Abstract
A number of sequencing studies identified the prognostic impact of somatic mutations in myelodysplastic syndrome (MDS). However the majority of them focused on methylation regulation, apoptosis and proliferation genes. Despite the number of experimental studies published on the role of micro-RNA processing and checkpoint genes in the development of MDS, the clinical data about mutational landscape in these genes is limited. We performed a pilot study which evaluated mutational burden in these genes and their association with common MDS mutations. High prevalence of mutations was observed in the genes studied: 54% had mutations in DICER1, 46% had mutations in LAG3, 20% in CTLA4, 23% in B7-H3, 17% in DROSHA, 14% in PD-1 and 3% in PD-1L. Cluster analysis that included these mutations along with mutations in ASXL1, DNMT3A, EZH2, IDH1, RUNX1, SF3B1, SRSF2, TET2 and TP53 effectively predicted overall survival in the study group (HR 4.2, 95%CI 1.3–13.6, p = 0.016). The study results create the rational for incorporating micro-RNA processing and checkpoint genes in the sequencing panels for MDS and evaluate their role in the multicenter studies.
Introduction
Myelodysplastic syndrome (MDS) is a heterogenic group of diseases characterized by accumulation of somatic mutations [1–3], alterations in the bone marrow niches [4], various pathological events in the immune system, including pyroptosis and autoimmune bone marrow damage [5, 6], tumor escape at later stages [7] and ineffective hematopoiesis as a result of aforementioned events. Genome instability and high incidence of secondary cancerogenic genetic events determines frequent transformation of MDS to acute myeloid leukemia (AML) [8]. The current standard of care in high-risk MDS are hypomethylating agents [9, 10], which significantly improve time to progression and survival, but only in the minority of patients they induce complete remission (CR). The only curative option is an allogeneic stem cell transplantation (SCT), but even in candidates in the modern era of advanced supportive care the results are generally worse than in CR of acute leukemia with only 30–40% of overall survival in 5 years [11, 12].
The relatively unfavorable outcomes after existing therapies drive the search for a novel therapeutic targets in high-risk MDS. One of the breakthroughs in modern oncology is the introduction of the checkpoint inhibitors into clinical practice [13]. The analysis of the checkpoint proteins expression in the bone marrow of MDS and AML patients demonstrated that myeloid cells express different checkpoint ligands and receptors, including CD80, CD86 and PD-1L [14–16]. However, the best response observed in the clinical studies of checkpoint inhibitors was “stable disease”, despite the fact that some patients had stabilization for a long period of time, indicating the potential efficacy of these agents in MDS [17]. The other checkpoint inhibitors, like anti-TIM3 and anti-CD47, have a more promising response rate, but longer follow up is required to determine whether this response translates into long-term remission [18, 19].
Another aspect of MDS pathogenesis is the changes in a bone marrow niche [20, 21]. The experimental studies indicate that knock out of the genes involved in micro-RNA processing and extracellular signaling, like DICER1, DROSHA and SBDS may lead to MDS-like phenotype [22]. Expression profile of these genes is also altered in MDS [23]. Despite current studies with next-generation sequencing (NGS) that include thousands of patients, these studies focus on 36–55 genes related to methylation, proliferation and apoptosis, while full exome sequencing is generally used to validate the results of panel sequencing in the subgroup of patients [24]. Despite some studies do focus on micro-RNA processing [25, 26] and checkpoint genes [27] in solid tumors, data in MDS regarding these additional potential mechanisms of MDS progression and resistance is limited. Thus we performed a pilot study evaluating interaction of mutations in the most commonly mutated genes, checkpoint and micro-RNA-associated genes.
Methods
Patients
The study included 35 patients with high-risk MDS consulted at the hematopoietic stem cell transplantation (HSCT) center at the time of diagnosis during 2008–2018. All patients provided informed consent for the use of their biological material in the research purposes. A total of 48 samples from the 35 enrolled patients were analyzed. The study was approved by the Ethical committee of the First Pavlov Medical University and performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments. All patients included in the study signed written informed consent for the use of their biological materials and medical records for research purposes before their inclusion in the study. The DNA samples were anonymized before sequencing. All samples, except longitudinal samples in six patients, were taken at diagnosis. The sequencing was performed in Apr 2020. The median age was 49 years (range 18–80). Eighty two percent had high or very high risk according to IPSS-R. Twenty five patients undergone HSCT the others received therapy with hypomethylating agents (Table 1).
