detection may allow the identification of small TP53 biallelic clones with a VAF of <1%.68 

Diagnostic criteria for MDS-del(5q)

Current classifications of hematologic neoplasms are based on a combination of clinical, morphologic, and genomic data.69 MDS-del(5q) is defined by cytopenia, presence of del(5q) alone or with 1 additional abnormality except monosomy 7 or del(7q), and absence of TP53 multihit lesions or excess blasts (Table 1). However, studies of genomic profiling have indicated that most patients carry additional genetic lesions that can variably affect clinical outcomes. So, we propose using blast count as a disease stage indicator and concomitant genetic lesions as molecular qualifiers to the MDS-del(5q) entity, as illustrated in Table 2 and Figure 4.

Risk stratification through the IPSS-M

For patients belonging to the del(5q) group of the molecular taxonomy of MDS, the IPSS-M provides valuable risk stratification as illustrated in the supplemental Figure 9. All patients with higher-risk MDS-del(5q), as defined by an IPSS-M score of >0, may be considered for allogeneic transplantation. To determine their eligibility, additional factors must be considered, including patient-related risk factors, donor availability, and the patient's values and wishes.70 

Therapeutic options and disease monitoring

The adoption of a patient-centered care model that leverages precision medicine criteria constitutes the optimal approach for the treatment of patients with MDS-del(5q).8 Therapeutic options include active surveillance, a medical treatment, and allogeneic transplantation; currently available medical treatments are summarized in Table 3. This table illustrates the complexity inherent to the approval processes by different regulatory agencies, namely, the US Food and Drug Administration and the European Medicines Agency, over time.71-73 

Therapeutic decision-making should be based on the IPSS-M risk, as illustrated in Figure 5. Patients with lower-risk MDS-del(5q) are initially appropriate candidates for follow-up care or medical treatments. In contrast, those with higher-risk MDS-del(5q) should be considered for allogeneic transplantation.

Active surveillance with deferred treatment is the optimal approach for older adult patients with lower-risk MDS-del(5q) who do not require regular red blood cell transfusions. Erythropoiesis stimulating agents may be used in patients with anemia with serum erythropoietin levels below 200 mU/mL.8 At present, no evidence-based recommendations can be made regarding the use of luspatercept, as registration trials specifically excluded patients with MDS-del(5q) (Table 3).

The use of lenalidomide should be limited to patients with symptomatic anemia who require regular red blood cell transfusions, (Table 3). Before deciding on using this treatment, information on TP53 mutation status is needed. A co-occurring monoallelic TP53 mutation with high VAF (ie, >20%) involves a higher risk of leukemic transformation under lenalidomide treatment, possibly because these patients may already carry small TP53 biallelic clones.57,68 This risk must be discussed carefully with the patient in a process that takes full account of his/her values and wishes. If lenalidomide treatment is initiated, genomic profiling should be used to closely monitor the TP53-mutant clone.49 

For patients with higher-risk MDS-del(5q), allogeneic hematopoietic cell transplantation is currently the sole treatment that has the potential to cure the disease by eradicating del(5q)-mutant hematopoietic cells and establishing normal donor hematopoiesis.70,74,75 Figure 6 and supplemental Table 2 illustrate the clinical outcomes of patients with MDS-del(5q) reported to the European Society for Blood and Marrow Transplantation registry between 2016 and 2020. The estimated overall survival rate is ∼50% at 4 years. For patients with higher-risk MDS-del(5q) who are not eligible for transplantation, treatment with a hypomethylating agent should be considered.76 

Figure 6.

European Society for Blood and Marrow Transplantation data on allogeneic transplantation in patients with MDS-del(5q). A total of 124 patients received transplantation between 2016 and 2020. The 4-year estimated overall survival was 50% (95% confidence interval [CI], 39-60). The 4-year cumulative incidence of relapse was 25% (95% CI, 16-33), whereas that of nonrelapse mortality was 28% (95% CI, 19-36). See also supplemental Table 2. Pts, patients.

Figure 6.

European Society for Blood and Marrow Transplantation data on allogeneic transplantation in patients with MDS-del(5q). A total of 124 patients received transplantation between 2016 and 2020. The 4-year estimated overall survival was 50% (95% confidence interval [CI], 39-60). The 4-year cumulative incidence of relapse was 25% (95% CI, 16-33), whereas that of nonrelapse mortality was 28% (95% CI, 19-36). See also supplemental Table 2. Pts, patients.

