Next Article in Journal
Sickle Cell Anemia and Inflammation: A Review of Stones and Landmarks Paving the Road in the Last 25 Years
Previous Article in Journal
Alloantibody Identification: The Importance of Temperature, Strength Reaction and Enzymes—A Practical Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome

1
Department of Pathology, Medical School, University of Pécs, Szigeti Str. 12, 7624 Pécs, Hungary
2
Department of Internal Medicine, Kaposi Mór Teaching Hospital of Somogy County, Tallián Gy. Str. 20-32, 7400 Kaposvár, Hungary
3
Department of 1st Internal Medicine, Medical School, University of Pécs, Ifjúság Str. 13, 7624 Pécs, Hungary
*
Author to whom correspondence should be addressed.
Hematol. Rep. 2025, 17(1), 1; https://doi.org/10.3390/hematolrep17010001
Submission received: 23 October 2024 / Revised: 19 December 2024 / Accepted: 24 December 2024 / Published: 6 January 2025

Abstract

:
Background: T-cell prolymphocytic leukemia (T-PLL) is a rare mature T-cell lymphoma that is usually associated with poor prognosis and short overall survival. Methods: We present a case of a 61-year-old woman presenting with T-PLL and the leukemic cells harboring BCR::ABL1 (BCR—breakpoint cluster region; ABL1—ABL protooncogene 1) fusion transcripts as the result of a variant of t(9;22)(q34;q11) called Philadelphia translocation: t(9;22;18)(q34;q11;q21). Sequencing revealed a rare BCR transcript with an exon 6 breakpoint corresponding to e6a2 transcripts, which has thus far been reported in only 26 cases of leukemias. Results: After 9 months of follow-up, the disease progressed and required treatment. Following alemtuzumab and chemotherapy, a short course of imatinib therapy stabilized the disease for six months, which was followed by progression and the demise of the patient. Conclusions: To the best of our knowledge, this is the first report of a mature T-cell lymphoma with a variant Philadelphia-translocation and a very rare type of BCR::ABL1 transcript. This case highlights the importance of comprehensive genetic testing of malignancies, as abnormal molecular pathways may be uncovered that may be specifically targeted by drugs.

1. Introduction

The BCR::ABL1 (BCR—breakpoint cluster region; ABL1—ABL protooncogene 1) translocation is one of the most extensively studied abnormalities in tumor cytogenetics, resulting in the iconic Philadelphia chromosome (Ph-chromosome), which is a shortened version of chromosome 22. This translocation is detectable in chronic myeloid leukemia (CML), a proportion of acute lymphoblastic leukemia (ALL), and very rarely in acute myeloid leukemia (AML) cases [1,2]. T-cell lineage involvement is very uncommon, and is restricted to uncommon cases of T-cell ALL and T-cell lymphoblastic crises of CML [3,4,5]. The BCR::ABL1 translocation is not associated with mature lymphoid neoplasms, although a few such cases have been reported, with a single report involving mature T-cell lymphoma [6,7,8].
Although in CML patients the BCR::ABL1 translocation is detectable not only in myeloid cells, but in hematopoietic stem cells and progenitors of both B and T-cells, it is not present in mature T-cells [9]. The explanation of this phenomenon is unclear. It seems that BCR::ABL1+ T-cell progenitors have a reduced differentiation capability and reduced survival [10].
Here, an exceptional case of BCR::ABL1-positive T-cell prolymphocytic leukemia case is reported. A very rare BCR::ABL1 transcript variant was identified in the neoplastic cells, as well as a variant Ph-chromosome with evidence of BCR::ABL1-related clonal evolution. Bone marrow progenitors and myeloid cells were BCR::ABL1-negative, and the abnormality was restricted to the neoplastic T-cells. The patient received various treatments, and the disease was stabilized temporarily while the patient was receiving tyrosine kinase inhibitor therapy; however, the patient later died following progression after 39 months of survival. The objective of this manuscript is to expand the knowledge base of known cancer drivers occurring in unusual clinical contexts by providing a description of this unique T-cell lymphoma case.

