Next Article in Journal
Metacyclogenesis as the Starting Point of Chagas Disease
Next Article in Special Issue
Metabolic Profiling as an Approach to Differentiate T-Cell Acute Lymphoblastic Leukemia Cell Lines Belonging to the Same Genetic Subgroup
Previous Article in Journal
PSEN1 His214Asn Mutation in a Korean Patient with Familial EOAD and the Importance of Histidine–Tryptophan Interactions in TM-4 Stability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

ETV6::ABL1-Positive Myeloid Neoplasm: A Case of a Durable Response to Imatinib Mesylate without Additional or Previous Treatment

by
Maria Teresa Bochicchio
1,*,
Giovanni Marconi
2,
Carmen Baldazzi
3,
Lorenza Bandini
3,
Francesca Ruggieri
1,4,
Alessandro Lucchesi
2,
Claudio Agostinelli
4,5,
Elena Sabattini
5,
Agnese Orsatti
5,
Anna Ferrari
1,
Giorgia Capirossi
1,
Chiara Servili
1,
Andrea Ghelli Luserna di Rorà
6,
Giovanni Martinelli
7,
Giorgia Simonetti
1 and
Gianantonio Rosti
2,*
1
Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy
2
Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy
3
Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy
4
Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40100 Bologna, BO, Italy
5
Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy
6
Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme, PI, Italy
7
Scientific Directorate, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 118; https://doi.org/10.3390/ijms25010118 (registering DOI)
Submission received: 27 October 2023 / Revised: 8 December 2023 / Accepted: 18 December 2023 / Published: 21 December 2023
(This article belongs to the Special Issue Hematological Malignancies: Molecular Mechanisms and Therapy)

Abstract

:
ETV6::ABL1 rearranged neoplasms are rare hematological diseases. To date, about 80 cases have been reported, including myeloid and lymphoid leukemias. The ETV6 gene codes for an ETS family transcription factor and several fusion partners have been described. When translocated, ETV6 causes the constitutive activation of the partner genes. Here, we report the case of a 54-year-old woman with a cryptic insertion of the 3′ region of ABL1 in the ETV6 gene. The patient was first diagnosed with idiopathic hypereosinophilic syndrome, according to the clinical history, conventional cytogenetics, standard molecular analyses and pathologist description. Next generation sequencing of diagnosis samples unexpectedly detected both ETV6::ABL1 type A and B fusion transcripts, which were then confirmed by FISH. The diagnosis was Myeloid/Lymphoid neoplasm with ETV6::ABL1 fusion, and the patient received imatinib mesylate treatment. In a follow-up after more than one year, the patient still maintained the molecular and complete hematological responses. This case highlights the importance of timely and proper diagnostics and prompt tyrosine kinase inhibitor treatment.

1. Introduction

ETV6::ABL1 (also known as TEL::ABL1) is a rare fusion that has been found in different types of hematological diseases. To date, about 80 cases of hematological neoplasms (more frequently, acute lymphoblastic leukemia (ALL), myeloproliferative neoplasms (MPNs), Philadelphia-negative chronic myeloid leukemia (CML) [1], atypical (a)CML and chronic myelomonocytic leukemia (CMML)) carrying ETV6::ABL1 translocation have been reported [2,3,4,5,6]. ETV6::ABL1 chimeric protein has been found in less than 1% of ALL cases; however, due to the lack of a systematic screening for this fusion transcript, its overall incidence in hematological neoplasms cannot be precisely defined [2]. The ETV6 (ETS variant 6) gene encodes for an ETS family transcription factor containing two functional domains: the N-terminus PNT (exons 3–4) and the C-terminus ETS (exons 6–7) domains, flanking the central domain coded by the exon 5 [7]. ETV6 is involved in the maintenance of the vascular network, hematogenesis, embryogenesis and development of different tissues [7]. The ABL1 gene encodes for a non-receptor tyrosine kinase containing different structural domains, including SRC-homology domains (SH1, SH2 and SH3) responsible for the regulation of their own activity, DNA-binding (DB) domains and actin-binding (AB) domains, in addition to a nuclear translocation signal (NTS) sequence, sites for phosphorylation by protein kinase C (PKC) and a proline-rich sequence [8]. Two types of ETV6::ABL1 in-frame fusion isoforms have been described: the so-called “type A”, involving exon 4 of ETV6 and exon 2 of ABL1, and the “type B” translocation, involving exon 5 of ETV6 and exon 2 of ABL1. The ETV6::ABL1 fusion protein retains both the SH domains and the tyrosine kinase domain of ABL1 [9], leading to the loss of ABL1 autoinhibitory activity, thus resulting in a constitutive active enzyme. Both transcripts encode for a chimeric non-receptor tyrosine kinase resembling the BCR::ABL1 structure. Moreover, in vitro studies have demonstrated that ETV6::ABL1 phosphorylates the same substrates activated by BCR::ABL1 chimeric proteins, suggesting that ETV6 may replace the BCR role and activate ABL1 [10], and explaining why patients harboring ETV6::ABL1 translocation are sensitive to TKI treatment [9]. Here, we present the case of a patient who received a diagnosis of Myeloid/Lymphoid neoplasm with ETV6::ABL1 fusion and reached a durable response by imatinib mesylate treatment.