Table 1. Characteristics of patients.
| Parameter | N, (%) |
|---|---|
| Age, years, median (range) | 49 (18–80) |
| Gender | |
| males | 21 (60%) |
| females | 14 (40%) |
| IPSS-R score | |
| Low | 1 (3%) |
| Intermediate | 5 (14%) |
| High | 18 (51%) |
| Very high | 11 (31%) |
| Blasts in bone marrow | |
| 0–4.9% | 8 (23%) |
| 5–10% | 16 (46%) |
| 10.1–20% | 11 (31%) |
| Karyotype | |
| Normal | 11 (31%) |
| Monosomal | 6 (17%) |
| Complex | 5 (14%) |
| Other | 13 (37%) |
| Neutrophils, median (range), 109/L | 1.0 (0–9.0) |
| Platelets, median (range), 109/L | 90 (10–518) |
| Transfusion dependence | 9 (26%) |
| SCT performed | 19 (54%) |
Targeted sequencing
Genomic DNA was extracted from the fresh bone marrow aspirates using TriZ reagent extraction Kit (Inogene, Russian Federation) and stored at -80°C until the day of the assay. Whole bone marrow was used to potentially capture the mutations in the bone marrow niche cells. Separation of subpopulations of cells was not performed before sequencing. The quality of the samples before the assay was analyzed using Qubit 4.0 (Thermo Fisher, CA, USA).
The libraries for target sequencing of genes were prepared using shotgun liquid hybridization method with DNA probes. KAPA HyperPlus Kit (Roche, Switzerland) according to the manufacturer instructions were used for library preparation. The enrichment of targeted genome sequences was performed using SeqCap EZ Target Enrichment System (Roche, Switzerland) according to the manufacturer instructions. The enrichment of the coding sequences was performed for the following genes: ASXL1, CD274 (Programmed cell death 1 ligand 1), CD276 (В7-Н3 ligand to CTLA4 and CD28 receptors), DICER1, DNMT3A, DROSHA, EZH2, IDH1, IDH2, LAG3, MFSD11 (Major facilitator superfamily domain containing 11), PDCD1 (Programmed cell death 1 receptor), PIKFYVE (Phosphoinositide kinase, FYVE-type zinc finger containing), RUNX1, SF3B1, SRSF2, TET2 and TP53. The coverage of the target genes was at least x1000. Sequencing was performed with MiSeq using MiSeq Reagent Kits v2 (Illumina, USA) with 2х250 n.p. complementary read regimen. The primary reads are available at BioProject, accession number PRJNA631513, https://www.ncbi.nlm.nih.gov/bioproject/631513.
Bioinformatics
Quality of reads was assessed using FastQC [28], they were aligned to the GRCh38 reference genome with BWA [29], next GATK 4.1.5.0 [30] duplicate marking, sorting, and base quality score recalibration tools were applied. SNP and INDEL calling was performed using Mutect2 algorithm [31] according to GATK Best Practices recommendations [32]. Effects of discovered variants was determined by Ensembl Variant Effect Predictor [33] and ANNOVAR [34], using RefSeq annotation [35], population and variant interpretation databases (COSMIC [36], GnomAD [37], ClinVar [38]), and prediction tools (PolyPhen-2 [39], SIFT [40], MutationTaster2 [41], MutationAssessor [42], PROVEAN [43], and CADD [44]). Variants with allele frequencies more than 1% according to GnomAD data were filtered.
Statistical analysis
The set of single nucleotide polymorphisms (SNPs) obtained with GATK was filtered according to the functionality and loci (synonymous, intronic and intergenic items were removed). The variant allele frequency (VAF) threshold of 5% was chosen to describe the frequency of common MDS-related mutations as the most frequently used presentation of the data. However the general analysis of mutation frequency in the microRNA processing genes and checkpoint genes was carried out with the 1% threshold, because these genes were described to have significant impact on microenvironment cells and tumor-infiltrating macrophages that comprise usually minor populations. The 1% threshold was supposed to capture these minor subpopulations. On the other hand, it ensured at least 10x reads per each mutation detected to avoid false positive results. Since the clinical relevance of the mutations in the microRNA processing and checkpoint genes is not determined, all mutations, exon and UTR, were included in the analysis. For clustering analysis those SNPs that were detected in only one patient, were excluded. In clustering analysis an every remained SNP we obtained a typical AF in the sample: a median AF among all the patients was calculated and rounded to the closest value from the set {0%, 50%, 100%}. This value was subtracted from every particular patient AF, thus providing an individual frequency shift bounded between -100% and +100%. A matrix of frequency shifts was composed, in which rows represented SNPs, and columns represented patients. A tree clustering was performed for the matrix columns and rows (Euclidian distance was used as a measure of items similarity). Top two patients clusters were taken for the downstream analysis: a set of clinical parameters were compared between clusters as well as survival characteristics. Quantitative parameters were compared with a Mann-Whitney test, survival analysis was performed with Kaplan-Meyer method. Heatmap was processed and visualized with pheatmap [45]. Circos plot was implemented with circlize package [46]. TCGAbiolinksGUI was used to visualize association of mutations [47].