2. Case Presentation

In May 2012, a 61-year-old female patient presented with complaints of weakness and itching for six weeks. Splenomegaly and mild leukocytosis (30 G/L) without lymphocytosis, normal platelet count and hemoglobin values were detected, no B symptoms were reported, and LDH (lactate dehydrogenase) was found to be elevated (607 U/L). A mild, generalized lymph node enlargement was demonstrated with imaging. Hepatosplenomegaly was not detected. Informed consent was given by the patient and the study was approved by the institutional review board (IRB), with reference number 6650/PTE-2017.
Peripheral blood smears showed 46% of atypical lymphocytes with slightly angulated, 8–10 µm large nuclei and prominent nucleoli (Figure 1A). Immunophenotyping of peripheral blood and bone marrow aspirates was performed by flow cytometry using a Beckman-Coulter Navios flow cytometer. Peripheral blood and bone marrow showed 44% and 33% of CD2+, CD3+, CD4+, CD5+, CD7+, CD8, CD52+, CD56 T-cells, respectively (Figure 1B). Bone marrow biopsy showed 10% of focal, interstitial lymphoid infiltrate showing CD3, CD4 and TCL1 (T-cell leukemia/lymphoma protein 1, protein product of TCL1A gene) positivity, as well as TDT (terminal deoxynucleotidyltransferase), CD10, and CD34 negativity with normal hematopoiesis in the background (Figure 1C,D). Monoclonal TCRG (T-cell receptor gamma) gene rearrangement was demonstrated using polymerase chain reaction. Next-generation sequencing (NGS) was performed on Novaseq 6000 platform (Illunina, San Diego, CA, USA) using a customized panel of 138 genes involved in myeloid and lymphoid leukemias with a median sequencing depth of 1052X and coverage of 92.4% at 500X depth (Supplementary File S1). No pathogenic variant of the JAK/STAT pathway was detected.
Karyotyping of peripheral blood cells stimulated by phytohemagglutinin for three days, and bone marrow karyotyping after 24 h of culturing and GTG banding revealed a complex karyotype with a typical inv(14)(q11;q32) along with del(6q), i(8q) and a variant Philadelphia translocation—t(9;22;18)(q34;q11;q12)—as well as duplication of the Philadelphia chromosome. All metaphases with clonal alterations (10/30 in peripheral blood and 11/18 in bone marrow) showed del(6q), i(8q), inv(14), as well as t(9;22;18) and two other structural aberrations (Figure 2A). Fluorescence in situ hybridization (FISH) was performed using BCR::ABL1 dual fusion (Abbott-Vysis), as well as TCL1A break apart (Cytocell) probe kits. Fusion of BCR and ABL1 genes (Figure 2B), as well as the disruption of the TCL1A gene, was demonstrated. Myeloid cells and megakaryocytes were found to be BCR::ABL1-negative based on FISH performed on bone marrow biopsy slides. Half of the leukemic cells showed a signal pattern indicating duplication of the Philadelphia chromosome. Following reverse-transcriptase polymerase chain reaction (PCR) using primers for BCR exon 1 and ABL1 exon 2, Sanger sequencing of the transcript revealed an e6a2 BCR::ABL1 transcript indicating an atypical BCR breakpoint (Figure 3). The cytogenetic and molecular findings remained unchanged during follow-up.
Initially, therapy was not started for 9 months due to minimal progression. Then, alemtuzumab therapy was initiated due to significant leukocytosis, night sweats, fever and mild splenomegaly, but no response was observed for 2 months. Cyclophosphamide therapy was started at a dose of 100 mg daily, and a reduction in leukocyte count as well as spleen size was achieved. Three months later, imatinib was initiated at a 400 mg dose. Both leukocyte count and spleen size improved; however, therapy was stopped due to the appearance of skin rashes six months later. No progression was noted during this time. Cyclophosphamide therapy was reinitiated. Twelve months later, a multiagent regimen containing cyclophosphamide, doxorubicin, vincristine and prednisone (CHOP) was started due to an increase in the patient’s spleen size and white blood cell count. Two months later, nilotinib was initiated due to progressive disease, but no significant clinical improvement was seen for four months. Eventually, a massive splenomegaly occurred, and the patient was lost 39 months after the initial diagnosis. Figure 4 summarizes the sequence of therapies (Figure 4).

