Next Article in Journal
Whole-Exome Sequencing of Pakistani Consanguineous Families Identified Pathogenic Variants in Genes of Intellectual Disability
Next Article in Special Issue
Case Report: Rare IKZF1 Gene Fusions Identified in Neonate with Congenital KMT2A-Rearranged Acute Lymphoblastic Leukemia
Previous Article in Journal
Analysis of Nuclear DNA Content and Karyotype of Phaseolus vulgaris L.
Previous Article in Special Issue
Status and Prospects of Fluorescence In Situ Hybridization Automation—A Survey among Laboratory Directors and Their Designates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Acute Promyelocytic Leukemia with Rare Genetic Aberrations: A Report of Three Cases

1
Genetics/Genomics Division, Sonora Quest Laboratories, University of Arizona College of Medicine, Phoenix, AZ 85034, USA
2
AmeriPath Indiana, Indianapolis, IN 46219, USA
3
AmeriPath Northeast, Shelton, CT 06484, USA
4
Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
5
Department of Pathology, University of Arizona College of Medicine, Sonora Quest Laboratories, Phoenix, AZ 85034, USA
*
Authors to whom correspondence should be addressed.
Genes 2023, 14(1), 46; https://doi.org/10.3390/genes14010046
Submission received: 30 November 2022 / Revised: 12 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Genomic Aberrations in Hematologic Malignancies)

Abstract

:
Acute promyelocytic leukemia (APL) is a unique subtype of acute myeloid leukemia (AML) that is characterized by the PML::RARA fusion or, more rarely, a variant RARA translocation. While APL can be clinically suspected, diagnosis of APL requires genetic confirmation. Targeted therapy such as all-trans-retinoic acid (ATRA) and arsenic trioxide (ATO) has dramatically improved the prognosis of APL patients, but this is dependent on timely genetic testing as different fusions and/or mutations can affect therapeutic outcomes. Here we report three APL cases with various genetic aberrations: cryptic PML::RARA fusion, variant RARA rearrangement, and typical PML::RARA fusion with co-existing FLT3-ITD mutation. They serve to illustrate the utility of integrating genetic testing, using chromosome analysis, fluorescence in situ hybridization (FISH), reverse transcriptase-polymerase chain reaction (RT-PCR), and next-generation sequencing (NGS) in providing a detailed understanding of the genetic alterations underlying each patient’s disease.