Close modal

In younger patients with MDS-del(5q), the patient's eligibility for allogeneic stem cell transplantation should be assessed at the time of diagnosis following the recommendations of the European Society for Blood and Marrow Transplantation.70 It is essential to consider disease-related and patient-related risk factors, donor availability, and the patient's choice based on personal goals. Once eligibility for transplantation has been established, the long-term therapeutic program can be defined. Supplemental Figure 10 illustrates a patient’s journey through MDS-del(5q), ending with a successful allogeneic transplantation from an unrelated donor.

Fifty years after the seminal paper by Van den Berghe et al based on chromosome banding analysis, molecular profiling in combination with conventional cytogenetics is now required for a precision medicine approach to MDS-del(5q).

Assessment of the TP53 allelic state is essential for distinguishing between MDS-del(5q) and TP53-mutant MDS, an aggressive myeloid malignancy characterized by TP53 multihit state. The IPSS-M risk assessment stratifies patients into lower-risk and higher-risk categories, enabling precise decisions regarding their eligibility for allogeneic transplantation and the optimal timing of this procedure. Clinical trials in patients with MDS-del(5q) should now incorporate genomic profiling into the trial design to elucidate the effects of investigational therapies on the clonal architecture of MDS-del(5q).67 Specifically, genomic profiling can document the suppression of mutant clones with the restoration of normal hematopoiesis, as well as the emergence or expansion of abnormal clones under selective pressure.

In conclusion, this work illustrates how integrating morphologic, clinical, and genomic data of well-characterized patient populations can lead to the identification of distinct entities associated with different clinical outcomes that warrant specific treatments.69 This is the next logical step in the process that Van den Berghe et al started by linking macrocytic anemia to the unusual morphology of megakaryocytes and a chromosomal abnormality in a study of 3 patients.1 

This special report is the result of a collaborative effort among investigators from the International Working Group for the Prognosis of MDS (IWG-PM), the European Society for Blood and Marrow Transplantation (EBMT), and MYNERVA (Myeloid Neoplasms Research Venture Associazione Italiana per la Ricerca sul Cancro [AIRC]. The IWG-PM consists of a group of international investigators whose focus is aimed at defining the clinical and biologic features of MDS. The Chronic Malignancies Working Party of the EBMT promotes clinical investigations on the role of hematopoietic cell transplantation in chronic malignancies. MYNERVA is a consortium of Italian investigators who, thanks to the financial support of the AIRC (Milan, Italy), have teamed up for a research initiative focused on myeloid neoplasms.

We dedicate this work to the memory of Herman Van den Berghe. After the description of a minute chromosome 22 in chronic myeloid leukemia by Nowell and Hungerford,77 the short letter by Van den Berghe et al1 represents the second cornerstone indicating that myeloid malignancies arise from somatic mutations of the genome.

Contribution: M.R., L. Malcovati, and M.C. conceptualized this special report, thoroughly reviewed the extant literature, performed statistical analyses of data available in the International Working Group for the Prognosis of MDS database, and drafted the manuscript; and all authors contributed to the design of the work and the interpretation of the data, reviewed the manuscript critically for intellectual content, and approved the final version to be published.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Mario Cazzola, Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy; email: mario.cazzola@unipv.it.