3. Discussion and Conclusions

The BCR::ABL1 translocation and the resulting fusion protein is an archetype of molecular pathological abnormalities that may be successfully targeted with specific drugs. BCR::ABL1 signaling results in increased survival and proliferation of hematopoietic progenitors, as seen in cases of CML, a proportion of ALL, and rarely in AML [1]. The BCR::ABL1 translocation is not associated with other cancer types, although BCR:ABL1 positivity was reported in three plasma cell myeloma cases [11,12,13], two diffuse large B-cell lymphomas [14,15], one hairy cell leukemia [6], and a single case of Sezáry syndrome transforming into large T-cell lymphoma [7].
The case reported herein is unique for several reasons. It is the first reported case of Philadelphia translocation-positive T-cell prolymphocytic leukemia. T-PLL is a rare, mature, post-thymic T-cell lymphoma with a poor prognosis and short overall survival [16]. The median age at diagnosis is 61 years. Although the clinical course is usually aggressive, with very high WBC count and splenomegaly from the onset of disease, approximately 15% of patients show an indolent phase of variable length, during which patients may be asymptomatic [16,17]. T-PLL cells are characteristically CD2+/CD3+/CD5+/CD7+/CD52+, and 60% of cases show CD4 positivity and CD8 negativity, although co-expression of CD4 and CD8, and rarely, CD4/CD8+ phenotypes is observed [18]. Additionally, 75% of cases show structural abnormalities of chromosome 14 involving the gene TCL1A. Inversion 14 is the most common of these abnormalities [12]. Trisomy or isochromosome 8 is seen in approximately 66% of cases [19].
In addition to the aforementioned characteristics, the neoplastic cells of the reported case showed duplication of the Philadelphia chromosome, a common additional cytogenetic abnormality seen associated with clonal evolution in cases of CML. This aberration indicates the importance of BCR::ABL1 signaling regarding survival and proliferation of the neoplastic T-cells, since the duplication may increase the level of BCR::ABL1 expression and is regarded as a potential cause of resistance to tyrosine kinase inhibitors [1,20].
The BCR::ABL1 fusion gene was the result of a variant Philadelphia translocation involving chromosomes 9, 18, and 22. Such variant translocations are observed in approximately 5–10% of CML cases. Molecular studies revealed a rare BCR::ABL1 transcript (e6a2). This transcript has been reported in less than 30 cases to date; 18 CML, 1 ALL, and 7 AML cases [21,22,23]. It has been suggested that this transcript may be associated with more aggressive clinical behavior in CML [22,24]. The limited number of reported cases does not allow for definitive conclusions to be drawn; however, four out seven patients reported to have received imatinib treatment and follow-up experienced treatment failure.
The patient received only a short course of tyrosine kinase inhibitor (TKI) therapy. Although a complete response was not observed, the patient’s disease stabilized; however, it was not possible to draw conclusions regarding the TKI sensitivity of the neoplastic T-cells. T-PLL is characterized by TCL1A fusion, usually in the form of inv(14), as was detected in the case discussed here. Amplification of 8q is a common secondary finding; complex karyotypes are detected in >70% of cases [25]. Mutations of the JAK/STAT pathway are common alterations which occur in 76% of cases, with JAK3 mutations being most common [26]; however, activation of the Jak/Stat pathway may be detected even without gain-of-function mutations [27]. ATM deletions and mutations, as well as mutations of methyl transferases affecting epigenetic modulation (e.g., KMT2C, KMT2D, KMT5A), are also common [28].