1. Introduction

Acute promyelocytic leukemia (APL) is a subset of acute myeloid leukemia (AML) in which abnormal promyelocytes predominate. It accounts for 5–8% of AML cases. The disease can occur at any age but mainly affects middle-aged adults [1,2]. APL cells may contain Auer rods and kidney-shaped or bilobed nuclei, along with high-level myeloperoxidase expression. The genetic hallmark of APL is the fusion of the RARA gene at chromosome 17q21.2 with either the PML gene at chromosome 15q24.1 or, rarely, another partner gene. Other chromosomal aberrations, such as 7q deletion and trisomy 8, can also be present. Alterations in FLT3, such as an internal tandem duplication (FLT3-ITD), tyrosine kinase domain mutation (FLT3-TKD), and others, have been reported in APL cases [3,4,5]. APL immunophenotypic characteristics include high side scatter, forward scatter, negative expression of CD34 and HLA-DR, and positive expression of CD13, CD33, and CD117 [6,7]. An important distinguishing feature of APL is the frequent association with disseminated intravascular coagulation (DIC), leading to a high rate of mortality and morbidity if not diagnosed and treated immediately [8]. Targeted therapy with all-trans retinoic acid (ATRA) in conjunction with arsenic trioxide (ATO) has fortunately reduced the risk of hemorrhagic complications and transformed APL into a highly curable neoplasm. Therefore, the rapid and accurate establishment of APL diagnosis is critical for timely and effective treatment [9].
While APL can be suspected based on clinical presentation, cellular morphology, and immunophenotypic features, its diagnosis requires genetic confirmation of PML::RARA fusion or variant RARA rearrangement. Most Cytogenetics laboratories perform FISH with probes for PML and RARA for rapid detection of PML::RARA fusion, followed by FISH with break-apart probes for RARA if no fusion is identified, but rather three signals for RARA are evident. Chromosome analysis is also performed to evaluate for the (15;17) translocation [or a less common (v;17) translocation] associated with APL, and to identify any additional karyotypic complexity [10,11]. Reverse transcriptase polymerase chain reaction (RT-PCR), or real-time quantitative PCR, primarily used in subsequent analyses to assess disease status, can also be utilized on initial diagnosis if the cytogenetics testing is negative but clinical suspicion of APL is high. NGS assays with RNA-seq capability, such as the NGS Oncomine Myeloid Assay used in our lab, can detect PML::RARA fusion and identify variant RARA rearrangements as well as uncover other molecular alterations.
Here we report three cases of APL that demonstrate the utility of comprehensive laboratory testing in rendering an accurate diagnosis and in providing a better understanding of the genetic profile underlying each patient’s disease.
Case 1
The patient is a 32-year-old female with night sweats, pancytopenia, decreased platelets, and low fibrinogen (57 mg/dL). Laboratory data showed the following abnormal findings: WBC = 2535/µL; RBC = 2.7 million cells/µL; Hgb = 8.5 gm/dL; Hct = 24.1%; and platelets = 13 K/µL. Her blood smear consisted predominantly of normochromic, normocytic red blood cells with scattered nucleated red blood cells, mild anisocytosis, and decreased granulocytes. Abnormal promyelocytes were occasionally identified. Her platelets were markedly decreased. No fibrin strands or platelet clumps were observed.
Her bone marrow aspirate was cellular and paucispicular. The H&E- and Giemsa-stained particle sections were composed of blood clots. The H&E- and Giemsa-stained decalcified biopsy was adequate and markedly hypercellular. The cellularity was >95%, and the myeloid-to-erythroid ratio was indeterminate. Most of the cells were abnormal promyelocytes and represented >90% of nucleated cells. The cells had reniform nuclei and variable amounts of granulation. Auer rods and APL cells were observed. Granulocytic differentiation to the neutrophil stage was not significant. Monocytic differentiation to the monocyte stage was not evident. Erythropoiesis and megakaryopoiesis were decreased. No background dyspoiesis was evident. Lymphocytes were not increased. No granulomas were identified. Iron stains of the aspicular aspirate and decalcified biopsy showed no stainable reticuloendothelial iron stores.