2. Case Description

In January 2019, a 54-year-old Caucasian woman was referred to our institution for leukocytosis. She had a mild increase in white blood cell (WBC) count over one year (mean WBC 12 × 109/L, mean neutrophils 8 × 109/L), basophilia (5%) and eosinophilia (14%). JAK2, CALR and MPL mutations and BCR::ABL1 rearrangements were negative (peripheral blood). The patient was asymptomatic and was not receiving any chronic treatment. She had no significant medical history, no history of smoking, no ongoing infections, negative inflammation markers, a normal chest and abdomen examination, and a normal abdomen ultrasonography (US). The patient underwent a regular follow-up (every 4 months) and did not receive any treatment. In the following 18 months, the WBC counts fluctuated around 11–13 × 109/L and the clinical patients’ conditions were stable. In June 2020, the WBC count was raised to 32 × 109/L (basophilia 4%, eosinophilia 11%), while hemoglobin and platelets count were within the normal range, spleen was not palpable and the abdomen US results were normal. BCR-ABL1 translocation and JAK2, CALR, and MPL mutations were confirmed to be negative (peripheral blood). Furthermore, no PDGFRA, PDGFRB or FGFR1 rearrangements were detected. A trephine biopsy showed hypercellular bone marrow (95%) with a diffuse eosinophilic infiltration, slightly reduced erythropoiesis, normal CD34+ cells and mastocytes (Figure 1). Cytogenetic examination showed a unique clone characterized by 47, XX, +12 (on 20 metaphases, Figure 2A). Chest X-ray and heart US excluded any significant organ involvement. Therefore, the patient was disagosed with idiopathic hypereosinophilic syndrome.
Next generation sequencing (NGS) was performed on the diagnosis peripheral blood sample, on both DNA and RNA, using the Oncomine Myeloid Assay (Thermo Fisher Scientific, Waltham, MA, USA), surprisingly revealing the presence of both ETV6::ABL1 type A and B fusion transcripts (Figure 2B and Table S1), while confirming the absence of the Philadelphia chromosome. The ETV6::ABL1 fusions were confirmed by RT-PCR (Figure 2C) and Sanger sequencing (Figure 2D,E). No additional DNA variants were found. Further information on the sample collection, nucleic acids isolation, library preparation and RT-PCR are reported in the Supplementary Methods. The variants and fusions tested are reported in Tables S2–S4.
In order to confirm the fusion transcripts revealed by NGS, we performed fluorescent in situ hybridization (FISH). FISH analysis using an ETV6 break-apart probe revealed three copies of the ETV6 gene without evidence of ETV6 rearrangement (Figure 3A). On the contrary, FISH analysis with a BCR::ABL1 Tricolor Color Dual Fusion (TCDF) probe confirmed the presence of the ABL1 rearrangement with ABL located on one chromosome 12 (Figure 3B). In order to confirm the ETV6::ABL1 fusion, we performed FISH analysis combining ETV6::RUNX1 ES Dual Color Dual Fusion and BCR::ABL1 TCDF. FISH analysis showed the presence of the ETV6::ABL1 fusion on chromosome 12 (Figure 3C). Therefore, we concluded that the ETV6::ABL1 fusion was the result of a cryptic insertion of the 3′ of ABL1 (q34) into the ETV6 locus (12p13). Details on chromosome banding analysis (CBA) and FISH are described in the Supplementary Methods. Based on the results, the diagnosis was modified to Myeloid/Lymphoid neoplasm with ETV6::ABL1 fusion, and the patient started imatinib mesylate treatment at the dose of 200 mg QD (8 September 2020).
We monitored the most abundant fusion transcript, ETV6exon5::ABL1 exon2, using both RT-PCR and Nested PCR. Follow-up peripheral blood samples were collected and analyzed at 3 months (time point B), at 6 months (time point C), at 12 months (time point D) and at 18 months (time point E) of therapy, according to clinical practice. RT-PCR results were negative in all the follow-up samples (Figure 4A). Negativity was confirmed by NGS at time points B and C using the Myeloid Plus Solution panel (Sophia Genetics). Nested-PCR negativity was reached at time point D, while samples at time points B and C were still weakly positive (1/2 replicates, Figure 4B). The complete hematological response was assessed by blood count, revealing all the parameters to be within the normal range. The last evaluation—after 3 years of imatinib mesylate treatment—showed a WBC count of around 5 × 109/L and a neutrophils count of 3.70 × 109/L (basophilia 0.6%, eosinophilia 2.4%). The main clinical and laboratory information are summarized in Figure 5.

