Abstract
ETV6::ACSL6 represents a rare genetic aberration in hematopoietic neoplasms and is often associated with severe eosinophilia, which confers an unfavorable prognosis requiring additional anti-inflammatory treatment. However, since the translocation is unlikely to produce a fusion protein, the mechanism of ETV6::ACSL6 action remains unclear. Here, we performed multi-omics analyses of primary leukemia cells and patient-derived xenografts from an acute lymphoblastic leukemia (ALL) patient with ETV6::ACSL6 translocation. We identified a super-enhancer located within the ETV6 gene locus, and revealed translocation and activation of the super-enhancer associated with the ETV6::ACSL6 fusion. The translocated super-enhancer exhibited intense interactions with genomic regions adjacent to and distal from the breakpoint at chromosomes 5 and 12, including genes coding inflammatory factors such as IL-3. This led to modulations in DNA methylation, histone modifications, and chromatin structures, triggering transcription of inflammatory factors leading to eosinophilia. Furthermore, the bromodomain and extraterminal domain (BET) inhibitor synergized with standard-of-care drugs for ALL, effectively reducing IL-3 expression and inhibiting ETV6::ACSL6 ALL growth in vitro and in vivo. Overall, our study revealed for the first time a cis-regulatory mechanism of super-enhancer translocation in ETV6::ACSL6ALL, leading to an ALL-accompanying clinical syndrome. These findings may stimulate novel treatment approaches for this challenging ALL subtype.
Introduction
Chromosomal rearrangements are common in cancers, particularly hematologic malignancies.1 In acute lymphoblastic leukemia (ALL), their characterization has led to significant improvements in risk stratification and the development of targeted therapy.2,3 ETV6 is reported to form fusion genes with over 30 different partners, representing one of the most frequently translocated genes in ALL.4 While ETV6::RUNX1, a common ETV6 fusion in children, indicates favorable outcomes,4 other ETV6 fusions indicate poor prognosis.5,6 In addition to standard-of-care chemotherapy, targeted therapies have been regularly applied in the clinic to inhibit the trans-regulatory activities of ETV6 partner proteins.5 ,7 Even though ETV6 is critical for hematopoiesis, its function in leukemogenesis may be underestimated compared to its partner proteins which play a dominant role in the dysregulation of downstream genes and pathways.7, 8 Moreover, the mechanisms by which various ETV6 fusions cause malignancy remain poorly understood.
ETV6::ACSL6 t(5;12)(q31;p13) is a rare ETV6 fusion, with only 17 cases reported in myeloid malignancies worldwide until 2022, and none in ALL. The prognosis of patients with ETV6::ACSL6 is usually unfavorable, with most patients surviving less than a year.9 Eosinophilia is a common complication of the disease, which often results in damage to various organs. The elevated eosinophils, in severe cases, can cause cerebral infarction and heart failure, posing life-threatening risks. This complicates the clinical care of these patients, and underscores the significance of prompt identification and treatment. Previous studies have found several pro-inflammatory mediators, including interleukin (IL)-3 and IL-5, to be up-regulated in acute myeloid leukemia (AML) with ETV6 t(5;12) translocations.10 While IL-5 is considered vital for the maturation and differentiation of eosinophils, IL-3 displays more potent effects on their functions.11 However, the mechanism of IL-3 and IL-5 upregulation in ETV6::ACSL6 is yet to be elucidated.
Translocation events can induce trans- or cis-regulatory activities, altering gene expression profiles of cancer cells.12 Most previous studies focused on trans-regulatory activities of oncogenic fusion proteins.13-15 However, ETV6::ACSL6 has been shown to be associated with frameshift mutation conferring a premature stop codon, thus unlikely to be translated into a full-length protein,16 suggesting an alternative mechanism is involved. Cis-regulatory elements play critical roles in oncogenesis through structural variations (SV). This may cause ‘regulatory rearrangements’ of promoters and enhancers, leading to dysregulation of oncogenes. For example, t(8;14)(q24;q32), the most common translocation in Burkitt lymphoma, leads to MYC overexpression due to the relocation of an enhancer from chromosome (chr) 14 to its nearby region.17 Furthermore, in 2004, it was shown that the ectopic expression of the homeobox gene CDX2 resulting from the t(12;13)(p13;q12) and not the expression of the ETV6::CDX2 fusion gene resulted in AML in a murine model.18 Recently, ETV6 was also demonstrated to regulate its partner gene MN1 via super-enhancer (SE) hijacking in AML.19 However, the mechanism is yet to be extended to other ETV6 fusions as well as genes apart from its partners, and the clinical relevance of ETV6-associated SE hijacking events remains unexplored.
Coding genes of inflammatory factors, including I L- 3, IL-5 and GM-CSF, are located adjacent to ACSL6 on chr5. This raises a question of whether and how ETV6::ACSL6 is involved in the dysregulation of these genes. Therefore, in this study, we used multi-omics approach to interrogate the ETV6 translocation-induced cis-regulatory mutation and changes in the 3D genome structure in ETV6::ACSL6 ALL to decipher mechanisms of inflammatory factor dysregulation and its associated clinical syndrome eosinophilia.