Immunohistochemistry for CD34 showed no interstitial increase or clustering. Flow cytometry showed that the aspirate contained 84% blast cells with a dim CD45(+)/CD34(−)/CD117(+)/CD13(+)/CD33(+)[99%]/HLA-DR(−) and myeloperoxidase (99.3%) immunophenotype indicative of acute myeloid leukemia and suggestive of acute promyelocytic leukemia. Fluorescence in situ hybridization (FISH) analysis was performed on bone marrow aspirate with the PML::RARA dual-color dual-fusion probe set (MetaSystems, Medford, MA, USA). Two hundred nuclei were examined, the results were negative for PML::RARA rearrangement. Subsequent analysis with RARA break-apart probes (XL RARA BA; Metasystems) was also negative for RARA rearrangement. Chromosome analysis revealed a normal female karyotype (Figure 1A). NGS Oncomine Myeloid Assay (Thermo Fisher Scientific, Waltham, MA, USA) was also performed, which detected PML::RARA fusion and a WT1 mutation (Figure 1B). RT-PCR was concurrently performed and revealed the long-form transcript of PML::RARA fusion at a transcript level of 1478.933 with ABL1 as the control gene. This assay detects the short form (bcr3), the long form (bcr1), and the variant exon 6 (bcr2) PML::RARA transcripts (Quest Diagnostics, San Juan Capistrano, CA, USA). Based on the molecular test results, FISH was re-evaluated. Enhancing probe signal strength with Metafer 4 imaging software (Version 4.1.5; Meta-Systems) allowed for a fusion signal with a very faint signal for RARA (green) to be evident on the long arm of chromosome 15. These FISH results demonstrate that the PML::RARA fusion in this patient has resulted from a cytogenetically cryptic insertion of RARA into the PML locus on 15q (Figure 1C,D).
Case 2
A 51-year-old male presented to the clinic due to fatigue and easy bruising. A morphologic examination of the peripheral blood smear demonstrated some immature/blast-like cells. These cells were of predominantly intermediate to large size with oval to folded/convoluted nuclei, fine nuclear chromatin, small nucleoli, and scanty, finely granular cytoplasm. Examination of the bone marrow aspirate smears revealed adequate cellularity for analysis. Granulocytic precursors consisted almost entirely of blasts with virtually no neutrophils. Erythroid precursors were markedly decreased but showed no significant morphologic changes. A differential count showed 78% blasts, 0% promyelocytes, 2% myelocytes, 2% metamyelocytes, 7% granulocytes, 1% monocytes, 0% eosinophils, 0% basophils, 7% erythroid cells, 3% lymphocytes, and 0% plasma cells. The M:E ratio was 12.7. Megakaryocytes were decreased. Prussian blue stain revealed decreased iron staining. Flow cytometry demonstrated 79% blasts that were CD13(+), CD33(+), CD117(+), CD34(−), CD14(−), CD64(−) and HLA-DR(−). A diagnosis of AML with immunophenotypic features suggestive of APL was rendered.
FISH was performed on bone marrow with the PML::RARA dual-color dual-fusion probe set (Abbott Diagnostics, Abbott Park, IL, USA). Two hundred nuclei were examined, and all were negative for PML::RARA rearrangement. However, three signals for the RARA gene were evident in 173/200 (86.5%) of the cells scored (Figure 2A). FISH with RARA dual-color break-apart probes (Abbott Diagnostics, Abbott Park, IL) was subsequently performed, which demonstrated split signals, consistent with rearrangement of RARA in 177/200 (88.5%) of cells scored (Figure 2B). FISH with an AML panel (Abbott Diagnostics, Abbott Park, IL), comprised of probes for RUNX1 (21q22.3) & RUNX1T1 (8q22), BCR (22q11) & ABL1 (9q34.12), KMT2A (11q23), and CBFB (16q22.1), was also performed. Two hundred nuclei were examined for each probe, and the results demonstrated three signals for RUNX1T1 in 68/200 (34.0%) of the cells scored, suggesting a gain of all or part of chromosome 8 (Figure 2C). Chromosome analysis identified a translocation between the long arms of chromosomes 11 and 17 [i.e., t(11;17)(q23;q21)]. Two cells also displayed an additional copy of chromosome 8 (Figure 2D). RT-PCR was negative for PML::RARA fusion (Quest Diagnostics, San Juan Capistrano, CA, USA). NGS analysis with the Oncomine Myeloid Assay (Thermo Fisher Scientific, Waltham, MA, USA) detected ZBTB16::RARA fusion and additional mutations in IDH2, TET2, and SRSF2 genes (Figure 2E).
Case 3
A 26-year-old male presented with a two-week history of fever and bleeding gums. The bone marrow biopsy was hypercellular marrow with left-shifted maturation in the myeloid lineage. The flow cytometry revealed a 77% blast population with dim CD45, bright CD33+, CD34+ (subset), HLA-DR(−), CD13+ (dim), CD117+, CD56+ (subset), and cytoplasmic MPO was positive. The morphology showed an increased nuclear cytoplasmic ratio and bilobed nuclei with sliding plate morphology. The clinicopathological findings were concerning for APL, and thus the clinical team initiated ATRA (all-trans-retinoic acid) treatment while awaiting genetic diagnosis.
FISH was performed on bone marrow aspirate using the PML::RARA dual-color dual-fusion probe set (Abbott Diagnostics, Abbott Park, IL), the results of which were positive for PML::RARA rearrangement in 187/200 (93.5%) of the cells scored (Figure 3A). FISH analysis with an AML panel for RUNX1 (21q22.3 & RUNX1T1 (8q22), BCR (22q11) & ABL1 (9q34.12), KMT2A (11q23), and CBFB (16q22.1) (Abbott) identified three signals for RUNX1T1, suggesting gain of all or part of chromosome 8 in 172/200 (86.0%) of the cells scored (Figure 3B). RT-PCR was also positive for PML::RARA fusion (Quest Diagnostics, San Juan Capistrano, CA). Chromosome analysis detected the (15;17) translocation associated with APL, and trisomy 8 in 15 of the 20 cells examined (Figure 3C). The concurrent NGS Oncomine Myeloid Assay (Thermo Fisher Scientific, Waltham, MA) detected PML::RARA fusion and FLT3-ITD mutation with 35.30% allele frequency (Figure 3D).