Results
Identified mutations
The pattern of common MDS mutations with ≥5% VAF was similar to the previous studies. Twenty percent of patients at diagnosis had mutations in the ASXL1, 17% in TP53, 14% in DNMT3A, 14% in SF3B1, 11% in RUNX1, 9% in IDH1 and 6% in IDH2 and EZH1 each. Single instances of TET2 and SRFS2 were indentified. No common mutations were found in 14% of patients (Fig 1, S1 and S2 Figs).
Fig 1. Prevalence of common exonic pathogenic mutations in the study group.
White color in the heatmap represents absence of mutations. Blue colors represent mutations with VAF<50%, orange and red with VAF>50%. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted. (*) indicates mutations which are present in the COSMIC database. (**) indicates mutations associated with oncohematological diseases in the COSMIC database.
Since there is no data about the pathogenic impact of mutations in the micro-RNA processing genes and checkpoint genes as well as the size of clinically relevant populations of cells with mutations the analysis selected all mutations that affected either protein sequence or modified gene expression. Polymorphisms effecting more than 50% of patients were not accounted, but displayed in the graphical form. In the studied set of genes 140 unique mutations were indentified that fit the selection criteria (S1 Table). A significant number of mutations was observed in the checkpoint genes: 46% of patients had mutations in LAG3, 20% in CTLA4, 23% in B7-H3, 14% in PD-1 and 3% in PD-1L. Also a significant number of patients with PD-1L polymorphisms previously described in relation to adverse cancer outcomes were determined: rs4742098 in 54%, rs2297136 in 63%, rs4143815 in 54%. No previously described polymorphisms in other genes were identified.
Furthermore the prevalence of mutations in the micro-RNA processing genes was also relatively high: 17% in DROSHA and 54% in DICER1. A number of polymorphisms in these genes were also high but their significance is still undetermined. The cumulative incidence of SNPs in the studied set of genes regardless of their established pathogenic impact was highest in ASXL1, TET2, DICER1 and RUNX1 (S3 Fig).
The majority of observed SNPs are not reported to be pathogenic, so the following steps were performed to evaluate the influence of mutations on the clinical course of the disease: 1) deviations in VAF from the median in the group were calculated. This step allowed to separate both patients with abnormal polymorphisms and clonal changes; 2) tree clustering analysis was performed based on these deviations from median VAF in the group; 3) two clusters were identified (S4 Fig). Since the multiple comparison correction did not allow to achieve statistically significant results in the frequency of certain mutations, they were ordered by descending significance of differences between clusters. The first 15 most significant mutations demonstrated that in cluster 1 there was a higher prevalence of SF3B1, less DICER1 mutations with high VAF and less B7-H3 (CD276) mutations. Cluster 2 harbored more ASXL1 mutations, RUNX1 mutations, more PD-1L (CD274) mutations.
There was no difference between the identified clusters in the rate of SCT performed (50% vs 56%, p = 0.75). There was also no association of identified mutation clusters with IPSS-R score (p = 0.58), WPSS score (p = 0.34), Armand et al score (p = 0.21), age of the patients (p = 0.43), percentage of blasts in the bone marrow at diagnosis (p = 0.2), hemoglobin level at diagnosis (p = 0.84) and platelet level at diagnosis (p = 0.085). Also the distribution of patients who received HSCT was not different between two clusters (69% vs 73%, p = 0.75). Thus, the fact of HSCT did not interfered in the results of the analysis. There was a week association with neutrophil levels at diagnosis. Patients in the cluster 1 had lower levels (median 670 vs 990 x10^9/L, p = 0.013) (S5 Fig). Nonetheless there was a significant difference in overall survival. The 5-year overall survival estimate was higher in cluster 1 patients: 72% (95%CI 42–89%) vs 27% (95%CI 8–51%), p = 0.029 (Fig 2A). In the multivariate analysis with correction for IPSS-R score (HR 1.5, 95%CI 1.0–2.3, р = 0.28), the clusterization remained a significant predictor of all-cause mortality (HR 4.2, 95%CI 1.3–13.6, p = 0.016, Fig 2B).