Methods
Bone marrow samples from patients
Bone marrow (BM) samples from ALL patients or the healthy donor (diagnosed with thrombocytopenia, but with both BM smear and flow cytometry showing no abnormalities) were obtained under informed consent from Ruijin Hospital. Mononuclear cells were enriched by density gradient centrifugation with Ficoll solution. The use of samples was approved by the institutional review board. All relevant ethical regulations were followed in this study.
scNMT-sequencing library preparation and sequencing
scNMT-sequencing (seq) libraries were prepared according to a previous protocol.20 Single cells were sorted using FACS Aria into 96-well plates containing 2.5 μl of reaction buffer: 1xM.CviPI buffer (NEB), 2 U M.CviPI (NEB), 160 μM S-adenosylmethionine (NEB), 1 U μl-1 RNasein (Thermo), 0.1% IGEPAL CA-630 (Sigma). After a 15-minute incubation at 37°C, RLT plus buffer (Qiagen) were added and samples stored at -80°C. Poly-A RNA was captured on oligo-dT and amplified cDNA was prepared according to Smart-seq2 protocols. The gDNA lysate was purified on AMPureXP beads before bisulfite-sequencing (BS-seq) libraries were prepared according to the scBS-seq protocol.21 Sequencing was carried out on a NovaSeq instrument, with a mean raw sequencing depth of 7.5 million (BS-seq) and 5 million (RNA-seq) paired-end reads per cell. BS-seq alignment and methylation/accessibility quantification was performed following a previous approach.20 Briefly, individual CpG or GpC sites of each cell were modeled using a binomial distribution, where the number of successes represented the reads supporting methylation, and the number of trials was the total read count. The CpG methylation or GpC accessibility rate for each site and cell was determined through maximum likelihood estimation. Subsequently, the rates were rounded to the nearest integer (0 or 1).
All other methods are described in detail in the Online Supplementary Appendix.
Results
Transcriptomic analysis of ETV6::ACSL6 acute lymphoblastic leukemia and its accompanied eosinophils
The ETV6::ACSL6 ALL patient is a 66-year-old male who presented to the clinic with a dry cough. The eosinophil count has consistently been greater than 1.5x109/L for over five years. The patient demonstrated elevated eosinophils (black arrow) in the peripheral blood and BM (Figure 1A). Two distinct populations of ALL cells and eosinophils were observed by flowcytometry analysis, with ALL cells exhibiting classic immunophenotype (CD19+CD45dimC-D10+CD20-CD38dimCD58+) (Figure 1B). Monitoring of routine blood tests showed similar dynamics of cell counts in ALL and eosinophils following induction therapy (Online Supplementary Figure S1). The karyotype revealed 46,XY,t(5;12) (q31;p13),del(11)(p15)[1]/46,XY[23]. RNA-seq was performed on the two populations individually after flow-sorting and ETV6::ACSL6 was only detected in lymphoblast, while eosinophils did not carry this fusion gene. This indicates that the eosinophil expansion seems to be reactive rather
Next, comparing GEP of the ETV6::ACSL6 ALL with previously published RNA-seq datasets of ALL,22,23 we found that the ETV6::ACSL6 ALL was clustered with eosinophils, mono-nuclear cells from a healthy BM, and GM12878 cells, but distinct from a well-studied ETV6 translocation, ET-V6::RUNX1 fusion, suggesting a distinct mechanism of ET-V6::ACSL6 in promoting malignancy transformation (Figure 1E). We further analyzed the top 10 genes that were differentially expressed (also enriched on chr5 and chr12) in the ETV6::ACSL6 ALL and other ALL subtypes. Interestingly, genes adjacent to the breakpoint of chr5 were significantly up-regulated, including P4HA2, SLC22A5, ACSL6, I L-3 and IL-5 (13 Kb, 300 Kb, 40 Kb, 7 Kb and 480 Kb from the breakpoint, respectively), while genes distant from ETV6 on chr12, such as PRH1 and APOLD1 (480 Kb and 1.1 Mb from the breakpoint, respectively), were down-regulated (Figure 1F). This indicates that the perturbations in the ALL transcriptome not only resulted from the potential intragenic SE of ETV6 promoting genes on chr5, but also from the inactivation of genes on chr12 with the deprivation of ETV6 regulation.
Multi-omics analysis of IL-3 activation in ETV6::ACSL6 acute lymphoblastic leukemia
To further delineate the genetic basis of the fusion gene, we performed long-read sequencing on ALL cells using Oxford Nanopore Technologies (ONT). Both RNA-seq (Figure 2A) and ONT-seq (Figure 2B) demonstrated a breakpoint at the first intron of ETV6. RNA-seq identified a fusion of the 1st exon of ETV6 with the 2nd exon of ACSL6 in mRNA (Figure 2A, C). However, instead of a DNA break at the ACSL6 locus, the ONT data revealed a breakpoint at the intergenic region of ACSL6 and I L- 3 on chr5 (Figure 2B). These data indicate that the fusion of ETV6 to ACSL6 revealed by RNA-seq may be due to alternative splicing which skipped the 1st exon of ACSL (Figure 2D). We also detected genetic aberrations at various genomic regions, the functions of which are yet to be defined (Figure 2E, Online Supplementary Table S1, S2, Online Supplementary Figure S2).