2. Discussion

When an APL diagnosis is clinically suspected, the affected patient should be managed as a medical emergency. Providing accurate genetic diagnosis as quickly as possible thus plays a critical role. In some cases, this can be complicated by rare genetic aberrations, such as cryptic PML::RARA rearrangements, variant RARA rearrangements, and/or co-existing targetable mutations such as FLT3. The cases presented in this study illustrate the importance of utilizing various genetic testing to ensure an accurate and comprehensive diagnosis of APL. A summary of genetic testing utilized in all three cases is shown in Table 1.
FISH is utilized to rapidly evaluate for the PML::RARA gene fusion or other RARA rearrangements. However, there are rare instances in which such rearrangements go undetected by this methodology as demonstrated in Case 1. Chromosome analysis provides a global assessment of the entire genome and therefore can identify translocations associated with APL. It also picks up secondary chromosomal alterations such as trisomy 8 and can identify clonal evolution, as in Cases 2 and 3. With this methodology, too, there are instances of rare alterations resulting in PML::RARA fusion or other RARA rearrangements that are not detectable at the resolution limits of this analysis. Such cytogenetically cryptic fusions are only detected by molecular techniques, such as RT-PCR or NGS.
Case 1 illustrates this point. The initial FISH analysis with both PML::RARA dual fusion probes and RARA break-apart probes were negative. Chromosome analysis showed a normal karyotype. NGS and quantitative RT-PCR, however, identified the PML::RARA fusion. Re-evaluation of FISH, upon enhancing probe signal strength, identified a faint signal for RARA colocalizing with signal for PML on the long arm of one chromosome 15, thus confirming PML::RARA rearrangement due to a cytogenetically cryptic insertion of RARA into the PML locus. The oncogenic PML::RARA fusion more often occurs on the derivative chromosome 15, as seen in this case, and less commonly on the derivative chromosome 17 or other chromosomes [12]. Although one report by Zacarria et al. showed that the PML::RARA fusion on 17q may indicate a poor response to ATRA [13], APL patients with cryptic PML::RARA fusion generally respond very well to the targeted therapies (including ATRA and ATO) and share a favorable prognosis, the same as the APL patients with a typical PML::RARA fusion [14,15].
Some cryptic fusions are only detectable by molecular techniques such as RT-PCR or NGS, which offer much higher sensitivity at the RNA level. NGS can also detect DNA mutations such as the WT1 mutation described in Case 1, which has been reported as a recurrent alteration in newly diagnosed APL, and can impact treatment decisions [16]. Therefore, Case 1 illustrates that whenever there is a strong clinical suspicion for APL but negative chromosome analysis and FISH results, it is important to use molecular techniques to ensure that a cryptic PML::RARA fusion is not missed.
Variant RARA translocations involving partner genes other than PML have been reported, including ZBTB16 (previously called PLZF) at 11q23.2, NUMA1 at 11q13.4, NPM1 at 5q35.1, and STAT5B at 17q21.2 [17]. Cases with these variant translocations should be diagnosed as APL with a variant RARA translocation [1]. ZBTB16::RARA translocation, shown in Case 2, is the most common RARA variant translocation reported [18]. In this rearrangement, the RARA gene (17q21) is fused to the zinc finger and BTB domain containing protein 16 (ZBTB16, 11q23), which is also called the PML zinc finger protein (PLZF) gene [18]. Patients with ZBTB16::RARA fusion usually have the typical clinical presentation of APL, and incidence of disseminated intravascular coagulation. However, they often have some morphological variations, such as predominance of cells with regular nuclei, many granules, absence of Auer rods, or increased number of Pelgeroid neutrophils [18,19]. Variant APL with t(11;17)(q23;q21), resulting in ZBTB16::RARA fusion, has been reported as being resistant to ATRA. Therefore, identifying this rearrangement is very important [19]. The FISH strategy employed by most, if not all, labs does not identify a fusion partner. Chromosome analysis can often help elucidate fusion partners involved, as demonstrated in Case 2, as long as the alteration is not cytogenetically cryptic. RT-PCR using primers specific for PML::RARA cannot detect variant fusion partners, as shown in Case 2. Depending on the platform, NGS testing, however, can detect RARA rearrangement with numerous partner genes and multiple types of transcripts as well as identify concurrent mutations in other genes such as IDH2, TET2, and SRSF2 genes in Case 2. Both IDH2 and SRSF2 have been described in the literature as recurrent secondary changes in APL with ZBTB16::RARA [20]. NGS provides comprehensive detection of mutations and variant fusion partners and should be used whenever there is a RARA rearrangement not involving PML.