Fig 2.
A. 5-year overall survival of patients in the identified clusters. B. Forrest plot with multivariate analysis of genetic clusters and IPSS-R. C. Example of clonal evolution with microRNA processing gene.
The analysis of mutation associations demonstrated uniform occurrence of mutations in the known MDS-associated genes and DICER1, DROSHA, and checkpoint genes. Except LAG3 with significant prevalence of missense mutations, SNPs in DICER1, DROSHA, CD274 and CD276 were predominantly documented in 5-UTR and 3-UTR regions (Fig 3, S6 Fig). The uniform distribution indicates that there is no pathogenic link between these mutations and they accumulate sporadically.
Fig 3. Associations between genetic abnormalities in the individual patients.
The left vertical bar indicate the percentage of patients with abnormalities in the gene. Red color indicate synonymous substitution. Upper horizontal bar characterize mutation burden in the individual patients per megabase pair. Middle boxplot indicates that type of mutations in the genes in individual patients. Mutations are ordered and presented based on their deleterious effect in the order as shown in the legend. Additional less deleterious mutations in the gene are not shown. Horizontal lower bar plot indicate the type of genetic alterations present in individual patient across all genes tested. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted.
In seven patients who did have longitudinal samples of the bone marrow aspirate the clonal evolution was traced with the same set of genes. The mutations appearing during the disease coarse affected tp53, SRSF2, DROSHA and DICER1 with high AF in the studied patients. The other mutations in the checkpoint genes were observed in the minor clones with predominant involvement of LAG3 (S7 Fig).
Discussion
MDS is the disease with one of the highest number of sequencing studies [48], however this is one of the first in MDS that confirmed the presence of mutations in checkpoint and micro-RNA processing genes and more than half of high-risk patients had these aberrations. This profile of mutations explains several observations from the experimental studies.
Both DROSHA and DICER1 are RNase III enzymes involved in the processing micro-RNA in the nucleus. It was demonstrated that alterations of micro-RNA signaling is due to abnormal functioning of these two enzymes [49]. In MDS multiple miRNAs were reported to be abnormally expressed, including pro-apoptotic miR-34a, anti-apoptotic miR-378 and miR-144, antioxidant miR-451 [50, 51], anti-DNMT1 miR-126 [52]. In mouse studies it was demonstrated that knock out of DICER1 in the mesenchymal cells in the bone marrow leads to abnormal expression of more than 10 miRNAs and MDS-like phenotype [53, 54]. Several miRNAs also regulate NLRP3 inflammasome which facilitates pyroptosis and hematopoiesis aging in MDS [5, 53, 54]. The observed VAFs and prevalence in the study group of DICER and DROSHA SNPs indicated that there were both polymorphisms and minor clones with somatic mutations, probably associated with the bone marrow niche cells. Further studies with selected subpopulations of cells are required to confirm the exact role of each genetic aberration in these genes.
Another aspect revealed in this study is the high frequency of mutations in the checkpoint genes. It was demonstrated that a number of checkpoint ligands, like PD-1L, PD-2L, B7, CD80 are overexpressed in MDS and in certain instances they are induced via inflammasome activation [55–57]. The accumulation of mutations in receptor genes leads to infective interaction with ligands and thus might represent the evolutionary protective changes against tumor progression in the setting of unstable genome and clonal hematopoiesis. The same finding may explain the moderate response to checkpoint blockade in MDS. The monoclonal antibodies may either not bind to the receptor with abnormal conformation or this receptor may not be expressed at all due to missense and frame shift mutations [57, 58].
The study does have several limitations, primary due to small number of patients. Particularly cautious should be the interpretation of clinical results. The difference in survival presented in the article was not to suggest the clinical predictive algorithm, but rather to demonstrate that when the results of the studied gene panel were analyzed mathematically there was some predictive power for the clinical outcomes. Also germline cells were not analyzed in parallel, which forced us to implement an advanced statistical methodology that facilitated interpretation of this data. However the major point of the study was not to identify and validate the significance of certain mutations, but rather highlight the importance of mutations in miRNA processing and checkpoint genes that should be included in the common MDS sequencing panels and evaluated in the large muticenter studies for their potential prognostic value and role in the pathogenesis.