The two derivative chromosomes in ETV6::ACSL6 ALL, i.e., an ETV6::ACSL6 (EA) strand and an IL3::ETV6 (IE) strand, are illustrated in Online Supplementary Figure S3 and verified by RT-PCR in Online Supplementary Figure S4A. Analyzing RNA-seq data, we identified a frameshift mutation leading to a premature stop codon (Online Supplementary Figure S4B). To confirm the result, we ectopically expressed a full-length EA transcript in Nalm6 (pre-B ALL) cells with a flag peptide at the end of ACSL6 (Online Supplementary Figure S4C). As a control, a wild-type ETV6-Flag vector was also transduced, which demonstrated an overexpression of ETV6 and flag proteins (ETV6-OE) (Online Supplementary Figure S4C). However, while expression of the EA transcript was verified by PCR (Online Supplementary Figure S4D), neither a size shift of the ETV6 protein nor the expression of flag was detected (Online Supplementary Figure S4E). In summary, ETV6::ACSL6 does not give rise to a fusion protein, consistent with previous reports.6
Given the scarcity of patient samples, we performed single cell multi-omics sequencing (scNMT-seq) on ALL cells and eosinophils (Online Supplementary Figure S5A). UMAP reduction of scRNA data revealed two clusters: a majority ALL cluster expressing cancer-associated genes like ZFP36L2, SF3B1, and ARHGDIB,24-26 and a smaller cluster comprising eosinophils and some ALL cells (Figure 2F, Online Supplementary Figure S5B). Gene set enrichment analysis (GSEA) revealed differentially expressed genes between ALL and eosinophils enriched in hypoxia, unfolded protein response, and DNA repair (p53) pathways, which are reported to be frequently activated in tumors27 (Figure 2G). Performing scNMT-seq in different subtypes showed that the IL-3 promoter (orange shading) revealed less enriched methylated-CpG and higher enriched methylated-GpC (accessibility) in the ETV6::ACSL6 ALL (RJ-9) compared to another ALL (RJ-10) with a normal karyotype (Figure 2H, Online Supplementary Figure S5C). The dot plots demonstrated CpG and GpC methylation status in each cell indicating a highly activated I L- 3 in ETV6::ACSL6 ALL single cells. Moreover, eosinophils exhibited less GpC methylation (accessibility) and higher CpG methylation at the I L- 3 locus than the ETV6::ACSL6 ALL cells (Online Supplementary Figure S5D). Taken together, our data suggest that the ETV6 translocation altered epigenetic features at genomic regions beyond its fusion partner at chr5.
Furthermore, we extracted enrichments of methylated-CpG and -GpC at promoters for individual cells. Integrating DNA methylation (mCpG), accessibility (GpCm), and RNA transcription through multi-omics factor analysis (MOFA) revealed 3 clusters (Online Supplementary Figure S5E), highlighting the epigenetic heterogeneity. mRNA contributed less to cluster identification compared to DNA methylation and chromatin accessibility (5%, 41% and 46% variance in top 5 MOFA factors) (Online Supplementary Figure S5F). We then profiled the top 50 differentially methylated regions, identifying two leukemia cells with higher chromatin accessibility and hypomethylation in genes such as TCF12, LIN52 (Online Supplementary Figure S5G) that were reported to promote tumor progression.28,29 Enrichment analysis revealed involvement of metabolic and energetic pathways (Online Supplementary Figure S5H), suggesting a connection between metabolic perturbance with epigenetic changes in ETV6::ACSL6 ALL.
Overall, our data suggest that the ETV6 translocation induced critical epigenetic changes at the gene locus adjacent to the breakpoint on the 2 derivative chromosomes; however, mechanisms causing the abnormal epigenetics remain to be identified.
Chromatin structural variation induced by ETV6 super-enhancer translocation
Next, we performed SE analysis to explore enhancer activities in diverse cell types. Extracting cell-type specific SE from the SEdb database, encompassing lymphoid, myeloid and other non-hematopoietic cell types, we observed distinctive blood-specific SE, notably the ETV6 locus (Figure 3A). This suggests that ETV6 serves not only as a crucial transcription factor, but also as an indispensable intragenic hematopoiesis-specific SE in both lymphoid and myeloid cells. In order to validate our findings in ETV6::ACSL6 ALL, we analyzed H3K4me1 (primed and active enhancers), H3K-27ac (activated enhancers), BRD4 and p300 enrichments at the ETV6 locus (Figure 3B). Prominent enrichments of H3K27ac and H3K4me1 were identified at the ETV6 locus, with BRD4, the reader of H3K27ac, also present in this region. The enriched H3K27ac was also observed in B-ALL with normal karyotype (ALL-50 from our previous studies22) and cell lines from ENCODE database,30 suggesting the ETV6 intragenic region functions as an SE in cis-regulatory machinery. Using the ROSE algorithm,31 the 200 Kb (chr12:11718965-11902194) region was recognized as a highly-confident SE (Figure 3C). Interestingly, compared to Nalm6, an ALL cell line with ETV6::PDGFR fusion, REH with ETV6::RUNX1 showed decreased H3K27ac enrichment at the ETV6 locus (Figure 3D), indicating a diminished SE activity in REH. Therefore, the activity of the intragenic SE seems to vary in different ETV6 fusions.