3. Discussion

In this study, we report the case of an ETV6::ABL1 rearranged patient with a diagnosis of Myeloid/Lymphoid neoplasm, who received imatinib mesylate treatment and achieved a durable response.
The formation of an in-frame ETV6::ABL1 fusion gene involves complex genomic rearrangements because ETV6 and ABL1 genes have opposite chromosome orientations. Conventional diagnostic techniques (such as conventional cytogenetics) sometimes fail to detect this rearrangement because of its cryptic nature due to the similar G-banding pattern of the distal long arm of chromosome 9 and the distal short arm of chromosome 12. Moreover, no ready-to-use ETV6::ABL1 FISH probes are commercially available, suggesting that the ETV6-ABL1 fusion may remain undetected in a number of patients [5,11].
In the case we present, the use of next generation sequencing allowed us to overcome the above-mentioned limitations and to detect this fusion transcript, which changed the initial diagnosis of hypereosinophilic syndrome into the proper diagnosis of Myeloid/Lymphoid neoplasm with ETV6::ABL1 fusion.
The patient showed the presence of both type “A” and “B” fusion transcripts, first detected by NGS and then confirmed by RT-PCR and Sanger sequencing. FISH analysis combining ETV6::RUNX1 DCDF and BCR-ABL1 TCDF probes performed on metaphases showed a cryptic insertion of the 3′ region of ABL1 in the ETV6 gene on one chromosome 12, in addition to a signal consistent with trisomy 12.
ETV6::ABL1 rearrangements have been reported in different hematological malignancies and, in particular, in ALL, followed by MPNs and acute myeloid leukemia (AML) [11,12]. Eosinophilia represents a common clinical feature and a hallmark of all ETV6::ABL1 rearranged MPNs cases reported in the literature [2], while the most frequent molecular alterations observed in ALL or in lymphoid blast crisis (LCB) patients are deletions of CDKN2A/CDKN2B, IKZF1 or PAX5 [2].
ETV6::ABL1 rearranged hematological neoplasms share many clinical features with CML. Indeed, the ETV6::ABL1 fusion protein functionally resembles the BCR::ABL1 ones, being characterized by a constitutive activation of the chimeric transcript [13], and sensitivity to both first- [14] and second-generation tyrosine kinase inhibitor (TKI) treatments [15]. For these reasons, the patient received imatinib therapy. Imatinib competitively binds the ABL1 ATP binding site in the ETV6::ABL1 fusion protein by the same mechanism of action described for the BCR::ABL1 protein.
Despite this, in the literature, few patients have received TKI treatment at first manifestation of the disease and/or at the first progression, and most of them died or relapsed/evolved [2]. Schwaab et al. presented data on ETV6::ABL1 rearranged MPN patients that received imatinib, nilotinib or dasatinib after a prior treatment with hydroxyurea and/or cytarabine or intensive chemotherapy. Patients receiving imatinib did not achieve a complete cytogenetic (CCR) or molecular (CMR) response, which was instead obtained by patients under nilotinib or dasatinib treatment [16]. Accordingly, previous reports showed that imatinib allowed only an initial reduction of disease followed by a mild neutrophilia, basophilia and eosinophilia after 6 months, with persistent ETV6::ABL1 positivity in FISH and nested PCR [13], or a transient response followed by transformation into ALL [17]. Moreover, most patients reported in the literature were diagnosed as atypical AML or Ph-like ALL.
Conversely, our patient presented without any blast excess at the diagnosis, and the clinical features were consistent with chronic diseases. She started TKI treatment as a frontline therapy after receiving the correct diagnosis and is still maintaining complete hematological and molecular responses after 36 months of imatinib mesylate. To our knowledge, this is the first case reporting a follow-up longer than one year without any additional [13] or previous chemotherapy treatment [18]. Our data likely rule out the co-occurrence of genomic events accounting for resistance in our patient and suggest a potential driver role for the ETV6::ABL1 rearrangement. Studies accounting for different sensitivity profiles, gene expressions and BCR-ABL1-like signatures are warranted in this case; however, an accurate and multicenter sample collection is needed to meet this endpoint.