Our case is unique, being the first and, presently, only reported incidence of BCR::ABL1-positive T-PLL. A single case of T-PLL with SEPT9::ABL1 fusion has been reported previously. Other cytogenetic or molecular alterations were not reported in that case, and the patient did not respond to imatinib or dasatinib [29].
T-PLL is a rare, mature T-cell lymphoma with an aggressive clinical course. Typically, patients present with leukocytosis, splenomegaly, and lymphadenopathy, often with anemia and thrombocytopenia. Initially, an indolent period may be detected; however, progression does occur, and the median overall survival is less than 3 years. The current recommended first-line therapy is alemtuzumab, which should be followed by hemopoietic stem cell transplantation if possible. There is no current consensus regarding the best treatment after alemtuzumab failure, which occurs almost universally following a median of 12 months. Purine analogues as monotherapy or in combination are generally recommended in cases of T-PLL [30].
Recent genomic profiling results have led to preclinical studies suggesting that epigenetic modulation by histone deacetylase inhibitors may be a potential therapeutic approach in T-PLL [30]. Antagonists of Mdm2 or Bcl2 family molecules as well as inhibitors of the Jak/Stat pathway show promise in vitro, with few clinical case reports or short case series available [31,32,33,34].
It is not yet clear how BCR::ABL1 affects the survival and proliferation of cells other than stem cells or progenitor cells. The BCR::ABL1 fusion results in constitutive activation of the Jak/Stat pathway, often due to direct phosphorylation of STAT5 [35,36], which may lead to activation of this crucial pathway even without other gain-of-function mutations, The fusion may result in increased proliferation or resistance to apoptosis of cells by activating other signal transduction pathways such as RAS/RAF/MAPK, WNT or PI3K/AKT/mTOR [37]. It is currently unclear why BCR::ABL1 fusion is rare outside the context of ALL and CML despite influencing several crucial signaling pathways. It is possible that genomic alterations may not maintain the same biomarker potential across different diagnoses. Investigating how BCR::ABL1 or other known driver mutations influence signal transduction pathways in unusual diagnoses may lead to a better understanding of the mechanics of targeted therapy, and even to more efficient control of minimal residual disease in BCR::ABL1-positive leukemias. Also, unexpected targetable genetic alterations appearing in T-cell malignancies may provide therapeutic as well as research opportunities, potentially leading to better understanding and thus, better control of these often difficult-to-treat malignancies.
In summary, the case reported here is, thus far, the only BCR::ABL1-positive T-PLL case recorded in the literature. The fusion transcript was rare, corresponding to e6a2 transcripts that have been reported in only 26 cases so far. The findings suggest that BCR::ABL1 fusion was a driver mutation in this case, as no other genetic alteration was noted to activate the Jak/Stat pathway. A single, unique case does not provide sufficient information to draw conclusions regarding the exact biological consequence of BCR::ABL1 fusion in mature T-cell lymphomas and the efficacy of targeted therapy in this context. Appropriate models have to be designed to assess the exact molecular biological role of BCR::ABL1 in the context of mature T-cell neoplasms. Our case demonstrates the crucial importance of karyotyping and comprehensive genomic testing of leukemias, since potential targets of efficient therapies may be uncovered. Continuous expansion of our knowledge base of genetic alterations in cancer is vital for increasing the efficiency of therapy.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/hematolrep17010001/s1, Supplementary File S1: List of next-generation sequencing genes used in the manuscript.