The ETV6 locus was split into 2 sections: Pro-SE1 (green rectangle) including the ETV6 promoter plus a minor SE on the left side; SE2 (blue rectangle) representing the major SE on the right side (Figure 3E). To verify the enhancer activities of 2 sections, we selected a 3Kb region (orange shading) from each section based on transcription factor (TF) binding density and chromatin accessibility (DNase I Hypersensitive Site, DHS) from the ENCODE database and inserted them into a luciferase reporter vector (Figure 3F). While both Pro-SE1- and SE2-inserted vectors revealed significantly enhanced luminescence compared to control, the SE2-inserted vector demonstrated a much more prominent luminescence signal indicating an inherited SE activity (Figure 3G). Motif analysis revealed potential binding sites for TF related to lymphocyte-development / malignancy at the SE, such as Smad3, PAX5 and others (Figure 3H). Overall, the 2 sections of the ETV6 locus derived from the translocation event maintain enhancer activities, indicating their potential role in regulating ALL genes.
Super-enhancers are clusters of enhancers tightly interacting with multiple adjacent or distal genes.32 Using high-throughput chromosome conformation capture (Hi-C), we investigated mechanisms of ETV6-SE interacting with target genes on a genome-wide scale. ETV6::ACSL6 ALL displayed a butterfly morphology at the intersection of chr5 and chr12 in the Hi-C heatmap (dotted circles in Figure 4A), whereas GM12878 showed blank, confirming translocation only in RJ-9. Next, using a web-based genome browser,33 we observed strong interactions between Pro-SE1 and ACSL6, enhancing ACSL6 transcription and H3K27Ac modification in RJ-9 (green dashed lines in Figure 4B). SE2 interacted with multiple downstream genes of ACSL6, including strong interactions with I L- 3, P4HA2 and a slight interaction with IL-5 (blue dashed lines in Figure 4B). Consistent with their normally silent state in lymphocytes, GM12878 displayed no active signals for these genes (Figure 4C). Hi-C contact intensities revealed the formation of new topologically associated domains (TAD) spanning the breakpoints with CTCF binding at boundaries, which constrained regulatory activities of Pro-SE1 and SE2, and restricted their target genes (Figure 4D). Neoloop finder identified 69 interchromosomal neoloops on IE and 7 on EA,34 suggesting uneven activities of Pro-SE1 and SE2 in regulating genes (Online Supplementary Figure S6). To validate the regulatory effects of the newly formed TAD, we compared gene expression profiles within and outside of the TAD. ETV6::ACSL6 ALL revealed an upregulation of genes located within the TAD compared to GM12878, whereas genes outside of the TAD did not exhibit significant expression perturbations (Figure 4E). Furthermore, the sustained activity of ETV6 was also observed in the patient sample with ETV6::RUNX1, where the translocated ETV6 exhibited strong interactions with its partner, RUNX1.35 In ETV6::ACSL6 ALL, among all genes that are activated due to enhancer hijacking, IL-3 and IL-5 are supposed to promote eosinophilia in patients; furthermore, P4HA2, a proline hydrolase, has been shown to be associated with poor prognosis in diffuse large B-cell lymphoma.36 Studying P4HA2 expression in our previously reported ALL patients37 by RNA-seq and qPCR, we found that patients with high expression of P4HA2 had lower survival rates (Online Supplementary Figure S7), indicating that P4HA2 dysregulation by SE2 may contribute to the poor prognosis of these patients. Overall, our data suggest that hijacking the 2 sections derived from ETV6-SE altered 3D genomic organization at new locations, triggering gene transcription exclusively in the ETV6::ACSL6 ALL and led to its unique clinical characteristics.
Bromodomain and extraterminal domain inhibitors partially reversed gene dysregulation in ETV6::ACSL6 acute lymphoblastic leukemia
The biological function of SE is often mediated by bromodomain proteins like BRD4 that recognizes highly-enriched acetylated histones at the SE. The bromodomain inhibitor JQ1 has been shown to disrupt enhancer functions, with more pronounced effect on SE.38 Performing in vitro treatment of JQ1 on a patient-derived xenograft (PDX) of the ETV6::ACSL6 ALL, 1,163 genes were significantly up-regulated and 1,925 down-regulated, including a decrease in IL-3 (fold change: 0.535887, P<0.001) and a mild decrease in IL-5 (Figure 5A, B). Notably, MYC and BCL2 were also down-regulated in agreement with previous reports on JQ1-induced gene regulation.39 GSEA revealed inhibited cytokine and Myc-targets pathways (Figure 5C). RT-qPCR confirmed that exposing cells to JQ1 decreased I L- 3 expression in a time- and concentration-dependent manner (Figure 5D). Consistent with this observation, IL-3 concentration in the medium of ETV6::ACSL6 PDX cell culture decreased after JQ1 treatment; in contrast, ETV6::RUNX1 cells showed no IL-3 in the medium before and after JQ1 treatment (Figure 5E). As a control, Jurkat cells were pre-stimulated by phytohemagglutinin and phorbol myristate acetate to activate IL3 expression and followed with JQ1 treatment for 12 hours. RT-qPCR showed a decrease of 50% in IL3 mRNA after JQ1 treatment, which was significantly lower than the IL3 decrease in the ETV6::ACSL6 ALL (Online Supplementary Figure S8). Since the IL3 expression in Jurkat cells without an SE translocation is thought to be regulated by a regular enhancer at IL3 upstream;40 this suggests a stronger dependency on BRD4 by SE. Moreover, BRD4 binding was substantially reduced at the whole-genome level and the ETV6 locus upon JQ1 treatment (Figure 5F-H). Consistent with previous reports,39 JQ1 altered the genome-wide BRD4 distribution, decreasing at promoters and increasing at intergenic regions (Figure 5 I). Therefore, JQ1 holds promise for dampening gene dysregulation in ETV6::ACSL6 ALL.