4. Conclusions

The detection of the ETV6::ABL1 rearrangement remains difficult, due to its cryptic nature.
A deeper genomic characterization of patients with ETV6::ABL1 fusion transcripts may improve our understanding of the biological complexity behind this disease. Although this could represent a limitation of our study, overall, our results underline the importance of timely and proper diagnostics, and the need to revise the current screening algorithms—for example, by recommending NGS RNA panels—in order to detect and monitor ETV6::ABL1 rearrangements. In this case, molecular biology was instrumental in the diagnosis and, consequently, in the selection of an appropriate therapy. Notably, in the absence of molecular biology results, supportive therapies would have been the only ones administered to the patient.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25010118/s1. References [19,20,21] are cited in the supplementary materials.

Author Contributions

M.T.B. and G.R. drafted the manuscript and prepared figures; M.T.B., C.B., L.B., A.F., G.C., A.G.L.d.R., F.R., C.A., E.S. and A.O. conducted molecular biology, cytogenetic experiments and anatomic pathology evaluation; C.S., G.R., G.M. (Giovanni Marconi) and A.L. collected patient samples and clinical data; M.T.B., G.S., G.M. (Giovanni Marconi) and G.R. conceived, designed and interpreted the data; M.T.B. and G.R. supervised the project; G.S. provided the funding; M.T.B., G.M. (Giovanni Marconi), G.M. (Giovanni Martinelli), G.S. and G.R. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partly supported by the contribution of Ricerca Corrente by the Italian Ministry of Health within the research line “Precision, gender and ethnicity-based medicine and geroscience: genetic-molecular mechanisms in the development, characterization and treatment of tumors” for IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, Meldola (FC), Italy. Part of the research activities by Carmen Baldazzi were supported by the Italian Ministry of Health, Ricerca Finalizzata GR-2018-12365278, within the expected project experimental work.

Institutional Review Board Statement

This study was approved by Comitato Etico della Romagna (protocol 5805/2019, #NCT04298892) and was carried out in accordance with the principles outlined in the 1964 Declaration of Helsinki.

Informed Consent Statement

Written informed consent was received from the patient prior to inclusion in the study. Moreover, written consent to publish the information contained in this article was also obtained from the patient.

Data Availability Statement

All data generated or analyzed during this study are included in this published article and in its Supplementary Materials.

Acknowledgments

The authors acknowledge Nicoletta Testoni and Emanuela Ottaviani from IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna (BO), Italy for their useful discussion on cytogenetics and molecular data.