Supporting information
The brighter colors indicate higher number of mutations in the patients’ genes. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH).
(PDF)
The left vertical bar indicate the percentage of patients with abnormalities in the gene. Upper horizontal bar characterize mutation burden in the individual patients per megabase pair. Middle boxplot indicates that type of mutations in the genes in individual patients. Mutations are ordered and presented based on their deleterious effect in the order as shown in the legend. Additional less deleterious mutations in the gene are not shown. Horizontal lower bar plot indicate the type of genetic alterations present in individual patient across all genes tested. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted.
(PDF)
The heat map includes both pathogeneic mutations and SNPs with undetermined significance. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Dark blue color represents high allele frequency shift downwards (up to -100%) from common allele frequency in the sample (the most part of the sample is homozygous with presence of this SNP, while the particular patient is homozygous with absence of this SNP). Similarly, red colors represent high allele frequency allele frequency shift upwards (the most part of the sample is homozygous with absence of this SNP, while the particular patient is homozygous with presence of this SNP). Yellow colors represent allele frequencies close to the median allele frequency for this gene in a patient. The patients are clustered according to their mutation patterns.
(PDF)
White color in the heatmap represents absence of mutations. Blue colors represent mutations with VAF<50%, orange and red with VAF>50%. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted. (*) indicates mutations which are present in the COSMIC database. (**) indicates mutations associated with oncohematological diseases in the COSMIC database.
(PDF)
(PDF)
Patients with only one mutated gene as well as genes with only one patient with mutation were excluded from analysis. The width of circus fragment represents the incidence of mutations in the specific genes. The width of the ribbon between the genes represents the rate of association.
(PDF)
(PDF)
(XLSX)
Data Availability
BioProject, accession number PRJNA631513, https://www.ncbi.nlm.nih.gov/bioproject/631513.
Funding Statement
This work was supported by Russian Science Foundation grant № 17-75-20145.
References
- 1.Busque L, Patel JP, Figueroa ME, Vasanthakumar A, Provost S, Hamilou Z et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet. 2012. November;44(11):1179–81. 10.1038/ng.2413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014. December 25;371(26):2477–87. 10.1056/NEJMoa1409405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015. July 2;126(1):9–16. 10.1182/blood-2015-03-631747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Montes P, Bernal M, Campo LN, González-Ramírez AR, Jiménez P, Garrido P et al. Tumor genetic alterations and features of the immune microenvironment drive myelodysplastic syndrome escape and progression. Cancer Immunol Immunother. 2019. December;68(12):2015–2027. 10.1007/s00262-019-02420-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Masters SL, Gerlic M, Metcalf D, Preston S, Pellegrini M, O’Donnell JA et al. NLRP1 inflammasome activation induces pyroptosis of hematopoietic progenitor cells. Immunity. 2012. December 14; 37(6):1009–23. 10.1016/j.immuni.2012.08.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sallman DA, Cluzeau T, Basiorka AA, List A. Unraveling the Pathogenesis of MDS: The NLRP3 Inflammasome and Pyroptosis Drive the MDS Phenotype. Front Oncol. 2016. June 16;6:151. 10.3389/fonc.2016.00151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sugimori C, List AF, Epling-Burnette PK. Immune dysregulation in myelodysplastic syndrome. Hematol Rep. 2010. January 26;2(1):e1. 10.4081/hr.2010.e1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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. September;102(9):1502–1510. 10.3324/haematol.2017.166173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kantarjian HM, Thomas XG, Dmoszynska A, Wierzbowska A, Mazur G, Mayer J et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012. July 20;30(21):2670–7. 10.1200/JCO.2011.38.9429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A et al.; International Vidaza High-Risk MDS Survival Study Group. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009. March;10(3):223–32. 