Cytotoxicity of bromodomain and extraterminal domain inhibitor treatment on ETV6::ACSL6 acute lymphoblastic leukemia in vitro and in vivo
We then assessed the cytotoxic effects of JQ1 on ET-V6::ACSL6 ALL in vitro and in vivo. Treatment with JQ1 at concentrations >100nM significantly reduced cell viability in vitro, with limited effect on other genetic lesions (Figure 6A, Online Supplementary Table S3). JQ1 was then tested in combination with standard-of-care drugs against the ETV6::ACSL6 ALL. Our data demonstrated synergistic effects of JQ1 with vincristine and doxorubicin, and an additive effect with dexamethasone in vitro, with ZIP scores of 24.9, 25.0, and 6.88, respectively (Figure 6B, Online Supplementary Figure S9A). Notably, while vincristine or doxorubicin alone achieved 50-70% inhibition of cell viability, the addition of JQ1 (1 μM) substantially decreased the IC50 and reduced cell viability to around 10% (Figure 6C, Online Supplementary Figure S9B). Furthermore, we tested 5 other commonly-used BET inhibitors: ABBV-744, Birabresib, I-BET151, Mivebresib and PFI-1, and observed their synergistic cytotoxicity with vincristine in treating ETV6::ACSL6 ALL (Table 1, Online Supplementary Figure S9C). Next, in vivo treatments were performed as shown in Figure 6D. RT-qPCR revealed a significant decrease in IL-3 expression following 24-hour in vivo treatment of JQ1 and vincristine (Figure 6E). The combination treatment significantly inhibited tumor growth in spleen, BM and peripheral blood at day 28, compared to single-drug treatment with vincristine or JQ1 (Figure 6F, Online Supplementary Figure S10A, B). Additionally, we observed that the spleens of the mice in the combination group were dramatically reduced in size after 28 days of treatment compared to those treated with vincristine alone (Online Supplementary Figure S10C). One-week post-treatment, the mice treated with vincristine experienced relapse, while the combination group maintained remission for another seven days (Figure 6G). Survival analysis also showed that the combination treatment significantly prolonged the median event-free survival of mice by 9.2 days compared to vincristine alone (Figure 6H). Overall, our data suggest that combining first-line chemotherapy with JQ1 is promising to improve the treatment of ETV6::ACSL6 B-ALL patients.
Discussion
Chromosome translocations often lead to fusion oncogenes, generating fusion proteins interfering with signaling transduction in cancer cells. However, our study confirmed that SV can impact tumor behavior by altering gene expression through enhancer hijacking.34 In ETV6::ACSL6 leukemia, a frameshift-induced stop codon prevents protein formation, rendering the fusion protein theory insufficient for explaining eosinophilia. Furthermore, the eosinophils did not carry ETV6::ACSL6, indicating eosinophilia as a paraneoplastic syndrome. Our study identified an SE at the ETV6 locus and revealed that in ETV6::ACSL6 ALL, the ETV6-SE was split into 2 parts, both exhibiting enhancer activities. This altered chromatin organization in derivative chromosomes, which enhanced the expression of IL-3 and other inflammatory factors, leading to eosinophilia in patients. Our study provides novel insights into cis-regulatory mutation mechanisms associated with this ALL subtype and its clinical complication.
In several ETV6-translocation subtypes, fusion proteins were reported to play a dominant role in oncogenesis, most of which function as transcription factors or kinases.41 For instance, ETV6::RUNX1, constituting 13% of B-ALL, was reported to mediate oncogenic activity as a transcription factor.14 However, mechanisms beyond the trans-regulation by fusion proteins have been poorly studied. In 2021, ETV6 was reported for the first time to contain an intragenic SE triggering expression of its partner gene MN1 in ETV6::MN1 AML.19 In our study, while Hi-C may not be the optimal technique for quantifying enhancer hijacking events, limiting our ability to accurately assess the interactions between regions, it is still useful for detecting new interactions from the translocated SE and target genes. The translocated ETV6 regulated not only its fusion partner ACSL6, but also distant genes such as I L-3, leading to a critical clinical syndrome (eosinophilia) in ALL. Our future work will extend our findings to other ETV6 fusion partners to gain a comprehensive understanding of the cis-regulatory mechanisms in ETV6 ALL subtypes.