1.
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
(
5474
):
437
-
438
.
2.
Sokal
G
,
Michaux
JL
,
Van Den Berghe
H
, et al
.
A new hematologic syndrome with a distinct karyotype: the 5 q--chromosome
.
Blood
.
1975
;
46
(
4
):
519
-
533
.
3.
Boultwood
J
,
Lewis
S
,
Wainscoat
JS
.
The 5q-syndrome
.
Blood
.
1994
;
84
(
10
):
3253
-
3260
.
4.
Jaffe
ES
,
Harris
NL
,
Stein
H
,
Vardiman
J
. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues. 3rd ed.
IARC
;
2001
.
5.
Arber
DA
,
Orazi
A
,
Hasserjian
R
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood
.
2016
;
127
(
20
):
2391
-
2405
.
6.
Arber
DA
,
Orazi
A
,
Hasserjian
RP
, et al
.
International Consensus Classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data
.
Blood
.
2022
;
140
(
11
):
1200
-
1228
.
7.
Khoury
JD
,
Solary
E
,
Abla
O
, et al
.
The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms
.
Leukemia
.
2022
;
36
(
7
):
1703
-
1719
.
8.
Cazzola
M
.
Myelodysplastic syndromes
.
N Engl J Med
.
2020
;
383
(
14
):
1358
-
1374
.
9.
Le Beau
MM
.
Deletions of chromosome 5 in malignant myeloid disorders
.
Cancer Surv
.
1992
;
15
:
143
-
159
.
10.
Mallo
M
,
Cervera
J
,
Schanz
J
, et al
.
Impact of adjunct cytogenetic abnormalities for prognostic stratification in patients with myelodysplastic syndrome and deletion 5q
.
Leukemia
.
2011
;
25
(
1
):
110
-
120
.
11.
Germing
U
,
Lauseker
M
,
Hildebrandt
B
, et al
.
Survival, prognostic factors and rates of leukemic transformation in 381 untreated patients with MDS and del(5q): a multicenter study
.
Leukemia
.
2012
;
26
(
6
):
1286
-
1292
.
12.
Boultwood
J
,
Fidler
C
,
Strickson
AJ
, et al
.
Narrowing and genomic annotation of the commonly deleted region of the 5q- syndrome
.
Blood
.
2002
;
99
(
12
):
4638
-
4641
.
13.
Boultwood
J
,
Pellagatti
A
,
McKenzie
AN
,
Wainscoat
JS
.
Advances in the 5q- syndrome
.
Blood
.
2010
;
116
(
26
):
5803
-
5811
.
14.
Lai
F
,
Godley
LA
,
Joslin
J
, 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
(
2
):
235
-
245
.
15.
Jerez
A
,
Gondek
LP
,
Jankowska
AM
, et al
.
Topography, clinical, and genomic correlates of 5q myeloid malignancies revisited
.
J Clin Oncol
.
2012
;
30
(
12
):
1343
-
1349
.
16.
Boultwood
J
,
Pellagatti
A
,
Cattan
H
, et al
.
Gene expression profiling of CD34+ cells in patients with the 5q- syndrome
.
Br J Haematol
.
2007
;
139
(
4
):
578
-
589
.
17.
Ebert
BL
,
Pretz
J
,
Bosco
J
, et al
.
Identification of RPS14 as a 5q- syndrome gene by RNA interference screen
.
Nature
.
2008
;
451
(
7176
):
335
-
339
.
18.
Da Costa
L
,
Leblanc
T
,
Mohandas
N
.
Diamond-Blackfan anemia
.
Blood
.
2020
;
136
(
11
):
1262
-
1273
.
19.
Barlow
JL
,
Drynan
LF
,
Hewett
DR
, et al
.
A p53-dependent mechanism underlies macrocytic anemia in a mouse model of human 5q- syndrome
.
Nat Med
.
2010
;
16
(
1
):
59
-
66
.
20.
Pellagatti
A
,
Marafioti
T
,
Paterson
JC
, et al
.
Induction of p53 and up-regulation of the p53 pathway in the human 5q- syndrome
.
Blood
.
2010
;
115
(
13
):
2721
-
2723
.
21.
Dutt
S
,
Narla
A
,
Lin
K
, et al
.
Haploinsufficiency for ribosomal protein genes causes selective activation of p53 in human erythroid progenitor cells
.
Blood
.
2011
;
117
(
9
):
2567
-
2576
.
22.
Schneider
RK
,
Schenone
M
,
Ferreira
MV
, et al
.
Rps14 haploinsufficiency causes a block in erythroid differentiation mediated by S100A8 and S100A9
.
Nat Med
.
2016
;
22
(
3
):
288
-
297
.
23.
Cazzola
M
.
Myelodysplastic syndrome with isolated 5q deletion (5q- syndrome). A clonal stem cell disorder characterized by defective ribosome biogenesis
.
Haematologica
.
2008
;
93
(
7
):
967
-
972
.