10.1016/S1470-2045(09)70003-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Heuser M, Gabdoulline R, Löffeld P, Dobbernack V, Kreimeyer H, Pankratz M et al. Individual outcome prediction for myelodysplastic syndrome (MDS) and secondary acute myeloid leukemia from MDS after allogeneic hematopoietic cell transplantation. Ann Hematol. 2017. August;96(8):1361–1372. 10.1007/s00277-017-3027-5 Epub 2017 Jun 13. . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Vij R, Le-Rademacher J, Laumann K, Hars V, Owzar K, Shore T et al. A Phase II Multicenter Study of the Addition of Azacitidine to Reduced-Intensity Conditioning Allogeneic Transplant for High-Risk Myelodysplasia (and Older Patients with Acute Myeloid Leukemia): Results of CALGB 100801 (Alliance). Biol Blood Marrow Transplant. 2019. October;25(10):1984–1992. 10.1016/j.bbmt.2019.06.007 Epub 2019 Jun 15. ; PMCID: PMC6790289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018. September;8(9):1069–1086. 10.1158/2159-8290.CD-18-0367 [DOI] [PubMed] [Google Scholar]
- 14.Costello RT, Mallet F, Sainty D, Maraninchi D, Gastaut JA, Olive D. Regulation of CD80/B7-1 and CD86/B7-2 molecule expression in human primary acute myeloid leukemia and their role in allogenic immune recognition. Eur J Immunol. 1998;28(1):90–103. [DOI] [PubMed] [Google Scholar]
- 15.Graf M, Reif S, Hecht K, Pelka-Fleischer R, Kroell T, Pfister K, et al. High expression of costimulatory molecules correlates with low relapse-free survival probability in acute myeloid leukemia (AML). Ann Hematol. 2005. May;84(5):287–97. 10.1007/s00277-004-0978-0 [DOI] [PubMed] [Google Scholar]
- 16.Yoyen-Ermis D, Tunali G, Tavukcuoglu E, Horzum U, Ozkazanc D, Sutlu T et al. Myeloid maturation potentiates STAT3-mediated atypical IFN-γ signaling and upregulation of PD-1 ligands in AML and MDS. Sci Rep. 2019. August 12;9(1):11697. 10.1038/s41598-019-48256-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Garcia-Manero G, Daver NG, Montalban-Bravo G, Jabbour EJ, DiNardo CD, Kornblau SM et al. A Phase II Study Evaluating the Combination of Nivolumab (Nivo) or Ipilimumab (Ipi) with Azacitidine in Pts with Previously Treated or Untreated Myelodysplastic Syndromes (MDS). Blood 2016; 128 (22): 344. [Google Scholar]
- 18.Borate U, Esteve J, Porkka K, Knapper S, Vey R, Scholl S et al. Phase Ib Study of the Anti-TIM-3 Antibody MBG453 in Combination with Decitabine in Patients with High-Risk Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML). Blood 2019; 134 (S1): 570. [Google Scholar]
- 19.Russ A, Hua AB, Montfort WR, Rahman B, Riaz IB, Khalid MU et al. Blocking "don’t eat me" signal of CD47-SIRPα in hematological malignancies, an in-depth review. Blood Rev. 2018. November;32(6):480–489. 10.1016/j.blre.2018.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pronk E, Raaijmakers MH. The mesenchymal niche in MDS. Blood 2019; 133: 1031–1038. 10.1182/blood-2018-10-844639 [DOI] [PubMed] [Google Scholar]
- 21.Raaijmakers MH. Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol. 2012. January;95(1):17–25. 10.1007/s12185-011-1001-x [DOI] [PubMed] [Google Scholar]
- 22.Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010. April 8;464(7290):852–7. 10.1038/nature08851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Santamaría C, Muntión S, Rosón B, Blanco B, López-Villar O, Carrancio S et al. Impaired expression of DICER, DROSHA, SBDS and some microRNAs in mesenchymal stromal cells from myelodysplastic syndrome patients. Haematologica. 2012. August;97(8):1218–24. 10.3324/haematol.2011.054437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nagata Y, Makishima H, Kerr CM, Przychodzen BP, Aly M, Goyal A et al. Invariant patterns of clonal succession determine specific clinical features of myelodysplastic syndromes. Nat Commun. 2019. November 26;10(1):5386. 10.1038/s41467-019-13001-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sand M, Bromba A, Sand D, Gambichler T, Hessam S, Becker JC et al. Dicer Sequencing, Whole Genome Methylation Profiling, mRNA and smallRNA Sequencing Analysis in Basal Cell Carcinoma. Cell Physiol Biochem. 2019;53(5):760–773. 10.33594/000000171 [DOI] [PubMed] [Google Scholar]
- 26.Lambo S, Gröbner SN, Rausch T, Waszak SM, Schmidt C, Gorthi A et al. The molecular landscape of ETMR at diagnosis and relapse. Nature. 2019. December;576(7786):274–280. 10.1038/s41586-019-1815-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Johnson DB, Frampton GM, Rioth MJ, Yusko E, Xu Y, Guo X et al. Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol Res. 2016. November;4(11):959–967. 10.1158/2326-6066.CIR-16-0143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Andrews S. FastQC: A Quality Control Tool for High Throughput Sequence Data [Online]. 2010; Available online at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/