Our data highlight cis-regulatory mutations controlling the expression of multiple genes, offering potential targets for precision therapy. IL-3, studied for over 30 years, induces proliferation of various cell types, including eosinophils and malignant hematopoietic cells like AML.42 IL-3 and GM-CSF are crucial in MYC-transduced human hematopoietic cells transitioning to AML, suggesting a crucial role of cytokines in tumorigenesis.43 Furthermore, ACSL6 and a distal gene P4HA2 were also activated by the SE translocation. ACSL6, a long-chain acyl-CoA synthetase, catalyzes long-chain fatty acids.44 Even though the function of ACSL6 in leukemia is not fully understood, its expression was shown to correlate with the prognosis of AML.45 Our preliminary studies indicated the association of P4HA2 with ALL prognosis, which aligns with previous findings in diffuse large B-cell lymphoma.36 However, the oncogenic role of SE-induced gene overexpression in ETV6::ACSL6 ALL remains unclear, and this will be addressed in further studies. Nevertheless, according to the ‘multi-hit’ theory in oncogenesis,46 SE-induced gene dysregulation may cooperate with other genetic variations driving leukemia transformation and progression. Eosinophilia accompanying hematologic malignancies has diverse causes, including neoplastic, reactive, or idiopathic. The WHO 2022 classification includes “Myeloid/Lymphoid neoplasms with eosinophilia” (M/LN-Eo) as a separate category, with gene rearrangements of PDGFRA, PDGFRB and others.47 In M/LN-Eo with clonal eosinophilia, gene rearrangements can be detected in eosinophils and other myeloid/lymphoid cell lineages, suggesting pluripotential hematopoietic progenitor cell origin. Conversely, reactive eosinophilia often results from tumors secreting cytokines, such as IL-3. Examples of such tumors include Hodgkin lymphoma48 and our case of ETV6::ACSL6 ALL. Therefore, it is critical to determine different treatment strategies, considering varied causal mechanisms of eosinophilia in hematologic malignancies. Besides glucocorticoids, the conventional treatment for eosinophilia, other targeted options are being explored. Clonal eosinophilia bearing fusion genes may benefit from tyrosine kinase inhibitors,47 but the choice is limited for reactive eosinophilia, which often causes severe symptoms and hinders primary disease treatment. In ETV6::ACSL6 ALL, with eosinophilia linked to elevated IL-3, drugs targeting IL-3 pathways are considered. Furthermore, BRD4 inhibitors have achieved promising anti-tumor effects in various preclinical studies.49 Since the translocated SE conferring IL-3 elevation was highly enriched with BRD4, its inhibition significantly dampened the SE-induced gene expression, and synergized with first-line chemotherapy drugs in treating ETV6::ACSL6 ALL in vitro and in vivo. This demonstrates the potential of new therapeutic approaches for ETV6::ACSL6 ALL.
Overall, our study reports for the first time that ETV6 translocation led to chromatin structural variations in ALL patients, which resulted in dysregulation of inflammatory factors conferring eosinophilia in ALL. This highlighted the crucial role of enhancer hijacking in oncogenesis and particularly its clinical complications, and provided insights into improving treatment strategies for this ALL subtype with unfavorable prognosis.
Footnotes
- Received August 22, 2023
- Accepted February 2, 2024
Correspondence
Disclosures
The authors have no conflicts of interest to disclose. Any opinions, findings, conclusions or recommendations expressed in this publication do not reflect the views of The Government of the Hong Kong SAR, the Innovation and Technology Commission or the Vetting Committee of the Mainland-Hong Kong Joint Funding Scheme of the Innovation and Technology Fund.
Funding
This research was funded by grants from the National Key R&D Program of China (2022YFE0200100), the Innovation Program of Shanghai Science and Technology Committee (23141903000, 21430711800), the National Natural Science Foundation of China (NSFC 82070144, 82270155, 82070227, 32271165, 82270187), the Mainland-Hong Kong Joint Funding Scheme supported by the Innovation and Technology Commission, the Government of Hong Kong SAR, China (MHP/054/21), the Ideas Grant funding of the National Health and Medical Research Council of Australia (APP1181666), and the Mobility Programme of the Joint Committee of the Sino-German Center for Research Promotion by the NSFC and the Deutsche Forschungsgemeinschaft (M-0337). RBL is supported by a fellowship from The National Health and Medical Research Council of Australia (NHMRC Fellowship APP1157871).
Acknowledgments
The authors thank Prof. Heather Lee (University of Newcastle, Australia) for assistance in establishing the single-cell technique. The authors acknowledge the ENCODE Consortium for generating DNaseI HS datasets and TF ChIP-seq datasets.