24.
Schneider
RK
,
Adema
V
,
Heckl
D
, et al
.
Role of casein kinase 1A1 in the biology and targeted therapy of del(5q) MDS
.
Cancer Cell
.
2014
;
26
(
4
):
509
-
520
.
25.
Bello
E
,
Pellagatti
A
,
Shaw
J
, et al
.
CSNK1A1 mutations and gene expression analysis in myelodysplastic syndromes with del(5q)
.
Br J Haematol
.
2015
;
171
(
2
):
210
-
214
.
26.
Smith
AE
,
Kulasekararaj
AG
,
Jiang
J
, et al
.
CSNK1A1 mutations and isolated del(5q) abnormality in myelodysplastic syndrome: a retrospective mutational analysis
.
Lancet Haematol
.
2015
;
2
(
5
):
e212
-
e221
.
27.
Stalmann
USA
,
Ticconi
F
,
Snoeren
IAM
, et al
.
Genetic barcoding systematically compares genes in del(5q) MDS and reveals a central role for CSNK1A1 in clonal expansion
.
Blood Adv
.
2022
;
6
(
6
):
1780
-
1796
.
28.
Eulalio
A
,
Huntzinger
E
,
Izaurralde
E
.
Getting to the root of miRNA-mediated gene silencing
.
Cell
.
2008
;
132
(
1
):
9
-
14
.
29.
Starczynowski
DT
,
Kuchenbauer
F
,
Argiropoulos
B
, et al
.
Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype
.
Nat Med
.
2010
;
16
(
1
):
49
-
58
.
30.
Kumar
MS
,
Narla
A
,
Nonami
A
, et al
.
Coordinate loss of a microRNA and protein-coding gene cooperate in the pathogenesis of 5q- syndrome
.
Blood
.
2011
;
118
(
17
):
4666
-
4673
.
31.
Woll
PS
,
Kjallquist
U
,
Chowdhury
O
, et al
.
Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo
.
Cancer Cell
.
2014
;
25
(
6
):
794
-
808
.
32.
Adema
V
,
Palomo
L
,
Walter
W
, et al
.
Pathophysiologic and clinical implications of molecular profiles resultant from deletion 5q
.
EBioMedicine
.
2022
;
80
:
104059
.
33.
Raza
A
,
Meyer
P
,
Dutt
D
, et al
.
Thalidomide produces transfusion independence in long-standing refractory anemias of patients with myelodysplastic syndromes
.
Blood
.
2001
;
98
(
4
):
958
-
965
.
34.
List
A
,
Kurtin
S
,
Roe
DJ
, et al
.
Efficacy of lenalidomide in myelodysplastic syndromes
.
N Engl J Med
.
2005
;
352
(
6
):
549
-
557
.
35.
List
A
,
Dewald
G
,
Bennett
J
, et al
.
Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion
.
N Engl J Med
.
2006
;
355
(
14
):
1456
-
1465
.
36.
Greenberg
P
,
Cox
C
,
LeBeau
MM
, et al
.
International scoring system for evaluating prognosis in myelodysplastic syndromes
.
Blood
.
1997
;
89
(
6
):
2079
-
2088
.
37.
Fenaux
P
,
Giagounidis
A
,
Selleslag
D
, et al
.
A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with low-/intermediate-1-risk myelodysplastic syndromes with del5q
.
Blood
.
2011
;
118
(
14
):
3765
-
3776
.
38.
Hellstrom-Lindberg
ES
,
Kroger
N
.
Clinical decision-making and treatment of myelodysplastic syndromes
.
Blood
.
2023
;
142
(
26
):
2268
-
2281
.
39.
Singh
A
,
Al-Kali
A
,
Foran
JM
, et al
.
Lenalidomide therapy for primary myelodysplastic syndromes with isolated del(5q): determinants of response and survival in a real-world setting
.
Am J Hematol
.
2022
;
97
(
10
):
E377
-
E379
.
40.
Crisa
E
,
Mora
E
,
Germing
U
, et al
.
Transfusion independence after lenalidomide discontinuation in patients with del(5q) myelodysplastic neoplasm: a HARMONY Alliance study
.
Leukemia
.
2024
;
38
(
10
):
2259
-
2265
.
41.
Diez-Campelo
M
,
Lopez-Cadenas
F
,
Xicoy
B
, et al
.
Low dose lenalidomide versus placebo in non-transfusion dependent patients with low risk, del(5q) myelodysplastic syndromes (SintraREV): a randomised, double-blind, phase 3 trial
.
Lancet Haematol
.
2024
;
11
(
9
):
e659
-
e670
.
42.
Greenberg
PL
,
Tuechler
H
,
Schanz
J
, et al
.
Revised international prognostic scoring system for myelodysplastic syndromes
.
Blood
.
2012
;
120
(
12
):
2454
-
2465
.
43.
Wei
S
,
Chen
X
,
Rocha
K