- 29.Li H & Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. 2009; Bioinformatics, 25(14), 1754–1760. 10.1093/bioinformatics/btp324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.McKenna A., Hanna M., Banks E. et al. The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Research, 2010; 20(9), 1297–1303. 10.1101/gr.107524.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013. March;31(3):213–9. 10.1038/nbt.2514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011. May;43(5):491–8. 10.1038/ng.806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A et al. The Ensembl Variant Effect Predictor. Genome Biol. 2016. June 6;17(1):122. 10.1186/s13059-016-0974-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang K, Li M, & Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Research. 2010;38(16), e164–e164. 10.1093/nar/gkq603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.O’Leary NA, Wright MW, Brister JR, Ciufo S, Haddad D, McVeigh R et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 2016. January 4;44(D1):D733–45. 10.1093/nar/gkv1189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019. January 8;47(D1):D941–D947. 10.1093/nar/gky1015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J, Wang Q et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020. May;581(7809):434–443. 10.1038/s41586-020-2308-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018. January 4;46(D1):D1062–D1067. 10.1093/nar/gkx1153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010. April;7(4):248–9. 10.1038/nmeth0410-248 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sim NL, Kumar P, Hu J, Henikoff S, Schneider G, Ng PC. SIFT web server: predicting effects of amino acid substitutions on proteins. Nucleic Acids Res. 2012. July;40(Web Server issue):W452–7. 10.1093/nar/gks539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Schwarz JM, Cooper DN, Schuelke M, Seelow D. MutationTaster2: mutation prediction for the deep-sequencing age. Nat Methods. 2014. April;11(4):361–2. 10.1038/nmeth.2890 [DOI] [PubMed] [Google Scholar]
- 42.Reva B, Antipin Y, & Sander C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Research. 2011;39(17), e118–e118. 10.1093/nar/gkr407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Choi Y, & Chan A P PROVEAN web server: a tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics. 2015; 31(16), 2745–2747. 10.1093/bioinformatics/btv195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rentzsch P, Witten D, Cooper GM, Shendure J, Kircher M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 2019. January 8;47(D1):D886–D894. 10.1093/nar/gky1016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Raivo Kolde R. pheatmap: Pretty Heatmaps. 2019. R package version 1.0.12. https://CRAN.R-project.org/package=pheatmap
- 46.Gu Z, Gu L, Eils R et al. Circlize implements and enhances circular visualization in R. Bioinformatics. 2014. October;30(19):2811–2. 10.1093/bioinformatics/btu393 [DOI] [PubMed] [Google Scholar]
- 47.Silva TC, Colaprico A, Olsen C et al. TCGAbiolinksGUI: A graphical user interface to analyze cancer molecular and clinical data [version 1; peer review: 1 approved, 1 approved with reservations]. F1000Research 2018, 7:439. [Google Scholar]
- 48.Tcvetkov NY, Epifanovskaya OS, Rudnitskaya YV, Morozova EV, Moiseev IS, Afanasyev BV. Meta-analysis of studies with genome sequencing in myelodysplastic syndrome treated with hypomethylating agents. Cellular Therapy and Transplantation. 2018. 7(1): 44–51. [Google Scholar]
- 49.Thomson JM, Newman M, Parker JS, Morin-Kensicki EM, Wright T, Hammond SM. Extensive post-transcriptional regulation of microRNAs and its implications for cancer. Genes Dev. 2006. August 15;20(16):2202–7. 10.1101/gad.1444406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Merkerova MD, Krejcik Z, Belickova M, Hrustincova A, Klema J, Stara E et al. Genome-wide miRNA profiling in myelodysplastic syndrome with del(5q) treated with lenalidomide. Eur J Haematol. 2015. July;95(1):35–43. 10.1111/ejh.12458 [DOI] [PubMed] [Google Scholar]
- 51.Qian W, Jin F, Zhao Y, Chen Y, Ge L, Liu L et al. Downregulation of microRNA-144 inhibits proliferation and promotes the apoptosis of myelodysplastic syndrome cells through the activation of the AKAP12-dependent ERK1/2 signaling pathway. Cell Signal. 2020. April;68:109493. 10.1016/j.cellsig.2019.109493 [DOI] [PubMed] [Google Scholar]