Conflicts of Interest

Maria Teresa Bochicchio received honoraria as consultant from Werfen, Instrumentation Laboratory, S.p.A. Giovanni Marconi received honoraria as a consultant from/participant in a speaker bureau of Abbvie, Astellas, Immunogen, Menarini-Stemline, Pfizer, Ryvu, Servier, Syros and Takeda and received research support from Abbvie, Astellas, Astrazeneca, Jazz, Pfizer. Giovanni Martinelli has competing interests with Novartis, BMS, Roche, Pfizer, ARIAD and MSD. Gianantonio Rosti participated in a speaker bureau of Novartis, Incyte, BMD and Pfizer; and is on steering committees and advisory boards (Novartis, Pfizer, Incyte). All the other authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. De Braekeleer, E.; Douet-Guilbert, N.; Morel, F.; Le Bris, M.J.; Basinko, A.; De Braekeleer, M. ETV6 Fusion Genes in Hematological Malignancies: A Review. Leuk. Res. 2012, 36, 945–961. [Google Scholar] [CrossRef] [PubMed]
  2. Zaliova, M.; Moorman, A.V.; Cazzaniga, G.; Stanulla, M.; Harvey, R.C.; Roberts, K.G.; Heatley, S.L.; Loh, M.L.; Konopleva, M.; Chen, I.M.; et al. Characterization of Leukemias with ETV6-ABL1 Fusion. Haematologica 2016, 101, 1082–1093. [Google Scholar] [CrossRef] [PubMed]
  3. Cessna, M.H.; Paulraj, P.; Hilton, B.; Sadre-Bazzaz, K.; Szankasi, P.; Cluff, A.; Patel, J.L.; Hoda, D.; Toydemir, R.M. Chronic Myelomonocytic Leukemia with ETV6-ABL1 Rearrangement and SMC1A Mutation. Cancer Genet. 2019, 238, 31–36. [Google Scholar] [CrossRef] [PubMed]
  4. Metzgeroth, G.; Steiner, L.; Naumann, N.; Lübke, J.; Kreil, S.; Fabarius, A.; Haferlach, C.; Haferlach, T.; Hofmann, W.K.; Cross, N.C.P.; et al. Myeloid/Lymphoid Neoplasms with Eosinophilia and Tyrosine Kinase Gene Fusions: Reevaluation of the Defining Characteristics in a Registry-Based Cohort. Leukemia 2023, 37, 1860–1867. [Google Scholar] [CrossRef] [PubMed]
  5. Qi, Z.; Smith, C.; Shah, N.P.; Yu, J. Complex Genomic Rearrangements Involving ETV6::ABL1 Gene Fusion in an Individual with Myeloid Neoplasm. Genes 2023, 14, 1851. [Google Scholar] [CrossRef] [PubMed]
  6. Choi, S.I.; Jang, M.A.; Jeong, W.J.; Jeon, B.R.; Lee, Y.W.; Shin, H.B.; Hong, D.S.; Lee, Y.K. A Case of Chronic Myeloid Leukemia with Rare Variant Etv6/Abl1 Rearrangement. Ann. Lab. Med. 2017, 37, 77–80. [Google Scholar] [CrossRef] [PubMed]
  7. Hock, H.; Shimamura, A. ETV6 in Hematopoiesis and Leukemia Predisposition. Semin. Hematol. 2017, 54, 98–104. [Google Scholar] [CrossRef] [PubMed]
  8. Amarante-Mendes, G.P.; Rana, A.; Datoguia, T.S.; Hamerschlak, N.; Brumatti, G. BCR-ABL1 Tyrosine Kinase Complex Signaling Transduction: Challenges to Overcome Resistance in Chronic Myeloid Leukemia. Pharmaceutics 2022, 14, 215. [Google Scholar] [CrossRef]
  9. Xie, W.; Wang, S.A.; Hu, S.; Xu, J.; Medeiros, L.J.; Tang, G. Myeloproliferative Neoplasm with ABL1/ETV6 Rearrangement Mimics Chronic Myeloid Leukemia and Responds to Tyrosine Kinase Inhibitors. Cancer Genet. 2018, 228, 41–46. [Google Scholar] [CrossRef]
  10. Okuda, K.; Golub, T.R.; Gilliland, D.G.; Griffin, J.D. P210BCR/ABL, P190BCR/ABL, and TEL/ABL Activate Similar Signal Transduction Pathways in Hematopoietic Cell Lines. Oncogene 1996, 13, 1147–1152. [Google Scholar]
  11. Tirado, C.A.; Siangchin, K.; Shabsovich, D.S.; Sharifian, M.; Schiller, G. A Novel Three-Way Rearrangement Involving ETV6 (12p13) and ABL1 (9q34) with an Unknown Partner on 3p25 Resulting in a Possible ETV6-ABL1 Fusion in a Patient with Acute Myeloid Leukemia: A Case Report and a Review of the Literature. Biomark. Res. 2016, 4, 16. [Google Scholar] [CrossRef] [PubMed]