Table 1. Summary of genetic testing utilized in all three cases.
Table 1. Summary of genetic testing utilized in all three cases.
Case No.KaryotypingFISHRT-PCR
(PML::RARA)
NGS
Case 146,XX [20]Negative initially; cryptic single fusion on 15q upon enhancementPositivePML::RARA fusion
A muation in WT1
Case 246,XY,t(11;17)(q23;q21) [18]/47,idem,+8 [2]Negative for PML::RARA fusion,
Positive for RARA rearrangment
Negative ZBTB16::RARA fusion
Mutations in IDH2, ET2 and SRSF2
Case 347,XY,+8,t(15;17)(q21;q21) [15]/46,XY [5]Positive for PML::RARA fusionPositivePML::RARA fusion
FLT3-ITD
The turnaround time (TAT) for NGS, RT-PCR, and chromosome analysis in many centers is generally more than 5 days. FISH results are often ready in 24 h. The latter is therefore heavily relied upon for timely confirmation of diagnosis and start to treatment.
When the FISH results are negative, NGS testing can be critical. Laboratories continue to work on reducing TAT for NGS. This can be of particularly importance for cases in which FISH and/or chromosome analysis are negative but clinicopathological suspicion for APL is high. NGS can serve as a rapid way of evaluating for an atypical PML::RARA fusion that was not picked up cytogenetically or in identifying the fusion partner in cases with variant RARA rearrangements.
Genetic testing including chromosome analysis, FISH, and NGS performed for Case 3 identified t(15;17) and its associated PML::RARA fusion. Chromosome analysis and FISH also identified the concurrent gain of chromosome 8, the most common secondary change in APL [1], while NGS detected FLT3-ITD mutation, which can help with risk stratification in patients with APL [21,22]. FLT3-ITD mutation with a high variant allele frequency (VAF), associated with PML::RARA gene fusion and trisomy 8, all of which represent a poor prognosis for APL, especially in young patients [23]. Mutations involving FLT3, including FLT3-ITD and FLT3-TKD mutations, occur in 30–40% of APL. Patients with FLT3-ITD mutation are more likely to have significantly higher white blood cell count at diagnosis, higher risk of induction deaths, and lower overall survival rates than those without this mutation [21,22,23,24]. A meta-analysis of 11 publications found that patients with the FLT3-ITD mutation had lower overall survival (OS) rates (RR = 0.59, p < 0.001) and a 1.70-fold higher likelihood of dying of any cause than those without this mutation [24]. One study reported that ATRA-ATO might abrogate the negative prognostic impact of FLT3-ITD [25]. In Case 3, the Mut/WT ratio is 0.563. According to Schlenk et al., an allelic ratio > 0.51 was associated with unfavorable relapse-free and overall survival [26]. This case serves to illustrate the utility of NGS testing in patients with APL, as detection of additional molecular alterations may help stratify risk more precisely and allow for a more individualized approach to treatment.
The treatment of patients with APL is different from those with other AML subtypes. Thanks to advances in the diagnosis and treatment of this disease, APL is currently considered the most curable adult leukemia. There are many treatment options for APL patients, including all-trans retinoic acid (ATRA), arsenic trioxide (ATO), anthracyclines, antimetabolites, stem cell transplantation, and clinical trials. Many studies have demonstrated that the combination of ATO and ATRA is superior to the former standard of care, which includes anthracyclines (daunorubicin and idarubicin), for patients with low-risk APL. For patients with high-risk APL, combinations of ATO, ATRA, and anthracyclines are widely used; the antimetabolite, e.g., cytarabine, can also be added to induction or consolidation regimens. Finally, for patients who have persistent minimal residual disease (MRD), stem cell transplantation and clinical trials are available (https://www.lls.org/treatment-acute-promyelocytic-leukemia, accessed on 1 October 2022). The use of a comprehensive genetic testing approach can help to accurately stratify patients’ risk and ensure that they receive the appropriate therapy.
These cases demonstrate the benefit of using a comprehensive genetic testing approach to identify cryptic PML::RARA fusions that may go undetected by conventional cytogenetics alone (i.e., chromosome analysis and FISH analysis), to characterize variant RARA rearrangements and uncover concurrent molecular alterations and secondary cytogenetic changes that can help guide treatment decisions and improve patient outcome. In addition, close follow-up and monitoring disease progression are recommended for these patients.