, et al
.
A critical role for phosphatase haplodeficiency in the selective suppression of deletion 5q MDS by lenalidomide
.
Proc Natl Acad Sci U S A
.
2009
;
106
(
31
):
12974
-
12979
.
44.
Kronke
J
,
Fink
EC
,
Hollenbach
PW
, et al
.
Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS
.
Nature
.
2015
;
523
(
7559
):
183
-
188
.
45.
Tehranchi
R
,
Woll
PS
,
Anderson
K
, et al
.
Persistent malignant stem cells in del(5q) myelodysplasia in remission
.
N Engl J Med
.
2010
;
363
(
11
):
1025
-
1037
.
46.
Jadersten
M
,
Saft
L
,
Smith
A
, et al
.
TP53 mutations in low-risk myelodysplastic syndromes with del(5q) predict disease progression
.
J Clin Oncol
.
2011
;
29
(
15
):
1971
-
1979
.
47.
Mallo
M
,
Del Rey
M
,
Ibanez
M
, et al
.
Response to lenalidomide in myelodysplastic syndromes with del(5q): influence of cytogenetics and mutations
.
Br J Haematol
.
2013
;
162
(
1
):
74
-
86
.
48.
Mossner
M
,
Jann
JC
,
Nowak
D
, et al
.
Prevalence, clonal dynamics and clinical impact of TP53 mutations in patients with myelodysplastic syndrome with isolated deletion (5q) treated with lenalidomide: results from a prospective multicenter study of the German MDS study group (GMDS)
.
Leukemia
.
2016
;
30
(
9
):
1956
-
1959
.
49.
Lode
L
,
Menard
A
,
Flet
L
, et al
.
Emergence and evolution of TP53 mutations are key features of disease progression in myelodysplastic patients with lower-risk del(5q) treated with lenalidomide
.
Haematologica
.
2018
;
103
(
4
):
e143
-
e146
.
50.
Fleti
F
,
Chan
O
,
Singh
A
, et al
.
TP53 mutations and variant allele frequency in myelodysplastic syndromes with del(5q): a Mayo-Moffitt study of 156 informative cases
.
Am J Hematol
.
2023
;
98
(
4
):
E76
-
E79
.
51.
Tefferi
A
,
Fleti
F
,
Chan
O
, et al
.
TP53 variant allele frequency and therapy-related setting independently predict survival in myelodysplastic syndromes with del(5q)
.
Br J Haematol
.
2024
;
204
(
4
):
1243
-
1248
.
52.
Abdallah
M
,
Reichard
K
,
Gangat
N
,
Tefferi
A
.
Treatment-emergent mutations in myelodysplastic syndrome with del(5q)-lenalidomide related or disease-intrinsic clonal evolution?
.
Blood Cancer J
.
2024
;
14
(
1
):
49
.
53.
Martinez-Hoyer
S
,
Deng
Y
,
Parker
J
, et al
.
Loss of lenalidomide-induced megakaryocytic differentiation leads to therapy resistance in del(5q) myelodysplastic syndrome
.
Nat Cell Biol
.
2020
;
22
(
5
):
526
-
533
.
54.
Sperling
AS
,
Guerra
VA
,
Kennedy
JA
, et al
.
Lenalidomide promotes the development of TP53-mutated therapy-related myeloid neoplasms
.
Blood
.
2022
;
140
(
16
):
1753
-
1763
.
55.
Bernard
E
,
Nannya
Y
,
Hasserjian
RP
, et al
.
Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes
.
Nat Med
.
2020
;
26
(
10
):
1549
-
1556
.
56.
Bernard
E
,
Hasserjian
RP
,
Greenberg
PL
, et al
.
Molecular taxonomy of myelodysplastic syndromes and its clinical implications
.
Blood
.
2024
;
144
(
15
):
1617
-
1632
.
57.
Montoro
MJ
,
Palomo
L
,
Haferlach
C
, et al
.
Influence of TP53 gene mutations and their allelic status in myelodysplastic syndromes with isolated 5q deletion
.
Blood
.
2024
;
144
(
16
):
1722
-
1731
.
58.
Cazzola
M
.
Risk stratifying MDS in the time of precision medicine
.
Hematol Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
375
-
381
.
59.
Bernard
E
,
Tuechler
H
,
Greenberg
PL
, et al
.
Molecular international prognostic scoring system for myelodysplastic syndromes
.
NEJM Evid
.
2022
;
1
(
7
):
EVIDoa2200008
.
60.
Meggendorfer
M
,
Haferlach
C
,
Kern
W
,
Haferlach
T
.
Molecular analysis of myelodysplastic syndrome with isolated deletion of the long arm of chromosome 5 reveals a specific spectrum of molecular mutations with prognostic impact: a study on 123 patients and 27 genes