References
- Mitelman F, Johansson B, Mertens F. The impact of translocations and gene fusions on cancer causation. Nat Rev Cancer. 2007; 7(4):233-245. Google Scholar
- Li JF, Dai YT, Lilljebjörn H. Transcriptional landscape of B cell precursor acute lymphoblastic leukemia based on an international study of 1,223 cases. Proc Natl Acad Sci U S A. 2018; 115(50):E11711-E11720. Google Scholar
- Chen B, Jiang L, Zhong ML. Identification of fusion genes and characterization of transcriptome features in T-cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A. 2018; 115(2):373-378. Google Scholar
- Rasighaemi P, Ward AC. ETV6 and ETV7: siblings in hematopoiesis and its disruption in disease. Crit Rev Oncol Hematol. 2017; 116(2):106-115. Google Scholar
- Zaliova M, Moorman AV, Cazzaniga G. Characterization of leukemias with ETV6-ABL1 fusion. Haematologica. 2016; 101(9):1082-1093. Google Scholar
- Zhou MH, Gao L, Jing Y. Detection of ETV6 gene rearrangements in adult acute lymphoblastic leukemia. Ann Hematol. 2012; 91(8):1235-1243. Google Scholar
- Bain BJ, Ahmad S. Should myeloid and lymphoid neoplasms with PCM1-JAK2 and other rearrangements of JAK2 be recognized as specific entities?. Br J Haematol. 2014; 166(6):809-817. Google Scholar
- Zimmermannova O, Doktorova E, Stuchly J. An activating mutation of GNB1 is associated with resistance to tyrosine kinase inhibitors in ETV6-ABL1-positive leukemia. Oncogene. 2017; 36(43):5985-5994. Google Scholar
- Su Z, Liu X, Hu W. Myeloid neoplasm with ETV6::ACSl6 fusion: landscape of molecular and clinical features. Hematology. 2022; 27(1):1010-1018. Google Scholar
- Cools J, Mentens N, Odero MD. Evidence for position effects as a variant ETV6-mediated leukemogenic mechanism in myeloid leukemias with a t(4;12)(q11-q12;p13) or t(5;12) (q31;p13). Blood. 2002; 99(5):1776-1784. Google Scholar
- Esnault S, Kelly EA. Essential mechanisms of differential activation of eosinophils by IL-3 compared to GM-CSF and IL-5. Crit Rev Immunol. 2016; 36(5):429-444. Google Scholar
- Patel B, Kang Y, Cui K. Aberrant TAL1 activation is mediated by an interchromosomal interaction in human T-cell acute lymphoblastic leukemia. Leukemia. 2014; 28(2):349-361. Google Scholar
- Huang Y, Mouttet B, Warnatz HJ. The leukemogenic TCF3-HLF complex rewires enhancers driving cellular identity and self-renewal conferring EP300 vulnerability. Cancer Cell. 2019; 36(6):630-644. Google Scholar
- Polak R, Bierings MB, van der Leije CS. Autophagy inhibition as a potential future targeted therapy for ETV6-RUNX1-driven B-cell precursor acute lymphoblastic leukemia. Haematologica. 2019; 104(4):738-748. Google Scholar
- Adnan-Awad S, Kim D, Hohtari H. Characterization of p190-Bcr-Abl chronic myeloid leukemia reveals specific signaling pathways and therapeutic targets. Leukemia. 2021; 35(7):1964-1975. Google Scholar
- Yagasaki F, Jinnai I, Yoshida S. Fusion of TEL/ETV6 to a novel ACS2 in myelodysplastic syndrome and acute myelogenous leukemia with t(5;12)(q31;p13). Genes Chromosomes Cancer. 1999; 26(3):192-202. Google Scholar
- López C, Burkhardt B, Chan JKC. Burkitt lymphoma. Nat Rev Dis Primers. 2022; 8(1):78. Google Scholar
- Rawat VP, Cusan M, Deshpande A. Ectopic expression of the homeobox gene Cdx2 is the transforming event in a mouse model of t(12;13)(p13;q12) acute myeloid leukemia. Proc Natl Acad Sci U S A. 2004; 101(3):817-822. Google Scholar
- Riedel SS, Lu C, Xie HM. Intrinsically disordered Meningioma-1 stabilizes the BAF complex to cause AML. Mol Cell. 2021; 81(11):2332-2348. Google Scholar
- Argelaguet R, Clark SJ, Mohammed H. Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature. 2019; 576(7787):487-491. Google Scholar
- Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G. Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat Protoc. 2017; 12(3):534-547. Google Scholar
- Jing D, Huang Y, Liu X. Lymphocyte-specific chromatin accessibility pre-determines glucocorticoid resistance in acute lymphoblastic leukemia. Cancer Cell. 2018; 34(6):906-921. Google Scholar
- Yu CH, Wu G, Chang CC. Sequential approach to improve the molecular classification of childhood acute lymphoblastic leukemia. J Mol Diagn. 2022; 24(11):1195-1206. Google Scholar
- Fang F, Lu J, Sang X. Super-enhancer profiling identifies novel critical and targetable cancer survival gene LYL1 in pediatric acute myeloid leukemia. J Exp Clin Cancer Res. 2022; 41(1):225. Google Scholar