- 52.Solly F, Koering C, Mohamed AM, Maucort-Boulch D, Robert G, Auberger P et al. An miRNA-DNMT1 Axis Is Involved in Azacitidine Resistance and Predicts Survival in Higher-Risk Myelodysplastic Syndrome and Low Blast Count Acute Myeloid Leukemia. Clin Cancer Res. 2017. June 15;23(12):3025–3034. 10.1158/1078-0432.CCR-16-2304 [DOI] [PubMed] [Google Scholar]
- 53.Chen L, Hou X, Zhang M, Zheng Y, Zheng X, Yang Q et al. MicroRNA-223-3p modulates dendritic cell function and ameliorates experimental autoimmune myocarditis by targeting the NLRP3 inflammasome. Mol Immunol. 2020. January;117:73–83. 10.1016/j.molimm.2019.10.027 [DOI] [PubMed] [Google Scholar]
- 54.Dong F, Dong S, Liang Y, Wang K, Qin Y, Zhao X. miR‑20b inhibits the senescence of human umbilical vein endothelial cells through regulating the Wnt/β‑catenin pathway via the TXNIP/NLRP3 axis. Int J Mol Med. 2020. March;45(3):847–857. 10.3892/ijmm.2020.4457 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dail M, Yang L, Green C, Ma C, Robert A, Kadel EE et al. Distinct Patterns of PD-L1 and PD-L2 Expression By Tumor and Non-Tumor Cells in Patients with MM, MDS and AML. Blood 2016; 128: 1340. [Google Scholar]
- 56.Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014. June;28(6):1280–8. 10.1038/leu.2013.355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cheng P, Eksioglu EA, Chen X, Kandell W, Le Trinh T, Cen L et al. S100A9-induced overexpression of PD-1/PD-L1 contributes to ineffective hematopoiesis in myelodysplastic syndromes. Leukemia. 2019. August;33(8):2034–2046. 10.1038/s41375-019-0397-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zeidan AM, Knaus HA, Robinson TM, Towlerton AMH, Warren EH, Zeidner JF et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin Cancer Res. 2018. August 1;24(15):3519–3527. 10.1158/1078-0432.CCR-17-3763 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
The brighter colors indicate higher number of mutations in the patients’ genes. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH).
(PDF)
The left vertical bar indicate the percentage of patients with abnormalities in the gene. Upper horizontal bar characterize mutation burden in the individual patients per megabase pair. Middle boxplot indicates that type of mutations in the genes in individual patients. Mutations are ordered and presented based on their deleterious effect in the order as shown in the legend. Additional less deleterious mutations in the gene are not shown. Horizontal lower bar plot indicate the type of genetic alterations present in individual patient across all genes tested. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted.
(PDF)
The heat map includes both pathogeneic mutations and SNPs with undetermined significance. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Dark blue color represents high allele frequency shift downwards (up to -100%) from common allele frequency in the sample (the most part of the sample is homozygous with presence of this SNP, while the particular patient is homozygous with absence of this SNP). Similarly, red colors represent high allele frequency allele frequency shift upwards (the most part of the sample is homozygous with absence of this SNP, while the particular patient is homozygous with presence of this SNP). Yellow colors represent allele frequencies close to the median allele frequency for this gene in a patient. The patients are clustered according to their mutation patterns.
(PDF)
White color in the heatmap represents absence of mutations. Blue colors represent mutations with VAF<50%, orange and red with VAF>50%. The risk line is the IPSS-R score presented by groups: low (L), intermediate (I), high (H), very high (VH). Transplantation line indicates whether the patient was allografted. (*) indicates mutations which are present in the COSMIC database. (**) indicates mutations associated with oncohematological diseases in the COSMIC database.
(PDF)
(PDF)
Patients with only one mutated gene as well as genes with only one patient with mutation were excluded from analysis. The width of circus fragment represents the incidence of mutations in the specific genes. The width of the ribbon between the genes represents the rate of association.
(PDF)
(PDF)
(XLSX)
Data Availability Statement
BioProject, accession number PRJNA631513, https://www.ncbi.nlm.nih.gov/bioproject/631513.