.
Haematologica
.
2017
;
102
(
9
):
1502
-
1510
.
61.
Zampini
M
,
Riva
E
,
Lanino
L
, et al
.
Characterization and clinical implications of p53 dysfunction in patients with myelodysplastic syndromes
.
J Clin Oncol
.
2025
;
43
(
18
):
2069
-
2083
.
62.
Malcovati
L
,
Cazzola
M
.
How I manage patients with unexplained cytopenia
.
Blood
.
2025
;
145
(
15
):
1610
-
1620
.
63.
Mallo
M
,
Arenillas
L
,
Espinet
B
, et al
.
Fluorescence in situ hybridization improves the detection of 5q31 deletion in myelodysplastic syndromes without cytogenetic evidence of 5q
.
Haematologica
.
2008
;
93
(
7
):
1001
-
1008
.
64.
Jaju
RJ
,
Boultwood
J
,
Oliver
FJ
, et al
.
Molecular cytogenetic delineation of the critical deleted region in the 5q- syndrome
.
Genes Chromosomes Cancer
.
1998
;
22
(
3
):
251
-
256
.
65.
Makishima
H
,
Rataul
M
,
Gondek
LP
, et al
.
FISH and SNP-A karyotyping in myelodysplastic syndromes: improving cytogenetic detection of del(5q), monosomy 7, del(7q), trisomy 8 and del(20q)
.
Leuk Res
.
2010
;
34
(
4
):
447
-
453
.
66.
Duncavage
EJ
,
Bagg
A
,
Hasserjian
RP
, et al
.
Genomic profiling for clinical decision making in myeloid neoplasms and acute leukemia
.
Blood
.
2022
;
140
(
21
):
2228
-
2247
.
67.
Cazzola
M
,
Malcovati
L
.
Genome sequencing in the management of myelodysplastic syndromes and related disorders
.
Haematologica
.
2025
;
110
(
2
):
312
-
329
.
68.
Saiki
R
,
Momozawa
Y
,
Nannya
Y
, et al
.
Combined landscape of single-nucleotide variants and copy number alterations in clonal hematopoiesis
.
Nat Med
.
2021
;
27
(
7
):
1239
-
1249
.
69.
Cazzola
M
,
Sehn
LH
.
Developing a classification of hematologic neoplasms in the era of precision medicine
.
Blood
.
2022
;
140
(
11
):
1193
-
1199
.
70.
Gurnari
C
,
Robin
M
,
Ades
L
, et al
.
Clinical-genomic profiling of MDS to inform allo-HCT: recommendations from an international panel on behalf of the EBMT
.
Blood
.
2025
;
145
(
18
):
1987
-
2001
.
71.
Fenaux
P
,
Platzbecker
U
,
Mufti
GJ
, et al
.
Luspatercept in patients with lower-risk myelodysplastic syndromes
.
N Engl J Med
.
2020
;
382
(
2
):
140
-
151
.
72.
Platzbecker
U
,
Della Porta
MG
,
Santini
V
, et al
.
Efficacy and safety of luspatercept versus epoetin alfa in erythropoiesis-stimulating agent-naive, transfusion-dependent, lower-risk myelodysplastic syndromes (COMMANDS): interim analysis of a phase 3, open-label, randomised controlled trial
.
Lancet
.
2023
;
402
(
10399
):
373
-
385
.
73.
Platzbecker
U
,
Santini
V
,
Fenaux
P
, et al
.
Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): a multinational, randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
.
2024
;
403
(
10423
):
249
-
260
.
74.
Garderet
L
,
Ziagkos
D
,
van Biezen
A
, et al
.
Allogeneic stem cell transplantation for myelodysplastic syndrome patients with a 5q deletion
.
Biol Blood Marrow Transpl
.
2018
;
24
(
3
):
507
-
513
.
75.
Kroger
N
.
Treatment of high-risk myelodysplastic syndromes
.
Haematologica
.
2025
;
110
(
2
):
339
-
349
.
76.
Rasmussen
B
,
Nilsson
L
,
Tobiasson
M
, et al
.
Influence of cytogenetics on the outcome of patients with high-risk myelodysplastic syndrome Including deletion 5q treated with azacitidine with or without lenalidomide
.
Genes Chromosomes Cancer
.
2025
;
64
(
2
):
e70029
.
77.
Nowell
P
,
Hungerford
D
.
A minute chromosome in human chronic granulocytic leukemia [abstract]
.
Science
.
1960
;
142
:
1497
.

Author notes

The online version of this article contains a data supplement.

Sign in via your Institution