  12. La Starza, R.; Trubia, M.; Testoni, N.; Ottaviani, E.; Belloni, E.; Crescenzi, B.; Martelli, M.F.; Flandrin, G.; Giuseppe Pelicci, P.; Mecucci, C. Clonal Eosinophils Are a Morphologic Hallmark of ETV6/ABL1 Positive Acute Myeloid Leukemia. Haematologica 2002, 87, 789–794. [Google Scholar] [PubMed]
  13. Gancheva, K.; Virchis, A.; Howard-Reeves, J.; Cross, N.C.; Brazma, D.; Grace, C.; Kotzampaltiris, P.; Partheniou, F.; Nacheva, E. Myeloproliferative Neoplasm with ETV6-ABL1 Fusion: A Case Report and Literature Review. Mol. Cytogenet. 2013, 6, 39. [Google Scholar] [CrossRef] [PubMed]
  14. Okuda, K.; Weisberg, E.; Gilliland, D.G.; Griffin, J.D. ARG Tyrosine Kinase Activity Is Inhibited by STI571. Blood 2001, 97, 2440–2448. [Google Scholar] [CrossRef] [PubMed]
  15. Nand, R.; Bryke, C.; Kroft, S.H.; Divgi, A.; Bredeson, C.; Atallah, E. Myeloproliferative Disorder with Eosinophilia and ETV6-ABL Gene Rearrangement: Efficacy of Second-Generation Tyrosine Kinase Inhibitors. Leuk. Res. 2009, 33, 1144–1146. [Google Scholar] [CrossRef] [PubMed]
  16. Schwaab, J.; Naumann, N.; Luebke, J.; Jawhar, M.; Somervaille, T.C.P.; Williams, M.S.; Frewin, R.; Jost, P.J.; Lichtenegger, F.S.; La Rosée, P.; et al. Response to Tyrosine Kinase Inhibitors in Myeloid Neoplasms Associated with PCM1-JAK2, BCR-JAK2 and ETV6-ABL1 Fusion Genes. Am. J. Hematol. 2020, 95, 824–833. [Google Scholar] [CrossRef] [PubMed]
  17. Kakadia, P.M.; Schmidmaier, R.; Völkl, A.; Schneider, I.; Huk, N.; Schneider, S.; Panzner, G.; Neidel, U.; Fritz, B.; Spiekermann, K.; et al. An ETV6-ABL1 Fusion in a Patient with Chronic Myeloproliferative Neoplasm: Initial Response to Imatinib Followed by Rapid Transformation into ALL. Leuk. Res. Rep. 2016, 6, 50–54. [Google Scholar] [CrossRef] [PubMed]
  18. Barbouti, A.; Ahlgren, T.; Johansson, B.; Höglund, M.; Lassen, C.; Turesson, I.; Mitelman, F.; Fioretos, T. Clinical and Genetic Studies of ETV6/ABL1-Positive Chronic Myeloid Leukaemia in Blast Crisis Treated with Imatinib Mesylate. Br. J. Haematol. 2003, 122, 85–93. [Google Scholar] [CrossRef]
  19. Righi, S.; Novero, D.; Godio, L.; Bertuzzi, C.; Bacci, F.; Agostinelli, C.; Sagramoso, C.; Rossi, M.; Piccioli, M.; Gazzola, A.; et al. Myeloid Nuclear Differentiation Antigen: An Aid in Differentiating Lymphoplasmacytic Lymphoma and Splenic Marginal Zone Lymphoma in Bone Marrow Biopsies at Presentation. Hum. Pathol. 2022, 124, 67–75. [Google Scholar] [CrossRef]
  20. Jeon, M.J.; Yu, E.S.; Kim, D.S.; Choi, C.W.; Kim, H.N.; Ah Kwon, J.; Yoon, S.Y.; Yoon, J. Performance Evaluation and Clinical Impact of the Oncomine Myeloid Research Assay for Gene Expression Analysis in Myeloid Haematologic Malignancies. J. Clin. Pathol. 2022, 76, 778–783. [Google Scholar] [CrossRef]
  21. Simonetti, G.; Padella, A.; do Valle, I.F.; Fontana, M.C.; Fonzi, E.; Bruno, S.; Baldazzi, C.; Guadagnuolo, V.; Manfrini, M.; Ferrari, A.; et al. Aneuploid Acute Myeloid Leukemia Exhibits a Signature of Genomic Alterations in the Cell Cycle and Protein Degradation Machinery. Cancer 2019, 125, 712–725. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Morphological characterization. Hematoxylin and eosin (H&E, 200×) and Giemsa stain (GM, magnification 200×; GM1, magnification 400×); immunohistochemistry (IHC) stains for CD34 and CD117 (magnification 200×) and Gomori stain for reticulin fibers (Reticulin, magnification 200×).