Funding

No funding.

Data Availability Statement

Exclude.

Conflicts of Interest

All authors have declared no conflict of interest.

References

  1. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Paulson, K.; Serebrin, A.; Lambert, P.; Bergeron, J.; Everett, J.; Kew, A.; Jones, D.; Mahmud, S.; Meloche, C.; Sabloff, M.; et al. Acute promyelocytic leukaemia is characterized by stable incidence and improved survival that is restricted to patients managed in leukaemia referral centres: A pan-Canadian epidemiological study. Br. J. Haematol. 2014, 166, 660–666. [Google Scholar] [CrossRef] [PubMed]
  3. Melnick, A.; Licht, J.D. Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood 1999, 93, 3167–3215. [Google Scholar] [CrossRef] [PubMed]
  4. Callens, C.; Chevret, S.; Cayuela, J.-M.; Cassinat, B.; Raffoux, E.; de Botton, S.; Thomas, X.; Guerci, A.; Fegueux, N.; Pigneux, A.; et al. Prognostic implication of FLT3 and Ras gene mutations in patients with acute promyelocytic leukemia (APL): A retrospective study from the European APL Group. Leukemia 2005, 19, 1153–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zelent, A.; Guidez, F.; Melnick, A.; Waxman, S.; Licht, J.D. Translocations of the RARalpha gene in acute promyelocytic leukemia. Oncogene 2001, 20, 7186–7203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ferrari, A.; Bussaglia, E.; Úbeda, J.; Facchini, L.; Aventin, A.; Sierra, J.; Nomdedéu, J.F. Immunophenotype distinction between acute promyelocytic leukaemia and CD15- CD34- HLA-DR- acute myeloid leukaemia with nucleophosmin mutations. Hematol. Oncol. 2012, 30, 109–114. [Google Scholar] [CrossRef]
  7. Horna, P.; Zhang, L.; Sotomayor, E.M.; Lancet, J.E.; Moscinski, L.C. Diagnostic immunophenotype of acute promyelocytic leukemia before and early during therapy with all-trans retinoic acid. Am. J. Clin. Pathol. 2014, 142, 546–552. [Google Scholar] [CrossRef] [Green Version]
  8. David, S.; Mathews, V. Mechanisms and management of coagulopathy in acute promyelocytic leukemia. Thromb. Res. 2018, 164 (Suppl. S1), S82–S88. [Google Scholar] [CrossRef]
  9. Shen, Z.X.; Shi, Z.Z.; Fang, J.; Gu, B.W.; Li, J.M.; Zhu, Y.M.; Shi, J.Y.; Zheng, P.Z.; Yan, H.; Liu, Y.F.; et al. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc. Natl. Acad. Sci. USA 2004, 101, 5328–5335. [Google Scholar] [CrossRef] [Green Version]
  10. Kim, M.J.; Yang, J.J.; Meyer, C.; Marschalek, R.; Park, T.S. Molecular methods for genomic analyses of variant PML-RARA or other RARA-related chromosomal translocations in acute promyelocytic leukemia. Korean J. Hematol. 2012, 47, 307–308. [Google Scholar] [CrossRef]
  11. Sanz, M.A.; Fenaux, P.; Tallman, M.S.; Estey, E.H.; Löwenberg, B.; Naoe, T.; Lengfelder, E.; Döhner, H.; Burnett, A.K.; Chen, S.J.; et al. Management of acute promyelocytic leukemia: Updated recommendations from an expert panel of the European LeukemiaNet. Blood 2019, 133, 1630–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Grimwade, D.; Biondi, A.; Mozziconacci, M.J.; Hagemeijer, A.; Berger, R.; Neat, M.; Howe, K.; Dastugue, N.; Jansen, J.; Radford-Weiss, I.; et al. Characterization of acute promyelocytic leukemia cases lacking the classic t(15;17): Results of the European Working Party. Groupe Francais de Cytogenetique Hematologique, Groupe de Francais d’Hematologie Cellulaire, UK Cancer Cytogenetics Group and BIOMED 1 European Community-Concerted Action “Molecular Cytogenetic Diagnosis in Haematological Malignancies”. Blood 2000, 96, 1297–1308. [Google Scholar]
  13. Zaccaria, A.; Valenti, A.; Toschi, M.; Salvucci, M.; Cipriani, R.; Ottaviani, E.; Martinelli, G. Cryptic translocation of PML/RARA on 17q. A rare event in acute promyelocytic leukemia. Cancer Genet. Cytogenet. 2002, 138, 169–173. [Google Scholar] [CrossRef] [PubMed]