- Han C, Khodadadi-Jamayran A, Lorch AH. SF3B1 homeostasis is critical for survival and therapeutic response in T cell leukemia. Sci Adv. 2022; 8(3):eabj8357. Google Scholar
- Zhu J, Tian Z, Li Y. ATG7 promotes bladder cancer invasion via autophagy-mediated increased ARHGDIB mRNA stability. Adv Sci (Weinh). 2021; 8(22):e2104365. Google Scholar
- Zhang Y, Wang S, Zhang J. Elucidating minimal residual disease of paediatric B-cell acute lymphoblastic leukaemia by single-cell analysis. Nat Cell Biol. 2022; 24(2):242-252. Google Scholar
- Zhang Y, Yu R, Li Q. SNHG1/miR-556-5p/TCF12 feedback loop enhances the tumorigenesis of meningioma through Wnt signaling pathway. J Cell Biochem. 2020; 121(2):1880-1889. Google Scholar
- Luo Z, Li X, Zhao Z, Yang X, Xiao S, Zhou Y. MicroRNA-146a affects the chemotherapeutic sensitivity and prognosis of advanced gastric cancer through the regulation of LIN52. Oncol Lett. 2017; 13(3):1386-1392. Google Scholar
- The ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012; 489(7414):57-74. Google Scholar
- Whyte WA, Orlando DA, Hnisz D. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell. 2013; 153(2):307-319. Google Scholar
- Hnisz D, Abraham BJ, Lee TI. Super-enhancers in the control of cell identity and disease. Cell. 2013; 155(4):934-947. Google Scholar
- Wang Y, Song F, Zhang B. The 3D Genome Browser: a web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol. 2018; 19(1):151. Google Scholar
- Wang X, Xu J, Zhang B. Genome-wide detection of enhancer-hijacking events from chromatin interaction data in rearranged genomes. Nat Methods. 2021; 18(6):661-668. Google Scholar
- Mallard C, Johnston MJ, Bobyn A. Hi-C detects genomic structural variants in peripheral blood of pediatric leukemia patients. Cold Spring Harb Mol Case Stud. 2022; 8(1):a006157. Google Scholar
- Jiang W, Zhou X, Li Z. Prolyl 4-hydroxylase 2 promotes B-cell lymphoma progression via hydroxylation of Carabin. Blood. 2018; 131(12):1325-1336. Google Scholar
- Gao M, Wang J, Rousseaux S. Metabolically controlled histone H4K5 acylation/acetylation ratio drives BRD4 genomic distribution. Cell Rep. 2021; 36(4):109460. Google Scholar
- Filippakopoulos P, Qi J, Picaud S. Selective inhibition of BET bromodomains. Nature. 2010; 468(7327):1067-1073. Google Scholar
- Lovén J, Hoke HA, Lin CY. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell. 2013; 153(2):320-334. Google Scholar
- Baxter EW, Mirabella F, Bowers SR. The inducible tissue-specific expression of the human IL-3/GM-CSF locus is controlled by a complex array of developmentally regulated enhancers. J Immunol. 2012; 189(9):4459-4469. Google Scholar
- De Braekeleer E, Douet-Guilbert N, Morel F, Le Bris MJ, Basinko A, De Braekeleer M. ETV6 fusion genes in hematological malignancies: a review. Leuk Res. 2012; 36(8):945-961. Google Scholar
- Aldoss I, Clark M, Song JY, Pullarkat V. Targeting the alpha subunit of IL-3 receptor (CD123) in patients with acute leukemia. Hum Vaccin Immunother. 2020; 16(10):2341-2348. Google Scholar
- Bulaeva E, Pellacani D, Nakamichi N. MYC-induced human acute myeloid leukemia requires a continuing IL-3/GM-CSF costimulus. Blood. 2020; 136(24):2764-2773. Google Scholar
- Rossi Sebastiano M, Konstantinidou G. Targeting long chain Acyl-CoA synthetases for cancer therapy. Int J Mol Sci. 2019; 20(15):3624. Google Scholar
- Chen WC, Wang CY, Hung YH, Weng TY, Yen MC, Lai MD. Systematic analysis of gene expression alterations and clinical outcomes for long-chain acyl-coenzyme A synthetase family in cancer. PLoS One. 2016; 11(5):e0155660. Google Scholar
- Gilliland DG, Tallman MS. Focus on acute leukemias. Cancer Cell. 2002; 1(5):417-420. Google Scholar
- Shomali W, Gotlib J. World Health Organization-defined eosinophilic disorders: 2022 update on diagnosis, risk stratification, and management. Am J Hematol. 2022; 97(1):129-148. Google Scholar
- Francischetti IMB, Alejo JC, Sivanandham R. Neutrophil and eosinophil extracellular traps in Hodgkin lymphoma. Hemasphere. 2021; 5(9):e633. Google Scholar
- Guo J, Zheng Q, Peng Y. BET proteins: biological functions and therapeutic interventions. Pharmacol Ther. 2023; 243:108354. Google Scholar
- Ianevski A, Giri AK, Aittokallio T. SynergyFinder 2.0: visual analytics of multi-drug combination synergies. Nucleic Acids Res. 2020; 48(W1):W488-W493. Google Scholar
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.