Figure 1. Morphological characterization. Hematoxylin and eosin (H&E, 200×) and Giemsa stain (GM, magnification 200×; GM1, magnification 400×); immunohistochemistry (IHC) stains for CD34 and CD117 (magnification 200×) and Gomori stain for reticulin fibers (Reticulin, magnification 200×).
Ijms 25 00118 g001
Figure 2. Cytogenetic and molecular characterization. (A) G-banded karyotype with trisomy involving the chromosome 12 (red arrow). (B) Overview of the ETV6::ABL1 fusion transcripts detected by NGS and involving exon 5 of ETV6 and exon 2 of ABL1, or exon 4 of ETV6 and exon 2 of ABL1, respectively. (C) RT-PCR revealing the 706 bp ETV6exon5::ABL1exon2 fusion transcript (upper band) and the 160 bp ETV6exon4::ABL1exon2 fusion transcript (lower band). A 100 bp molecular weight marker was used. (D,E) Electropherograms of the sequences spanning the breakpoint confirming in-frame fusions involving ETV6 exon 5 or ETV6 exon 4 and ABL1 exon 2. The dashed lines indicate the breakpoint regions. Each peak represents a single nucleotide in the DNA sequence, and each nucleotide has a different colour; A is green, T is red, C is blue and G is black. MM: Molecular Marker; A: diagnosis; B: 3 months follow-up; C: 6 months follow-up; D: 12 months follow-up; E: RT negative; F: PCR negative.
Figure 2. Cytogenetic and molecular characterization. (A) G-banded karyotype with trisomy involving the chromosome 12 (red arrow). (B) Overview of the ETV6::ABL1 fusion transcripts detected by NGS and involving exon 5 of ETV6 and exon 2 of ABL1, or exon 4 of ETV6 and exon 2 of ABL1, respectively. (C) RT-PCR revealing the 706 bp ETV6exon5::ABL1exon2 fusion transcript (upper band) and the 160 bp ETV6exon4::ABL1exon2 fusion transcript (lower band). A 100 bp molecular weight marker was used. (D,E) Electropherograms of the sequences spanning the breakpoint confirming in-frame fusions involving ETV6 exon 5 or ETV6 exon 4 and ABL1 exon 2. The dashed lines indicate the breakpoint regions. Each peak represents a single nucleotide in the DNA sequence, and each nucleotide has a different colour; A is green, T is red, C is blue and G is black. MM: Molecular Marker; A: diagnosis; B: 3 months follow-up; C: 6 months follow-up; D: 12 months follow-up; E: RT negative; F: PCR negative.
Ijms 25 00118 g002
Figure 3. FISH analyses (100× magnification). (A) FISH analysis with ETV6 break-apart on previously G-banded metaphase showing 3 fusion signals on 3 chromosomes 12, indicating 3 copies of ETV6. (B) FISH analysis with BCR-ABL1 TCDF probes on previously G-banded metaphase showing 2 green signals on chromosome 22, two blue/red signals on chromosome 9 and an extra red signal on the short arm of chromosome 12, indicating ABL1 rearrangement. (C) FISH analysis combining ETV6-RUNX1 DCDF and BCR-ABL1 TCDF probes on metaphase showing a fusion between ETV6 marked in spectrum green and ABL1 marked in spectrum orange on derivative chromosome 12, confirming ETV6::ABL1 rearrangement. The arrows indicate the derivatives chromosome.
Figure 3. FISH analyses (100× magnification). (A) FISH analysis with ETV6 break-apart on previously G-banded metaphase showing 3 fusion signals on 3 chromosomes 12, indicating 3 copies of ETV6. (B) FISH analysis with BCR-ABL1 TCDF probes on previously G-banded metaphase showing 2 green signals on chromosome 22, two blue/red signals on chromosome 9 and an extra red signal on the short arm of chromosome 12, indicating ABL1 rearrangement. (C) FISH analysis combining ETV6-RUNX1 DCDF and BCR-ABL1 TCDF probes on metaphase showing a fusion between ETV6 marked in spectrum green and ABL1 marked in spectrum orange on derivative chromosome 12, confirming ETV6::ABL1 rearrangement. The arrows indicate the derivatives chromosome.