  14. Blanco, E.M.; Curry, C.V.; Lu, X.Y.; Sarabia, S.F.; Redell, M.S.; Lopez-Terrada, D.H.; Roy, A. Cytogenetically cryptic and FISH-negative PML/RARA rearrangement in acute promyelocytic leukemia detected only by PCR: An exceedingly rare phenomenon. Cancer Genet. 2014, 207, 48–49. [Google Scholar] [CrossRef] [PubMed]
  15. Karlin, K.; Bryke, C.; Dias, A.; Michaels, P. Cytogenetically cryptic PML::RARA fusion in acute promyelocytic leukemia: Testing strategies in the modern era. Leuk. Res. Rep. 2022, 17, 100320. [Google Scholar] [CrossRef] [PubMed]
  16. Liquori, A.; Ibañez, M.; Sargas, C.; Sanz, M.A.; Barragán, E.; Cervera, J. Acute Promyelocytic Leukemia: A Constellation of Molecular Events around a Single PML-RARA Fusion Gene. Cancers 2020, 12, 624. [Google Scholar] [CrossRef] [Green Version]
  17. Langabeer, S.E.; Preston, L.; Kelly, J.; Goodyer, M.; Elhassadi, E.; Hayat, A. Molecular Profiling: A Case of ZBTB16-RARA Acute Promyelocytic Leukemia. Case Rep. Hematol. 2017, 2017, 7657393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Sainty, D.; Liso, V.; Cantuù-Rajnoldi, A.; Head, D.; Mozziconacci, M.J.; Arnoulet, C.; Benattar, L.; Fenu, S.; Mancini, M.; Duchayne, E.; et al. A new morphologic classification system for acute promyelocytic leukemia distinguishes cases with underlying PLZF/RARA gene rearrangements. Blood 2000, 96, 1287–1296. [Google Scholar]
  19. Mannan, A.; Muhsen, I.N.; Barragán, E.; Sanz, M.A.; Mohty, M.; Hashmi, S.K.; Aljurf, M. Genotypic and Phenotypic Characteristics of Acute Promyelocytic Leukemia Translocation Variants. Hematol. Oncol. Stem Cell Ther. 2020, 13, 189–201. [Google Scholar] [CrossRef] [PubMed]
  20. Pardo Gambarte, L.; Franganillo Suárez, A.; Cornago Navascués, J.; Soto de Ozaeta, C.; Blas López, C.; Atance Pasarisas, M.; Salgado Sánchez, R.N.; Serrano del Castillo, C.; Mata Serna, R.; Velasco-Rodríguez, D.; et al. ZBTB16-RARα-Positive Atypical Promyelocytic Leukemia: A Case Report. Medicina 2022, 58, 520. [Google Scholar] [CrossRef]
  21. Breccia, M.; Loglisci, G.; Loglisci, M.G.; Ricci, R.; Diverio, D.; Latagliata, R.; Foà, R.; Lo-Coco, F. FLT3-ITD confers poor prognosis in patients with acute promyelocytic leukemia treated with AIDA protocols: Long-term follow-up analysis. Haematologica 2013, 98, e161–e163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lucena-Araujo, A.R.; Kim, H.T.; Jacomo, R.H.; Melo, R.A.; Bittencourt, R.; Pasquini, R.; Pagnano, K.; Fagundes, E.M.; Chauffaille, M.D.L.; Chiattone, C.S.; et al. Internal tandem duplication of the FLT3 gene confers poor overall survival in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and anthracycline-based chemotherapy: An International Consortium on Acute Promyelocytic Leukemia study. Ann. Hematol. 2014, 93, 2001–2010. [Google Scholar] [PubMed]
  23. Tripon, F.; Crauciuc, G.A.; Bogliş, A.; Moldovan, V.; Sándor-Kéri, J.; Trifa, A.P.; Bănescu, C. Co-occurrence of PML-RARA gene fusion, chromosome 8 trisomy, and FLT3 ITD mutation in a young female patient with de novo acute myeloid leukemia and early death: A CARE case report. Medicine 2020, 99, e19730. [Google Scholar] [CrossRef] [PubMed]
  24. Picharski, G.L.; Andrade, D.P.; Fabro, A.L.M.R.; Lenzi, L.; Tonin, F.S.; Ribeiro, R.C.; Figueiredo, B.C. The Impact of Flt3 Gene Mutations in Acute Promyelocytic Leukemia: A Meta-Analysis. Cancers 2019, 11, 1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cicconi, L.; Divona, M.; Ciardi, C.; Ottone, T.; Ferrantini, A.; Lavorgna, S.; Alfonso, V.; Paoloni, F.; Piciocchi, A.; Avvisati, G.; et al. PML-RARalpha kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia 2016, 30, 1987–1992. [Google Scholar] [CrossRef] [PubMed]
  26. Schlenk, R.F.; Kayser, S.; Bullinger, L.; Kobbe, G.; Casper, J.; Ringhoffer, M.; Held, G.; Brossart, P.; Luebbert, M.; Salih, H.R.; et al. Differential impact of allelic ratio and insertion site in FLT3-ITD–positive AML with respect to transplantation. Blood 2014, 124, 3441–3449. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chromosome analysis of the bone marrow revealed a normal female karyotype. ISCN: 46,XX (A). NGS detected PML::RARA fusion and a WT1 mutation (B). Initial FISH analysis, which failed to identify the PML::RARA fusion (C). FISH re-evaluation, upon enhancing signal strength, identified a subtle cryptic insertion of RARA (green) into PML (red). Arrows indicate PML::RARA fusion at 15q (D).
Figure 1. Chromosome analysis of the bone marrow revealed a normal female karyotype. ISCN: 46,XX (A). NGS detected PML::RARA fusion and a WT1 mutation (B). Initial FISH analysis, which failed to identify the PML::RARA fusion (C). FISH re-evaluation, upon enhancing signal strength, identified a subtle cryptic insertion of RARA (green) into PML (red). Arrows indicate PML::RARA fusion at 15q (D).
Genes 14 00046 g001
Figure 2. FISH with PML::RARA dual-color dual-fusion probes, negative for PML::RARA fusion, displaying three RARA (green) signals (A). FISH with RARA dual color break-apart probes displaying split signal, indicative of RARA rearrangement (B). FISH with the AML panel exhibiting three red signals for the pericentromeric region of chromosome 8 (C). Chromosome analysis identified gain of chromosome 8 and an (11;17) translocation. ISCN: 47,XY,+8, t(11;17)(q23;q21) (D). NGS analysis detected ZBTB16::RARA fusion and mutations in IDH2, TET2, and SRSF2 genes (E).
Figure 2. FISH with PML::RARA dual-color dual-fusion probes, negative for PML::RARA fusion, displaying three RARA (green) signals (A). FISH with RARA dual color break-apart probes displaying split signal, indicative of RARA rearrangement (B). FISH with the AML panel exhibiting three red signals for the pericentromeric region of chromosome 8 (C). Chromosome analysis identified gain of chromosome 8 and an (11;17) translocation. ISCN: 47,XY,+8, t(11;17)(q23;q21) (D). NGS analysis detected ZBTB16::RARA fusion and mutations in IDH2, TET2, and SRSF2 genes (E).
Genes 14 00046 g002
Figure 3. FISH with PML::RARA dual-color dual-fusion probes identified PML::RARA fusion (A). FISH with the AML panel displayed three RUNX1T1 signals (red), suggesting a gain of chromosome 8 (B). Chromosome analysis showed a (15;17) translocation and concurrent gain of chromosome 8, consistent with the FISH analysis. ISCN: 47,XY,+8,t(15;17)(q21;q21) (C). NGS detected PML::RARA fusion and FLT3-ITD mutation (D).
Figure 3. FISH with PML::RARA dual-color dual-fusion probes identified PML::RARA fusion (A). FISH with the AML panel displayed three RUNX1T1 signals (red), suggesting a gain of chromosome 8 (B). Chromosome analysis showed a (15;17) translocation and concurrent gain of chromosome 8, consistent with the FISH analysis. ISCN: 47,XY,+8,t(15;17)(q21;q21) (C). NGS detected PML::RARA fusion and FLT3-ITD mutation (D).
Genes 14 00046 g003
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

Liu, G.; Liu, L.; Bartolo, D.D.; Li, K.Y.; Li, X. Acute Promyelocytic Leukemia with Rare Genetic Aberrations: A Report of Three Cases. Genes 2023, 14, 46. https://doi.org/10.3390/genes14010046

AMA Style

Liu G, Liu L, Bartolo DD, Li KY, Li X. Acute Promyelocytic Leukemia with Rare Genetic Aberrations: A Report of Three Cases. Genes. 2023; 14(1):46. https://doi.org/10.3390/genes14010046

Chicago/Turabian Style

Liu, Guang, Lanting Liu, Daniel Di Bartolo, Katie Y. Li, and Xia Li. 2023. "Acute Promyelocytic Leukemia with Rare Genetic Aberrations: A Report of Three Cases" Genes 14, no. 1: 46. https://doi.org/10.3390/genes14010046

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