Author Contributions

Conceptualization, L.V. and B.K.; methodology, L.V., B.H., M.E., B.K., and H.A.; data curation, H.A. and B.K.; writing—original draft preparation, L.V. and B.K.; writing—review and editing, L.V., B.H., M.E., B.K. and H.A.; supervision, H.A. and B.K.; funding acquisition, B.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Internal Research Grant of the University of Pécs, Hungary, Grant Number 009_2023_PTE_RK/26 (B.K.). The article processing charge was provided by B.K.

Institutional Review Board Statement

The study was approved by the Regional Research Ethical Committee of University of Pécs, Clinical Centre (6650/PTE-2017).

Informed Consent Statement

Written informed consent was obtained from the patient(s) to publish this paper.

Data Availability Statement

Dataset available on request from the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Quintás-Cardama, A.; Cortes, J. Molecular biology of bcr-abl1–positive chronic myeloid leukemia. Blood 2009, 113, 1619–1630. [Google Scholar] [CrossRef] [PubMed]
  2. Reboursiere, E.; Chantepie, S.; Gac, A.C.; Reman, O. Rare but authentic Philadelphia-positive acute myeloblastic leukemia: Two case reports and a literature review of characteristics, treatment and outcome. Hematol. Oncol. Stem Cell Ther. 2014, 8, 28–33. [Google Scholar] [CrossRef]
  3. Ye, C.C.; Echeverri, C.; Anderson, J.E.; Smith, J.L.; Glassman, A.; Gulley, M.L.; Claxton, D.; Craig, F.E. T-cell blast crisis of chronic myelogenous leukemia manifesting as a large mediastinal tumor. Hum. Pathol. 2002, 33, 770–773. [Google Scholar] [CrossRef] [PubMed]
  4. Lim, L.C.; Heng, K.K.; Vellupillai, M.; Tan, L.T.; Boey, B.C.; Lau, L.C.; How, G.F. Molecular and phenotypic spectrum of de novo Philadelphia positive acute leukemia. Int. J. Mol. Med. 1999, 4, 665–667. [Google Scholar] [CrossRef] [PubMed]
  5. Mongia, A.; Romano, F.; Ciullini Mannurita, S.; Peruzzi, B.; Bencini, S.; Parrini, D.; Fasano, L.; Fanelli, A. A Rare Onset of T-Lymphoid Blast Crisis in Chronic Myeloid Leukemia with Two Distinct Blast Populations. Hematol. Rep. 2024, 16, 413–420. [Google Scholar] [CrossRef] [PubMed]
  6. Won, Y.W.; Kim, S.J.; Park, T.S.; Oh, S.H.; Wan, T.S.; Baek, E.J. De novo hairy cell leukemia with a major BCR/ABL1 rearrangement: A case report with a literature review. Pathol. Int. 2014, 64, 142–147. [Google Scholar] [CrossRef]
  7. Callet-Bauchu, E.; Salles, G.; Gazzo, S.; Dalle, S.; Berger, F.; Hayette, S. Identification of a novel e8/a4 BCR/ABL fusion transcript in a case of a transformed Sézary syndrome. Haematologica 2007, 97, 1277–1278. [Google Scholar] [CrossRef]
  8. Ragupathi, L.; Najfeld, V.; Chari, A.; Petersen, B.; Jagannath, S.; Mascarenhas, J. A case report of chronic myelogenous leukemia in a patient with multiple myeloma and a review of the literature. Clin. Lymphoma Myeloma Leuk. 2013, 13, 175–179. [Google Scholar] [CrossRef]
  9. MacKinney, A.A., Jr.; Clark, S.S.; Borcherding, W.; Fizzotti, M.; Hong, R. Simultaneous demonstration of the Philadelphia chromosome in T, B, and myeloid cells. Am. J. Hematol. 1993, 44, 48–52. [Google Scholar] [CrossRef] [PubMed]
  10. Takahashi, N.; Miura, I.; Saitoh, K.; Miura, A.B. Lineage Involvement of Stem Cells Bearing the Philadelphia Chromosome in Chronic Myeloid Leukemia in the Chronic Phase as Shown by a Combination of Fluorescence-Activated Cell Sorting and Fluorescence In Situ Hybridization. Blood 1998, 92, 4758–4763. [Google Scholar] [CrossRef]
  11. Van Den Berghe, H.; Louwagie, A.; Broeckaert-Van Orshoven, A.; David, G.; Verwilghen, R.; Michaux, J.L.; Sokal, G. Philadelphia chromosome in human multiple myeloma. J. Natl. Cancer Inst. 1979, 63, 11–16. [Google Scholar] [PubMed]
  12. Tanaka, M.; Kimura, R.; Matsutani, A.; Zaitsu, K.; Oka, Y.; Oizumi, K. Coexistence of chronic myelogenous leukemia and multiple myeloma. Case report and review of the literature. Acta Haematol. 1998, 99, 221–223. [Google Scholar] [CrossRef] [PubMed]
  13. Martiat, P.; Mecucci, C.; Nizet, Y.; Stul, M.; Philippe, M.; Cassiman, J.J.; Michaux, J.L.; Van den Berghe, H.; Sokal, G. P190 BCR/ABL transcript in a case of Philadelphia-positive multiple myeloma. Leukemia 1990, 4, 751–754. [Google Scholar]
  14. Mitani, K.; Sato, Y.; Tojo, A.; Ishikawa, F.; Kobayashi, Y.; Miura, Y.; Miyazono, K.; Urabe, A.; Takaku, F. Philadelphia chromosome positive B-cell type malignant lymphoma expressing an aberrant 190 kDa bcr-abl protein. Br. J. Haematol. 1990, 76, 221–225. [Google Scholar] [CrossRef]
  15. Cerretini, R.; Noriega, M.F.; Narbaitz, M.; Slavutsky, I. New chromosome abnormalities and lack of BCL-6 gene rearrangements in Argentinean diffuse large B-cell lymphomas. Eur. J. Haematol. 2006, 76, 284–293. [Google Scholar] [CrossRef]
  16. Dearden, C. How I treat prolymphocytic leukemia. Blood 2012, 120, 538–551. [Google Scholar] [CrossRef] [PubMed]
  17. Garand, R.; Goasguen, J.; Brizard, A.; Buisine, J.; Charpentier, A.; Claisse, J.F.; Duchayne, E.; Lagrange, M.; Segonds, C.; Troussard, X.; et al. Indolent course as a relatively frequent presentation in T-prolymphocytic leukaemia: Groupe Francais d’Hematologie Cellulaire. Br. J. Haematol. 1998, 103, 488–494. [Google Scholar] [CrossRef]
  18. Matutes, E.; Brito-Babapulle, V.; Swansbury, J.; Ellis, J.; Morilla, R.; Dearden, C.; Sempere, A.; Catovsky, D. Clinical and laboratory features of 78 cases of T-prolymphocytic leukaemia. Blood 1991, 78, 3269–3274. [Google Scholar] [CrossRef] [PubMed]
  19. Maljaei, S.H.; Brito-Babapulle, V.; Hiorns, L.R.; Catovsky, D. Abnormalities of chromosomes 8,11,14 and X in T-prolymphocytic leukaemia studied by fluorescence in situ hybridization. Cancer Genet. Cytogenet. 1998, 103, 110–116. [Google Scholar] [CrossRef]
  20. Assouline, S.; Lipton, J.H. Monitoring response and resistance to treatment in chronic myeloid leukemia. Curr. Oncol. 2011, 18, e71–e83. [Google Scholar] [CrossRef] [PubMed]
  21. Hochhaus, A.; Reiter, A.; Skladny, H.; Melo, J.V.; Sick, C.; Berger, U.; Guo, J.Q.; Arlinghaus, R.B.; Hehlmann, R.; Goldman, J.M.; et al. A novel BCR-ABL fusion gene (e6a2) in a patient with Philadelphia chromosome-negative chronic myelogenous leukemia. Blood 1996, 88, 2236–2240. [Google Scholar] [CrossRef] [PubMed]
  22. Piedimonte, M.; Ottone, T.; Alfonso, V.; Ferrari, A.; Conte, E.; Divona, M.; Bianchi, M.P.; Ricciardi, M.R.; Mirabilii, S.; Licchetta, R.; et al. A rare BCR-ABL1 transcript in Philadelphia-positive acute myeloid leukemia: Case report and literature review. BMC Cancer 2019, 19, 50. [Google Scholar] [CrossRef] [PubMed]
  23. Manzella, L.; Tirrò, E.; Vitale, S.R.; Puma, A.; Consoli, M.L.; Tambè, L.; Pennisi, M.S.; DIGregorio, S.; Romano, C.; Tomarchio, C.; et al. Optimal Response in a Patient with CML Expressing BCR-ABL1 E6A2 Fusion Transcript with Nilotinib Therapy: A Case Report. Vivo 2020, 34, 1481–1486. [Google Scholar] [CrossRef]
  24. Colla, S.; Sammarelli, G.; Voltolini, S.; Crugnola, M.; Sebastio, P.; Giuliani, N. e6a2 BCR-ABL transcript in chronic myeloid leukemia: Is it associated with aggressive disease? Haematologica 2004, 89, 611–613. [Google Scholar] [PubMed]
  25. Braun, T.; Dechow, A.; Friedrich, G.; Seifert, M.; Stachelscheid, J.; Herling, M. Advanced Pathogenetic Concepts in T-Cell Prolymphocytic Leukemia and Their Translational Impact. Front. Oncol. 2021, 11, 775363. [Google Scholar] [CrossRef]
  26. Kiel, M.J.; Velusamy, T.; Rolland, D.; Sahasrabuddhe, A.A.; Chung, F.; Bailey, N.G.; Schrader, A.; Li, B.; Li, J.Z.; Ozel, A.B.; et al. Integrated genomic sequencing reveals mutational landscape of T-cell prolymphocytic leukemia. Blood 2014, 124, 1460–1472. [Google Scholar] [CrossRef]
  27. Wahnschaffe, L.; Braun, T.; Timonen, S.; Giri, A.K.; Schrader, A.; Wagle, P.; Almusa, H.; Johansson, P.; Bellanger, D.; López, C.; et al. Herling MJAK/STAT-Activating Genomic Alterations Are a Hallmark of T-PLL. Cancers 2019, 11, 1833. [Google Scholar] [CrossRef]
  28. Schrader, A.; Crispatzu, G.; Oberbeck, S.; Mayer, P.; Pützer, S.; von Jan, J.; Vasyutina, E.; Warner, K.; Weit, N.; Pflug, N.; et al. Actionable perturbations of damage responses by TCL1/ATM and epigenetic lesions form the basis of T-PLL. Nat. Commun. 2018, 9, 697. [Google Scholar] [CrossRef] [PubMed]
  29. Suzuki, R.; Matsushita, H.; Kawai, H.; Matsuzawa, H.; Tsuboi, K.; Watanabe, S.; Kawada, H.; Ogawa, Y.; Ando, K. Identification of a novel SEPT9-ABL1 fusion gene in a patient with T-cell prolymphocytic leukemia. Leuk. Res. Rep. 2014, 3, 54–57. [Google Scholar] [CrossRef] [PubMed]
  30. Braun, T.; von Jan, J.; Wahnschaffe, L.; Herling, M. Advances and Perspectives in the Treatment of T-PLL. Curr. Hematol. Malig. Rep. 2020, 15, 113–124. [Google Scholar] [CrossRef]
  31. Brothers, J.; Castillo, D.R.; Jeon, W.J.; Joung, B.; Linhares, Y. Partial response to venetoclax and ruxolitinib combination in a case of refractory T-prolymphocytic leukemia. Hematology 2023, 28, 2237342. [Google Scholar] [CrossRef] [PubMed]
  32. Colon Ramos, A.; Tarekegn, K.; Aujla, A.; Garcia de de Jesus, K.; Gupta, S. T-Cell Prolymphocytic Leukemia: An Overview of Current and Future Approaches. Cureus 2021, 13, e13237. [Google Scholar] [CrossRef]
  33. von Jan, J.; Timonen, S.; Braun, T.; Jiang, Q.; Ianevski, A.; Peng, Y.; McConnell, K.; Sindaco, P.; Müller, T.A.; Pützer, S.; et al. Optimizing drug combinations for T-PLL: Restoring DNA damage and P53-mediated apoptotic responses. Blood 2024, 144, 1595–1610. [Google Scholar] [CrossRef] [PubMed]
  34. Herling, M.; Dearden, C.; Zaja, F.; El-Sharkawi, D.; Ding, W.; Bellido, M.; Khot, A.; Tick, L.; Jacobsen, E.; Eyre, T.A.; et al. Limited efficacy for ibrutinib and venetoclax in T-prolymphocytic leukemia: Results from a phase 2 international study. Blood Adv. 2024, 8, 842–845. [Google Scholar] [CrossRef]
  35. Carlesso, N.; Frank, D.A.; Griffin, J.D. Tyrosyl phosphorylation and DNA binding activity of signal transducers and activators of transcription (STAT) proteins in hematopoietic cell lines transformed by Bcr/Abl. J. Exp. Med. 1996, 183, 811–820. [Google Scholar] [CrossRef]
  36. Berger, A.; Sexl, V.; Valent, P.; Moriggl, R. Inhibition of STAT5: A therapeutic option in BCR-ABL1-driven leukemia. Oncotarget 2014, 5, 9564–9576. [Google Scholar] [CrossRef] [PubMed]
  37. 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] [PubMed]
Figure 1. Morphology and immunophenotype of neoplastic cells in peripheral blood and bone marrow. (A) Peripheral smears revealed neoplastic lymphocytes of 9–10 µm, with irregular, often angulated nuclei and prominent nucleoli (1000× magnification). (B) Flow cytometry at diagnosis showed 54% lymphocytes, the majority of which demonstrated the following phenotypes: CD2+/CD3+/CD4+/CD5+/CD7+/CD8/CD52+/CD56. SSC indicates side scatter. Green gate: lymphocytes; orange gate: monocytes; blue gate: granulocytes. The numbers on the other scatter plots represent percentages of the quadrants. (C) Histology showed normocellular bone marrow with approximately 15% interstitial and vaguely nodular lymphoid infiltrate in the bone marrow (200× magnification, Naphthol AS-D chloracetate esterase staining. (D) Immunohistochemistry using TCL1 antibody (Abcam, Cambridge, UK, 1:100 dilution), 200× magnification.
Figure 1. Morphology and immunophenotype of neoplastic cells in peripheral blood and bone marrow. (A) Peripheral smears revealed neoplastic lymphocytes of 9–10 µm, with irregular, often angulated nuclei and prominent nucleoli (1000× magnification). (B) Flow cytometry at diagnosis showed 54% lymphocytes, the majority of which demonstrated the following phenotypes: CD2+/CD3+/CD4+/CD5+/CD7+/CD8/CD52+/CD56. SSC indicates side scatter. Green gate: lymphocytes; orange gate: monocytes; blue gate: granulocytes. The numbers on the other scatter plots represent percentages of the quadrants. (C) Histology showed normocellular bone marrow with approximately 15% interstitial and vaguely nodular lymphoid infiltrate in the bone marrow (200× magnification, Naphthol AS-D chloracetate esterase staining. (D) Immunohistochemistry using TCL1 antibody (Abcam, Cambridge, UK, 1:100 dilution), 200× magnification.
Hematolrep 17 00001 g001
Figure 2. Cytogenetic findings. (A) 47,XX, del(6)(q22q23), i(8)(q10), +i(8)(q10), t(9;22;18)(q34;q11;q21), inv(14)(q11;q32), der(15)t(8;15)(q22;p13), add(16)(p13) karyotype was seen in 5 out of 30 metaphases obtained from peripheral blood (an arrow marks the Philadelphia chromosome). Four metaphases showed the same abnormalities with the addition of add(17)(q25), while one metaphase showed the addition of +der(22). (B) Interphase cells showed one fusion, two red, and two green signals with BCR::ABL1 dual fusion probe (Abbott, Chicago, United States of America). Approximately half of the cells showed an additional fusion signal representing +der(22) (630×).
Figure 2. Cytogenetic findings. (A) 47,XX, del(6)(q22q23), i(8)(q10), +i(8)(q10), t(9;22;18)(q34;q11;q21), inv(14)(q11;q32), der(15)t(8;15)(q22;p13), add(16)(p13) karyotype was seen in 5 out of 30 metaphases obtained from peripheral blood (an arrow marks the Philadelphia chromosome). Four metaphases showed the same abnormalities with the addition of add(17)(q25), while one metaphase showed the addition of +der(22). (B) Interphase cells showed one fusion, two red, and two green signals with BCR::ABL1 dual fusion probe (Abbott, Chicago, United States of America). Approximately half of the cells showed an additional fusion signal representing +der(22) (630×).
Hematolrep 17 00001 g002
Figure 3. Results of Sanger sequencing. Based on alignment with reference RNA sequences, the transcript was identified as in-frame with the breakpoint showing fusion of BCR exon 6 to ABL1 exon 2.
Figure 3. Results of Sanger sequencing. Based on alignment with reference RNA sequences, the transcript was identified as in-frame with the breakpoint showing fusion of BCR exon 6 to ABL1 exon 2.
Hematolrep 17 00001 g003
Figure 4. Summary of therapies and clinical data. ALEM: alemtuzumab; CYCLO: cyclophosphamide; IMA: imatinib; CHOP: cyclophosphamide-, doxorubicin-, vincristine-, and prednisone-containing regimen; NILO: nilotinib.
Figure 4. Summary of therapies and clinical data. ALEM: alemtuzumab; CYCLO: cyclophosphamide; IMA: imatinib; CHOP: cyclophosphamide-, doxorubicin-, vincristine-, and prednisone-containing regimen; NILO: nilotinib.
Hematolrep 17 00001 g004
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

Vida, L.; Horváth, B.; Egyed, M.; Kajtár, B.; Alizadeh, H. The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome. Hematol. Rep. 2025, 17, 1. https://doi.org/10.3390/hematolrep17010001

AMA Style

Vida L, Horváth B, Egyed M, Kajtár B, Alizadeh H. The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome. Hematology Reports. 2025; 17(1):1. https://doi.org/10.3390/hematolrep17010001

Chicago/Turabian Style

Vida, Livia, Bálint Horváth, Miklós Egyed, Béla Kajtár, and Hussain Alizadeh. 2025. "The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome" Hematology Reports 17, no. 1: 1. https://doi.org/10.3390/hematolrep17010001

APA Style

Vida, L., Horváth, B., Egyed, M., Kajtár, B., & Alizadeh, H. (2025). The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome. Hematology Reports, 17(1), 1. https://doi.org/10.3390/hematolrep17010001

Article Metrics

Back to TopTop