Ijms 25 00118 g003
Figure 4. Monitoring of ETV6exon5::ABL1exon2 fusion overtime on peripheral blood samples. (A) RT-PCR revealing the 421-bp ETV6exon5-ABL1exon2 fusion transcript. (B) Nested PCR revealing the 321-bp ETV6exon5::ABL1exon2 fusion transcript. A 100 bp molecular weight marker was used. (C) Electropherogram of the sequence spanning the breakpoint that confirmed in-frame fusion involving ETV6 exon 5 and ABL1 exon 2. The dashed lines indicate the breakpoint regions. Each peak represents a single nucleotide in the DNA sequence, and each nucleotide has a different colour; A is green, T is red, C is blue and G is black. MM: Molecular Marker; A: diagnosis; B: 3 months follow-up; C: 6 months follow-up; D: 12 months follow-up; E: 18 months follow-up; F: RT negative; G: PCR negative; H: Nested-PCR negative.
Figure 4. Monitoring of ETV6exon5::ABL1exon2 fusion overtime on peripheral blood samples. (A) RT-PCR revealing the 421-bp ETV6exon5-ABL1exon2 fusion transcript. (B) Nested PCR revealing the 321-bp ETV6exon5::ABL1exon2 fusion transcript. A 100 bp molecular weight marker was used. (C) Electropherogram of the sequence spanning the breakpoint that confirmed in-frame fusion involving ETV6 exon 5 and ABL1 exon 2. The dashed lines indicate the breakpoint regions. Each peak represents a single nucleotide in the DNA sequence, and each nucleotide has a different colour; A is green, T is red, C is blue and G is black. MM: Molecular Marker; A: diagnosis; B: 3 months follow-up; C: 6 months follow-up; D: 12 months follow-up; E: 18 months follow-up; F: RT negative; G: PCR negative; H: Nested-PCR negative.
Ijms 25 00118 g004
Figure 5. Timeline displaying clinical and therapy schedule (upper part) and laboratory data (bottom part).
Figure 5. Timeline displaying clinical and therapy schedule (upper part) and laboratory data (bottom part).
Ijms 25 00118 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bochicchio, M.T.; Marconi, G.; Baldazzi, C.; Bandini, L.; Ruggieri, F.; Lucchesi, A.; Agostinelli, C.; Sabattini, E.; Orsatti, A.; Ferrari, A.; et al. ETV6::ABL1-Positive Myeloid Neoplasm: A Case of a Durable Response to Imatinib Mesylate without Additional or Previous Treatment. Int. J. Mol. Sci. 2024, 25, 118. https://doi.org/10.3390/ijms25010118

AMA Style

Bochicchio MT, Marconi G, Baldazzi C, Bandini L, Ruggieri F, Lucchesi A, Agostinelli C, Sabattini E, Orsatti A, Ferrari A, et al. ETV6::ABL1-Positive Myeloid Neoplasm: A Case of a Durable Response to Imatinib Mesylate without Additional or Previous Treatment. International Journal of Molecular Sciences. 2024; 25(1):118. https://doi.org/10.3390/ijms25010118

Chicago/Turabian Style

Bochicchio, Maria Teresa, Giovanni Marconi, Carmen Baldazzi, Lorenza Bandini, Francesca Ruggieri, Alessandro Lucchesi, Claudio Agostinelli, Elena Sabattini, Agnese Orsatti, Anna Ferrari, and et al. 2024. "ETV6::ABL1-Positive Myeloid Neoplasm: A Case of a Durable Response to Imatinib Mesylate without Additional or Previous Treatment" International Journal of Molecular Sciences 25, no. 1: 118. https://doi.org/10.3390/ijms25010118

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop