1
|
Bardelli V, Arniani S, Pierini V, Nardelli C, Matteucci C, Lema Fernandez AG, Crocioni M, Cerrano M, Salutari P, Papayanidis C, Trappolini S, Giglio F, Mastaglio S, Zappasodi P, Pasciolla C, Defina M, Piccini M, Lanzarone G, Di Giacomo D, Sica S, Montefiori LE, Mullighan CG, Mecucci C, La Starza R. Repurposing the Whole Expression Transcriptome Assay for the Genetic Diagnosis of T-Cell Acute Lymphoblastic Leukemia and Lymphoma. J Mol Diagn 2025:S1525-1578(25)00040-6. [PMID: 39984035 DOI: 10.1016/j.jmoldx.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/25/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
Unlike other cases of acute leukemia, the diagnosis of T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL) is uniquely based on morphology and flow cytometry. Although the genomic background has been broadly uncovered, the large spectrum of genes involved and the variability of the molecular mechanisms underlying gene deregulation have delayed the introduction of molecular cytogenetics into diagnostic flowcharts. To overcome these limitations and implement a genetic diagnosis of T-ALL/LBLs, a whole transcriptome expression assay (WTEa) was repurposed as a "priority test" to classify T-ALL/LBLs into the major genetic subtypes. A WTEa classifier based on a set of 312 probes on 215 T-ALL/LBLs was set up and applied, which properly assigned >95% of cases with subtype-defining alterations to the corresponding subgroups (ie, TAL/LMO, HOXA, TLX1, TLX3, BCL11B). It pinpointed cases that harbored cryptic alterations, such as noncoding mutations that generate new enhancer at TAL1 and LMO2 loci (8% of TAL/LMO), and duplications of noncoding element downstream BCL11B (BETA) (18% of BCL11B). It was also suitable to classify lymphoma cases for which only formalin-fixed embedded tissues were available, as confirmed in cases harboring TLX1 or TLX3 rearrangements, and distinguished new putative subtypes. WTEa offers a unifying tool to provide a genetic classification of T-ALL/LBLs. If introduced in multicenter prospective studies, it will facilitate evaluation of the clinical impact of genetic classification.
Collapse
Affiliation(s)
- Valentina Bardelli
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Silvia Arniani
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Valentina Pierini
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Carlotta Nardelli
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Caterina Matteucci
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Anair Graciela Lema Fernandez
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Maria Crocioni
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Marco Cerrano
- Division of Hematology, A.O.U. Città della Salute e della Scienza, Turin, Italy
| | | | - Cristina Papayanidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "L. & A. Seràgnoli," Bologna, Italy
| | | | - Fabio Giglio
- OncoHematology Division, European Institute of Oncology, Milan, Italy
| | - Sara Mastaglio
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Patrizia Zappasodi
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Marzia Defina
- Hematology Unit, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Matteo Piccini
- Hematology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giuseppe Lanzarone
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Turin, Italy
| | - Danika Di Giacomo
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy; Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Simona Sica
- Haematology Unit, Department of Radiological and Hematological Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lindsey E Montefiori
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Section, Department of Medicine and Surgery, University of Perugia, Centro di Ricerche EmatoOncologiche, Azienda di Perugia (CREO A.O.), Perugia, Italy.
| |
Collapse
|
2
|
Pastorczak A, Urbanska Z, Styka B, Miarka-Walczyk K, Sedek L, Wypyszczak K, Wakulinska A, Nowicka Z, Szczepański T, Stańczak M, Fendler W, Kowalczyk J, Młynarski W, Lejman M. Genetic hallmarks and clinical implications of chromothripsis in childhood T-cell acute lymphoblastic leukemia. Leukemia 2024; 38:2344-2354. [PMID: 39192035 PMCID: PMC11518979 DOI: 10.1038/s41375-024-02370-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024]
Abstract
Chromothripsis (cth) is a form of genomic instability leading to massive de novo structural chromosome rearrangements in a one-time catastrophic event. It can cause cancer-promoting alterations, such as loss of sequences for tumor-suppressor genes, formation of oncogenic fusions, and oncogene amplifications. We investigated the genetic background and clinical significance of cth in childhood T-cell acute lymphoblastic leukemia (T-ALL) patients. For this purpose, whole-genome copy number alterations were analyzed in 173 children with newly diagnosed T-ALL using high-density microarrays. Cth was identified in 10 T-ALL samples (5.78%). In six of them, cth occurred in a constitutional background of Nijmegen breakage syndrome (n = 5) or Li-Fraumeni syndrome (n = 1). Cth generated alterations, including deletions of CDKN2A/B (n = 4) and EZH2 (n = 4), amplifications of CDK6 (n = 2), and NUP214::ABL1 and TFG::GPR128 fusions. Cth-positive leukemias exhibited deletions involving the tumor-suppressor genes RB1 (n = 3), TP53 (n = 1) and MED12 (n = 2). Cth-positive T-ALL patients had a lower probability of 5-year overall survival (OS) [0.56 vs. 0.81; hazard ratio (HR) = 4.14 (1.42-12.02) p = 0.017] as did 5-year event-free survival [0.45 vs. 0.74; HR = 3.91 (1.52-10.08); p = 0.012]. Chromothripsis is an infrequent genomic phenomenon in pediatric T-ALL but is significantly associated with cancer-predisposing syndromes and may associate with inferior prognosis.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland.
- Department of Genetic Predisposition to Cancer, Medical University of Lodz, Lodz, Poland.
| | - Zuzanna Urbanska
- Department of Genetic Predisposition to Cancer, Medical University of Lodz, Lodz, Poland
| | - Borys Styka
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| | | | - Lukasz Sedek
- Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Poland
| | - Kamila Wypyszczak
- Department of Genetic Predisposition to Cancer, Medical University of Lodz, Lodz, Poland
| | - Anna Wakulinska
- Department of Oncology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Poland
| | - Marcin Stańczak
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Jerzy Kowalczyk
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Wojciech Młynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
- Institute of Medical Expertises, Lodz, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
3
|
Hu X, Wang Z, Qin Y, Xu J, Xu N, Wang Q, Lin R, Zhao K, Zhou H, Xuan L, Yu S, Liu Q. Allogeneic haematopoietic stem cell transplantation might overcome the poor prognosis of adolescents and adult patients with T-lineage acute lymphoblastic leukaemia and CDKN2 deletion. Bone Marrow Transplant 2024; 59:1146-1153. [PMID: 38769349 DOI: 10.1038/s41409-024-02306-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
This study delves into the clinical implications of cyclin-dependent kinase inhibitor 2 (CDKN2) deletion in adult T-lineage acute lymphoblastic leukemia (T-ALL). Among 241 patients included in this study, 57 had CDKN2 deletion and 184 had CDKN2 wild-type (WT), and 165 underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) and 76 did not undergo allo-HSCT. CDKN2 deletion correlated with higher white blood cell count, more high-risk diseases, and complex karyotype. The 5-year overall survival (OS) was 36.8% and 58.2% (P < 0.001), 5-year disease-free survival (DFS) was 47.1% and 59.3% (P = 0.018), and 5-year cumulative incidence of relapse (CIR) was 33.7% and 22.3% (P = 0.019) in patients with CDKN2 deletion and WT, respectively. Multivariate analysis identified CDKN2 deletion as an independent adverse prognostic factor for OS (HR 2.11, P = 0.003). In the CDKN2 deletion subgroup, landmark analysis showed that the 5-year OS was 56.7% and 19% (P = 0.002) for patients who underwent allo-HSCT and those who did not, respectively. And multivariate analysis confirmed the beneficial role of allo-HSCT in OS (HR 0.23, P < 0.001). In conclusion, CDKN2 deletion was associated with a poor prognosis in adult T-ALL, and allo-HSCT might be beneficial for this population.
Collapse
Affiliation(s)
- Xiaoshan Hu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Zhixiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Yuting Qin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Jun Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Qiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Ke Zhao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Sijian Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China.
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China.
| |
Collapse
|
4
|
Balciuniene J, Ning Y, Lazarus HM, Aikawa V, Sherpa S, Zhang Y, Morrissette JJD. Cancer cytogenetics in a genomics world: Wedding the old with the new. Blood Rev 2024; 66:101209. [PMID: 38852016 DOI: 10.1016/j.blre.2024.101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/10/2024]
Abstract
Since the discovery of the Philadelphia chromosome in 1960, cytogenetic studies have been instrumental in detecting chromosomal abnormalities that can inform cancer diagnosis, treatment, and risk assessment efforts. The initial expansion of cancer cytogenetics was with fluorescence in situ hybridization (FISH) to assess submicroscopic alterations in dividing or non-dividing cells and has grown into the incorporation of chromosomal microarrays (CMA), and next generation sequencing (NGS). These molecular technologies add additional dimensions to the genomic assessment of cancers by uncovering cytogenetically invisible molecular markers. Rapid technological and bioinformatic advances in NGS are so promising that the idea of performing whole genome sequencing as part of routine patient care may soon become economically and logistically feasible. However, for now cytogenetic studies continue to play a major role in the diagnostic testing and subsequent assessments in leukemia with other genomic studies serving as complementary testing options for detection of actionable genomic abnormalities. In this review, we discuss the role of conventional cytogenetics (karyotyping, chromosome analysis) and FISH studies in hematological malignancies, highlighting the continued clinical utility of these techniques, the subtleties and complexities that are relevant to treating physicians and the unique strengths of cytogenetics that cannot yet be paralleled by the current high-throughput molecular technologies. Additionally, we describe how CMA, optical genome mapping (OGM), and NGS detect abnormalities that were beyond the capacity of cytogenetic studies and how an integrated approach (broad molecular testing) can contribute to the detection of actionable targets and variants in malignancies. Finally, we discuss advances in the field of genomic testing that are bridging the advantages of individual (single) cell based cytogenetic testing and broad genomic testing.
Collapse
Affiliation(s)
- Jorune Balciuniene
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yi Ning
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Vania Aikawa
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarina Sherpa
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Lizcova L, Prihodova E, Pavlistova L, Svobodova K, Mejstrikova E, Hrusak O, Luknarova P, Janotova I, Sramkova L, Stary J, Zemanova Z. Cytogenomic characterization of pediatric T-cell acute lymphoblastic leukemia reveals TCR rearrangements as predictive factors for exceptional prognosis. Mol Cytogenet 2024; 17:14. [PMID: 38783324 PMCID: PMC11118568 DOI: 10.1186/s13039-024-00682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND T-cell acute lymphoblastic leukemia (T-ALL) represents a rare and clinically and genetically heterogeneous disease that constitutes 10-15% of newly diagnosed pediatric ALL cases. Despite improved outcomes of these children, the survival rate after relapse is extremely poor. Moreover, the survivors must also endure the acute and long-term effects of intensive therapy. Although recent studies have identified a number of recurrent genomic aberrations in pediatric T-ALL, none of the changes is known to have prognostic significance. The aim of our study was to analyze the cytogenomic changes and their various combinations in bone marrow cells of children with T-ALL and to correlate our findings with the clinical features of the subjects and their treatment responses. RESULTS We performed a retrospective and prospective comprehensive cytogenomic analysis of consecutive cohort of 66 children (46 boys and 20 girls) with T-ALL treated according to BFM-based protocols and centrally investigated cytogenetics and immunophenotypes. Using combinations of cytogenomic methods (conventional cytogenetics, FISH, mFISH/mBAND, arrayCGH/SNP and MLPA), we identified chromosomal aberrations in vast majority of patients (91%). The most frequent findings involved the deletion of CDKN2A/CDKN2B genes (71%), T-cell receptor (TCR) loci translocations (27%), and TLX3 gene rearrangements (23%). All chromosomal changes occurred in various combinations and were rarely found as a single abnormality. Children with aberrations of TCR loci had a significantly better event free (p = 0.0034) and overall survival (p = 0.0074), all these patients are living in the first complete remission. None of the abnormalities was an independent predictor of an increased risk of relapse. CONCLUSIONS We identified a subgroup of patients with TCR aberrations (both TRA/TRD and TRB), who had an excellent prognosis in our cohort with 5-year EFS and OS of 100%, regardless of the presence of other abnormality or the translocation partner. Our data suggest that escalation of treatment intensity, which may be considered in subsets of T-ALL is not needed for nonHR (non-high risk) patients with TCR aberrations.
Collapse
Affiliation(s)
- Libuse Lizcova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - Eva Prihodova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Lenka Pavlistova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Karla Svobodova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Ester Mejstrikova
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Ondrej Hrusak
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Pavla Luknarova
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Iveta Janotova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Lucie Sramkova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Zuzana Zemanova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
6
|
Alabed HBR, Pellegrino RM, Buratta S, Lema Fernandez AG, La Starza R, Urbanelli L, Mecucci C, Emiliani C, Gorello P. Metabolic Profiling as an Approach to Differentiate T-Cell Acute Lymphoblastic Leukemia Cell Lines Belonging to the Same Genetic Subgroup. Int J Mol Sci 2024; 25:3921. [PMID: 38612731 PMCID: PMC11011837 DOI: 10.3390/ijms25073921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/24/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive tumor mainly affecting children and adolescents. It is driven by multiple genetic mutations that together define the leukemic phenotype. Interestingly, based on genetic alterations and/or deregulated expression, at least six genetic subgroups have been recognized. The TAL/LMO subgroup is one of the most represented genetic subgroups, characterizing 30-45% of pediatric T-ALL cases. The study of lipid and metabolic profiles is increasingly recognized as a valuable tool for comprehending the development and progression of tumors. In this study, metabolic and lipidomic analysis via LC/MS have been carried out on four T-ALL cell lines belonging to the TAL/LMO subgroup (Jurkat, Molt-4, Molt-16, and CCRF-CEM) to identify new potential metabolic biomarkers and to provide a subclassification of T-ALL cell lines belonging to the same subgroup. A total of 343 metabolites were annotated, including 126 polar metabolites and 217 lipid molecules. The statistical analysis, for both metabolic and lipid profiles, shows significant differences and similarities among the four cell lines. The Molt-4 cell line is the most distant cell line and CCRF-CEM shows a high activity in specific pathways when compared to the other cell lines, while Molt-16 and Jurkat show a similar metabolic profile. Additionally, this study highlighted the pathways that differ in each cell line and the possible enzymes involved using bioinformatic tools, capable of predicting the pathways involved by studying the differences in the metabolic profiles. This experiment offers an approach to differentiate T-ALL cell lines and could open the way to verify and confirm the obtained results directly in patients.
Collapse
Affiliation(s)
- Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Anair Graciela Lema Fernandez
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (A.G.L.F.); (C.M.)
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (A.G.L.F.); (C.M.)
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Unit, Laboratory of Molecular Medicine (CREO), Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (A.G.L.F.); (C.M.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Paolo Gorello
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06100 Perugia, Italy (R.M.P.); (S.B.); (L.U.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
7
|
Almeida A, T'Sas S, Pagliaro L, Fijalkowski I, Sleeckx W, Van Steenberge H, Zamponi R, Lintermans B, Van Loocke W, Palhais B, Reekmans A, Bardelli V, Demoen L, Reunes L, Deforce D, Van Nieuwerburgh F, Kentsis A, Ntziachristos P, Van Roy N, De Moerloose B, Mecucci C, La Starza R, Roti G, Goossens S, Van Vlierberghe P, Pieters T. Myb overexpression synergizes with the loss of Pten and is a dependency factor and therapeutic target in T-cell lymphoblastic leukemia. Hemasphere 2024; 8:e51. [PMID: 38463444 PMCID: PMC10924755 DOI: 10.1002/hem3.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/28/2024] [Indexed: 03/12/2024] Open
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has improved over time, the outcome of T-ALL patients with primary resistant or relapsed leukemia remains poor. Therefore, further progress in the treatment of T-ALL requires a better understanding of its biology and the development of more effective precision oncologic therapies. The proto-oncogene MYB is highly expressed in diverse hematologic malignancies, including T-ALLs with genomic aberrations that further potentiate its expression and activity. Previous studies have associated MYB with a malignant role in the pathogenesis of several cancers. However, its role in the induction and maintenance of T-ALL remains relatively poorly understood. In this study, we found that an increased copy number of MYB is associated with higher MYB expression levels, and might be associated with inferior event-free survival of pediatric T-ALL patients. Using our previously described conditional Myb overexpression mice, we generated two distinct MYB-driven T-ALL mouse models. We demonstrated that the overexpression of Myb synergizes with Pten deletion but not with the overexpression of Lmo2 to accelerate the development of T-cell lymphoblastic leukemias. We also showed that MYB is a dependency factor in T-ALL since RNA interference of Myb blocked cell cycle progression and induced apoptosis in both human and murine T-ALL cell lines. Finally, we provide preclinical evidence that targeting the transcriptional activity of MYB can be a useful therapeutic strategy for the treatment of T-ALL.
Collapse
Affiliation(s)
- André Almeida
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Sara T'Sas
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Luca Pagliaro
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Igor Fijalkowski
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Wouter Sleeckx
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Hannah Van Steenberge
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | | | - Béatrice Lintermans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Wouter Van Loocke
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Bruno Palhais
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Alexandra Reekmans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Valentina Bardelli
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Lisa Demoen
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Lindy Reunes
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical BiotechnologyGhent UniversityGhentBelgium
| | | | - Alex Kentsis
- Tow Center for Developmental Oncology, Sloan Kettering Institute and Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Panagiotis Ntziachristos
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Lab for Translational Oncogenomics and Bioinformatics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Pediatric Precision Oncology Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Pediatric Hematology‐OncologyGhent University HospitalGhentBelgium
| | - Cristina Mecucci
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Roberta La Starza
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Giovanni Roti
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Pieter Van Vlierberghe
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Tim Pieters
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| |
Collapse
|
8
|
De Bie J, Quessada J, Tueur G, Lefebvre C, Luquet I, Toujani S, Cuccuini W, Lafage-Pochitaloff M, Michaux L. Cytogenetics in the management of T-cell acute lymphoblastic leukemia (T-ALL): Guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103431. [PMID: 38016418 DOI: 10.1016/j.retram.2023.103431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
Molecular analysis is the hallmark of T-cell acute lymphoblastic leukemia (T-ALL) categorization. Several T-ALL sub-groups are well recognized based on the aberrant expression of specific transcription factors. This recently resulted in the implementation of eight provisional T-ALL entities into the novel 2022 International Consensus Classification, albeit not into the updated World Health Organization classification system. Despite this extensive molecular characterization, cytogenetic analysis remains the backbone of T-ALL diagnosis in many countries as chromosome banding analysis and fluorescence in situ hybridization are relatively inexpensive techniques to obtain results of diagnostic, prognostic and therapeutic interest. Here, we provide an overview of recurrent chromosomal abnormalities detectable in T-ALL patients and propose guidelines regarding their detection. By referring in parallel to the more general molecular classification approach, we hope to offer a diagnostic framework useful in a broad clinical genetic setting.
Collapse
Affiliation(s)
- Jolien De Bie
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Julie Quessada
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France; CRCM, Inserm UMR1068, CNRS UMR7258, Aix Marseille Université U105, Institut Paoli Calmettes, Marseille 13009, France
| | - Giulia Tueur
- Laboratoire d'hématologie, Hôpital Avicenne, AP-HP, Bobigny 93000, France
| | - Christine Lefebvre
- Unité de Génétique des Hémopathies, Service d'Hématologie Biologique, CHU Grenoble Alpes, Grenoble 38000, France
| | - Isabelle Luquet
- Laboratoire d'Hématologie, CHU Toulouse (IUCT-O), Toulouse 31000, France
| | - Saloua Toujani
- Service de Cytogénétique et Biologie Cellulaire, CHU de Rennes, Rennes 35033, France
| | - Wendy Cuccuini
- Laboratoire d'Hématologie, Unité de Cytogénétique, Hôpital Saint-Louis, AP-HP, Paris 75010, France
| | - Marina Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Lucienne Michaux
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium; Katholieke Universiteit Leuven, Leuven 3000, Belgium.
| |
Collapse
|
9
|
Zhang X, Cui B, Li Y, Li Z, Zheng J, Chu X, Xiao P, Lu J, Wang Z, Cen J, Liu Y, Hu S. Transcriptome sequencing identifies novel EVX fusions involved in transcriptional activation of HOX family genes in pediatric immature T-cell acute lymphoblastic leukemia: two cases reports and a literature review. Int J Hematol 2023; 118:508-513. [PMID: 37243888 DOI: 10.1007/s12185-023-03619-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
Driver genomic alterations in pediatric immature T-cell acute lymphoblastic leukemia are not fully known. We report two cases of novel EVX fusions involved in the transcriptional activation of HOX family genes, ETV6::EVX2 and MSI2::EVX1/HOXA13, which activate HOXD and HOXA cluster genes transcription through enhancer hijacking. HOXA and HOXD were the only key transcription factors activated in these cases, which indicates their important roles in leukemogenesis. Our findings elucidate potential drivers for development of T-cell lymphoblastic leukemia, and are valuable for diagnosis and risk stratification of pediatric T-ALL in the era of precision medicine.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Bowen Cui
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yizhen Li
- Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place MS313, Memphis, TN, 38105, USA
| | - Zhiheng Li
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Jiajia Zheng
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xinran Chu
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Peifang Xiao
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Jun Lu
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zheng Wang
- Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jiannong Cen
- Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yu Liu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Shaoyan Hu
- Department of Hematology, Jiangsu Children's Hematology and Oncology Center, and the Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
10
|
Liu Q, Yu X, Wen J, Yin N, Liao X, Zou P, Guo Y, Song L, Xiao J. Genomic signatures and prognosis of advanced stage Chinese pediatric T cell lymphoblastic lymphoma by whole exome sequencing. Front Pediatr 2023; 11:1224966. [PMID: 37664545 PMCID: PMC10469305 DOI: 10.3389/fped.2023.1224966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Objective To investigate the genomic signatures and prognosis of advanced-stage T cell lymphoblastic lymphoma (T-LBL) and to examine the relationship between T-LBL and T cell acute lymphoblastic leukemia (T-ALL). Methods 35 Chinese T-LBL children with stage III or IV disease were recruited for this study. They were treated with combination chemotherapy and whole exome sequencing. The relationship of the clinical features, prognosis and specific gene mutations was researched. Gene chips of T-LBL and T-ALL were downloaded from a database, and differential gene expression was analyzed. Results Germline causal gene mutations (CARS or MAP2K2) were detected in 2 patients; 3.06 ± 2.21 somatic causal gene mutations were identified in the 35 patients, and somatic mutations were observed in the NOTCH1, FBXW7, PHF6 and JAK3 genes. NOTCH1 mutations were significantly associated with FBXW7 mutations, and the age at diagnosis of patients with NOTCH1-FBXW7 mutations was less than that of patients without such mutations (P < 0.05). 32 patients achieved complete remission (CR), and 14 and 18 patients were classified into the intermediate risk (IR) group and high risk (HR) group. During a median follow-up of 44 months, 3 patients relapsed. Three-year prospective event free survival (pEFS) was 82.286%, and no significant differences of pEFS were found for different sexes, ages, or statuses of NOTCH1-FBXW7 mutations, (P > 0.05); however, the mean survival time of the IR group was longer than that of the HR group (P < 0.05). Differential expression of genes in the T-LBL and/or T-ALL datasets was analyzed using the R package limma, and 1/3 of the differentially expressed genes were found in both the T-ALL and T-LBL datasets. High expression of PI3K-Akt signal pathway genes and the USP34 gene was found in the T-LBL dataset. Conclusion Although T-ALL and T-LBL both originate from precursor T-cells and are considered different manifestations of the same disease and the outcome of T-LBL is favorable when using T-ALL-based chemotherapy, there are differences in the gene distribution between T-LBL and T-ALL. It seems that the PI3K-Akt signaling pathway and the USP34 gene play important roles in T-LBL, but medicines targeting the USP34 gene or the PI3K-Akt pathway may be invalid.
Collapse
Affiliation(s)
- Qinglin Liu
- First Clinical College of Chongqing Medical University, Chongqing, China
| | - Xiang Yu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jinquan Wen
- Department of Pediatric Hematology, Hospital of Xianyang Caihong, Shaanxi, China
| | - Nange Yin
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Department of Pharmacy, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Liao
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Department of Hematology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Pinli Zou
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Department of Hematology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxia Guo
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Department of Hematology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Song
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Department of Pharmacy, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jianwen Xiao
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Hematology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|
11
|
Weichert-Leahey N, Shi H, Tao T, Oldridge DA, Durbin AD, Abraham BJ, Zimmerman MW, Zhu S, Wood AC, Reyon D, Joung JK, Young RA, Diskin SJ, Maris JM, Look AT. Genetic predisposition to neuroblastoma results from a regulatory polymorphism that promotes the adrenergic cell state. J Clin Invest 2023; 133:e166919. [PMID: 37183825 PMCID: PMC10178836 DOI: 10.1172/jci166919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/14/2023] [Indexed: 05/16/2023] Open
Abstract
Childhood neuroblastomas exhibit plasticity between an undifferentiated neural crest-like mesenchymal cell state and a more differentiated sympathetic adrenergic cell state. These cell states are governed by autoregulatory transcriptional loops called core regulatory circuitries (CRCs), which drive the early development of sympathetic neuronal progenitors from migratory neural crest cells during embryogenesis. The adrenergic cell identity of neuroblastoma requires LMO1 as a transcriptional cofactor. Both LMO1 expression levels and the risk of developing neuroblastoma in children are associated with a single nucleotide polymorphism, G/T, that affects a GATA motif in the first intron of LMO1. Here, we showed that WT zebrafish with the GATA genotype developed adrenergic neuroblastoma, while knock-in of the protective TATA allele at this locus reduced the penetrance of MYCN-driven tumors, which were restricted to the mesenchymal cell state. Whole genome sequencing of childhood neuroblastomas demonstrated that TATA/TATA tumors also exhibited a mesenchymal cell state and were low risk at diagnosis. Thus, conversion of the regulatory GATA to a TATA allele in the first intron of LMO1 reduced the neuroblastoma-initiation rate by preventing formation of the adrenergic cell state. This mechanism was conserved over 400 million years of evolution, separating zebrafish and humans.
Collapse
Affiliation(s)
- Nina Weichert-Leahey
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Hui Shi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ting Tao
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Derek A. Oldridge
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam D. Durbin
- Department of Oncology and Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Brian J. Abraham
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Mark W. Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center, Rochester, Minnesota, USA
| | - Andrew C. Wood
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand
| | - Deepak Reyon
- Molecular Pathology Unit, Center for Computational and Integrative Biology, and Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Keith Joung
- Molecular Pathology Unit, Center for Computational and Integrative Biology, and Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Biology Department, MIT, Cambridge, Massachusetts, USA
| | - Sharon J. Diskin
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - A. Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Weichert-Leahey N, Shi H, Tao T, Oldridge DA, Durbin AD, Abraham BJ, Zimmerman MW, Zhu S, Wood AC, Reyon D, Joung JK, Young RA, Diskin SJ, Maris JM, Look AT. Genetic Predisposition to Neuroblastoma Results from a Regulatory Polymorphism that Promotes the Adrenergic Cell State. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530457. [PMID: 36909587 PMCID: PMC10002714 DOI: 10.1101/2023.02.28.530457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Childhood neuroblastomas exhibit plasticity between an undifferentiated neural crest-like "mesenchymal" cell state and a more differentiated sympathetic "adrenergic" cell state. These cell states are governed by autoregulatory transcriptional loops called core regulatory circuitries (CRCs), which drive the early development of sympathetic neuronal progenitors from migratory neural crest cells during embryogenesis. The adrenergic cell identity of neuroblastoma requires LMO1 as a transcriptional co-factor. Both LMO1 expression levels and the risk of developing neuroblastoma in children are associated with a single nucleotide polymorphism G/T that affects a G ATA motif in the first intron of LMO1. Here we show that wild-type zebrafish with the G ATA genotype develop adrenergic neuroblastoma, while knock-in of the protective T ATA allele at this locus reduces the penetrance of MYCN-driven tumors, which are restricted to the mesenchymal cell state. Whole genome sequencing of childhood neuroblastomas demonstrates that T ATA/ T ATA tumors also exhibit a mesenchymal cell state and are low risk at diagnosis. Thus, conversion of the regulatory G ATA to a T ATA allele in the first intron of LMO1 reduces the neuroblastoma initiation rate by preventing formation of the adrenergic cell state, a mechanism that is conserved over 400 million years of evolution separating zebrafish and humans.
Collapse
|
13
|
Thomas X. T-cell acute lymphoblastic leukemia: promising experimental drugs in clinical development. Expert Opin Investig Drugs 2023; 32:37-52. [PMID: 36541671 DOI: 10.1080/13543784.2023.2161361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Despite advances in treatment approaches in acute lymphoblastic leukemia (ALL), the prognosis of adults with newly diagnosed T-ALL remains poor, as well as that of adults and children with relapsed disease. Novel targeted therapies are therefore needed. AREAS COVERED This review summarizes promising emerging strategies for the treatment of T-ALL. EXPERT OPINION The recent molecular characterization of T-ALL has led to the identification of new therapeutic targets. Small-molecules inhibitors and other targeted therapies have therefore been recently developed and are currently under clinical investigations. Similarly, first studies involving monoclonal antibodies and chimeric antigen receptor (CAR) T cells have shown encouraging results. Improvement of outcome with these novel approaches, eventually combined with current standard chemotherapy, is therefore expected in a near future in T-ALL.
Collapse
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Department of Clinical Hematology, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
14
|
Leoncin M, La Starza R, Roti G, Pagliaro L, Bassan R, Mecucci C. Modern treatment approaches to adult acute T-lymphoblastic and myeloid/T-lymphoblastic leukemia: from current standards to precision medicine. Curr Opin Oncol 2022; 34:738-747. [PMID: 36017547 DOI: 10.1097/cco.0000000000000900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To review the most recent advancements in the management of adult T-cell acute lymphoblastic leukemia (T-ALL), we summarize insights into molecular diagnostics, immunotherapy, targeted therapy and new techniques of drug sensitivity profiling that may support further therapeutic progress in T-ALL subsets. RECENT FINDINGS With current induction/consolidation chemotherapy and/or risk-oriented allogeneic stem cell transplantation programs up to 95% adult T-ALL patients achieve a remission and >50% (up to 80% in adolescents and young adults) are cured. The group of patients who fail upfront therapy, between 25% and 40%, is enriched in high-risk characteristics (unfavorable genetics, persistent minimal residual disease) and represents the ideal setting for the study of molecular mechanisms of disease resistance, and consequently explore novel ways of restoration of drug sensitivity and assess patient/subset-specific patterns of drug vulnerability to targeting agents, immunotherapy and cell therapy. SUMMARY The emerging evidence supports the contention that precision medicine may soon allow valuable therapeutic chances to adult patients with high-risk T-ALL. The ongoing challenge is to identify the best way to integrate all these new data into the therapeutic path of newly diagnosed patients, with a view to optimize the individual treatment plan and increase the cure rate.
Collapse
Affiliation(s)
- Matteo Leoncin
- Hematology Unit, Azienda Ulss3 Serenissima, Ospedale dell'Angelo, Venezia-Mestre
| | | | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Renato Bassan
- Hematology Unit, Azienda Ulss3 Serenissima, Ospedale dell'Angelo, Venezia-Mestre
| | - Cristina Mecucci
- Department of Medicine and Surgery, University of Perugia, Perugia
| |
Collapse
|
15
|
Early T-Cell Precursor ALL and Beyond: Immature and Ambiguous Lineage T-ALL Subsets. Cancers (Basel) 2022; 14:cancers14081873. [PMID: 35454781 PMCID: PMC9030030 DOI: 10.3390/cancers14081873] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immature T-cell acute lymphoblastic leukemias englobes a wide range of low prevalence subtypes, not well identified, that in some cases overlap with myeloid lineage subtypes. Globally, this “grey zone” of immature leukemias, are difficult to precisely diagnose using a classical immunophenotypic approach. Interesting, genomic data collected during last years has shown that these subtypes share several genomic alterations, raising the question of how their phenotypes reflect distinct AL entities. Here we provide a systematic overview of the genetic events associated with immature T-ALL and outline their relationship with treatment choices and outcomes. Our goal is to offer a basis for using the genetic information for new diagnostic algorithms. An immunogenetic classification of these immature subtypes will better stratify patients and improve their management with more efficient and personalized therapeutic options. Abstract A wide range of immature acute leukemias (AL), ranging from acute myeloid leukemias with minimal differentiation to acute leukemias with an ambiguous lineage, i.e., acute undifferentiated leukemias and mixed phenotype acute leukemia with T- or B-plus myeloid markers, cannot be definitely assigned to a single cell lineage. This somewhat “grey zone” of AL expresses partly overlapping features with the most immature forms of T-cell acute lymphoblastic leukemia (T-ALL), i.e., early T-cell precursor ALL (ETP-ALL), near-ETP-ALL, and pro-T ALL. These are troublesome cases in terms of precise diagnosis because of their similarities and overlapping phenotypic features. Moreover, it has become evident that they share several genomic alterations, raising the question of how their phenotypes reflect distinct AL entities. The aim of this review was to provide a systematic overview of the genetic events associated with immature T-ALL and outline their relationship with treatment choices and outcomes, especially looking at the most recent preclinical and clinical studies. We wish to offer a basis for using the genetic information for new diagnostic algorithms, in order to better stratify patients and improve their management with more efficient and personalized therapeutic options. Understanding the genetic profile of this high-risk T-ALL subset is a prerequisite for changing the current clinical scenario.
Collapse
|
16
|
Chromothripsis is a frequent event and underlies typical genetic changes in early T-cell precursor lymphoblastic leukemia in adults. Leukemia 2022; 36:2577-2585. [PMID: 35974102 PMCID: PMC9613476 DOI: 10.1038/s41375-022-01671-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/08/2022]
Abstract
Chromothripsis is a mitotic catastrophe that arises from multiple double strand breaks and incorrect re-joining of one or a few chromosomes. We report on incidence, distribution, and features of chromothriptic events in T-cell acute lymphoblastic leukemias (T-ALL). SNP array was performed in 103 T-ALL (39 ETP/near ETP, 59 non-ETP, and 5 with unknown stage of differentiation), including 38 children and 65 adults. Chromothripsis was detected in 11.6% of all T-ALL and occurred only in adult cases with an immature phenotype (12/39 cases; 30%). It affected 1 to 4 chromosomes, and recurrently involved chromosomes 1, 6, 7, and 17. Abnormalities of genes typically associated with T-ALL were found at breakpoints of chromothripsis. In addition, it gave rise to new/rare alterations, such as, the SFPQ::ZFP36L2 fusion, reported in pediatric T-ALL, deletions of putative suppressors, such as IKZF2 and CSMD1, and amplification of the BCL2 gene. Compared to negative cases, chromothripsis positive T-ALL had a significantly higher level of MYCN expression, and a significant downregulation of RGCC, which is typically induced by TP53 in response to DNA damage. Furthermore we identified mutations and/or deletions of DNA repair/genome stability genes in all cases, and an association with NUP214 rearrangements in 33% of cases.
Collapse
|
17
|
Mallard C, Johnston M, Bobyn A, Nikolic A, Argiropoulos B, Chan J, Guilcher G, Gallo M. Hi-C detects genomic structural variants in peripheral blood of pediatric leukemia patients. Cold Spring Harb Mol Case Stud 2021; 8:mcs.a006157. [PMID: 34819303 PMCID: PMC8744495 DOI: 10.1101/mcs.a006157] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 11/25/2022] Open
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is often driven by chromosome translocations that result in recurrent and well-studied gene fusions. Currently, fluorescent in-situ hybridization probes are employed to detect candidate translocations in bone marrow samples from B-ALL patients. Recently Hi-C, a sequencing-based technique originally designed to reconstruct the three-dimensional architecture of the nuclear genome, was shown to effectively recognize structural variants. Here, we demonstrate that Hi-C can be used as a genome-wide assay to detect translocations and other structural variants of potential clinical interest. Structural variants were identified in both bone marrow and peripheral blood samples, including an ETV6-RUNX1 translocation present in one pediatric B-ALL patient. Our report provides proof-of-principle that Hi-C could be an effective strategy to globally detect driver structural variants in B-ALL peripheral blood specimens, reducing the need for invasive bone marrow biopsies and candidate-based clinical tests.
Collapse
|
18
|
T-Cell Acute Lymphoblastic Leukemia: Biomarkers and Their Clinical Usefulness. Genes (Basel) 2021; 12:genes12081118. [PMID: 34440292 PMCID: PMC8394887 DOI: 10.3390/genes12081118] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
T-cell acute lymphoblastic leukemias (T-ALL) are immature lymphoid tumors localizing in the bone marrow, mediastinum, central nervous system, and lymphoid organs. They account for 10-15% of pediatric and about 25% of adult acute lymphoblastic leukemia (ALL) cases. It is a widely heterogeneous disease that is caused by the co-occurrence of multiple genetic abnormalities, which are acquired over time, and once accumulated, lead to full-blown leukemia. Recurrently affected genes deregulate pivotal cell processes, such as cycling (CDKN1B, RB1, TP53), signaling transduction (RAS pathway, IL7R/JAK/STAT, PI3K/AKT), epigenetics (PRC2 members, PHF6), and protein translation (RPL10, CNOT3). A remarkable role is played by NOTCH1 and CDKN2A, as they are altered in more than half of the cases. The activation of the NOTCH1 signaling affects thymocyte specification and development, while CDKN2A haploinsufficiency/inactivation, promotes cell cycle progression. Among recurrently involved oncogenes, a major role is exerted by T-cell-specific transcription factors, whose deregulated expression interferes with normal thymocyte development and causes a stage-specific differentiation arrest. Hence, TAL and/or LMO deregulation is typical of T-ALL with a mature phenotype (sCD3 positive) that of TLX1, NKX2-1, or TLX3, of cortical T-ALL (CD1a positive); HOXA and MEF2C are instead over-expressed in subsets of Early T-cell Precursor (ETP; immature phenotype) and early T-ALL. Among immature T-ALL, genomic alterations, that cause BCL11B transcriptional deregulation, identify a specific genetic subgroup. Although comprehensive cytogenetic and molecular studies have shed light on the genetic background of T-ALL, biomarkers are not currently adopted in the diagnostic workup of T-ALL, and only a limited number of studies have assessed their clinical implications. In this review, we will focus on recurrent T-ALL abnormalities that define specific leukemogenic pathways and on oncogenes/oncosuppressors that can serve as diagnostic biomarkers. Moreover, we will discuss how the complex genomic profile of T-ALL can be used to address and test innovative/targeted therapeutic options.
Collapse
|
19
|
Bardelli V, Arniani S, Pierini V, Pierini T, Di Giacomo D, Gorello P, Moretti M, Pellanera F, Elia L, Vitale A, Storlazzi CT, Tolomeo D, Mastrodicasa E, Caniglia M, Chiaretti S, Ruggeri L, Roti G, Schwab C, Harrison CJ, Almeida A, Pieters T, Van Vlierberghe P, Mecucci C, La Starza R. MYB rearrangements and over-expression in T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2021; 60:482-488. [PMID: 33611795 DOI: 10.1002/gcc.22943] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022] Open
Abstract
We investigated MYB rearrangements (MYB-R) and the levels of MYB expression, in 331 pediatric and adult patients with T-cell acute lymphoblastic leukemia (T-ALL). MYB-R were detected in 17 cases and consisted of MYB tandem duplication (tdup) (= 14) or T cell receptor beta locus (TRB)-MYB (= 3). As previously reported, TRB-MYB was found only in children (1.6%) while MYB tdup occurred in both age groups, although it was slightly more frequent in children (5.2% vs 2.8%). Shared features of MYB-R T-ALL were a non-early T-cell precursor (ETP) phenotype, a high incidence of NOTCH1/FBXW7 mutations (81%) and CDKN2AB deletions (70.5%). Moreover, they mainly belonged to HOXA (=8), NKX2-1/2-2/TLX1 (=4), and TLX3 (=3) homeobox-related subgroups. Overall, MYB-R cases had significantly higher levels of MYB expression than MYB wild type (MYB-wt) cases, although high levels of MYB were detected in ~ 30% of MYB-wt T-ALL. Consistent with the transcriptional regulatory networks, cases with high MYB expression were significantly enriched within the TAL/LMO subgroup (P = .017). Interestingly, analysis of paired diagnosis/remission samples demonstrated that a high MYB expression was restricted to the leukemic clone. Our study has indicated that different mechanisms underlie MYB deregulation in 30%-40% of T-ALL and highlighted that, MYB has potential as predictive/prognostic marker and/or target for tailored therapy.
Collapse
Affiliation(s)
- Valentina Bardelli
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Silvia Arniani
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Valentina Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Tiziana Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Danika Di Giacomo
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Paolo Gorello
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Martina Moretti
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Fabrizia Pellanera
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Antonella Vitale
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | | | - Doron Tolomeo
- Department of Biology, University of Bari "Aldo Moro", Bari, Italy
| | - Elena Mastrodicasa
- Department of pediatric and gynecology, Pediatric Onco-hematology, Perugia Regional hospital, Perugia, Italy
| | - Maurizio Caniglia
- Department of pediatric and gynecology, Pediatric Onco-hematology, Perugia Regional hospital, Perugia, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Loredana Ruggeri
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Giovanni Roti
- Hematology and Bone Marrow Transplantation Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Claire Schwab
- Leukaemia Research Cytogenetic Group, Newcastle University Centre for Cancer, Newcastle-upon-Tyne, UK
| | - Christine J Harrison
- Leukaemia Research Cytogenetic Group, Newcastle University Centre for Cancer, Newcastle-upon-Tyne, UK
| | - André Almeida
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Tim Pieters
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| |
Collapse
|
20
|
Candoni A, Lazzarotto D, Ferrara F, Curti A, Lussana F, Papayannidis C, Del Principe MI, Bonifacio M, Mosna F, Delia M, Minetto P, Gottardi M, Fracchiolla N, Mancini V, Forghieri F, Zappasodi P, Cerrano M, Vitale A, Audisio E, Trappolini S, Romani C, Defina M, Imbergamo S, Ciccone N, Santoro L, Cambò B, Iaccarino S, Dargenio M, Aprile L, Chiaretti S, Fanin R, Pizzolo G, Foà R. Nelarabine as salvage therapy and bridge to allogeneic stem cell transplant in 118 adult patients with relapsed/refractory T-cell acute lymphoblastic leukemia/lymphoma. A CAMPUS ALL study. Am J Hematol 2020; 95:1466-1472. [PMID: 32777149 DOI: 10.1002/ajh.25957] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
The outcome of relapsed or refractory (R/R) T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/T-LBL) in adults is poor, with less than 20% of patients surviving at 5 years. Nelarabine is the only drug specifically approved for R/R T-ALL/T-LBL, but the information to support its use is based on limited available data. The aim of this observational phase four study was to provide recent additional data on the efficacy and safety of nelarabine in adults with R/R T-ALL/T-LBL and to evaluate the feasibility and outcome of allogeneic hematopoietic stem cell transplant (SCT) after salvage with nelarabine therapy. The primary endpoints were overall response rate (ORR) and overall survival (OS). Additional endpoints were safety, SCT rate and post-SCT OS. Between May 2007 and November 2018, 118 patients received nelarabine salvage therapy at 27 Italian hematology sites. The median age was 37 years (range 18-74 years), 73% were male, 77 had a diagnosis of T-ALL and 41 of T-LBL, and 65/118 (55%) had received more than two lines of therapy. The median number of nelarabine cycles was two (range 1-4); 43/118 (36%) patients had complete remission (CR), 16 had partial remission (14%) and 59 (50%) were refractory, with an ORR of 50%. The probability of OS, from the first dose of nelarabine, was 37% at 1 year with a median survival of 8 months. The OS at 1 year was significantly better for the 47 patients (40%) who underwent SCT after nelarabine salvage therapy (58% vs 22%, log-rank P < .001). The probability of OS at 2 and 5 years from SCT was 46% and 38%, respectively. Seventy-five patients (64%) experienced one or more drug-related adverse events (AE). Grade III-IV neurologic toxicities were observed in 9/118 (8%) of cases and thrombocytopenia or/and neutropenia (grade III-IV) were reported in 41% and 43% of cases, respectively. In conclusion, this is one of the largest cohorts of adult patients with R/R T-ALL/T-LBL treated in real life with nelarabine. Taking into account the poor prognosis of this patient population, nelarabine represents an effective option with an ORR of 50% and a CR rate of 36%. In addition, 40% of cases following nelarabine salvage therapy could undergo SCT with an expected OS at 2 and 5 years of 46% and 38%, respectively. The safety profile of nelarabine was acceptable with only 8% of cases showing grade III-IV neurological AE.
Collapse
Affiliation(s)
- Anna Candoni
- Clinica Ematologica Azienda Sanitaria Universitaria Integrata di Udine Udine Italy
| | - Davide Lazzarotto
- Clinica Ematologica Azienda Sanitaria Universitaria Integrata di Udine Udine Italy
| | | | - Antonio Curti
- Dipartimento di Oncologia ed Ematologia Policlinico S. Orsola‐Malpighi Bologna Italy
| | - Federico Lussana
- U.O.C. Ematologia Ospedale di Bergamo ASST Papa Giovanni XXIII Bergamo Italy
| | - Cristina Papayannidis
- Dipartimento di Oncologia ed Ematologia Policlinico S. Orsola‐Malpighi Bologna Italy
| | | | | | - Federico Mosna
- Ematologia e Centro Trapianto di Midollo Osseo Ospedale Regionale San Maurizio Bolzano Italy
| | - Mario Delia
- U.O. Ematologia con Trapianto Azienda Ospedaliero‐Universitaria Consorziale Bari Italy
| | - Paola Minetto
- Clinica Ematologica Ospedale San Martino Genoa Italy
| | | | - Nicola Fracchiolla
- U.O. Ematologia IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano Milan Italy
| | - Valentina Mancini
- Dipartimento di Ematologia e Oncologia Ospedale Niguarda Milan Italy
| | - Fabio Forghieri
- S.C. Ematologia Azienda Ospedaliero Universitaria di Modena Modena Italy
| | | | - Marco Cerrano
- S.C. Ematologia 1 A.O.U. Citta della Salute e della Scienza di Torino (Presidio Molinette) Torino Italy
| | - Antonella Vitale
- Ematologia, Dipartimento di Medicina Traslazionale e di Precisione "Sapienza" Università di Roma Rome Italy
| | | | - Silvia Trappolini
- S.O.D. Clinica Ematologica Azienda Ospedaliero Universitaria Ospedali Riuniti Ancona Italy
| | | | - Marzia Defina
- UOC Ematologia Azienda Ospedaliero Universitaria Senese Siena Italy
| | - Silvia Imbergamo
- U.O. di Ematologia e Immunologia Clinica Università degli Studi di Padova Padova Italy
| | - Nadia Ciccone
- S.C. Ematologia Azienda Ospedaliero Universitaria di Ferrara Ferrara Italy
| | - Lidia Santoro
- U.O.C. Ematologia e Trapianto di Midollo Osseo Avellino Italy
| | - Benedetta Cambò
- UOC Ematologia e Centro Trapianti di Midollo Osseo Azienda Ospedaliero Universitaria di Parma Parma Italy
| | | | | | - Lara Aprile
- S.C. Ematologia Ospedale S.G. Moscati Taranto Italy
| | - Sabina Chiaretti
- Ematologia, Dipartimento di Medicina Traslazionale e di Precisione "Sapienza" Università di Roma Rome Italy
| | - Renato Fanin
- Clinica Ematologica Azienda Sanitaria Universitaria Integrata di Udine Udine Italy
| | | | - Roberto Foà
- Ematologia, Dipartimento di Medicina Traslazionale e di Precisione "Sapienza" Università di Roma Rome Italy
| |
Collapse
|